PHARMACEUTICAL CHEMISTRY FACULTY OF PHARMACEUTICAL … · i pharmaceutical chemistry ukwueze,...

187
i PHARMACEUTICAL CHEMISTRY UKWUEZE, STANLEY EJIKE (PG/PhD/05/40421) PHYTOCHEMICAL AND BIOACTIVITY-GUIDED EVALUATION OF THE ANTIBACTERIAL CONSTITUENTS OF PSIDIUM GUAJAVA (LINN.) AND LORANTHUS MICRANTHUS (LINN.) LEAVES Digitally Signed by: Content manager’s Name DN : CN = Webmaster’s name O = University of Nigeria, Nsukka OU = Innovation Centre Ugwoke Oluchi C. FACULTY OF PHARMACEUTICAL SCIENCES

Transcript of PHARMACEUTICAL CHEMISTRY FACULTY OF PHARMACEUTICAL … · i pharmaceutical chemistry ukwueze,...

i

PHARMACEUTICAL CHEMISTRY

UKWUEZE, STANLEY EJIKE

(PG/PhD/05/40421)

PHYTOCHEMICAL AND BIOACTIVITY-GUIDED

EVALUATION OF THE ANTIBACTERIAL CONSTITUENTS OF

PSIDIUM GUAJAVA (LINN.) AND LORANTHUS MICRANTHUS

(LINN.) LEAVES

Digitally Signed by: Content manager’s Name

DN : CN = Webmaster’s name

O = University of Nigeria, Nsukka

OU = Innovation Centre

Ugwoke Oluchi C.

FACULTY OF PHARMACEUTICAL SCIENCES

ii

PHYTOCHEMICAL AND BIOACTIVITY-GUIDED EVALUATION OF

THE ANTIBACTERIAL CONSTITUENTS OF PSIDIUM GUAJAVA

(LINN.) AND LORANTHUS MICRANTHUS (LINN.) LEAVES

UKWUEZE, STANLEY EJIKE

(PG/PhD/05/40421)

DEPARTMENT OF PHARMACEUTICAL CHEMISTRY,

FACULTY OF PHARMACEUTICAL SCIENCES,

UNIVERSITY OF NIGERIA, NSUKKA.

SUPERVISOR: PROF. (MRS.) P. O. OSADEBE

MARCH, 2014.

iii

PHYTOCHEMICAL AND BIOACTIVITY-GUIDED EVALUATION OF

THE ANTIBACTERIAL CONSTITUENTS OF PSIDIUM GUAJAVA

(LINN.) AND LORANTHUS MICRANTHUS (LINN.) LEAVES

UKWUEZE, STANLEY EJIKE

(PG/PhD/05/40421)

A THESIS SUBMITTED TO

THE DEPARTMENT OF PHARMACEUTICAL AND MEDICINAL

CHEMISTRY, FACULTY OF PHARMACEUTICAL SCIENCES,

UNIVERSITY OF NIGERIA, NSUKKA.

IN FULFILMENT OF THE REQUIREMENTS FOR THE AWARD OF

DOCTOR OF PHILOSOPHY (PhD) DEGREE IN PHARMACEUTICAL

& MEDICINAL CHEMISTRY.

iv

CERTIFICATION

Ukwueze, Stanley Ejike, a postgraduate student with registration number:

PG/PhD/05/40421 has satisfactorily completed the requirement for the award

of the degree of Doctor of Philosophy (PhD) of the Department of

Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences,

University of Nigeria, Nsukka.

The research embodied in this thesis is original and has not been submitted in

part or in full for the award of any other diploma or degree of this or any other

University.

_____________________________ ______________________

Prof. (Mrs.) P. O. Osadebe Prof. C. J. Mbah

(Supervisor) (Head of Department)

_____________________________

(External Examiner)

v

DEDICATION

To my beloved wife (Odimnobi), Nchedo, and my lovely roots: Osinachi,

Munachiso, Oluoma and Uchechi who have remained the priceless jewels and

adorable ornaments of my life.

vi

ACKNOWLEDGEMENT

To God be the Glory, great thing He has done! He has shown me His

faithfulness once again by making it possible for me to attain the zenith of this

tortuous and seemingly endless race. I will forever consecrate my life to Him!

My immeasurable gratitude goes to my supervisor, Prof. Mrs. P. O. Osadebe,

who God has used to guide and illuminate my academic path since my

undergraduate years. She and her inspirational husband, Rev. Prof. N. N.

Osadebe, have been more of motivators and mentors not just to me, but also to

my entire family in diverse realms. I will always remain indebted.

My appreciation also goes to my ever loving parents, Ichie and Mrs. S. K. C.

Ukwueze, who God has kept alive to witness their life-long dream of having

one of their children attain this academic summit. May God continue to

preserve their life for us!

My special acknowledgement goes to my brother, friend, colleague and

schoolmate, Dr. Okoye, F. B. C. who has always been of great support in times

of need. His role in carrying out the spectral analysis in Germany and the

eventual structure elucidations cannot be quantified. My appreciation also goes

to Dr. Peters at the Institute of Organic Chemistry, Heinrich-Heine-University

for the NMR measurements. I also acknowledge the contributions of the

Institute of Pharmaceutical Biology and Biotechnology, University of Heirich-

Heine-Düsseldorf for the HPLC/ESI-MS recordings.

vii

I would like at this juncture to appreciate the professional and elderly roles

played by some of my senior colleagues who kept prodding me on even when I

appeared distracted. Worthy of mention here include Prof. C. O. Onyeji (my

VC), Prof. Cyril Usifo (my amiable Prof), Assoc. Prof. Eseyin (my Oga), Prof.

Orisakwe, Prof. S. I. Ofoefule, Prof. Vincent Okore and Prof. O. K. Udeala (my

able Dean). The list is quite endless!

It will be so unfair if I shall end this eulogy without the appreciation of all the

laboratory staff that played one role or the other towards the accomplishment of

this study. Names like Chief Hassan and Mrs. Caleb come tops here. Also, my

colleagues in the Department of Pharmaceutical and Medicinal Chemistry,

University of Port Harcourt deserve special recognition.

Finally, my greatest felicitation goes to my beloved wife, Nchedo, and my

vibrant children: Osinachi, Munachiso, Oluomachukwu and Uchechi. I will

always remember the angelic voice of my sweet one (Odimnaheart) constantly

reminding me of her joy in seeing this work completed. Thank God it finally

came to a glorious end! My siblings equally deserve special recognition: Sis.

Vicky, Rev. Leo, Eby, Ify, Iyke, Chika and Eyinwa.

To anyone I might have forgotten to mention, please accept my apology and lets

all join hands to say three ‘Gbosas’ to one another for a job well done.

Merci! Danke!!

Ukwueze Stanley Ejike. February, 2014.

viii

ABSTRACT

Antibiotics have remained the mainstay of drug therapy of infectious diseases

worldwide. Their use is, however, limited by their numerous adverse effects and rapid

development of microbial resistance. Identification of natural products from plants that

may serve as valuable sources of antimicrobial agents for medicinal or agricultural uses

seems to be a viable alternative to the conventional antibiotics. To achieve this goal,

biological assays should be carried out in order to identify promising plant extracts,

guide the separation and isolation, and to evaluate "lead" compounds. Psidium guajava

Linn. and Loranthus micranthus Linn. have been employed traditionally in Nigeria and

other parts of the globe for the treatment of various human ailments such as wounds,

gastrointestinal tract disorders and other forms of infective and non-infective disorders.

The main objective of this study was to identify and isolate the antibacterial compounds

from the leaves of Psidium guajava and Loranthus micranthus L. The specific objectives

were to: (i) carry out phytochemical evaluation and isolation of antibacterial

constituents of the plants using standard methods, (ii) elucidate the structures of the

isolated secondary metabolites, and (iii) carry out antibacterial assay of the isolated

compounds.

Fresh leaves of Loranthus micranthus (Linn.) parasitic on the stem of Persea americana

were collected at Nsukka while those of Psidium guajava were collected from the bio-

resource area of the University of Port Harcourt in June 2010. The leaves were then

cleaned, air-dried for 14 days and milled to coarse powder. The powdered materials

(800 g each) were defatted with n-hexane (5 L) and extracted in a soxhlet extractor with

90.0 % methanol. The methanol extract was further fractionated to yield the

chloroform, ethyl acetate, acetone and methanol soluble fractions. Each of the fractions

was screened for antibacterial activity using Agar-well diffusion method. Phytochemical

tests were carried out using standard procedures. The fractions that had the best

antibacterial activity were subjected to column chromatographic separation and

monitored by analytical thin layer chromatography (TLC). The ethyl acetate fraction

(PsG-EF) from P. guajava that gave satisfactory bioassay result was subjected to further

Sephadex-LH 20 chromatographic fractionation and purification to afford ten fractions

(PsG-EF1 to PsG-EF10) which were pooled. Fractions PsG-EF4, PsG-EF5 and PsG-EF7

that had good antibacterial activity were subjected to semi-preparative reverse phase

high pressure liquid chromatography (HPLC) purification to isolate the phenolic

compounds; I-V. The structures of these compounds were elucidated by analytical and

spectral techniques which included: ultra violet (UV), proton nuclear magnetic

resonance (1H-NMR), carbon-13 nuclear magnetic resonance (13C-NMR), distortionless

ix

enhancement by polarization transfer (DEPT), proton-proton correlation spectroscopy

(1H-1HCOSY), heteronuclear multiple quantum correlation (HMQC), heteronuclear

multiple bond correlation (HMBC) and electron spray ionization-mass spectroscopy

(ESI-MS) analyses. The isolated compounds were screened against standard strains of

Staphylococus aureus (ATCC 25923) and Escherichia coli (ATCC 35219) using broth

dilution assay method, and the MIC values determined and compared with ceftriaxone.

All data obtained were analyzed by GraphPad Prism® 5 using differences in mean by

two-way ANOVA and further subjected to Bonferroni post-tests to compare replicate

means. The results were presented as mean ± SEM. Differences between means were

considered significant at P<0.05.

The results showed that the ethyl acetate fraction (PsG-EF) from P. guajava yielded one

known compound (IV) and four novel phenolic compounds (I, II, III & V). The isolated

compounds were elucidated as : 2,4-dihydroxy-6-O-βD-glucopyranosyl benzophenone

(I); 2,4-dihydroxy-3-methyl-6-O-βD-glucopyranosylbenzophenone (II); 2,4-dihydro xy-

3-methyl-6-O-βD-glucopyranosylbenzophenone (4→5", 6'→1") benzene-2",3",4",5"-

tetraol (III); quercertin-3-O-αL-arabinofuranoside (IV) and 2,4-dihydroxy-6-O-βD-

glucopyranosylbenzophenone (4→5", 6'→1") benzene-2",3",4",5"-tetraol (V).

Compounds I, II, III, and V are new natural products which have not been previously

reported in literature for this plant, guava, and the trivial names Guajaphenone A, B, C

and D were proposed, while Compound IV has been previously reported as Guaijaverin.

The various fractions of Psidium guajava L. exhibited significant (p < 0.05) antibacterial

activities while for Loranthus micranthus L., its various fractions showed significantly

lower values (p > 0.001) when compared with the control (ceftriaxone) suggestive of a

generally weak or negligible antibacterial action. All the isolated compounds from P.

guajava were also found to have moderate antibacterial activities against E. coli and S.

aureus in comparison with ceftriaxone whlie I and IV showed lower MICs than those of

the other isolates against the test organisms.

x

Table of Contents

Page

TITLE PAGE i

CERTIFICATION iii

DEDICATION iv

ACKNOWLEDGEMENT v

ABSTRACT vii

TABLE OF CONTENT ix

LIST OF FIGURES xiv

LIST OF TABLES xvii

CHAPTER ONE: GENERAL INTRODUCTION 1

1.0 PREAMBLE 1

1.1 INFECTIOUS DISEASES AND CONVENTIONAL ANTIBIOTIC THERAPY 2

1.2 LIMITATIONS OF CONVENTIONAL ANTIBACTERIAL

AGENTS 6

1.2.1 Bacterial Resistance 6

1.2.2 Adverse Reactions by Agents 7

1.3 HIGHER PLANTS AS ANTIMICROBIAL AGENTS 8

1.3.1 Medicinal Plants with Antibacterial Activity 9

xi

1.3.2 Plant Secondary Metabolites Associated with Antibacterial Effects 10

1.3.2.1 Flavones, Flavonoids and Flavonols 11

1.3.2.2 Alkaloids 12

1.3.2.3 Terpenoids and Essential Oils 13

1.3.2.4 Tannins 15

1.3.2.5 Miscellaneous Plant Constituents 17

1.4 LITERATURE REVIEWS OF PLANTS USED 21

1.4.1 Loranthus micranthus Linn 21

1.4.1.1 Taxonomy of L. Micranthus 21

1.4.1.2 Description of the Family, Genus and Species of L. micranthus 23

1.4.1.3 Ethnomedicinal Uses and Pharmacological Studies on

L. micranthus 24

1.4.2 Psidium guajava Linn 25

1.4.2.1 Taxonomy of Psidium guajava 25

1.4.2.2 Morphology of Psidium guajava 26

1.4.2.3 Ethnomedicinal Uses and Pharmacological Studies of

Psidium guajava 28

1.4.2.4 The Phytochemistry of Psidium guajava 32

1.5 BIOASSAY-GUIDED CHARACTERIZATION OF ANTI-BACTERIAL CONSTITUENTS FROM HIGHER PLANTS 37

1.5.1 Principles of Bioassay 38

xii

1.5.2 Screening Methods for Antibacterial Agents from Higher Plants 38

1.5.2.1 Test Organisms and Culture Media 38

1.5.2.2 Antibacterial Testing 40

1.6 STRUCTURE ELUCIDATION OF BIOACTIVE PLANT METABOLITES 42

1.6.1 Preliminary Analysis 42

1.6.2 Application of Modern Analytical Techniques 43

1.7 STATEMENT OF PROBLEM 44

1.8 JUSTIFICATION OF THE STUDY 44

1.9 AIMS AND SCOPE OF THE WORK 45

CHAPTER TWO: MATERIALS AND METHODS 46

2.1 MATERIALS 46

2.1.1 Plant Materials 46

2.1.2 Microorganisms Used 46

2.1.3 Solvents and Reagents 47

2.1.4 Materials and General Instruments 48

2.2 METHODS 50

2.2.1 Extraction and Fractionation of Plant Materials 50

xiii

2.2.2 Isolation and Purification of the Active Constituents from PsG-EF 54

2.2.3 HPLC Analysis of Active Constituents from PsG-EF 55

2.2.4 Electron Spray Ionization Mass Spectrometry (HPLC/ESI-MS)

of Isolates 55

2.2.5 Nuclear Magnetic Resonance (NMR) Spectroscopy of Isolates 56

2.2.6 Preliminary Screening of Extracts and Fractions

for Antimicrobial Activity 56

2.2.7 Phytochemical Screening of Plant Extracts and Fractions 57

2.2.8 Antibacterial Screening of Isolates 57

2.2.9 Statistical Analyses 58

CHAPTER THREE: RESULTS 59

3.1 Extraction and Solvent Fractionation 59

3.2 Preliminary Phytochemical and Antibacterial Screening Results 61

3.3 Isolation of Bioactive Constituents 73

3.4 Structure Elucidation of Isolated Compounds 74

3.4.1 PsG-EF4A (Compound I) 74

3.4.2 PsG-EF4B (Compound II) 76

3.4.3 PsG-EF7A (Compound III) 78

3.4.4 PsG-EF7D (Compound IV) 80

xiv

3.4.5 PsG-EF7E (Compound V) 81

3.5 Antibacterial Profile of Bioactive Constituents 83

CHAPTER FOUR: DISCUSSION AND CONCLUSION 84

4.1 Discussion 84

4.2 Conclusion 98

REFERENCES 100

APPENDICES 113

xv

LIST OF FIGURES

page

Fig. 1.0: Flowers and Leaves of Loranthus micranthus 22

Fig. 2.0: Loranthus micranthus parasitic on a host tree 23

Fig. 3.0 Psidium guajava Tree 27

Fig. 4.0 Leaves and Flowers of Psidium guajava 27

Fig. 5.0 Fruit of Psidium guajava 28

Fig. 6: Schematic Diagram of the Extraction/Fractionation 52

Procedure for L. micranthus Leaves.

Fig. 7: Schematic Diagram of the Extraction/Fractionation 53

Procedure for P. guajava Leaves.

Fig. 8: Graph of Mean IZD (mm) ± SEM of the extracts/fractions of

L. micranthus leaves against bacteria 68

Fig. 9a: Structure of Compound I 74

Fig. 9b: Numbering of Carbon Skeleton of Compound I 75

Fig. 10a: Structure of Compound II 76

Fig. 10b: Numbering of Carbon Skeleton of Compound II 77

Fig. 11a: Structure of Compound III 78

Fig. 11b: Numbering of Carbon Skeleton of Compound III 79

Fig. 12: Structure of Compound IV 80

Fig. 13: Structure of Compound V 81

xvi

Appendix 1: ESI-MS /UV Spectra of Compound I 113

Appendix 2: H-NMR Spectrum (500MHz; MeOD) of Compound I 114

Appendix 3: H-NMR Spectrum (600MHz; MeOD) of Compound I 115

Appendix 4: H-NMR Spectrum (600MHz; DMSO-d6) of Compound I 116

Appendix 5: 2-D COSY of Compound I 117

Appendix 6: C-13 NMR Spectrum of Compound I 118

Appendix 7: HMQC Spectrum of Compound I 119

Appendix 8: HMBC Spectrum of Compound I 120

Appendix 9: ESI-MS /UV Spectra Compound II 121

Appendix 10: H-NMR Spectrum (500MHz; MeOD) of Compound II 122

Appendix 11: H-NMR Spectrum (600MHz; DMSO) of Compound II 123

Appendix 12: 2-D COSY of Compound II 124

Appendix 13: C-13 NMR Spectrum of Compound II 125

Appendix 14: ESI-MS /UV Spectra Compound III 126

Appendix 15: H-NMR Spectrum (500MHz; MeOD) of Compound III 127

Appendix 16: H-NMR Spectrum (600MHz; DMSO) of Compound III 128

Appendix 17: 2-D COSY of Compound III 129

Appendix 18: C-13 NMR Spectrum of Compound III 130

Appendix 19: HMQC Spectrum of Compound III 131

Appendix 20: HMBC Spectrum of Compound III 132

Appendix 21: UV Spectrum of Compound IV 133

Appendix 22: ESI-MS Spectra of Compound IV 134

xvii

Appendix 23: H-NMR Spectrum (500MHz; MeOD) of Compound IV 135

Appendix 24: 2-D COSY of Compound IV 136

Appendix 25: H-NMR Spectrum (500MHz; MeOD) of Compound V 137

Appendix 26: 2-D COSY of Compound V 138

Appendix 27: 2-D COSY [aromatic region] of Compound V 139

xviii

LIST OF TABLES Page

Table 1: Some plant species with potential antimicrobial activities... 9

Table 2: Yield from extracts/fractions of the leaves of L. micranthus. 59

Table 3: Yield from extracts/fractions of the leaves of P. guajava …. 60

Table 4: Results of phytochemical tests on the leaf extract of

Loranthus micranthus parasitic on different host trees.... 61

Table 5: Results of the anti-microbial screening of extracts of

mistletoe from six different host plants... 62

Table 6: Results of phytochemical tests on the leaf extract of

Loranthus micranthus harvested at different seasons... 63

Table 7: Results of the anti-microbial screening of leaf extracts of

Loranthus harvested at different seasons... 64

Table 8: Result of MICs of leaf extracts of African mistletoe harvested

from P. americana against some fungi... 65

Table 9: Results of phytochemical tests on the solvent fractions of L.

micranthus leaves harvested from P.americana... 66

Table 10: Result of mean IZD (mm) ± SEM for L. micranthus

extracts/fraction… 67

Table 11: Results of phytochemical tests on the leaf extract of

P. guajava harvested at different seasons.... 69

Table 12: Results of the anti-microbial screening of leaf extracts of

P. guajava harvested at different seasons... 70

Table 13: Results of phytochemical tests on the solvent fractions

of Psidium guajava leaves... 71

Table 14: Table of Mean IZD (mm) +/- SEM for P. guajava

extracts/fractions... 72

Table 15: 1H and

13C-NMR data of Compound I... 75

Table 16: 1H and

13C-NMR data of Compound II... 77

Table 17: 1H and

13C-NMR data of Compound III... 79

xix

Table 18: 1H and

13C-NMR data of Compound V... 82

Table 19: Antibacterial profile of the isolated compounds against

Staphylococcus aureus and E. coli... 83

xx

CHAPTER ONE

GENERAL INTRODUCTION

1.0 PREAMBLE

Over the years, medicines and medicinal agents derived from plants have

made large contributions to human health and well-being. This is because they

are either used directly as phytomedicines for the treatment of various

ailments or they may become the base and the natural blueprint for the

development of new drugs (Cseke et al, 2006).

Herbal medicine also called phytotherapy or phytomedicine has been around

since the beginning of recorded history. It has also been described as the

therapeutic use of medicinal plants referred to as herbs (Thea et al, 2008).

Herbal medicine has become an integral part of standard health care, based on

a combination of time honored traditional usage and ongoing scientific

research. Surging interest in medicinal herbs has increased scientific scrutiny of

their therapeutic potential and safety. Some of the medicinal plants are

believed to enhance the natural resistance of the body to infections (Atal et al,

1986).

According to the World Health Organisation (WHO), herbal medicines could

also be referred to as phytopharmaceuticals sold as over the counter products

in modern dosage forms such as tablets, capsules, syrups or liquids for oral use

or dietary supplements containing herbal products, also called nutraceuticals

xxi

available in modern dosage forms, or even referred to as medicines consisting

of other crude, semi processed or processed medicines, which have a vital

place in primary health care and developing countries like Nigeria.

Traditional medicines are finished drug products intended for self-medication

or application that contain, as the active principles, herbal ingredients that

have received relatively little attention in world scientific literature, but for

which traditional or folkloric use is well documented in herbal references. It

may contain chemically defined or herbal based materials in addition to the

active principles (Canada, 1989).

The medicinal properties of plant have been investigated in the light of recent

scientific development throughout the world due to their potent

pharmaceutical activities and low toxicity. Today many countries still rely on

the medical values of herbs and use of medicinal plants for their therapeutic

practices (Thea et al, 2008). In this same vein, Nigeria which is having a vast

heritage of knowledge and expertise in herbal medicines is not an exception.

Finally, it has been variously established that the identification of these natural

products from plants that may serve as valuable sources of bioactive agents for

medicinal and agricultural uses largely depends on bioactivity-directed

isolation (Cseke et al, 2006).

1.1 INFECTIOUS DISEASES AND CONVENTIONAL ANTIBIOTIC THERAPY

xxii

Infectious diseases, also known as contagious diseases or transmissible

diseases, and include communicable diseases, comprise clinically evident

illness (i.e., characteristic medical signs and/or symptoms of disease) resulting

from the infection, presence and growth of pathogenic biological agents in an

individual host organism. In certain cases, infectious diseases may be

asymptomatic for much or their entire course. Infectious pathogens include

some viruses, bacteria, fungi, protozoa, multicellular parasites, and aberrant

proteins known as prions. These pathogens are the cause of disease epidemics,

in the sense that without the pathogen, no infectious epidemic occurs.

Transmission of pathogen can occur in various ways including physical contact,

contaminated food, body fluids, objects, airborne inhalation, or through vector

organisms (Ryan and Ray, 2004). Infectious diseases that are especially

infective are sometimes called contagious and can be easily transmitted by

contact with an ill person or their secretions. Infectious diseases with more

specialized routes of infection, such as vector transmission or sexual

transmission, are usually regarded as contagious but do not require medical

quarantine of victims.

The term infectivity describes the ability of an organism to enter, survive and

multiply in the host, while the infectiousness of a disease indicates the

comparative ease with which the disease is transmitted to other hosts; and as

such, an infection is not synonymous with an infectious disease, as some

infections do not cause illness in a host (Ryan and Ray, 2004).

xxiii

Among the almost infinite varieties of microorganisms, relatively few cause

disease in otherwise healthy individuals. Infectious disease results from the

interplay between those few pathogens and the defenses of the hosts they

infect. The appearance and severity of disease resulting from any pathogen

depends upon the ability of that pathogen to damage the host as well as the

ability of the host to resist the pathogen. Clinicians therefore classify infectious

microorganisms or microbes according to the status of host defenses - either

as primary pathogens or as opportunistic pathogens:

Primary pathogens cause disease as a result of their presence or activity within

the normal, healthy host, and their intrinsic virulence (the severity of the

disease they cause) is, in part, a necessary consequence of their need to

reproduce and spread. Many of the most common primary pathogens of

humans only infect humans; however many serious diseases are caused by

organisms acquired from the environment or which infect non-human hosts.

Organisms which cause an infectious disease in a host with depressed

resistance are classified as opportunistic pathogens. Opportunistic disease may

be caused by microbes that are ordinarily in contact with the host, such as

pathogenic bacteria or fungi in the gastrointestinal or the upper respiratory

tract, and they may also result from (otherwise innocuous) microbes acquired

from other hosts (as in Clostridium difficile colitis) or from the environment as

a result of traumatic introduction (as in surgical wound infections or compound

fractures). An opportunistic disease requires impairment of host defenses,

which may occur as a result of genetic defects (such as chronic granulomatous

xxiv

disease), exposure to antimicrobial drugs or immunosuppressive chemicals (as

might occur following poisoning or cancer chemotherapy), exposure to ionizing

radiation, or as a result of an infectious disease with immunosuppressive

activity (such as with measles, malaria or HIV disease). Primary pathogens may

also cause more severe disease in a host with depressed resistance than would

normally occur in an immunosufficient host.

Many human diseases are caused by pathogenic organisms resulting

sometimes in high mortality figures. Among these pathogens, bacteria account

for a reasonable percentage of causative organisms implicated in human

infectious diseases. Over the years, infections have been managed by the

conventional antibiotics. Antibiotics are microbial metabolites or synthetic

analogues inspired by them that, in small doses, inhibit the growth and survival

of microorganisms without serious toxicity to the host. They therefore exhibit

selective toxicity. In many cases, the clinical utility of natural antibiotics has

been through medicinal chemistry manipulations of the original structure

leading to broader antimicrobial spectrum, greater potency, lesser toxicity,

more convenient administration, and additional pharmacokinetic advantages.

Through customary usage, the many synthetic substances that are unrelated to

natural products but still inhibit or kill microorganisms are referred to as

antimicrobial agents instead (Martin, 1998).

Antibiotics are used to treat infections caused by organisms that are sensitive

to them, usually bacteria or fungi. They may alter the normal microbial content

xxv

of the body (e.g. in the intestine, lungs, bladder) by destroying one or more

groups of harmless or beneficial organisms, which may result in infections

(such as thrush in women) due to overgrowth of resistant organisms. These

side-effects are most likely to occur with broad-spectrum antibiotics (those

active against a wide variety of organisms). Resistance may also develop in the

microorganisms being treated; for example, through incorrect dosage or over-

prescription. Antibiotics should, therefore, not be used to treat minor

infections, which will clear up unaided. Some antibiotics may, in addition,

cause allergic reactions (Hendricks and Nemeth, 2010).

1.2 Limitations of Conventional Antibacterial Agents

1.2.1 Bacterial Resistance

Resistance is the failure of microorganisms to be killed or inhibited by

antimicrobial treatment. Resistance can either be intrinsic (exist before

exposure to drugs) or acquired (develop subsequent to exposure to a drug).

Resistance of bacteria to the toxic effects of antimicrobial agents and to

antibiotics develops fairly easily both in the laboratory and in the clinic and is

an ever-increasing public health hazard.

In clinical practice, resistance more commonly takes place by Resistance (R)

factor mechanisms. In more lurid examples, enzymes are elaborated that

attack the antibiotic and inactivate it. Mutations leading to resistance occur by

many mechanisms. They can result from point mutations, insertions, deletions,

xxvi

inversions, duplications and transpositions of segments of genes or by

acquisition of foreign DNA from plasmids, bacteriophages, and transposable

genetic elements.

These mechanisms can convert an antibiotic-sensitive cell to an antibiotic -

resistant cell. This can take place many times in a bacterium's already short

generation time.

Bacterial resistance generally is mediated through one of three mechanisms:

• Failure of the drug to penetrate into or stay in the cell

• Destruction of the drug by defensive enzymes, or

• Alterations in the cellular targets of the enzymes.

All these call for conservative but aggressive application of appropriate

antimicrobial chemotherapy. In many cases, however, a resistant

microorganism can still be controlled by achievable, though higher, doses than

are required to control sensitive populations. These higher doses must be

cautiously employed as they may predispose the patient to antibiotic adverse

reactions that can be life-threatening.

1.2.2 Adverse Reactions by Agents

Many patients placed on conventional antibiotics have reported various cases

of adverse drug reactions ranging from mild to severe/life-threatening

reactions including arrhythmias, hepatotoxicity, acute renal failure, and

antiretroviral therapy-induced lactic acidosis (Granowitz et al, 2008). Adverse

reactions associated with drug use include allergies, toxicities, and side effects.

xxvii

An allergy is a hypersensitivity reaction to a drug. Many allergies are IgE-

mediated and occur soon after drug administration. Examples of IgE-mediated

type 1 hypersensitivity reactions include early-onset urticaria, anaphylaxis,

bronchospasm, and angioedema. Non-IgE-mediated reactions include

hemolytic anemia, thrombocytopenia, acute interstitial nephritis, serum

sickness, vasculitis, erythema multiforme, Stevens-Johnson syndrome, and

toxic epidermal necrolysis. Toxicity, which is generally due to either excessive

dosing or impaired drug metabolism, is a consequence of administering a drug

in quantities exceeding those capable of being physiologically ‘‘managed’’ by

the host. Examples of toxicity caused by excessive dosing include penicillin-

related neurotoxicity (e.g. twitching, seizures) and the toxicities caused by

aminoglycosides. Side effects include adverse reactions that are neither

immunologically mediated nor related to toxic levels of the drug. An example is

the dyspepsia caused by erythromycin.

Various forms of frequently encountered toxicities/adverse reactions include

anaphylaxis, cardiotoxicity, nephrotoxicity, adverse heamatological and

dermatological reactions, neurotoxicity, hepatotoxicity, muscoskeletal

tocxicity, electrolyte and glucose abnormalities, fever, antibiotic-associated

diarrhea/colitis, etc (Granowitz et al, 2008).

1.3 HIGHER PLANTS AS ANTIMICROBIAL AGENTS

The emergence of pathogenic microbes with increased resistance to

established antibiotics provides a major incentive for the discovery of new

xxviii

antimicrobial agents. Antimicrobial screening of plant extracts and

phytochemicals then represents a starting point for antimicrobial drug

discovery.

Main-stream medicine is increasingly receptive to the use of antimicrobial and

other drugs derived from plants, as traditional antibiotics (products of

microorganisms or their synthesized derivative) become ineffective and as

new, particularly viral, diseases remain intractable to this type of drug. Another

driving factor for the renewed interest in plant antimicrobials in the past 20

years has been the rapid rate of (plant) species extinction (Lewis and Elvin-

Lewis, 1995). There is a feeling among natural-products chemists and

microbiologists alike that the multitude of potentially useful phytochemical

structures which could be synthesized chemically is at risk of being lost

irretrievably. There is a scientific discipline known as ethnobotany (or

ethnopharmacology), whose goal is to utilize the impressive array of

knowledge assembled by indigenous peoples about the plant and animal

products they have used to maintain health (Rojas et al, 1992). Lastly, the

ascendancy of the human immunodeficiency virus (HIV) has spurred intensive

investigation into the plant derivatives which may be effective, especially for

use in underdeveloped nations with little access to expensive Western

medicines.

1.3.1 Medicinal Plants with Antibacterial Activity

xxix

Many tropical and non-tropical plants have been evaluated for their

antimicrobial activity. For want of space, representatives of diverse plant

families with documented antibacterial activity and the organisms tested are

tabulated below.

Table 1: Some plant species with potential antimicrobial activities

Common/Botanical name Family Bacterial Susceptibility

Celosia argentea L. Amaranthaceae K. pneumoniae

Tylophora indica (Burm.f.) Merr. Asclepiadaceae K. Pneumoniae

Vernonia anthelmintica (L.) Willd. Asteraceae K. Pneumoniae

Balanites aegyptiaca (L.) Del. Balanitaceae K. Pneumoniae, S. typhimurium

Spathodea campanulata Beauv Bignonaceae K. Pneumoniae

Cassia fistula L. Caesalpiniaceae K. Pneumoniae, P. mirabilis

Beta vulgaris L. Chenopodiaceae K. Pneumoniae

Spinacia oleracea L. Chenopodiaceae K. Pneumoniae, P. mirabilis

Commelina benghalensis L. Commelinaceae K. Pneumoniae

Rourea santaloides (Vahl.) Connaraceae E. aerogenes; K. Pneumoniae; P.

mirabilis

Cressa cretica L. Convolvulaceae K. Pneumoniae

Lepidium sativum L. Cruciferae S. typhimurium

xxx

Momordica charantia L. Cucurbitaceae E. aerogenes; K. Pneumoniae; P.

mirabilis

Cyperus scarious R.Br. Cyperaceae E. aerogenes; K. Pneumoniae; P.

mirabilis

Ricinus communis L. Euphorbiaceae K. Pneumoniae, P. mirabilis

Arachis hypogaea L. Fabaceae E. aerogenes; K. Pneumoniae;

Vigna radiata L. Fabaceae K. Pneumoniae; P. mirabilis

Fumaria indica (Haussk.) Pugsley. Fumariaceae K. Pneumoniae; P. mirabilis

Ocimum kilimanjaricum L. Labiatae E. aerogenes; E. coli; K. Pneumoniae; P.

mirabilis; P. vulgaris.

Artocarpus hetrophyllus Lam. Moraceae E. aerogenes; P. mirabilis

Ficus elastica Roxb. Moraceae K. Pneumoniae; P. mirabilis

Piper longum L. Piperaceae E. aerogenes; K. Pneumoniae; P.

mirabilis; S. typhimurium .

Gardenia resinifera Roth. Rubiaceae K. Pneumoniae; P. mirabilis

Mesua ferra Linn. Guttiferae E. aerogenes; K. Pneumoniae; P.

mirabilis; P. vulgaris.

Alchornea cordifolia Euphorbiaceae K. Pneumoniae; E. coli; B. subtilis; S.

aureus

Chromolaena odorata Asteraceae Propionibacterium canes;

Mycobacterium spps.

(Parekh and Chanda, 2007; Okoye and Ebi, 2007)

1.3.2 Plant Secondary Metabolites Associated with Antimicrobial Effect

xxxi

Plants have been shown to possess an amazing potential to synthesize

aromatic substances, most of which are phenols or their oxygenated-

substituted derivatives. These substances have been reported to consist of

mostly secondary metabolites, of which at least 12,000 have been isolated, a

number estimated to still be less than 10% of the total (Schultes, 1978).

Plant secondary metabolites, in many cases, serve as plant defense

mechanisms against predation by microorganisms, insects, and herbivores.

Some, such as terpenoids give plants their odours; others (e.g. quinones and

tannins) are responsible for plant pigmentation. Many of these compounds are

also responsible for plant flavour. It is therefore not surprising that useful

antimicrobial phytochemicals have been derived from these plant secondary

metabolites.

1.3.2.1 Flavones, Flavonoids and Flavonols

Flavones are phenolic structures containing one carbonyl group (as opposed to

the two carbonyls in quinones). Addition of a 3-hydroxy group yields a flavonol

while flavonoids are also hydroxylated phenolic substances but occur as a C6-

C3 unit linked to an aromatic ring. These compounds have been known to be

synthesized by plants in response to microbial infection and have equally been

found in vitro to be effective antimicrobial substances against a wide array of

microorganisms (Dixon et al, 1983; Cowan, 1999). Their activity is probably due

to their ability to complex with extracellular and soluble proteins and to

complex with bacterial cell walls, often leading to the inactivation of the

xxxii

proteins, loss of function and cell lysis (Stern et al, 1996). More lipophilic

flavonoids may also disrupt microbial membrane (Tsuchiya et al, 1996).

Example of flavonoid compounds with known antimicrobial activity includes

the catechins. These are the most reduced form of the C-3 unit in flavonoid

compounds that have been extensively researched due to their occurrence in

oolong green teas. Teas have been reported to exert antimicrobial activity and

also contain a mixture of catechin compounds which inhibited in vitro activity

of Vibrio cholerae 01, Streptococcus mutans, Shigella, and other bacteria and

microorganisms (Toda et al, 1989; Batista et al, 1994; Borris, 1996; Sakanaka et

al, 1989; Sakanaka et al, 1992; Vijaya et al, 1995).

Examples of compounds in these groups with antimicrobial activity include

flavone (I), catechin (II), chrysin (III) and quercetin (IV).

I II

xxxiii

III IV

1.3.2.2 Alkaloids

These are basically heterocyclic nitrogenous compounds. Many of these

compounds found in higher plants have shown promising antibacterial activity.

For instance, diterpenoid alkaloids, commonly isolated from the plants of the

Ranunculaceae, or buttercup family, are commonly found to have

antimicrobial properties (Omulokoli et al, 1997). Some of the highly aromatic

planar quaternary alkaloids such as berberine (V) have their mechanism of

action attributable to their ability to intercalate with DNA (Phillipson and

O'Neill, 1987). Other alkaloids with antimicrobial actions are Harmane (VI),

Piperine (VII), etc.

V VI

xxxiv

VII

1.3.2.3 Terpenoids and Essential Oils

Essential oils are secondary metabolites that are highly enriched in compounds

based on an isoprene structure. The observed fragrance in most plants is

contained in their essential oil fraction. They consist mainly of compounds

belonging to the chemical group called terpenes. When the compounds

contain additional elements, usually oxygen, they are called terpenoids.

Terpenoids, even though synthesized from acetate units, differ from fatty acids

by their extensive branching and cyclization.

Many plant terpenoids have been found to be active against bacteria, fungi,

viruses and protozoa (Amaral et al, 1998; Habtemariam et al, 1993; Himejima

et al, 1992; Mendoza et al, 1997; Kubo et al, 1993; Hasegawa et al, 1994;

Ghoshal et al, 1996; Rao et al, 1993; Sun et al, 1996; Tassou et al, 1995; 2000).

Although the mechanism of their antibacterial action is not yet fully

understood, terpenes are speculated to act by disrupting cell membranes due

to their lipophilic nature. In fact, it has been reported as far back as 1977 that

of all the essential oil derivatives being examined, 30% were inhibitory to

xxxv

bacteria while 60% inhibited fungi (Chaurasia and Vyas, 1977). For instance,

capsaicin [(VIII); a terpenoid constituent found in Chiles peppers] in addition to

its wide range of biological activities in humans, has been shown to clearly

inhibit various bacteria to differing extents; and although possibly detrimental

to the human gastric mucosa, it is bactericidal to Helicobacter pylori (Cichewicz

and Thorpe, 1996; Jones et al, 1997). Also, the ethanol-soluble fraction of

purple prairie clover yields a terpenoid called petalostemumol, which showed

excellent activity against Bacillus subtilis and Staphylococcus aureus but lesser

activity against Gram-negative bacteria as well as Candida albicans (Hufford et

al, 1993). In the same vein, two diterpenoid compounds isolated from the

roots of Plectranthus hereroensis were found to have good activity against S.

aureus, V. cholerae, P. aeruginosa, and Candida spp (Batista et al, 1994); while

another diterpene, trichorabdal A, isolated from a Japanese herb by Kadota et

al (1997) was found to directly inhibit H. Pylori. Terpeoids like menthol (IX) and

artemisin (X) have also shown antimicrobial activity.

VIII

xxxvi

IX X

1.3.2.4 Tannins

The term 'tannin' is used to describe a group of polymeric phenolic substances

capable of tanning leather or precipitating gelatin from solution. The property

is known as astringency. Tannins which are found in almost every plant part

are divided into hydrolyzable and condensed tannins. Hydrolyzable tannins are

based on gallic acid, usually as multiple esters with D-glucose; while the more

numerous condensed tannins (often called proanthocyanidins) are derived

from flavonoid monomers (Cowan, 1999). Generally, tannins may be formed

by condensation of flavan derivatives which have been transported to woody

tissues of plant, or alternatively, by polymerization of quinone units (Geissman,

1963).

Tannins have been shown to act at the molecular levels by complexing with

microbial proteins through so-called non-specific forces such as hydrogen

bonding and hydrophobic effects, as well as covalent bond formation (Haslam,

1996; Stern et al, 1996). Thus, their mode of antimicrobial action may be

related to their ability to inactivate microbial adhesins, enzymes, cell envelope

xxxvii

transport proteins, etc. They may also complex with polysaccharide (Ya et al,

1988). Tannins have also shown to act via direct inactivation of microorganisms

(eg. low tannin concentrations modify the morphology of germ tubes of

Crinipellis perniciosa (Brownlee et al, 1990).

According to various documented studies reviewed by Scalbert (1991), tannins

were found to be toxic to filamentous fungi, yeasts and bacteria. Condensed

tannins have been described to bind cell walls of ruminal bacteria, preventing

growth and protease activity (Jones et al, 1994). Though still speculative,

tannins are considered to be wholly or partially responsible for the antibiotic

activity of various aqueous and solvent extracts of many tropical and

temperate plants scattered across the globe (Taylor et al, 1996). Pentagalloyl

glucose (XI; hydrolysable tannin) and procyanidine (XII; condensed tannin)

have shown remarkable antimicrobial activities (Cowan, 1999).

XI XII

1.3.2.5 Miscellaneous Plant Constituents

xxxviii

Other major groups of antimicrobial compounds from plants include the simple

phenols and phenolic acids, quinones, coumarins, lectins and polypeptides;

and mixtures of all these groups. There abound many documented plant

derivatives belonging to these chemical groups that have proven antimicrobial

activity.

The common herbs terragon and thyme both contain caffeic acid (XIII; a

phenylpropane-derived phenolic compound), which is effective against viruses,

bacteria and fungi (Wild, 1994; Brantner et al, 1996). Catechol (XIV) and

pyrogallol both are hydroxylated phenols, shown to be toxic to

microorganisms; while eugenol (XV; a phenolic compound possessing a C3 side

chain at lower level of oxidation but also classified as an essential oil) is

considered bacteriostatic against both fungi and bacteria (Duke, 1985). Gallic

acid (XVI) has also proven to be toxic to some microorganisms.

XIII XIV

XV XVI

xxxix

Quinones are aromatic rings with two ketone substitutions which are

ubiquitous in nature and are characteristically highly reactive. In addition to

providing a source of stable free radicals, they are known to complex

irreversibly with nucleophilic amino acids in proteins, often leading to

inactivation of the protein and loss of action (Stern et al, 1996). For these

reasons, the potential range of quinone antimicrobial effects is great. Kazmi et

al (1994) described an anthraquinone from Cassia italica, a Pakistani tree,

which was bacteriostatic for Bacillus anthracis, Corynebacterium

pseudodiphthericum and Pseudomonas aeruginosa but bactericidal for

Pseudomonas pseudomalliae. Also, Hypericin (XVII), an anthraquinone from St.

John’s wort (Hypericum perforatum), has received much attention in the

scientific journals lately as an antidepressant. This compound has, however,

been reported by Duke (1985) to possess general antimicrobial properties.

Cowan (1999) reported that rhein (XVIII) which is an anthraquinone compound

has broad antimicrobial effects.

XVII XVIII

xl

Coumarins (XIX) are phenolic substances made up of fused benzene and α-

pyrone rings. They are responsible for the characteristic odour of hay. As a

group, coumarins have been found to stimulate macrophages which could

have an indirect negative effect on infections (Casley-Smith, 1997).

Hydroxycinamic acids, related to coumarins, seem to be inhibitory to Gram-

positive bacteria (Fernandez et al, 1996). Also, phytoalexins, which are

hydroxylated derivatives of coumarins, are produced in carrots in response to

fungal infection and can be presumed to have antifungal activity (Hoult and

Paya, 1996). General antimicrobial activity was equally documented in

coumarin compounds found in woodruff (Galium odoratum) extracts

(Thompson, 1978). Although data about specific antibiotic properties of

coumarins are scarce, many reports give reasons to believe that some utility

may reside in these phytochemicals (Cowan, 1999; Hamburger and

Hostettmann, 1991).

XIX

xli

Peptides which are inhibitory to microorganisms were first reported by Balls et

al (1942). They are often positively charged and contain disulfide bonds. Their

mechanism of action may be the formation of ion channels in the microbial

membrane, or by competitive inhibition of adhesion of microbial proteins to

host polysaccharide receptors (Zhang and Lewis, 1997; Sharon and Ofek, 1986).

Inhibition of bacteria and fungi by these macromolecules (e.g. peptides from

the herbaceous Amaranthus) has been documented (De Bolle et al, 1996).

Also, thionins, which are peptides commonly found in barley and wheat,

consisting of 47 amino acid residues are toxic to yeasts and both Gram-

negative and Gram-positive bacteria (Fernandes de Caleya et al, 1972).

Fabatin, a recently identified 47-residue peptide from fava beans, appears to

be structurally related to γ-thionins from grains and inhibits E. coli, P.

aeruginosa and Enterococcus hirae but not Candida or Saccharomyces (Zhang

and Lewis, 1997).

The antimicrobial activity of several extracts from plants has been linked to

compounds belonging to more than one chemical group. For instance, the

chewing sticks which are widely used in many African countries as an oral

hygiene aid come from different species of plants, and within one stick, the

chemically active component may be heterogeneous (Akpata and Akinrimisi,

1977). Crude extracts of one species used for this purpose, Serindeia

werneckei, inhibited the periodontal pathogens Porphyromonas gingivalis and

Bacteroides melaninogenicus in vitro (Rotimi et al, 1988). Also, the active

component of one of the Nigerian chewing sticks (Fagara zanthoxyloides) was

xlii

found to consist of various alkaloids (Odebiyi and Sofowora, 1979). Pawpaw

(Carica papaya) yields a milky sap, often called latex, which is a complex

mixture of chemicals (Cowan, 1999). Chief among them is papain, a well-

known proteolytic enzyme. It also contains carpaine (an alkaloid) and

terpenoids (Thomson, 1978). All these compounds in papaya have been shown

to contribute to the antimicrobial properties of its latex which was found to be

bacteriostatic to B. subtilis, Enterobacter cloacae, E. coli, Salmonella typhi,

Staphylococcus aureus and Proteus vulgaris (Osato et al, 1993).

1.4 LITERATURE REVIEWS OF PLANTS USED

1.4.1 Loranthus micranthus Linn

1.4.1.1 Taxonomy of L. Micranthus

The botanical profile of Loranthus micranthus is as summarized below:

Kingdom: Plantae

Phylum: Angiosperm

Sub-Phylum: Dicotyledons

Order: Santalales

Family: Loranthaceae

Sub-Family: Lorantheae

Genus: Loranthus

xliii

Species: micranthus

The mistletoe plant is an evergreen obligate parasite with over 700 species

which depends on its hosts for minerals and water only, as it can

photosynthesize its carbohydrate by means of its green leaves (Gill, 1973;

Griggs, 1991).

The most common species include: European mistletoe (Viscum album L.);

American mistletoe (Phoradendron flavescens); Australian/Argentine mistletoe

(Ligaria cuneifolia R et. T); African mistletoe, e.t.c.

Figure 1.0: Flowers and Leaves of Loranthus micranthus

xliv

Figure 2.0: Loranthus micranthus parasitic on a host tree

1.4.1.2 Description of the Family, Genus and Species of L. micranthus

The Loranthaceae family consists of parasites with green leaves found in both

tropical and temperate regions. They are mostly small semi-parasitic shrubs

attached to their hosts by suckers or haustoria (usually regarded as modified

adventurous roots). The family is fairly large with over 36 genera and 130

species, most of which are quite omnivorous in their choice of hosts, but a few

are restricted to one or two. Few members of Loranthaceae family root in the

earth (e.g. the Western Australia Christmas tree -Nuytsia floribunda, which

grows into a small tree of up to 10 metres high). For most others that root on

hosts, there is commonly an outgrowth, often of considerable size and

xlv

complicated in shape, where the parasite root joins the host. The roots of the

parasites often branch within the tissue of the host (as in Viscum).

The genus, Loranthus, consists of several species scattered in many parts of

Africa. It belongs to the sub-family, Lorantheae, which is characterized by the

presence of stem without secretory canals and has extraxylary phloem. Their

flowers have below the petals an outgrowth from the axis in form of small ring

or fringe called calyculus. After some weeks of the seeds germinating on

branches of its host, the Loranthus plant produces proper flowers which are

generally bright red and conspicuous, although one species produces yellow

flowers with red tips. The flowers are soon followed by small, fleshy,

drupaceous fruits which are much sought after by birds. The red Loranthus is a

common sight in many parts of West Africa, particularly in cocoa and cola

plantations, where whole branches are often covered with this medicinal herb.

The African mistletoe species, micranthus, is found mainly in the Southeastern

part of Nigeria. It grows on a large number of hosts including kola nut (Kola

acuminata), avocado (Persea americana), dogoyaro/neem (Azadirachta

indica), oil bean (Pentaclethra macrophylla), ogbono (Irvigia gabonensis),

lemons/citrus, etc. It produces sympodial, often dichasical, stem and the leaves

are usually evergreen and leathery. The cymose inflorescences are in spikes,

with the flowers on the internodes as well as on the nodes. Thus, they have

clusters of narrowly tubular flowers that are bright-red which appear as

clusters of coloured ‘matches’.

xlvi

1.4.1.3 Ethnomedicinal Uses and Pharmacological Studies on L. micranthus

Several ethnomedicinal usages have been attributed to Loranthus micranthus.

These include: blood pressure control (antihypertensive activity), anti-diabetic

activity, anticancer, antimicrobial and in many other metabolic diseases which

qualified mistletoe as an “all-purpose herb” (Kafaru, 1993; Obatomi et al, 1994;

Oliver-Bever, 1986; Dalziel, 1955). Different research works have been carried

out on the several species of the plant to demonstrate and support the

existence of many of the ethno medicinal claims (Obatomi et al, 1996;

Osadebe and Ukwueze, 2004; Osadebe and Akabogu, 2006; Osadebe et al,

2004, 2010, 2012; Ukwueze and Osadebe, 2012; Agbo et al, 2013; Omeje et al,

2012). Previous works on Loranthus micranthus have equally shown that some

of its medicinal activities vary with the particular host tree from which it is

harvested (Osadebe and Ukwueze, 2004; Osadebe et al, 2004). Other factors

that have been shown to affect the phytochemical composition and

pharmacological activities of mistletoe plant include species, harvesting

season, etc. (Obatomi et al, 1994; Wagner et al, 1996; Osadebe et al, 2008).

1.4.2 Psidium Guajava Linn

1.4.2.1 Taxonomy of P. guajava

The botanical profile of Psidium guajava is as summarized below:

xlvii

Kingdom: Plantae

Phylum: Angiosperms

Sub-Phylum: Eudicots

(unranked): Rosids

Order: Myrtales

Family: Myrtaceae

Subfamily: Myrtoideae

Tribe: Myrteae

Genus: Psidium

Species: guajava

Binomial name: Psidium guajava L.

Psidium guajava L, a fruit-bearing tree commonly known as guava, of the

family Myrtaceae, is a native of tropical America. The French call it goyave or

goyavier; the Dutch, guyaba or goeajaaba; the Surinamese, guave or goejaba;

and the Portuguese, goiaba or goaibeira. Hawaiians call it guava or kuawa. In

Guam, it is abas. In Malaya, it is generally known either as guava or jambu batu

(Morton, 1987).

1.4.2.2 Morphology of P. guajava

xlviii

Cultivated varieties grow about 10 m in height and produce fruits within 4

years. Wild trees grow up to 20 m high and are well branched. The guava tree

can be easily identified by its distinctive thin, smooth, copper-colored bark that

flakes off, showing a greenish layer beneath. The trees might have spread

widely throughout the tropics because they thrive in a variety of soils,

propagate easily and bear fruits quickly. The fruits are enjoyed by humans,

birds and monkeys, which disperse guava seeds and cause spontaneous dumps

of guava saplings to grow throughout the rainforest (Wealth of India, 2003).

Figure 3.0: Psidium guajava Tree

xlix

Figure 4.0: Leaves and Flowers of Psidium guajava

l

Figure 5.0: Fruit of Psidium guajava

1.4.2.3 Ethnomedicinal Uses and Pharmacological Studies of P. guajava

Psidium guajava is a medicinal plant used in tropical and subtropical countries

to treat many health disorders. In the indigenous system of medicine, different

parts of the plant are used for the treatment of various human ailments such as

wounds, ulcers, bowels and cholera (Begum et al., 2002a). Investigations have

indicated that its bark, fruit and leaves possess antibacterial, hypoglycaemic,

anti-inflammatory, analgesic, antipyretic, spasmolytic and CNS depressant

activities ( Begum et al., 2002b). It has indeed been variously reported that

Psidium guajava leaf extract has a wide spectrum of biological activities such as

anticough, antibacterial, haemostasis (Jaiarj et al., 1999; 2000), antidiarrhoeal

and narcotic properties (Lozoya et al.,1990), and antioxidant properties (Qian

and Nihorimbere, 2004). According to Lutterodt and Maleque (1998) and

Meckes et al., 1996, the leaf extract is used to treat diarrhoea, abdominal pain,

convulsions, epilepsy, cholera, insomnia and has hypnotic effect.

The long history of guava use has led modern-day researchers to intensify their

study on guava extracts. Its traditional use against diarrhea, gastroenteritis and

other digestive complaints has been validated in numerous clinical studies. In a

study including 17 Thai medicinal plants on anti-proliferative effects on human

li

mouth epidermal carcinoma and murine leukemia cells using MIT assay, guava

leaf showed anti-proliferative activity, which was 4.37 times more than

vincristine (Manosroi et al. , 2006).

Bark and leaf extracts were shown to have in vitro toxic action against

numerous bacteria. Gallocatechin isolated from the methanol extract of guava

leaf showed antimutagenic activity against E. coli (Matsuo et al., 1994). Water

and chloroform extracts of guava were effective in activating the mutagenicity

of Salmonella typhimurium (Grover and Bala, 1993). The antimicrobial activities

of P. guajava and leaf extracts, determined by disk diffusion method (zone of

inhibition), were compared to tea tree oil (TTO), doxycycline and clindamycin

antibiotics. It was shown that P. guajava leaf extracts might be beneficial in

treating acne especially those that have anti-inflammatory activities (Qadan et

al., 2005). The active flavonoid compound-quercetin-3-O-alpha-l-

arabinopyranoside (guaijaverin) - extracted from guava leaves has high

potential antiplaque activity by inhibiting the growth of Streptococcus mutans

(Limsong et al., 2004). Guava leaf extract also inhibited the growth of

Streptococcus aureus in a study carried out by disc diffusion method

(Abdelrahim et al., 2002). In several other studies, guava showed significant

antibacterial activity against common diarrhea-causing bacteria such as

Staphylococcus, Shigella, Salmonella, Bacillus, E. coli, Clostridium and

Pseudomonas. Indeed, the aqueous, alcohol and chloroform extracts have

been found to be effective against Aeromonas hydrophila, Shigella spp. and

Vibrio spp., Staphylococcus aureus, Sarcinta lutea and Microbacterium phlei

(Jaiarj et al., 1999). In a more recent study, the aqueous and ethanol:water

extracts of P. guajava leaves, roots and stem bark were found to be active

against the Gram-positive bacteria Staphylococcus aureus and Bacillus subtilis,

lii

but virtually inactive against the Gram-negative bacteria Escherichia coli and

Pseudomonas aeruginosa (Sanches et al.,2005).

A double-blind clinical study of the effects of a Phytodrug (QG-5) developed

from guava leaf showed a decrease in duration of abdominal pain, which was

attributed to antispasmodic effect of quercetin present in leaf extract (Xavier

et al., 2002). Guava leaf extracts and fruit juices have also been clinically

studied for infantile diarrhea. In a clinical study with 62 infants with infantile

rotaviral enteritis, the recovery rate was 3 days (87.1%) in those treated with

guava, and diarrhea ceased in a shorter period than controls. It was concluded

in the study that guava has 'good curative effect on infantile rotaviral enteritis'

(Wei et al., 2000). Lectin chemicals in guava were shown to bind to E. coli (a

common diarrhea-causing organism), preventing its adhesion to the intestinal

wall and thus preventing infection and resulting diarrhea (Rodriguez et al.,

2001). Guava leaf extract has also shown to have tranquilizing effect on

intestinal smooth muscle, inhibit chemical processes found in diarrhea and aid

in the re-absorption of water in intestines. In another research, an alcoholic

leaf extract was reported to have a morphine-like effect, by inhibiting the

gastrointestinal release of chemicals in acute diarrheal disease. This morphine-

like effect was thought to be related to a chemical, quercetin. The effective use

of guava in diarrhea, dysentery and gastroenteritis can also be related to

guava's documented antibacterial properties (Tona et al., 2000). In a study

carried out with leaf extract of the plant, inhibition of gastrointestinal release

liii

of acetylcholine by quercetin present in extract was suggested as a possible

mode of action in the treatment of acute diarrheal disease (Lutterodt, 1992).

Guava fruit and leaf showed antioxidant and free radical scavenging capacity

(Hui-Yin and Gow-Chin, 2007). A study of aqueous extract of P. guajava in

acute experimental liver injury induced by carbon tetrachloride, paracetamol

and thioacetamide, showed its hepatoprotective activity. The effects observed

were compared with a known hepatoprotective agent, silymarin. Histological

examination of the liver tissues supported hepatoprotection (Roy et al., 2006).

During various episodes of screening of medicinal plants, extract from P.

guajava leaves was found to exhibit significantly inhibitory effect on the

protein tyrosine phosphatase1B (PTP1B). Significant blood glucose lowering

effects of the extract were observed after intraperitoneal injection of the

extract at a dose of 10mg/kg in both 1-and 3-month-old Lepr(db)/Lepr(db)

mice (Oh et al. , 2005). In a study undertaken to investigate the hypoglycemic

and hypotensive effects of P. guajava leaf aqueous extract in rats, it showed

hypoglycemic activity. The hypoglycemic effect of plant extract was examined

in normal and diabetic rats, using streptozotocin (STZ)-induced diabetes

mellitus model (Ojewole, 2005). Also, i.p. treatment with 1g/kg guava juice

produced a marked hypoglycemic action in normal and alloxan-treated

diabetic mice (Cheng and Yang, 1983). In two randomized human studies, the

consumption of guava fruit for 12 weeks was shown to reduce blood pressure

by an average 8%, decrease total cholesterol level by 9%, decrease

triglycerides by almost 8% and increase HDL cholesterol by 8%; while a

liv

randomized, single-blind, controlled trial conducted to examine the effects of

guava fruit intake on blood pressure and blood lipids in patients with essential

hypertension showed the possibility that an increased consumption of guava

fruit can cause a substantial reduction in blood pressure and blood lipids

without decreasing HDL-cholesterol level (Singh et al., 1992, 1993).

Leaf extract of guava had shown ionotropic effect on guinea pig atrium

(Conde-Garcia et al., 2003). Some studies reported that the leaf extract and its

derivative identified as quercetin has effect on the intracellular calcium levels

in gastrointestinal smooth muscle (Lozoya et al., 1990), in cardiac muscle cell

(Apisariyakul et al., 1999) and in neuromuscular junction (Chaichana and

Apisariyakul, 1996). In other animal studies, guava leaf extracts have shown

central nervous system (CNS) depressant activity (Shaheen, 2000). Guava leaf

extract showed anticough activity by reducing the frequency of cough induced

by capsaicin aerosol (Jaiarj et al., 1999).

1.4.2.4 The Phytochemistry of Psidium guajava

Guava has been found to be rich in tannins, phenols, triterpenes, flavonoids,

essential oils, saponins, carotenoids, lectins, vitamins, fibre and fatty acids.

According to Olajide et al (1999), the leaves of P. guajava contain an essential

oil rich in cineol, tannins, triterpenes and flavonoids. Various reports of

phytochemical screening of Psidium guajava leaf showed tannins in aqueous

extract; and anthocyans, alkaloids, flavonoids, tannins and steroids/terpenoids in

ethanolic extract.

lv

More than twenty identified compounds from Psidium guajava leaf have been

reported (Seshadri and Vasishta, 1965; Osman et al., 1974; Lutterodt and

Maleque, 1988). The major components are: β-selinene (XX), β-caryophyllene

(XXI), caryophyllene oxide (XXII), squalene (XXIII), selin-11-en-4α-ol

(XXIV), guaijavarin (XXV), isoquercetin (XXVI), hyperin (XXVII), quercitrin

(XXVIII) and quercetin-3-O-gentobioside; morin-3-O-α-L-lyxopyranoside

(XXIX), morin-3-O-α-L-arabopyranoside (XXX); β-sitosterol (XXXI), uvaol

(XXXII), oleanolic acid (XXXIII), ursolic acid (XXXIV) and one new

pentacyclic triterpenoid: guajanoic acid (Lozoya et al., 1994; Meckes et al.,

1996; Arima and Danno, 2002; Begum et al., 2004).

XX XXI

XXII XXIII

lvi

XXIV XXV

XXVI XXVII

XXVIII XXIX

XXX XXXI

lvii

XXXII XXXIII

XXXIV

Guava fruit is higher in vitamin C than citrus fruits (80 mg of vitamin C in 100g

of fruit) and contains appreciable amounts of Vitamin A as well and is also a

good source of pectin (Sunttornusk, 2002).

The bark of guava tree contains considerable amounts of tannins (11-27%), and

hence is used for tanning and dyeing purposes. Leucocyanidin (XXXV), luectic

acid, ellagic acid (XXXVI) and amritoside (XXXVII) have been isolated from the

stem bark.

Other compounds that have been isolated from guava plant include avicularin

(XXXVIII; 3-L-4-4-arabinofuranoside), α-pinene (XXXIX), β-pinene (XL), limonene

(XLI), terpenyl acetate (XLII), isopropyl alcohol (XLIII), longicyclene (XLIV), β-

lviii

bisabolene, β-copanene, farnesene, humulene, cardinene, curcumene, mallic

acids, ursolic, crategolic, guayavolic acids, cineol, etc (Shruthi et al., 2013).

XXXV XXXVI

XXXVII XXXVIII

XXXIX XL XLI

lix

XLII XLIII XLIV

1.5 BIOASSAYBIOASSAYBIOASSAYBIOASSAY----GUIDED CHARACTERIZATION OF ANTIGUIDED CHARACTERIZATION OF ANTIGUIDED CHARACTERIZATION OF ANTIGUIDED CHARACTERIZATION OF ANTIBACTERIALBACTERIALBACTERIALBACTERIAL CONSTITUENTS CONSTITUENTS CONSTITUENTS CONSTITUENTS

FROM HIGHER PLANTSFROM HIGHER PLANTSFROM HIGHER PLANTSFROM HIGHER PLANTS

The driving force behind much phytochemical research is the discovery of new

biological active compounds for medicinal or agricultural uses. Biological assays

then must be carried out in order to identify promising plant extracts, to guide

the separation and isolation, and to evaluate lead compounds. Identification of

natural products from plants that may serve as valuable sources of bioactive

agents for medicinal and agricultural uses largely depends on bioactivity-

directed isolation (Cseke et al, 2006).

The choices of bioassays depend a great deal on the amounts of materials to

be tested and the time and effort necessary to carry out the assays. Obviously,

an in vivo assay using the organism afflicted (humans or animals) would

provide the most meaningful results. However, exploratory screening using

whole animals is impractical (or unethical), and various in vitro screening

methods have been developed to provide guided separation and identification

of lead compounds. The latter have the advantage in that they can be

lx

automated with robotics and miniaturized, leading to rapid throughput

screening of large numbers of samples. In addition, the in vitro bioassays may

provide activity information that is precluded by poor bioavailability using a

whole-animal in vivo assay. For instance, natural products that inhibit the

growth of tumor cells or bacteria in an in vitro assay may identify promising

molecular structures that would benefit from semi-synthetic modifications

(Cseke et al, 2006).

1.5.1 Principles of Bioassay

Bioassay (or biological assay) is the estimation of the activity or potency of a

drug or other substance (e.g. plant extract) by comparing its effects on a test

organism with that of a standard preparation. It is a type of scientific

experiment conducted to measure the effects of a substance on a living

organism and is essential in the development of new drugs and other scientific

monitoring. Bioassays may be qualitative or quantitative.

1.5.2 Screening Methods for Antibacterial Agents from Higher Plants

The discovery of promising plant extracts and the subsequent activity-guided

isolation of constituents put specific requirements on the bioassays to be used

for that purpose. They have to be simple, rapid, reproducible and inexpensive

in order to be compatible with the large number of assays to be performed.

lxi

Antimicrobial activity of plants can be detected by observing the growth

response of various microorganisms to those plant tissues or extracts which

are placed in contact with them. Many methods for detecting such activity are

available, but since they are not equally sensitive or even based upon the same

principle, the results obtained will also be profoundly influenced not only by

the method selected, but also by the microorganisms used to carry out the test

(Vanden-Berghe and Vlietinck, 1991). In general, biological assays or evaluation

can be carried out much more efficiently on water-soluble, pure crystalline

substances than on mixtures like plant extracts.

1.5.2.1 Test Organisms and Culture Media

The purpose of any antimicrobial investigation will obviously determine to a

great extent the choice of test organisms to be used. For an investigation of a

general character, the test organisms selected should be as diverse as possible

and preferably representative of all important groups of pathogenic bacteria

according to their physical and chemical composition and resistance pattern.

Most screening studies on plant extracts, however, have been carried out on

one or two bacteria, including strains of Staphylococcus aureus and Escherichia

coli, although such findings may not adequately predict an interesting broad-

spectrum activity or a selective but pronounced activity against some of the

problem-pathogenic bacteria in chemotherapy such as resistant S. aureus, P.

aeruginosa, Proteus vulgaris, Klebsiella pneumoniae, Neisseria gonorrhoeae,

Candida albicans and others.

lxii

Most bacteria (and yeasts) can be cultivated on standard Mueller-Hinton agar

or diagnostic sensitivity test agar (DST) and American type culture collection

(ATCC) or similar standard microorganisms are available. Only few bacteria

(e.g. Neisseria gonorrhoeae and Campylobacter fetus) require special growth

factors which should be included in the standard medium.

In general, standard microorganisms should be preferably used as test bacteria

during screening for new antimicrobially-active plant components for ease of

reproducibility of results by other researchers. If the interest, however, is in

finding new products which are selectively active against problem

microorganisms causing certain diseases, e.g. resistant P. aeruginosa, it is

clearly appropriate to employ the corresponding isolated pathogenic

microorganisms (Rwangabo et al., 1988).

1.5.2.2 Antibacterial Testing

The currently available antimicrobial screening methods fall into three broad

groups, including diffusion, dilution and bioautographic methods (Rios et al,

1988). These testing methods will only give an idea of the presence or absence

of substances with antimicrobial activity in the plant extracts, as the potency of

the active ingredients can only be determined on pure compounds using

standardized methodologies. The results obtained using any of the methods,

lxiii

however, are influenced by such factors such as extraction method, inoculum

volume, culture medium composition, pH and incubation temperature.

In the diffusion technique, a reservoir (e.g. filter paper disc, porcelain/stainless

steel cylinder or hole punched in the media) containing the plant extract to be

tested is brought into contact with an inoculated medium (e.g. agar) and, after

incubation, the diameter of the clear zone around the reservoir (inhibition

zone diameter) is measured. In order to lower the detection limit using this

method, the inoculated system is kept at low a temperature during several

hours before incubation, which favors diffusion over microbial growth and thus

increases the inhibition diameter. In most studies, the inhibition zones

obtained are compared with those obtained for antibiotics so as to establish

the sensitivity of the test organism to the extract. Advantages of the diffusion

methods are the small size of the sample used in the screening and the

possibility of testing up to five or six compounds per plate against a single

microorganism.

For the dilution methods, samples being tested are mixed with a suitable

medium, which has previously been inoculated with the test organism. After

incubation, growth of the microorganism may be determined by direct visual

or turbidimetric comparison of the test culture with a control culture which did

not receive an addition of the sample being tested, or by plating out both test

and control cultures (Kavanagh, 1963). Usually a series of dilutions of the

original sample in the culture medium is made and then inoculated with the

test organism. After inoculation, the endpoint of the test (MIC-value) is taken

lxiv

as the highest dilution which will just prevent perceptible growth of the test

organism (Vanden-Berghe and Vlietinck, 1991). In comparison, several

different test microorganisms may be tested simultaneously on the same

dilution as against diffusion methods in which several substances or dilutions

of one substance may be tested simultaneously against one test

microorganism. The agar dilution method is thus very quick, time saving and

also very useful to guide the isolation of antimicrobially active components

from plant extracts (Bakana et al, 1987; Rwangabo et al, 1988).

Bioautographic methods are employed to localize antibacterial activity on a

chromatoGram. The procedures are based on the agar diffusion technique,

whereby the antimicrobial agent is transferred from the thin layer or paper

chromatoGram to an inoculated agar plate through a diffusion process. Zones

of inhibition are then visualized by appropriate vital stains. Although very

suitable for testing highly active antibiotics (MIC-values < 10ug/ml),

bioautographic methods might not be very promising for testing plant extracts,

which often contain much less potent antimicrobial agents than the currently

available antibiotics (Vanden-Berghe and Vlietinck, 1991).

1.6 STRUCTURE ELUCIDATION OF BIOACTIVE PLANT METABOLITES

Chemical compounds, usually derived from plants and other natural sources,

have been used by humans for thousands of years to alleviate pain, diarrhea,

infection, and various other maladies. Until recently, these ''remedies" were

lxv

primarily crude preparations of plant material of unknown constitution. The

revolution in the synthetic organic chemistry during the nineteenth century

produced a concerted effort towards identification of the structures of the

active constituents of these naturally derived medicinals and synthesis of what

were hoped to be more efficacious agents.

By determining the molecular structures of the active components of these

complex mixtures, it is hoped that a better understanding of how these

components work can be elucidated (Knittel and Zavod, 2008).

1.6.1 Preliminary Analysis

Bioassay-directed fractionation is the process of isolating pure active

constituents from some type of biomass (eg. plants, microbes, marine

invertebrates, etc.) using a decision tree that is dictated solely by bioactivity

(Kinghorn, 2008). A variety of chromatographic separation techniques are

available for these purposes, including those based on adsorption on sorbents,

such as silica gel, alumina, Sephadex, and more specialized solid phases, and

methods involving partition chromatography inclusive of counter-current

chromatography. Recent improvements have been made in column

technology, automation of high-performance liquid chromatography (HPLC; a

technique often used for final compound purification) and compatibility with

HTS methodology (Butler, 2004).

1.6.2 Application of Modern Analytical Techniques

lxvi

Routine structure elucidation is performed using combinations of

spectroscopic procedures, with particular emphasis on one and two-

dimensional 1H- and

13C-nuclear magnetic resonance (NMR) spectroscopy and

mass spectroscopy (MS). 2D NMR spectra, generally, provide more information

about a molecule than 1D NMR spectra and are especially useful in

determining the structure of a molecule, particular for molecules that are too

complicated to work with using 1D NMR. Thus, for the structural elucidation of

a compound, a simple 1D NMR (H1 and C

13) may not be sufficient. Advanced

techniques like correlation spectroscopic methods, when analyzed properly,

provide the perfect way to determine the structure of a given formula or an

entirely unknown compound. The combination of some or all of these 1D and

multi-dimensional NMR spectra are all very useful in identification and

characterization of the structure, and orientation of bonds in a molecule

(Becker, 2000).

Considerable progress has been made in the development of cryogenic and

microcoil NMR probe technology for the determination of structures in sub-

milligram amounts of natural products (Koehn and Carter, 2005). In addition,

the automated processing of spectroscopic data for the structure elucidation

of natural products is a practical proposition (Steinbeck, 2004).

Another significant advance is the use of "hyphenated" analytical techniques

for rapid determination of the structure of natural products without the need

for a separate isolation step, such as liquid chromatograph-nuclear magnetic

resonance (LC-NMR) and LC-NMR-MS (Koehn and Carter, 2005; Butler, 2004).

lxvii

1.7 STATEMENT OF PROBLEM

Antibiotics have remained the mainstay of clinical therapy of infectious

diseases worldwide. Many microorganisms, however, are becoming

increasingly resistant to most of these agents (Jawetz et al., 1989). Another

serious limitation of these chemotherapeutic agents is the numerous adverse

effects associated with their administration (Snavely and Hodges, 1984). These

limitations have, thus, made it imperative that extensive efforts must be made

to uncover new antimicrobial agents from alternative sources whose

structures and modes of action may very likely differ from those of microbial

sources (antibiotics). Compounds extracted from higher plants have the

potential of fulfilling this purpose and that has led to the increasing screening

studies on several plant extracts in search of new ‘leads’. Unfortunately, most

of these works are preliminary investigations of the pharmacological actions of

the crude plant extracts, rather than the isolation, identification or the

characterization of the active constituents of these extracts.

1.8 JUSTIFICATION OF THE STUDY

Several works have been carried out to verify and confirm the traditional

antibacterial uses of Psidium guajava and Loranthus micranthus growing in the

tropical rain forest region of Nigeria, but researches on the isolation,

characterization and detailed chemical investigations of their

pharmacologically active components have remained inexhaustive.

lxviii

There is, therefore, the need for comprehensive chemical and bio-molecular

studies on these active constituents, and hence the justifications for this

present research.

1.9 AIMS AND SCOPE OF THE WORK

Aims of the Study

• To carry out a detailed bioactivity-guided phytochemical evaluation of

the extracts/solvent fractions of the leaves of Psidium guajava and

Loranthus micranthus.

• To identify and isolate the antibacterial compounds from the leaves of

the plants.

• To characterize and elucidate the structure of the isolates.

• To compare the antibacterial potentials of these isolates with standard

antibiotics with a view to developing derivatives of the ‘lead’

compounds and optimize same for activity with respect to potency and

selectivity, especially against bacteria causing opportunistic AIDS

infections [ e.g. resistant strains of Pseudomonas aeruginosa, and

Staphylococcus aureus; including the multi-drug resistant (MDR) strains

of methicillin-resistant Staphylococcus aureus (MRSA), and

Mycobacterium avium complex (MAC)].

lxix

CHAPTER TWO

MATERIALS AND METHODS

2.1 MATERIALS

2.1.1 Plant Materials

The leaves of Loranthus micranthus L. were collected in mid-June at Nsukka,

South-Eastern Nigeria, from the stem of Persea americana and authenticated

by Mr. A. Ozioko, a taxonomist with the Bio-resources Development and

Conservation Project (BDCP), Nsukka, Nigeria. Voucher specimen was

deposited in the herbarium of the Faculty of Pharmaceutical Sciences,

University of Nigeria, Nsukka. The leaves of Psidium guajava L. were collected

at the same period from the bio-resource area of the University of Port

Harcourt in South-South region of Nigeria and authenticated by the Plant

Science Biology (PSB) Department of the University. The voucher specimen was

deposited in the herbarium of the Faculty of Pharmaceutical Sciences,

University of Port Harcourt.

The leaves were air-dried at room temperature to a constant weight,

pulverized and passed through a 1mm sieve. The powdered materials were

stored in air-tight containers and kept in a refrigerator.

2.1.2 Microorganisms Used

The studies were performed with standard cultures of Staphylococus aureus

(ATCC 25923), Pseudomonas aeruginosa (ATCC 27833) and Escherichia coli

lxx

(ATCC 35219) obtained from the Nigerian Institute of Medical Research

(NIMR),Yaba, Lagos, Nigeria. The clinical isolates of other microorganisms were

used. All the microorganisms were grown in nutrient broth (Biotec, Suffolk, UK)

at 37oC and maintained on nutrient agar (Biotec, Suffolk, UK) slants at 4

oC. The

standardized cultures of the organisms were used throughout the experiment.

2.1.3 Solvents and Reagents

N-Hexane (Sigma-Aldrich, South Africa), Chloroform (Sigma-Aldrich, South

Africa), Ethyl acetate (BDH, England), Methanol (Sigma-Aldrich, South

Africa), Acetone (BDH, England), DMSO (Sigma-Aldrich, USA), Acetic acid

(Hopkins and Williams, England). Cutter mill (Manesty, England), Electronic

Balance (Sartorius, Germany) and Weighing balance (Ohans, USA). Other

reagents used were freshly prepared in the laboratory according to official

specifications.

Solvents Used for Isolation and Characterization of Active Ingredients

Dichloromethane (DCM), ethyl acetate, hexane and methanol. These solvents

were purchased from the Institute of Chemistry, University of Duesseldorf,

Germany. They were distilled before use and special grades were used for

spectroscopic measurements.

Solvents for HPLC

Methanol (LiChroSolv HPLC; Merck) and nano-pure water (distilled and heavy

metals free water obtained by passing distilled water through nano- and

ionexchange filter cells; Barnstead, France).

Solvents for NMR

lxxi

Deuterated methanol (Uvasol, Merck) and DMSO-d6 were used for NMR

measurements.

2.1.4 Materials and General Instruments

Chromatography

Pre-coated TLC plates (Silica Gel 60 F254, layer thickness 0.2mm) Merck

Silica Gel 60, 40−63 μm mesh size Merck

Sephadex LH 20, 25−100 μm mesh size Merck

General instruments

Analytical balances MC-1 Sartorious

Half-micro and analytical balance MC-1 Sartorious

Glass ware Schott Duran

Drying Oven ET6130 Heraeus

Ultra sonicator RK 510H Bandelin

UV-Lamp (254 and 366 nm) Camag

Rotary evaporator Büchi Rotavapor R-200

Vacuum pump CVC 2000 Vacuubrand

Centrifuge Pico Heraeus

lxxii

Nitrogen generator UHPN 3001 Nitrox

Air generator ZA 20 WGA

Fraction collector Retriever II ISCO

Lyvac GT2 (Freeze dryer) Steris

Vacuum pump Trivag D10E (Freeze dryer) Leybol

Syringe Hamilton 1701 RSN

Magnetic stirrer Variomag

HP Behrotest PH 10-Set Multipoint

Semi-preparative HPLC

Pump: L-7100 Merck/Hitachi

Detector: UV-L7400 (Photodiode array detector) Merck/Hitachi

Printer: Chromato-Intergartor D-2000

Merck/Hitachi

Column: Eurospher 100-C18, [10 μm; 300 mm × 8 mm] Knauer

Pre-column: Eurospher 100-C18, [10 μm; 30 mm × 8 mm] Knauer

Analytical HPLC

Pump: P 580A LPG Dionex

lxxiii

Autosampler: ASI-100T (injection volume = 20 μL) Dionex

Detector: UVD 340S (Photodiode array detector) Dionex

Column oven: STH 585 Dionex

Column: Eurospher 100-C18, [5 μm; 125 mm × 4 mm] Knauer

Pre-column: Vertex column, Eurospher 100-5 C18 [5-4 mm] Knauer

Software: Chromeleon (V. 6.30)

HPLC-MS

Analytical HPLC: Agilent 1100 series (Photodiode array detector) Agilent

MS: Finigan LCQ-DECA Thermoquest

Ionizer: ESI and APCI Thermoquest

Vacuum pump: Edwards 30 BOC

Column: Eurospher 100-C18, [5 μm; 227 mm × 2 mm] Knauer

Pre-column: Vertex column, Eurospher 100-5 C18 [5−4 mm] Knauer

NMR

DRX-500 Bruker

lxxiv

2.2 METHODS

2.2.1 Extraction and Fractionation of Plant of Materials

A portion of the powdered material (800 g) from L. micranthus plant was

defatted with n-hexane (4.5 L; to yield HFM) and the dried marc (540 g)

extracted in a soxhlet extractor with absolute methanol (3.5L). The dried

methanol soluble extract (MFM; 60.0 g) was reconstituted in 30 mL of

methanol, made up to 250 mL with distilled water, shaken for about 30

minutes and subjected to successive liquid-liquid extraction with chloroform (3

X 750 ml), ethyl acetate (3 X 750 ml) and acetone (3 X 750 ml) to yield the

chloroform (CFM; 11.4 g), ethyl acetate (EFM; 21.3 g) and acetone (AFM; 17.4

g) soluble fractions respectively. An aqueous portion (8.7 g) was left behind

after the fractionation process. The fractions were concentrated by

evaporating over water-baths set at 40oC. The concentrated/dried fractions

were stored in a refrigerator for further analysis.

The pulverized air-dried leaves (600 g) from Psidium guajava were defatted

with n-hexane (2.5L) and the dried marc (450 g) extracted with 5 L of 90 %

methanol for 4 days at room temperature (250C) and the extract concentrated

in vacuo with rotary evaporator. The dried methanol extract (35 g, 7.7 % w/w)

was reconstituted in 20 mL of methanol, made up to 200 mL with distilled

water, shaken for about 30 minutes and subjected to successive liquid-liquid

extraction with chloroform (3 x 750 ml), ethyl acetate (3 x 750 ml) and acetone

(3 x 750 ml) to yield PsG-CF (6.0 g; 1.3% w/w), PsG-EF (10.7g; 2.4% w/w) and

lxxv

PsG-AF (8.6 g; 1.9 % w/w) fractions respectively. An aqueous portion (6.8 g)

was left behind after the fractionation process.

Another portion (300 mg of the crude powder from L. micranthus) was

macerated in 90% methanol (3.5 L) for 48 hours and the filtrate concentrated

in vacuo to yield the crude methanol extract, CMFM.

Similarly, 100 mg of the crude powder from P. guajava was macerated in 90 %

methanol (1.5 L) for 48 hours and the filtrate concentrated in vacuo to yield

the crude methanol extract, PsG-CMF.

The schematic representaions for the extraction and fractionation procedures

for the plants are presented in Figures 6 & 7.

lxxvi

Dried Marc; 540g

Methanol; 5.5L

Hexane Fraction

(HFM); 44.2g

Chloroform; 3 X 750 ml

Methanol Fraction

(MFM); 60.0g

N-Hexane; 4.5L

Dried Marc

Acetone Fraction

(AFM); 17.4g

Ethyl acetate Fraction

(EFM); 21.3g

Chloroform Fraction

(CFM); 11.4g

Acetone; 3 X 750 ml

Ethyl acetate; 3 X 750 ml

Methanol;

3.5L

Crude Methanol

Fraction (CMFM);

31.4g 800g

PLANT MATERIAL

300g

Fig. 6: Schematic Diagram of the Extraction/Fractionation Procedure for L. micranthus Leaves.

Aqueous Residue

(8.7g)

lxxvii

Plant Material (100g)

Dried Marc; 450g

Methanol; 5L

Hexane Fraction (PsG-

HF); 32.6g

Chloroform; 3 X 750 ml

Methanol Fraction

(PsG-MF); 35g

N-Hexane; 2.5L

Dried Marc

Acetone Fraction

(PsG-AF); 8.9g

Ethyl acetate Fraction

(PsG-EF); 10.9g

Chloroform Fraction

(PsG-CF); 6.2g

Acetone; 3 X 750 ml

Ethyl acetate; 3 X 750 ml

Methanol;

1.5L

Crude Methanol

Fraction (PsG-

CMF); 14.4g 600g

Fig. 7: Schematic Diagram of the Extraction/Fractionation Procedure for P. guajava Leaves.

Aqueous Residue

(6.8g)

lxxviii

2.2.2 Isolation and Purification of the Active Constituents from PsG-EF

Thin layer and column chromatography

TLC was performed on pre-coated TLC plates with Silica gel 60 F254 (layer

thickness 0.2 mm, E. Merck, Darmstadt, Germany) with either CH2Cl2: MeOH

(9:1) for semi-polar compounds or n-Hexane: EtOAc (8:2) for non-polar

compounds as mobile phase. The compounds were detected by their UV

absorbtion at 254 and 366 nm or by spraying the TLC plates with anisaldehyde

reagent followed by heating at 1100C.

Column chromatography was carried out using Sephadex® LH-20. The ethyl

acetate fraction (PsG-EF) was separated on a sephadex LH-20 column (3 X 60

cm) eluted with Dichloromethane: MeOH (1:1) to afford 10 pooled fractions

PsG-EF1 to PsG-EF 10. Briefly, 200 g of Sephadex LH-20 was sonicated with 300

mL of DCM: MeOH (1:1) for about 30 min and the gel gradually transferred into

column (3 X 60 cm) with the tab open. Solvent was allowed to drain until the

gel was firmly formed. The sample to be separated was dispersed in 2 mL of

DCM: MeOH (1:1) sonicated for 10 min and then centrifuged. The supernatant

was transferred into the column and allowed to permeate the gel, when fresh

DCM: MeOH (1:1) was added and the whole set up connected to Fraction

collector (Retriever II ISCO, Germany) and adjusted to the flow rate of 0.2

mL/min.

lxxix

2.2.3 HPLC Analysis of Active Constituents from PsG-EF

Analytical high pressure liquid chromatography (HPLC)

The fractions (PsG-EF1 to PsG-EF 10) were subjected to analytical HPLC. The

solvent gradient used started with 10:90 (MeOH: nanopure water (adjusted to

pH 2 with phosphoric acid) increasing to 100 % MeOH in 45 minutes. The

compounds were detected by an UV-VIS Diode Array detector.

Semi-preparative high pressure liquid chromatography (HPLC)

Antibacterial bioactive fractions PsG-EF4 and PsG-EF7 were subjected to semi-

preparative HPLC purification to isolate the compounds PsG-EF4A, PsG-EF4B,

PsG-EF7A, PsG-EF7D and PsG-EF7E. Briefly, the separation column (125 x 21.4

mm, ID) was prefilled with Eurospher C-18 (Knauer, Berlin, Germany) or

Dynmax (250 x 21.4 mm, L.ID). The mobile phase used comprised a linear

gradient of nanopure water and methanol. 50 µL of approximately 40 mg/mL

solution of the substance was injected for each time. The flow rate was

stabilized at 5 mL/min, and the paper speed of the recorder was 5 mm/min.

The eluted peaks were collected respectively by manual work based on the

records of a UV-vis detector.

2.2.4 Electron Spray Ionization Mass Spectrometry (HPLC/ESI-MS) of Isolates

lxxx

HPLC/ESI-MS was carried out using a ThermoFinningan LCQ-Deca mass

spectrometer connected to an UV detector. The samples were each dissolved

in MeOH and injected to the HPLC/ESI-MS set up. A solution of the sample was

then sprayed at atmospheric pressure through a 2-5 kV potential. HPLC was

run on a Eurospher C-18 (6 x 2 mm, L. ID) reversed phase column. The mobile

phase was H2O containing 0.1% Formic acid (A), to which MeOH (B) or ACN (C)

was added by a linear gradient: initial, 0% of B; 45 min, 80% of B; 55 min, 100%

of B. The flow rate was at 400 μL/min and the absorbance detected at 254 nm.

ESI (electrospray ionization) was performed at a capillary temperature of 200

0C and drift voltage of 20eV. Since the molecular ion peak is the most abundant

ion in ESI spectra, it is also possible to perform MS/MS experiments.

Measurements were done at Institute of Pharmaceutical Biology and

Biotechnology, University of Heirich-Heine-Düsseldorf.

2.2.5 Nuclear magnetic resonance spectroscopy (NMR) of Isolates

The 1H NMR and

13C NMR spectra were recorded at 300

0K on DRX 500 or NMR

spectrometers. All 1D and 2D spectra were obtained using the standard Brüker

software. The sample was dissolved in a deuterated methanol (CD3OD), or

hexadeuterated dimethylsulfoxide (DMSO-d6) the choice of which is dependent

on the solubility of the sample. TMS was used as internal standard reference

signal. The observed chemical shifts (δ) were recorded in ppm and the coupling

constants (J) were recorded in Hz.

lxxxi

2.2.6 Preliminary Screening of Extracts and Fractions for Antimicrobial

Activity

Antibacterial Activity Testing

Each extract or fraction was screened for antibacterial activity using the

microorganisms provided. The inhibition zone diameters (IZDs) of the extracts

and reference antibiotics (Ceftriaxone - MAY & BAKER PLC, Nigeria) were

determined by agar-well diffusion method (Lovian, 1980). The standardized

broth culture (0.1ml) of the test microorganism was introduced into a sterile

petri-dish and 20ml of molten agar added. The content was mixed thoroughly

and allowed to solidify. Four 2-fold serial dilutions of each extract (including

the fractions/ sub-fractions) and reference antibiotics were obtained from

their stock solutions (20mg/ml in DMSO). Four quadrants were marked on

each petri-dish and a cup (6mm) was bored on each quadrant using a sterile

cork-borer. Two drops of each dilution were placed in each cup using Pasteur

pipettes, allowed to diffuse for about an hour and then incubated at 37oC. This

procedure was repeated for each of the microorganisms. The IZDs were

recorded after 24 hours of incubation.

2.2.7 Phytochemical Screening of Plant Extracts and Fractions

Phytochemical Tests

Phytochemical tests were carried out to detect the presence of steroids,

alkaloids, tannins, glycosides, reducing sugars, flavonoids and saponins. These

were carried out according to the procedures outlined by Harbourne (1998).

lxxxii

2.2.8 Antibacterial Screening of Isolates

Standard cultures of S. aureus and E. coli in Mueller-Hinton broth that had

been incubated for 24 hours were diluted 1000-fold with the same broth.

Aliquots of the dilution were mixed with 0.1 ml of the solutions of the isolated

compounds that have been dissolved or suspended in 10% aqueous dimethyl

sulfoxide (DMSO) in sterilized culture tubes. The mixtures were incubated for

24 hours at 37oC and the growth of each test bacterium determined by

turbidity. The minimum inhibitory concentration (MIC) of each compound

against a particular organism was expressed as the least concentration of that

compound which did not show turbidity. Ceftriaxone was used as the standard

antibacterial agent.

2.2.9 Statistical Analyses

Statistical Analysis

All the data obtained were analyzed by GraphPad Prism® (Model 5) using two-

way ANOVA and subjected to Bonferroni post-tests to compare replicate

means. The statistical results were presented as mean ± SEM. Differences

between means were considered significant at P<0.05.

lxxxiii

CHAPTER THREE

RESULTS

3.1 Extraction and Solvent Fractionation Result

The yield from the extraction and fractionation carried out on the leaves of

Loranthus micranthus using various solvents is presented in Table 2 below.

Table 2: Yield from Extracts/Fractions of the leaves of L. micranthus.

Extract/Fraction Weight of

Powder/Marc (g)

Yield (g) % Yield

HFM 800.0 44.2 5.5

MFM 540.0* 61.0 11.3

CFM 540.0* 11.4 2.1

EFM 540.0* 21.3 3.9

lxxxiv

AFM 540.0* 17.4 3.2

CMFM 300.0 31.4 10.5

Key: HFM = Hexane extract, MFM = Defatted Methanol extract, EFM= Ethyl acetate soluble

fraction, AFM = Acetone soluble fraction, CFM = Chloroform soluble fraction, CMFM

= Crude Methanolic Extract; * = Portion of marc obtained after defatting with

hexane.

lxxxv

The yield from the extraction and fractionation carried out on the leaves of

Psidium guajava using various solvents is presented in Table 3 below.

Table 3: Yield from Extracts/Fractions of the leaves of P. guajava.

Extract/Fraction Weight of

Powder/Marc

(g)

Yield (g) % Yield

PsG-HF 600.0 32.6 5.3

PsG-MF 450.0* 35.7 7.9

PSG-CF 450.0* 6.2 1.3

PsG-EF 450.0* 10.9 2.4

PsG-AF 450.0* 8.9 2.0

PsG-CMF 100.0 14.8 14.8

Key: PsG-HF = Hexane extract, PsG-MF = Methanol soluble fraction, PsG-EF=Ethyl acetate soluble

fraction, PsG-AF = Acetone soluble fraction, PsG-CF = Chloroform soluble fraction, PsG-CMF =

Crude Methanolic Extract; * = Portion of marc obtained after defatting with hexane.

lxxxvi

3.2 Preliminary Phytochemical and Antibacterial Screening Results

The results of the preliminary investigations carried out on the leaf

extracts/fractions of Loranthus micranthus and Psidium guajava are presented

below.

The result of the comparative study on the phytochemical and anti-microbial

properties of leaves of Loranthus micanthus harvested from different host

trees is shown in Tables 4 & 5.

Table 4: Results of Phytochemical Tests on the Leaf extract of Loranthus

micranthus parasitic on different host trees.

HOST TREE Steriods Alkaloids Glycosides Reducing sugars Flavonoids Saponins Tannins

K. acuminata + ++++ + + ++ + ++

P. americana + ++++ ++ + + + ++

B. nitida - + + + + + +

P.macrophylla - + + + + + +

I. gabonensis - ++ + + + + ++

A. indica + + ‽ + + - +

- = Absent; + = Present in small quantity; ++ = moderately present; +++ =

Present in large quantity; ‽ = not detected.

lxxxvii

Table 5: Results of the anti-microbial screening of extracts of mistletoe from

six different host plants.

Each value represents the mean ± SEM;*: P <.05, **: P < 0.01, ***: P < 0.001 significantly lower

when compared with control 1; blank spaces indicate no observable inhibition; IG = I. gabonensis;

PM = P. macrophylla; KA = K. acuminata; AI = A. indica; PA = P. americana; BN = B. nitida; AMX =

amoxicillin; KTZ = ketoconazole

Host tree Minimum Inhibitory Concentrations (MIC, mg/ml) ± SEM

S. aureus B. subtilis P. aeruginosa S. typhi A. niger C. albicans

IG 4.45±0.20* 4.19±0.30 6.08 ± 2.34 5.55± 0.55 - -

PM 5.76 ± 1.27 4.13 ± 0.59 7.97±1.81 - - -

KA 4.01 ± 0.17* 3.99 ± 0.30** 5.16± 2.03 6.48±.0.68 2.56± 0.14 7.98±1.86

AI 6.08 ± 0.23 4.53 ± 0.19 7.41±1.21 - - -

PA 3.76±0.25* 3.53±0.84** 4.53 ± 2.34*** 4.94± 0.2 4.63±1.0 6.05±0.32

BN 7.28±0.50 7.70±1.38 10.32±0.32 - - -

AMX 4.95±0.64 4.19±0.04 7.05±0.74 2.43± 0.49 - -

KTZ - - - - 0.44±0.07 2.26± 0.05

lxxxviii

The result of the study on the seasonal variations of the phytochemical and

anti-microbial constituents of L. miranthus is presented in Tables 6 & 7.

Table 6: Results of Phytochemical Tests on the Leaf extract of Loranthus

micranthus harvested at different seasons

MONTHS Tannins Flavonoids Alkaloids Reducing

sugars

Terpenoids Saponins Glycosides

Jan + + - + + + +

April ++ ++ ++ + + + +

July ++ +++ ++ + + + +

Nov + ++ - + + + +

Key: - = Absent; + = Present in small quantity; ++ = Moderately present; +++ = present in large

quantity.

lxxxix

Table 7: Results of the anti-microbial screening of leaf extracts of Loranthus

harvested at different seasons.

Months Inhibition Zone Diameter (IZD, mm) ± SEM

S. aureus B. subtilis P. aeruginosa S. kapemba E. coli

Jan. 14.7±0.0.67 13.3±0.33*** 16.0±0.58 10.7±0.67*** 18.3±0.88

April 16.7±1.67 17.0±0.58 17.3±0.67 20.7±0.67 16.0±0.58*

July 16.7±0.67 18.0±0.00 16.0± 0.58 19.7±.0.33 19.0± 0.58

Nov. 14.7±0.33 16.3±0.33* 16.0±1.0 18.7±0.33* 14.7±0.33**

Each value represents the mean ± SEM,*: P < 0.05, **: P < 0.01, ***: P < 0.001 significantly lower

when compared with values obtained at the other months.

xc

The result of the preliminary study to verify the folkloric utilization of the

leaves of the African mistletoe (L. micranthus) as antifungal agent is presented

in Table 8.

Table 8: Result of MIC of L. micranthus against some fungi

Each value represents the mean ± SEM; n = 3.

Extract MIC (mg/ml)

Candida Aspergillus

K. acuminata 7.73 ± 0.59 4.98 ± 2.02

P. americana 5.85 ± 0.26 4.07 ± 1.17

I. gabonensis 9.73 ± 2.31 8.14 ± 1.96

Ketoconazole (control) 1.09 ± 0.13 0.22 ± 0.11

xci

The results of the phytochemical and antimicrobial investigations on the

fractions from Loranthus micranthus are presented in Tables 9 & 10 below.

Table 9: Results of Phytochemical Tests on the solvent fractions of Loranthus

micranthus leaves harvested from Persea Americana

FRACTIONS Tannins Flavonoids Alkaloids Reducing

sugars

Terpenoids Saponins Glycosides

CMFM + ++ ++ + + + +

HFM + + - - + - -

MFM + + + + + + +

EFM + + - + - - +

AFM + ++ + + + + +

CFM + + - - + - -

Key: - = Absent; + = Present in small quantity; ++ = Moderately present; +++ = Present in large

quantity; CMFM = Crude Methanolic Extract, HFM = Hexane extract, MFM = Defatted

Methanol extract, EFM= Ethyl acetate soluble fraction, AFM = Acetone soluble fraction,

CFM = Chloroform soluble fraction.

xcii

xciii

Table 10: Result of Mean IZD (mm) +/- SEM for L. micranthus

Extracts/Fraction

FRACTIONS

{20mg/ml} S. aureus B. subtilis P. aeruginosa E. coli

CMFM 16.00 ± 0.58*** 18.00 ± 0.58 18.00 ± 0.58*** 10.00 ± 1.16***

HFM 1.98 ± 0.88*** 3.82 ± 1.20*** 2.10 ± 0.33*** 0.00 ± 0.00***

MFM 9.67 ± 1.20*** 13.00 ± 2.31*** 10.00 ± 2.08*** 5.67 ± 1.20***

EFM 4.67 ± 1.20*** 6.67 ± 1.20*** 6.67 ± 0.33*** 6.33 ± 0.88***

AFM 7.33 ± 1.20*** 9.67 ± 1.20*** 7.67 ± 0.88*** 5.00 ± 1.15***

CFM 2.67 ± 0.33*** 5.67 ± 0.67*** 3.00 ± 0.58*** 1.33 ± 0.33***

Control (10mg/ml)

26.00 ± 0.58 20.00 ± 0.58 33.67 ± 2.03 27.00 ± 1.15

CMFM = Crude Methanolic Extract, HFM = Hexane extract, MFM = Defatted Methanol

extract, EFM= Ethyl acetate soluble fraction, AFM = Acetone soluble fraction,

CFM = Chloroform soluble fraction, Control = Ceftriaxone; * P<0.05; ** P<0.01 and

*** P<0.001 significantly lower when compared with Control

xciv

Fig. 8.0: Graph of Mean IZD (mm) ± SEM of the extracts/fractions of L.

micranthus leaves against bacteria.

xcv

The results of the phytochemical and antimicrobial investigations on the leaves

of Psidium guajava harvested at differents seasons prevalent in Nigeria are

presented in Tables 11 & 12 below.

Table 11: Results of Phytochemical Tests on the Leaf extract of Psidium

guajava harvested at different seasons

MONTHS Tannins Flavonoids Alkaloids Reducing

sugars

Terpenoids Saponins Glycosides

Jan ++ - + + + - +

April ++ + - ++ + + +

July ++ ++ - ++ + + +

Nov ++ + + ++ + - +

- = Absent; + = Present in small quantity; ++ = Moderately present; +++ = Present in large quantity

xcvi

Table 12: Results of the anti-microbial screening of leaf extracts of Psidium

guajava harvested at different seasons

Months Inhibition Zone Diameter (IZD, mm) ± SEM

S. aureus B. subtilis P. aeruginosa S. kapemba E. coli

Jan. 31.0 ± 1.40 19.3 ± 0.47* 16.0 ± 1.40*** 26.0 ± 1.41 20.33 ± 0.40

April 31.0 ± 1.0 20.3 ± 0.44 20.0 ± 0.00 29.5 ± 0.45 22.3 ± 0.45

July 25.7 ± 0.24* 24.0 ± 1.41 18.7 ± 0.40* 15.3 ± 0.04*** 18.7 ± 0.59*

Nov. 19.3 ± 0.45*** 20.3 ± 0.04 24.7 ± 0.45 18.7 ± 0.04** 21.7 ± 0.47

Each value represents the mean ± SEM,*: P < 0.05, **: P < 0.01, ***: P < 0.001 significantly lower

when compared with values obtained at the other months

xcvii

The results of the phytochemical and antimicrobial investigations on the

fractions from Psidium guajava leaf are presented in Tables 13 & 14 below.

Table 13: Results of Phytochemical Tests on the solvent fractions of Psidium

guajava leaves

FRACTIONS

Tannins Flavonoids Alkaloids Reducing sugars

Terpenoids Saponins Glycosides

PSG-CMF ++ ++ + ++ + + ++

PSG-MF ++ ++ + + + + ++

PSG-EF ++ ++ - + - + ++

PSG-AF ++ + - + - + +

PSG-CF + - - + + + +

Key: PSG-CMF = Crude Methanolic Extract, PSG-MF=Methanol soluble fraction, PSG-EF=Ethyl

acetate soluble fraction, PSG-AF = Acetone soluble fraction, PSG-CF = Chloroform soluble fraction;-

= Absent; + = Present in small quantity; ++ = Moderately present; +++ = Present in large quantity.

xcviii

Table 14: Table of Mean IZD (mm) +/- SEM for P. guajava Extracts/Fractions

Fractions

(10mg/ml)

S. aureus B. subtilis P. aeruginosa E. coli

PSG-CMF 23.67 +/- 0.88 17.67 +/- 0.88 14.33 +/- 0.88*** 6.67 +/- 0.67***

PSG-MF 16.67 +/- 0.72*** 14.67 +/- 0.54*** 12.33 +/- 0.62*** 10.00 +/- 0.94***

PSG-EF 25.00 +/- 1.25 19.00 +/- 0.82 15.00 +/- 1.25*** 12.00 +/- 0.94***

PSG-AF 21.00 +/- 0.94** 17.33 +/- 0.27 14.33 +/- 0.72*** 9.67 +/- 0.54***

PSG-CF 9.67 +/- 0.54*** 7.00 +/- 0.94*** 5.67 +/- 0.72*** 1.67 +/- 0.27***

Control (10mg/ml)

26.00 +/- 0.58 20.00 +/- 0.58 33.67 +/- 2.03 27.00 +/- 1.15

Key: PSG-CMF = Crude Methanolic Extract, PSG-MF=Methanol soluble fraction, PSG-EF=Ethyl

acetate soluble fraction, PSG-AF = Acetone soluble fraction, PSG-CF = Chloroform soluble

fraction; Control = Ceftriaxone; * P<0.05; ** P<0.01 & *** P<0.001 significantly lower when

compared with Control.

xcix

3.3 Isolation of Bioactive Constituents

The ethyl acetate fraction (PsG-EF) from P. guajava that gave the best

antibacterial bioassay result was subjected to further Sephadex-LH 20

chromatographic fractionation and purifications to afford ten pooled fractions

(PsG-EF1 to PsG-EF10). Fractions PsG-EF4, PsG-EF5 and PsG-EF7 that had good

antibacterial potential were subjected to semi-preparative reverse phase HPLC

purification to isolate the phenolic compounds I - V.

The structures of these compounds were elucidated using a combination of

analytical and spectral techniques which included: UV, 1H-NMR,

13C-NMR,

DEPT, 1H

1HCOSY, HMQC, HMBC and ESI-MS analyses (see Appendices for

spectra details).

A summary of the spectral data and the elucidated structures of the isolates

are presented in pages 74- 82 while the detailed interpretation of analytical

data leading to the predicted structures is presented in chapter 4 under

“Discussion”.

c

3.4 Structure Elucidation of Isolated Compounds

3.4.1 PsG-EF4A (Compound I: Yellow semi-solid; 3.0mg)

1H-NMR (500 MHz, MeOD) δ = 7.70 (m, 2H), 7.52 (t, J=7.4, 1H), 7.41 (t, J=7.7, 2H), 6.21 (d, J=2.1, 1H),

6.07 (d, J=2.0, 1H), 4.81 (d, J=7.7, 1H), 3.85 (m, 1H), 3.66 (dd, J=5.7, 12.1, 1H), 3.22 (m, 1H), 2.83 (m,

1H).

1H-NMR (600 MHz, MeOD) δ = 7.70 (m, 2H), 7.52 (m, 1H), 7.42 (t, J=7.8, 2H), 6.22 (d, J=2.1, 1H), 6.07

(d, J=2.1, 1H), 4.81 (d, J=7.8, 3H), 3.85 (dd, J=2.2, 12.1, 1H), 3.66 (dd, J=5.6, 12.1, 1H), 3.22 (dd, J=6.6,

11.1, 1H), 2.84 (dd, J=7.8, 9.1, 1H).

EI.MS m/z: 392.8 [M+1], 391.3 [M-1], 231.1 [M+1 - 162]

UV λmax (MeOH) nm (Ɛ): 254; 296.

O O

HO OH

O

OH

OHHO

OH

[Molecular Formula: C19H20O9; Formula Mass = 392.4]

ci

Fig. 9a: Structure of Compound I

cii

Table 15: 1H and

13C-NMR data of compound I

Position δδδδH δδδδC HMBC

1 - 107.7

2 - 161.0

3 6.07 d (2.1) 96.6 1, 2, 4, 5

4 - 162.0

5 6.22 d (2.1) 94.2 1, 3, 4, 6

6 - 156.9

7 - 194.8

8 - 138.7

9/13 7.7 d 128.9 7, 11, 10, 12

10/12 7.42 t (7.8) 128.2 8, 9, 13

11 7.52 t 132.4 9, 13

1' 4.81 d (7.8) 100.6 6

2' 2.83 dd 73.1 1', 3'

3' 3.32 # 76.5 2', 4'

4' 3.22 t 69.4 6'

5' 3.34 # 77.1 4'

6' 3.66 dd Ha

3.86 dd Hb

60.6 5'

4'

NMR was measured at 500 MHz (1H) and 150 MHz (13C) (CD3OD).

# = overlapped with the broad solvent peak.

ciii

Fig. 9b: Numbering of Carbon Skeleton for Compound I [XXVII]

civ

3.4.2 PsG-EF4B (Compound II: Yellow semi-solid; 3.5mg)

1H NMR (500 MHz, MeOD) δ = 7.57 (m, 2H), 7.48 (t, J=7.4, 2H), 7.38 (dd, J=7.7, 15.3, 2H), 6.20 (s,

1H), 4.72 (d, J=7.7, 1H), 3.87 (dd, J=2.1, 12.0, 1H), 3.65 (dd, J=5.9, 12.0, 1H), 3.25 (d, J=9.1, 1H), 3.14

(t, J=9.4, 1H), 2.51 (dt, J=7.1, 14.3, 1H), 2.02 (s, 3H).

1H NMR (600 MHz, MeOD) δ = 7.58 (dd, J=3.4, 5.0, 2H), 7.49 (m, 1H), 7.39 (m, 3H), 6.20 (s, 1H), 4.73

(d, J=7.7, 1H), 3.87 (dd, J=2.2, 12.0, 1H), 3.66 (dd, J=6.0, 12.0, 1H), 3.28 (ddd, J=5.8, 10.9, 19.6, 2H),

3.16 (m, 2H), 2.52 (dt, J=8.2, 16.3, 1H), 2.02 (d, J=5.7, 3H).

EI.MS m/z: 406.8 [M+1], 405.3 [M-1], 245.2 [M+1 - 162]

UV λmax (MeOH) nm (Ɛ): 250; 302.

O O

HO OH

O

OH

OHHO

OH

[Molecular Formula: C20H22O9; Formula Mass- 406.38328]

Fig. 10a: Structure of Compound II

cv

cvi

Table 16: 1H and

13C-NMR data of compound II

Position δH δC HMBC

1 - 107.5

2 - 158.0

3 - 113.6

4 - 160.1

5 6.20 d (2.1) 94.2 1, 3, 4, 6

6 - 155.3

7 - 197.1

8 - 140.4

9/13 7.58 d (3.4; 5.0) 128.7 7, 11, 10, 12

10/12 7.39 m 127.6 8, 9, 13

11 7.49 m 131.5 9, 13

1' 4.73 d (7.7) 100.1 6

2' 2.52 dt 72.9 1', 3'

3' 3.28 # 73.4 2', 4'

4' 3.16 m 69.9 6'

5' 3.30 dd 77.0 4'

6'

3-CH3

3.66 dd Ha

3.87 dd Hb

2.02 s (5.7)

61.4

7.9

5'

4'

2, 3, 4

cvii

NMR was measured at 500 MHz (1H) and 150 MHz (

13C) (CD3OD).

# = overlapped with the broad solvent peak.

Fig. 10b: Numbering of Carbon Skeleton for Compound II

cviii

3.4.3 PsG-EF7A (Compound III: Brownish-yellow semi-solid; 2.5mg)

1H NMR (500 MHz, MeOD) δ = 7.50 (m, 2H), 7.41 (d, J=7.4, 1H), 7.27 (t, J=7.7, 2H), 7.10 (s, 1H), 6.15

(s, 1H), 4.76 (d, J=7.7, 1H), 4.55 (d, J=11.9, 1H), 4.34 (m, 1H), 3.50 (s, 2H), 2.50 (m, 1H), 2.02 (s, 3H).

1H NMR (600 MHz, MeOD) δ = 7.50 (d, J=7.3, 2H), 7.41 (q, J=7.4, 1H), 7.27 (t, J=7.7, 2H), 7.10 (s, 1H),

6.15 (s, 1H), 4.77 (d, J=7.7, 2H), 4.55 (dd, J=1.9, 11.9, 2H), 4.35 (dd, J=4.6, 12.0, 1H), 3.51 (m, 1H),

3.32 (dd, 1H), 3. 28 (dd, 1H), 2.51 (dd, J=9.1, 17.8, 1H), 2.02 (s, 3H).

EI.MS m/z: 545.0 [M+1], 543.3 [M-1]

UV λmax (MeOH) nm (Ɛ): 262; 362

[Molecular Formula: C26H24O13; Formula Mass- 544.46]

Fig. 11a: Structure of Compound III

cix

cx

Table 17: 1H and

13C-NMR data of compound III

Position δH δC HMBC

1 - 105.0

2 - 157.3

3 - 115.5

4 - 161.3

5 6.15 s 93.6 1, 3, 4, 6

6 - 156.4

7 - 177.7

8 - 138.5

9/13 7.50 d (7.3) 128.4 7, 11, 10, 12

10/12 7.27 t (7.7) 127.6 8, 9, 13

11 7.41 q (7.4) 134.2 9, 13

1' 4.77 d (7.7) 101.8 6

2' 2.51 dd 72.8 1', 3'

3' 3.28 dd 76.2 2', 4'

4' 3.32 dd 70.3 6'

5' 3.51 m 73.7 4'

6'

1''

2''

3''

4''

5''

4.35 Ha (dd)

4.55 Hb (dd)

-

-

-

-

-

69.0

148.4

145.6

120.7

145.2

165.8

5'

4'

cxi

6''

3-CH3

7.10 s

2.02 d (5.7)

108.6

8.0

4, 1'', 2'', 4'', 5''

2, 3, 4

NMR was measured at 600 MHz (1H) and 150 MHz (

13C) (CD3OD).

Fig. 11b: Numbering of Carbon Skeleton for Compound III

cxii

3.4.4 PsG-EF7D (Compound IV: Brownish-Yellow, needle-like solid; 4.5mg).

1H NMR (500 MHz, MeOD) δ = 7.52 (1H,s), 7.50 (1H,d,J=8.3), 6.9 (1H,d,J=8.4),

6.39 (1H,s), 6.20 (1H,s), 5.47 (1H,s), 4.85 (1H, d, J=7.3), 4.33 (1H, d, J=2.8), 3.90

(1H, d, J=3.1),3.87 (1H, m), 3.81 (1H, m), 3.50 (2H, t, J=3.6), 3.35 (1H, m).

EI.MS m/z: 435.0 [M+1], 433.1 [M-1], 303.1 [(M+1) - 132]

UV λmax (MeOH) nm (Ɛ): 257 (26, 595), 354 (23, 595).; m.p. 252-2630C.

PsG-EF7D was elucidated as the previously reported compound: Quercertin-3-

O-αL- arabinofuranoside with the chemical name: 2-(3,4-Dihydroxy phenyl)-

5,7-dihydroxy-4-oxo-4H-chromen-3-yl-α-L-arabinofuranoside. [Trivial name:

guaijaverin].

cxiii

Fig. 12: Structure of Compound IV

cxiv

3.4.5 PsG-EF7E (Compound V: Yellow semi-solid; 2.1mg)

1H NMR (500 MHz, MeOD) δ = 7.61 (d, J=8.4, 2H), 7.44 (s, 1H), 7.31 (t, J=7.8,

2H), 7.09 (s, 1H), 6.16 (s, 1H), 6.05 (s, 1H), 4.50 (s, 1H), 4.33 (s, 1H), 3.58 (s, 1H),

2.77 (s, 1H).

UV λλλλmax (MeOH) nm (ƐƐƐƐ): 265; 294.

O O

OH

O

OH

OHOH

O

O O

OH

OH

OH

[Molecular Formula: C25H22O13; Formula Mass- 530.43]

cxv

Fig. 13: Structure of Compound V

cxvi

Table 18: 1H and

13C-NMR data of compound V

Position δH δC HMBC

1 - 105.0

2 - 157.3

3 6.16 s 115.5

4 - 161.3

5 6.05 s 93.6 1, 3, 4, 6

6 - 156.4

7 - 177.7

8 - 138.5

9/13 7.61 d (8.4) 128.4 7, 11, 10, 12

10/12 7.31 t (7.8) 127.6 8, 9, 13

11 7.44 s 134.2 9, 13

1' 4.70 s 101.8 6

2' 2.77 s 72.8 1', 3'

3' 3.18 s 76.2 2', 4'

4' 3.42 s 70.3 6'

5' 3.58 s 73.7 4'

6'

1''

2''

3''

4''

5''

4.33 dd Ha

4.50 dd Hb

-

-

-

-

-

69.0

148.4

145.6

120.7

145.2

165.8

5'

4'

cxvii

6'' 7.09 s

108.6 4, 1'', 2'', 4'', 5''

NMR was measured at 500 MHz (1H) and 150 MHz (

13C) (CD3OD).

cxviii

3.5 Antibacterial Profile of Bioactive Constituents

Table 19: Antibacterial profile of the isolated compounds against

Staphylococcus aureus and E. coli.

Compound

MIC (µg/ml)

S. aureus E. coli

I 250 650

II 400 900

III 850 1950

IV 250 750

V 650 1200

Ceftriaxone 20 25

cxix

CHAPTER FOUR

DISCUSSION AND CONCLUSION

4.1 Discussion

The results of the preliminary investigations carried out on the leaf

extracts/fractions of Loranthus micranthus and Psidium gajava gave some

basic but revealing data that proved very vital for further assay on the plant

materials.

First, the comparative study of the phytochemical and anti-microbial

properties of leaves of Loranthus micanthus harvested from different host

trees showed significant variations in the parameters reviewed (Tables 4

& 5). The extracts from K. acuminata and P. americana exhibited appreciable

activities against some of the bacteria, but, showed only mild activity against

the fungi. The extract from P. americana showed significant activity (P < 0.05)

against some Gram-negative bacteria (P. aeruginosa) while maintaining

moderate activity against the Gram-positive bacteria. Also, alkaloids,

terpenoids and tannins were found to be relatively preponderant in the

extracts from P. americana and K. acuminata as against that from other hosts.

Thus, the study indicated that L. micranthus leaves to be used for the

treatment of non-specific infections should be preferentially sourced from

either P. americana or K. acuminata.

cxx

In the same vein, the study on the seasonal variations of the phytochemical

and anti-microbial constituents of the plants revealed some interesting

parameters. The investigation on leaves of L. micranthus (parasitic on P.

americana) harvested at different seasons of the year showed variations across

the seasons (Tables 6 & 7). Some phytoconstituents like alkaloids, terpenoids

and tannins were found to show significant variations across the seasons.

These constituents were found to be more abumdant in the extracts from the

leaves harvested in April and July. Higher antimicrobial activities were also

observed within these seasons. In effect, better antimicrobial activity was

exhibited by the extracts of the leaves harvested in April and July. This might

then suggest that Loranthus leaves intended for use in the treatment of non-

specific infections may be preferentially harvested either at the onset (April) or

the peak (July) of the rainy season. On the other hand, the seasonal variation

study data (Tables 11& 12) on the phytochemical and antimicrobial activities of

Psidium guajava were non-significant, even though the extracts harvested in

April (onset of rainy season) showed slightly better bioactivities than the other

seasons. This may be as a result of the relatively similar amount of the major

phytoconstituents like terpenoids, tannins, flavonoids, etc. found in the

extracts across the seasons.

The results (Table 8) of the study for determination of the folkloric utilization

of the leaves of the African mistletoe (L. micranthus) as an all-purpose

antimicrobial agent showed that it had negligible antifungal activity and thus

its paste should not be recommended for the treatment of wounds or any

cxxi

other topical infections where pathogenic fungal organisms might be

implicated.

Tables 9 & 10 on the investigation of the crude extract and various fractions of

L. mircranthus for antibacterial activity showed that although the plant had

some level of activity against the test organisms, when compared to the

control, all the extracts/fractions showed statistically lower values (up to

P<0.001) suggestive of a generally weak antibacterial action. Among the

extracts/fractions, there was a conspicuous and progressive loss of

antibacterial activity with the initial methanol extracts (CMFM and MFM)

having the highest activities while the other solvent fractions showed diverse

but significantly lower values (Figure 8.0). For the fractions, that of acetone

(AFM) exhibited the highest level of antibacterial activity. The activity trend is

therefore CMFM > MFM > AFM > EFM > CFM > HFM. An attempt at further

fractionation of the acetone soluble fraction (AFM) from Loranthus yielded

sub-fractions with no observable zones of inhibition. This suggested total loss

of antibacterial activity at this stage, and consequently, the bioactivity-guided

isolation process could not proceed any further. The phytochemical screening

results followed the same trend as above with CMFM and MFM showing the

presence of most of the constituents screened (Table 9). CFM and HFM only

showed traces of terpenoids and phenolic compounds. AFM with a slightly

higher activity than the other fractions tested positive for such constituents

like tannins, alkaloids, terpenoids and saponins, while the presence of tannins

cxxii

and flavonoids in EFM might have contributed to its improved antibacterial

activity over CFM and HFM.

The prevailing data for L. micranthus assay showed therefore that the

observed antibacterial activity in mistletoe might have arisen as a result of

interactions among the various plant constituents identified rather than that of

any one in isolation. As previously reported by different researchers, the

antimicrobial activity of several extracts from plants has been linked to

compounds belonging to more than one chemical group. For instance, the

chewing sticks which are widely used in many African countries as an oral

hygiene aid come from different species of plants, and within one stick, the

chemically active component may be heterogeneous (Akpata and Akinrimisi,

1977). Also, the active component of one of the Nigerian chewing sticks

(Fagara zanthoxyloides) was found to consist of various alkaloids (Odebiyi and

Sofowora, 1979). Pawpaw (Carica papaya) yields a milky sap, often called latex,

which is a complex mixture of chemicals (Cowan, 1999). All the compounds in

papaya have been shown to contribute to the antimicrobial properties of its

latex which was found to be bacteriostatic to B. subtilis, Enterobacter cloacae,

E. coli, Salmonella typhi, Staphylococcus aureus and Proteus vulgaris (Osato et

al, 1993). It follows therefore, using bioassay as a guide, that the utilization, in

isolation, of any of the constituents present in Loranthus for the management

of microbial infections might not only lead to treatment failure but could

equally result to the development of resistance among pathogenic organisms

being treated. We therefore recommended the use of the crude, aqueous or

cxxiii

alcoholic extracts of mistletoe leaves in the management of non-complicated

community acquired bacterial infections (Ukwueze et al, 2013; Ukwueze and

Osadebe, 2013).

Conversely, the results of the antimicrobial screening of the solvent fractions

and extracts from P. guajava yielded very positive significant data (Table 14).

Compared with control, all the test materials (except the chloroform-soluble

fraction, PsG-CF) showed antibacterial activities comparable to that of

ceftriaxone at similar concentrations. Most of the fractions from P. guajava

showed similar or slightly higher antibacterial activity than the crude methanol

extract (PsG-CMF) with the ethylacetate-soluble fraction (PsG-EF) exhibiting

the highest antibacterial activity against the test organisms and also showing a

statistically comparable activity with the control. Phytochemical analysis of the

P. guajava extracts/fractions showed that flavonoids and phenolic compounds

might be mainly responsible for the antimicrobial activities as the highly active

fractions had a preponderance of these phytoconstituents (Table 13). These

findings were in consonance with literature reports on the plant (Limsong et al.,

2004; Olajide et al 1999; Xavier et al., 2002; Arima and Danno, 2002; Begum

et al., 2004).

The ethyl acetate fraction (PsG-EF) from P. guajava that gave a satisfactory

bioassay result was subjected to further Sephadex-LH 20 chromatographic

fractionation and purifications to afford ten pooled fractions (PsG-EF1 to PsG-

EF10). Fractions PsG-EF4, PsG-EF5 and PsG-EF7 that had good antibacterial

yield were subjected to semi-preparative reverse phase HPLC purification to

isolate the phenolic compounds I - V. The structures of these compounds were

elucidated using a combination of analytical and spectral techniques which

included: UV, 1H-NMR,

13C-NMR, DEPT,

1H

1HCOSY, HMQC, HMBC and ESI-MS

analyses.

cxxiv

The phytochemical and spectral analysis of the isolated compounds indicated

that they were mainly phenolic glycosides having either hexose or pentose

sugars as the glycone portion. Ultraviolet (UV) spectral analysis of most

reported phenolic glycosides show that the nucleus of chromone (benzo-γ-

pyrone) constitutes the parent chromophore in flavonoids and other related

phenolic compounds like xanthones and anthocyanins. These chromones

which occur naturally in the hydroxylated or glucosylated forms have in

general two or three UV bands near 250, 300 and 350 nm. They represent the

principal bands but could be modified by substitutions as could be seen in the

UV spectral of these isolates. Also, the presence of prominent mass peaks of

fragments in the EI-MS spectra of the compounds indicative of loss of either

pentose (-132 m/z) or pyranose (-162 m/z) molecules confirm that they were

glycosides. Other spectral data of the compounds equally supported this

assertion (figures 9-13 and the appendices).

Compound I was isolated as yellowish viscous liquid with UV maxima at λmax

254.0 and 296.0 nm. The molecular formula of I was deduced as C19H20O9

based on the ESI-MS molecular ion peak at 392.8 [M+H]+. The

1H-NMR

spectrum of compound I showed aromatic signals of an AA’BB’X system at δH

7.7 (m, 2H), δH 7.42 (t, J=7.8 Hz) and δH 7.52 ((t, J=7.4 Hz) respectively. Each of

the AA’BB’ signals were found to be integrating for two protons assigned to H-

9/H-13 and H-10/H-12 respectively while the X signal at δH 7.52 integrated for

one proton assigned to H-11 (Table 15 & figure 9b). These five protons are

assigned to ring A of the aglycone nucleus as shown in the structure below

(XLIII).

cxxv

XLV

Another set of aromatic signals at δH 6.22 (d, J=2.1) and δH 6.07 (d, J=2.1)

integrating for one proton each were assigned to H-5 and H-3 respectively

attached to ring B of the phenolic nuclei (fig. 9b). The 2D-COSY spectrum and

the coupling constant (J = 2.1) of these protons indicated that they were meta-

coupled, while the HMBC showed no correlation between these protons and

the carbonyl carbon (δC 194.8; C-7). The various spectral correlations including

the fragmentation pattern of the ESI-MS spectrum correctly predicted the

structure of ring B of the aglycone backbone as shown (XLVI).

XLVI

cxxvi

The correlations equally showed that unlike in many traditional flavonoid

nuclei where we have fused structures, rings A & B were actually linked by a

carbonyl group. A careful correlation of the HMBC spectrum as presented in

Table 15 informed our proposal of a benzophenone skeleton as shown (XLVII).

XLVII

The 1H-NMR of compound I further showed signals from oxygenated C-Hs (δH

2.84-4.81). This strongly suggested the presence of a sugar moiety [Hobley et

al., 1996]. The signal at δH 4.81 is typical of proton attached to the anomeric

carbon (δC 100.6) assigned to H-1'. The signals at δH 3.85 (dd, J=2.2, 12.1), 3.66

(dd, J=5.6, 12.1), 3.34 (dd), 3.32 (dd), 3.22 (dd, J=6.6, 11.1), 2.84 (dd, J=7.8, 9.1)

were assigned to H-6'b, H-6'a, H-5', H-3', H-4' and H-2' respectively. The

spectral correlations and the molecular ion fragment peak at 231.1 m/z [M+1 (-

162)] indicated the presence of a pyranose sugar, which could be either

glucose or galactose [Qian et al., 2008; Al-Fadhli et al., 2006]. The HMBC

spectrum of the compound that showed no correlation between H-4'of the

cxxvii

sugar moiety and signal at δH 6.07 (H-5) of ring B confirmed this sugar to be d-

glucose [Li et al., 2013]. Galactose with its proton at C-4' projecting outwards

would have produced the reverse correlation. This is evident from the

structures of these sugars (XLVIII).

XLVIII

The attachment of the d-glucopyranosyl moiety (sugar unit) at the C-6 position

of the benzophenone was based on the HMBC correlations of H-5 signal to C-1,

C-3, C-4 and C-6 and that of H-1' to only C-6. Also, in the 1H-NMR spectrum, the

anomeric proton signal of glucose was observed as a doublet at δH 4.81 with

diaxial coupling constant J1’,2’ = 7.7 Hz, which confirmed a β configuration and

the pyranose form of sugar unit [Markham and Geiger, 1994; Olszewska and

Wolbis, 2002]. Compound I was thus elucidated as a benzophenone glycoside

with the name 2,4-dihydroxy-6-O-β-D-glucopyranosylbenzophenone for which

the trivial name guajaphenone A is proposed. To the best of our knowledge,

this compound is being isolated for the first time from Psidium guajava plant.

cxxviii

Our search also revealed that compound 1 is closely related to another

benzophenone glycoside, 2,6-dihydroxy-4-O-β-d-

glucopyranosylbenzophenone, which was isolated for the first time from the

leaves of Psidium guajava. (Fu et al., 2010).

Compound II was isolated as brownish-yellow viscous liquid with UV maxima at

λmax 250.0 and 302.0 nm and a molecular formula of C20H22O9 which was

deduced based on the ESI-MS molecular ion peak at 406.8 [M+H]+. This puts

the difference between the mass of I and II at exactly +14. This value is

equivalent to the mass of a methylene group (-CH2) and could imply the

replacement of a proton in compound I with a methyl (-CH3) group. A

comparison of the 1H and

13C NMR data of I and II showed that their spectra

have similar chemically equivalent signals in almost all the regions with just

few variations (Table 16). The 1H-NMR spectrum of compound II showed only

one signal at δH 6.20 (1H, s) assigned to H-5 of ring B as against two signals at

δH 6.22 (d, J=2.1, 1H) and 6.07 (d, J=2.1, 1H) assigned respectively to H-5 and H-

3 in compound I (figure 10b). This confirms the replacement of a proton in

compound II at the ring B region. The 1H-NMR spectrum of II also showed an

additional signal at the SP3

hybridized C-H region (δH 2.02: d, J=5.7) integrating

for three protons indicating the presence of a methyl group. 2-D COSY, DEPT,

HMQC and HMBC correlations confirmed the assignment of groups in ring B of

the compound as illustrated in this structure (XLIX):

cxxix

XLIX

A strong HMBC correlation of the methyl proton signals to C-3 supported a

methyl group substitution at this position. Further 1H-

1H COSY, DEPT, HMQC

and HMBC correlations confirmed II as a methyl substituted analogue of I and

was elucidated as 2,4-dihydroxy-3-methyl-6-O-β-D-glucopyranosylbenzophen-

one, for which we have proposed the trivial name guajaphenone B. This

compound is being reported for the first time in literature.

Compound III spectra are a little bit different from the previous ones, even

though there exists reasonable degree of symmetry among the spectra. It was

isolated as brownish viscous liquid with UV maxima at λmax 262.0 and 362.0 nm

and a molecular formula of C26H24O13 which was deduced based on the ESI-MS

molecular ion peak at 545.0 [M+H+]. The

1H-NMR spectrum of this compound

showed signals similar to that of Compound II in most of the analytical regions.

The proton signals at δH 7.50, 7.41 and 7.27 were basically identical as those

found at the same region in the previous compounds which corresponded to

cxxx

the proton arrangement already discussed for ring A of the glycosides.

Compared to compound II, however, there was an aromatic singlet proton

signal at δH 7.10 which neither 2D-COSY nor other heteronuclear correlations

were able to assign it as part of rings A or B of that compound. This suggested

the presence of another aromatic nucleus (ring C) in compound III. Its 13

C-NMR

spectrum confirmed this ring with signals between δC 115-125 ppm which were

clearly absent in the spectra of Compounds I & II (Appendices 6, 13 & 18).

HMBC spectrum of the compound showed that this ring C proton is remotely

linked to the signals at δC 108.6. Thus, HMBC and COSY correctly predicted the

structure of ring C of Compound III as in (L) below:

L

The EI-MS spectrum also showed a prominent fragment peak of about 153 m/z

which was closely linked to ring C fragment (Appendix 14). A combination of

all the spectral correlations thus elucidated Compound III as 2,4-dihydroxy-3-

cxxxi

methyl-6-O-βD-glucopyranosylbenzophenone (4→5", 6'→1") benzene-

2",3",4",5"-tetraol.

Compound IV spectra presented quite a different correlation from the

compounds so far discussed. The positive ionization from the EI-MS data was

435 [M+1] while the negative ionization gave 433 [M-1] implying that the

molar mass was 434. The major fragment of 435 was 303 [(M+1) -132]. This

signified a loss of 132 m/z corresponding to the loss of a pentose sugar. The

mass of the aglycone was thus 302 m/z. This with the UV absorption spectrum

confirmed the compound to be a glycoside. The 1H-NMR spectrum suggested

that there were two aromatic spin systems [one ABX (δ 7.52, 7.50 and 6.90:

two are otho-coupled while two are meta-coupled) and one AX (δ 6.39 and

6.20: the two are meta-coupled)]. The 2D-COSY therefore predicted the

structures (LI) of the AX (ring A) and ABX (ring C) aromatic systems to be:

LI

However, the EI-MS data and other spectral information indicated the

presence of another conjugated system (ring B) without proton signals. This

cxxxii

presented the possibility of a fused system, typical of most flavonoids as

indicated below (LII):

LII

The spin system from δH 5.45, 4.33, 3.90, 3.87 and 3.50 belonged to the sugar

(a pentose) protons. From the spectral correlations and literature reports, this

could either be rhamnose or arabinose. The absence of a C-methyl signal in the

1H-NMR spectrum rules out the presence of rahmnose whose occurrence could

equally have caused an upfield shift of H-2' and H-6' signals of the flavonoid

nucleus (Markham and Geiger, 1994). Comprehensive analysis of the NMR data

of IV showed that the compound is the previously reported quercertin-3-O-α-L-

arabinofuranoside with the trivial name, guaijaverin. The structure of the

compound was confirmed by comparison of its 1D and 2D NMR data with

those reported in the literature (Olszewska and Wolbis, 2002; Sanches et al.,

2005; Begum et al., 2002a).

Compound V was isolated as golden-brown viscous liquid with UV maxima at

λmax 265.0 and 294.0 nm and a molecular formula of C25H22O13 which was

cxxxiii

deduced based on the ESI-MS molecular ion peak at 531.4 [M+H+]. As observed

with compound II, the difference between the calculated positive ionization

peak for III and V is -14, a value that is equivalent to the mass of a methylene

group (-CH2) and which could imply the loss of a methyl (-CH3) group for a

proton in this instance. The 1H-NMR spectrum of the compound showed signal

for this additional proton at δH 6.05 (1H, s) assigned to H-3 of ring B as against

the single signal at the same aromatic region in compound III (Table 18). Other

analytical data were very similar to that of Compound III in most of the

regions. Compound V was thus elucidated as the demethylated analogue of

Compound III with the name 2,4-dihydroxy-6-O-βD-

glucopyranosylbenzophenone (4→5", 6'→1") benzene-2",3",4",5"-tetraol.

The antibacterial profiling of the isolated compounds showed that they all had

activities against S. aureus and E. coli but at different degrees. Compounds I &

IV showed activities closest to that of ceftriaxone against both organisms at

similar concentrations (Table 19). It appeared, however, that substitution (in

this case methylation) generally decreased antibacterial action in each case as

can be seen between I versus II and V versus III with the demethylated forms

showing lower MIC values against the bacteria. It was equally observed that

the compounds all had better antibacterial action against Staphylococcus

aureus than against Escherichia coli.

cxxxiv

4.2 Conclusion

Several medicinal plants have been used in traditional practice across Africa to

combat infectious disorders prevalent in that region. Currently, many of these

herbal remedies are available in Nigerian and other West African markets with

bogus claims of efficacy, hence the need for scientific validation of these claims

and establishment of the principle components and mechanisms behind their

activities.

Several works have been carried out in the past to verify the folkloric use of

the African mistletoe in the management of microbial infections. Previous

works by our research team on the crude plant powder and some of its

fractions have established low levels of antibacterial activity by L. micranthus,

but with negligible anti-fungal property. The optimal harvesting seasons as well

as the host trees of choice for mistletoe as an antimicrobial agent were equally

well documented. Further bioactivity-guided analysis on the antibacterial

potential of mistletoe, however, showed that the observed antibacterial

activity in mistletoe might have arisen as a result of interactions among the

various plant constituents identified rather than that of any one in isolation.

We therefore recommended the use of the crude, aqueous or alcoholic

extracts of mistletoe leaves in the management of non-complicated

community acquired bacterial infections.

The ethyl acetate fraction, as well as the Sephadex LH-20 fractions from P.

guajava exhibited significant (P < 0.05) antibacterial activity. The isolated

cxxxv

compounds (I-V) from the active fractions of P. guajava displayed moderate

antibacterial activities. The chemical names of the isolated compounds were

elucidated as : 2,4-dihydroxy-6-O-βD-glucopyranosylbenzophenone (I); 2,4-

dihydroxy-3-methyl-6-O-βD-glucopyranosylbenzophenone (II); 2,4-dihydroxy-

3-methyl-6-O-βD-glucopyranosylbenzophenone (4→5", 6'→1") benzene-

2",3",4",5"-tetraol (III); quercertin-3-O-α-L-arabinofuranoside (IV) and 2,4-

dihydroxy-6-O-β-D-glucopyranosylbenzophenone (4→5", 6'→1") benzene-

2",3",4",5"-tetraol (V). Compounds I, II, III and V are new natural products

which have not been previously reported in literature, and the trivial names

guajaphenone A, B, C and D were proposed, while Compound IV has been

previously reported. Compound I was found to be closely related to a new

benzophenone glycoside, 2,6-dihydroxy-4-O-β-d-

glucopyranosylbenzophenone, isolated for the first time from the leaves of

Psidium guajava (Fu et al., 2010). All the isolated compounds from P. guajava

were also found to have moderate antibacterial activities against E. coli and S.

aureus in comparison to ceftriaxone, with I and IV showing lower MICs than

that of the other isolates against the organisms.

The results of this study are therefore a major contribution to the field of

medicinal chemistry of drugs used in infectious disease states, considering the

observed limitations of the contemporary antibiotic agents. It is expected that

these isolated compounds could be optimized and developed into useful

therapeutic agents in management of infectious disease states and that further

cxxxvi

works should be carried out in order to maximize the full potentials of this

study.

cxxxvii

REFERENCES

Abdelrahim SI, Almagboul AZ, Omer ME, Elegami A. (2002). Antimicrobial

activity of Psidium guajava L. Fitoterapia, 73(7-8): 713-715.

Agbo MO, Lai D, Okoye FBC, Osadebe PO, Proksch P. (2013). Antioxidative

polyphenols from Nigerian mistletoe Loranthus micranthus (Linn.)

parasitizing on Hevea brasiliensis. Fitoterapia; 86:78–83

Akpata ES, Akinrimisi EO. (1977). Antibacterial activity of extracts from some

African chewing sticks. Oral Surg. Oral Med. Oral Pathol, 44:717-722.

Al-Fadhli A, Wahidulla S, D’Souza L. (2006). Glycolipids from the red alga

Chondria armata (Kütz.) Okamura. Glycobiology, 16 (10): 902-915.

Amaral JA, Ekins A, Richards SR, Knowles R. (1998). Effect of selected

monoterpenes on methane oxidation, denitrification, and aerobic

metabolism by bacteria in pure culture. Appl Environ Microbiol, 64:520-

525.

Apisariyakul A, Chaichana N, Takemura H. (1999). Dual effects of quercetin on

contraction in cardiac and skeletal muscle preparations. Res Commun

Molecul Pathol P, 105:129-138.

Arima H, Danno GI. (2002). Isolation of antimicrobial compounds from guava

(Psidium guajava L.) and their structural elucidation. Biosci Biotechnol

Biochem, 66:1727-1730.

Atal CK, Sharma ML, Kaul A, Khajuria A. (1986). Immunomodulating agents of

plant origin. I: Preliminary screening. J. Ethnopharmacol, 18: 133-141.

Bakana P, Claeys M, Totte J, Pieters LA, Van Hoof C, Tamba-Vemba L, van Den

Berghe DA., Vlie-Tink AJ. (1987). Structure and chemotherapeutical

activity of a polyisoprenylated benzophenone from the stem bark of

Garcinia huillensis. Journal of Ethnopharmacology 21, 75–84.

cxxxviii

Balls AK, Hale WS, Harris TH. (1942). A crystalline protein obtained from a

lipoprotein of wheat flour. Cereal Chem, 19:279-288.

Batista O, Duarte A, Nascimento J, Simones MF. (1994). Structure and

antimicrobial activity of diterpenes from the roots of Plectranthus

hereroensis. J Nat Prod 57:858-861.

Becker ED. (2000). High Resolution NMR-Theory and Chemical Applications.

Maryland: Academy Press; pp 251-277.

Begum S, Hassan SI, Ali SN, Siddiqui BS. (2004). Chemical constituents from the

leaves of Psidium guajava. Nat Prod. Res, 18(2): 135-140.

Begum S, Hassan SI, Siddiqui BS. (2002b). Two new triterpenoids from the

fresh leaves of Psidium guajava. Planta Med, 68, 1149-1152.

Begum S, Hassan SI, Siddiqui BS, Shaheen F, Ghayur AH. (2002a). Triterpenoids

from the leaves of Psidium guajava. Phytochemistry, 61, 399-403.

Borris RP. (1996). Natural products research: perspectives from a major

pharmaceutical company. J Ethnopharmacol, 51:29-38.

Brantner A, Males Z, Pepeljnjak S, Antolic A. (1996). Antimicrobial activity of

Paliurus spina-christi mill. J Ethnopharmacol, 52:119-122.

Brownlee HE, McEuen AR, Hedger J, Scott IM. (1990). Antifungal effects of

cocoa tannin on the witches’ broom pathogen Crinipellis perniciosa.

Physiol Mol Plant Pathol, 36:39-48.

Butler MS. (2004). The role of natural product chemistry in drug discovery. J

Nat Prod, 67:2141-2153.

Canada. (1989). Drugs Directorate guidelines. Ottawa: Health Protection

Branch, Health and Welfare Canada.

Casley-Smith JR. (1997). Coumarin in the treatment of lymphoedema and

other high-protein oedemas. In R. O’Kennedy and R. D. Thornes (eds.),

Coumarins: Biology, Applications and Mode of Action. John Wiley & Sons,

Inc., New York, p. 348.

cxxxix

Chaichana N, Apisariyakul A. (1996). Cholinergic blocking effect of quercetin.

Thai J Pharmacol, 18:1-15.

Chaurasia SC, Vyas KK. (1977). In vitro effect of some volatile oil against

Phytophthora parasitica var. piperina. J Res Indian Med Yoga Homeopath,

24-26.

Cheng JT, Yang RS. (1983). Hypoglycemic effect of guava juice in mice and

human subjects. Am. J. Clin Med, 11(1-4): 74-76.

Cichewicz RH, Thorpe PA. (1996). The antimicrobial properties of chile peppers

(Capsicum species) and their uses in Mayan medicine. J Ethnopharmacol,

52:61-70.

Conde-Garcia EA, Nascimento VT, Santiago-Santos AB. (2003). Inotropic effects

of extracts of Psidium guajava L (guava) leaves on the guinea pig atrium.

Braz J Med Biol Res, 36: 661-668.

Cowan, MM. (1999). Plant products as antimicrobial agents. Clinical

Microbiology Reviews, 12 (4): 564-582

Cseke LJ, Kirakosyan A, Kaufman PB, Warba S, Duke JA, Brielmann HL. (2006).

Natural Products from Plants (2nd. ed.), CRC/Taylor & Francis, Boca

Raton, FL.

Dalziel JM. The Useful Plant of West Tropical Africa. (1955). Crown Agents for

Overseas Governments and Administration: London, p. 297.

De Bolle MF, Osborn RW, Goderis IJ, Noe L, Acland D, Hart CA, Torrekens S,

Van Leuven F, Broekart NF. (1996). Antimicrobial properties from

Mirablis jalapa and Amaranthus caudalus: expression, processing,

localization and biological activity in transgenic tobacco. Plant Mol Biol,

31:993-1008.

Dixon RA, Dey PM, Lamb CJ. (1983). Phytoalexins: enzymology and molecular

biology. Adv Enzymol, 55:1-69.

Duke JA. (1985). Handbook of medicinal herbs. CRC Press, Inc.: Boca Raton, Fla.

cxl

Fernandes de Caleya R., Gonzalez-Pascual B. Garcia-Olmedo F, Carbonero, P.

(1972). Susceptibility of phytopathogenic bacteria to wheat purothionins

in vitro. Appl Microbiol, 23: 998-1000.

Fernandez MA, Garcia MD, Saenz MT. (1996). Antibacterial activity of the

phenolic acids fraction of Scrophularia frutescens and Scrophularia

sambucifolia. J Ethnopharmacol, 53:11-14.

Fu HZ, Luo YM, Li CJ, Yang JZ, Zhang DM. (2010). Psidials A-C, three unusual

meroterpenoids from the leaves of Psidium guajava L . Org Lett, 12: 656-

659.

Geissman TA. (1963). Flavonoid compounds, tannins, lignins and related

compounds. In M. Florkin and E. H. Stotz (eds.), Pyrrole pigments,

isoprenoid compounds and phenolic plant constituents, vol. 9. Elsevier:

New York, p. 265.

Ghoshal S, Krishna Prasad BN, Lakshmi V. (1996). Antiamoebic activity of Piper

longum fruits against Entamoeba histolytica in vitro and in vivo. J

Ethnopharmacol, 50:167-170.

Gill FB. (1973). The Machiavellian Fronter’s, 38 (2), pp. 2-5.

Granowitz EV, Brown RB. (2008). Antibiotic adverse reactions and drug

interactions. Crit Care Clin, 24:421-42.

Griggs P. (1991). Mistletoe, Myth, magic and medicine. The Biochemist, 13:3-4.

Grover IS, Bala S. (1993). Study on anti-mutagenic effects of guava in S.

typhimurium. Mutation Research/Genetic Toxicology, 300(1): 1-3.

Habtemariam S, Gray AI, Waterman PG. (1993). A new antibacterial

sesquiterpene from Premna oligotricha. J Nat Prod, 56:140-143.

Hamburger H, Hostettmann K. (1991). The link between phytochemistry and

medicine. Phytochemistry, 30:3864-3874.

Harborne, JB. (1998). Phytochemical methods: a guide to modern techniques of

plant analysis, 3rd

ed. Chapman and Hall, London.

cxli

Hasegawa H, Matsumiya S, Uchiyama M, Kurokawa T, Inouye Y, Kasai R,

Ishibashi S, Yamasaki K. (1994). Inhibitory effect of some triterpenoid

saponins on glucose transport in tumor cells and its application to in vitro

cytotoxic and antiviral activities. Planta Med, 6:240-243.

Haslam E. (1996). Natural polyphenols (vegetable tannins) as drugs: possible

modes of action. J Nat Prod, 59:205-215.

Hendricks K, Nemeth L. (Eds.). (2010). Handmark concise medical dictionary

(8th ed.). Oxford: Market House Books Limited.

Himejima M, Hobson KR., Otsuka T, Wood DL, Kubo I. (1992). Antimicrobial

terpenes from oleoresin of ponderosa pine tree Pinus ponderosa: a

defense mechanism against microbial invasion. J Chem Ecol, 18:1809-

1818.

Hobley P, Howarth O, Ibbett RN. (1996). 1H and

13C-NMR shifts for

aldopyranose and aldofuranose monosaccharides: Conformational

analysis and solvent dependence. Magn. Reson. Chem., 34:755–760.

Hoult JRS, Paya M. (1996). Pharmacological and biochemical actions of simple

coumarins: natural products with therapeutic potential. Gen Pharmacol,

27:713-722.

Hufford CD, Jia Y, Croom EM Jr., Muhammed I, Okunade AL, Clark AM, Rogers

RD. (1993). Antimicrobial compounds from Petalostemum purpureum. J

Nat Prod, 56:1878-1889.

Hui-Yin Chen, Gow-Chin Yen. (2007). Antioxidant activity and free radical-

scavenging capacity of extracts from guava (Psidium guajava L.) leaves,

Food Chemistry, 101(2): 686-694.

Jaiarj P, Khoohaswan P, Wongkrajang Y, Peungvicha P, Suriyawong P, Saraya

MLS, Ruangsomboo O. (1999). Anticough and antimicrobial activities of

Psidium guajava Linn leaf extract. J Ethnopharmacol, 67: 203-212.

cxlii

Jaiarj P, Wongkrajang Y, Thongpraditchote S, Peungvicha P, Bunyapraphatsara

N, Opartkiattikul N. (2000). Guava leaf extract and topical haemostasis.

Phytother Res, 14: 388-391.

Jawetz E, Melnick JL, Adelberg EA. (1989). Medical Microbiology; 8th

ed. USA:

Prentice-Hall Int’l Inc., pp. 143-172.

Jones GA, McAllister TA, Muir AD, Cheng KJ. (1994). Effects of sainfoin

(Onobrychis viciifolia scop.) condensed tannins on growth and proteolysis

by four strains of ruminal bacteria. Appl Environ Microbiol, 60:1374-1378.

Jones NL, Shabib S, Sherman PM. (1997). Capsaicin as an inhibitor of the

growth of the gastric pathogen Helicobacter pylori. FEMS Microbiol Lett,

146:223-227.

Kadota S, Basnet P, Ishii E, Tamura T, Namba T. (1997). Antibacterial activity of

trichorabdal from Rabdosia trichocarpa against Helicobacter pylori. Zentbl

Bakteriol, 286: 63-67.

Kafaru E. (1993). Herbal Remedies, The Guardian, Thursday June 3, p 24.

Kavanagh F. (1963). Analytical Microbiology, Vol. 1; California: Academic Press.

Kazmi MH, Malik A, Hameed S, Akhtar N, Noor Ali S. (1994). An anthraquinone

derivative from Cassia italica. Phytochemistry, 36:761-763.

Kinghorn AD. (2008). Drug Discovery from Natural Products, In, Lemke TL,

Williams DA (eds.) FOYE's Principles of Medicinal Chemistry (6th Ed.) India:

Wolters Kluwer (India ) Pvt. Ltd, Pp 12-25.

Knittel JJ, Zavod RM. (2008). Drug design and relationship of functional groups

to pharmacologic activity. In: Lemke TL, Williams DA (Eds.), Foye's

Principles of Medicinal Chemistry, (6th ed.). Philadelphia, PA: Lipincott

Williams & Wilkins, pp. 26-53.

Koehn FE, Carter GT. (2005). The evolving role of natural products in drug

discovery. Nat Rev Drug Discov, 4:206-220.

cxliii

Kubo I, Muroi H, Himejima M. (1993). Combination effects of antifungal

nagilactones against Candida albicans and two other fungi with

phenylpropanoids. J Nat Prod, 56:220-226.

Li X, Huang G, Zhao G, Chen W, Li J, Sun L. (2013). Two new monoterpenes

from Tithonia diversifolia and their anti-hyperglycemic activity. Rec. Nat.

Prod., 7: 351-354.

Lewis WH, Elvin-Lewis MP. (1995). Medicinal plants as sources of new

therapeutics. Ann Mol Bot Gard, 82:16-24.

Limsong J, Benjavong kulchai E, Kuvataanasuchati J. (2004). Inhibitory effects

of some herbal extracts on adherence of S. mutans. J Ethnopharmcol,

92(2-3): 281-289.

Lovian I. (1980). Antibiotics in Laboratory Medicine. London: Williams and

Williams, Baltimore. 7-22 p.

Lozoya X, Becerril G, Martinez ML. (1990). Model of intraluminal perfusion of

the guinea pig ileum in vitro in the study of the antidiarrheal properties of

the guava (Psidium guajava). Arch Invest Med, 21(2): 155-162.

Lozoya X, Meckes M, Abou-Zaid M, Tortoriello J, Nozzolillo C, Amason JT.

(1994). Quercetin glycosides in Psidium guajava L. leaves and

determination of spasmolytic principle. Arch Med Res; 25:11-15.

Lutterodt GD. (1992). Inhibition of Microlax-induced experimental diarrhea

with narcotic-like extracts of Psidium guajava leaf in rats. J

Ethnopharmacol, 37(2): 151-157.

Lutterodt GD, Maleque A. (1988). Effects on mice locomotor activity of a

narcotic-like principle from Psidium guajava leaves. J Ethnopharmacol.

24(2-3): 219-231.

Manosroi J, Dhumtanom P, Manosroi A. (2006). Anti-proliferative activity of

essential oil extracted from Thai medicinal plants on KB and P38 cell lines.

Cancer lett, 235(18): 114-120.

cxliv

Martin AR. (1988) Antibacterial Antibiotics, In: Delgado JN, Remers WA (eds.),

Wilson and Gisvold's Textbook of Organic Medicinal and Pharmaceutical

Chemistry (10th Ed.). USA: Lippincott-Raven Publishers, pp 253-325.

Markham KR, Geiger N. (1994) 1H nuclear magnetic resonance spectroscopy of

flavonoids and their glycosides in hexadeuterodimethylsulfoxide, In:

Harborne JB (ed.), The Flavonoids Advances in Research since 1986. New

York: Chapman and Hall; pp 441-473.

Matsuo N, Hanamure, Koyoko SY, Nakamura, Tomita I. (1994) Identification of

(+) gallocatechin as a bio-antimutagenic compound in Psidium guajava

leaves, Phytochemistry, 36(4): 1027-1029.

Meckes M, Calzada F, Tortoriello J, González JL, Martinez M. (1996).

Terpenoids isolated from Psidium guajava hexane extract with depressant

activity on central nervous system. Phytother Res, 10: 600-603.

Mendoza L, Wilkens M, Urzua A. (1997). Antimicrobial study of the resinous

exudates and of diterpenoids and flavonoids isolated from some Chilean

Pseudognaphalium (Asteraceae). J Ethnopharmacol, 58:85-88.

Mitscher LA, Lemke TL, Gentry EJ. (2008). Antibiotics and Antimicrobial

Agents, In: Lemke TL, Williams DA (eds.), FOYE's Principles of Medicinal

Chemistry (6th Ed.) India: Wolters Kluwer (India ) Pvt. Ltd, Pp 1028-1083.

Morton J. (1987). Guava. In: Fruits of warm climates, Julia F. Morton, Miami FL,

356-363

Obatomi DK, Aina VO, Temple VJ. (1996). Effect of African Mistletoe on blood

pressure in spontaneously hypertensive rats. Intl J pharmacog, 34 (2):

124-127.

Obatomi DK, Bikomo EO, Temple VJ. (1994). Anti-diabetic properties of African

Mistletoe in streptozotocin-induced diabetic rats. J Ethnopharmacol,

43:13-17.

Odebiyi OO, Sofowora EA. (1979). Antimicrobial alkaloids from a Nigerian

chewing sticks (Fagara zanthoxyloides). Planta Med, 36:204-207.

cxlv

Oh WK, Lee CH, Lee MS, Bae EY, Sohn CB, Oh H, Kim BY, Ahn JS. (2005).

Antidiabetic effects of extracts from Psidium guajava. J Ethnopharmacol,

96(3): 411-415.

Ojewole JA. (2005). Hypoglycemic and hypotensive effects of Psidium guajava

L, (Myrtaceae) leaf aqueous extracts. Methods Find Exp Clin Pharmacol,

27(10): 689-695.

Okoye FBC, Ebi GC. (2007). Preliminary antimicrobial and phytochemical

investigation of the plant extracts and column fractions of Alchornea

floribunda leaves. J Pharm Allied Sci, 4(1): 395-402.

Olajide OA, Awe SO, Makinde JM. (1999). Pharmacological studies on the

leaves of Psidtum guajava. Fitoterapia, 70: 25-31.

Oliver-Bever B. Medicinal Plants of Tropical West Africa. (1986). Cambridge

University: London, p. 375.

Olszewska M, Wolbis M. (2002). Further flavonoids from the flowers of Prunus

spinosa L, Acta Polon Pharm-Drug Res, 59 (2): 133-137.

Omeje EO, Osadebe PO, Esimone CO, Nworu CS, Kawamura A, Proksch P.

(2012). Three hydroxylatedlupeol-based triterpenoid esters isolated from

the Eastern Nigeria mistletoe parasitic on Kola acuminata. Natural

Product Research; 26 (19):1775-1781.

Omulokoli E, Khan B, Chhabra SC. (1997). Antiplasmodial activity of four

Kenyan medicinal plants. J Ethnopharmacol, 56:133-137.

Osadebe PO, Abba CC, Agbo MO. (2012). Antimotility effects of extracts and

fractions of Eastern Nigeria mistletoe (Loranthus micranthus Linn). Asian

Pacific Journal of Tropical Medicine; 5(7):556-560

Osadebe PO, Akabogu IC. (2006). Antimicrobial activity of Loranthus

micranthus harvested from Kolanut tree. Fitoterapia, 77: 54-56.

Osadebe PO, Dieke CA, Okoye, FBC. (2008). A study of the seasonal variation in

the antimicrobial constituents of the leaves of the leaves of Loranthus

cxlvi

micranthus sourced from Persea americana. Research Journal of

Medicinal Plant, 2 (1): 48-52.

Osadebe PO, Okide GB, Akabogu IC. (2004). Study on anti-diabetic activities of

crude methanolic extracts of Loranthus micranthus (Linn.) sourced from

five different host trees. J Ethnopharmacol, 95: 133-138.

Osadebe PO, Omeje EO, Uzor PF, David EK, Obiorah DC. (2010). Seasonal

variation for the antidiabetic activity of Loranthus micranthus methanol

extract. Asian Pacific Journal of Tropical Medicine, 3(3):196-199.

Osadebe PO, Ukwueze SE. (2004). A comparative study of the Phytochemical

and Antimicrobial properties of the Eastern Nigeria species of African

Mistletoe (Loranthus micranthus) sourced from different host trees.

Journal of Biological Research and Biotechnology, 2(1): 18-23.

Osato JA, Santiago LA, Remo GM, Cuadra MS, Mori A. (1993). Antimicrobial

and antioxidant activities of unripe papaya. Life Sci, 53: 1383-1389.

Osman AM, Younes ME, Sheta AE. (1974). Triterpenoids of the leaves of

Psidium guajava . Phytochemistry, 13: 2015-2016.

Phillipson JD, O’Neill MJ. (1987). New leads to the treatment of protozoal

infections based on natural product molecules. Acta Pharm. Nord, 1:131-

144.

Parekh J, Chanda, S. (2007). In vitro screening of antibacterial activity of

aqueous and alcoholic extracts of various Indian plant species against

selected pathogens from Enterobacteriaceae. Afr J Micro Res, 1(6):92-99.

Qadan F, Thewaini AJ, Ali DA, Afifi R, Elkhawad A, Matalka KZ. (2005). The

antimicrobial activities of Psidium guajava and Juglans regia leaf extracts

to acne-developing organisms. Am J Chin Med, 33(2): 197-204.

Qian H, Nihorimbere V. (2004). Antioxidant power of phytochemicals from

Psidium guajava leaf. J Zhejiang Univ Sci; 5(6): 676-683.

Qian WL, Khan Z, Watson DG, Fearnley J. (2008). Analysis of sugars in bee

pollen and propolis by ligand exchange chromatography in combination

cxlvii

with pulsed amperometric detection and mass spectrometry. Journal of

Food Composition and Analysis, 21: 78–83.

Rao, KV, Sreeramulu K, Gunasekar D, Ramesh D. (1993). Two new

sesquiterpene lactones from Ceiba pentandra. J Nat Prod, 56: 2041-2045.

Ríos JL, Recio MC, Villar A. (1988). Screening methods for natural products with

antimicrobial activity: a review of the literature. Journal of

ethnopharmacology, 23: 127-149.

Rodriguez RC, Cruz PH, Rios HG. (2001). Lectins in fruits having gastrointestinal

activity: their participation in hemagglunating property of Escherichia coli

O157. Arch.Med.Res, 32(4): 251-257.

Rojas A, Hernandez L, Pereda-Miranda R, Mata R. (1992). Screening for

antimicrobial activity of crude drug extracts and pure natural products

from Mexican medicinal plants. J Ethnopharmacol. 35:275-283.

Rotimi VO, Laughon BE, Bartlett JG, Mosadami HA. (1988). Activities of

Nigerian chewing stick extracts against Bacteroides gingivalis and

Bacteroides melaninogenicus. Antimicrob Agents Chemother, 32: 598-

600.

Roy CK, Kamath JV, Asad M. (2006). Hepatoprotective activity of Psidium

guajava L leaf extract, Indian J Exp Biol, 44(4): 305-311.

Rwangabo PC, Claeys M, Pieters L, Corthout J. (1988). Umuhengerin, new

antimicrobially active flavanoid from Lantana trifolia. J Nat Prod, 51: 966-

968.

Ryan KJ, Ray CG (eds.) (2004). Sherris Medical Microbiology (4th ed.). McGraw

Hill. ISBN 0-8385-8529-9.

Sakanaka S, Kim M, Taniguchi M, Yamamoto T. (1989). Antibacterial substances

in Japanese green tea extract against Streptococcus mutans, a cariogenic

bacterium. Agric Biol Chem, 53: 2307-2311.

cxlviii

Sakanaka S, Shimura N, Aizawa M, Kim M, Yamamoto T. (1992). Preventive

effect of green tea polyphenols against dental caries in conventional rats.

Biosci Biotechnol Biochem, 56: 592-594.

Sanches NR, Cortez DAG, Schiavini MS, Nakamura CV, Filho BPD. (2005). An

evaluation of antibacterial activities of Psidium guajava (L.). Brazilian Arch

Biol Tech, 48 (3): 429-436.

Scalbert A. (1991). Antimicrobial properties of tannins. Phytochemistry, 30:

3875-3883.

Schultes RE. (1978). The kingdom of plants, In: Thomson WAR (ed.), Medicines

from the Earth. McGraw-Hill Book Co.: New York, N.Y, p. 208.

Seshadri TR, Vasishta K. (1965). Polyphenols of the leaves of Psidium guajava:

quercetin, guaijiaverin, leucocyanidin and amritoside. Phytochemistry, 4:

989-992.

Shaheen HM. (2000). Effect of Psidium guajava leaves on some aspects of

central nervous system in mice. Phytother Res, 14(2): 107-111.

Sharon N, Ofek I. (1986). Mannose specific bacterial surface lectins, In:

Mirelman D (ed.), Microbial lectins and agglutinins. John Wiley & Sons,

Inc: New York, N.Y, p. 55-82.

Shruthi SD, Roshan A, Timilsina SS, Sunita S. (2013). A review on the medicinal

plant Psidium guajava Linn. (Myrtaceae). Journal of Drug Delivery &

Therapeutics, 3(2): 162-168.

Singh RB, Rastogi SS, Singh NK, Ghosh S, Gupta S, Niaz MA. (1993). Can guava

fruit intake decrease blood pressure and blood lipids? J Hum Hypertens,

7(1): 33-38.

Singh RB, Rastogi SS, Singh NK, Ghosh S, Niaz MA. (1992). Effects of guava

intake on serum total and high-density lipoprotein cholesterol levels and

on systemic blood pressure. Am J Cardiol, 70(15): 1287-1291.

Snavely SR, Hodges GR. (1984). The neurotoxicity of antibacterial agents. Arch

Intern Med, 101: 92-104.

cxlix

Steinbeck C. (2004). Recent developments in automated structure elucidation

of natural products. Nat Prod Rep, 21:512-518.

Stern JL, Hagerman AE, Steinberg PD, Mason PK. (1996). Phlorotannin-protein

interactions. J Chem Ecol, 22:1887-1899.

Sun, HD, Qiu SX, Lin LZ, Wang ZY, Lin ZW, Pengsuparp T, Pezzuto JM, Fong

HH, Cordell GA, Farnsworth NR. (1996). Nigranoic acid, a triterpenoid

from Schisandra sphaerandra that inhibits HIV-1 reverse transcriptase. J

Nat Prod, 59: 525-527.

Sunttornusk L. (2002). Quantitation of vitamin C content in herbal juice using

direct titration. J Pharm Biomed Anal, 28(5): 849-55.

Tassou C, Drosinos EH, Nychas GJE. (1995). Effects of essential oils from mint

(Mentha piperita) on Salmonella enteritidis and Listeria monocytogenes in

model food systems at 4°C and 10°C. Journal of Applied Bacteriology, 78:

593–600.

Tassou C, Koutsoumanis K, Nychas GJE. (2000). Inhibition of Salmonella

enteritidis and Staphylococcus aureus in nutrient broth by mint essential

oil. Food Research International, 33: 273–280.

Taylor RSL, Edel F, Manandhar NP, Towers GHN. (1996). Antimicrobial activities

of southern Nepalese medicinal plants. J Ethnopharmacol, 50: 97-102.

The wealth of India (Vol X). (2003). National Institute of science

communication and Information Resources, New Delhi, 411-413.

Thea LC, Kelvin MW, Chris M. (2008). Herbal Medicine, 212 (4) Retrieved from

http:// www.kcc.cc.il.us/full/url.

Thomson WAR (ed.). (1978). Medicines from the Earth. McGraw-Hill Book Co.:

Maidenhead, United Kingdom.

Toda M, Okubo S, Ohnishi R, Shimamura T. (1989). Antibacterial and

bactericidal activities of Japanese green tea. Jpn J Bacteriol, 45: 561-566.

cl

Tona L, Kambu K, Ngimbi N, Mesia K, Penge O, Lusakibanza M, Cimanga K, De

Bruyne T, Apers S, Totte J, Pieters L, Vlietinck AJ. (2000). Antiamoebic and

spasmolytic activities of extracts from some antidiarrhoeal traditional

preparations used in Kinshasa, Congo. Phytomedicine, 7(1): 31-38.

Tsuchiya H, Sato M, Miyazaki T, Fujiwara S, Tanigaki S, Ohyama M, Tanaka T,

Iinuma M. (1996). Comparative study on the antibacterial activity of

phytochemical flavanones against methicillin-resistant Staphylococcus

aureus. J Ethnopharmacol, 50:27-34.

Ukwueze SE, Osadebe PO. (2012). Determination of anti-fungal properties of

the African mistletoe species: Loranthus micranthus L. International

Journal of Pharma and Bio Sciences, 3(1): 454-458.

Ukwueze SE, Osadebe PO. (2013). Bioassay-guided fractionation for the

isolation of the antibacterial compounds from Loranthus species of the

mistletoe plant, In: Gupta VK. (ed.); Utilization and Management of

Medicinal Plants Vol. 2; New Delhi: Daya Publishing House, pp 421-430.

Ukwueze SE, Osadebe PO, Ezenobi NO. (2013). Bioassay-guided evaluation of

the antibacterial activity of Loranthus species of the African mistletoe.

Intl J Pharm Biomed Research, 4(2): 79-82.

Vanden-Berghe DA, Vlietinck AJ. (1991). Screening for antibacterial and

antiviral agents, In: Hostettmann K. (ed.); Methods in Plant Biochemistry,

Vol. 6, Assays for Bioactivity; London: Academic Press, pp 47-69.

Vijaya K, Ananthan S, Nalini R. (1995). Antibacterial effect of the aflavin,

polyphenon 60 (Camellia sinensis) and Euphorbia hirta on Shigella spp-a

cell culture study. J Ethnopharmacol, 49: 115-118.

Wagner ML, Teresa F, Elida A, Rafeal AG. (1996). Micromolecular and

macromolecular comparison of Argentina mistletoe (Ligaria Cuneifolia).

Acta Farmacentica Babaerense, 15 (2): 99 – 105.

Wei L, Li Z, Chen B. (2000). Clinical study on treatment of infantile rotaviral

enteritis with Psidium guajava L. Zhongguo Zhong Xi Yi Jie He Za Zhi,

20(12): 893-895.

cli

Wild R. (ed.) (1994). The complete Book of Natural and Medicinal Cures. Rodale

Press, Inc., Emmaus, Pa.

Xavier Lozoya, Hortesia Reyes, Soto-Gonzalez Y, Doubova SV. (2002). Intestinal

anti-spasmodic effect of a phytodrug of Psidium guajava folia in the

treatment of acute diarrheic disease. J Ethnopharmacol, 83(1-2): 19-24.

Ya, C, Gaffney SH, Lilley TH, Haslam E. (1988). Carbohydrate-polyphenol

complexation, In: R. W. Hemingway and J. J. Karchesy (eds.), Chemistry

and Significance of Condensed Tannins. Plenum Press, New York, N.Y., p.

553.

Zhang Y, Lewis K. (1997). Fabatins: new antimicrobial plant peptides. FEMS

Microbiol. Lett., 149:59–64

clii

APPENDICES

cliii

Appendix 1: ESI-MS /UV Spectra of Compound I

cliv

Appendix 2: H-NMR Spectrum (500MHz; MeOD) of Compound I

clv

clvi

Appendix 3: H-NMR Spectrum (600MHz; MeOD) of Compound I

clvii

Appendix 4: H-NMR Spectrum (600MHz; DMSO-d6) of Compound I

clviii

clix

Appendix 5: 2-D COSY of Compound I

clx

clxi

Appendix 6: C-13 NMR Spectrum of Compound I

clxii

Appendix 7: HMQC Spectrum of Compound I

clxiii

clxiv

Appendix 8: HMBC Spectrum of Compound I

clxv

clxvi

Appendix 9: ESI-MS /UV Spectra Compound II

clxvii

Appendix 10: H-NMR Spectrum (500MHz;MeOD) of Compound II

clxviii

clxix

Appendix 11: H-NMR Spectrum (600MHz; DMSO) of Compound II

clxx

Appendix 12: 2-D COSY of Compound II

clxxi

Appendix 13: C-13 NMR Spectrum of Compound II

clxxii

Appendix 14: ESI-MS /UV Spectra Compound III

clxxiii

Appendix 15: H-NMR Spectrum (500MHz; MeOD) of Compound III

clxxiv

Appendix 16: H-NMR Spectrum (600MHz; DMSO) of Compound III

clxxv

Appendix 17: 2-D COSY of Compound III

clxxvi

Appendix 18: C-13 NMR Spectrum of Compound III

clxxvii

Appendix 19: HMQC Spectrum of Compound III

clxxviii

Appendix 20: HMBC Spectrum of Compound III

clxxix

clxxx

Appendix 21: UV Spectrum of Compound IV

clxxxi

Appendix 22: ESI-MS Spectra of Compound IV

clxxxii

Appendix 23: H-NMR Spectrum (500MHz; MeOD) of Compound IV

clxxxiii

Appendix 24: 2-D COSY of Compound IV

clxxxiv

Appendix 25: H-NMR Spectrum (500MHz; MeOD) of Compound V

clxxxv

Appendix 26: 2-D COSY of Compound V

clxxxvi

clxxxvii

Appendix 27: 2-D COSY [aromatic region] of Compound V