, fury!- n. Humid. ‘ 3 l . g. , .. VF“: . h. .1: r I'. I. ,0... r. ”Mk I , t k t- if... .‘flz. g 4 |. furl". ... ho. ...\I‘cl. A .3. 5.1.. . . , . 11'- I!!!» D { . .. (1‘ ' mullllfil’filllllllllllwill mam 3129301787 9747 Michigan State University This is to certify that the dissertation entitled BIOACTIVE COMPOUNDS FROM OCIMUM SANCTUM LINN. LAMIACEAE presented by MARK A. KELM has been accepted towards fulfillment of the requirements for Ph.D. Horticulture degree in Major professor Date May 12, 1999 MS U is an Affirmative Action/Equal Opportunity Institution 0-12771 PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINE return on or before date due. MAY BE RECAU£D with earlier due date if requested. DATE DUE DATE DUE DATE DUE use muss-nu BIOACTIVE COMPOUNDS FROM OCIMUM SANCTUM LINN. LAMIACEAE By Mark Allen Kelm A DISSERTATION Submitted to Michigan State University in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Horticulture 1999 ABSTRACT BIOACTIVE COMPOUNDS FROM OCIMUM SANCTUM LINN. LAMIACEAE By Mark Allen Kelm A phytochemical investigation was conducted on various solvent extracts of the herb Ocimum sanctum Linn. Lamiaceae resulted in the isolation and characterization of mosquitocidal, anti-oxidant, and anti-inflammatory compounds. Mosquito-bioassay-guided fractionation led to the isolation of two mosquitocidal compounds, eugenol and (E)-6-hydroxy-4,6-dimethyl-3-heptene-2- one with minimum inhibitory concentrations at 200 and 6.25 ug-ml", respectively. A novel triglyceride, 1,3-dilinoleneoyl-Z-palmitin also was identified. In two other separate studies, anti-oxidant and anti-inflammatory assays yielded eugenol, cirsilineol, cirsimaritin, isothymonin, isothymonin, apigenin, and rosmarinic acid, which were isolated from the acetone extract of fresh leaves and stems of O. sanctum. Anti-oxidant activities of many of these compounds namely cirsilineol, isothymonin, and rosmarinic acid were comparable to synthetic anti-oxidants when assayed at 10 uM concentrations. At 1000 uM concentration nearly all of these compounds demonstrated very good anti-inflammatory activity as demonstrated by the inhibition of cyclo-oxygenase-1. In another study, corn eanNorm-bioassay-guided fractionation led to the characterization of a bioactive porphyrin that exhibited toxicity/anti-feedant activity at 100 ppm concentration. Structural identification and characterization was facilitated by the use of various spectroscopic experiments (NMR, MS, IR, and UV). Due to the plant’s anti-oxidant activity, it could potentially be used as a cancer preventative agent or even to slow the growth of existing malignant tissues. The anti-inflammatory activity 0. sanctum, in particular its ability to inhibit cyclo-oxygenase-2 could provide an alternative to currently available drugs. Incorporation of O. sanctum either as a ground powder or crude extracts into diet supplements or phytoceuticals could aid in the treatment of the aforementioned maladies. The use of O. sanctum has potential in the control of insect pests. Furthermore, because 0. sanctum is a plant, the acceptance and marketability of it would be enhanced considering the current lean towards natural remedies for human and agriculture pest management. ”GM/6m, @Md/Iw/ng ACKNOWLEDGMENTS I wish to thank my best friend and future wife, Sarah Breitkreutz for all the help, encouragement, understanding, and love that she has given through the course of my doctorate program. She has made a difficult task a lot less burdensome. I also wish to thank my family for the support they have given me during the last several years. The encouragement and support that they gave were deeply appreciated The advice and discussions academic and othenNise, provided by my lab mates; Dr. Amitabh Chandra, Dr. James Nitao, Dr. Russel Ramsewak, Dr. S. Balasubramanian (a.k.a. “Bala”), Andy Erickson, Dr. Haibo Wang, Dr. Yu-Chen Chang, and Dr. Holly Herner were also greatly valued and appreciated. A special thanks also goes to Di Zhang, Laura Clifford, Di Wu, and Rafikali Momin. Thanks to the guys at the NMR facility, Drs. Kermit Johnson and Long Lee. Finally I want like to thank the members of my graduate committee, Drs. Jack Kelly, Doug Gage, Bob Schutzki, Jim Miller, and my major advisor Dr. Muralee Nair for the guidance an advice they were able to offer. TABLE OF CONTENTS LIST OF FIGURES .............................................................................. vii LIST OF ABBREVIATIONS .................................................................. viii Introduction ........................................................................................... 1 CHAPTER I — Literature Review ................................................................ 5 CHAPTER II - Mosquitocidal compounds and a triglyceride, 1, 3-dilinoleneoyl-Z-palmitin from Ocimum sanctum Linn ............................... 28 CHAPTER III — Anti-oxidants from Ocimum sanctum Linn ............................. 39 CHAPTER IV - Anti-inflammatory compounds from Ocimum sanctum Linn ...... 60 CHAPTER V — A porphyrin compound from Ocimum sanctum Linn. with corn earworm anti-feedant/toxicity activity ........................................................ 68 CHAPTER VI - Summary ...................................................................... 76 REFERENCES .................................................................................... 80 vi LIST OF FIGURES Figure 1. Anti-oxidant activity of synthetic and isolated compounds assayed at 10 uM ................................................................................................ 59 Figure 2. Anti-oxidant activity at 21 minutes for synthetic and isolated compounds at 10 uM ............................................................................ 59 Figure 3. In vitro anti-inflammatory assay of eugenol, compounds 1-6 and synthetics. Eugenol, compounds 1-6, and aspirin tested at 1000 uM and ibuprofen and naproxen tested at 10 pM ................................................... 64 Figure 4. In vitro anti-inflammatory activity of eugenol, compounds 1,2,5,6 and aspirin tested at 1000 uM and ibuprofen tested at 10 uM ........................ 66 Figure 5. Corn eanNonn anti-feedant/toxic activity of two fractions containing isolated porphyrin. Significant at P < 0.005 ................................................ 73 vii BHA BHT CHCb cox 13CNMR DMSO DQFCOSY d dd DPA-PA DEPT E EIMS Et20 EtOH EtOAc FABMS GC 1HNMR HPLC LC m MeOH MIC MOPS MPLC MS m/z NMR PG PGHS-1 PGSH-2 PTLC s TBA TBHQ TLC UV 6 J VLC LIST OF ABBREVIATIONS Butylated hydroxyanisole Butylated hydroxytoluene Chloroform Cyclooxygenase Carbon nuclear magnetic resonance Dimethyl sulfoxide Double quantum filtered correlated spectroscopy Doublet Doublet of doublet 3-(p-(6-phenyl)-1 ,3,5-hexatrienyl)phenylpropionic acid Distortionless enhancement polarization transfer Entgegen Electron impact ionization mass spectroscopy Diethyl ether Ethanol Ethyl acetate Fast atom bombardment mass spectroscopy Gas chromatography Proton nuclear magnetic resonance High pressure lipid chromatography Lethal concentration Mulitplet Methanol Minimum inhibitory concentration 3-[N-morpholino] propanesulfonic acid Medium pressure lipid chromatography Mass spectroscopy Mass-to-charge ratio Nuclear magnetic resonance Prostaglandin Prostaglandin endoperoxide H synthase-1 Prostaglandin endoperoxide H synthase-2 Preparative thin layer chromatography Singlet 2-thiobarbituric acid tert-butylhydroquinone Thin layer chromatography Ultraviolet Chemical shifts Coupling constant Vacuum liquid chromatography viii INTRODUCTION Ocimum sanctum Linn. Lamiaceae has been used for generations in Southeast Asian traditional medicine and cuisine. O. sanctum (synonym O. tenuiflorum) more commonly known by its numerous vernacular names; Holy Basil, Sacred Basil, Tulsi (Unani), Tulasi, Tulssi, Surasa, Krishnamul, Vishnu- priya (Sanskrit), Kala-tulasi (Hindi), Krishna-tulasi (Bengali), and Thulasi (Tamil), has been used to treat a wide variety of ailments. This plant, as many of the common names imply, holds religious significance as well. Sacred Basil is thought to be the transformed nymph, Tulsi, the beloved of Lord Krishna, and thus has been regarded as sacred and as a consequence, it can be found in and around many Hindu temples and homes (Mallick et al., 1989). There are even some Christian sects that consider the herb as holy, since it can be found growing near the tomb of Christ. According to Ayurveda, the traditional Indian system of medicine, teas made from the leaves are prescribed for colds, stomach disorders, and as a source of nourishment for the old and weak (Thakur, 1989). In addition, the roots are alleged to posses aphrodisiac qualities. A decoction made from the roots has been used to treat malarial fever. The leaf juices have been applied to the skin for treatment of ringworms and other cutaneous afflictions (Butani, 1982). According to African folk medicine, 0. sanctum was used to repel mosquitoes and have a subterfuge and poultice-like effects (Batta and Santhakumari, 1970). II Besides its use in plantings around Hindu temples and homes, 0. sanctum has little horticultural importance. However, the plant has been considered as an alternative and cheaper source of the highly valued phenyl propanoid, eugenol (Asthana and Gupta, 1984). Eugenol can comprise 30-70% (Asthana and Gupta, 1984) of the essential oil and is used mainly in pharmaceutical formulations, perfumes, toiletries, and as a flavoring agent in foods. Currently eugenol is obtained from the costly spices clove, Eugenia caryophyllata L. and cinnamon, Cinnamon zeylanicum L. Much of the impetus for research on O. sanctum has been due to its role in traditional folk medicine. Most of these studies on O. sanctum were focused primarily on the bioactivity of crude extracts. These studies not only examined pharmacological activities, but also activities pertaining to agriculturally related pest problems such as insects, nematodes, and fungal diseases. Interestingly, a limited number of papers published on O. sanctum dealt with the isolation and identification of bioactive compounds and the determination of bioactivity of compounds previously identified in O. sanctum. Many studies pertaining to various biological activities have been completed on crude extracts from O. sanctum, as discussed in Chapter II. Also, a number of articles have focused more specifically on the bioactivities of compounds identified from O. sanctum, are discussed in Chapter II. It has been my hypothesis that given the body of pre-existing research, as well as preliminary studies carried out in the Bioactive Natural Products Laboratories (BNPL), O. sanctum has the potential to generate bioactive compounds for human and agricultural pest control. Therefore, the primary purpose of the current research was to isolate and identify the presence of biologically active compounds not previously examined. In addition, it was hoped that the present work will add to the body of phytochemcial knowledge that already exists for O. sanctum. O. sanctum plants were obtained initially from the Hare Krishna of Detroit and grown in the BNPL greenhouses, Department of Horticulture and National Food Safety and Toxicology Research Center, Michigan State University. As previously stated, primarily the crude extracts of O. sanctum have been examined for pharmacological as well as agricultural pest related studies. The potential health and pesticidal benefits from O. sanctum are therefore apparent given this body of literature and its long use as a traditional medicine. Therefore, I hypothesized that O. sanctum contains compound(s) capable of demonstrating antioxidant and anti-inflammatory activities. In addition, 0. sanctum contains compounds that possess insecticidal and/or antifeedant activity. This study, extracts and compounds were examined for biological activities. Insect bioassays utilizing mosquito larvae (Aedes aegyptii Linn.) and corn ean~orm (Heliocoverpa zea Hubner) were carried out to determine toxicity and antifeedant activities, respectively. Antioxidant and anti-inflammatory assays were conducted on isolated compounds. The body of this dissertation is comprised of several chapters. Each chapter, excluding the literature review is organized in a scientific journal format, which includes an abstract, introduction, materials and methods, and a results and discussion section. Chapter 1 LITERATURE REVIEW This chapter reviews the breadth of literature on the bioactivities of crude extracts and pure compounds from O. sanctum. Among the pharmacological activities reported for O. sanctum crude extracts were anti-carcinogenic, chemopreventative, anti-oxidant, radioprotective, anti-mutagenic, analgesic, anti- pyretic, anti-inflammatory, anti-spermatogenic, anti-fertility, hypoglycemic, anti- ulcerogenic, immunostimulatory, anti-stress, CNS effects, anti-bacterial, and effect on blood lipid profiles. Bioactivities associated with agricultural pest and other pest problems were; anti-fungal, anti-bacterial, insecticidal, pulicidal, nematicidal, and herbicidal activities. Papers about bioactivities of pure compounds from O. sanctum were far fewer, these consisted of antioxidant, hepatoprotective, anti-inflammatory, anti-hyperlipidemia, anti-tumor promotion, and anti-stress. Other miscellaneous articles pertaining to phytochemical studies on O. sanctum are presented. Kelm, M. A. and Nair, M. G. (submitted 1999) Bioactivity and Phytochemical review of Ocimum sanctum Linn. J. Ethnopannacology Pharmacologically related studies An investigation of the anticarcinogenic effects of the commonly consumed Indian herb, Tulsi revealed that it significantly decreased the occurrence of benzo[a]pyrene-induced neoplasia and 3'methyl-4- dimethylaminoazo benzene induced hepatomas in Swiss mice and Vlfinstar rats, respectively (Aruna and Sivaramakrishnan, 1992). Also, 0. sanctum exhibited chemopreventative activity against chemical dimethylbenz[a]anthracene (DMBA) induced carcinogenesis. In one study, a topical application of alcoholic O. sanctum leaf extract followed by application of DMBA resulted in a significant reduction in skin papillomagenesis of male Swiss albino mice (Prashar et al., 1994). The authors also noted a 25% increase in glutathione-S-transferase activity and a decrease in glutathione content in mouse skin following the topical application of O. sanctum leaf extract. In a later study, treatment of mice with the ethanolic extract introduced via gastric intubation and orally were found to be more effective than topical applications in the prevention of DMBA-induced tumor formation (Prashar and Kumar, 1995). Similar results were obtained using Syrian golden hamsters (Karthikeyan et al., 1999). Researchers determined the influence of alcoholic extracts on the activity of certain detoxifying enzymes (Banerjee et al., 1996). Oral doses of O. sanctum extract at 400 and 800 mg-kg'1 body weight for 15 days significantly elevated the activities of cytochrome p-450, cytochrome b5, aryl hydrocarbon hydroxylase, and glutathione-S-transferase (Banerjee et al., 1996). These enzymes are considered to be very important in the detoxification process of carcinogens, mutagens and other poisons. The anti-thyrodic and anti-oxidant activity of oral doses of aqueous extracts of O. sanctum was demonstrated by observing a rise in serum thyroxine concentrations and an increase in hepatic lipid peroxidation, glucose-6- phosphate, and endogenous anti-oxidant enzymes super oxide dismutase and catalase (Panda and Kar, 1998). In another study, it was determined that O. sanctum extract possesses free radical scavenging activity in vitro (Maulik et al., 1999). It is well known that free radicals, in particular oxygen free radicals, play a major role in the pathology of numerous diseases which include heart disease, arrhythmia, stroke, brain damage, respiratory syndrome, liver damage, cancer, and influenza (Mukhopadhyay et al., 1994 and Das and Maulik 1994). Ursolic acid, a triterpene, isolated from O. sanctum demonstrated protective effect against lipid peroxidation (Balanehru and Nagarajan, 1991). In a follow up article, Balanehru and Nagarajan (1992) found that ursolic acid prevented the cardiotoxicity of adriamycin by acting as an antioxidant. Furthermore, a pharmacological review article on ursolic acid discussed its hepatoprotective, anti-inflammatory, anti-tumor, and anti-hyperlipidemia activity (Liu, 1995). lntraperitoneal injections of 50 mg-kg‘1 followed by 10 mg-kg‘1 per day of O. sanctum extract for five consecutive days gave the best protection (70% survival) against whole body exposure to 60Co gamma radiation in albino mice (Devi et al., 1995). In a following report it was found that O. sanctum extracts gave in vivo protection against 60Co-gamma-radiation induced cytogenic damage (Ganasoundari et al., 1997). The authors speculated that the likely mechanism of the observed radioprotection involved free radical scavenging activity by the O. sanctum extracts administered interperitoneally. In a related article, researchers found that the same extracts were beneficial in the protection of bone marrow stem cells in adult Swiss mice against the deleterious effects of 6°Co gamma radiation (Ganasoundari et al., 1997). This report also demonstrated the lack of a toxic effect on the part of O. sanctum water extracts as compared to the well established radioprotector, WR-2721 (amifostine), an organic thiophosphate. Given this, 0. sanctum extracts may have an advantage over toxic WR-2721 in clinical uses as a natural and potentially benign substance. In a follow-up report, the same authors had shown the radioprotective effect of bone marrow by the administration of O. sanctum water extracts, in addition, the toxicity of WR-2721 was reduced by the presence of O. sanctum extracts which also resulted in an increase in the beneficial radioprotective effects synergistically (Ganasoundari et al., 1998). The authors also demonstrated a significant rise in chromosome protection by the use of O. sanctum extracts when administered intraperitoneally to Swiss mice. Again, it was suggested that the radioprotection was caused by the free radical scavenging qualities of O. sanctum extracts (Ganasoundari et al., 1998). The anti-mutagenic effect of O. sanctum juice were demonstrated in mice that were administered the bone marrow cell mutagens; methylmetnaesulfonate, mitomycin C, and dimethylnitroamine (Lim-Sylianco et al., 1988). O sanctum extracts were given orally by gavage, whereas the mutagens were given intraperitoneally. The analgesic activity of the oil from the seeds of O. sanctum was determined in rats and mice (Singh and Majumdar, 1995). Using the tail flick, tail clip, and tail immersion methods, researchers concluded that O. sanctum was not effective in raising the pain threshold, indicating that the O. sanctum is not centrally active. However, using the acetic-acid-induced writhing method, the oil showed a significantly elevated responses in a dose-dependent manner, which is indicative of peripherally mediated pain. A dose of 3 ml-kg‘1 of O. sanctum oil gave similar results as produced by a 100 mg-kg‘1 of aspirin. These treatments were given intraperitoneally. In an earlier study, it was observed that rises in hypouricemic and uricosuric effect followed by administration of Tulsi leaves or seeds in rabbits may result in a reduction of uric acid levels (Sarkar et al., 1990). High levels of uric acid are associated with gouty arthritis. These results may, therefore, substantiate one of the traditional uses of Tulsi as a treatment for gouty arthritis or other joint inflammations. Methanolic and water suspensions of O. sanctum both showed analgesic activity in the mouse hot plate method, anti-inflammatory activity in rats, and anti- pyretic activity in rats (Godwani et al., 1987). 500 mg-kg‘1 doses of the methanolic extract and water suspensions were found to achieve the same response as seen with 300 mg-kg’1 aspirin. However, at the same dose levels, the methanolic extract and the crude water suspension displayed weaker and shorter duration of analgesic activity than that of aspirin. The fixed oil of O. sanctum was 100% effective in the treatment of bovine mastitis after five days alone and after three days when combined with cloxacillin (Singh et al., 1995). Bovine mastitis effects the udder and is characterized by inflammation and bacterial infections largely due to Staphylococcus aureus. In another study, the anti-asthmatic and anti-inflammatory of ethanol extracts of both dry and fresh leaves of O. sanctum were analyzed. Guinea pigs were used for the study. Interestingly, only the fresh extracts along with the volatile and fixed oils had shown protection against histamine-induced acetyl choline pre- convulsive dyspnea (Singh and Agrawal, 1991). Results from anti-inflammatory studies involving carrageenan-induced hind paw edema indicated inhibition by fresh-leaf volatile and fixed oils. Furthermore, in a later study, anti-inflammatory activity was demonstrated by the oil from O. sanctum, since it inhibited arachidonic acid and leukotriene-induced paw edema. Furthermore, the authors postulated that O. sanctum may be used as a potential anti-inflammatory agent that blocks both cyclooxygenase and lipoxygenase pathways in arachidonic acid metabolism (Singh et al., 1996). 10 Non-polar (benzene, petroleum ether, and ether) and polar extracts (acetone and EtOH) of O.sanctum had shown 80-60% and less than 50% anti- fertility, respectively. Anti-fertility activity of O. sanctum extracts was determined by the examination of fertile female rats for the presence of spermatozoa in vaginal smears, followed by laparomety to determine the number of implantation sites (Batta and Santhakumari, 1970). High amounts of O.sanctum leaves in the diets of male albino mice decreased in seminal plasma pH, which corresponded to an increase in reducing substances such as alkaline phosphatase, acid phosphatase, mucoproteins, and electrolytes (Kasinathan et al., 1972). Histologically, there was a slight impairment of spermatogenesis. Taken together, the observed sterility was in large part due to these changes. A more recent report compliments these findings (Seth et al., 1981) in which a benzene leaf extract of O. sanctum reduced the count and motility of sperm in adult male rats. A reduction in the weight of testes was also observed. Furthermore, over the course of three months of diets including Tulsi leaves, the mating behavior of both male and female rats was severely inhibited (Khanna et al., 1986). This study also showed a decreased sperm count and sperm motility and weight reduction in male sex organs of rats over long-term exposure to Tulsi diets. Contrary to these studies by Khanna et al. (1986) and Kasinathan et al. (1972), researchers found an increase in testes size vs. controls and an increase in cone weight vs. control in 11 White leghorn male birds (Arneja et al., 1987). These studies, therefore, suggested that antifertility activity is species dependent. Incorporation of seeds and leaves of O. sanctum into artificial diets displayed a progressive and significant hypoglycemic effect during the first week in adult albino rabbits. Both seed- and leaf-containing diets reduced blood sugar levels when compared with the control. However, on a dry weight basis, the leaves were more effective at reducing blood sugar (Sarkar and Pant, 1989). Doses of 100, 150, 200, and 400 mg-kg'1 of Tulsi extract were given along with a normal diet to adult male Wistar rats. Significant decreases in sexual behavior were observed by those rats receiving 200 mg-kg'1 and 400 mg-kg'1 (Kantak and Gogate, 1992). Sexual behavior was observed and scored followed by presenting male rats with a primed ovariectomise female in which 2 pg of estrogen were administered the previous three days followed by progesterone on the fourth day. Reghunandanan et al., (1995) reported a reduction in testicular sperm count and glutamyl transpeptidase activity in Wistar rats after 24 h following intraperitoneal injections of benzene extract of O. sanctum leaves (300 make"). A definite lowering of blood sugar level in normal, glucose-fed hypoglycemic and streptozotocin-induced diabetic rats was observed following oral administration of alcoholic extracts of O.sanctum. Enhancement of the 12 action of exogenous insulin in normal rats also was observed (Chattopadhyay, 1993) Researchers concluded that O.sanctum extracts have anti-ulcerogenic properties against experimental ulcers, since it can cause a decrease in acid secretion and increased mucous secretion in VWstar albino rats (Mandel et al., 1993). O.sanctum-pretreated rat stomach mucosa was able to resist increases in gastric lesions caused by HCl-ethanol treatments. Pretreated rats also displayed a decrease in acidity and an increase in mucosal defense. Both studies (Mandel et al., 1993 and Janardhanan, et al., 1999) seem to justify the use of O.sanctum as an anti-ulcerogenic compound. Steam distillates from fresh 0. sanctum leaves were used in a study to determine its effects on humoral immune responses in vitro. It was found that O.sanctum could keep in check or maintain the humoral immune responses by acting on different aspects involved with the immune system such as antibody production, the release of mediators of hypersensitivity reactions, and tissue responses to mediators in target organs (Mediratta et al., 1988). A more recent study reported on the humoral immune response in albino rats by the quantification of agglutinating antibodies and E-rosette formation. Results showned an increase in antibody titre, E-rosette formation and lymphocytosis which are indicative of immunostimulation of humoral immunological responses (Godhwani et al., 1988). Diets containing dry powdered leaf of O.sanctum were 13 fed to birds at 500 mg-day'1 orally. lmmunomodulating and immunopotentiating activities by dried leaf of O.sanctum were shown in naturally IBD (infectious bursal disease) virus-infected poultry (Sadekar et al., 1998). IBD virus is one of the poultry industry's toughest problems. Connected in part with the immuno-stimulatory properties, researchers also discovered that O. sanctum possess anti-stress or adaptogenic properties (Wagner, et al., 1994, Sen et al., 1982, Bahrgava and Singh, 1981,) . O.sanctum was found to enhance physical endurance of swimming mice, inhibit stress- induced ulcers in rats, protect from carbon tetrachloride-induced hepatotoxicity in mice and rats, and inhibit milk-induced leucocytosis (Bahrgava and Singh, 1981). Sen and coworkers (1992) analyzed the effects of O. sanctum extract and eugenol (major component in O.sanctum) on various biochemical stress-induced changes. Both O.sanctum and eugenol reduced cholesterol levels caused by restraint stress in male Wistar rats (Sen et al., 1992). Also, O.sanctum and eugenol effectively lowered lactate dehydrogenase and alkaline phosphatase levels typically caused by stress (Sen et al., 1992). Red blood cell membrane dynamics also changed due to induction by restraint stress. Increased membrane protein clusterization, increased membrane fluidity and reduced membrane thickness occurred in response to stress; these are lessened or reversed in the presence of either O.sanctum or eugenol (Sen et al., 1992). 14 Action of O. sanctum on the central nervous system (CNS) was studied in mouse models (Ahumada et al., 1991 and Sakina et al., 1990). In these studies, it was found that O. sanctum extracts acted in synergy with the drug pentobarbital in depressing CNS responses. Sakina et al. (1990) reported a decrease in recovery time and severity due to electroshock and pentylenetetrazole-induced convulsions, in decreased apomorphine-induced fighting time and ability to walk in rodents. This study also indicated a possible interaction of O. sanctum extracts with dopaminergic neurons and a synergistic action when combined with bromocryptine, a potent Dz-receptor antagonist. In addition, longer narcosis times were observed in male mice compared to female mice (Ahumada et al., 1991). Anti-bacterial studies showed ethanolic extracts of O. sanctum to be effective against Staphylococcus aureus (100 rig-disc“), S. albus (100 jig-disc“), S. citreus (250 jig-disc“) and E. coli NCTC strain (500 jig-disc") (Phadke and Kulkarni, 1989). Water extracts of O. sanctum leaves exhibited anti-biotic activity against several strains of tuberculosis bacteria which included: Mycobacterium tuberculosis H37 Rv TMC102, M. tuberculosis SmR10(N)-1, and M. tuberculosis 1NHR Rv6 (Reddi et al., 1986). Also, 0. sanctum juice (30 ill-disc") exhibited inhibitory activity in assays utilizing Sarcina lutea. However, activity was not observed on Bacillus cereus, B. meaten'um, B. subtillis, S. epidermis, Proteus sp., Salmonella typhosa, and Vibrio cholerae (Ferdous et al., 1990). 15 The blood lipid profile of albino rats was significantly lower following the administration of fresh 0. sanctum leaves (Sarkar et al., 1994). Total serum cholesterol, triglycerides, phospholipids, and LDL cholesterol levels all were reduced significantly. In conjunction, blood HDL cholesterol and total fecal sterol contents were significantly greater. Together, these results suggest that O. sanctum has the potential to prevent or treat heart disease as well as other disorders associated with high fat intake. Agricultural Pest and Insect-Related Studies In addition to the pharmacologically related studies, agricultural insect control and related studies utilizing O. sanctum have been conducted. Much of the driving force behind this research was due to the widespread use of O. sanctum as a pest-managing agent. Only crude extracts of O. sanctum were studied for pest-managing activity. Methanol extracts of O. sanctum leaves exhibited some fungicidal and bacteriacidal activities on Tn'chophyton mertagrophytes and Bacillus subtilis (Ehrenburg) Cohn at concentrations greater than 104 ppm but were not active on Collectotn'chum Iagenan'um (Passerini) Ellis et Halstead, Pyn'culan'a oryzae Cavara, and Cochliobolus miyabeanus (Ito et Kuribayashi) Drechsler ex Dastur (Sukari and Takahashi, 1988). However, contradicting results were obtained with ethanolic leaf extracts of O. sanctum. These extracts inhibited the growth of 16 three fungal rice pathogens: rice blast; P. oryzae, brown spot; C. miyabeanus, and sheath blight; Rhizoctonia solani Kiihn when applied as a spray (2.5 g-l") on infected rice plants and in agar plate media at 5.0, 10.0, and 10.0 g-I", respectively (Tewari and Nayak, 1991). Ethanolic extracts and essential oil of O. sanctum showed fungicidal activity against another species of rice blast, P. gn’sea Sac. in both conidial germination and mycelial growth (Tewari, 1995). Ethanolic extracts also showed good performance on par with some synthetic fungicides (ediphenphos and carbendazim) in field studies (Tewar, 1995). Bananas, Musa spientum var. Malbhog pretreated with aqueous leaf extracts of O. sanctum then infected with the fungi; Botryodiplodia theobroma, Fusan'um theobromae, F. oxysporum, Helminthospon'um spiciferum, Culvularia lunata, Aspergillus flavus, and Tn'chothecium roseum resulted in significant retardation of fungal pathogen growth (Singh et al., 1993). Aqueous leaf extracts of O. sanctum effectively prevented conidial germination and slowed the growth of pre- and post-infected by H. spiceferum and F. scirpi in sponge gourd, Luffa cyclindn'ca L. (Ahmad and Prasad, 1995). Spore germination, spore growth, and cell wall degrading enzymes of the fruit rot pathogens, Botryodiplodia theobromae Pat. and Rhizopus anhizus Fisher were inhibited by O. sanctum leaf extracts (Patil, et al., 1992). A reduction of total protein content of the fungi also were observed following treatment with O. sanctum leaf extracts. Soxhletted ethanol extracts of both white and red strains of O. sanctum did not show any activity on the fish and shrimp bacterial pathogens, Acromonas 17 hydrophila, Streptococcus sp., and ten strains of Vibrio (Direkbusarakom et al., 1998). Contact toxicity bioassays on cockroach (Blatella gennanica L.) and red bean weevil (Callosbrunchus chinesis L.) gave 100% mortality due to application of 10 mg of O. sanctum essential oil (Sukari and Takahashi, 1988). Methanol leaf extracts of O. sanctum proved to be effective as antifeedants and repellents but were, not insecticidal against Anomis sabulifera Guen., jute semilooper larvae (Mallick and Banerji, 1989). Ethanolic and methanolic extracts of O. sanctum were bioassayed against the following aphid species: Myzus persicae (Sulzer), Metopolophium dirhodum (Walker), Aphis fabae (Scopoli), Sitobion avenae (Fabricius), and Acyrthosiphon pisum (Harris). Mortality of all species was observed following application of the test extract (Stein et al., 1988). The authors concluded that the observed aphidicidal activity cannot be due to eugenol, rather different compounds are more likely causing the observed effect. 0. sanctum extracts applied at a rate of 10 kg-ha‘1 on rice TKM9 seedlings resulted in only 5% survival of green leafnopper, Nephotettix virescens Dist. (Narasimhan and Mariappan, 1988). Only 33% of transmission rate for rice tungro virus was observed for plants treated with O. sanctum compared to the 75% observed in the control. When O. sanctum seed are placed in water, they exuded a mucilaginous substance, a polysaccharide of unknown structure. This substance has been found to effectively trap mosquito larvae (Culex fatigens) resulting in their death (Hasan and Dec, 1994). Hexane and acetone leaf 18 extracts also showed excellent to moderate toxicity on C. fatigans larvae, respectively (Deshmukh et al., 1982). Arrekul et al., (1988) found that crude extracts of O. sanctum a demonstrated repellent effect against the oriental fruit fly, Daucus doralis. Paradoxically, Roomi and coworkers (1993) found that O. sanctum leaf extracts were acting as attractants in field studies. Methyl eugenol, present in the essential oil, was used as a positive control which attracted Daucus spp. The oviposition deterrent effect of O. sanctum leaf extracts on spotted bollworm (En‘as Vite/Ia Fabr.) was demonstrated in a study by Sojitra and Patel (1992). A 5% leaf extract suspension of O. sanctum applied to okra plants (Hibiscus esculentus L.) was effective at reducing the number of eggs laid. In a later study, researchers (Adiroubane and Letchoumane, 1988) found that 2.5 and 5% water suspensions of O. sanctum ground leaf alone and in combination with synthetic pesticides (carbaryl 0.05% and endosulfan 0.035%) were significantly effective in controlling the population of leafhoppers, Amrasca bigulatta in the field. Using similar treatments, infestation of spotted bollworm, Ean'as spp. was significantly less than in the nontreated controls. Hexane and acetone leaf extracts of O. sanctum demonstrated pulicidal properties with LC50 at 0.05 and 1.45, respectively (Renapurkar and Deshmukh, 1984). Activities were comparable to dichloro-diphenyl-trichloroethane (DDT), 19 dieldrin, malathion, and fenthion with L050 values at 0.31, 0.39, 0.44, and 0.34, respectively. Chatterjee et al., (1982) reported nematicidal activity of eugenol and linalool against the nematode, Meliodogyne incognita. In another article, eugenol, linalool, cineole, and geraniol (see structures below) were found to be toxic against nematodes; Anguina tritici, Tylenchulus semipenetrans, M. javanica, and Heterodera cajani (Sangwan et al., 1990). Okra plants inoculated with M. incognita followed by treatments with water decoctions of O. sanctum leaves resulted in significant reductions in root protein levels and in the number of root galls (Santi et al., 1984). Also, 0. sanctum extracts were found to inhibit several enzymes completely, namely mitochondrial malate dehydrogenase and malic enzyme in the metabolic pathways of the filarial worm, Setan’a digitata (Banu et al., 1992). Phytochemical Studies The earliest phytochemical investigations of O. sanctum leaves were conducted by Nair et al., (1982). Among the compounds identified were as follows: ursolic acid, apigenin, luteolin, apigenin-7-O-glucuronide, luteolin-7-O- glucoronide, orientin, and molludistin. Oleanic acid also was reported from O. sanctum (Wagner, et al., 1994). 20 OH COOH R0 0 HO OH 0 ursolic acid R = H; apigenin R = glucose; apigenin-7-O-glucoside OH OH R0 0 OH O R =H; luteolin R = glucose; luteolin-7-O-glucoside OH OH HO OH Glc O OH HO HO O CH3O O OH O OH O orientin molludistin 21 Later the chemical composition of O. sanctum seed oil was determined (Malik et al., 1987). Seed oil represented 18.2% by weight of the seed. GC indicated the presence of lauric (2.84%), myristic (1.90%), palmitic (5.24%), stearic (3.12%), oleic (6.0%), linoleic (59.1%), and linolenic (21.7%) acids. lauric acid palmitic acid myristic acid 0 0 HOW Ito/“1% oleic acid steric acid HO 6 — 2 HO 6 —- 3 linoleic acid linolenic acid A reinvestigation of O. sanctum leaves led researchers to discover the presence of vicenin-2 (apigenin-6, 8-C-diglucoside), rosmarinic acid, galuteolin (luteolin-5-O-glucoside), cirsilineol (5, 4'-dihydrxy-6, 7, 3’-trimethoxyflavone), and OH 0 HO \ 0 OH COOH HO rosmarinic acid OH O vicenin-Z 22 Glc OR R = H; 4-allyl- l-O-B-Dglucopyranosyl- 2-hydroxybenzene R = CH3; 4-allyl-l-O-B-D-glucopyranosyl 2-methoxybenzene cirsilineol galuteolin two phenyl propane glucosides; 4-allyl-1-O-j3—D-glucopyranosyl-2- hydroxybenzene and -allyl-1-O-B—D-glucopyranosyl-2-methoxybenzene (Nbrr and Wagner, 1992). Also, gallic acid, gallic acid methyl ester, gallic acid ethyl ester, protocatechuic acid, vanillic acid, 4-hydroxybenzoic acid, vanillin, hydroxybenzaldehyde, caffeic acid, and chlorogenic acid were identified by UV spectroscopy and HPLC (Nbrr and Wagner, 1992). 23 l i 2.. OH HO OH HO\¢rOH HO : ,OH COOH coocn, COOCHZCH3 gallic acid gallic acid methyl ester gallic acid ethyl ester OH OH OH OH : ,OMC © COOH COOH COOH protocatechuic 301d vanillic acid 4-hydroxybenzoic acid OH OH OMc : OH CHO CHO .. OH vamllm 4-hydroxybenzaldehyde OH / OH O O HO H OH // HOOC H coon 0” caffeic acid chlorogenic acid 24 Sukari and coworkers (1995) isolated and identified stigmasterol, B—sitosterol and triacontanol ferulate from the dried bark of O. sanctum. stigmasterol OH CH O 09' I W“ l HO O B—sitosterol triacontanol ferulate GC-MS and FTIR experiments of the essential oils of O. sanctum led to the identification of 20 compounds in one study (Laakso et al., 1990) and 12 compounds in another study (Vasudevan et al., 1997). In the former study eugenol, B—bisabolenes, methyl eugenol, cineole, ocimene, humulene, and 8- caryophyllene were the major components derived from the steam and water- distilled oils. Linalool and geraniol were also identified. In the latter study, eugenol, and B-caryophyllene, methyl eugenol, and a number of unidentified compounds the major compounds found in the subcritical fluid extract. 25 OH OMe OMe : ,OMe eugenol methyl eugenol B—bisabolenes cineole / \ / / / l I“ B—ocimene a—humulene B—caryophyllene OH OH linalool geraniol 26 Given the above studies and the lack of studies pertaining to bioactive compounds, this work has shed more light on this deficient area of phytochemical research on O. sanctum. Through the process of bioassay-guided fractionation, compounds possessing insecticidal, anti-oxidant, and anti-inflammatory activity were discovered. 27 Chapter 2 MOSQUITOCIDAL COMPOUNDS AND A TRILGLYCERIDE, 1,3- DILlNOLENEOYL-Z-PALMITIN FROM OCIMUM SANCTUM LINN.* Abstract - The hexane extract of Ocimum sanctum was investigated using mosquito bioassay-guided fractionation and yielded compounds 1 and 2. The isolation of the triglyceride, 1, 3-dilinoleneoyI-2-palmitin (3) from O. sanctum leaves and stems is novel. The structures of these compounds were established using 1H- and 13CNMR spectral data. Compounds eugenol (1) and E-6-hydroxy- 4, 6-dimethyl-3-heptene-2-one (2) exhibited mosquitocidal activity at 200 and 6.25 ug-ml'1 in 24 h, respectively, on fourth instar Aedes aegyptii larvae. * M. A. Kelm and M. G. Nair (1998) Mosquitocidal compounds and a triglyceride, 1, 3-dilinoleneoyl-Z-palmitin, from Ocimum sanctum. J. Ag. Food Chem. 46, 3092-3094. 28 Introduction Ocimum sanctum L. (Lamiaceae) has been used for generations in Southeast Asian medicine and cuisine. O. sanctum or “Sacred Basil”, as the Ayurvedic herbal drug, has been used to treat a variety of human ailments (Thakur et al., 1989; Butani et al., 1982). Malarial fevers, ringworms, and other cutaneous afflictions also have been treated with this plant (Butani et al., 1982). According to African folk medicine, 0. sanctum was reported to repel mosquitoes, and have subterfuge and poultice effects (Batta and Santhakumari, 1970). Also, the crude extracts from O. sanctum have demonstrated biological activities against certain insects (Risvi, 1981). For example, crude alcoholic extracts exhibited aphidcidal properties (Stein et al., 1988), antifeedant activities on Jute semilooper, Anomis sabulifera (Malik and Rafique, 1989), and mosquito repellent and toxic properties (Batta and Santhakumari, 1970; Deshmukh et al., 1982). Previous phytochemical studies of O. sanctum have led to the isolation of flavones and flavone glycosides, including; apigenin (Nair et al., 1982), apigenin- 7-O-glucoside (Nair et al., 1982), cirsilineol (Nbrr and Wagner, 1992), galuteolin (Nbrr and Wagner, 1992), luteolin (Nair et al., 1982), luteolin-7-O-glucoside (Nair et al., 1982), molludistin (Nair et al., 1982), orientin (Nair et al., 1982), and vicenin-2 (Nbrr and Wagner, 1992). Other compounds identified in O. sanctum were eugenol (Laakso et al., 1990), rosmarinic acid (Nbrr and Wagner, 1992), 8- sitosterol (Sukari et al., 1995), stigmasterol (Sukari et al., 1995), triacontanol 29 ferulate (Sukari et al., 1995), and ursolic acid (Nair et al., 1982). Subsequently, the phenyl propane glycosides 4-allyl—1-O-8-D-glucopyranosyI-2-hydroxybenzene and 4-allyl-1-O-I3-D-glucopyranosyl-2-methoxybenzene were isolated (Nbrr and Wagner, 1992). GC-MS and GC-FTIR analyses of the essential oil of O. sanctum revealed the presence of 20 compounds (Laakso et al., 1990). From these studies eugenol, methyl chavicol, and B-bisbolenes were found to be the major components in the essential oil. Past research on O. sanctum focused on the biological activity of crude extract. Typically, phytochemical investigations carried out on O. sanctum extracts did not lend to the isolation and structural identification of biologically active compounds. The work in our laboratory, in part, involves the preliminary screening of many plant and microbial extracts in order to determine the presence of any biologically active compound. In this paper, we report, for the first time, the isolation and structure determination of a novel triglyceride (3) and two mosquitocidal compounds (1, 2) from the leaf and stem hexane extract of O. sanctum. Materials and Methods General Experimental. 1HNMR, DQFCOSY, NOESY, and HMQC spectra were recorded at 300 and 500MHz. 13CNMR and DEPT spectra were recorded at 126 MHz. Chemical shifts were recorded in CDCI3, and the values are in 6 30 (ppm) based on residual of CHCI3 7.24 and CDCI3 77.0. Coupling constants, J, are in Hz. EIMS were recorded at 70 eV. Particle size of silica gel used in VLC and MPLC was 35-70 pm. All PLTC purifications were carried out on 250- and or 500-um silica gel plates. Spots and bands were visualized under UV light (366 and 254 nm). Gas chromatographic analyses were performed utilizing a flame ionization detector which was set at 260°C. The capillary column used for the analysis was a DB-5 (30 m x 0.25 mm ID). The temperature for the analysis was programmed from 150° (4 min) to 250°C (5 min) at 4°C-min‘1 with a helium carrier gas at a linear velocity of 34 cm-sec’1 and with split injections. Plant Material. A voucher plant specimen (MSC 360851) was filed with the Beal-Darlington herbarium, Department of Botany and Plant Pathology, Michigan State University. Leaves and stems of O. sanctum were harvested from plants maintained in the Pesticide Research Center greenhouses at Michigan State University. Plant materials were then freeze-dried, milled, and stored at -20°C until extraction. Mosquitocidal Bioassay. Fourth instar mosquito larvae, Aedes aegyptii L. were reared in our laboratory from eggs (courtesy of Drs. Alexander Raikal and Alan Hays, Department of Entomology. Michigan State University). Eggs were hatched in 500 mL of distilled, degassed water prepared by sonication (30 min). 31 Approximately 5 mg of bovine liver powder was added to the water to provide a food source. After four days, the fourth instar mosquito larvae were ready for bioassay. At least 10 larvae were placed in 980 uL of degassed, distilled H20. To this, 20 pL of DMSO containing the appropriate concentration of test extract or purified compounds was added and left at room temperature. Extracts were tested at 250 ppm. Pure compounds were tested initially at 100-250 ppm then were diluted serially and subsequently bioassayed to determine LC1oo. 4 mL test tubes were used for the bioassay. There were three replicates per treatment. The number of dead larvae were recorded at 2-, 4- and 24-h intervals. The control tube containing at least 10 larvae received 20 uL of DMSO alone, and mortality was recorded as in the case of test compounds. These bioassays were conducted according to previously published works (Roth et al., 1998; Nitao et al., 1991; Nair et al., 1989). Saponification and Methylation of Compound 3. With stirring, 6.6 mg of 3 was treated with 5% NaOH in MeOH (1 mL) for 5 min. Methanolic 6N HCI then was added to acidify this solution. This material was dried under a stream of nitrogen. Diazomethane was prepared by reacting N-nitotroso-N-methyl urea with concentrated KOH solution under ether. As the diazomethane product formed, it dissolved into the organic ether phase. This yellow-colored ether solution containing the diazomethane product then was collected and used to methylate the free fatty acids obtained in the previous step. The methylated product was filtered to remove any solids prior to G0 analyses. 32 Extraction and Isolation of Compounds 1, 2, and 3. The freeze-dried O. sanctum leaves and stems (440 g) were extracted sequentially with hexane, EtOAc, and MeOH. 750 mL of each solvent was used for the first 12 hours. Thereafter, a second 750 mL of fresh solvent was used to extract the plant material for an additional 12 hours. The hexane extraction afforded 13.1 g residue upon removal of solvent. A portion of this residue (10.4 g) was fractionated on a silica gel VLC (200 9) using: 4:1 pentane-EtZO as the mobile phase. A 600 mL sintered funnel was used for the VLC. Dimensions of the MP column were 250 mm x 25 mm. Fraction collection was based on the color of bands observed through the VLC sintered funnel as well as the MP column. Flow rates for MPLC experiments were approximately 1-2 mL per min. Fractions collected in the VLC experiment were 1 (375 mL), 2 (200 mL), and 3 (300 mL). Next, elution with 2:1 pentane-EtZO yielded fractions 4 (650 mL) and 5 (250 mL). This was followed by elution with 1:1 pentane-EtZO, which gave fractions 6 (250 mL), 7 (500 mL), and 8 (300 mL), and then elution with 320 which gave fractions 9 (325 mL) and 10 (200 mL). Finally, elution with CHCI3 and MeOH afforded fractions 11 (500 mL) and 12 (875 mL), respectively. All fractions were bioassayed, and only fraction 2 was found to be mosquitocidal. Fraction 2 was purified further with silica gel MPLC using hexane-MeZCO solvent systems to yield 7 fractions. The seven solvent systems used and the volume of each fraction collected were as follows: 1 (hexane; 110 ml), 2 (40:1; hexanezacetone; 90 mL), 3 (25:1; hexanezacetone; 90 mL), 4 (10:1; hexanezacetone; 90 mL), 5 (4:1; hexanezacetone; 190 mL), 6 (acetone; 95 mL), and 7 (acetone; 250 mL). 33 The seventh fraction was found to be mosquitocidal. The MeOH-soluble portion of this fraction was finally purified by repeated preparative TLC to give compounds 1 (31.9 mg), 2 (10.1 mg) and 3 (8.8 mg). Initially, compound 1 was purified using 100% EtOAc as the mobile phase. 8:1 hexane-EtOAc followed by 10:1 hexane-acetone mobile phase ultimately led to the isolation of compound 1. Like compound 1‘, compounds 2 and 3 were purified initially with 100% EtOAc as the mobile phase. Isolation of compounds 2 and 3 was accomplished using 2:1 and 1:1 EtOAc-hexane mobile phases, respectively. Compound 1, a pale brown oil: 1HNMR (CDCI3) 6: 3.30 (d, 2H, J=6.6 Hz, H-7), 3.86 (s, 3H, -OCH3), 5.04 (dd, 1H, J=9.0 Hz, J=1.5 Hz, H-9 cis), 5.05 (dd, 1H, J=18.0 Hz, J=2.4 Hz, H-9 trans), 5.47 (s, 1H, -OH), 5.93 (m, 1H, H-8), 6.67 (d, 1H, J=7.8 Hz, H-6), 6.67 (s, 1H, H-3), 6.83 (dd, 1H, J=8.7 Hz, J=1.5 Hz, H-5); 13CNMR (CDCI3) 6: 39.9 (C-7), 55.8 (-OCH3), 111.0 (C-9), 114.1 (C-8), 115.5 (C- 6), 121.1 (C-5), 131.9 (C-3), 137.8 (C-4), 143.7 (C-2), 146.3 (C-1). The spectral data of this compound were identical to an authentic sample purchased from Aldrich. OMe HO \ 1 Compound 2, a colorless oil: 1HNMR (CDCI3) 6: 1.23 (s, 6H, H-7, 9), 1.88 (s, 3H, H-8), 2.14 (s, 3H, H-1), 2.56 (s, 2H, H-5), 4.25 (s, 1H, -OH), 6.01 (s, 1H, H-3); 13CNMR (00013) a: 21.0 (08), 27.8 (01), 29.3 (07, 9), 53.8 (05), 69.8 34 (C-6). 124.5 (C-3), 157.4 (C-4), 202.3 (C-2). Therefore, it is identified as 56- Hydroxy-4, 6-dimethyl-3-heptene—2-one. 1HNMR data were found to be in agreement with previously published data (Kimura et al., 1982). O 6 / 4 123 5 OH 2 7 Compound 3, a pale yellow oil: El-MS m/z (rel. int.): 611 (M"—C16H31O]" (9), 262 [C18H290+H, 85]+ (85), 261 [C13H2901" (42), 239 (15), 230 (15), 108 (69), 95 (100), 81 (96), 53 (93); 1HNMR (CDCI3) 6: 0.86 (bt, 3H, H-16"), 0.95 (t, 6H, J=7.5 Hz, H-18'), 1.25 [m, 40H, H-(4'-7')><2 and H-(4"-15")], 1.58 [m, 6H, H-(3'XZ), 3"], 2.01 [m, 8H, H-(8', 17')XZ], 2.29 (t, 4H, J=7.5 Hz, H-2'XZ), 2.28 (t, 2H, J=7.5 Hz, H-2"), 2.76 (m, 8H, H-11', 14'XZ), 4.12 (dd, 2H, J=18.0, 6.0 Hz, H-1a, 33), 4.36 (dd, 2H, J=16.2, 4.2 Hz, H-1b, 3b), 5.24 (m, 1H, H-2), 5.34 (m, 12H, H-9', 10' 12', 13', 15', 16'XZ); 13'CNMR (CDCI3) 6: 14.1 (C-16"), 14.3 (C-18'XZ), 27.2 (C-8', 17'XZ), 22.6, 22.7, 24.8, 24.9, 29.0-29.7 (C-4'-7'xz, 4"-15"), 25.5, 25.6 (C- 11', 14'XZ), 31.5 (C-3'XZ), 31.9 (C-3"), 34.0 (C-2'x2), 34.2 (C-2"), 62.1 (C-1, 3), 68.9 (C-2), 127.1-131.9 (C-9', 10', 12', 13', 15', 16'XZ), 172.3 (C-1'XZ), 173.3 (C- 1"). The spectral data confirmed that this compound is 1, 3-dilinoleneoyl-2- palmitin. 35 0 6 —3 O O 14 O O 6 —3 3 Results and Discussion Three compounds, 1-3, were isolated from the leaf and stem hexane extract of O. sanctum by successive silica gel VLC, MPLC, and preparative TLC. The 1HNMR spectrum of compound 2 contained only five singlets. The peak furthest upfield at 6 1.23, integrated for 6 protons, indicated the presence of two deshielded magnetically equivalent methyl groups, C-7 and C-8. Deshielding of these methyl groups occurred as a result of a hydroxy group at C-6. Methyl protons at C-1 and C-9 at 6 2.14 and 1.88, respectively, were indicative of methyls attached to a carbonyl carbon and an olefinic carbon. From the 13CNMR spectrum, it was concluded that compound 2 contained an a, 8 unsaturated ketone moiety (C-3, C-4, and C-2, respectively) with an additional oxygenated carbon (C-6). This oxygenated carbon was determined to be a tertiary alcohol as indicated by the disappearance of a singlet at 62.56 following a 020 shake in the 1HNMR spectrum. The DEPT spectrum supported the 1HNMR and 13CNMR data as well as indicating the presence of three non-protonated carbons at 202.3, 157.4, and 69.8 ppm for one carbonyl, one olefinic, and one oxygenated carbon, respectively. Finally, the stereochemistry of 2 was determined to be E by 36 NOESY since no correlations were observed for the olefinic proton on C-3 and the methyl protons on C4. Therefore, compound 2, was confirmed to be E-6- hydroxy-4, 6-dimethyl-3-heptene—2-one. The 1HNMR spectrum of 2 was identical to previously published findings (Kimura et al., 1982). The structure of compound 3 was determined using 1H- and 13'CNMR, DEPT, and DQFCOSY spectral data. Also, two-dimensional HMQC proton-carbon correlations facilitated the assigning of saturated carbons in the fatty acid side chains and provided further evidence to 1D-NMR experiments. The 1HNMR signals at 6 4.12, 4.36 and 5.24 and 13CNMR signals at 6 62.09 and 68.87 along with MS fragments at m/z 611 [M*-C15H31O]+, 262 [C13H290+H]", and 261 [C13H2901” confirmed that compound 3 is a triglyceride with C16 and C18 fatty acid esters. The overlapping multiplets at 6 5.34 for twelve protons correlated with unsaturated carbons at 6 127.09 to 131.94 in the HMQC spectrum indicated the presence of six double bonds in this molecule. Support for the chemical nature of the side chains came from the GC analyses of the methyl esters of fatty acids obtained from the hydrolyzed compound 3. The GC profile confirmed the presence of the methyl esters of linolenic and palmitic acids with a ratio of 2:1, respectively. Also, retention times for both methyl esters were identical to those of authentic samples of linolenic and palmitic acid methyl esters analyzed under the same conditions. The novel triglyceride, compound 3, was not mosquitocidal. LD100 were 200 and 6.25 ug-mL'1 in 24 h for compounds 1 and 2, respectively, when tested 37 against fourth instar Aedes aegyptii larvae. There was no mortality for control larvae. In a previous report the phenylpropanoid, eugenol was found to act as an attractant to the beetle, Maladera matn'da (Ben-Yakir et al., 1995). Eugenol, in previous studies, was found to comprise 30-70% of the essential oil in O. sanctum (Asthana et al., 1984). Compound 2 originally was isolated from green and red bell peppers, Capsicum annum L. (Kimura et al., 1982) and later identified in the culture broth CHCI3 extract of Streptomyces olivaceus (Grote et al., 1990). The synthesis of 2 has been reported by Duperrier et al. (1975). To the best of our knowledge, compound 3 has not been reported previously as a natural product. The isolation and identification of mosquitocidal compounds in O. sanctum supports earlier findings that had shown insecticidal activity in crude extracts (Batta and Santhakumari, 1970; Risvi, 1981; Stein et al., 1988; Malik and Rafique, 1989; Deshmukh et al., 1982). Additional insecticidal compounds will be isolated from extractions of O. sanctum leaves and stems using more polar solvents such as acetone. Chapters 3, 4 and 5 describe other biologically active compounds in the acetone extracts of O. sanctum. 38 Chapter 3 ANTIOXIDANT COMPOUNDS FROM OCIMUM SANCTUM LINN.* Abstract —Antioxidant bioassay-directed extraction of the fresh leaves and stems of Ocimum sanctum and purification of the extract yielded compounds, cirsilineol (1), cirsimaritin (2), isothymusin (3), isothymonin (4), apigenin (5) and rosmarinic acid (6). Appreciable quantities of eugenol (70-80%) also were isolated along with these compounds. The structures of compounds 1-6 were established by spectroscopic methods. Compounds 1, 5, and 6 were isolated previously from O. sanctum, whereas compounds 2, 3, and 4 are identified for the first time in O. sanctum. Eugenol and compounds 1, 3, 4, and 6 exhibited good antioxidant activity at 10 uM concentration. * M. A. Kelm, G. M. Strasburg, and M. G. Nair Antioxidant compounds from Ocimum sanctum Linn. Phytomedicine. 39 Introduction Flavonoids represent one of the most ubiquitous classes of polyphenolic secondary compounds found in higher plants. Many common fruits, vegetables, herbs, and plant products such as wine, juices, dried fruits are rich sources of flavonoids. More importantly, many of these compounds have demonstrated rather potent anti-oxidant activity by being able to block and or scavenge free radicals (Saija et al., 1995). The formation of oxygen free radicals is a normal phenomenon carried out in aerobic cells. The interaction of these free radicals with lipids produces hydroperoxides and peroxides, which in turn may act adversely with biological systems, resulting in cancer. By way of the free radical scavenging mechanism, flavonoids effectively negate the deleterious effects of hydroperoxides and peroxides. Free radical formation also can result from the presence of transition metal ions such as Fe”, which can act as free radical initiators. Alternatively, flavonoids can prevent the formation of free radicals by chelation or complexation with the transition metal free radical initiator. Concerning structural activity relationships (SAR) of flavanoids, hydroxyl groups at the C5 and C7 and the C2 - 03 double bond were shown to be neceSsary for high inhibitory activity on xanthine oxidase (Cos et al., 1998). Xanthine oxidase forms superoxide radicals and hydrogen peroxide and is involved in the oxidation of hypoxanthine to xanthine to uric acid. Researchers also have found that a hydroxyl group at C3' and at 03 were essential for high 40 superoxide scavenging activity (Cos et al., 1998). In addition, substitution patterns on the B ring were important to antioxidant activity of flavonoids, with hydroxyl groups increasing activity by preventing lipid peroxidation (Arora et al., 1998 and Arora et al., 1997). Many of the previous studies on Ocimum sanctum Linn. Lamiaceae have focused largely on the biological activity of crude extracts. A triterpene, ursolic acid, isolated from O. sanctum was shown to be effective in protecting against lipid peroxidation (Balenehru and Nagarajan, 1991). We have reported mosquitocidal compounds eugenol and (E)-6-hydroxy-4, 6-dimethyl-3-heptene-2- one from O. sanctum (Kelm and Nair, 1998). In the present study, we report for the first time the extraction, purification, and structural identification of anti- oxidant compounds from the fresh leaves and stems of O. sanctum. Also, the importance of the A ring substitution relative to anti-oxidant/free-radical scavenging activity are discussed. Materials and Methods General Experimental. 1HNMR spectra were recorded at 300 and 500MHz. 13CNMR and DEPT spectra were recorded at 126 MHz. Chemical shifts were recorded in DMSO-d5, CD300, and CDCI3. The values are in 6 (ppm) based on residual of DMSO 2.29; DMSO-d5 39.7 and CHCI3 7.24; CDCI3 77.0. Coupling constants, J, are in Hz. EIMS were recorded at 70 eV. UV 41 experiments were carried out on a Shimadzu UV-260 spectrophotometer. Shift reagents were prepared and used according to Markham (1982). UV samples were prepared at 125-50 ppm concentrations. Particle size of silica gel used in VLC was 35-70 um. PLTC purifications were carried out on 250, 500, 1000, and 2000 um silica gel plates and 200 pm KC18 silica gel plates (60 A). Spots and bands were visualized under UV light (366 and 254 nm). Anti-oxidant assays. Anti-oxidant assays were conducted on crude extracts and pure compounds by analysis of model liposome oxidation by fluorescence spectroscopy peroxidation (Arora et al., 1998 and Arora et al., 1997). The procedure for the anti-oxidant assay is as follows. A mixture containing 5 pmol of 1-steroyl-2-Iinoleneoyl-sn-glycerol-3-phosphocholine and 5 umol of fluorescence probe 3-(p-6-phenyl)-1, 3, 5-hexatrienyl) phenyl propionic acid was dried in a foil-covered round-bottom flask (to prevent degradation of probe) on a rotary evaporator. The resulting lipid film was suspended in 500 uL of MBSE buffer. The MBSE buffer contained 0.15 M NaCl, 0.1 mM EDTA (ethylenediamine tetra acetic acid di-sodium salt), 10 mM MOPS [3-(N- morpholine propane sulfonic acid), adjusted to pH 7.0] buffer and then treated with Chelex 100 chelating resin to remove trace-metal ions. The lipid-buffer mixture was subjected to ten-freeze thaw cycles using a dry ice/ethanol bath. Following the last thaw, the lipid-buffer suspension was extruded 29 times through a Liposofast extruder containing a polycarbonate membrane with a 100 nm pore size to produce unilamellar liposome. A 20 uL aliquot of this liposome 42 suspension was diluted to 2 mL with 200 uL of HEPES buffer (adjusted to pH 7.0), 100 uL of 1M NaCl (treated with Chelex 100), and 1.68 mL nanopure H20 then incubated for 5 min at room temperature, followed by incubation at 23°C in a spectrophotometric cuvette. Peroxidation then was initiated by the addition of 20 uL of 0.5 mM FeClz stock solution to achieve a final concentration of 5 um of Fe+2 in the presence of test compounds or crude extracts (dissolved in DMSO). Two controls were used in which one contained test solvent (DMSO), liposome, and buffer, whereas the second one contained Fe‘z. Fluorescence intensity of these liposome solutions were measured at an excitation wavelength of 384 nm for every 3 min over a period of 21 min. The decrease in relative fluorescence intensity with time indicated the rate of peroxidation. TBHQ, BHT, and Vitamin E were used as positive controls for this study. Plant Material. A voucher plant specimen (MSC 360851) of O. sanctum was filed with the Beal-Darlington herbarium, Department of Botany and Plant Pathology, Michigan State University. Leaves and stems of O. sanctum were harvested from plants maintained in the Center for Integrated Plant Systems greenhouses at Michigan State University. 0. sanctum plants were grown in 11- inch plastic pots, using a mixture of 50% loam and 50% Bacto mix. Plants were watered on a daily basis and fertilized once a week with 20-20-20 Peters brand fertilizer. Leaves and stems were harvested periodically from 4 to 6 month-old plants. 43 Generalized Extraction of Plant Material. Three separate extractions (l-lll) were carried out for the isolation of eugenol and compounds 1-6. The following is a representative protocol used for the extraction of the plant material. 2.58 kg of fresh 0. sanctum leaves and stems were chopped, blended with acetone (6.5 L), and allowed to set for 24 h. The acetone was removed in vacuo to yield an aqueous green suspension. Solids were collected in a Bfichner funnel lined with a No. 4 Whatman filter paper to give extract A (18.33 g). The aqueous filtrate was sequentially extracted with CHCI3 (5 x 250 mL) followed by EtOAc (5 x 250 mL) to provide extracts B (4.58 g) and C (12.12 g), respectively. The remaining aqueous phase was dried in vacuo to yield extract D (56.40 g). Extraction l. 763.9 g of fresh leaves and stems were chopped into pieces approximately 10-15 mm long. These pieces were blended with acetone (2 L) the allowed to soak for 24 h. Thereafter, the mixture was filtered through a Bfichner funnel lined with a No. 4 Whatman filter paper. The plant material was washed with several aliquots of acetone (3x50 mL). The filtrate was distilled to remove acetone, resulting in the production of a green precipitate suspended in an aqueous solution. The solids (Extract A; 10.5 g) were recovered by centrifuging at 10,000 rpm for 10 min at 4°C. The remaining water was extracted with CHCI3 (3 x 50 mL) to give upon drying in vacuo, Extract B (0.786 9). Extract C (17.9 g) was obtained simply by drying the water-soluble phase in vacuo. 44 Extraction II. The second extraction carried out was essentially the same as the previous with the exception of several modifications as described below. 450 g of fresh 0. sanctum was chopped, blended with acetone (1.2 L), and allowed to set for 24 h. The acetone was removed in vacuo to yield a green precipitate suspended in an aqueous solution. Solids were collected in a Biichner funnel lined with a No. 4 Whatman filter paper to give Extract A (4.53 g). The aqueous filtrate was sequentially extracted with CHCI3 (3 x 50 mL) followed by EtOAc (3 x 50 mL) to provide Extract B (0.49 g) and Extract C (1.16 g), respectively. The remaining aqueous phase was dried in vacuo to yield Extract D (7.1 g). Extraction III. The third extraction utilized 1.37 kg of plant material. The same procedure was followed as for the second extraction; however, larger volumes of solvents were used. Acetone (3.3 L), CHCI3 (5 x 200 mL), and EtOAc (5 x 200 mL) were used to obtain Extracts A, B, and C, respectively. Weights of Extracts A, B, C, and D were 3.3, 3.3, 9.1 , and 33.3 g, respectively. Isolation of eugenol and compound 1. Preparative TLC (silica gel; 3 x 2000 um) of Extract B (477.3 mg) using CHClgzMeOH (15:1) as the mobile phase, afforded fractions, i-v. Fraction i (342 mg) and ii (14.4 mg) were determined to be pure by TLC. Fraction iii was purified further (silica gel; 500 pm) using CHClgzMeOH (25:1) which gave fractions 1-4. Here, fraction 3 (9.7 mg) was determined by TLC to be identical to fraction ii. A second preparative 45 TLC (silica gel; 4 x 2000 pm) of Extract B (290 mg) using CHCI3zMeOH (15:1) resulted in fractions i-v. In this purification fractions i (195 mg) and ii (6.6 mg) determined to be pure, by TLC, were identical to fractions obtained from the first purification of Extract B. Fractions i from the first and final purification were combined and subsequently determined by NMR to be eugenol. Eugenol, a pale brown oil: 1HNMR (C003) 6: 3.30 (d, 2H, J=6.6 Hz, H-7), 3.86 (s, 3H, -OCH3), 5.04 (dd, 1H, J=9.0 Hz, J=1.5 Hz, H-9 cis), 5.05 (dd, 1H, J=18.0 Hz, J=2.4 Hz, H-9 trans), 5.47 (s, 1H, -OH), 5.93 (m, 1H, H-8), 6.67 (d, 1H, J=7.8 Hz, H-6), 6.67 (s, 1H, H-3), 6.83 (dd, 1H, J=8.7 Hz, J=1.5 Hz, H-5); 13CNMR (CDCI3) 6: 39.9 (C-7), 55.8 (-OCH3), 111.0 (C-9), 114.1 (C-8), 115.5 (C-6), 121.1 (C-5), 131.9 (C-3), 137.8 (C-4), 143.7 (C-2), 146.3 (C-1). The spectral data of this compound was identical to an authentic sample purchased from Aldrich Chemical Company, Inc. Milwaukee, WI 53233. OMe HO \ eugenol Fractions ii, iii, and ii from the first, second, and third purifications, respectively were combined and subsequently determined to be compound 1 by NMR. Cirsilineol; 5, 4'-dihydroxy-6, 7, 3’-trimethoxyflavone (1). UV kmax (MeOH) 341 nm, 275 nm; (5% AlCl3/MeOH) 378 nm, 284 nm, 262 nm; (AICl3/HCI) 366 nm, 287 nm, 260 nm. 1HNMR (DMSO-d5) 6: 12.98 (bs, 1H, 5- OH), 7.67 (d, 1H, J=9 Hz, H-6’), 7.54 (s, 1H, H-2’), 6.92 (s, 1H, H-3), 6.90 (d, 1H, 46 J=8.0 Hz, H-5'), 6.89 (s, 1H, H-8), 3.91 (s, 3H, -OCH3), 3.87 (s, 3H, -OCH3), 3.72 (s, 3H, -OCH3). 13CNMR (DMSO—d5) 6: 56.56 (-OCH3), 57.09 (-OCH3), 60.70 (—OCH3), 92.28 (C-8), 103.25 (03), 105.65 (010), 115.57 (05'), 120.95 (C-6'), 121.35 (01'), 132.44 (C-6), 148.92 (0.5), 152.45 (09), 152.82 (0.3), 153.31 (cw), 159.13 (07'), 184.78 (0.2), 182.68 (04). Isolation of compound 2. Extract B from Extraction I was fractionated twice as previously mentioned. The third fraction (iii) from each of the purifications, 4.2 and 22.5 mg, respectively were found to be identical by TLC. Extract B (490.4 mg) from Extraction II was loaded onto preparative silica gel TLC plates (3x2000 um) and developed with CHCI3: MeOH (15:1) as the mobile phase. Bands i-v were isolated. Band i (318 mg) was determined to be compound 1 by comparing its TLC with a previously identified sample obtained. Band ii (10 mg) was found to be identical to both fractions iii above and these subsequently were combined. The combined fractions were subjected to further purification by preparative TLC (2x250 pm) with CHClgzMeOH (20:1) as the mobile phase. Bands i-iv were obtained. Fraction iii (3.5 mg) was found to be suitable for NMR and aftenivards was determined to be compound 2. Cirsimaritin; 5, 4’-dihydroxy-6, 7-dimethoxyflavone (2). UV kmax (MeOH) 329 47 nm, 278 nm; (5% AlClg/MeOH) 382 nm, 284 nm; (AlClg/HCI) 358 nm, 288 nm. ‘HNMR (DMSO-d5) 6: 7.94 (d, 2H, J=9.0 Hz, H-2’, 6'), 8.92 (s, 1H, 1+3), 8.90 (d, 2H, J= 9Hz, H-3’, 5'), 8.82 (s, 1H, H-8), 3.91 (s, 3H, -OCH3-6), 3.72 (s, 3H, - OCH3-7). ”CNMR (DMSO-d5) 6: 56.51 (-oc113-7), 80.10 (-OCH3-6), 91.55 (C-8), 102.30 (03), 105.04 (010), 118.20 (03', 5'), 120.30 (or), 128.48 (02', 6’), 131.83 (C-6), 152.09 (0.5), 152.81 (0.9), 158.55 (07), 182.35 (C-4'), 184.23 (c- 2), 182.18 (C-4). Isolation of compounds 3 and 4. Fraction iii from Extract B of Extraction l and fraction iv from Extract B of Extraction II were combined (17.6 mg). Preparative TLC silica gel (2x250 um) using CHClgzMeOH (10:1) as the mobile phase was carried out on the mixture. Bands 1-4 were obtained, and fraction 4 was determined to be pure by TLC and subsequently was determined to be compound 3 (6.7 mg) by NMR studies. lsothymusin; 5, 8, 4’-trihydroxy-6, 7- dimethoxyflavone (3). UV kmax (MeOH) 328 nm, 306 nm; (5% AlClg/MeOH) 360 nm, 322 nm, 289 nm, 234 nm; (AICI3/HCI) 352 nm, 322 nm, 288 nm. 1HNMR (00300) 6: 7.93 (d, 2H, J= 9 Hz, H-2', 6'), 6.92 (d, 2H, J=9.3 Hz, H-3’, 5’), 6.63 (s, 1H, H-3), 4.02 (s, 3H, -oc113), 3.91 (s, 3H, -oc1-13). 13CNMR (coaoo) 48 6: 61.35 (—OCH3), 61.99 (—OCH3), 104.46 (C-3), 107.94 (C-10), 116.99 (C-3’, 5’), 123.30 (C-2’, 6’), 132.25 (C-8), 137.81 (C-6), 142.86 (C-7), 146.42 (C-5), 149.20 (C-9), 162.92 (C-4’), 166.74 (C-2), 184.84 (C-4). Band 4 from both purifications of Extract B, Extraction l were combined (5.2 mg) and purified using silica gel preparative TLC (1x250 um) and CHClazMeOH (15:1) as the mobile phase. Bands 1-3 were isolated. The NMR analysis of fraction l resulted in the identification of compound 4 (1 mg). Isothymonin; 5, 8, 4’-trihydroxy-6, 7, 3’-trimethoxyflavone (4). UV kmax (MeOH) 340 nm, 288 nm; (5% AlClg/MeOH) 450 nm, 371 nm, 327 nm, 292 nm, 263 nm; (AICI3/HCI) 450 nm, 364 nm, 331 nm, 293 nm, 259 nm. 1HNMR I (CD3OD) 6: 7.61 (dd, J= 2.1, 9.0 Hz, 1H, H-6’), 7.60 (d, J=1.8 Hz, 1H, H-2’), 6.93 (d, J=8.7 Hz, 1H, H-5'), 4.03 (s, 3H, -OCH3-6), 3.96 (s, 3H, -OCH3-7), 3.91 (s, 3H, -OCH3-3’). 49 Isolation of compound 5. Fractionation of Extract B from Extraction III was carried out using VLC. Extract B (3 g) was loaded onto a bed of silica gel (75 g) preconditioned in hexane. Fractions were collected in aliquots of 125 (1), 150 (2), 100(3), 120(4), 160(5), 160(6), 150 (7), 210 (8), and 1,200 (9) mL using hexane, hexanezacetone (1 :1), hexanezacetone (1 :1), hexanezacetone (1 :1), acetone, CHCI3, CHClgzMeOH (1:1), CHClazMeOH (1:1), and MeOH, respectively. The acetone-soluble portion of fraction 3 (190 mg) was dissolved in MeOH and concentrated. This was allowed to stand overnight at room temperature, resulting in the precipitation of solids. The mother liquor was recovered and dried in vacuo to yield a brown gum (160 mg). A portion of this was subjected to repeated silica gel PTLCs using CHClgzMeOH (30:1, 10:1), to afford compound 5, apigenin (1.6 mg). Apigenin; 5, 7, 4’-trihydroxyflavone (5). UV kmax (MeOH); (5% AlCl3/MeOH); (AICl3/HCI). 1HNMR (DMSO-d5) 6: 12.86 (bs, 1H, 5-OH), 7.73 (d, 1H, J=8.7 Hz, H-6’), 7.70 (d, 1H, J=8.7 Hz, H-2’), 6.79 (cl, 1H, J=8.7 Hz, H-5’), 6.76 (d, 1H, J=8.7 Hz, H-3’), 6.34 (s, 1H, H-3), 5.80 (s, 1H, H-6), 5.57 (s, 1H, H-8), 3.44 (bm, 2H, 7, 4’-OH). 50 Isolation of Compound 6. Prior to fractionation, the EtOAc extract C (1.16 g) was first dissolved in MeOH (1 mL), stirred with CHCI3 (16 mL) and refrigerated overnight. The precipitate (630 mg) was recovered and dried in vacuo. This precipitate (630 mg) was purified by medium pressure silica gel column chromatography. Fractions were collected in aliquots of 750 (1), 1000 (2), 1000(3), 1000(4), 1500 (5), 200(6), 900 (7), and 850 (8) mL using CHCI3zMeOH (3:1), MeOHzCHCla (2:1, 4:1, 4:1), 100% MeOH, 2 x 1% HCOOH (in MeOH), 100% MeOH, respectively. The active fraction 2 (233 mg) was dissolved in MeOH (5 mL) and precipitated with CHCI3. The pale yellow solids were recovered and dried in vacuo to yield compound 6 (62.4 mg). Rosmarinic acid (6). UV Amax (MeOH) 349 nm, 338 nm, 223nm. IR vmax cm'1 3350, 3250,1689, 1600, 1524. 1H-NMR (DMSO-d6) 6: 7.37 (d, 1H, J=15.9 Hz, H-7), 7.04 (d, 1H, J=1.8 Hz, H-2), 6.92 (dd, 1H, J=9.0, 2.1, 2.1 Hz, H-6), 6.75 (d, 1H, J=2.1 Hz, H-5), 6.67 (d, 1H, J=2.1 Hz, H-2'), 6.60 (d, 1H, J=7.8 Hz, H-5'), 6.49 (dd, 1H, J=9.0, 2.1, 2.1 Hz, H-6'), 6.18 (d, 1H, J=16.2 Hz, H-8), 4.89 (1H, m H- 10), 3.04 (1H,, m, H-11), 2.77 (1H,, m, H-11). 13c-NMR (DMSO-d5) 8: 173.23 (C-12), 166.46 (C-9), 148.38 (C-4), 145.73 (C-3), 144.82 (C-1), 144.50 (C-4'), 143.49 (C-3'), 130.01 (C-1'), 125.76 (C-8), 121.24 (C-7), 119.93 (C-2'), 116.70 (C-2), 116.03 (C-5'), 115.47 (C-5), 114.99 (C-6'), 114.87 (C-6), 75.98 (C-10), 30.76 (C-11). 51 Results and Discussion. Seven compounds, eugenol and 1-6, were isolated from the fresh leaf and stem extracts of O. sanctum. Structure determination was facilitated by 1H and 13CNMR experiments. Final position assignments of flavone hydroxyl groups were achieved by the use of different shift reagents in UV fluorescence experiments. (Markham, 1982). According to Markham’s Interpretation of UV spectra, compound 1 in MeOH indicated the presence of a flavone with strong absorption at 341 and 275 nm. In fact, the UV spectra of compounds 2-5 indicated the presence of flavone. Following the addition of AICI3 stock solution, a +37 nm shift in Band I in the spectrum of compound 1 was observed. This shift was indicative of a hydroxyl substitution at the C-5 position. The presence of the 5-OH hydrogen bonded to the carbonyl oxygen at C-4 was confirmed by the chemical shift of the —OH signal at 6 12.98. As deduced from the AlCl3/HCI spectrum, the presence of A or B ring o—diOH substitutions was not observed. Two doublets at 6 7.67 (J = 9.0 Hz) and at 6.90 (J = 8.0 Hz) corresponded to protons H-6’ and H-5’, respectively. Singlets at 6 6.92 and 6 6.89 represented 52 protons at H-3 and H-8, respectively. Intense, strong three proton singlets at 6 3.91, 3.87, and 3.72 were assigned to methoxy protons. These proton data were found to be in agreement with those previously published (Martinez, et al., 1987). Compound 2, cirsimaritin was isolated previously from the methanolic extract of aerial parts of Baccharis trimera (Asteraceae) Less. (Grayer and Veitch, 1998), the diethyl ether extract of freeze-dried leaves of Becium grandiflorum (Lamiaceae) Pic. Serrn. (Yousseff and Frahm, 1995), the ethanolic extract of the aerial parts of Centaurea scoparia (Asteraceae) Sieb. (Zhu et al., 1996), the ethanolic (70%) extract of the air-dried root bark of Clerodendrum mandarinomm (Verbenaceae) Diels. (Cuvelier et al., 1996), and was identified by HPLC in two genera of the Lamiaceae family, Salvia officinalis L. and Rosmarinus officinalis L. (Barberan et al., 1996). The 5-OH and the 4’-OH for compound 2 was observed at 6 12.93 and 10.34, respectively. Two three-proton singlets at 6 3.91 and 3.72 were assigned to methoxy groups at C-7 and C-6, respectively. Two proton doublets 6 7.94 (J=9.0 Hz) and 6.90 (J=9.0 Hz) indicated the presence of a para substituted aromatic ring. These data were found to be in agreement with previously published data (Zhu et al., 1996). A +25 nrn shift in the UV spectrum following the addition of AlCl3/HCI further indicated the presence of the 5-OH substitution. With AICI3 alone, shifts in band I were not large enough to indicate the presence of any o-diOH substitution, in particular the A ring of the molecule. 53 lsothymusin, compound 3, was isolated from a natural source, B. grandiflorum for the first time (Yousseff and Frahm, 1995). lsothymusin (6, 7- dimethoxy-5, 8, 4’-trihydroxyflavone) was previously known as a conversion product via a Wessely-Moser rearrangement of the natural product, isothymusin (7, 8-dimethoxy-5, 6, 4’-trihydroxyflavone). A protocol for this conversion was reported (Horie et al., 1995). In the present study, the identification of compound 3 relied largely on UV experiments, since 1HNMR data for thymusin and isothymusin are interchangeable. A comparison of AICI3 and AlCl3/HCI spectra of compound 3 revealed only a small change, 8 nm in Band I, which indicated the lack of o—diOH substitution pattern on the A ring. Our results are in agreement with those previously published by Barberan et al. (1985). Two 2H proton doublets at 6 7.93 (J = 9.0 Hz) and 6.92 (J = 9.3 Hz) were indicative of a para disubstituted aromatic ring. The above chemical shifts were assigned to H-2', 6’ and H-3’, 5', respectively. Two intense three-proton singlets at 6 4.02 and 3.91 were assigned to methoxy groups on the B ring at C-6 and C-7, respectively. 1HNMR data is consistent with previously published work (Horie et al., 1995). lsothymusin can be formed from its isomer by a Wessely-Moser rearrangement (Ferreres et al., 1985). Compound 4, isothymonin, is reported herein for the first time as a natural product. Isothymonin, like isothymusin is obtained by means of acidic treatment 54 and Wessely-Moser rearrangement. The UV spectral interpretation for compound 4 was much like that carried out for compound 3. 1HNMR spectral data were very similar to data obtained for isothymusin with the addition of a three-proton singlet at 6 3.91 which was assigned to a methoxy group. The UV and 1HNMR data for compound 4 were found to match closely previously published data (Barberan et al., 1985 and Horie et al., 1995). Apigenin is probably one of the most common of the flavones. Apigenin, compound 5, was previously identified in O. sanctum leaves (N6rr and Wagner, 1992), O. basilicum (Grayer et al., 1996), B. trimera (Youseff and Frahm, 1995), and in the extracts of S. offlcinalis and R. offincinalis (Cuvelier et al., 1996). The antioxidant activity of apigenin was reported previously (Cholbi et al., 1991). However, in another study (Chen et al., 1996), researchers found that apigenin was unable to prevent the oxidation of lipids in canola oil. The isolation and identification of compound 6, rosmarinic acid was reported from O. sanctum (Nbrr and Wagner, 1992) and S. officinalis (Wang et al., 1998). Portions of the 1HNMR spectrum, namely chemical shifts at 6 7.37 (J=15.9 Hz), 7.04 (J=1.8 Hz), 6.92 (J = 9.0, 2.1, 1.8 Hz), 6.75 (J = 8.4 Hz), and 6.18 (J=16.2 Hz) matched closely with those of caffeic acid. Furthermore, the acidic hydrolysis of 6 yielded caffeic acid, as indicated by comparative TLC. The chemical shifts at 6 7.37 (J = 15.9 Hz) and 6.18 (J = 16.2 Hz) were for doublets that integrated for one proton each. Coupling constants for both of these olefinic 55 protons (H-7, 8) indicated that they existed in a trans geometry. Protons at positions 2, 5, and 6 gave identical splitting patterns with 2’, 5', and 6’, respectively. Chemical shifts for H-2 and 2' were 6 7.04 (J = 1.8 Hz) and 6.67 (J = 2.1 Hz), respectively. Their J values were indicative of meta coupling protons. Ortho coupling was observed for H-5 and 5’ doublets. A 1H doublet of doublet was assigned to H-6 and 6’ at 6 6.92 and 6.49, respectively. Both ortho and meta coupling was observed for H-6 and 6’ where J values were 9.0, 2.1, 1.8 Hz and 9.0, 2.1, 2.1 Hz, respectively. 13CNMR experiments indicated the presence of two carbons at 6 173.23 and 166.46 which corresponded to the carboxylic acid and ester functional groups, respectively. Both 1HNMR and 13CNMR data agreed with published data (Wang et al., 1998). Anti-oxidant assays were conducted according to previously published procedures (Arora et al, 1998 and Arora et al., 1997). Results for anti-oxidant bioassay are presented in Figure 1 and 2. All flavones with the exception of compound 2 demonstrated excellent anti-oxidant activity. Eugenol and rosmarinic acid (6) also displayed good anti-oxidant activity. The most notable compound in regards to anti-oxidant activity was compound 3, isothymusin. These results suggest that hydroxy substitution on the A ring at C-8 enhances anti-oxidant activity, whereas the addition of a methoxy group at C-3’ position inhibits the activity, as in the case of compounds 3 and 4. However, in the case of compounds 1 and 2, when the hydroxy at C-8 is lacking, the more active compound contains a methoxy at the C-3' position. lsothymusin, compound 3, 56 was the best performer in regards to its anti-oxidant activity. Compound 3 was about 50% more active than the synthetic antioxidants TBHQ and BHT at the same 10 0M concentration. Compound 4 performed as well as compound 1 and better than BHT and TBHQ at 10 uM concentration. Compound 1 performed equally well if not better than TBHQ and BHT in our anti-oxidant assays (Figure 1, 2). Surprisingly, due to structural similarities, compound 2 displayed poor anti- oxidant activity in our anti-oxidant assays (Figure 1, 2). The presence of eugenol in O. sanctum is well known. In fact, eugenol can comprise 38% of the water distilled essential oil of O. sanctum (Laakso et al., 1990) and up to 86% in the steam distillate (Sukari et al., 1988). In the same study, the essential oil of O. sanctum showed some inhibitory activity on Tn'chophyton mertagrophytes (Robin) Blanchard and Bacillus subtilis (Ehrenberg) Cohn at 10‘ ppm and 100% mortality on BIatteI/a gennanica L. and Callosobrunchus chinensis L. in the presence of 10 mg of the neat oil. In a later study, it was found that eugenol could demonstrate anti-stress activity by effecting stress-induced changes brought about by various biochemical parameters and cell membrane dynamics utilizing red blood cells (Sen et al., 1992). Eugenol’s anti-oxidant activity prior to this paper was reported by Priyadarsini et al., 1998. In their study, inhibition of lipid peroxidation of rat brain homogenates induced by ferric ion, ferrous ion, and cumene hydroperoxide was demonstrated by eugenol at leo 74.2, 11.3, 136.8 umol-dm", respectively. Eugenol’s anti-oxidant activity was comparable to compound 6 (Figure 1, 2). The 57 presence of eugenol’s 4-hydroxy group could be involved in the observed activity by formation of a less reactive phenoxyl radical. Compound 1, cirsilineol was previously identified in O. sanctum (Nbrr and Wagner, 1992), O. basilicum (Grayer et al., 1996), and Artemisia assoana VVIIIK. (Martinez et al., 1987). Cirsilineol was found to have potent 3’, 5’-cyclic adenosine monophosphate phosphodiesterase (cAMP-PDE) inhibitory activity (Nagasugi et al., 1998). In this study the authors were able to correlate high cAMP-PDE-inhibitory activity with averaged 13CNMR chemical shifts. Cirsimaritin showed a 77% antimutagenic activity at 25 0M per plate in a modified Ames test. Apigenin was 85.2% effective at the same concentration in the same study (Grayer and Veitch, 1998). S. officinalis and R. officinalis extracts that contained cirsimaritin and apigenin demonstrated anti-oxidant activity as measured by accelerated auto-oxidation of methyl linoleneate (Barberén et al., 1996). Compound 6 compared to other compounds in the study demonstrated high anti-oxidant activity. Compound 6 certainly was more active than Vitamin E, and compounds 2 and 5 (Figure 1, 2). The anti-oxidant activity of rosmarinic acid was reported previously from sage, Salvia officinalis where it was found to scavenge DPPH (2, 2-diphenyl picryhydrazyl) free radical-induced oxidation (Wang et al., 1998). 58 Relative Fluorescence Relative Fluorescence Figure 1. Anti-oxidant activity of synthetic and isolated oorrpounds assayed at 10uM. Figure 2. Anti-oxidant activity at 21 minutes for synthetic and isolated compounds at 10 0M. 59 Chapter 4 ANTI-INFLAMMATORY COMPOUNDS FROM OCIMUM SANCTUM LINN. Abstract — Eugenol and compounds 1-6 described in the previous chapter were subjected to anti-inflammatory assays. Eugenol demonstrated 97% COX-1 inhibitory activity when assayed at 1000 uM concentrations. Compounds 1, 2, and 4-6 displayed 37, 50, 37, 65, and 58 % COX-1 inhibitory activity, respectively, when assayed at 1000 uM. Eugenol and compounds 1, 2, 5, and 6 demonstrated COX-2 inhibitory activity at slightly higher levels when assayed at 1000 uM. The activities of compounds 1-6 were comparable to ibuprofen, naproxen, and aspirin tested at 10, 10, and 100 uM, respectively. 60 Introduction The formation of prostaglandins from arachidonic acid by prostaglandin synthase is a well studied process. The formation and subsequent effect of prostaglandins can result in the stimulation of inflammation and associated pain (Stryer, 1988). Prostaglandin synthase contains both cyclo-oxygenase and hydroperoxidase components; however, it is well understood that two distinct isoforms of cyclo-oxygenase (COX) exist, namely COX-1 and COX-2 which are both involved in the conversion of arachidonic acid to prostaglandins (Lipsky et al., 1998). COX-1 is found throughout the body, particularly in the gastrointestinal tract, kidneys and platelets, whereas COX-2 is found predominantly in inflamed tissues. Both COX-1 and COX-2 are inhibited by traditional non-steroidal anti-inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, naproxen, sulindac, diclofenac, etc. Many of these drugs are taken to releive inflammation pain such as musculoskeletal pain including arthritis and tendonitis, as well as other general aches and pains. Since traditional NSAIDS are not specific in the inhibition of both COX forms, the inhibition of COX-1 seems to be associated with gastrointestal damage, renal dysfunction, and platelet abnormalities (Simon et al., 1998). Therefore, much of the research in the 1990s has been directed toward the discovery of NSAIDS with specific inhibition towards the enzyme COX-2. As stated in the previous chapter, flavonoids are widespread components in the plant portion of the human diet. And, many of these have low toxicity in 61 mammals. Their action as anti-inflammatory agents and their low toxicity make them prime candidates in plant drug research. The anti-inflammatory activity of flavonoids is believed to be controlled, at least in part, by the addition of cyclo- oxygenase (Kim et al., 1998). Flavonoids such as 3-hydroxyflavone, galangin, quercetin, and kaempferol demonstrated good cyclo-oxygenase inhibitory activity in assays utilizing rat mixed peritoneal leukocytes (Hoult et al., 1994). Other researchers have reported similar findings (Kim et al., 1998, Tordera et al., 1994, Middleton and Kandaswani, 1992). In a study looking at structural activity relationships of flavonoids, it was determined that flavonoids lacking 3’, 4’- dihydroxy substitution as well as fewer overall hydroxy groups had greater cyclo- oxygenase inhibitory activity (Moroney et al., 1988). More specifically, it was demonstrated that B-ring hydroxyl substitution decreased a flavonoid’s ability to inhibit cyclo-oxygenase. Baumann et al., (1979) indicated that ortho dihydroxy- substituted phenols can act as initiators of cyclo-oxgenase by acting as cofactors in prostaglandin generation, specifically in cell-free assays. The compounds isolated from O. sanctum are described in Chapter 3 were eugenol, cirsilineol (1), cirsimaritin (2), isothymusin (3), isothymonin (4), apigenin (5) and rosmarinic acid (6). These compounds were subjected to anti- inflammatory assays in order to determine COX-1 and COX—2 inhibitory activities. Of the compounds tested, eugenol (Saeed et al., 1995), apigenin (5) (Todera et al., 1994 and Kim et al., 1998), and rosmarinic acid (6) have demonstrated anti-inflammatory activity. The COX-1 inhibitory activity of flavones, 1, 2, and 4 are described here for the first time. Eugenol, compounds 62 1, 2, 5, and 6 demonstrated better COX-2 inhibitory activity compared to COX-1 inhibitory activity. This would indicate specific enzyme inhibition towards COX-2. Materials and Methods Refer to Chapter 3, Materials and Methods section for General Experimental, Plant Materials, Extraction, and Fractionation procedures. Anti-inflammatory Assay. Human prostaglandin H synthase isozymes (hPGHS-1) were expressed in cos-1 cells as described previously (Laneuville et al., 1994; Meade et al., 1993). Cyclo-oxygenase activity (COX) was measured by utilizing microsomal membranes (ca. 5 mg protein/ml in 0.1 M TrisHCL, pH 7.4) from sham-transfected cos-1 cells or from cos-1 cells transfected with the plasmid pOSML - PGHS - 1. Cycle-oxygenase assays were performed at 37 °C by monitoring the initial rate of 02 uptake using an 02 electrode (Instech Laboratories, Inc., 5209 Militia Hill Road, Plymouth Meeting PA 19462-1216). Each assay mixture contained 3 mL of 0.1 M TrisHCl, pH 8.0, 1 mmol phenol, 85 pg hemoglobin and 100 umol arachidonic acid. Reactions were initiated by adding 5 to 25 pg of microsomal protein in a volume of 10-20 uL. Instantaneous inhibition was determined by measuring the cyclooxygenase activity initiated by adding aliquots of microsomal suspensions of hPGHS—1 (10 pmol Ozlmin cyclooxygenase activity/aliquot) to assay mixtures containing 10 umol arachidonate and 1000 (N concentrations of the test 63 hPGHS-1 (10 pmol Ozlmin cyclooxygenase activity/aliquot) to assay mixtures containing 10 umol arachidonate and 1000 (N concentrations of the test compounds (Figure 3, 4). Ibuprofen and naproxen were assayed at 10 pM and aspirin at 1000 (M. Results and Discussion The in vitro anti-inflammatory activity of eugenol, compounds 1, 2 and 4—6 all demonstrated inhibitory activity against COX-1 as expressed in Figure 3. Figure 3. In vitro anti-inflammatory assay of eugenol, compounds 1-6 and synthetics. Eugenol, compounds 1-6, and aspirin tested at 1000 HM and ibuprofen and naproxen tested at 10 (M. 120 % COX-1 inhibitory activity 64 rosmarinic acid demonstrated 58% COX-1 inhibitory activity in comparison to the other compounds. The activities of eugenol, apigenin (5), and rosmarinic acid (6) were reported by Saeed et al., 1995, Todera et al., 1994, and Kim et al., 1998. The COX-1 inhibitory activities of compounds 1, 2, and 4 are reported here for the first time. At 1000 uM, compounds 1-6 demonstrated COX-1 inhibitory activity comparable to ibuprofen, naproxen, and aspirin at 10, 10, and 100 uM concentrations, respectively. Ibuprofen, naproxen, and aspirin demonstrated 33, 58, and 46% COX-1 inhibitory activity, respectively. COX-2 inhibitory activities were demonstrated by eugenol, compounds 1, 2, 5, and 6 when assayed at 1000 (M (Figure 4). Compound 4 was unavailable for testing COX-2 inhibitory activity. Compound 3 did not inhibit COX-1 or COX-2 activity. This present study supports earlier findings and suggests possible phytochemicals relative to research on the anti-inflammatory activity of O. sanctum: Early studies had shown that methanolic and aqueous suspensions of O. sanctum showed anti- inflammatory activity in rats (Godwani, 1987); however, anti-inflammatory activity was found to be less active than aspirin. Singh et al., 1996, found that the volatile oil of O. sanctum could inhibit arachidonic and leukotriene-induced inflammation. They concluded that inhibition of cyclo-oxygenase and lipo- 65 Figure 4. In vitro anti-inflammatory activity of eugenol, compounds 1, 2, 5, 6, and aspirin tested at 1000 M and ibuprofen tested at 10 M. .4? 4?; 120 O “l 100 - _ , . E 80 a- 73:17: _ ___ g 60 g, 7 1.3155; __ ‘ .E 40 . . N 1‘ . 5 20 o 0 — * 8\° f oxygenase pathways in arachidonic acid metabolism might be occurring. The present findings tend to support these earlier findings; however, inhibition of enzymatic pathways might be only part of the overall activity. Tulsi has been shown to decrease levels of uric acid in rabbits (Sarkar et al., 1990). Elevated levels of uric acid are associated with gouty arthritis and other joint inflammation. Structural activity relationships (SAR) of flavonoids 1-5, relative to COX-1 inhibitory activity seemed dependent on number and position of hydroxy and methoxy groups on their A and B rings. Similar conclusions regarding SAR can be said of compounds 1, 2, 5, and 6 regarding COX-2 inhibitory activity. Due to the small number of compounds being compared, unequivocal deductions 66 regarding SAR for these compounds cannot be made. Regardless, we can discuss to some extent, the results obtained. The 5, 7, 4’-trihydroxyflavone, apigenin (6) had the greatest COX-1 inhibitory relative to the other flavones. Methoxy groups at 6, 7, and 3’ positions resulted in a decrease in activity. Compound 2, slightly more active that 1, had an additional methoxy group at the 3' position. One notable exception to this is the activity of compounds 3 and 4. Compound 4 showed modest COX-1 enzyme-inhibitory activity, whereas compound 3 totally lacked COX-1 enzyme inhibitory activity. The only difference between these two compounds was a methoxy group at the 3’ position. These findings present some compounds that lend support to earlier observed anti-inflammatory activities of crude extracts from O. sanctum. Furthermore, these results support the traditional use of Tulsi as a remedy for inflammation and pain. 67 Chapter 5 A PORPHYRIN COMPOUND FROM OCIMUM SANCTUM LINN. WITH CORN EARWORM ANTI-FEEDANT/TOXICITY ACTIVITY Abstract- A porphyrin compound characterized as phylloerythrin like compound was isolated from the crude acetone extract of O. sanctum by bioassay-guided fractionation. This compound was anti-feedant to corn eanivorm, Helicoverpa zea at 100 ppm. 68 Introduction Insect toxicity (Narasimhan and Marriappan; 1988, Stein et al., 1988; Sukari and Takahashi, 1988), anti-feedant, repellant (Mallick and Banerji, 1989, Arrekul, et al., 1988) and oviposition deterrent activities (Sojitra and Patel, 1992) were reported for extracts of O. sanctum. In their investigations, only crude extracts were analyzed for bioactivity against insects. Biologically active components of these crude extracts, remain unknown. Preliminary bioassays indicated that the crude acetone extract from O. sanctum contained a compound or compounds that were demonstrating anti- feedant activity against Helicoverpa zea, corn earworm. This study undertook the bioassay-guided fractionation approach to determine the presence of insect anti-feedants or toxins utilizing H. zea, corn earworm bioassays. My initial hypothesis of finding compounds responsible for corn earworm anti-feedant activity evolved into a search for a porphyrin type compound as the fractionation progressed. A red fluorescing compound always seemed to be associated with the active fractions. Materials and Methods Refer to Chapter 3, Materials and Methods section for General Experimental, Plant Materials, and Extraction procedures. 69 Fractionation of crude acetone extract. A quantity of crude acetone extract (12.5 g) was suspended in acetone (125 mL) and then kept in a freezer at -20°C overnight to cause precipitation. After 24 h, the suspension was filtered through a bed of celite. The bioactive filtrate (3.3 g) subsequently was fractionated using reverse phase (Supelco bonded phase silica C-18) medium pressure column chromatography (MPLC). Solvents used and corresponding fractions collected were as follows: A (70:30; MeOHszo), B (90:10; MeOHzH20), C-l (100% MeOH). Two fractions, H and l were found to be active and identical according to TLC and subsequently were combined. The combined fractions (173.2 mg) were subjected to preparative TLC (6x200 um, KCF18 silica gel plates, 60 A) using 80:20 MeOHzHZO as the mobile phase which resulted in five fractions (A-E). The bioactive fraction, fraction D (6 mg) was subjected to a final preparative TLC experiment (1 x 200 um, KCF 18 silica gel plates, 60 A) that resulted in two bands, A (1.6 mg) and B (1.9 mg). Band A was pure enough to conduct UV experiments. The UV absorption spectrum obtained was found to be identical to the published data for the porphyrin, phylloerythrin (Perrin, 1958); however, this band may contain other related porphyrin compounds as impurities with similar UV absorption spectra profiles. These other porphyrins may have similar biological activities. Similarities between 1H-NMR data of porphyrins provided by Kenner et al. (1973) and the isolated compound were found. The tentative structure for the active compound was elucidated using UV and NMR studies. Band A; Porphyrin: UV Amax (MeOH) 659 nm, 584 nm, 557 nm, 512 nm, 420 nm. 1H-NMR(DMSO-d6) 8: 12.84 (1H, s, N-H), 13.99 (1H, s, N-H), 70 12.84 (1H, 8, COOH), 9.10, 8.52, 8.36 (3H, s, olefinic), 3.47 (2H, s, -CH2CO-), 3.25 (4H, s, -CH3), 3.37 (4H, m, CH2CH3), 1.09 (6H, t, CH20H3). 3.25 1.09 9.10 Com earwonn Anti-feedant Assay. Helicoverpa zea Boddie Noctuidae eggs were hatched in an incubator at 27°C. Dry diet (940 mg) obtained from North Carolina lnsectory was placed into glass vials. Crude extracts were dissolved in DMSO to give a concentration of 1250 mg-20pL". Subsequent fractions were assayed at progressively lower concentrations. DMSO test solutions (20 uL) were mixed thoroughly with the dry diet (940 mg). DMSO (20uL) plus dry diet (940 mg) was used as the control. Agar solution/suspension (1.4%) was autoclaved 5 min at 15 psi at 125°C to encourage melting of the agar. The temperature of the agar was dropped to 45°C by the use of a water bath set. The agar solution then was pipetted into individual vials containing the test mixtures until a weight of 5 g was obtained. The vials were mixed thoroughly and poured into 3.5 mL polystyrene vials. One neonate larvae was place in each 71 vial. Vials were capped and placed in a growth chamber with a photoperiod of 16-h days and 8-h nights. Day and night temperatures averaged 27°C. There were 15 replications per treatment. Larvae were weighed after six days. Bioassay protocols were adapted from previously published work (Bell, R. A., and Joachin, F. G., 1976 and Joyner, K. and Gould, F., 1985). An analysis of variance (ANOVA) was conducted on the data using a completely randomized design (CRD). Results and Discussion The porphyrin-containing fraction, responsible for the corn earworm anti- feedant activity, has a UV absorption spectrum identical to previously published data (Perrin, 1958); however, several other compounds including phylloerythrin methyl ester, have similar UV absorption spectra profiles (Wolf and Scheer, 1973). 1HNMR data indicated a mixture ofcompounds similar to previously published data (Kenner et al., 1973) namely, peaks for the ethyl, methyl, and unsaturated protons. The results of the corn earworm bioassay indicated that the treatments assayed at 100 ppm had a highly significant (p < 0.005) effect on the caterpillar weight (Figure 5). It is important to point out that the fractions assayed (C and D) contained the isolated compound in addition to smaller amounts of other 72 porphyrin-type compounds. Therefore, in actuality, biological activity might be due to other porphyrin type compounds working in concert or independently. Figure 5. Corn earworm antifeedant/toxic activity of two fractions containing isolated porphyrin. Significant at P < 0.005. A 37 0’ 1 gsti-.2 e s f 4,-2 ,.z____z :5, 2 l-_ _ i ,v , a i i m , 3 15 L 4 9 v i i i i , L 7 8, 1: J“? * +2 2 A _ L“ g0.5,-f _ s -5 E. 2 z < 01—2 , ,. .- - i c D Porphyrin-based compounds, such as phylloerythrin, result from the normal rumen microbial breakdown of chlorophyll from ingested forage plants in livestock animals. The phylloerythrin is absorbed and transported to the liver. Healthy livers transfer the phylloerythrin to the bile for excretion. Livers damaged by the fungal toxin, sporidesmin, cannot properly metabolize phylloerythrin, which then accumulates in peripheral blood. Therefore, circulating phylloerythrin in the blood causes the photosensitization reaction in nonpigmented skin (Cheeke, 1997). Photosensitization manifests itself often as facial eczema. The observed eczema is an example of "secondary photosensitization," in which the skin 73 lesions are really the secondary result of liver damage, rather than the direct result of a plant toxin (Hansen et al., 1994). Extrapolation of the above information to our corn earworm bioassays and the activity that was observed, may not be necessarily relative to photoxicity to H. zea caterpillars. However, it is possible that phototoxicity resulting from abnormal chlorophyll metabolism might be occurring in the H. zea gut and resulting in mortality and weight loss. As of 1995, phototoxin-mediated effects and phototoxicity in insects resulting from abnormal chlorophyll metabolism is not know to exist (Berenbaum, 1995); however, akin to this phenomenon, involved the administering of synthetic compounds which interfere with heme biosynthesis from protoporphyrin IX (Rebeiz et al., 1990 and 1988). Lethal accumulation of protoporphyrin lX in Trichoplusia ni Hubner and H. zea larvae was found to result following the administration of 2, 2’-dipyridyl alone or in combination with 6-aminolevulenic acid (Rebeiz et al., 1988). Lethal accumulation of protoporphyrin IX in the insect was associated with regurgitation, convulsions, and loss of body fluids. Larval death occurred in the dark and light, however, the treatment appeared to be photodynamic in nature. Also, exogenous applications of protoporphyrin and Mg- protoporphyrin also exhibited photodynamic damage. The compound, 1, 10- phenanthroline exhibited similar effects in T. ni (Rebeiz et al., 1990). The preliminary evidence in this study indicates that a porphyrin like compound is responsible for the observed mortality of corn earworm larvae. Whether or not a phototoxic or related effect is occurring, remains unknown. 74 Whatever the mode of action, this work may provide a basis for further research on the insecticidal activity of porphyrins. 75 Chapter 6 SUMMARY The body of information about 0. sanctum prior to this work was presented in Chapter 1 and also formed much of the impetus for the current work. Much of the literature summarized in Chapter 1 focused primarily on the biological activity of crude extracts of O. sanctum. This early work stimulated the formulation of a multidimensional hypothesis namely that, O. sanctum contains compounds capable of exhibiting anti-oxidant and anti-inflammatory as well as insecticidal and anti-feedant activities. Compounds possessing these activities were isolated and identified as discussed in Chapters 2-5. Identification of isolated compounds was determined by the use of various spectral techniques. Chapters 2 and 5 focused on mosquitocidal and corn earworm anti- feedant activities, respectively. Isolated as oils, eugenol and E—6—hydr0xy-4, 6- dimethyl-3-heptene-2-one demonstrated mosquitocidal activity at 200 and 6.25 pg-mL’1 in 24 h, respectively on fourth instar A. aegyptii larvae. Both compounds with the addition of a novel triglyceride, 1, 3-dilinoleneoyI-2-palmitin were isolated from the leaf and stem hexane extract of O. sanctum. Bioassay directed fractionation experiments conducted in Chapter 5 resulted in the identification of a porphyrin compound that displayed toxic activity against corn eanNorm larvae. Porphyrins are known for their photochemical activity. Therefore, the porphyrin isolated may possess a phototoxic or photosensitizing effect that results in the 76 death and lower weigh of the test insect by either interfering with heme biosynthesis or interaction with other chemical aspects of the insect. Given these preliminary data, further research into the insect toxicology of porphyrins is warranted. Preliminary anti-oxidant assays on crude extracts from O. sanctum had shown excellent anti-oxidant activity. With antioxidant activity in mind, the isolation and identification of eugenol, cirsilineol, cirsimaritin, isothymusin, isothymonin, apigenin, and rosmarinic acid from acetone extracts of fresh 0. sanctum leaves and stems was carried out in Chapter 3. Eugenol, cirsilineol, isothymusin and rosmarinic acid demonstrated good antioxidant activity when assayed at 10 uM. Cirsimaritin, isothymusin, and isothymonin were identified for the first time in O. sanctum. lsothymonin was also reported as a natural product for the first time. The antioxidant compounds isolated and identified in Chapter 3 were subjected to COX-1 inhibitory assays (Chapter 4). Eugenol was by far the most active, showing 97% COX-1 inhibitory activity when assayed at 1000 (M. The activity of eugenol exceeded the activity of aspirin by 51% at the same molar concentration. Apigenin and rosmarinic acid also performed well, demonstrating 65 and 58% COX-1 inhibitory activity, respectively. The other compounds demonstrated comparable activity with ibuprofen and aspirin at 10 pM and 1000 77 pM, respectively. Some of the same compounds demonstrated slightly higher levels of COX-2 inhibition at the 1000 uM thereby suggesting specificity. Natural mosquito control could conceivably be accomplished by utilizing conventional aerosol sprays or novel chemical dispersion technologies in the deployment of essential oils from O. sanctum. Human health concerns as well as an impact on the environment would be alleviated by the use of O. sanctum as a natural form of insect pest control. The anti-feedant and/or toxic nature of the porphyrin compound against corn earworm is new and therefore warrants further investigation. Determination of mode of action, phototoxicity, and its possible applications would be worthwhile endeavors for additional research. These research results could provide support for the use of O. sanctum in food supplements or nutraceuticals in the prevention of cancer and in the reduction of inflammation. The acceptance of O. sanctum as an alternative to conventional treatments would be well received by the public considering the current trend towards natural alternatives to drug treatments. This research has added to the body of literature dealing with O. sanctum. More specifically, the current work has led to the first identification of biologically active compounds from O. sanctum. Additional studies should be undertaken to 78 determine the presence of other biologically active compounds contained in O. sanctum. These compounds may hold potential for pharmaceutical and or agricultural applications in addition to being a worthwhile research endeavors. 79 REFERENCES Adiroubane, A. and Letchoumanane, S. (1998) Field efficacy of botanical extracts controlling major insect pests of Okra (Abelmoschus esculentus). Ind. J. Ag. Sci. 68, 168-170. Ahmad, S. K. and Prasad, J. S. (1995) Efficacy of foliar extracts against pre- and post-harvest diseases of sponge-gourd fruits. Left. App. Micro. 21, 373- 375. Ahumada, F., Trincado, M. A., and Arcellano, J. A. (1991) Effect of certain adaptogenic plant extracts on drug induced narcosis in female and male mice. Phytotherapy Res. 5, 29-30. Areekul, S., Sinchaisri, P., and Tigavatananon, S. (1988) Effects of Thai plant extracts on the Oriental fruit fly II. Repellency Test. Kaetsart J. (Nat. Sci.) 22, 56-61. Arneja, V., Sharma, D. N., Chand, D., and Singal, S. P. (1987) Some reproductive traits of white leghorn male birds as affected by Ocimum sanctum (Tulsi) feeding. Indian. J. Poult. Sci. 22, 279-282. Arora, A.; Nair, M. G. and Strasburg, G. M. (1998) Structure-activity relationships for antioxidant activities of a series of flavonoids in liposomal system. Free Radic. Biol. Med. 24, 1355-1363. Arora, A.; Nair, M. G. and Strasburg, G. M. (1997) A novel Fluorescence Assay to Evaluate Antioxidants Efficiency: Application to Flavonoids and 80 lsoflavonoids; Aruma, O. l. and Cuppett, S. 0., Ed.; AOCS Press, Illinois, pp 205-222. Aruna, K. and Sivaramakrishnan, V. M. (1992) Anticarcinogenic effects of some Indian plant products. Fd. Chem. Toxic 30, 953-956. Asthana, O. P. and Gupta, R. S. (1984) Effects of NP levels on physiological parameters at three growth stages in Sacred Basil (Ocimum sanctum). Indian Perfum 28, 49-53. Balanehru, S. and Nagarajan, B. (1991) Protective effect of oleanic acid and ursolic acid against lipid peroxidation. Biochem. Int. 24, 981-990. Balanehru, S. and Nagarajan, B. (1992) Intervention of adriamycin induced free radical damage. Biochem. Int. 70, 735-744. Banu, M. J., Nellaippan, K., and Dhandayuthapani, S. (1992) Mitochondrial malate dehydrogenase and malic enzyme of a filarial worm Setaria digitata: Some properties and effects of drugs and herbal extracts. Jpn. J. Med. Sci. Biol. 45, 137-150. Banerjee, S., Prashar, R., Kumar, A., and Rao, A. R. (1996) Modulatory influence of alcoholic extract of Ocimum leaves on Carcinogen- metabolizing enzyme activities and reduced glutathione levels in mouse. Nut. Can. 25, 205-217. Barberan, F. A. T., Ferres, F., and Tomas, F. (1985) TLC, UV and acidic treatment in the differentiation of 5,6-dihydroxyflavones and 5,8- dihydroxyflavones, 3-methoxyflavones and flavonols. Tetrahedron 41, 5733-5740. 81 Batta, S. K. and Santhakumari, G. (1970) The antifertility effect of Ocimum sanctum and Hibiscus Rosa Sinensis. Ind. J. Med. Res. 59, 777-781. Baumann, J., von Bruchhausen, F ., and Wurm, G. (1979) A structure activity on the influence of phenolic compounds and bioflavonoids on rat renal prostaglandin synthase. Naunyn Schmiedeberg’s Arch. Pharrncol. 307, 73-78. Bell, R. A., and Joachin, F. G. (1976) Techniques for rearing laboratory colonies of tobacco hornworrns and pink bollworrns. Ann. Entomol. Soc. Amer. 69, 365-373. Ben-Yakir, D., Bazar, A., and Chen, M. (1995) Attraction of Maladera matn'da (Coleoptera: Scarabaeidae) to eugenol and other lures. J. Econ. Bot. 88, 415-420. Berenbaum, M. (1995) Phototoxicity of plant secondary metabolites: Insect and mammalian perspectives. Arch Insect Biochem. Physiol. 29, 119-134. Butani, D. K. (1982) Insect pests of Tulsi (Ocimum sanctum Linnaeus) and their controls. Pesticides 16, 11-12. Bhargava, K. P. and Singh, N. (1981) Anti-stress activity of Ocimum sanctum Linn. Ind. J. Med. Res. 73, 443-451. Chatterjee, A., Sukul, N. C., Laskar, S., and Ghoshamajumdar, S. (1982) Nematicidal principles from two species of Lamiaceae. J. Nematology 14, 118-120. 82 Chattopadhyay, R. R. (1993) Hypoglycemic effect of Ocimum sanctum leaf extract in normal and streptozotocin diabetic rats. Ind. J. Exp. Biol. 31, 891-893. Cheeke, PR. (1997) Natural Toxicants in Feeds, Forages, and Poisonous Plants. Interstate Pub., Danville, IL. Chen, Z. Y., Chan, P. T., Ho, K. Y., and Wang, F. J. (1996) Antioxidant activity of natural flavonoids is governed by number and location of their aromatic hydroxyl groups. Chem. Phys. Lipids 79, 157-163. Cholbi, M. R., Paya, M., and Alcaraz, M. J. (1991) Inhibitory effects of phenolic compounds on CCl4-induced microsomal lipid peroxidation. Experentia 47, 195-199. Cos, P., Ying, L., Calomme, M., Hu, J. P., Cimanga,K., Van Poel, B., Pieters, L., Vlietinck, A. J., and Berghe, D. V. (1998) Structure-activity relationships and classification of flavonoids as inhibitors of xanthine oxidase and superoxide scavengers. J. Nat. Prod. 61, 71-76. Cuvelier, M. E., Richard, H., and Berset, C. (1996) Antioxidant activity and phenolic composition of pilot-plant and commercial extracts of sage and rosemary. J. Am. Oil Chem. Soc. 73, 645-652. Das, D. K. and Maulik, G. (1994) Evaluation of antioxidant effectiveness in ischemia reperfusion tissue injury. Methods in Enzymology 233, 601-610. Devi, P. U. and Ganasoundari, A. (1995) Radioprotective effect of leaf extract of Indian medicinal plant Ocimum sanctum. Ind. J. Exp. Biol. 33, 205-208. 83 Dey, B. B. and Choudhuri, M. A. (1983) Effect of leaf development stage on changes in essential oil of Ocimum sanctum L. Biochem. Physiol. Pflanzen. 178, 331-335. Deshmukh, P. B., Chavan, S. R., and Renapurkar, D. M. (1982) A study of insecticidal activity of twenty indigenous plants. Pesticides 12, 7-10. Direkbusarakom, S., Ezura, Y., Yoshimizu, M., and Herunsalee, A. (1998) Efficacy of Thai traditional herb extracts against fish and shrimp pathogenic bacteria. Fish Path. 33, 437-441. Duperrier, A., Moreau, M., and Dreux, J. (1975) Action des organomagnesiens mixtes et de l’hydrure de lithium sur les dihydropyrones. lI-Dihydro-3, 6- pyrones-2. Bull. Soc. Chim. 2307. Ferdous, A. J., Islam, N., Aiisan, M., and Faroque, A. B. M. (1990) Antibacterial activity of the leaves of Adhatoda vasica, Calotropis gigantea, Nerium odorum, and Ocimum sanctum. Bangladesh J. Bot, 19, 227-229. Ferreres, F., Barberan, F. A. T., and Tomas, F. (1985) 5, 6, 4’-trihydroxy-7, 8- dimethoxyflavone from Thymus membranaceus. Phytochemistry 24, 1869-1871. Godhwani, S., Godhwani, J. L., and Vyas, D. S. (1987) Ocimum sanctum: An experimental study evaluating its anti-inflammatory, analgesic and antipyretic activity in animals. J. Ethnophann. 21, 153-163. Godhwani, S., Godhwani, J. L., and Vyas, D. S. (1988) Ocimum sanctum - A preliminary study evaluating its immunoregulatory profile in albino rats. J. Ethnophann. 24, 193-198. 84 Grayer, R. J., Bryan, S. E., Veitch, N. C., Goldstone, F. J., Paton, A., and Wollenweber, E. (1996) External flavones in sweet basil, Ocimum basilicum, and related taxa. Phytochemistry 43, 1041-1047. Grayer, R. J. and Veitch, N. C. (1998) An 8-hydroxylated external flavone and its 8-O-glucoside from Becium grandiflorum. Phytochemistry 47, 779-782. Grote, R., Zeeck, A., Stilmpfel, J., and Zéhmer, H. (1990) Pyrrolizidinones Produced by Streptomyces olivaceus. Liebigs Ann. Chem. 256, 525-530. Hansen, D.E., R.D. McCoy, O.R. Hedstrom, S.P. Snyder, and P.J. Ballerstedt. (1994). Photosensitization associated with exposure to Pithomyces chartarum in lambs. J. Amer. Vet. Med. Assoc. 204,1668-1671. Hasan, S. B. and Deo, P. G. (1994) Ocimum sanctum seeds for mosquito control. Int. Pest Control Jan/Feb, 20-21. Horie, T., Kawamura, Y., Yamamoto, Kitou, T., and Yamashita, K. (1995) Synthesis of 5, 8—dihydroxy-6, 7-dimethoxyflavones and revised structures for some natural flavones. Phytochemistry 39, 1201-1210. Hoult, J. R. S., Moroney, M. A., and Paya, M. (1994) Actions of flavonoids and coumarins on lipoxygenase and cyclooxygenase. Meth. Enzymology 234, 443-455. Joyner, K. and Gould, F. (1985) Developmental consequences of cannabalism in Heliothis zea (Lepidoptera: Noctuidae). Ann. Entomol. Soc. Amer. 78, 24-28. 85 Kantak, N. M. and Gogate, M. G. (1992) Effect of short administration of Tulsi (Ocimum sanctum Linn.) on reproductive behavior of adult male rats. Ind. J. Physiol. Phannacol. 36, 109-111. Kasinathan, S., Ramakrishnan, S., and Basu, S. L. (1972) Antifertility effect of Ocimum sanctum L. Ind. J. Exp. Biol. 10, 23-25. Kelm, M. A. and Nair, M. G. (1998) Mosquitocidal compounds and a triglyceride, 1, 3-dilinoleneoyl-2-palmitin, from Ocimum sanctum. J. Ag. Food Chem. 46, 3092-3094. Kenner, G. W., McCombie, S. W., and Smith, K. M. (1973) Pyrroles and related compounds. Part XXIV. Separation and oxidative degradation of chlorophyll derivatives. J. C. S. Perkin [2517-2523. Khanna, S., Gupta, S. R., and Grover, J. K. (1986) Effect of long term feeding of Tulsi (Ocimum sanctum Linn.) On reproductive performance of adult albino rats. Ind. J. Exp. Biol. 24, 302-304. Kim, H. P., Mani, l., lversen, L., and Ziboh, V. A. (1998) Effects of naturally occurring flavonoids and biflavonoids on epidermal cyclooxygenase and lipooxygenase from guinea pigs. Prostaglandins, Leukotrienes, Ess. Fatty Acids. 58, 17-24. Kimura, K., Wada, T., Endo, M., and lwata, l. (1982) Lipids in Green and Red Bell Peppers. J. Jpn. Soc. Food Nutr. 35, 95-101. Laaskso, l., Seppanen-Laasko, T., Herrmann-Wolf, B., Kiihnel, N., and Knobloch, K. (1990) Constituents of the essential oil from the holy basil or tulsi plant, Ocimum sanctum. Planta Med. 56, 527. 86 Laneuville, O., Breuer, D. K., Dewitt, D. L., Hla, T., Funk, C. D., and Smith, W. L. (1994) Differential inhibition of human prostaglandin endoperoxidase H syntheses-1 and —2 by nonsteroidal anti-inflammatory drugs. J. Pharmacol. Exp. Ther. 271 (2), 927-934. Lim-Sylianco, C. Y., Jocano, A. P., and Lim, C. M. (1988) Antimutagenicity of twenty Philippine plants using the micronucleus test in mice. Philippine J. Sci. 117, 231-235. Lipsky, C. P. E., Abramson, S. B., Crofford, L., et al. (1998) The classification of cyclooxygenase inhibitors. J. Reumatology 25, 2298-2303. Malik, M. S. and Rafique, M. (1989) Effects of methanol extracts of Ocimum sanctum Linn. on jute semilooper, Anomis sabulifera Guen. Indian J. Ent. 51, 84-89. Malik, M. S., Rafique, M., Sattar, A., and Khan, S. A. (1987) The fatty acids of indigenous resources for possible industrial applications. Part XII. The fatty acids composition of the fixed oils of Ocimum sanctum and Salvia aegyptica seeds. Pakistan J. Sci. Ind. Res. 30, 369-371. Mandal, 8., Das, D. N., Kamala D. E., Ray, K., Roy, G., Chaudhuri, S. B., Sahana, C. C., Chaudhuri, S. B., and Chowdhuri, M. K. (1993) Ocimum sanctum Linn. - A study on gastric ulceration and gastric secretion in rats. Ind. J. Physiol. Phanncol. 37, 91-92. Markham, K. R. (1982) Techniques of Flavonoid Identification; Academic Press Inc. London, p 39-45. 87 Martinez, V., Barbera, O., Sanchez-Paraneda, J., and Marco, J. A. (1987) Phenolic and acetylenic metabolites from Artemisia assoana. Phytochemistry, 26, 2619-2624. Maulik, G., Maulik, N., Bhandari, V., Kagan, V. E., Pakrashi, S., and Das, D. K. (1997) Evaluation of antioxidant effectiveness of a few herbal plants. Free Rad. Res. 27, 221-228. Meade, E. A. ; Smith, W. L. ; and DeWItt, D. L. (1993) Differential inhibition of prostaglandin endoperoxide synthase (cyclooxygenase) isozymes by aspirin and other non-steroidal anti-inflammatory drugs. J. Biol. Chem, 268, 6610-6614. Mediratta, P. K., Dewan, V., Bhattacharya, S. K., Gupta, V. S., Maiti, P. C., and Sen, P. (1988) Effect of Ocimum sanctum Linn. on humoral immune responses. Ind. J. Med. Res. 87, 384-386. Mukhopadhyay, S. N. and Das, D. K. (1994) Oxygen Responses, Reactivities, and Measurements in Biosystems CRC Press, Florida. Nair, A. G. R., Gunasegaran, R., and Joshi, B. S. (1982) Chemical Investigation of Certain South Indian Plants. Ind. J. Chem. 213, 979-980. Nair, M. G., Putnam, A. R., Mishra, S. K., Mulks, M. H., Taft, W. H., Keller, B. E., Miller, J. R., Zhu, P. -P., Meinhart, D., and Lynn, D. G. (1989) Faeriefungin: A new broad-spectrum antibiotic from Streptomyces griseus Var. Autotrophicus. J. Nat. Pmd. 52, 797-809. Nakasugi, T. and Komai, K. (1998) Antimutagens in the Brazilian folk plant Carqueja (Baccharis trimera Less). J. Agric. Food Chem, 46, 2560-2564. 88 Narasimhan, V. and Mariappan, V. (1988) Effect of plant derivatives on green leafhopper (GLP) and rice tungro (RTV) transmission. IRRN. 13, 28-29. Nitao, J. K., Nair, M. G., Thorogood, D. L., Johnson, K. S., and Scriber, M. J. (1991) Bioactive Neolignans from the Leaves of Magnolia virginiana. Phytochemistry, 30, 2193-2195. N6rr, H. and Wagner, H. (1992) New constituents from Ocimum sanctum. Planta Med. 58, 574. Patil, R. K., Sharma, A., and Pathak, V. N. (1992) Inhibition of polyamine biosynthesis in Botryodiplodia theobromae Pat. and Rhizopus anhizus Fisher by Ocimum sanctum leaf extract. Indian J. Mycol. Pl. Pathol. 22, 201-202. Perrin, D. D. (1958) The determination of phylloerythrin in blood. Biochem. J. 68, 314-31 8. Phadke, S. A. and Kulkarni, S. D. (1989) Screening of in vitro antibacterial activity of Tenninala chebula, Eclapta alba, and Ocimum sanctum. Ind. Sci. Med. Sci. 43, 113-117. Prashar, R. and Kumar, A. (1995) Chemopreventive action of Ocimum sanctum on 2, 12-dimethylbenz(a)anthracene DMBA-induced papillomagenesis in the skin of mice. Int. J. Phannacog. 33, 181-187. Prashar, R., Kumar, A., Banerjee, S., and Rao, A. R. (1994) Chemopreventative action by an extract from Ocimum sanctum on mouse skin papillomagenesis and its enhancement of skin glutathione S-transferase activity and acid sulfdryl level. Anti-cancer Drugs 5, 567-572. 89 Priyadarsini, K. Y., Guha, S. N., and Rao, M. N. A. (1998) Physico-chemical properties and antioxidant activities of methoxyphenols. Free Radic. Biol. Med. 24, 933-941. Rebeiz, C. A., Juvik, J. A., and Rebeiz, C. C. (1990). Porphyric insecticides 2. 1, 10-phenanthroline, a potent porphyric insecticide modulator. Pestic. Biochem. Physiol. 36, 201-207. Rebeiz, C. A., Juvik, J. A., and Rebeiz, C. C. (1990). Porphyric insecticides 1. concept and phenomenology. Pestic. Biochem. Physiol. 24, 375-378. Reddi, G. S., Shukla, N. P., and Singh, K. V. (1986) Chemotherapy of tuberculosis - Antitubercular activity of Ocimum sanctum leafy extract. Fitoterpia 57, 114-115. Reghunandanan, R. Sood, S., Reghunandanan, V. Mehta, R. M., and Singh, G. P. (1995) Effect of Ocimum sanctum Linn. (Tulsi) extract on testicular formation. Indian J. Med. Sci. 49, 83-87. Renapurkar, D. M. and Deshmukh, P. B. (1984) Pulicidal activity of some indigenous plants. Insect Sci. Applic. 5, 101-102. Risvi, S. J. H. Discovery of natural herbicides. (1981) Pesticides, 3, 40-42. Roth, G. N., Chandra, A., and Nair, M. G. (1998) Novel Bioactivities of Curcuma longa Constituents. J. Nat. Prod. 61, 542-545. Roomi, M. W., Abbas, T., Shah, A. H., Robina, S., Qureshi, S. A., Hussain, S. S., and Nasir, K. A. (1993) Control of fruit-flies (Daucus spp.) by attractants of plant origin. Anzeiger fuer Schaedlingskunde Pflanzenschutz Umweltschutz. 66, 155-157. 90 Saeed, S. A., Simjee, R. U., Shamin, G., et al. (1995) Eugenol: A dual inhibitor of platelet-activating factor and arachidonic acid metabolism. Phytomedicine 2 (1), 23-28. Sadekar, R. D., Pimprikar, N. M., Bhandarkar, A. G., and Barmase, B. S. (1998) lmmunomodulating effect of Ocimum sanctum Linn. Dry leaf powder on humoral immune responses in poultry infected with IBD virus. Ind. Vet. J. 75, 73-74. Sadekar, R. D., Pimprikar, N. M., Bhandarkar, A. G., and Barmase, B. S. (1998) lmmunomodulating effect of Ocimum sanctum Linn. Dry leaf powder on cell mediated immune (CMI) responses in poultry, naturally infected with IBD virus. Ind. Vet. J. 75, 168-169. Saija, A., Scalese, M., Lanza, M., Marzullo, D., Bonina, F., Castilli, F. (1995) Flavonoids as antioxidant agents: Importance of their interaction with biomembranes. Free Rad. Biol. Med. 19, 481-486. Sakina, M. R., Dandiya, P. C., Hamdard, M. E., and Hameed, A. (1990) Preliminary psychopharmacological evaluation of Ocimum sanctum leaf extract. J. Ethnopharrn. 28, 143-150. Sangwan, N. K., Verma, B. S., Verma, K. K., and Dhindsa, K. S. (1990) Nematicidal activity of some essential plant oils. Pestic. Sci. 28, 331-335. Sarkar, A., Lavania, S. C., Pandley, D. N., and Pant, M. C. (1994) Changes in the blood lipid profile after administration of Ocimum sanctum (Tulsi) leaves in the normal albino rat. Ind. J. Physiol. Pharmacol. 38, 311-312. 91 Sarkar, A., Pandey, D. N., and Pant, M. C. (1990) A report on the effects of Ocimum sanctum (Tulsi) leaves and seeds on blood and urinary uric acid, urea, and urine volume in normal albino rabbits. Ind. J. Physiol. Pharmacol. 34, 61-62. Sarkar, A. and Pant, M. C. (1989) A comparative study of the hypoglycemic action of the seeds and fresh leaves of Ocimum sanctum (Tulsi). Ind. J. Physiol. Phannac. 33, 197-1 98. Sen, P., Maiti, P. 0., Ray, A., Audolov, N. A., and Valdman, A. V. (1992) Mechanism of anti-stress activity of Ocimum sanctum Linn., eugenol and Tinospora malabarica in experimental animals. Ind. J. Exp. Biol. 30, 592- 596. Seth, S. D., Johri, N., Sundaram, K. R. (1981). Antispermatogenic effect of Ocimum sanctum. Ind. J. Exp. Biol. 19,975-976. Simon, L. S., Lanza, F. L., Lipsky, P. E., et al. (1998) Preliminary study of safety and efficacy of SC-58635, a novel cyclooxygenase 2-inhibitor. Arthritis Rheumatism 41, 1591-1602. Sinhababu, S. P. and Sukul, N. C. (1984) Influence of inoculum densities of root- knot nematode on the total protein content of lady's finger plants and amelioration of root-knot disease by Ocimum sanctum. Indian Phytopath. 37, 188-191. Singh, S., Majumdar, D. K., and Rehan, H. M. S. (1996) Evaluation of anti- inflammatory potential of fixed oil of Ocimum sanctum (Holybasil) and its possible mechanism of action. J. Ethnopharmacog. 54, 19-26. 92 Singh, H. N. P., Prasad, M. M., and Sinha, K. K. (1993) Efficacy of leaf extracts of some medicinal plants against disease development in banana. Lett. App. Micro. 17, 269-271. Singh, S. and Majumdar, D. K. (1995) Analgesic activity of Ocimum sanctum and its possible mechanism of action. Int. J. Phannacog. 33, 188-192. Singh, S. and Majumdar, D. K. (1995) Anti-inflammatory and antipyretic activities of Ocimum sanctum fixed oil. Int. J. Phannacog. 33, 288-292. Singh, S., Majumdar, D. K., and Singh, J. P. (1995) Studies on therapeutic efficacy of fixed oil of Ocimum sanctum in bovine mastitis. Ind. Vet. J. 72, 867-869. Singh, S. and Agrawal, S. (1991) Anti-asthmatic and anti-inflammatory activity of Ocimum sanctum. Int. J. Phannacog. 29, 306-310. Sojitra, l. R. and Patel, J. R. (1992) Effect of plant extracts on oviposition behavior of spotted bollworm (Earias Vite/la) infesting okra (Hibiscus esculentus). Ind. J. Ag. Sci. 62, 848-849. Stein, U., Sayaampol, B., Klingauf, F., Bestmann, H-. J., Vostrowsky, O., Classen, B. (1988) Ahpidizide Wrrkung ethanolisher Extrakte aus dem Heiligen Basilikum, Ocimum sanctum. Entomol. Gener. 13, 229-237. Stryer, L. (1988) Biochemistry 3rd ed. W. H. Freeman and Company, New York p. 992. Sukari, M. A. and Takahashi, S. (1988) Biological activity of some Malaysian plant extracts. Pertanika 11, 249-253. 93 Tewari, S. N. (1995) Ocimum sanctum L., a botanical fungicide for rice blast control. Trop. Sci. 35, 263-273. Tewari, S. N. and Nayak, M. (1990) Activity of four plant leaf extracts against three fungal pathogens of rice. Trop. Agric. 68, 373-375. Todera, M., Ferrandiz, M. L., and Alcaraz, M. J. (1994) Influence of flavonoids on degranulation and arachidonic acid release in rat neutrophils. Z. Naturforsch. 49, 235-240. Vaishnav, H. B. (1988) Pharmacognostic studies of Ocimum basilicum L. and comparison with O. sanctum. Acta. Botanica Indica 16, 251-254. Vasudevan, P., Tandon, M., Namrata, P., Nuennerich, P., Mueller, F., Mele, A., and Lentz, H. (1997) Fluid CO; extraction and hydrodistillation of certain biocidal essential oils and their constituents. J. Sci. Ind. Res. 56, 662- 672. Yousseff, D., and Frahm, A. W. (1995) Constituents of the Eygptian Centaurea scopariazlll. Phenolic constituents of the aerial parts. Planta Med, 61, 570-573. Wagner, H., N6rr, H., and Vlfinterhoff, H. (1994) Plant Adaptogens. Phytomedicine 1, 63-76. Wang, M., Li, J., Rangarajan, M., Shao, Y., LaVoie, E. J., Huang, T-C., and Ho, C-H. (1998) Antioxidative phenolic compounds from sage (Salvia officinalis). J. Agric. Food Chem. 46, 4869-4873. Wolf, H and Scheer, H. Photochemishe Hydrierung von Phéioporphyrinen: 7, 8- cis-Phéiophorbide. Liebigs Ann. Chem. 1710-1740. 94 Zhu, M., Phillipson, J. D., Greengrass, P. M., and Bowery, N. G. (1996) Chemical and biological investigation of the root bark of Clerodendrum mandarinorum. Planta Med. 62, 393-396. 95 "1111111111111)