Download IHIM, STELLA AMARACHI - It works

Document related concepts

Complement system wikipedia , lookup

Macrophage wikipedia , lookup

DNA vaccination wikipedia , lookup

T cell wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Lymphopoiesis wikipedia , lookup

Phagocyte wikipedia , lookup

Molecular mimicry wikipedia , lookup

Immune system wikipedia , lookup

Adaptive immune system wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Immunomics wikipedia , lookup

Innate immune system wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Transcript
i
EVALUATION OF THE IMMUNOMODULATORY ACTIVITIES OF THE AQUEOUS
EXTRACT AND THE BETA–D-GLUCAN-RICH POLYSACCHARIDE FRACTION OF
Pleurotustuberregium(Pleurotaceae)
BY
IHIM, STELLA AMARACHI
PG/MPHARM/13/64775
DEPARTMENT OF PHARMACOLOGY AND TOXICOLOGY
FACULTY OF PHARMACEUTICAL SCIENCES
UNIVERSITY OF NIGERIA, NSUKKA
APRIL, 2015.
ii
TITLE PAGE
EVALUATION OF THE IMMUNOMODULk2ATORY ACTIVITIES OF THE AQUEOUS
EXTRACT AND THE BETA–D-GLUCAN- RICH POLYSACCHARIDE FRACTION
OFPleurotustuberregium (Pleurotaceae)
iii
CERTIFICATION
IHIM, STELLA AMARACHI, a postgraduate student in the Department of Pharmacology and
Toxicology with registration number PG/MPHARM/13/64775has satisfactorily completed the
requirements for the award of the degree of Master of Pharmacy (M.PHARM) of the Department
of Pharmacology and Toxicology, Faculty of Pharmaceutical Sciences, University of Nigeria,
Nsukka. The work embodied in this dissertation is original and has not been submitted for the
award of any other diploma or degree in this or any other University.
---------------------------------------Dr. C.SNworu
Dr.T.C Okoye
(Supervisor)
----------------------------------------(Head of Department)
iv
DEDICATION
This work is dedicated to God Almighty for his mercies and love in my life and to my supervisor,
Dr.ChukwuemekaS. Nworu,for his fatherly support in making this work a success.
v
ACKNOWLEDGEMENTS
I wish to express my profound gratitude to my supervisor, DrChukwuemekaNworufor making
this research work a reality. I also want to appreciate Prof. Charles Okoye, Prof. P. A.Akah, Prof.
C.O.Esimone, Dr. T.COkoye, Dr. (Mrs) A. C. Ezike, Rev (Pharm) E.Ezea,Dr (Mrs)Odoh, Dr.
NickObitte, Mr Austin Okorieand other academic, technical and administrative staff in the
Department of Pharmacology, University of Nigeria, Nsukka for the part they played in making
this work successful. I will want to appreciate my colleagues and friends - Ife, Nenye, Azom,
Lydia, Lawrence, Victor, Ebuka, Lovelyn, EzeChinenye, Chigo, Ebere, Peter, Oge, Nnamdi,
Uche, Chika, John and others too numerous to mention for their effort and encouragement in
making this master’s programme a reality. I wish to acknowledge the help of the academic and
technical staff of Pharmaceutical Microbiology and Biotechnology,Faculty of Pharmaceutical
Sciences,NnamdiAzikiwe University (Agulu Campus)
I will not fail to acknowledge and thank my family; Engr and Mrs George Ihim- my Parents, my
siblings- Miki, Urchman, Victor, Chioma, Promise and my friend Kelvin for their support,
understanding and prayers during this period.
Finally, I thank the Almighty God for his favour, for the good health and for everything He has
done for me.
Ihim, StellaAmarachi
April, 2015
vi
ABSTRACT
Mushroom cuisines are global delicacies and have been used for decades, not only as food, but
mostly for the health benefits which are claimed or scientifically proven. The present study
explored the immunomodulatoryactivites of the hot aqueous extract of a local oyster mushroom,
Pleurotustuberregium (Fr.) Singer (Pleurotaceae) (PT) and its β–D-Glucan-Rich Polysaccharide
fraction (BGP).The effects of the aqueous extract (PT) and the β–D-Glucan-Rich Polysaccharide
fraction (BGP) were tested on some specific and non-specific immune responses in immunecompetent mice and in culture of RAW 264.7 macrophage cells. The effect of the PT and BGP on
specific cell mediated immune response was investigated by the delayed type hypersensitivity
response (DTHR) whilethe effect of the extract on specific humoral immune responses of treated
mice was determined by prime-boost immunization protocol using ovalbumin as antigen and
followed by the determination of antibody titers in the sera using the enzyme-linked
immunosorbent assay (ELISA).Their effect on non–specific immune responses was determined
by the colloidal carbon-clearance assay in mice. The effect of BGP on functional maturation and
activation of the monocytic cells was also determined by measuring the level of tumour necrosis
factor (TNF)-α and inducible oxygen (iNO) expressed into culture supernatant using cytokine
capture ELISA and Griess reagent respectively.Short-term oral administration of PT (100, 200
and 400mg/kg) or BGP (100 and 200mg/kg) elicited a dose-related increase in DTHR and
increased the mean phagocytic clearance of colloidal carbon in mice as much as 7 – 10 fold when
compared to the clearance in the untreated group of mice. In a homologous prime-boost
immunization schedule, oral supplementation with PT (100, 200 and 400mg/kg) or BGP (100 and
200mg/kg) significantly (P<0.05) increased primary and secondary ovalbumin (OVA) – specific
total IgG, IgG1 and IgG2a titres in treated mice as much as 2 – 4 folds compared to the untreated
control. In cell culture, stimulation of RAW 264.7 macrophages with BGP (5-1000 µg/ml)
induced a remarkable and significant (P<0.05) proliferation of total spleenocytes in a
concentration-related manner. Stimulation of RAW 264.7 cells with BGP (5-100 µg/ml)
significantly (P<0.05) increased the levels of expressed TNF-α and iNO in culture. Similarly,
prestimulation of RAW 264.7 cells significantly (P<0.05) increased the phagocytic capacity of
the macrophages in a neutral dye uptake assay. Acute toxicity tests revealed that even at doses of
PT up to 5000mg/kg body weight (per os), there was no lethality or any sign of acute intoxication
in the mice after 24 h. Put together, the result of these investigations suggest that hot aqueous
extract of P tuberegium (PT) and its β–D-Glucan-rich polysaccharide fraction (BGP) possess
some immunomodulatory activities in mice and could be further investigated and harnessed for
therapeutic purposes in immunodeficiency conditions.
vii
TABLE OF CONTENTS
TITLE PAGE -
-
-
-
-
-
-
-
-
-
i
CERTIFICATION
-
-
-
-
-
-
-
-
-
iii
DEDICATION
-
-
-
-
-
-
-
-
-
iv
ACKNOWLEDGMENTS
-
-
-
-
-
-
-
-
v
ABSTRACT -
-
-
-
-
-
-
-
-
vi
TABLE OF CONTENTS
-
-
-
-
-
-
-
-
vii
LIST OF FIGURES -
-
-
-
-
-
-
-
-
xi
LIST OF TABLES
-
-
-
-
-
-
-
-
xiii
-
-
-
-
-
-
-
-
xiv
-
LIST OF APPENDICES
CHAPTER ONE: INTRODUCTION AND LITERATURE REVIEW
1.1
Scientific Background -
1.2
-
-
-
-
-
-
1
Overview of the Immune System
-
-
-
-
-
3
1.2.1
Organs of the Immune System
-
-
-
-
-
3
1.2.1.1
Primary Lymphoid Organs
-
-
-
-
-
3
1.2.1.2
Secondary Lymphoid Organ -
-
-
-
-
-
5
1.2.2
Cells of the Immune System
-
-
-
-
-
6
1.2.2.1 Lymphoid Stem Cells -
-
-
-
-
-
-
9
1.2.2.2 Myeloid Stem Cells
-
-
-
-
-
-
11
Arms of the Immune System -
-
-
-
-
-
-
-
-
-
14
-
-
-
-
23
-
-
-
-
-
26
-
-
-
-
-
28
-
-
-
-
-
29
1.2.3
-
1.2.3.1 Innate Immunity
-
-
1.2.3.2 Adaptive Immunity
-
-
-
1.2.4
Overview of Antibodies
1.2.4.1
Classes of Immunoglobulin
1.2.4.2
Roles of Antibodies
1.3
Mediators of the Immune System
-
-
-
-
-
31
1.3.1
Cytokines
-
-
-
-
-
-
31
1.3.2
Complement System
-
-
-
-
-
35
Disorders of the Immune System
-
-
-
-
-
38
1.4.1
Hypersensitivity
-
-
-
-
-
-
38
1.4.2
Immune Deficiency Diseases -
-
-
-
-
-
39
1.4
-
-
-
-
-
14
-
viii
1.4.3
Autoimmune Diseases
-
-
-
-
-
-
41
1.4.4
Graft Versus Host Diseases -
-
-
-
-
-
42
1.4.5
Immune Complex Diseases
-
-
-
-
-
42
1.5
The Concept of Immunomodulation, Immunosuppression, Immunostimulation and
-
Immunotolerance
-
-
-
-
-
-
-
43
1.5.1
Immunostimulation
-
-
-
-
-
-
-
43
1.5.2
Immunosuppression -
-
-
-
-
-
-
44
1.5.3
Tolerance
-
-
-
-
-
-
45
1.6
Potentials of Mushroom as Immunomodulatory Substance of Natural Origin45
1.7
Beta (B) Glucans
-
-
-
-
-
48
1.7.1
Beta Glucans and the Immune System
-
-
-
-
48
1.7.1.1
Beta GlucanImmunostimulating Activity
-
-
-
-
49
1.7.1.2
Beta Glucan Increases Resistance to Infectious Challenge -
-
50
1.7.1.3
Beta GlucanAnticarcinogenic Activity
-
51
1.7.1.4
Beta Glucan as Adjuvant to Cancer Chemotherapy and Radiotherapy
51
1.8
Botanical Profile and Review Of Pleurotustuberregium
-
-
52
1.8.1
Taxonomy of Pleurotustuberregium -
-
-
-
53
1.8.2
Botanical Description of Pleurotustuberregium
-
-
-
53
1.8.3
Geographical Distribution of Pleurotustuberregium -
-
-
55
1.8.4
Ethnomedicinal and Folkloric Uses of Pleurotustuberregium
-
55
1.8.5
Pharmacological Studies, Phytochemical and Proximate Constituents
-
ofP. tuberregium
-
-
-
-
-
-
-
-
-
-
-
-
-
-
56
CHAPTER TWO: MATERIALS AND METHODS
2.1
Materials
2.1.1
-
-
-
-
-
-
-
-
57
Chemicals and Reagents
-
-
-
-
-
-
57
2.1.2
Equipment
-
-
-
-
-
-
-
-
57
2.2
Method
-
-
-
-
-
-
-
-
57
2.2.1
Collection and Authentication of Plant Materials
-
-
-
57
2.2.2
Preparation of Plant Material -
-
-
-
58
2.2.3
Bioactivity Guided Fractionation of Crude Extract -
-
-
58
2.2.4
Extraction of the β glucan rich Polysaccharide Fraction of
-
-
ix
-
-
-
-
-
60
2.2.5
Phytochemical Analysis of Extracts -
-
-
-
-
60
2.2.6
Pharmacological Studies
-
-
-
-
-
-
61
2.2.6.1
Animals
-
-
-
-
-
-
-
-
61
2.2.6.2
Antigens
-
-
-
-
-
-
-
-
61
2.2.6.3
Acute Toxicity (LD50) Test of Extracts
-
-
-
-
61
2.2.7
Studies on the Hot Aqueous Extract of Pleurotustuberregium(PT)
Pleurotustuberregium -
-
and the βGlucan Enriched Polysaccharide Fraction of Pleurotus
tuberregium(BGP)
-
-
-
-
-
-
-
62
2.2.7.1
Studies on Relative Spleen Weight
-
-
-
-
-
62
2.2.7.2
Studies on Phagocytic Index -
-
-
-
-
-
62
2.2.7.3
Studies on Delayed Type Hypersensitivity Response (DTHR)
-
63
2.2.7.4
Studies on Humoral Antibody Response Induced by Ovalbumin
-
63
2.2.8
In vitro Studies
2.2.8.1
Spleenocytes Proliferation Assay
-
-
-
-
-
-
65
-
-
-
-
-
66
2.2.8.2
Effect of BGP on Inducible Nitric Oxide (iNO) Production/Release -
66
2.2.8.3
Effect of BGP on Tumour Necrosis Factor (TNF- Α)
Production /Release -
-
-
-
-
-
-
-
67
2.2.8.4
Effect of BGP on Rate of Phagocytosis of Macrophages
-
-
67
2.9.0
Statistical Analysis
-
-
-
-
-
-
-
68
-
-
-
-
-
-
-
69
CHAPTER THREE: RESULTS
3.1
Extraction
3.2
Phytochemical Analysis of Extract and Fraction
-
-
-
69
3.3
Acute Toxicity Test
-
-
-
69
3.4
Effect of Aqueous Extract of Pleurotustuberregium on Phagocytic Activity in
Mice -
3.5
-
-
-
-
-
-
-
-
-
-
69
-
-
-
-
-
-
-
73
The Effect of Pleurotustuberregium Extracts on Delayed Type Hypersensitivity
response (DTHR)
3.7
-
-
The Effect of Hot Aqueous Extract of P. tuberregium on the Relative Spleen
Weight ofMice
3.6
-
-
-
-
-
-
-
The Effect of Hot aqueous extract (PT) and the β Glucan rich
-
75
x
Polysaccharide Fraction of Pleurotustuberregium(BGP)on Humoral
Antibody Synthesis Induced by Ovalbumin 3.7.1
-
-
-
78
The Effect of Hot of Aqueous Extract of Pleurotustuberregium on
Primary and Secondary Humoral Immune Responses to Ovalbumin
in Mice
3.8
-
-
-
-
-
-
-
-
78
Effect of Βeta (β)-Glucan rich Polysaccharide Fraction of
Pleurotustuberregium (BGP) on Primary and Secondary Humoral immune
Responses to Ovalbumin in Mice
-
-
-
-
-
82
3.9
Invitro studies -
-
-
-
-
-
89
3.9.1
Effect of Βeta (β)-Glucan rich Polysaccharide Fraction of P.
-
-
tuberregium (BGP) on Spleenocytes Proliferation
-
-
-
89
3.9.2
Efect of BGP ON TNF-α Production by RAW264.7 -
-
-
89
3.9.3
Effect of BGP on Nitric Oxide Production by Raw264.7
-
-
90
3.9.4
Effect of BGP on the rate of Phagocytosis (Neutral Red Uptake)
RAW264.7
-
-
-
-
-
-
-
-
90
CHAPTER FOUR: DISCUSSION AND CONCLUSION
4.1
Discussion
-
-
-
-
-
-
-
-
95
4.2
Conclusion
-
-
-
-
-
-
-
-
103
References
Appendices
xi
LIST OF FIGURES
Figure 1: The immune system cells -
-
-
Figure 2: The Fruiting Bodies of Pleurotustuberregium
-
-
-
-
8
-
-
-
-
54
Figure 3: Schematic Representation of the Extraction of Pleurotustuberregium (PT)
and its Beta Glucan Polysaccharide Enriched Fraction (BGP)
-
-
59
Figure 4: The Effect of Hot Aqueous Extract of Pleurotustuberregium(PT) on Phagocytic
Index in Swiss Albino Mice
-
-
-
-
-
-
72
Figure 5: The Effect of Hot Aqueous Extract of P. tuberregium on the Relative Spleen
Weight of Mice
-
-
-
-
-
-
-
-
74
Figure 6: The Effect of Hot Aqueous Extract of Pleurotustuberregium (BGP) on Delayed
Type Hypersensitivity Response in Mice -
-
-
-
-
76
Figure 7: The Effect of Beta (β) GlucanrichPolysacharide Fraction of Pleurotustuberregium
(BGP) on Delayed Type Hypersensitivity in Mice
-
-
-
77
Figure 8: The Effect of Hot Aqueous Extract of P. tuberregium (PT) on Ovalbumin Specific
Total IgG Response in Mice
-
-
-
-
-
-
79
Figure 9: The Effect of Hot Aqueous Extract of P. tuberregium (PT) on ovalbumin-specific
total IgG1 response in mice
-
-
-
-
-
-
80
Figure 10: The Effect of Hot Aqueous Extract of P. tuberregium (PT) on ovalbumin-specific
total IgG2a response in mice
-
-
-
-
-
-
81
Figure 11a: The Effect of Β-Glucan Rich Polysaccharide Fraction of P. tuberregium on
Ovalbumin – Specific Total IgG Primary Response in Mice
-
-
83
Figure 11b: The Effect of Βeta (β) Glucan Rich Polysaccharide Fraction ofP. tuberregium on
Ovalbumin – Specific Total IgG Secondary Response in Mice
-
-
84
Figure 12a: The Effect of Βeta (β) GlucanRich Polysaccharide Fraction ofP. tuberregium on
Ovalbumin – Specific IgG1 Primary Response in Mice -
-
-
85
Figure 12b: The Effect of Βeta (β) Glucan Rich Polysaccharide Fraction of P. tuberregiumon
Ovalbumin – Specific IgG1 Secondary Response in Mice
-
-
86
Figure 13a: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium on
Ovalbumin – Specific IgG2a Primary Response in Mice -
-
-
87
xii
Figure 13b: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium on
Ovalbumin – Specific IgG2a Secondary Response in Mice
-
-
88
Figure 14: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium(BGP)
on Spleenocytes Proliferation
-
-
-
-
-
-
91
Figure 15: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium(BGP)
onTNF-α Production by RAW264.7
-
-
-
-
-
92
Figure 16: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium(BGP)
on Nitric Oxide Production by RAW264.7
-
-
-
-
93
Figure 17: The Effect of Beta Glucan Rich Polysaccharide Fraction of P. tuberregium on Rate of
Phagocytosis by Raw 264.7
-
-
-
-
-
-
94
xiii
LIST OF TABLES
Table 1: Sources and Activity of Cytokines -
-
-
-
-
-
34
Table 2: The Biological Functions of Complement and its Fragments
-
-
37
Table 3: Some Mushrooms with Identified Immunomodulatory Activities -
-
47
Table 4: The Yield and Phytochemical Constituents of Immune Active Extracts and
Fractions of Pleurotustuberregium -
-
-
-
-
-
71
xiv
LIST OF APPENDICES
Appendix 1: The Effect of Hot Aqueous Extract of Pleurotustuberregium (PT) on
Phagocytic Activity in Swiss Albino Mice -
-
-
-
127
Appendix 2: The effect of Hot Aqueous Extract of P. tuberregium on the Relative
Spleen Weight of Mice
-
-
-
-
-
-
-
128
Appendix 3: The Effect of Hot Aqueous Extract of P. tuberregium on Delayed
Type Hypersensitivity Response in Mice
-
-
-
-
129
Appendix 4: The Effect of BGP on Delayed Type Hypersensitivity Response in Mice
130
Appendix 5: The Effect of Hot Aqueous Extract of Pleurotustuberregium (PT) on
Primaryand Secondary IgGHumoral Antibody Response to Ovalbumin Mice 131
Appendix 6: The Effects of Hot Aqueous Extract of Pleurotustuberregium (PT) on
Primaryand Secondary IgG1 Humoral Antibody Response to Ovalbumin in Mice
132
Appendix 7: The Effects of Hot Aqueous Extract of Pleurotustuberregium (PT) on
Primary and Secondary IgG2a Humoral Antibody Response to Ovalbumin in Mice
133
Appendix 8: The Effects of ΒGP on Primary and Secondary Total IgGHumoral
Antibody Response to Ovalbumin in Mice
-
-
-
-
134
-
135
Appendix 9: The Effects of ΒGP on Primary and Secondary Total IgG1 Humoral
Antibody Response to Ovalbumin in Mice
-
-
-
Appendix 10: The Effects of BGP on Primary and Secondary IgG2a Humoral
Antibody Response to Ovalbumin in Mice
-
-
-
-
136
-
-
-
137
-
-
138
Appendix 13: The Effect of BGP on Nitric Oxide Production by RAW264.7
-
139
Appendix 14: The Effect of BGP on Rate of Phagocytosis RAW264.7
-
140
Appendix 11: The Effect of BGP on Spleenocytes Proliferation
Appendix 12: The Effect of BGP on TNF-α Production by RAW264.7
-
xv
CHAPTER ONE
INTRODUCTION AND REVIEW OF LITERATURE
1.1
Scientific Background
The immune system is a network of cells,tissues and organs that work together to defend the body
against attacks by “foreign invaders”. The human body provides an ideal environment for many microbes.
The immune system consists of two categories of defense mechanisms- the innate (non- specific) and the
adaptive (specific) systems (Janeway et al., 2005). Scientists continue to study how the body launches
attacks that destroy invading microbes.
The emergence of acquired immunodeficiency syndrome (AIDS) and the concern about bioterrorism have
also increased the emphasis on the role of the immune system in defending individuals against infections.
The two mechanisms of the immune system can be modified by substances to either enhance or
suppress their ability to resist invasion by pathogens (William, 2001). There are however, limited
strategies available to efficiently modulate the immune response. Nutritional interventions that involve
optimizing the intake of essential nutrients and utilizing promising functional foods have become an
increasingly favoured approach to the modulation of immune cell function. Mushrooms have long been
suggested to possess immunomodulatory properties. More recent studies, however, report that oral
supplementation with a variety of different mushroom species are effective in modulating certain
immune functions.It has also been observed that immunomodulatory substances of natural origin could
play a significant role in human disease prevention and treatment (Dixon, 2001)
Of significant relevance and importance is the ability of particular mushroom-derived compounds to
modulate the human immune response and to inhibit certain tumour growths (Wasser and Weis, 1999a,
1999b). Medicinal mushroom research has focused on discovering compounds that can modulate
xvi
positively or negatively the biologic response of immune cells. Thosecompounds which appear to
stimulate the human immune response are being sought for the treatment of cancer, immunodeficiency
diseases, or for generalized immunosuppression following drug treatment; for combinational therapy
with antibiotics; and as adjuncts for vaccines (Jong and Birmingham, 1992). Those compounds that
suppress immune reactions are potentially useful in the remedy of autoimmune (an abnormal immune
response against self-antigens) or certain gastro-intestinal tract diseases (e.g. Crohns) (Badger,
1983).Compounds that are capable of interacting with the immune system to upregulate or
downregulate specific aspects of the host response can be classified as immunomodulators. One of the
most significant factors of many of the derived bioactive polymers from medicinal mushrooms is their
role as immunomodulators (Yamaguchi, 1992).
P. tuberregium is a popular edible mushroom and has been considered a profound health promoting
mushroom in traditional Chinese medicine (Isikhuemhen et al., 2000a, 2000b; Huang, 2002). It is used in
the preparation of cures for headache, stomach ailments, chest pain, colds and fever, asthma, small pox,
and high blood pressure (Okhuoya and Okogbo, 1991; Fasidi and Olorunmaiye, 1994; Alobo,
2003).Pharmacologically, P. tuberregium polysaccharide attenuates hyperglycemia and oxidative stress in
experimental diabetic rats (Hui-Yuet al., 2012).
For most other species of Pleurotus, several medicinal properties have been reported. They include
properties attributable to their polysaccharides; antigenotoxic, biomutagenic activities (Fillipie and Umek,
2002) anti-inflammatory activity, antilipidemic, antihypertensive and antihyperglycemic activities (Huet
al., 2006), antibacterial and antifungal activities (Hu et al., 2006; Lee et al., 2010;Dahech et al., 2011;
Wong et al., 2011)as well as hepato- and nephro-protective effects (Nworu et al., 2014).
However there is dearth of scientific information on the effect of P. tuber-regium on the immune system.
1.2
Overview of the Immune System
xvii
The architecture of the immune system is multilayered with defenses on several levels. Basically, there
are two divisions of immune protection - the natural (innate/nonspecific) and the adaptive (specific)
mechanisms (Abbas and Lichtman, 2004). Both systems consist of a multitude of cells and molecules that
interact in a complex manner to detect and eliminate pathogens, basically distinguishing between nonself
and self(Matzinger, 1994; Matzinger, 1998). The immune system is closely tied to the lymphatic system.
The term innate immunity is sometimes used to include physical, chemical and microbiological barriers,
but more usually encompasses the elements of the immune system (Neutrophils, Monocytes,
Macrophages, Complement, Cytokines and acute phase proteins) which provide immediate host defense.
The highly conserved nature of the response which is seen in even the smallest animals, confirms its
importance in survival. Adaptive immunity is the hall mark of the immune system of higher animals. Its
response consists of antigen specific reactions through T lymphocytes and B lymphocytes (Delves and
Roitt, 2000).
1.2.1
Organs of the Immune System
Several organs existing in the human body bear responsibility for the synthesis and maturation of
immunocompetent cells. These include the bone marrow, thymus and lymph nodes.
1.2.1.1 Primary Lymphoid Organs
The primary lymphoid organs (bone marrow and thymus)are concerned with production and maturation
of lymphoid cells.
a.
Bone Marrow
xviii
All the cells of the immune system are initially derived from the bone marrow through a process known
as hematopoiesis. During foetal development hematopoiesis occurs initially in yolk sac and para-aortic
mesenchyma and later in the liver and spleen. This function is taken over gradually by the bone marrow.
During hematopoiesis, bone marrow derived stem cells differentiate into either mature cells or into
precursor of cells that migrate out of the bone marrow to continue their maturation in thymus.
The bone marrow produces B cells, natural killer cells, granulocytes and immature thymocytes, in
addition to red blood cells and platelets (Fu and Chaplin, 1999). Other white blood cells produced in the
marrow include neutrophils, basophils, eosinophils, monocytes, and lymphocytes.The bone marrow also
contains antibody secreting plasma cells which have migrated from the peripheral lymphoid tissue.
b.
Thymus
The thymus is a gland located in the anterior mediastinum just above the heart which reaches its greatest
size just prior to birth then atrophies with age. Immature lymphocytes begin to accumulate in the thymus
of human embryos at about 90 -100 days after fertilization. Initially, most of these immature lymphocytes
have come from the yolk sac and foetal liver rather than the bone marrow. Cells from the bone marrow
later migrate to the thymus as precursors and develop into mature thymocytes through a remarkable
maturation process sometimes referred to as “thymus Education”. Mature T cells migrate from the
thymus into the blood stream (Fu and Chaplain, 1999; Abbas and Lichtman, 2004) and some to secondary
lymphoid organs such as lymph node, peyer’s patches and spleen. About 98% of all T cells die in the
thymus.
The greatest rate of T cells production occurs before puberty. After puberty, the thymus shrinks and the
production of new T cells in the adult thymus drops away. Children with no development of the thymus
suffer from Digeorge’s syndrome that is characterized by deficiency in T cell development but normal
number of B cells.
xix
1.2.1.2 Secondary Lymphoid Organ
The secondary lymphoid organ plays an important role in the immune system. The secondary or
peripheral lymphoid organs are the lymph nodes and the spleen. Others are the Tonsils, Peyer’s patch.
They are sites where lymphocytes localize, recognize foreign antigen and mount response against it.
a.
Spleen
The spleen is situated in the upper left quadrant of the abdomen. It is the largest single lymphoid organ
in the body. It is an immunologic filter of the blood. The spleen is ductless, vertebrate gland that is closely
associated with the circulatory system where it functions in the destruction of old reds blood cells in
holding a reservoir of blood. In addition to capturing foreign materials (antigens) migratory macrophages
and dendritic cells, bring antigen to the spleen via the blood stream. An immune response is initiated
when the macrophages or dendritic cells present the antigen to the appropriate B or T cells. In the
spleen, B cells become activated and produce large amounts of antibodies (Abbas and Lichtman, 2004;
USC, 2006).
b.
Lymph Nodes
These are small oval shaped structures located along lymphatic vessels throughout the body. They are
about 1-25mm in diameter. It functions as an immunologic filter for the body fluid known as lymph
composed mainly of T cells, B cells, dendritic cells, and macrophages. The nodes drain fluid from most
body tissues. Antigens are filtered out of the lymph in the lymph node before returning the lymph to the
circulation. In a similar fashion as the spleen, the macrophages and dendritic cells that capture antigens
present these foreign materials to T cells and B cells consequently an immune response is initiated
(Abbas and Lichtmann, 2004; USC, 2006).
c.
Tonsils
xx
The tonsils are often the first organ to encounter pathogens and antigens that come into the body by
mouth or nose. There are basically three pairs of tonsils in a ring above the pharynx. The fundamental
immunological roles of tonsils have yet to be understood. Tonsil size may have a more significant impact
on upper airway obstruction for obese children than those of average weight (Wang et al., 2010).
d.
Peyer’s Patches
This is located in the wall of the intestine and the appendix, attached to the cecum of the large intestine.
It intercepts pathogens that come into the body through the intestinal tract. In adults, lymphocytes
predominate in peyer’s patch. T cell exposed to antigen in peyers patches also migrate into the lamina
propria and the epithelium, where they mature to cytotoxic T cells, providing another mechanism for
containing microbial assaults (Nagler, 2001).
1.2.2
Cells of the Immune System
All cells of the immune system are derived from pluripotent stem cells in the bone marrow by a process
known as haematopoiesis. Under the influence of cytokines, the pluripotent stem cells may become a
lymphoid stem cell or a myeloid stem cell. The lymphoid stem cell develops into B cells and T cells.
Natural killer (NK) cells are lymphocytes that act in a similar manner to cytotoxic T cells. These cells,
however, are not T cells (USC, 2006). The lineage of the Nk cells is not well understood. The myeloid stem
cell develops into platelets, red blood cells or the granulocyte-monocyte line. Monocytic cells include
monocytes and macrophages.
The granulocytes include neutrophils, eosinophils, mast cells and basophils. Monocytes circulate in the
blood and lymphatics but become macrophages when they enter the tissues. The development of these
cells from a stem cell involves cytokine growth factors and the development of a progenitor cell in each
line. The progenitor cell is a committed cell, meaning that it is committed to that line of cell growth (A
xxi
neutrophil progenitor must become a neutrophil, it cannot become an eosinophil for instance) (USC,
2006)(Figure 1).
xxii
Figure 1: The immune system cells
xxiii
1.2.2.1 Lymphoid Stem Cells
a.
B Cells/Lymphocytes
The development and maturation of this cell group occurs in the bone marrow. In mammals, the early
stage of Bcell maturation occurs in the fetal liver and the bone marrow. B cell development begins in the
fetal liver and continues in the bone marrow throughout life. They account for 5 -15% of lymphocytes in
circulation and 80 -90% in bone marrow, 20% -30% in lymph node and 50 -60% in the spleen. The most
important surface marker on the surface of mature B cell is the surface immunoglobulin usually of IgM
and IgD type(Roitt and Delves, 2001).
The major function of B lymphocytes is the production of antibodies in response to foreign proteins of
bacteria, viruses and tumor cells. Antibodies are specialized proteins that specifically recognize and bind
to one particular protein. Antibody production and binding to a foreign substance or antigen, often is
critical as a means of signaling other cells to engulf, kill or remove that substance from the body (Roitt
and Delves, 2001). B cells may differentiate into plasma cells (which secrete large amounts of antibodies)
or into memory B cells. Memory cells can survive 20years or more. Plasma cells are the effector cells of
the B cell lineage and are specialized in secreting immunoglobulin. When activated, B cells divide; some
of its progeny become memory cells and the remainder becomes immunoglobulin- secreting plasma cells
(Allman et al., 2004).
b.
T Cells
T cell is an abbreviation of “thymus dependent lymphocytes”. T lymphocytes arise in the bone marrow as
T cell precursors then migrate to and mature in the thymus (A lymphoid organ beneath the breast bone).
After entry into the thymus, the T cell precursorare also referred to as the “thymocytes”. T cells express
different receptors on the surface (T cell receptors) which is followed by expression of either CD8 or CD4
xxiv
surface molecule. The T cells accounts for 70-80% lymphocytes in peripheral blood, 5-10% in bone
marrow, 70-80% in lymph node and 30-40% in spleen.
T lymphocytes have two major subsets that are functionally and phenotypically different; Helper (CD4)
and cytotoxic/suppressor (CD8) T cells (Jacqueline and Bryon, 2001).
Helper T cells (Th) also called CD4+ T cell is a pertinent coordinator of immune regulation. The main
function of the T helper cell is to augument or potentiate immune responses by the secretion of
specialized factors (cytokines) that activate and promote the proliferation and differentiation of other
white blood cells; cytotoxic T cell, B cells and macrophages and activation of inflammatory leucocytes.
They are identified by the presence of the CD4 marker. They recognize antigen when presented along
with class 11 MHC molecules. Based on the kind of cytokines produced T lymphocytes (Th) are further
divided into Th1 and Th2 subsets(Jacqueline and Bryony, 2001).
Cytotoxic /T killer/ suppressor subset (TC) or CD8+ T cells are important in directly killing certain tumor
cells, viral infected cells and sometimes parasites. They lyse cells with foreign antigens. They are
identified by the presence of the CD8 Marker. They recognize antigen when presented along with class 1
MHC molecules(Jacqueline and Bryony, 2001).
The suppressor T cells have a role in the downregulation of immune response. Both types of T cells can
be found throughout the bodies. They often depend on the secondary lymphoid organs (the lymph nodes
and spleen) as sites where activation occurs but they are also found in other tissues of the body, most
conspicuously the liver, lung, intestinal and reproductive tracts (USC, 2006).
c.
Non Specific Killer Cells
xxv
Several different cells including NK and LAK cells, K cells, activated macrophages and eosinophils are
capable of killing foreign and altered self target cells in a non-specific manner. These cells play an
important role in the innate immune system (Abbas and Lichtman 2004; USC, 2006).
1.2.2.2 Myeloid Stem Cells
The myeloid stem cells develop into platelets, red blood cells or granulocyte-monocyte line. Monocytic
cells include monocytes and macrophages. The granulocytes include neutrophils, eosinophils, mast cells
and basophils.
a.
Granulocyte or Polymorphonuclear (PMN) Leucocytes
Another group of white blood cells is collectively referred to as granulocytes or polymorphonuclear
(PMNs) leucocytes. These granulocytes are so called because they have irregularly shaped nuclei.
Granulocytes are composed of three cell types identified as neutrophils, eosinophils and basophils based
on their staining characteristics with certain dyes. These cells are predominantly important in the
removal of bacteria and parasites from the body. They engulf foreign bodies and degrade them using
powerful enzymes (Peakman and Vergani, 1997).
i.
Neutrophils
These cells have short life span. They circulate in the blood for 6-7 hours then migrate through the
endothelial cell functions and reside in tissue spaces where they live for only for a few days and do not
multiply. Neutrophils are the most abundant of the leukocytes normally accounting for 54-75% of the
white blood cell. They are called neutrophils because their granules stain poorly with the mixture of dyes
used in staining leucocytes. They are effector cells of innate immunity that enter sites of infection and
work in anaerobic conditions. The content of neutrophils granules bind to neitheracidic nor basic stains.
Neutrophils phagocytose and kill microbes upon phagolysosomal fusion (Urban et al., 2006).
xxvi
ii. Eosinophils
These granulocytes are the second most abundant, 1-6% of proportion of leucocytes. They defend
against helminthes worms and other intestinal parasites. They also stimulate the release of cytokines
such as IL-1, IL-2, IL-4, IL-5, IL-6, IL-8, IL-13 and TNF alpha (α)(Rothenberg and Hogan, 2006; Hogan et al.,
2008). The eosinophilic granules contain basic substances that bind to the acidic stain eosin. Eosinophils
bind to the antibody using low affinity receptors(FCER11). They are not phagocytic (Jacqueline and
Bryony, 2001).
iii. Basophils:
It constitutes less than 1% of the proportion of leucocytes making them the least abundant. It is also
implicated in regulating the immune response to parasites and allergy but is so rare that relatively little is
known of its contribution to immune defense. The basophil cytoplasmic granule contains acidic substance
that binds basic stains such as hematoxylin during histological stains. They are involved in some of the
most severe immunologic reactions such as angioedema and anaphylaxis (Jacqueline and Bryony, 2001).
b.
Macrophages
Macrophages originate from the monocytes. They function both in innate and adaptive immunity. Once
monocyte leaves circulation and enter tissue, they are called macrophages. They are distinguished from
the granulocytes by being bigger, having a distinctive indented nucleus. The monocytes are the mobile
progenitors of sedentary tissue cells called macrophages. They travel in the blood to tissues, where they
mature into macrophages and take up residence. Tissue macrophages are large, irregularly shaped cells
characterized by an extensive cytoplasm with numerous vacuoles often containing engulfed materials.
They survive for months and can multiply(Mosser and Edwards, 2008).
xxvii
There are two types of macrophages: one that wanders in the tissue spaces and the other that are fixed
to vascular endothelium of liver, spleen, lymph node and other tissue. Macrophages display remarkable
plasticity and can change their physiology in response to environmental cues. These changes can give rise
to different populations of cells with distinct function (Mosser and Edwards, 2008). Macrophages present
in different organs have been given different names. They are histiocytes (in tissues), kupffer cells (in
liver), alveolar macrophages (in lungs), peritoneal macrophages (in peritoneum), microglial cells (in
brain), mesangial cells (in kidneys) and osteoclastis (in bone). Macrophages kill microbes, infected cells,
tumor cells; secrete immunolodulatory cytokines, antigen processing and presentation to T cells.
Macrophages respond to infections as quickly as neutrophils but persist much longer. Hence they are
dominant effector cells at the later part of infections. They are also often referred to as scavengers or
antigen presenting cells (APC) because they pick up and ingest foreign materials and present these
antigens to other cells of the immune system such as T cells and B cells. This is one of the important first
steps in the initiation of an immune response. Stimulated macrophages exhibit increased levels of
phagocytosis and are also secretory (Abbas and Lichtman, 2004; USC, 2006).
c.
Dendritic Cells
They are also known as antigen presenting cells (APC) and are more efficient APC’s than the
macrophages. These are efficient stimulators of B and T lymphocytes (Jacques and Ralph, 1998). B cells,
the precursor of antibody secreting cells, can directly recognize native antigen through their B cells
receptors. T lymphocytes however need the antigen to be processed and presented to them by an APC.
Dendritic cells in the periphery capture and process antigens, express lymphocyte co-stimulatory
molecules, migrate to lymphoid organs and secrete cytokines to initiate immune responses. They not
only activate lymphocytes, they also tolerize T cells to antigens that are innate to the body (self antigens),
thereby minimizing autoimmune reactions (Jacque and Ralph, 1998). Not much is known about dendritic
xxviii
cells because they are extremely hard to isolate which is often a prequisite for the study of functional
qualities of specific cell types. Of particular importance is the recent finding that dendritic cells bind high
amount of HIV and may be a reservoir of virus that is transmitted to CD4+ T cells during an activation
event (Janeway et al., 2005; USC, 2006).
1.2.3
Arms of the Immune System
There are two major subdivisions of the immune system; the innate or non specific immune system and
the adaptive or specific immune system.
The innate immune system is the body’s first line of defense against invading organisms while the
adaptive immune system act as second line of defense and also affords protection against re-exposure to
the same pathogen. Each of the major sub divisions of the immune system acts as second line of defense
and also affords protection against re-exposure to the same pathogen. Each of the major subdivisions of
the immune system has both cellular and humoral components by which they carry out their protective
function. There is usually interplay between the two arms of the immune system(Abbas and Lichtman,
2004).
1.2.3.1 Innate Immunity
This is also known as non-specific immune system and is the first line of defense (Janeway et al., 2005). It
does not confer a long lasting or protective immunity to the host (Alberts et al., 2002). The elements of
non specific (innate) immune system include anatomical barriers, secretory molecules and cellular
components.
A.
Anatomical Barriers to Infection
xxix
The skin, external epithelial layers, the movement of the intestines and the oscillation of bronchopulmonary cilia are the mechanical anatomical barriers (Roitt et al., 2001). Associated with these
protective surfaces are chemical and biological agents.
i.
Mechanical Factors
The epithelial surfaces form a physical barrier that is very impermeable to most infectious agents. Thus
the skin acts as our first line of defense against invading organisms. The regular desquamation of skin
epithelium also helps remove bacteria and other infectious agents that have adhered to the epithelial
surfaces (Goldsby et al., 2000). Movement due to cilia or peristalsis helps to keep air passages and the
gastrointestinal tract free from microorganisms. The flushing action of tears and saliva helps prevent
infection of the eyes and mouth. The trapping action of mucus that lines the respiratory and
gastrointestinal tracts helps to protect the lungs and digestive systems from infection.
ii.
Chemical Factors
Fatty acids in sweat inhibit the growth of bacteria. Lysozyme and phospholipase found in tears, saliva and
nasal secretions can breakdown the cell wall of bacteria and destabilise bacterial membranes. The low pH
of sweat and gastric secretions prevents growth of bacteria. Defensins (low molecular weight proteins)
found in the lung and gastrointestinal tract have antimicrobial activity. Surfactants in the lung act as
opsonins (substances that promote phagocytosis of particles by phagocytic cell) (USC, 2006).
xxx
iii.
Biological Factors
The normal flora of the skin and in the gastrointestinal tract can prevent the colonization of pathogenic
bacteria by secreting toxic substances or by competing with pathogenic bacteria for nutrients or
attachment to cell surfaces.
B.
Humoral Barriers to Infection
The anatomical barriers are very effective in preventing colonization of tissues by microorganisms.
However when there is damage to tissues, the anatomical barriers are breached and infection may occur.
Once infectious agents have penetrated tissues, another innate defense mechanism comes into play,
namely acute inflammation. Humoral factors play an important role in inflammation, which is
characterized by edema and the recruitment of phagocytic cells. These humoral factors are found in
serum or they are formed at the site of infection.
i. Complement System
The complement system is the major humoral non specific defense mechanism. Once activated,
complement can lead to increased vascular permeability, recruitment of phagocytic cells, lysis and
opsonisation of bacteria (Goldsby et al., 2000; USC, 2006).
ii. Coagulation System
Depending on the severity of the tissue injury, the coagulation system may or may not be activated.
Some products of the coagulation system contribute to the non specific defenses because of their ability
to increase vascular permeability and act as chemotactic agents for phagocytic cells. In addition, some of
the products of the coagulation system are directly antimicrobial. For example, beta-lysin, a protein
produced by platelets during coagulation can lyse many gram positive bacteria by acting as cationic
detergent (Goldsby et al., 2000; USC, 2006).
xxxi
iii. Lactoferrin and Transferrin
Lactoferrin is a 703- amino acid glycoprotein originally isolated frommilk. Plasma lactoferrin is
predominantly neutrophil derived but indications are that it may also be produced by other cells (Levay
and Viljoen, 1995). By binding iron, an essential nutrient for bacteria, these proteins limit bacteria
growth.
iv. Interferons
Interferons are protein that can limit virus replication in cells. They are a major class of cytokine that have
a role in immunity. They are so called because they inhibit viral replication but they have many other
functions. For instance, they also activate NK cells to kill virus infected host cells. They are divided into
two: type 1(α and β interferons) and type 2(ɣ or immune interferon). Type 1 interferons have potent
antiviral activity and are produced mainly by fibroblast and monocytes as a reaction to infection. They
also have antiproliferative function and useful in the treatment of chronic hepatitis B and C infections in
combination with antiviral drugs (Hoofnagle and Di, 1997) as well as in some forms of leukeamia
(Brustein and McGlave, 2001; Minet al., 2001).
v. Lysozyme
Lysozyme is an enzyme found in the protective fluid (tears, saliva and mucus) of most animals. Lysozyme
breaks down the cell wall of bacteria.
vi. Interleukin 1
More than any other cytokine family, the interleukin (IL) – 1 family is closely linked to the innate immune
response (Dinarello, 2009). IL -1 beta is the most studied member of the IL-1 family because of its role in
mediating autoinflammatory diseases. IL 1 induces fever and the production of acute phase proteins,
some of which are antimicrobial because they can opsonise bacteria.
xxxii
C.
Cellular Barriers to Infection
Part of the inflammatory response is the recruitment of polymorphonuclear cells, eosinophils and
macrophages to sites of infection. These cells are the main line of defense in the non-specific immune
system. Polymorphonuclear neutrophils (PMNs) are recruited to the site of infection where they
phagocytose invading organisms and kill them intracelllularly. In addition, PMNs contribute to collateral
tissue damage that occurs during inflammation(USC, 2006).
Tissue macrophages and newly recruited monocytes which differentiate into macrophages also function
in phagocytosis and intracellular killing of microorganism. In addition, macrophages are capable of
extracellular killing of infected or altered self-target cells. Furthermore, macrophages contribute to tissue
repair and act as antigen presenting cells, which are required for the induction of specific immune
responses (USC, 2006).
Natural killer (NK) and lymphokine activated killer (LAK) cells can non-specifically kill virus infected and
tumor cells. These cells are not part of the inflammatory response but they are important in nonspecific
immunity to viral infections and tumor surveillance. Eosinophils have proteins in their granules that are
effective in killing certain parasites.
D.
Phagocytosis and Intracellular Killing
Polymorphonuclear leucocytes (PMNs) are motile phagocytic cells that have lobed nuclei are identified by
their characteristic nucleus or by an antigen present on the cell surface called CD66 (Goldsby et al., 2000;
USC, 2006). They contain two kinds of granules, the content of which are involved in antimicrobial
properties of these cells. The primary or azurophilic granules which are abundant in young newly formed
PMNs contain cationic proteins and defensins that can kill bacteria, proteolytic enzymes like elastase and
cathepsin G to break down proteins, lysozyme to break down bacteria cell walls and characteristically,
xxxiii
myelopreoxidase, which is involved in the generation of bacteriocidal compounds (Goldsby et al., 2000;
USC, 2006).
The second type of granule found in more mature PMNs is the secondary or specific granule. These
contain lysozyme, NADPH oxidase components which are involved in the generation of toxic oxygen
products and characteristically lactoferrin an iron chelaring protein and B12- binding protein (USC, 2006).
Monocytes and macrophages are phagocytic cells that have characteristic kidney shaped nucleus. Unlike
PMNs they do not contain granules but they have numerous lysosomes which have contents similar to
the PMNs granules. Phagocytosis occurs through different steps and processes as shown below
i.
Circulating PMNs and monocytes respond to danger signals generated at the site of an infection.
The signals include N-formyl methionine containing peptides released by bacteria, clotting
system peptides, complement products and cytokines released from tissue macrophages that
have encountered bacteria in tissue (Bona and Bonilla 1995; Goldsby et al., 2000). Some of the
signals stimulate endothelial cells near the site of the infection to express cell adhesion molecules
such as ICAM 1 and selectins which bind components on the surface of phagocytic cells and cause
the phagocytes to adhere to the endothelium (Goldsby et al., 2000; USC, 2006). Vasodilators
produced at the site of the infection causes the junctions between endothelial cells to loosen and
the phagocytes then cross the endothelial barrier by squeezing between the endothelial cells in a
process known as diapedesis.
ii
Phagocytosis: phagocytic cells have a variety of receptors on their membrane through which
infectious agents bind to cells. These include Fc receptors, complement receptors, scavenger
receptors and toll like receptors. Bacteria with IgG antibody on their surface have the Fc region
exposed and this part of the Ig molecule can bind to the receptor on phagocytes. Binding to the
Fc receptor requires prior interaction of the antibody with the antigen. Binding of the IgG coated
xxxiv
bacteria to Fc receptors results in enhanced phagocytosis and activation of the metabolic activity
of phagocytes (respiratory burst)(Goldsby et al., 2000; USC, 2006). Phagocytic cells have a
receptor for the 3rd component of the complement C3b. Binding of C3b coated bacteria to this
receptor also results in enhanced phagocytosis and stimulation of the respiratory burst.
Scavenger receptors bind a variety of polyanions on bacterial surfaces resulting in phagocytosis
of bacteria. Phagocytes have a variety of Toll like receptors (pattern recognition receptors or
PRRs) which recognize broad molecular patterns called PAMPS (pathogen associated molecular
patterns) on infectious agents. Binding of infectious agents via Toll like receptors results in
phagocytosis and the release of inflammatory cytokines (IL-1, TNF-alpha, IL-6) by the phagocytes
(Peakman and Vergani, 1997;Goldsby et al., 2000). After attachment of a bacterium, the
phagocytes begin to extend pseudopods around the bacterium. The pseudopods eventually
surround the bacterium and engulf it and the bacterium is enclosed in a phagosome. During
phagocytosis the granules or the lysosomes of the phagocyte fuse with the phagosome and
empty their contents. The result is a bacterium in a phagolysosome which contains the contents
of the granules or lysosomes (Roitt and Delves, 2001; USC, 2006).
iii.
Respiratory burst: During phagocytosis, there is an increase in glucose and oxygen
consumption. This is referred to as respiratory burst. The consequences of the respiratory
burst is that a number of oxygen containing compounds are produced which kill the
bacteria being phagocytosed. This is referred to as oxygen dependent intracellular killing.
In addition, bacteria can be killed by preformed substances released from granules or
lysosomes when they fuse with the phagosome. This is referred to as oxygen independent
intracellular killing (USC, 2006).
a.
Oxygen Dependent Myeloperoxidase Independent Intracellular Killing
xxxv
During phagocytosis, glucose is metabolized via the pentose monophosphate shunt and NADPH is
formed (Janeway et al., 2005). Cytochrome B which was part of the specific granule combines
with the plasma membrane NADPH oxidase and activates it. The activated NADPH oxidase uses
oxygen to oxidize the NADPH. The result is the production of superoxide ions. Some of the
superoxide ions is converted to H2O2and singlet oxygen by superoxide dismutate. In addition,
superoxide ions react with the H2O2 resulting in the formation of hydroxyl radical and more
singlet oxygen.
The result of these reactions is the production of the toxic oxygen compounds superoxide
anion(O2-), H2O2, singlet oxygen(1O2) and the hydroxyl radical (OH+) (Roitt et al., 2001; USC,
2006).
b.
Oxygen Dependent Myeloperoxidase Dependent Intracellular Killing
As the
azurophilic granules fuses with the phagosome, myeloperoxidase is released into the
phagolysosome. Myeloperoxidase utilizes H2O2 and the halide ions (usually Cl-) to produce
hypochlorite a highly toxic substance. Some of the hypochlorite can spontaneously breakdown to
yield toxic substance. Some of the hypochlorite can spontaneously break down to yield singlet
oxygen. The result of these reactions is the production of toxic hypochlorite (OCl-) and singlet
oxygen (1O2) (Roitt and Delves, 2001; USC, 2006).
c.
Detoxification Reaction
PMNs and macrophages have means to protect themselves from the toxic oxygen intermediates.
These reactions involve the dismutation of superoxide anion to hydrogen peroxide by superoxide
dismutase and the conversion of hydrogen peroxide to water by catalase (USC, 2006).
d.
OxygenIndependent Intracellular Killing
xxxvi
In addition to the oxygen dependent mechanisms of killing there are also oxygen independent
killing mechanisms in phagocytes: cationic proteins (cathepsin) released into the phagolysosome
can damage bacterial membranes: lysosome breaks down bacterial cell walls, lactoferrin chelates
iron, which deprives bacteria of this required nutrient: hydrolytic enzymes breakdown bacterial
proteins. Thus, even patients who have defects in the oxygen dependent killing pathways are
able to kill bacteria. However since the oxygen dependent mechanisms are much more efficient
in killing, patients with defects in these pathways are more susceptible and get more serious
infections (USC, 2006).
e.
Nitric Oxide Dependent Killing
Binding of bacteria to macrophages particularly binding via Toll like receptors results in the
production of TNF –alpha, which acts in an autocrine manner to induce the expression of the
inducible nitric oxide synthetase gene resulting in the production of nitric oxide (NO). If the cell is
also exposed to interferon gamma (IFN-gamma) additional nitric oxide will be produced. Nitric
oxide released by the cell is toxic and can kill microorganisms in the vicinity of the macrophage
(Roitt and Delves, 2001; USC, 2006).
1.2.3.2 Adaptive Immunity
Adaptive immunity is another major subdivision of the immune system. It is the hall mark of the immune
system of higher animals. The adaptive immune system evolved in early vertebrates. It allows for a
stronger immune response as well as immunological memory where each pathogen is remembered by a
marker antigen (Pancer and Cooper, 2006). Adaptive immunity is so called because it adapts or learns to
recognize specific kinds of pathogens and retains memory of them for speeding up future responses. It is
xxxvii
the second barrier against infections. It is acquired later in life and is precise. The responseconsists of
antigen specific reactions through T lymphocytes and B lymphocytes. The adaptive response has memory
so that subsequent exposure leads to a more vigorous and rapid response but this is not immediate
(Delves and Roitt, 2001).
The cells of the adaptive immune system are special types of leucocytes, lymphocytes, B cells, T cells that
are derived from heamopoeitic stem cell in the bone marrow. There are two basic components of the
adaptive immune system; the cell mediated immunity and the humoral component of the adaptive
immunity. B cells are involved in the humoral immune responses whereas T cells are involved in the cell
mediated immune response (Janeway et al., 2005). Both T and B cells carry receptor molecules that
recognize specific targets.
A.
Cell Mediated Immunity
Adaptive T cell mediated immunity is driven by activation of T cells. Cytotoxic T cell is activated
byendogenous antigen to stimulate macrophage killing of endosomal pathogens. The pathogens targeted
by cellular immunity are protected from antibody and complement binding by their intracellular
locations. T cells recognize a non-self-target such as a pathogen only after antigen (small fragments of the
pathogen) have being processed and presented in combination with a self-receptor called a major
histocompactibility complex(MHC) molecule.
There are two major subtypes of T cells; the cytotoxic T cells (killer T cells) and the helper T cell. Killer T
cells only recognize antigens coupled to class 1 MHC molecules while helper T cells only recognize
antigens coupled to class 11 MHC molecules. These two mechanisms of antigen presentation reflect the
different roles of the two major types of T cells. Only professional antigen presenting cells (dendritic cells,
macrophages and B cells) have both class 1 and class 11 MHC and can deliver co stimulatory signals.
xxxviii
Activated T cells perform their effector functions when they encounter MHC presented peptide on their
target cells.
A third major subtype is the ɣð T cells that recognize intact antigens that are not bound to MHC receptors
(Girardi, 2006).
i.
Helper T Cells (Th)
The helper T cells regulate both innate and adaptive immune responses and help determine which types
of immune response the body will make to a particular pathogen (Abbas et al., 1996). These cells have no
cytotoxic potentials and do not kill infected cells or clear pathogens directly. They control the immune
response by directing other cells to perform these tasks. Helper T cells express T cell receptors (TCR) that
recognize antigen bound to class 11 MHC molecules. Helper T cells require long duration of engagement
with an antigen presenting cell (Kovacs et al.,2002).
Th cells are subdivided functionally by the pattern of cytokines they produce. On stimulation, precursor
Th O lymphocytes become either Th1 or Th2 cells. The difference between these cells is only in the
cytokines secreted. They are morphologically indistinguishable. However the response they generate is
very different. Th1 cells produce interleukin 2 which induces T cell proliferation. Interleukin 2 stimulates
CD8 T cell division and cytotoxicity by decreasing activation threshold. The other major cytokine
produced by Th1 cells, interferon ɣ actiavtes macrophages to kill intracellular pathogens such as
mycobacteria, fungi and protozoan and induces natural killer cells to cytotoxicity.
The Th1 cytokines therefore induces mainly a cell mediated inflammatory response.There is a positive
feedback loop as interferon ɣ stimulates other Th 0 cells to become Th 1 and inhibits Th 2 differentiation
(Swain et al., 1991). The activation of a resting helper T cell causes it to release cytokines that influence
xxxix
the activity of many cell types. Cytokine signal produced by helper T cells enhance the microbiocidal
function of macrophages and the activity of killer T cells (Albertset al., 2002).
ii.
Cytotoxic T Cells (Tc)
Cytotoxic T cells (killer T cells) mediate antigen specific, MHC restricted cytotoxicity and are important for
killing intra-cytoplasmic parasites that are not accessible to secreted antibody or to phagocytes. Examples
include all viruses, rickettsias, some obligate intracellular bacteria (Chlamydia) and some
protozoanparasites which export their proteins from macrophage vesicles to the cytoplasm (Toxoplasma
gondii). The only way to eliminate these pathogens is to kill their host cells (Harty et al., 2000). As with B
cells each type of T cell recognizes a different antigen. Killer T cells are activated when their T cell
receptor (TCR) binds to this specific in a complex with the MHC class 1 receptor of another cell.
Recognition of this MHC antigen complex is aided by a co-receptor on the T cell called CD8. The T cell
then travels throughout the body in search of cells where the MHC 1 receptor bears this antigen.
When an activated T cell contacts such cells it releases cytotoxins that form pores in the target cells
plasma membrane allowing ions, water and toxins to enter. This causes the target cell to undergo
apoptosis (Radoja et al., 2006). T cell killing of host cell is particularly important in preventing the
replication of viruses. T cell activation is tightly controlled and generally requires a very strong MHC
antigen activation signal, or additional activation signals provided by helper T cells (Radoja et al., 2006).
iii.
γgamma T Cells
γ gamma T Cells posses an alternative T cell receptor(TCR) as opposed to CD4+ and CD8+ T cells and share
the characteristics of helper T cells, cytotoxic T cells and NK cells. The conditions that produce responses
from γ gamma T cells are not fully understood. Like other unconventional T cells subsets bearing invariant
TCRs such as CD1d- restricted natural killer cells, γ gamma T cells straddle the border between innate and
xl
adaptive immunity (Girardi, 2006). On the other hand, γ gamma T cells are a component of adaptive
immunity as they rearrange TCR genes to produce receptor diversity and can also develop a memory
phenotype. On the other hand, the various subsets are also part of the innate immune system, as
restricted TCR or NK receptors may use as pattern recognition receptors(Girardi, 2006).
B.
Humoral Immunity
This is also referred to as antibody mediated immunity. It is regulated by B cells and the antibodies they
produce. Antibody mediated reactions defend against invading viruses and bacteria. Antibodies are
produced and secreted by plasma cells which are found mainly within lymph node and which do not
circulate. Plasma cells are derived from B lymphocytes. Adaptive humoral immunity also usually involves
T cell activation to produce cytokines that stimulate B cell antibody synthesis (Janeway et al., 2005).
1.2.4
Overview of Antibodies
Antibodies (immunoglobulins abbreviated Ig) are proteins of molecular weight 150,000-900,000kd
(Janeway et al., 2005; Abbas and Lichtman, 2004). They are also referred to as gammaglobulins.
Antibodies vary from molecule to molecule with respect to which organism they bind to or with their
specialized function in the body. Antibodies have a ‘Y’ shaped structure with three different regions. The
arms of ‘Y’ are termed the variable regions while the tail of ‘Y’ is the constant region. The variable
regionsare randomly generated so that they bind to specific pathogen epitopes(Janeway et al., 2005).
The constant region on the other hand is not randomly generated but comes in a few structural varieties
called isotypes.
The constant region is the part of the antibody to which other immune system cells such as macrophages
bind. Depending on the isotype, of the constant region, different responses will be triggered upon
binding, so that it is this part of the antibody that determines effect function. One end of the
xli
immunoglobulin binds to antigens (Fab portion so called because it is the fragment of the molecule which
is antigen binding) and the other end which is crystallizable and therefore called Fc is responsible for
effector function. The immunoglobulin molecule consists of two heavy chains and two light chains each
of approximate molecular weight 50000kd (Abbas and Lichtman, 2004).
The heavy and light chain consist of amino acids sequences. In the regions concerned with antigen
binding these regions are extremely variable, whereas in other regions of the molecule, they are
relatively constant. Thus every heavy and light chain possesses a variable and constant region.
The isotype of an Ig is determined by the constant region. There are five classes (isotypes) of
immunoglobulins; IgM, IgG, IgA, IgD, and IgE plus four subtypes of IgG (Ig G1-4) and two of IgA (IgA1,
IgA2). Immunoglobulins are found in serum and in secretions from mucosal surfaces (Abbas and
Lichtman, 2004).
1.2.4.1 Classes of Immunoglobulin
When a B cell encounters the kind of antigen that triggers it to become active, it gives rise to many large
cells known as plasma cells which produce antibodies (Pier et al., 2004). Each immunoglobulin class has a
special characteristic that provides it with special advantage:
i)
Immunoglobulin G (IgG): is a kind of antibody that works efficiently to coat microbes speeding
their uptake by other cells in the immune system. It is formed in large quantity and last for over a
month and travel from the blood stream to tissue easily. It is the only immunoglobulin class that
crosses the placenta and passes immunity from the mother to the new born (Pier et al., 2004).
xlii
ii)
Immunoglobulin M (IgM): They are the first antibodies formed in response to infection. They are
important in protection during the early days of an infection. IgM are the most effective against
bacterial infection (Pier et al., 2004).
iii)
Immunoglobulin A (IgA): This immunoglobulin is produced near the mucus membrane and find its
way to the saliva, mucus and digestive tract- guarding the entrance to the body and giving
protection against infection in the respiratory tract and intestines.
iv)
Immunoglobulin E (IgE):The natural job of this immunoglobulin is to protect against parasitic
infections and is responsible for allergic reactions (Pier et al., 2004).
v)
Immunoglobulin D (IgD): They are present in the surface of most but not all B cells. Early in their
development but little IgD is ever released into the circulation. It is not clear what function Ig D
performs though it may play a role in determining whether antigen activate the B cell.
1.2.4.2
Roles of Antibodies
Antibodies exist free in body fluids eg serum, and membrane bound to B lymphocytes. Their function
when membrane boundis to capture antigen for which they have specificity, after which the B
lymphocytes will take the antigen into its cytoplasm for further processing and presentation. Free
antibodies perform the functions listed below (Roitt et al., 2001; USC, 2006).
i)
Agglutination of particulate matter, including bacteria and virus. IgM is particularly suitable for
this as it is able to change its shape from a star form to a form resembling a crab.
ii)
Opsonisation (i.e coating) of bacteria for which the antibody’s Fab region has specificity
(especially IgG). This facilitates subsequent phagocytosis by cells possessing Fc receptor e.g.
xliii
neutrophil polymorphonuclear, leucocyte, (polymorphs). Thus it can be seen that in opsonisation
and phagocytosis, both the Fab and the Fc portions of immunoglobulin molecules are involved.
iii)
Neutralization: neutralization of toxin released by immunoglobulin G (eg tetanus toxin) is
neutralized when specific IgG antibody binds, thus preventing the toxin binding to motor end
plate and causing persistent stimulation, manifest as sustained muscular contraction, which is the
hall mark of titanic spasm). In the case of viruses antibody can hinder the ability to attach to
receptors on host cells. Here, only Fab is involved.
iv)
Immobilization of bacteria: antibodies against bacteria ciliae or flagella will hinder their
movement and ability to escape the attention of phagocytic cells. Again only Fab is involved.
v)
Activation of complement (classical pathway) especially by the Fc region of IgM and IgG, leads
eventually to death of bacteria by the terminal complement component which punch holes in the
cell wall leading to an osmotic death. Complement component also facilitate phagocytosis by
cells possessing a receptor for C3b e.g. polymorphs.
vi)
Mucosal protection: This is provided mainly by IgA and to a lesser degree IgG. IgA acts mainly by
inhibiting pathogens from attaching to mucosal surfaces. This function is attributed to Fab.
vii)
Expulsion as a consequence of mast cell degranulation: As a consequence of antigen e.g. parasitic
worms, binding to specific immunoglobulin E, attached to mast cell by their receptor for IgE Fc,
there is release of mediators from the mast cell. This leads to contraction of smooth muscles
which can result in diarrhoea and expulsion of parasite. Here we see involvement of both Fab
parasite antigen and Fc anchoring the reacting participant.
viii)
Precipitation of soluble antigen by immune complex formation: This consists of antigen linked to
antibody. Depending on the ratio of antigen to antibody, these can be of varying size. When fixed
at one site they can be removed by phagocytic cells. They may also circulate prior to localization
and removal, and can fix complement. Here Fab and Fc involved.
xliv
ix)
Antibody dependent cell mediated cytotoxicity (ADCC): Antibodies bind to organisms via their
Fab region. Large granular lymphocytes (Natural killer cells - NK cells), attach via Fc receptors and
kill these organism not by phagocytosis but by release of toxic substances called perforins.
x)
Conferring immunity to the foetus by the transplantal passage of IgG: IgG is the only class
(isotope) of immunoglobulin that can cross the placenta and enter the foetal circulation where it
confers passive immune protection. This is of great importance to the foetus in the first 3
months.
1.3
Mediators of the Immune System
1.3.1
Cytokines
Cytokines are small molecular weight messengers secreted by cell to alter the behavior of itself (Jacquelin
and Bryony, 2001). Cytokines are a group of proteins and peptides that are used as signaling compounds
in the body(Lee et al., 2004). These chemical signals are similar to hormones and neurotransmitters and
are used to allow one cell to communicate with another. The cytokine family consists of smaller water
soluble proteins and glycoproteins with a mass of 8-30kDa (Roitt et al., 2001; USC, 2006). They are
particularly important in both innate and adaptive immune responses.
Due to their central role in the immune system, cytokines are involved in a variety of immunological,
inflammatory and infectious diseases. Cytokines are produced by a wide variety of cell types (both
haemopoeitic and non haemopoeitic) and can have effects on both nearby cells or throughout the
organism. Sometimes these effects are strongly dependent on the presence of other chemicals and
cytokines(Leeet al., 2004)(Table1).
Each cytokine binds to a specific cell surface receptor. Subsequent cascades of intracellular signaling then
alter functions. This may include the upregulation and / or downregulation of several genes and their
transcription factors in turn resulting in the production of other cytokines, an increase in the number of
xlv
surface receptors for other molecules or the suppression of their own effect by feedback inhibition (Roitt
and Delves, 2001; Janeway et al., 2005). Interestingly, cytokines are characterized by considerable
“redundancy” in that many cytokines appear to share similar functions. Generalization of functions is not
possible with cytokines; nonetheless, their actions may be comfortably grouped as; autocrine, if the
cytokine acts on the cell that secretes it; paracrine, if the action is restricted to the immediate vicinity of a
cytokine’s secretion; endocrine, if the cytokine diffuses to distant regions of the body(carried by blood or
plasma) to affect different tissues.
Cytokines are made by many cell populations, but the predominant producers are helper T cells (Th) and
macrophages (Roitt and Delves, 2001). Cytokines have been variously named as lymphokines,
interleukins and chemokines, based on their presumed function, cell of secretion or target of action.
Because cytokines are characterized by considerable redundancy and pleiotropism, such distinction,
allowing for exception, are obsolete. The term interleukin was initially used by researchers for those
cytokines whose presumed target are principally leukocytes. It is now used largely for designation of
newer cytokines molecules discovered every day and bears little relation to their presumed
function(Roitt et al., 2001). The term chemokine refers to a specific class of cytokines that mediates
chemoattraction (chemotaxis) between cells.
A more clinically and experimentally useful classification divides immunological cytokines into those that
promote the proliferation and functioning of helper T-cells, type 1 (Th1) (e.g. IFN-y) and type 2 (Th2) (e.g.
IL-4, IL-10, IL-13, TGF-β), respectively (Goldsby et al., 2000). A classification of cytokines receptors based
on their three-dimensional structure has therefore been attempted. This classification, though seemingly
cumbersome, provides with several unique perspectives for attractive pharmacotherapeutic target
(Janeway et al., 2005).
xlvi
Immunoglobulin (Ig) superfamily which are ubiquitously present throughout several cells and tissues of
the vertebrate body, and share structure homology with immunoglobulin (antibodies), cell adhesion
molecules, and even some cytokines. Example: IL-1 receptor types. Haemopoitic Growth Factor (type 1)
family, whose members have certain conserved motifs in their extracellular amino-acid domain. The IL-2
receptor belongs to this chain, whose γ-chain (common to several other cytokines) deficiency is directly
responsible for the x-linked form of severe combined immunodeficiency (X-SCID). Interferon (type 2)
family: whose members are receptors for IFN β and γ. Tumour Necrosis Factor (TNF) (type 3) family:
whose members share a cysteine-rich common extracellular binding domain, seven transmembrane helix
familes, the ubiquitous receptor type of the animal kingdom. All G-protein coupled receptors (for
hormones and neurotransmitters) belong to this family (Goldsby et al., 2000)). It is important to note that
chemokine receptors, two of which act as binding protein for HIV (CXCR4 and CCR5), also belong to this
family.
xlvii
Table 1: Sources and Activity of Cytokines
Cytokine
Primary sources
Primary activities
IL-1
Macrophage
T,B cell activation; fever, inflammation
IL-2
T cells
T cell proliferation
IL-3
T cells
Growth of many cell types
IL-4
T cells
B cell growth and differentiation
IL-5
T cells
B cell eosinophil
IL-6
Macrophages , T Cells
B cell stimulation, inflammation
IL-7
Stromal cells
Early B and T cell differentiation
IL-8
Macrophages
Neutrophil (PMN) attraction
IL-9
T cells
Mitogen
IL-10
T cells
Inhibits Th 1 cytokine production
IL-11
Bone marrow stroma
Hematopoeisis
IL-12
APC
Stimulates T, NK cells
IL-13
T cells
Similar to IL-4
IL-14
Dendritics cells, T cells
B cell memory
IL-15
T cells
Same as IL-2
IFNα
Most cells
Anti-viral
IFNβ
Most cells
Anti-viral
IFNγ
T, NK cells
Inflammation, activates macrophages
TGFb
Macrophages, lymphocytes
Depends on target
TNFa
Macrophages
Inflammation, tumor killing
TNFb
T cells
Inflammation; tumor killing; enhance phagocytosis
GM-CSF
Th cell
Act on progenitor cells to stimulate growth and differentiation of monocytes
and DC
MIP-1α
Macrophages
Chemotasis of monocytes and T cells
MIP-1β
Lymphocytes
Chemotasis of monocytes and T cells
(Adapted from immunology briefcase, an e-book of Dalhousie University, 2006).
xlviii
1.3.2
Complement System
The complement system is a biochemical cascade that attacks the surface of foreign cells. Complementis
the major humoral component of the immune response (Rus et al., 2003; USC, 2006) and contains over
30 different proteins and is named for its ability to “complement” the killing of pathogens by antibodies.
Complement was discovered by Jules Bordet in 1900. It is produced by a variety of cells including,
hepatocytes, macrophages and gut epithelial cells (Anderson, 2003).
In human, this response is activated by the binding of complement proteins to carbohydrates on the
surface of microbes or by complement binding to antibodies that have attached to these microbes. This
recognition signal triggers rapid killing response (Liszewski and Atkinson, 1993). The speed of the
response is a result of signal amplification that occurs following sequential proteolytic activation of
complement molecules, which are also proteases. After complement proteins initially bind to the
microbes, they activate their protease activity which in turn activates other complement proteases, and
so on. This produces a catalytic cascade that amplifies the initial signal by controlled positive feedback
(Sim and Tsiftsoglou, 2004).
The cascade results in the production of peptides that attract immune cells, increase vascular
permeability and opsonize (coat) the surface of a pathogens, marking it for destruction. This deposition
of complement can also kill cells directly by disrupting their plasma membrane (Ruset al., 2003).
Some complement proteins bind to immunoglobulin or to membrane components of cells. Others are
proenzymes that, when activated, cleave one or more other complement proteins. Upon cleavage some
of the complement proteins yield fragments that activate cells, increase vascular permeability or
opsonize bacteria. (Abbas et al., 1994)(Table 2). Complement activation can be divided into four
pathways: the classical pathway, the alternative pathway, the lectin pathway, and the membrane attack
xlix
(or lytic) pathway. Both classical and alternative pathways lead to the activation of C5 convertase and
result in the production of C5b which is essential for the activation of the membrane attack pathway.
l
Table 2: The Biological Functions of Complement and its Fragments
Components and fragments
C5a, C3a, C5a-desArg, C4a
Activity
Increase of vascular permeability; smooth muscle contraction,
degranulation of mast cells and eosinophils
C5a, C5a-desArg
Neutrophil activation and chemotaxis; stimulation of prostaglandin
and leukotriene production
C3b, C4b
Opsonization of bacteris and immune complexes leading to
phagocytosis
C3a, C5a-desArg, Clq
Stimulation of the respiratoty burst of professional phagocytes
C5bC6C7C8C9 (membrane attack complex)
Lysis of bacteria and foreign cells
C3b, CR l
Solubilization of circulating immune complexes
(Adapted with modifications from Roitt et al., 2001; Francis et al., 2003).
1.4
Disorders of the Immune System
li
The immune system is a remarkably effective structure that incorporates specificity, inducibility and
adaptation. However, failure of host defenses can occur and fall into five broad categories:
hypersensitivities, immunodeficiencies, autoimmunity, immune complex disorders and host-graft
reaction.
1.4.1
Hypersensitivity
Hypersensitivity is an immune response that damages the body’s own tissues. They are divided into four
classes (Type I-IV) based on the mechanisms involved and the time course of the hypersensitive reaction.
Type I hypersensitivity is an immediate or anaphylactic reaction, often associated with allergy. Type I
hypersensitivity is mediated by IgE released from mast cells and basophils (USC, 2006). Usually harmless
substances can provoke inappropriate immune responses. The most common types of allergic reactionshay fever, some kinds of asthma, and hives are produced when the immune system responds to a false
alarm.
The role of IgE in the natural order is not known, although some scientists suspects that is developed as a
defense against infection by parasitic worms. The first time an allergy-prone person is exposed to an
allergen, he or she makes large amounts of the corresponding IgE antibody. These IgE molecules attach
to the surfaces of mast cells (in tissue) or basophils (in the circulation). When an IgE antibody sitting on a
mast cell or basophil encounters its specific allergen, the IgE antibody signals the mast cell or basophil to
release chemicals mediators stored within its granules(Roitt and Delves, 2001; USC, 2006). These
mediators include histamine, heparin and substances that activate blood platelets and attract secondary
cells such as eosinphilis and neutrophils. The activated mast cell or basophil also synthesizes new
mediators, including prostaglandins and leukotrines, on the spot. It is such chemical mediators that cause
the symptoms of allergy, including wheezing, itching, sneezing and runny eyes. They can also produce
lii
anaphylactic shock, a life threatening allergic reaction characterized by swelling of body tissues, including
the throat, and a sudden fall in blood pressure.
Type II hypersensitivity occurs when antibodies bind to antigens on the patient’s own cells, marking them
for destruction. This is also called antibody-dependent (or cytotoxic) hypersensitivity, and is mediated by
IgG and IgM antibodies (Roitt and Delves, 2001; USC, 2006). Immune complexes (aggregations of
antigens, complement proteins, and IgG and IgM antibodies) deposited in various tissues trigger Type III
hypersensitivity reactions (Roitt and Delves, 2001).
Type IV hypersensitivity (also known as cell-mediated or delayed type hypersensitivity) usually takes
between two and three days to develop. Type IV reactions are involved in many autoimmune and
infectious diseases, but may also involve contact dermatitis (poison ivy). These reactions are mediated by
T cells, monocytes, and macrophages (USC, 2006).
1.4.2
Immune Deficiency Diseases
An immune deficiency disease occurs when one or more cells within the immune system do not operate
properly, or the system is absent altogether. Some immune deficiency diseases are relatively common,
while others are extremely rare(Janeway et al., 2005). A primary immune deficiency occurs when the
abnormalities of the immune system develop from an inborn defect in the cells. Affected cells include Tcells, B-cells, B-cells, phagocytic cells or the complement system (Janeway et al., 2005). Most primary
immune deficiencies are inherited diseases; examples include X-linked agammaglobuliemia (XLA) and
severe combined immunodeficiency (SCID) which appear to run in families. Other primary immune
deficiencies, such as common variable immunodeficiency (CVID), appear less obviously inherited, but the
causes are unknown and genetic factors cannot be ruled out.
Secondary, immune deficiencies occur when damage is caused by environmental factors. Radiation,
chemotherapy, burns and infections contribute to the many causes of secondary immune deficiencies
liii
(Goldsby et al., 2000; Roitt et al., 2001). Acquired Immune Deficiency Syndrome (AIDS) is a secondary
immune deficiency caused by the Human Immunodeficiency Virus (HIV) that destroys T4 cells. AIDS
infections are known as “opportunistic” because they are produced by “commonplace” organisms that
are harmless in people with healthy immune system, but which take advantage of the “opportunity”
provided by immunocompromised condition. The most common infection is an unusual and lifethreatening form of pneumonia; Pneumocystis jerovecii (a protozoan infection).
AIDS patients are also susceptible to unusual lymphomas and Kaposi’s sarcoma, a rare cancer that results
from the abnormal proliferation of endothelial cells in the blood vessels. Although there are quite a
number of antiretroviral drugs which is used successfully in reducing viral burden, there is still no definite
cure for AIDS.
The use of immunomodulators, especially those of natural origin in tackling the menace of HIV holds a lot
of promise in preventing or treating opportunistic infections (Yamaguchi, 1992). In Leukemia and multiple
myeloma, abnormal, cancerous immune cells crowd out the normal stem cells of the bone marrow.
These abnormal cells reduce the number of B cells and lead to hypogammaglobulienemia or secondary
immune deficiency (Roitt and Delves, 2001; Janeway et al., 2005).
Diets lacking sufficient protein are associated with impaired cell-mediated immunity, complement
activity, phagocytes function, IgA antibody concentrations, and cytokine production. Deficiency of single
nutrients such zinc; selenium; iron; copper; vitamins A, C, E, and B6; and folic acid (vitamin B9) also
reduces immune responses (Chandra, 1997).
1.4.3
Autoimmune Diseases
Sometimes the immune system’s recognition apparatus breaks down, and the body begins to
manufacture antibodies and T cells directed against the body’s own constituents-cells, cell components,
or specific organs. Such antibodies are known as autoantibodies, and the diseases they produce are
liv
called autoimmune diseases. Autoimmune reactions contribute to many enigmatic diseases. For instance,
autoantibodies to red blood cells can cause anemia, autoantibodies to pancreas cells contribute to Type I
diabetes, and autoantibodies to nerve and muscle cells found in patients with myasthenia gravis.
Autoantibody known as rheumatoid factor is common in persons with rheumatoid arthritis. Persons with
systemic lupus erythematosus (SLE), whose symptoms encompass many systems, have autobodies that
affect the immune system at several levels. In patients with SLE, for instance, B cells are hyperactive
while suppressor cells are underactive. Moreover, production of IL-2 is low, while levels of gamma
interferon are high (USC, 2006).
The cause of autoimmune disease is not well understood, but several factors are likely to be involved.
These may include viruses and environmental factors such as exposure to sunlight, certain chemicals, and
some drugs, all of which may damage or alter body cells so that they areno longer recognizable as self.
Sex hormones may be important, too, since most autoimmune diseases are far more common in women
than in men (USC, 2006).
Heredity also appears to play a role. Autoimmune reactions, like many other immune responses, are
influenced by the genes of the MHC. A high proportion of human patients with autoimmune disease have
particular histocompatibility types. For example, many persons with rheumatoid arthritis display the selfmarker known as HLA-DR4 (Goldsby et al., 2000).
Many types of therapies are being used to combat autoimmune diseases. These include corticosteroids,
immunosuppressive drugs developed as anticancer agents, radiation of the lymph nodes, and
plasmapheresis, a sort of “blood washing” that removes diseased cells and harmful molecules from the
circulation (Goldsbyet al., 2000).
1.4.4
Graft Versus Host Diseases
lv
Transplantation of tissues and organs can cause life-threatening reactions in recipients (Janeway et al.,
2005). Graft cell reaction is specific and immunologic and has memory. It has been shown to be mediated
by lymphocytes. The antigens that serve as the principal targets of rejection are proteins encoded in the
major histocompatibility complex (MHC). The mainstay of preventing and treating the rejection of organ
transplant is immunosuppression, designed mainly to inhibit T cell activation and effector function (Abbas
and Lichtman, 2004).
1.4.5
Immune Complex Diseases
Immune complexes are clusters of interlocking antigens and antibodies. Under normal conditions
immune complexes are rapidly removed from the bloodstream by macrophages in the spleen and Kupffer
cells in the liver. In some circumstances, however, immune complexes continue to circulate. Eventually
they become trapped in the tissues of the kidneys, lung, skin, joints, or blood vessels depending on the
nature of the antigen, the class of antibody, and the size of the complex. There they set off reactions that
lead to inflammation andtissue damage (Roitt et al., 2001). Sometimes, as is the case with malaria and
viral hepatitis, they reflect persistent low-grade infections. Frequently, immune complexes develop in
autoimmune disease, where the continuous production of autoantibodies overloads the immune
complex removal system (Goldsby et al., 2000).
1.5
The
Concept
of
Immunomodulation,
Immunosuppression,
Immunostimulation
and
Immunotolerance
The innate and adaptive immune mechanisms could be modified by substances to either enhance or
suppress their ability to resist invasion by pathogens (William, 2001). Both immunostimulation and
immunosuppression are clinically relevant and search for substances and leads with these activities are
becoming a field of major interest all over the world (Naved et al., 2005).
lvi
Those compounds which appear to stimulate the human immune response are being sought for the
treatment of cancer, immunodeficiency diseases, or for generalized immunosuppression following drug
treatment; for combination therapy with antibiotics; and as adjuncts for vaccines (Jong and Birmingham,
1992). Those compounds that suppress immune reactions are potentially useful in the remedy of
autoimmune (an abnormal immune response against self-antigens) or certain gastro-intestinal tract
diseases (e.g. Crohns) (Badger, 1983). Whether immunomodulators enhance or suppress immune
responses can depend on a number of factors such as dosage, route of administration, and timing and
frequency of administration.
1.5.1
Immunostimulation
The immune response can be modified to enhance the humoral or cell-mediated immune response
against antigens. Immunostimulation reinforces the immune system. The mechanisms of potentiation of
immune responses may include one or all of the following situations: an increase in the rate; a
heightened intensity or level; an extension of normal immune response; and the induction of immune
response to an otherwise non-immunogenic substance (Jong and Birmingham, 1992).
1.5.2
Immunosuppression
The term immunosuppression refers to impairment of any component of the immune system resulting in
decreased immune function. Clinical indicators of immunosuppression include: myelosuppression, such
as pancytopenia, leucopenia, lymphopenia, and other blood dyscrasias; hypocellularity of immune
system tissues and decreased organ weight (such as the thymus, spleen, lymph nodes, or bone marrow);
decreased serum globulin levels; increased incidence of infections; and increased incidence of
tumors(Badger, 1983).
It is important to differentiate between unintended (adverse) immunosuppressive effects and intended
(pharmacodynamic) effects. For example, many antitumor drugs are toxic to rapidly dividing cells.
lvii
Immunosuppression due to bone marrow toxicity would be considered an adverse effect during the
treatment of a solid tumor, but not necessarily during treatment of a haematologic malignancy. For drugs
intended to be used for prevention of transplant rejection (e.g. cyclosporine), immunosuppression is the
intended pharmacodynamic effect.
Although this distinction appears to be relatively obvious, there are examples of drugs in which the
relationship between immunosuppression and pharmacodynamic effects appears subtly, yet is important
e.g., nonsteroidal anti-inflammatory drugs, 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors
and testosterone (Fimmel and Zouboulis, 2005). Methods to enhance detection of immunosuppression in
standard toxicology studies have been described, including exact tissues that should be examined and
effects that should be noted. These include determination of serum biochemical markers such as globulin
levels, hematology (including differential), gross pathology findings, immune system-related organ
weights, and histologic examination of immune system-related tissues.
1.5.3
Tolerance
Tolerance refers to the specific immunological non-reactivity to an antigen resulting from a previous
exposure to the same antigen (Miller, 1993). While the most important form of tolerance is nonreactivity to self-antigens, it is possible to induce tolerance to non-self-antigens. When an antigen or
substance induces tolerance, it is termed tolerogen. Tolerance can be induced to antigenic components
on both soluble proteins and cells (tissues) by injecting these materials into animals.
Tolerance to tissue and cell antigens can be induced by injection of hemopoietic (stem) cells in neonatal
or severely immunocompromised (achieved by lethal irradiation or drug treatment) animals. A state of
tolerance to a variety of T-dependent and T-independent antigens has been achieved using some
substances in various experimental models (Roitt et al., 2001).
1.6
Potentials of Mushroom as Immunomodulatory Substance of Natural Origin
lviii
The value of medicinal mushrooms to human health is increasingly gaining acceptance as researchers
provide data on the array of bioactive compounds found within these fascinating fungi. Mushrooms grow
wild in many parts of the world and also are commercially cultivated. It is low in calories and
carbohydrate; high in vegetable proteins and essential amino acids; a source of some fibre; and rich in
some important vitamins and minerals, including B vitamins, iron, potassium, selenium and zinc (Nworu
et al., 2011).
Researchers have found that mushrooms can directly stimulate both the lymphocytes, neutrophils and
secondary immune responses (immunoglobulin IgE, IgG) of the immune system. This stimulus can
increase production of immune defenders such as cytokines and macrophages, which play important
roles in recognizing and removing foreign antigens, as well as releasing chemical mediators including
interleukin-I (Adachiet al., 1994).
Some substances with immunomodulatory activities have been isolated from different species of
mushroom and include beta-glucans, lentins, polysaccharides, polysaccharide-peptide complexes,
triterpenoids, nucleosides and other secondary metabolites. Many of these bioactive substances, through
their stimulatory effects on the immune system, are showing powerful antitumour, antimutagenic and
anticancer activity (Borchers et al., 2008).
Of the hundreds of known mushroom varieties, several have been studied for their ability to enhance the
human immune system and fight infections. Some well-known medicinal mushrooms with benefits for
the immunesystem are presented in table 3.
The healing and immunostimulating properties of mushrooms have been known for thousands of years.
The extracts of these mushrooms were widely used for treatment purpose indifferentcountries. The
number of mushrooms on earth is estimated to be 140000: approximately10%(about 14,000 named
lix
species) is known. These mushrooms contain biologically active polysaccharides in fruit bodies and
cultured mycelium.
lx
Table3: Some Mushrooms with Identified Immunomodulatory Activities
Name of Mushroom
Common Name
References on Immuno-Activity
Cordyceps sinensis
Cordyceps (Caterpillar fungus)
Bo And Bau, 1980; Chang, 1981; Lui et al., 1992;
Ying et al., 1987
Ganoderma Iucidum
Reishi
Chang, 1981; Lin et al, 2006; Dharmananda, 1988;
Lui 1993, Miyazaki and Nishijima, 1981; Willard,
1990
Trametes versicolor
Kawaratake (Turkey Tail)
Arora, 1986
Auricularia auricular
Kikurage
Arora, 1986; Bo and Bau, 1980; Ying et al., 1987.
Agaricus blazeii
Himematsutake
Arora, 1986; Bo and Bau, 1980; Ying et al., 1987.
Trametes versicolor
Kawaratake (Turkey Tail)
Arora, 1986
Auricularia aruricular
Kikurage
Arora, 1986; Bo and Bau, 1980.
Agaricus blazeii
Himematsutake
Arora, 1986; Bo and Bau, 1980; Ying et al., 1987.
Lentinula edodes
Shitake (Snake butter)
Liu et al., 1992
Grifola frondosus
Maitake
Arora, 1986; Adachi et al.,1994;
Hericium erinaceus
Lion’s mane Hericium
Yang and Jong, 1989
Grifola umbellata
Choreimaitake (zhu ling)
Arora, 1986; Suzuki, 1990
Flamulina velutipes
Enokitake (Velvet Foot)
Arora, 1986; Zhou et al, 1989
Tremella fuciformis
White fungus
Arora, 1986; Ying et al., 1987
Poria cocos
Bukuryo (Hoelen)
Narui et al., 1980
lxi
1.7
Beta (B) Glucans
Polysachandes is produced universally by living organisms. They exhibit a large variety of complex
chemical structures, physiological functions and a wide range of potential application. They are polymers,
of sugars (monosaccharides) joined to each other by glycosidic linkages. These are very complex
molecules because sometimes covalent bonds occur between many pairs of carbon atoms(Bohn and
Miller, 1995; Celine and Philippe, 2007). Consequently, one sugar unit can be joined to more than two
other sugars, which results in the formation of highly branched enormous macromolecules(Bohn and
Miller, 1995; Celine and Philippe, 2007). The polysaccharides of mushrooms occur mostly as glucans.
Some of which are linked by ß-(1-3), (1-6) glycosidic bonds and α – (1-3) glycosidic bonds but many are
true heteroglycans.
Beta glucans are found in such foods as oats, barley, cereals, seaweeds, mush rooms and yeasts (Bohn
and Miller, 1995; Celine and Philippe, 2007).
ß – (1,3) D glucans are also found both in the prokaryotes and eucaryotes.
Betaglucans from fungi
and yeast are found as cell wall components. They have a common structure comprising a main chain of
(1,3) - Linked ß-D- glucopyranosyl units attached by (1, 6) linkages.
Glucans from barley oats or wheat are also found as cell wall components of grains but exhibit a different
structure, with a mixed ß- (1,3) and ß – (1,4) glycosidic bonds in the backbone.
1.7.1 Beta Glucans and the Immune System
Beta glucans do not in themselves cure disease. Rather, they help stimulate the immune system to work
better so that diseases can be prevented from attacking the body. Recent studies help us understand
how β gucans stimulate the immune system: β glucans from fungi bind to specific membrane receptors of
phagocytic cells and natural killer (NK) cells, stimulating their germ-killing abilities.
lxii
It also appears that β glucan molecules resemble the molecules found on bacterial cell walls. In effect, β
glucan molecules make the body believe it is being invaded by a bacterium. β glucan have been known to
scientists as a plant constituent for decade. It has been common knowledge in the scientific community
that βglucan is the most powerful immune stimulant and a very powerful antagonist to both benign and
malignant tumors (Dalia et al.,2007).
β glucan are naturally occurring polysacharides. These glucose polymers are the constituents of the cell
wall of certain pathogenic bacteria (Pneumocystis jerovecii, Cryptococus neoformans, Aspergillus
fumigatus, Histoplasma capsulatum, Candida albicans) and fungi (Saccharomyces cerevisiae). The main
components of the fungal cell wall are polysacharides and glycoproteins.
Sacharomyces cerevisiae cell wall consists of three layers: an inner layer of soluble B-glucan (30-35%),
middle layer of soluble β Glucan (20-22%), external layer of glycoprotein (30%) (Tokunaka et al., 2002). B
glucan has been purified from brewers and baker’s yeast, oats and barley bran(Baur and Geisler,
1996;Tokunaka et al., 2000).
1.7.1.1 Beta Glucan Immunostimulating Activity.
Persons who suffer from systemic fungal infections including those caused by Candida, Aspergillus and
Cryptococcus species have being described to possess high levels of circulating b-glucans in their plasma.
It is possible that they may have modulating effects on the immune system by activatingmacrophages,
phagocytosis of the pathogens, release of proinflammatory cytokines(Sato et al., 2006).B glucans have
been established as a key molecular pattern recognized by neutrophils(or polymorphonuclear leukocytesPMNs) in response to Candida albicans, because antibody specific for β glucan, a major component of
yeast cell wall, blocks this response(Lavigne et al., 2006).
lxiii
This mechanism to recognize and respond to their conserved structural components, particularly B –
glucans has evolved in mammals as defense against fungal pathogen.
Macrophages play a critical role in all phase of host defense that are both innate and adaptive immune
response in case of an infection. Lysosomal enzymes and phagocyticactivity determine the macrophage
function. The secreting of cytokines (1L-1,1L-6,1L-81,1L-12, TNF-α) and inflammatory mediators (nitric
oxide-NO and hydrogen perodixe H2o2) are other effects of these cells. Therefore activation of
macrophage functions by β glucans increase host immune defense. However, polysaccharides stimulate a
dose dependent increase in NO and TNF – α but not in reactive oxygen intermediate production in
peritonuem macrophages (Kim et al., 2004).
It is suggested that the ability of polysaccharides upon the up regulation of these surface molecules
involved in antigen presenting processes may by inference activate T cell mediated immunity against
malignant cells invivo. Taken together, these results suggest that β glucan acts as an effective
immunodulator and enhances the antitumoral activity of peritoneum macrophages (Kim et al., 2004).
1.7.1.2 Beta Glucan Increases Resistance to Infectious Challenge
B glucan itself can elicit broad anti-infective effects. Staphylococcus aureus, Echerichia coli, Candida
albican, Pneumocystis jerovecii, Listeria monocytogenes, Leishmonvia donovani, influenza virus are the
micro-organisms against which a protective effect of β glucan has been established(Dalia et al., 2007).
The potential antiviral effect of Sacharromyces cerevisiae b glucan was investigated on the pneumonia
induced by swine influenza virus (SIV). The microscopic lung lesions induced by SIV infection were
significantly more severe than those induced by infect in animals preadministered β glucan(Dalia et al.,
2007). Studies have also shownthat systemic β glucan treatment would result in enhanced migration of
neutrophils into a site of inflammation and improve antimicrobial function(Leblanc et al., 2006)as well as
lxiv
a priming effect for chemotaxis and respiratory burst in circulating neutrophils isolated from β glucantreated animals(Leblanc et al., 2006).
1.7.1.3 Beta Glucan AnticarcinogenicActivity
Carcinogenesis can be separated to different stages. The initiation phase involves exposure to a mutagen
and often requires its subsequent metabolic transformation into a biologically active form. This exposure
even if resulting in permanent damage to DNA is often insufficient by itself to cause cancer. Studies have
shown that polysaccharide mushroom extracts can inhibit cell transformation induced by a defined
oncogene through a novel non cytocidal route(Wendy et al., 2004; Dalia et al., 2007).
Natural killer cells (NK) aredirectly cytotoxic for tumor cells and play a primary role in regulating immune
response. Experimental studies have also shown that β glucan may mediate their anticarcinogenic activity
through one pathway called “Angiogenesis” which is a crucial to tumor growth and metastasis and
interruption of this process is a prime avenue for therapeutic intervention of tumor proliferation. Natural
killer(NK) cells are also directly cytotoxic for tumor cells and play a primary role in regulating immune
responses. As immunostimulating agents which acts through the activation of macrophages and NK cells
cytotoxicity, β glucan can inhibit tumor growth in promotion stage(Kodama et al., 2002; Ho et al., 2004;
Kodama et al., 2005).
1.7.1.4 Beta Glucan as Adjuvant to Cancer Chemotherapy and Radiotherapy
The major side effect of most chemotherapeutic drugs is neutropenia. The administration of anticancer
drugs often impairs blood forming functions. These functions are important to maintain thedefense
system ofthe individual. As a result, chemotherapy may accelerate risk of infection that decreases the
quality of life for cancer patients. β glucan extracted from certain mushroom have been shown to reduce
cyclophosphamide induced leucopenia(Harada et al., 2002).
lxv
Radiotherapy often results in hematopoietic and immune depletion. Patients often experience anemia,
lymphocytopenia and thrombocytopenia. This leads to high risk of development of serious and lethal
infections andincreasing the mortality andmorbidity of these patients. Experiment has demonstrated that
soluble yeast β glucan could enhance the proliferation ofhaematopoietic cells transplantation in a CR3
dependent manner. Taken together these observations suggest a novel role for complement CR3 and b
glucan in the restoration of hematopoeisis following bone marrow injury(Jin et al., 2003; Cramer et al.,
2006).
1.8 Botanical Profile and Review of Pleurotus tuberregium
Pleurotus tuberregium(Fr) Sing, is an edible basidiomycetes which grows in both tropical and subtropical
regions of the world(Okhuoya and Okogbo, 1991). It is found in many parts of tropical Africa and forms a
part of the local people’s food system. The sclerotia is collected from the forests, dried and stored for
future use as food(Isikhuemhen and Okhuoya, 1995).
The tuberous sclerotium can be used as a partial replacement for melon seed (Citrulus ianatus) or
groundnut (Arachis hypogea) cake in traditional preparation of sauces and soups. P. tuberregium is used
for medicinal purposes by traditional medical practitioners(Isikhuemhen and Okhuoya, 1995).
lxvi
1.8.1 Taxonomy of Pleurotus tuberregium
Kingdom:
Fungi
Phylum:
Basidiomycetes
Class:
Agaricomycetes
Order:
Agaricales
Family :
Pleurotaceae
Genus:
Pleurotus
Species:
P. tuberregium
Synonyms:
Pachyma tuberegium Fr.1822
Lentinus tuber-regium(Fr) 1836
Common vernacular names:
1.8.2
usu(Igbo), awu(Igala), katala(Hausa), umoho(Igede).
Botanical Description of Pleurotus tuberregium
P. tuberregium is found in many parts of the world especially the tropical and subtropical regions of the
world (Okhuoya and Okogbo, 1991). It produces mushroom from a unique globose sclerotium that is
more like a giant truffle. The sclerotia or underground tuber are usually spherical to ovoid, subterranean
sclerotia which sometimes measure up to 3cm in diameter (Okhuoya and Okogbo, 1991; Fasidi and
Olorunmaiye, 1994). The sclerotium is dark brown on the outside and whitish on the inside (Okhuoya and
Okogbo, 1991;Oso, 1977). The fruiting bodies of P. tuberregium is shown in figure 2
lxvii
Figure 2: The fruiting bodies of Pleurotus tuberregium
lxviii
1.8.3
Geographical Distribution ofPleurotus tuberregium
P. tuberregium (Fr) Sing, occurs in both tropical and subtropical regions of the world (Okhuoya and
Okogbo, 1991).
The geographical distribution of P. tuberregium includes most of equatorial Africa, India, Sri Lanka, South
East Asia and North Australia(Oso, 1977).
1.8.4
Ethnomedicinal and Folkloric Uses of Pleurotus tuberregium
P. tuberregium is used for medicinal purposes by traditional medical practitioners(Zoberi, 1973; Oso,
1977; Okhuoya and Okogbo, 1990; Okhuoya and Isikhuemhen, 1995). Most studies on P. tuberregium
have concentrated on the food and nutritional contents of the sclerotium and sporophores.
In Nigeria, both the sclerotium and the mushroom are eaten. The sclerotium of P. tuber-regium has long
being used for food and medicine by various tribes in Nigeria(Isikhuemhen and Okhuoya, 1995;Akpaja et
al., 2003). The Igbo use it to treat heart problems while it is used to treat asthma cough and obesity
among people of Edo State(Isikhuemhen and Okhuoya, 1995; Isikhuemhen et al., 2000a, 2000b). The
sclerotium which is hard is peeled and ground for use in a vegetable soup(Fasidi and Olorunmaiye, 1994).
lxix
It is used in the preparation of cures for headache, stomach ailments, chest pain, colds and fever, asthma,
small pox, and high blood pressure (Okhuoya and Okogbo, 1991; Fasidi and Olorunmaiye, 1994; Alobo,
2003). It is sometimes used for weight gains in malnourished babies. It is also used in the Asaba area of
Nigeria in herbal preparation for pregnant women to aid the development of foetus. In Ghana, the
slerotium is used mainly for fattening of malnourished babies and as one of the ingredients in the
embalming of dead bodies(Okhuoya et al., 1998).
It has also being reported that the pure culture of this fungus is able to kill and feed on
nematodes(Hibbett and Thom, 1994).
1.8.5
Pharmacological Studies, Phytochemical and Proximate Constituents of P. tuber
regium
P. tuberregium is considered a popular edible mushroom and has been considered a profound health
promoting mushroom in traditional Chinese medicine(Isikhuemhen et al., 2000a, 2000b; Huang, 2002).P.
tuberregium polysaccharideattenuates hyperglycemia and oxidative stress in experimental diabetic
rats(Hui et al., 2012). The fruit bodies ofP. tuberregium are rich in protein while the sclerotium is rich in
fiber especially non starch polysaccharides(Kadiri and Fasidi, 1990) mainly composed of bioactive β
glucans responsible for pharmacological actions(Cheung and Lee, 2000; Tao et al., 2006). For most other
species of Pleurotus, several medicinal properties have been reported. They include properties
attributable to their polysaccharides;antigenotoxic, biomutagenic activities(Fillipie and Umek, 2002) antiinflammatory activity, antilipidemic, antihypertensive and antihyperglycemic activities(Huet al., 2006)
antibacterial and antifungal activities(Hu et al., 2006;Ngai and Ng, 2006; Lee at al., 2010; Dahech et al.,
2011; Wong et al., 2011).
lxx
CHAPTER TWO
MATERIALS AND METHODS
2.1 Materials
2.1.1 Chemicals and Reagents
Ovalbumin (Sigma, Germany), Tween 20 (Sigma, Germany),
ABTS Peroxidase Substrate System,
Peroxidase-labelled antibodies (HRP anti-Mouse IgG (H+L), anti-mouse IgG1 (H+L); anti-mouse IgG2a)
(Kirkegaard & Perry Laboratories, KPL, Gaithersburg, MD, USA), Peroxidase Stop Solution Concentrate
(KPL, USA), Coating Buffer Concentrate (KPL, USA), Blocking Solution Concentrate (KPL, USA), absolute
ethanol (sigma), Indian ink, aluminium hydroxide (Al(OH)3), sodium bicarbonate (Na2CO3 ), pyrogen free
sterile normal saline, distilled water (service training centre unn), Fehlings solution I and II, Dragendorff’s
reagent, Wagner’s reagent, Mayer’s reagent, Hagner’s reagent, tetraoxosulphate (vi) acid, ammonium
hydroxide, ferric chloride, ethanol, lead subacetate, 2% dinitrobenzoic acid in 90% ethanol, glacial aceytic
acid, aluminium chloride, arachis oil. All reagents and solvents used were of analytical grade.
2.1.2 Equipment
Thermomax Microplate ELISA Reader (Molecular Device, USA), 96 well ELISA plates (Titertek®),centrifuge
(Gallenkamp, Germany), UV spectrophotometer(UnicoTM UV2102PC).
2.2 Methods
2.2.1 Collection and Authentication of Plant Materials
The fruiting bodies of the mushroom, Pleurotus tuberregium (Figure 2) were purchased commercially
from a local market – “Eke Atta” in Ikeduru Local Government Area of Imo State in the months of
November to January. The plant material was authenticated by Mr Alfred O. Ozioko, a taxonomist of the
lxxi
international Center for Ethnomedicine and Drug Development (Inter CEDD), Nsukka, Enugu State,
Nigeria.
2.2.2 Preparation of Plant Material
Collected fruiting bodies of the mushroom plant, Pleurotus tuberregiumwere thoroughly cleaned and
dried on filter paper sheets under shade at room temperature for about 2 weeks. After drying, the plant
materials were properly pulverized using a hammer mill. Thoroughly shade dried finely powdered parts
of the plant were extracted using aqueous hot water extraction method by heating the powder with
distilled water at 98oC for 4 h(Jones, 1998). The extract was then allowed to cool and filtered using
Whatman filterpaper no 4. The extract concentration was then determined and aliquoted in air tight
containers and stored at -20oC for further experimental studies. The extract yield was determined as
18.45%, w/w.
2.2.3 Bioactivity Guided Fractionation of Crude Extract
Based on the result of a prior preliminary immunomodulatory screening of the crude extract of Pleurotus
tuberregium (PT) as well as the evidence of immunomodulatory activity of β-glucan polysacharide rich
fractions of some mushrooms (Pugh et al., 2001; Wasser, 2002; Borchers et al., 1999), aβ−1,3−D-glucanrich polysaccharide fractionof P. tuberregium was extractedand studied for immunomodulatory
properties. The scheme used for this separation and activity guide is shown in Figure 3.
lxxii
PLANT MATERIAL
(P.tuber-regium)
Mascerate with 96% ethanol +
80oC + 3 hrs + allow to cool +
filter
Residue
Filtrate
Residue air
dried+ hot
distilled water +
water + 98oC + 4
Hrs + Cool +filter
Filtrate
Residue
Centrifuge at
5000 rpm for 10
min
Residue
supernatant
1.5 × Equal volume of
ethanol + 4oC+Centrifuge
Supernatant
Residue
Freeze dry
β-glucan rich polysaccharide for further
immunomodulatory activity screening
lxxiii
Figure 3: Schematic Representation of the Extraction of Pleurotus tuber-regium(PT)and
its Beta Glucan Rich PolysaccharideFraction (BGP)
2.2.4 Extraction of the Beta-glucan (β) Rich Polysaccharide Fraction of Pleurotus tuberregium
The β-glucan rich polysaccharide fraction of Pleurotus tuberregium was extracted from about 500 g
portion of the powdered Pleurotus tuberregium.
The plant material (500 g) was extracted with 96% ethanol (11.25 L) at 80oC for 3 h to remove low
molecular mass compounds (Robyt, 1998). The air dried residue was extracted with 2.5 L of hot distilled
water (98oC) for 4 h. The filtrate was centrifuged and the supernatant (PT) recovered after centrifugation.
Subsequently, the β-glucan rich polysaccharide extract (BGP) was obtained using the modification of a
previously described protocol(Wasterlund et al., 1993). Briefly BGP was precipitated from PT with the
addition of 1.5 volumes of absolute ethanol. The precipitate was stored overnight at 4oC the precipitate
was centrifuged and freeze dried to obtain BGP powder (10g, 2.0%[w/w]).
A water soluble beta glucan rich polysaccharide extract of P. tuberregium was extracted and studied for
immunomodulatory properties.
2.2.5 Phytochemical Analysis of Extracts
Phytochemical studies were carried out on the hot aqueous crude extract and on the β-glucan rich
polysaccharide fraction of Pleurotus tuberregium. Phytochemical studies were carried out using
lxxiv
appropriate procedures (Harborne, 1998;Evans, 2009). The extracts were screened for carbohydrate
using Molisch test; reducing sugar using Fehling’s test; alkaloids using Dragendorff’s, Meyers and
Wagner’s reagent; glycosides using hydrolysis, Bontrager’s, Modified Bontrager, Keddel’s and KellerKiliani test; flavonoids using ammonia and ammonium chloride tests; saponins using frothing and
emulsion tests, tannins using ferric chloride and lead subacetate tests; sterols and triterpenes using
Moleschott’s and Salkowski’s tests.
2.2.6 Pharmacological Studies
2.2.6.1 Animals
Adult swiss albino mice of either sexes of 7-8 weeks old weighing 20 g - 30 g obtained locally from the
laboratory animal facility of the Department of Pharmacology and Toxicology, University of Nigeria,
Nsukka were used in the study. The animals were acclimatized for at least 5 days in the institutional
animal facility under standard conditions of temperature (23oC ± 2oC) and 12:12 h of light and dark cycle.
The animals were fed with standard animal feed (Livestock Feed PLc, Lagos, Nigeria) and water ad
libitum.
2.2.6.2 Antigen
Ovalbumin antigen (Sigma, Germany) was used for immunization and challenge.
2.2.6.3 Acute Toxicity (LD50) Test of Extracts
The acute toxicity of PT and BGPwas estimated in mice using the appropriate method (Lorke, 1983). The
procedure was carried out using the oral route. The first phase was determination of the toxic range. The
mice were placed in three groups of 3 mice each and were given 10, 100 or 1000 mg/kg of PT or BGP. The
treated mice were observed for 24 h for number of deaths.
lxxv
The death pattern in the first phase determined the doses used for the second phase. Since there was no
death recorded in the first phase, for the oral route of administration, four mice received 1000, 1600,
2900 or 5000 mg/kg of the extracts at one dose per mouse. The treated animals were observed for 24 h
for lethality and other signs of acute intoxication.
The LD50 was then calculated as the geometric mean of the highest non-lethal dose and the least toxic
dose.
2.2.7
Studies on the Hot Aqueous Extract of Pleurotus tuberregium (PT) and the Beta (β)-Glucan
RichPolysaccharide Fraction of Pleurotus tuberregium (BGP)
The hot aqueous extract of Pleurotus tuberregium was screened for immunomodulatory activity in mice
using different models which showed the effect of the extract on some specific and non-specific immune
responses. The effect on delayed type hypersensitivity response, specific primary and secondary antibody
synthesis, carbon clearance, relative organ weight as well as in vivo leukocytes mobilization was studied
at this stage.
Further immunomodulatory studies were carried out on BGP using delayed type hypersensitivity model
and antibody ovalbumin – specific determination (total IgG, IgG1 and IgG2a) in mice.
2.2.7.1 Studies on Relative Spleen Weight
PT (100, 200 or 400 mg/kg)or Morinda citrifolia extract (Noni®, 100 mg/kg, per os; Good ′N Natural,
Ronkonkoma, NY)were administered orally to each mouse group for 7 days. On the Day 8, the animals
were sacrificed 24 h after the last dose and the spleen were freshly excised and weighed. The results are
expressed as the relative organ weight (organ weight/100 g of body weight) for each animal(Davis and
Kuttan, 2000).
2.2.7.2 Studies on Phagocytic Index
lxxvi
Phagocytic activity of the reticuloendothelial system in vivo was determined by carbon clearance test.
PT (100, 200 or 400 mg/kg) or Noni® (100 mg/kg)were administered orally to each group of animal for 8
days. On the Day 8, after the last dose, the animals received an intravenous injection of carbon
suspension (1:50 dilution of Indian ink, Hi-Media Laboratories Pvt. Ltd, Mumbai, India, in a dose of 1
ml/200 g of body weight through the tail vein). Blood was withdrawn from the retro orbital plexus before
injection at 0 min and 15 min after injection of carbon suspension. Each blood sample (50 µl) was lysed
with 4ml of 1% Na2CO3 solution. The absorbance was measured spectrophotometrically at 680nm
wavelength. The results are expressed as phagocytic index:
K = (Ln A t15 min) – (Ln At0 min) / (t 15 min– t0 min)
Where, A t15 min and A t 0 minare the Absorbances at 15 min and 0 min respectively.Phagocytic indices (K)
were normalized to to the mean index for the untreated control group.
2.2.7.3 Studies on Delayed Type Hypersensitivity Response (DTHR)
Delayed type hypersensitivity was induced in mice using ovalbumin as antigen. PT (100, 200 or400 mg/kg)
or BGP were administered orally 3 days prior to sensitization and continued daily after the challenge.
Animals were sensitized by subcutaneous injection of 100 µg ovalbumin (OVA; 1 µg/µl; Sigma-Aldrich,
Munich, Germany) in normal saline (Day 3) in the plantar region of the right hind foot paw and
challenged on Day 13. After 24 h the oedema produced by antigenic challenge in the left hind paw was
measured with a micrometer screw gauge(AticoTM, Advanced Technocracy Inc., Haryana, India). The DTH
response was determined from the increase (swelling) relative to initial footpad size and expressed as
mean percent thickness/edema (Barcotti et al., 1984).
2.2.7.4 Studies on Humoral Antibody Response Induced by Ovalbumin
lxxvii
The effect of PT and BGP on humoral immune responses was determined in mice in two separate
experiments using ovalbumin (OVA) as the antigen in a homologous prime-boost strategy. Mice were
immunized by an intraperitoneal (i.p.) injection of 100 µg of ovalbumin into their hind footpads (50
µl/footpad)on day 3 and challenged by injecting the same amount (i.p.) on Day 14. Primary antibody titer
was obtained on day 14 before the boost immunization, and secondary antibody titres were determined
twice on days 21 and day 28(7 and 14 days post booster)by the Enzyme Linked Immunosorbent Assay
(ELISA).
PT (100, 200 or 400 mg/kg) or BGP (100 or 200mg/kg) were administered orally 3 days prior to
sensitization and continued on alternate daysafter the challenge.Blood samples were collected from the
retro-orbital plexus of individual sensitized mouse on days 14, 21 and 28and centrifuged to obtain the
serum specific immunoglobulins (total IgG, IgG1, and IgG2a) titres against ovalbumin which was
determined by Enzyme Linked Immunosorbent Assay. Microtiter plates with 96 wells were coated with
100 µg OVA in bicarbonate buffer pH 9.6 and incubated overnight at 4oC with a proper cover. After the
incubation the unbound OVA was washed off thrice with 0.01 M PBS buffer containing 0.05% Tween-20
(PBS-T). The nonspecific binding sites were blocked with 1% solution bovine serum albumin (BSA) in PBS
and incubated for 1 h at room temperature. The wells were washed again with PBS-T and incubated with
100 μl of 1 in 25 diluted sera samples in duplicates for 1 h at 37oC. The unbound serum proteins and
other constituents were washed off. To assess the antibody levels, 100 μl of HRP-conjugated goat antimouse total IgG, IgG1, and IgG2a secondary antibodies were added at a dilution of 1:1000 was and
incubated for another 1 h at room temperature. Finally the unbound conjugates were washed off with
PBS-T and 100 μl/well of freshly prepared ABTS Peroxidase Substrate System and stopped with 100 μl of
peroxidase stop solution. The colour developed was read at 405 nm using an automatic ELISA reader
(Thermomax ® ELISA Plate Reader). The data represent the mean absorbance values of the sera samples
lxxviii
assayed in duplicates. Naïve mice sera or preimmune sera were included in the assay as control (Beck and
Spiegelberg, 1989).
2.2.8
InvitroStudies
The immunomodulatory activities of BGP were further evaluated by in vitro studies in the laboratory
facility of Prof. Andreal Gamboto, Vector Core Facility, Rangos Research Center, University of Pittsburgh
Medical Center, Pennyslyvania, USA. The assistance of my project supervisor; Dr C.S Nworu in getting
these studies done is acknowledged with gratitude.
Cell lines and Culture medium
Spleen cell and RAW264.7 cells were cultured in R-10, consisting of RPMI 1640 medium (Corning cellgro®
RPMI; Mediatech Inc., Manassas, VA, USA) supplemented with 10% heat-FBS, 50 µM 2-mercaptoethanol
(Gibco, Invitrogen, USA), 100 U/ml penicillin, and 100 µg/ml streptomycin
Preparation of lymphocytes from mouse spleens
BALB/c mice, 8-10 weeks old, obtained from Charles River Laboratories, Inc., Wilmington, MA, USA, fed
and kept in institutional animal facility were used for the study. Mice were euthanized and the spleens
were removed aseptically. The spleens were minced and single cell suspension was prepared by gentle
dispersion of the cells and straining through 70-μm BD Falcon cell strainer. Residual red blood was
haemolysed with acetic acid lysing buffer. The spleen cell suspension was washed twice with Hank’s
Balanced salt Solution (Corning cellgro® HBSS; Mediatech Inc., Manassas, VA, USA), and centrifuged twice
at 1200 rpm at 4°C for 5 min. The resulting cell pellet was resuspended in R-10 medium and re-suspended
lxxix
in R-10 medium for subsequent experiments. Cell viability was determined by trypan-blue dye exclusion
technique on a Neubauer hemocytometer and the culture adjusted to 1 × 106 viable cells/ml (De Jesus et
al., 2013).
2.2.8.1 Spleenocytes Proliferation Assay
The lymphoproliferative (mitogenic) effect of the BGP on murine splenic cells was determined by the
MTT(3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide) reduction assay. MTT is reduced to
blue formazan product by mitochondrial dehydrogenase in viable cells which reflects the normal
functioning of mitochondria and hence the metabolic rate of cells. Spleenocytes (1×105cells in 100 µl)
were seeded in a 96-well U-bottom plates. Graded concentrations of BGP (5, 25, 50, 100, 250 or 1000
μg/ml).or 1 μg lipopolysaccharide/ml (LPS; serotype 0128:B12, L 4255; Sigma) as a standard mitogen. The
negative control consists of similarly un-stimulated wells in which R-10 medium was added. The cells
were incubated at 37°C in 5% CO2 for 72 h.
Cell proliferation was measured by the MTT cell viability assay. Briefly, 20 μl of MTT tetrazolium bromide)
(5 mg/ml in PBS) was added to each well and incubated for another 4 h at 37oC. The culture plate was
centrifuged and culture uumedium gently discarded. Formazan crystals formed were dissolved by the
addition of 150 μl of DMSO to each well. The plate was agitated for 30 minutes on a shaker and the
optical density (OD) of the developed colour was read at 570 nm in ELISA plate reader (Bio-Kinetic
Reader-E312e®; Bio-Tech Instruments, Winooski, VT) (Mossman T,1983). The proliferation index (PI) was
calculated according to this relationship:
=
[ . .]
[ . .]
lxxx
2.2.8.2 Effect of BGP on Inducible Nitric Oxide (iNO) Production/Release
Nitric oxide production and release by macrophages was determined by nitrite accumulation in culture
supernatant measured by Griess reagent (Green, 1981). RAW264.7 cells (1×105 cells/well) were seeded in
96-well flat plate for 24 h. And then cells were treated for 24 h with BGP (0, 5, 25, 50 or 100 μg/ml)or LPS
(1 μg/ml). After incubation, 100 μl of cell-free culture supernatants were mixed with an equal volume of
freshly prepared Griess reagent in 96-well plate and incubated at 25℃ for 10 min. Griess reagent
contains equal volume of 0.2% naphthylethylenediamine dihydrochloride and 2% sulphanilamide in 5%
phosphoric acid. The absorbance was measured on a microplate reader at 570 nm (Bio-Kinetic ReaderE312e®; Bio-Tech Instruments, Winooski, VT). Nitrite concentration was quantified by extrapolation on a
standard NaNO2 curve generated by different concentrations of NaNO2 included in the Griess reagent
assay. Nitrite concentrations in culture supernatants were measured to assess iNO production in
RAW264.7 cells.
2.2.8.3 Effect of BGP on Tumour Necrosis Factor (TNF- Α) Production /Release
The effect of BGP on tumour necrosis factor (TNF-α) by macrophages was determined by ELISA.
RAW264.7 cells 1×105 cells/well) were seeded in 96-well flat bottom plate for 24 h. The cells were pretreated for 2 h with BGP (0, 5, 25, 50 or 100 μg/ml) or LPS (1 μg/ml) was added to the wells. After
incubation, cell-free supernatant was harvested and stored at -20°C for future analysis (Khanittha et al.,
2004). The concentrations of TNF-α in the conditioned culture supernatant was measured by cytokine
capture ELISA kits according to the manufacturer’s instruction (R&D Systems, Inc., Minneapolis, MN,
USA). The concentration of TNF-α was calculated from a standard curve of the standard mouse TNF-α
included ELISA assay.The level of sensitivity of the kit was 31.2 pg TNFα/ml.
2.2.8.4 Effect of BGP on Rate of Phagocytosis of Macrophages
lxxxi
The phagocytic ability of macrophage was measured by neutral red uptake (Kim et al., 2004; Chen et al.,
2010). RAW264.7 cells were cultured in 96-well flat-bottom plate at a density of 1×105 cells/well at 37oC
in a 5% CO2 atmosphere for 24 h. Thereafter, the cells were incubated with various concentrations of BGP
(5, 25, 50, and 100 μg/ml) and LPS (1 μg/ml) at 37℃ for 48 h. Neutral red solution (100 μl; 0.01%) was
added and incubated further for 1 h. The supernatant was discarded and the cells were washed twice
with PBS and lysed at room temperature with 100 μl of cell lysate solution (1:1 ratio of ethanol and 0.01%
acetic acid) added into 96-well plate for 2 h. The optical density was measured with a microplate reader
at 570 nm.
ℎ ! "# " " $ #
2.9.0
=
[%. &. ]'()''*(+',(-'
[%. &. ].-)'/,.0+'(123-'*30
Statistical Analysis
To demonstrate statistical significance of data, One-way Analysis of Variance (ANOVA; Fisher LSD post
hoc test) using GraphPad Prism 5 software (GraphPad Software, Inc., San Diego, CA). Generally,
differences between test and control treatments are considered significant at P< 0.05.
lxxxii
CHAPTER THREE
RESULTS
3.1 Extraction
The extraction of the mushroom plant, Pleurotus tuberregium (PT) with hot water gave a yield of 90.2 g
of aqueous extract which represents 18.45% w/w. Further extraction and purification of PT yielded 10 g
(2%,w/w) of the β-D glucan rich polysaccharide fraction (BGP).
3.2 Phytochemical Analysis of Extract and Fraction
Phytochemical tests on P. tuberregium gave positive reactions for alkaloids, acidic compounds,
carbohydrates, flavonoids, proteins, reducing sugars, resins, saponins, steroids, tannins and terpinoids.
The β-glucan rich polysaccharide fraction showed positive reactions for alkaloids, carbohydrates,
glycosides, proteins, saponins and steroids. The phytochemical constituents of PT and BGP are shown in
Table 4.
3.3 Acute Toxicity Test
In the acute toxicity tests in mice, PT and BGPadministered orally at doses up to 5 g/kg body weight did
not cause deaths in the mice after 24 h observation period in the two phases of the tests. There was
generally the absence ofsymptoms of toxicity. The LD50 is therefore deemed to be greater than 5000
mg/kg.
3.4 Effect of Aqueous Extract of Pleurotus tuberregium on Phagocytic Activity in Mice
The phagocytic activity of the reticuloendothelial system invivo which is generally determined by the
carbon clearance test was increased in the treated group of mice in a dose related manner when
compared to the untreated control group.The mean nomalised phagocytic index was increased to 7.47,
lxxxiii
10.07and 7.70 in the groups that were treated with PT (100, 200 or 400 mg/kg). Daily oral administration
of PT (100, 200 or 400 mg/kg), increased the mean phagocytic clearance of colloidal carbon in miceby as
much as 7-fold, 10-fold and 7-folds respectivelywhencompared to the clearance in the untreated group
of mice(Appendix 1, Figure 4).
lxxxiv
Table 4: The Yield and Phytochemical Constituents of Immune Active Extracts and Fractions of
Pleurotus tuberregium.
Extract/Fractions
Yield
Phytochemical Constituents
PT
90.2g
Alkaloids, acidic compounds,
(18.45%w/w)
Carbohydrate, flavonoids, proteins,
Reducing sugar, resins, saponins,
Steriods, tannins, terpinoids
BGP
10 g
Alkaloids, carbohydrates,
(2.0%w/w)
glycosides, Proteins, saponins,
sterols.
Percentage yield in parenthesis
lxxxv
Phagocytic activity
index
20
15
10
5
)
g/
kg
00
(4
PT
PT
(2
00
m
g/
kg
m
m
00
(1
PT
)
)
g/
kg
on
N
U
nt
re
at
ed
co
nt
ro
l
i
0
Treatment
Figure 4: The Effect of Hot Aqueous Extract of Pleurotus tuberregium (PT)on Phagocytic Index in Swiss
Albino Mice
lxxxvi
3.5
The Effect of Hot Aqueous Extract of P. tuberregium on the Relative Spleen Weight of Mice.
Short-term daily oral supplementation with PT (100 and 200 mg/kg) and Noni® (100mg/kg, p.o) for 7 days
caused a remarkable and dose related increase in the mean relative spleen weight of mice. The mean
spleen weight was increased from 0.46±0.04 in the untreated mice group to 0.494±0.012, 0.611±0.026
and 0.567±0.036 in the group that were treated with PT (100 200 and 400 mg/kg) respectively
representing a 6.87%, 32.20% and 22.76% increase (Appendix 2, Figure 5).
Treatment
e
10
0
(4
00
(2
00
(1
00
N
on
i(
PT
PT
PT
N
eg
at
iv
m
m
m
m
g/
kg
)
g/
kg
)
g/
kg
)
g/
kg
)
co
nt
ro
l
Specific spleen weight
(g/100 g body weight)
lxxxvii
0.70
0.65
0.60
0.55
0.50
0.45
0.40
lxxxviii
Figure 5: The Effect of Hot Aqueous Extract of P. tuberregium on the Relative Spleen Weight of Mice.
3.6 The Effect of Pleurotus tuberregium Extracts on Delayed Type Hypersensitivity Response (DTHR)
Treatment with PT (100, 200 and 400 mg/kg) produced a marked stimulation of DTHR induced by
ovalbumin in a dose dependent mannerwith paw thickness increasing from 0.288±0.049 mm,
0.420±0.032 and 0.402±0.101 respectively representing a 1.4%, 47.8% and 40.8% increase when
compared to the untreated control mice group (Appendix 3, Figure 6 ).
In a similar manner, BGP (100 and 200 mg/kg) caused a marked stimulation of DTHR induced by
ovalbumin in mice in a dose dependent manner with paw thickness increasing from 0.618±0.084 mm and
0.658±0.052representing a 36.7% and 45.5% increase (Appendix 4, Figure7). The cell mediated immunity
caused by PT and BGP is comparable to that produced by an immunostimulant drug Morinda citrifolia
(Noni) used as a standard control.
0.6
0.5
0.4
0.3
0.2
kg
)
PT
(4
0
(2
0
00
m
m
g/
gk
g
kg
)
g/
PT
PT
(1
0
0
m
m
N
on
i

(1
00
C
g/
kg
)
0.1
on
tr
ol
Paw thickness (mm)
lxxxix
Treatment
Figure 6: The Effect of Hot Aqueous Extract of Pleurotus tuberregium (PT)on Delayed Type
Hypersensitivity Response in Mice as Measured by Hind Paw Thickness
B
G
P
P
(2
00
m
m
tr
o
l
g/
kg
g/
kg
Treatment
)
)
g/
kg
)
on
(1
00
m
(1
00
i
G
on
B
N
C
Paw thickness (mm)
xc
0.65
0.60
0.55
0.50
0.45
xci
Figure 7: The Effect of Beta (β) Glucan rich Polysacharide Fraction of Pleurotus tuberregium (BGP) on
Delayed Type Hypersensitivity in Mice as Measured by Hind Paw Thickness
3.7
The Effect of Hot of Aqueous Extract of Pleurotus tuberregium(PT) on Primary and Secondary
Humoral Immune Responses to Ovalbumin in Mice
Alternate day oral administration of PT (100, 200 and 400mg/kg body weight) significantly (P<0.05)
caused an increase in the levels of ovalbumin (OVA) – specific total IgG, IgG1 and IgG2a in the sera of
treated mice which were immunized with OVA. The boost in specific immunization was observed on the
14th day after prime immunization – primary response, and on the 21st and 28th day after boost
immunization (ie 7th and 14th day post booster- Secondary response) (Appendix 5, 6, 7) 2-4 folds increases
in anti-OVA total IgG, IgG1 and IgG2a were observed (Figure 8, 9, 10). Maximum boost in the antibody
response immune was observed in the group of mice administered PT 200mg/kg. The increases in antiOVA antibody titers were similar to the increases caused by daily oral administration of Noni® as the
standard immunostimulant agent.
Response in Mice
Primary response
Boost response (7th day)
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
Anti-OVA total IgG titre
(Normalised to preimmune naïve sera)
xcii
12
10
8
6
4
2
0
Boost response (14th day)
Figure 8: The Effect of Hot Aqueous Extract of P. tuberregium (PT) on Ovalbumin Specific Total IgG
response in mice
Primary response
Boost response (7th day)
Boost response (14th day)
Figure 9: The Effect of Hot Aqueous Extract of P. tuberregium (PT)on ovalbumin-specific total IgG1
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
Anti-OVA total IgG1 titre
(Normalised to preimmune naïve sera)
xciii
14
12
10
8
6
4
2
0
response in mice
Primary response
Boost response (7th day)
Figure 10: The Effect of Hot Aqueous Extract of P. tuberregium (PT)on ovalbumin-specific total IgG2a
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
PT (400 mg/kg)
PT (200 mg/kg)
PT (100 mg/kg)
Noni® (100 mg/kg)
Negative control
Anti-OVA total IgG2a titre
(Normalised to preimmune naïve sera)
xciv
8
6
4
2
0
Boost response (14th day)
xcv
3.8
Effect of Βeta (β)-GlucanRichPolysaccharide Fraction of Pleurotus tuberregium (BGP) on
Primary and Secondary Humoral Antibody Responses to Ovalbumin in Mice
In a homologous prime boost schedule, alternate day oral supplementation with BGP (100 and 200mg/kg
body weight) promoted an increase in the anti-OVA antibody titers of specific total IgG, IgG1 and IgG2a in
the sera of mice treated and immunized with OVA. Observation recorded on the 14th day before boost
immunization-primary response, as well as the 7th and 14th day after boost immunization-secondary
response indicated a surge in the anti-OVA specific total IgG, IgG1 and IgG2a antibody sera level
(Appendix 8, 9, 10). Increases as high as 1-4 folds were recorded (Figure 11, 12, 13). Administration of
BGP 200mg/kg body weight was observed to show the maximum boost in the antibody immune
response. The increases in anti-OVA antibody titers were similar to the increases caused by oral
administration with Noni® as standard control. The result was significant at P<0.05.
B
B
G
P
P
ni
G
No
(2
(1
(1
00
00
00
m
m
g/
g/
kg
kg
Treatment
)
)
)
ol
kg
tr
g/
on
m
C
Total IgG titre
xcvi
0.40
0.35
0.30
0.25
0.20
xcvii
Figure 11a: The Effect of β-Glucan Rich PolysaccharideFractionof P. tuberregium on Ovalbumin –
Specific Total IgG Primary Response in Mice
BG
BG
ni
P
P
No
(2
(1
00
00
00
(1
m
m
m
g/
g/
kg
kg
l
Treatment
)
)
)
ro
kg
nt
g/
Co
Total IgG titre
xcviii
1.2
1.0
0.8
0.6
0.4
xcix
Figure 11b: The Effect of Βeta (β) Glucan Rich Polysaccharide Fraction ofP. tuberregium on Ovalbumin –
Specific Total IgG Secondary Response in Mice
N
on
i(
10
0m
g/
kg
)
B
G
P
(1
00
m
g/
kg
)
B
G
P
(2
00
m
g/
kg
)
C
on
tr
ol
IgG titre
c
1.5
1.0
0.5
0.0
Treatment
ci
Figure 12a: The Effect of Βeta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium on Ovalbumin
– Specific IgG1 Primary Response in Mice
3.5
IgG1 titre
3.0
2.5
2.0
1.5
Treatment
m
g/
kg
)
(2
00
B
G
P
B
G
P
(1
00
m
g/
kg
)
m
g/
kg
)
10
0
N
on
i(
N
eg
at
iv
e
co
nt
ro
l
1.0
cii
Figure 12b: The Effect of Βeta (β) Glucan RichPolysaccharide Fraction of P. tuberregium on Ovalbumin –
Specific IgG1 Secondary Response in Mice
N
on
i(
10
0m
g/
kg
)
B
G
P
(1
00
m
g/
kg
B
)
G
P
(2
00
m
g/
kg
)
C
on
tr
ol
IgG2a titre
ciii
1.0
0.8
0.6
0.4
Treatment
civ
Figure 13a: The Effect of Beta (β) Glucan RichPolysaccharideFraction of P. tuberregium
on Ovalbumin – Specific IgG2a Primary Response in Mice
2
1
)
)
kg
kg
m
g/
g/
00
(2
BG
P
(1
P
BG
No
ni
(1
00
00
m
m
g/
nt
kg
ro
)
l
0
Co
IgG2a titre
3
cv
Figure 13b: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium on Ovalbumin
– Specific IgG2a Secondary Response in Mice
cvi
3.9
INVITRO STUDIES
3.9.1
Effect of Βeta (β)-GlucanRich Polysaccharide Fraction of P. tuberregium (BGP) on Spleenocytes
Proliferation
Treatment with BGP (5 - 1000 µg/ml) produced a remarkable proliferation of spleen cells. Treatment of
spleenocytes with BGP produced a marked increase of spleenocytes. Stimulation with BGP (5, 25, 50,
100, 250 and 1000 µg/ml) showed spleenocyte proliferation indices of 1.44±0.02, 1.57±0.01, 1.90±0.05,
2.25±0.14, 1.53±0.07 and 1.28±0.11 respectively representing a 44.4, 57.3, 90.2, 125.1, 53.1, 28.4 and
86.1% increase when compared to the unstimulated control. The response at P< 0.001 was significant
(Appendix 11, Figure 14). The highest spleenocyte proliferation was produced by treatment of cell line
with 100 µg/ml BGP.
The proliferation of spleenocytes produced by β-glucan rich polysaccharide extract of P. tuberregium
(BGP) is comparable to that produced by a Lipopolysaccharide used as a standard mitogen control.
3.9.2
Effect of BGP ON TNF-α Production by RAW264.7
Stimulation of RAW264.7 cells with BGP significantly increased (P<0.001) production of TNF-α by
RAW264.7.Treatment with BGP (5, 25, 50 or100 µg/ml) increased the mean TNF-α levels in culture
supernatant in a concentration-dependent mannerfrom 0.18±0.01 ng/ml in the unstimulated control to
1.180±0.029 ng/ml, 1.580±0.017 ng/ml, 2.064±0.088 ng/ml and 1.987±0.022 ng/ml, respectively. These
represents 5-10 fold increases in the concentration of TNF-α over the unstimulated control (Appendix 12,
Figure 15). Stimulation of TNF-α expression by 50 and 100 µg BGP/ml are comparable to the production
elicited by by LPS (1 µg/ml) used as standard mitogen.
3.9.3
Effect ofBGP on Nitric Oxide Production by Raw264.7
cvii
Treatment with BGPproduced in a significant (P<0.001) increase in the production of nitric oxide by
RAW264.7. Stimulation of RAW264.7 with BGP (5, 25, 50 or100 µg/ml) increased the expression of nitrite
in the culture supernatant from 0.600±0.057 µM in the unstimulated control to 6.933±0.088 µM,
9.767±0.218 µM, 16.200±0.230 µM and 19.670±0.433 µM respectively. These represent 10 – 30 folds
increases in the nitric oxide release compared to the unstimulated control treatment (Appendix 13,
Figure 16). Nitric oxide production by 100 µg BGP/ml is comparable to that elicited by LPS (1µg/ml) used
as a standard mitogen.
3.9.4
Effect ofBGP on the rate ofPhagocytosis (Neutral Red Uptake) RAW264.7
Raw264.7 preteated with BGP (5, 25, 50 or100µg/ml) induced remarkably higher rate of phagocytosis
measured indirectly by neutral red uptake assay. Relative phagocytic indices were increased in RAW264.7
pretreated with BGP from1.000±0.013 in the unstimulated cells to 1.119±0.011, 1.165±0.016, 1.181±
0.001, and 1.083±0.010, respectively. These represent 11.9, 16.5, 18.1, and 8.3% increases compared
mean phagocytic index of untreated control cells (Appendix 14, Figure 17). The highest ratesof phagocytic
indiceswere shown byRAW264.7pretreated with 25 and 50µg BGP/ml and these were similar to the
modest increase in phagocytic index of 14.9 % produce by cells stimulated with LPS (1µg/ml).
cviii
Proliferation index (PI)
(Spleenocytes)
2.50
2.25
2.00
1.75
1.50
1.25
1.00
LP
C
on
tr
S
ol
(1
B
µg
G
/m
P
(5
l)
µg
25 /ml
(µ )
g/
m
50
l
(µ )
g/
10
m
l
0
(µ )
g/
25
m
l
0
(µ )
g/
10
m
00
l
(µ )
g/
m
l)
0.75
Figure 14: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium (BGP) on
Spleenocytes Proliferation
cix
α (ng/ml)
TNFα
2.5
2.0
1.5
1.0
0.5
C
on
LP
tr
ol
S
(1
µ
g/
B
m
G
l)
P
(5
µ
B
g/
G
m
P
l)
(2
5
µ
B
g/
G
m
P
l)
(5
0
µ
B
g/
G
P
m
l)
(1
00
µ
g/
m
l)
0.0
Treatment
Figure 15: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium (BGP) onTNF-α
Production by RAW264.7
cx
20
15
10
5
C
on
LP
tr
ol
S
(1
µg
B
/m
G
l)
P
(5
µg
B
G
/m
P
l)
(2
5
µg
B
G
/m
P
l)
(5
0
B
µg
G
/m
P
(1
l)
00
µg
/m
l)
µ M)
NO conc. (µ
25
Treatment
Figure 16: The Effect of Beta (β) Glucan Rich Polysaccharide Fraction of P. tuberregium (BGP) onNitric
Oxide Production by RAW264.7
cxi
1.1
1.0
0.9
0.8
U
ns
tim
ul
at
LP
ed
S
(1
µg
B
/m
G
P
l)
(5
µg
B
G
/m
P
l)
(2
5
µg
B
G
/m
P
l)
(5
0
B
µg
G
P
/m
(1
l)
00
µg
/m
l)
Phagocytic activity
index
1.2
Treatment
Figure 17: The Effect of Beta Glucan Rich Polysaccharide Fraction of P. tuberregium on Rate of
Phagocytosis by Raw264.7
cxii
cxiii
CHAPTER FOUR
DISCUSSION AND CONCLUSION
4.1 DISCUSSION
In this study the effects of a hot aqueous extract of Pleurotus tuberregium (PT) on some specific and nonspecific immune responses were investigated.Noni®,a polysaccharide-rich precipitate derived from the
fruit of Morinda citrifolia,and beneficial in the treatment of immuno-inflammatory disorders (Hirazumi et
al., 1996; Zhang et al., 2009; Brooks et al., 2009) was used as the positive control in the in vivo
experiments. This popular edible mushroom has been considered as a profound health promoting
mushroom. It is reputed as effective in a number of therapeutic areas (Hui et al., 2012). From the
preliminary phytochemical studies, the crude aqueous extract of P. tuber-regium contains saponins,
alkaloids, flavonoids (aurones, chalcones, flavones, flavonols and leucoanthocyanins), oligigosaccharides
as well as phytates and tannins (Basu et al., 2007; Ijeh et al., 2009). All these have potential health
promoting effects at least under some circumstances (Basu et al., 2007).
In the traditional setting, P. tuberregium has been known for its nutritive and medicinal properties in a
wide range of diseases such as in treating colds, malnutrition, anaemia, obesity, cough, asthma etc.
Modulation of the immune system response through stimulation or suppression may help in maintaining
a disease free state (Ghule et al., 2006).
The crude Pleurotus tuberregiumextract showed significant activity on both the humoral and cell
mediated immune system. This result encouraged further bioactivity guided fractionation of the crude
extract. Many bioactive substances with immunomodulatory properties have been extracted from
different mushroom species which include; β-glucans, lentins, polysaccharides, polysaccharide-peptide
complexes, triterpenoids, nucleosides, and other secondary metabolites (Luiet al., 1998; Eoet al., 1999;
cxiv
Zhu et al., 1999).Most of the bioactive polysaccharides isolated from mushroom are water-soluble (1→3)β-D-glucans with (1→ 6)-β-linked side branches (Schmid et al., 2001). Thus, for the present study, the β-Dglucan-rich polysaccharide fraction of P. tuberregium (BGP) was extracted and subjected to analyses of
immunomodulatory effects in mice and in cultures of monocytic/macrophage cells (RAW267.4).
In general, there were someremarkable effects of PT on theimmune system of treated mice.DTHR
induced by ovalbumin was stimulated by P. tuberregium extract in a dose related manner. DTHR which
was measured as an indicator of T cell mediated immune response assesses the effect of P. tuberregium
(PT) on cell mediated immune response which is initiated by actively sensitized T lymphocytes and is
expressed locally by cellular infiltration and inflammation (Sharma et al., 1996). DTHR is an autoimmune
response mediated by T cells, macrophages and monocytes. It requires the specific recognition of a given
antigen by activated T lymphocytes, which subsequently proliferate and release cytokines. The released
cytokines and other molecules increase vascular permeability, induce vasodilatation, macrophage
accumulation, and activation, promoting increased phagocytic activity and increased concentrations of
lytic enzymes for more effective killing (Elgert, 1996; Descotes, 1999). DTH comprises of two phases, an
initial sensitization phase after the primary contact with ovalbumin antigen. A subsequent exposure to
the ovalbumin antigen induces the effectors phase of the DTH response, where TH1 cells secrete a
variety of cytokines that recruits and activates macrophages and other non-specific inflammatory
mediators. The delay in the onset of the response reflects the time required for the cytokines to induce
the recruitment and activation of macrophages.
Results of this study shows that the extract at all doses increased the immune reaction to ovalbumin
indicating that the extract could affect the cellular component of the local immune reaction including
lymphocytes and monocytes recruited at the site of interaction (Ghazanfaret al., 2002).
cxv
Since cell mediated immune response is known to be critical to defense against infectious organism,
infection of foreign graft, tumor immunity and immunity to many intracellular infections of
microorganisms, especially those carrying chronic disease e.g Tuberculosis and DTHR (Elgert, 1972;Mitra
et al., 1999; Dashputre and Naikwade, 2010; Wahi, 2010), the increase in DTHR indicates that short term
administration of PT could augment the cell mediated immune responses. This implies enhanced ability
to protect host against conditons requiring robust cellular immune protection, such as viral infections,
tumors, and intracellular pathogens.
Similarly, oral supplementation withBGP increased stimulatory activity on the ovalbumin- induced
delayed type hypersensitivity reaction.Oral administration was possible in the study sinceβ-D-glucan is
resistant to gastric acid and pass through the stomach virtually unchanged (Rahar et al., 2011) This result
demonstrates and suggests that the P. tuberregium extract could owe its stimulatory activity on delayed
type hypersensitivity reaction to its abundant beta glucan rich proteo-polysaccharide constituent.
P. tuberregium extract showed an overall stimulatory effect on the reticuloendothelial system as
indicated by the phagocytic index. Phagocytosis is a non-specific immune defense and is very important
in the overall immune protection against a variety of infections (Janeway et al., 2005) as active
phagocytosis is the major defense mechanism against infection. The clearance rate of granular foreign
bodies from circulation reflects the phagocytic function of mononuclear macrophages. Increased carbon
clearance is an indicator of enhanced competency of the reticuloendothelial system and mononuclear
macrophages in removal of foreign particle, dead or injured cells, thereby an indicator of good
immunological response against foreign particles or antigens (Ponkshe and Indap, 2002; Thakur et al.,
2007; Roitt et al., 1993; Furthvan and Bergvanden 1991; Sangle et al., 2004).Pretreatment with P.
tuberregium extract resulted in an increase in clearance rate of carbon from the system of the animals
compared to the untreated mice group. P. tuberregium was found to stimulate the phagoytic activity of
cxvi
the macrophages as evidenced by an increase in the rate of carbon clearance. This activity implies that PT
increased phagocytic responses of host against debris and foreign invaders. Therefore, this result
indicates significant enhancement in the phagocytic function of macrophages hence response of host
against debris and foreign invaders and thus that P. tuberregium may have properties of influencing the
non specific immunity.
The spleen is an important organ of the immune system, and so the level of immune competence of an
organism is closely correlated with the developmental and functional integrity of this organ.Among the
different organs of the immune system, the spleen represents a major secondary lymphoid organ
involved in elicitation of immune response. The relative organ weight is fundamental to diagnose
whether the organ was exposed to an injury or not (Vaghasiya et al., 2010). The relative weight of each
organ of the immune system to the body weight of an individual is a commonly used index to reflect the
developmental status of the organ (Dong et al., 2007). Also in the absence of any infection and obvious
pathology, the relative weight of the spleen reflects overall immune function. Immunostimulatory agents
are known to increase the relative weights of lymphoid organs such as the spleen (Zhang et al., 2011). In
this study, administration of PT showed an increase in organ weight (spleen). Unlike the lymph nodes
which are specialized to trap localized antigen from regional tissue spaces, the spleen serves as a
reservoir for blood and filters, traps blood borne antigen and purifies the blood and lymphatic fluid that
flows through it, and thus respond to systemic infections (Anamika et al., 2010). The increase in spleen
weight may be due to the stimulatory effect of the plant extract on the lymphocytes and bone marrow
haematopoeitic system.
Oral supplemmentattion with aqueous extract of PT and BGP boosted the titres of IgG and its isoforms
(IgG1 and IgG2a). This indicates the enhanced effect of PT and BGP on the antibody producing humoral
immune system of Ova sensitized animals. The estimation of serum immunoglobulin level is a direct
cxvii
measure of humoral immunity and is an index of the functional status of developmental phases of the
humoral immune response viz antigen recognition, activation, and expression (Benacerral, 1978).
Basically humoral immunity is mediated by secreted antibodies and its physiological function is defense
against foreign bodies (Surendra et al., 2010).The IgG antibody isotype provides the majority of antibodybased immunity against invading pathogens. It is remarkable that PTand BGP were able to boost the
titers of IgG and its isoforms (IgG1 and IgG2a) thereby indicating that the antibody-producing humoral
immune system is strengthened with their short-term supplementation.
Spleenocyte proliferation is a crucial event in the activation cascade of both cellular and humoral immune
responses (Borchers et al., 2008) MTT assay thus provides a means of accessing the effect of beta glucan
rich proteo-polysaccharide on cellular immune response (Yang et al., 2011). Resultsfrom the present
study indicates that the proliferation of mice spleenocytes in beta glucan rich proteo-polysaccharide
treated cell lines is significantly increased in a dose dependent manner compared with the control and
concavalin treated spleenocyte cell lines. Stimulation of spleen proliferation could be associated with
elevated production of proinflammatory cytokines by lymphocytes (B cells) as well as increased
production of T cells (Ho et al., 2004). This suggests that BGP may modulate Th1-mediated immune
response (Andres et al., 2011). Also it suggests the possibility of the BGP extract being used as a possible
mitogen in murine spleen lymphocytes since its effect on spleenocyte proliferation was comparably
higher than the standard mitogen concavalin (Ho et al., 2004). Although the surface expression of
molecular activation markers CD69 and CD25 on spleenocytes were not ascertained, the measurement of
the expression of these molecules which are inducible cell surface glycoproteins acquired by immune
cells during lymphoid activation and involved in lymphocyte proliferation in further research studies may
shed more light on the ability of such treatment with beta glucan rich proteo-polysaccharide to produce
activation of these important immune cells (Nworu et al., 2012).
cxviii
Result shows that the production of nitric oxide by monocytic/ macrophage cell line (RAW264.7) in beta
glucan rich proteo-polysaccharide treated cell lines was significantly elevated in a dose dependent
manner compared with the control mice group and lipopolysaccharide treated spleenocyte cell lines.
Lipopolysaccharide (LPS) is a potent inducer of inflammation in cell lines and systemically in whole animal
experiment (Tokunaka et al., 2000). Particularly, LPS can quantitatively promote the stimulation of
macrophages to secrete pro-inflammatory cytokines and secondary mediatory, such as nitric oxide (NO)
as well as synthesis of reactive oxygen intermediates (Kilbourn et al., 1984; Duval et al., 1996).NO is a
gaseous molecule synthesized from L-arginine by nitric oxide systhase (NOS) (Xle et al., 1992; Korhonen
et al., 2005). It is a highly reactive free radical and it can form a number of oxidation products such as
NO2, NO2¯, N2O3 and S-nitrosothiols. NO participates in the physiology and pathophysiology of many
systems (Diouf et al., 2009). It is an important mediator of the non-specific host defense against invading
microbes and tumors. Thus NO can be used as a quantitative index of macrophage activation. It mediates
diverse functions including vasodilation, neurotransmission and inflammation. The role of NO in host
defense against microorganisms and tumor cells is well recognised. NO has been shown to be the
principal effector molecule produced by macrophages for cytotoxic activity and thus a quantitative index
of macrophage activation (Kilbourn et al., 1984; Ding et al., 1988).Although the mechanism was not
evaluated, it is speculative that part of the immunomodulatory activity of BGP extract may be due to
activation of macrophages through augmentation in NO production and other cytokines by induction of
iNOS mRNA gene expression.This is in agreement withseveral investigations indicating the biological
properties of the extracts/or compounds derived from other medicinal plants occurs through this
mechanism (Stimpel et al., 1984; Kang et al., 2002; Park et al., 2004; Kim et al., 2007).In addition, the
increased carbon clearance index invivo which reflects the enhancement of phagocytic function of
mononuclear macrophages and non specific immunity probably act through the release of NO as
previously described in the invivo carbon clearance model .
cxix
Inflammation is a complex biological response of the body to harmful stimuli including microbial
infections and chemical toxins. The inflammatory response is characterized by several steps including
coordinate activation of signaling pathways, expression of proinflammatory cytokines, chemokines and
adhesion molecules in resident tissue cells, as well as infiltration of leukocytes mainly macrophages,
neutrophils and dendritic cells and mediators of inflammation from the vascular system to remove the
harmful stimuli and to initiate the healing process (Sacca et al., 1997; Dinarello, 2010).Leukocytes or
endothelial cells produce proinflammatory or anti-inflammatory cytokines depending on their regulatory
activities on regulating inflammation. Proinflammatory cytokines includetumor necrosis factor-α (TNF-α)
and interleukin-1β (IL-1β). Tumor necrosis factor-α (TNF-α) have been shown to play central roles in the
initiation and amplification of inflammatory response (Hehlgans and Pfeffer, 2005; Park and Bowers,
2010). The effect of treatment with BGP on the expression of key inflammatory cytokine tumor necrosis
factor-α (TNF-α) was evaluated and found to increase its production.TNF- α has being implicated in
inflammatory processes. Inflammatory response is characterized by increased blood supply carrying more
leukocytes (mainly activated monocytes/macrophages) and plasma molecules, increased permeability of
capillaries allowing exudation of plasma proteins and increased migration of leukocytes to the affected
tissue (Male et al., 2007). These activated macrophages produce TNF- α, IL-1, IL-6 and IL-12 (Evans, 1996;
Bone et al., 1997). In order to initialize the inflammatory response, TNF-α and IL-1 induce vasodilation,
increase vascular permeability and increase blood flow to the affected site thereby resulting in edema,
redness and heat (Thao et al., 2008, Nworu et al., 2010; Nworu et al., 2011) leading to subsequent
elimination of the antigen. Helper T cells are designated as Th1 and Th2 subpopulations based on the
patterns of their cytokine secretion. Th1 cells, characterized by IL-2, IFNγ, and TNFα production, are
mainly involved in cell-mediated immunity to intracellular infection (Borchers et al., 2004). Enhanced Th1
response is beneficial for antimicrobial and antitumor defense, increased innate immunity to tumors and
cxx
viral infections and it is likely responsible for the immunomodulatory activity of the extract as mediated
by TNFα though this was not ascertained specifically.
BGP significantly increased the proliferation of monocytic/macrophage cell line (RAW264.7) in a dose
dependent manner. The result is comparable with the lipopolisaccharide proliferation of
monocytic/macrophage cell line (RAW264.7). Macrophages are the first line of defense in the interaction
between innate and specific immunity against infection (Yoon et al.,2003;Allan and Anders, 2008; Zhao et
al., 2010). They therefore play an important role in host defense against infections (Yu et al., 2008). The
extent of phagocytosis is a major indicator of macrophage effector activity as well as an important
indispensable step in the immunological defense system (Campelo et al., 2002). Increased proliferation of
monocytic/macrophage cell line (RAW264.7) further indicates the activity of BGP on cell mediated
immunity.
β-D-Glucans are recognized by “pattern recognition receptors” (PRR) in innate immune cells, including
dectin-1, complement receptor-3 (CR3 or CD11b/CD18) and TLR2/6, thereby activating and triggering
responses in several immune cells including monocytes, macrophages, dendritic cells, neutrophils, and
NK cells (Brown and Gordon, 2001). It is therefore plausible to suggest that upon stimulation by β-DGlucans content of the mushroom extracts through the PRR, macrophages exhibit increased expression
of iNO, TNFα, and phagocytic capability which are signs of improved functional maturation (Brown et al.,
2002).
These in vitro results with BGP strongly agrees with the in vivo models using the crude extract of Pleurotus
tuberregium (PT). This is also indicative of the better and enhanced immunomodulatory potentials of the BGP.
These benefits might be replicated in humans taking sufficient P. tuberregium for an appropriate period of time.
However, the clinical immunomodulatory potential of PT and BGPrequires further study.
4.2 CONCLUSION
cxxi
AlthoughP. tuberregium is a popular folk medicine and has attracted great attention due to its role in the
immune system activity against infections, there is little information available about this action. This
report provides both in vitro and in vivo evidences of the therapeutic potentials of this medicinal
mushroom plant.
This study has shown that the PTpossesses immunomodulatory properties. Pleurotus tuberregium extract
showed specific and non-specific immunomodulatory activity on both innate and acquired immune
mechanisms. The study also revealed that BGP, a glycopolysaccharide was responsible for the
immunodulatory activities of the mushroom P. tuberregium. BGP was shown to possess stimulatory
effects on the innate and adaptive components of the immune system depending on the type of immune
cells involved. These properties of P. tuberregium are clinically important and could be employed to
regulate normal immunologic functioning. Thus cellular and humoral immunomodulatory mechanisms of
action for the mushroom, P. tuberregium, justifies its inclusion in dietary supplement and local use in
food. Hence the cultivation and consumption of this Oyster mushroom should be encouraged not only for
its nutritive value but also for the health benefits in strengthening the immune system.
cxxii
REFERENCES
Abbas Ak, Murphy K, Sher A (1996). Functional diversity of helper T lymphocytes.Nature383(6603):78793 PMID 8893001.
Abbas AK, Lichtman AH, Jordan PS (1994). The Complement system; In: cellular and molecular
immunology, WB Saunders, Philadelphia, p. 260-281.
Abbas AK, Litchman AH (2004).Cellular and molecular immunology.Molecular Biology Education, 32:6566.
Adachi, Y, Okazaki M, Ohno N, Yadomae Y, (1994). Enhancement of cytokine production by macrophages
stimulated with (1-3)-D glucan grifolan (GRN), Isolated from Grifola frondosa.Biological and
PharmaceuticalBulletin., 17:1554-60.
Akpaja EO, Iskhuemhen OS, Okhuoya JA (2003). Ethno mycology and uses of edible and medicinal
mushrooms among the Igbo people of Nigeria. International Journal of Medicinal Mushroom
5:313-319.
Alberts B, Alexander J, Julian L, Martin R, Keith Roberts, Peter W (2002). Molecular biology of the cell:
Fourth Edition. Garland Science, New York and London ISBN 0-8153-3218-1.
Allan R, Anders HJ (2008). The role of innate immunityin autoimmune tissue injury.Current Opinion in
Rheumatology20:538-44
cxxiii
Allman D, Srivastava B, Lindsley R (2004). Alternative routes to maturity.branch points and pathways for
generating follicular and marginal zone b cell. Immunological Review197:147-60.
Alobo AP (2003). Proximate composition and functional properties of Pleurotus tuber-regium Sclerotia
flour and protein concentrate. Plant foods for human nutrition58(3):1-9
Anamika G., Manish KG,, Rahus KS, Vijaykumar M, Rao CHV, Goel Rk, Shampa A (2010).
Immunomodulatory effect of Morgina Oleifera Lam. extract on cyclophosphamide induced
toxicity in mice.Indian Journal of Experimental Boilogy 48:1157-1160.
Anderson JW(2003). Whole grains protect against atherosclerotic cardiovascular disease. Proceedings of
Nutrition Society62 (1): 135-42
Andres J, Shailesh D, Quing-Li W, James S, Daniel S (2011). Anti-inflammatory activity of edible Oyster
mushroom is mediated through inhibition of NF-KB and AP-1 Signaling. Nutrition Journal10:52.
Arora D (1986). Mushrooms demystified. Berkeley: Ten Speed Press.
Badger AM (1983). Development in industrial microbiology, proceedings of the 40th general meeting of
the Society for industrial microbiology, Sarasota, FL. Nash CH and underKofler LA, eds., Arlington
VA 25:274.
cxxiv
Barcotti A, Welker RD, Piccoli M, Schlick E, Papademetriou Hartung K, Chirigos MA (1984).
Immunosuppressive effects of human chorionic gonadotropin on delayed type hypersensitivity in
mice In : Immune Modulation Agents and their Mechanisms. Fenichel RL and Chirigos MA (eds)
Marcel Decker Inc New York and Basel. 25: 597-605
Basu SK, Thomas JE, Acharya SN (2007).Prospects for growth in global nutraceutical and functional food
markets : a Canadian Perspective” Australian Journal of Basic and Applied Science 1(4):637-649
Baur SK, Geisler G (1996). Variability of beta glucan content in oat caryposis of 132 cultivated oat
Genotypes and 39 wild oat genotypes. Journal of Agronomy and Crop Science 176:151-7.
Beck L, Spiegelberg HL (1989). The Polyclonal and antigen-specific IgG and IgEsubclass reponse of mice
injected with ovalbumin in alum or complete Freund’s adjuvant. Cell Immunol 123: 1 - 8
Benacerral R (1978). A Hypothesis to relate the specificity of T-Lymphocytes and the activity of I Region
and Ir genes in macrophages and B-lymphocytes. Journal of Immunology 120:1809-1812.
Bo L, Bau Y (1980). Fungi Pharmacopoeia (Sinica). Oakland: Kinoko Co.
Bohn JA, Miller JN (1995). B (1-3) Glucan as biological response modifies: a review of structure functional
activity relationships. Frontiers of carbohydrate research28:3-14.
Bona CA, Bonilla FA (1995). Textbook of Immunology, 2nd Edition, Hardwood: Academic publishers, ISBN
371850596, p.1-431
cxxv
Bone RC, Grodzin CJ, Balk RA (1997). Sepsis: a new hypothesis for pathogenesis of the disease process.
Chest 112: 235 -243.
Borchers AT, Stern JS, Hackman RM, Keen CL, Greshwin ME (1999).Mushrooms, tumors and
immunity.Proceedings of the Society of Experimental Biology and Medicine 221:281–293.
Borchers AT, Keen CL, Gershwin ME (2004). Mushrooms, Tumor and Immunity: An Update.Experimental
Boilogy and Medicine (Maywood) 229(5):393-406.
Borchers AT, Krishnamurthy A, Keen CH, Meyers FJ, Gershwin ME (2008).The Immunobiology of
mushroom.Experimental Biology and Medicine233(3):259-76. PMID 18296732.
Brooks VJ, Schafer M, Sharp P, Xu J, Cai J, Keuler NS, Godbee RG, Peek SF, Schultz RD,
Darien BJ. (2009). Effects of Morinda citrifolia (Noni) on CD4+ and CD8+ T-
Suresh
M,
cellactivation in neonatal
calves.Professional Animal Scientist20:202–206.
Brown GD, Gordon S. (2001) Immune recognition. A new receptor for β-glucans.Nature413
(6851):
36–37.
Brown GD, Taylor PR, Reid DM, Willment JA, Williams DL, Martinez- Pomares L, Wong SY,
S.
(2002).
Dectin-1
is
a
Major
β-glucan
Receptor
on
Macrophages.
Gordon
Journal
of
ExperimentalMedicine196(3): 407-12.
Brustein CG, McGlave PB (2001). The biology and treatment of Chronic Myclogeneous
Leukaemia.Oncology 1:23-31
cxxvi
Campelo MS,Oliveira, MB, Leão AM, Carbonero ER, Gorin PA, Iacomini M (2002). Effect of a Soluble α-Dglucan from the lichenized fungus Ramalina celastri on macrophage activity. International
Immunopharmacology 2: 691-698.
Celine L, Philippe M (2007). New developments and prospective applications for β (1,3) Glucans. Recent
Advances on Biotechnology1:59-73.
Chandra RK (1997). Nutrition and immune system: An Introduction. American Journal of Clinical
Nutrition66: 4605-4635. PMID 9250133.
Chen JR, Yang ZQ, Hu TJ, Yan ZT, Niu TX, Wang L. (2010). Immunomodulatory activity in vitro and in vivo
of polysaccharide from Potentilla anserina.Fitoterapia 81:1117-1124.
Cheung, Pc, Lee MY (2000). Fractionalization and characterization of mushroom dietary (Non Starch
polysaccharides) as Potential Nutraceuticals from Sclerotia of Pleurotus tuberregium
(Fries).Singer Journal of Agricultural and Food Chemistry 48(8):3148-3151.
Chang K (1981). Lenitinan-mediated resistance against VSV-encephalitis, abalson virus-induced tumor,
and trophoblastic tumor in mice.In Manipulation of hot defense mechanisms. Aoki T (eds.).
Amsterdam: Excerpta medica (Internationa Congress Series 576)
Cramer DA, Allendorf DJ, Baran JT, Hasen J, Marroquin J, LI B, Ratajczak J, Ratajczak MZ, Yan J (2006).
Beta glucan enhances complement - mediated hematopoietic recovery after bone marrow injury.
Blood 107:835-40. PMID 161793702005
cxxvii
Dahech I, Belghith Ks, Hamden K, Peki A, Belghith H, Mejdoub H (2011). Oral administration of levan
polysaccharide reduces the alloxan-induced oxidative stress in rats. International Journal of
Biological Macromolecules49(3):942-947.
Dalia A, Anatolijus K, Janina D, Egidijus K (2007). Effects of β-glucans on the immune system.Medicinal
(Kaunas) 43(8):597-606.
Dashputre NL, Naikwade NS (2010). Immunomodulatory activity of Abutilon Indicum linn on Albino
Mice.International Journal of PharmaSciences and Research.1(3):178-184.
Davis
L,
Kuttan
G.
(2000).Immunomodulatory
activity
of
Withania
somnifera.Journal
of
Ethnopharmacology71(1-2): 193–200
De Jesus M, Ahlawat S, Mantis N (2013). Isolating and immunostaining lymphocytes and dendritic cells
from murine peyer’s patches. Journal Visualised Experiment (73); e50167, doi:103791/50167
Delves PJ, Roitt IM (2000). Understanding modern immunology: The development of the human immune
system. N Engl J Med. 343(1):37-49.
Descotes J (1999). An introduction to immunotoxicology.Taylor, Francis editors.London 235.
Dharmananda S(1988). Medicinal mushrooms. Bestways, p. 54-58
cxxviii
Dinarello CA (2009). Immunological and inflammatory functions of the interleukin – 1 – Family.Annual
Reviews of Immunology27:519-50.
Dinarello CA (2010). Anti-inflammatory agents: present and future. Cell 140: 935-950
Ding AH, Nathan CF, Stuehr DJ (1988). Release of reactive nitrogen intermediates and reactive oxygen
intermediates from mouse peritoneal macrophages: comparisons of activating cytokines and
evidence for independent productionJournal of Immunology 141.
Diouf PN, Stevanovic T, Boutin Y (2009). The effect of extraction process on polyphenol content,
triterpene composition and bioactivity of yellow birch (Betula alleghaniensis Britton)
extracts.Industrial Crops and Products. 30: 297-303.
Dixon RA (2001).Natural products and plants disease resistance.Nature411:843-847.
Dong XF, SK Wang, Rn SA, Zhang Q, Tong JM (2007). Effect of immunosuppressant cyclophosphamide on
performance and incretion in broiler.Acta Veterinaria et Zootechnica Sinica. 38: 993-998.
Duval D L, Miller DR, Collier J, Billing RE (1996). Characteristics of hepatic nitric oxide synthase:
identification
as
the
cytokine
inducible
form
primarily
regulated
by
oxidants.MolecularPharmacology50:277-284.
Elgert KD (1972). Immunology: Understanding the immune system. Clinical Immunologyand
Immunopathology52:50-61.
Elgert KD (1996). Immunology understanding the immune system.Wiley (NY) 306
cxxix
Eo SK, Kim YS, Lee CK, Han SS. (1999). Antiviral activities of various water
and methanol soluble
substances isolated from Ganoderma lucidum. Journal ofEthnopharmacology68: 129–36.
Evans TJ (1996). Therole of macrophages in septic shock.Immunobiology 195: 6559
Evans WC. (2009). Trease and Evans Pharmacognosy, 16th ed. Edinburgh: WB Saunders, 616
Fasidi IA, Olorunmaiye KS (1994).Studies on the requirements for vegetative growth ofPleurotus
tuberregim (Fr.) Singer, a Nigerian Mushroom.Food Chemistry50:397-401.
Fillipie M, Umek A (2002). Screening of basidomycetes mushroom extracts for antigenotoxic and
biomutagenic activity.Die Pharmazie 57: 416-420
Fimmel S, Zouboulis CC (2005). Influence of physiological androgen levels on wound healing and immune
status in men. Aging Male8(3-4): 166-174. PMID 16390741.
Fraker PJ, Gershwin ME, Good RA, Prasad A (1986). Interrelationships between zinc and immune
function.Federation Proceedings Journal 45: 1474-9.
Francis K, Van Beck J, Neal JW, Gasque P (2003). Activation and regulation of the complement system;
Expert Reviews in Molecular Medicine.5(15): 1 - 19
Fu YX, Chaplain DD (1999). Development and maturation of secondary lymphoid tissues.Annual Review of
Immunology17:399-433.
cxxx
Furthvan R, Bergvanden BM (1991). Clinical Immunology. Ist Ed London: Gower Med. Pub
Ghazanfar T, Hassan ZM, Ebrahimi M (2002). Immunomodulatory activity of a protein isolated from garlic
extract on delayed type by hypersensitivity. International Immunopharmacology2:1541-9.
Ghule BV, Murugananthan G, Nakhat PD, Yeole PG (2006).Immunostimulant effect of Capparis zeylanica
Linn Leaves.Journal ofEthnopharmcol108:311-315.
Girardi M (2006). Immunosurveillance and immunoregulation by YðT cells.Journal of Investigative
Dermatology126(1): 25-31, PMID 16417214.
Goldsby RA, Kindt TJ, Osborne BA (2000). Kuby Immunology. AWH Freeman’s e book, Custom Publishing
Green LC (1981). Nitrite Biosynthesis in Man. Proceedings of the National Academy of Science18: 77647768.
Harada T, Miura N, Adachi Y, Nakajima M, Yadomae T, Ohno N (2002). Effect of SCG, 1, 3-B-glucan from
Sparassis Crispa on the hematopoietic response in cyclophosphamide induced leukopenic mice.
Biology PharmaceuticalBuletinl25:931-9.
Harborne JB (1998). Phytochemical methods – A guide to modern techniques of plant analysis. 1st edn:
Chapman and Hall; London, Uk
cxxxi
Harty J, Tvinnereim A, White D (2000).CD8+ T cell effector mechanisms in resistance to infection.Annual
Review of Immunology18:275-308 PMID 10837060.
Hehlgans T, Pfeffer K (2005). The intriguing biology of the tumour necrosis factor/tumour necrosis factor
receptor superfamily: players, rules and the games.Immunology115: 1-20
Hibbett DS, Thorn RG (1994).Nematode trapping in Pleurotus tuberregium. Mycologia 86:696-699.
Hirazumi A, Furusawa E, Chou SC, Hokama Y. (1996). Immunomodulation contributes to the anticancer
activity of Morinda citrifolia (Noni) Fruit Juice. Proceedings of the WesternPharmacology Society
39:7–9.
Ho CY, Clara BS, Kim CF, Leung KN, Fung KP, Tse TF (2004). Differential effect of Corilus versicolor (Yun
Zhi)
extract
on
cytokine
production
by
murine
lymphocytesinvitro.
International
Immunopharmacology4(12):1549-1557.
Ho JCK, Konerding MA, Gaumann A, Groth M, Liu WK (2004). Fungal polysaccharopeptide inhibits tumor
angiogenesis and tumor growth in mice. Life Science 75:1343-56.
Hogan S, Rosenberg H, Moqbel R, Phipps S, Foster PS, Lacy P, Kay Ab, Rothenberg ME (2008). Eosinophils:
biological properties and role in health and disease. Clinical and Experimental Allergy 38(5):70950.
Hoofinagle JH, Di BM (1997). The treatment of chronic viral hepatitis. New England Journal
ofMedicine336:347-56.
cxxxii
Hu SH, Wang JC, Lien J, Liaw ET, Lee MY (2006).Antihyperglycemic effect of polysaccharide from
fermented broth of Pleurotus citrinopileatus.Applied Microbiology and Biotechnology 70(1):107113.
Huang NL (2002).Inonotus obliquin” Edible fungi of China.21(7): 7-8
Hui-Yu, Mallikarjuna K, Ying-Ying C, Ting-Yi, Ying Chich T (2012).Pleurotus tuber-regium Polysaccharides
Attenuates Hyperglycemia and Oxidative Stress in Experimental Diabetic Rats.Evidence-Based
Complementary and Alternative Medicine. Article ID 856381.
Ijeh I, Okwujiako I, Nwosu P, Nnodim H (2009). Phytochemical composition of Pleurotus tuber-regium and
effects of the dietary incorporation on body/organ weights and serum trigcylglycerols role in
albino mice.Journal of Medicinal Plants Research 3(11):939-943.
Isikhuemhen OJ, Okhuoya JA (1995). A low cost technique for the cultivation of Pleurotus tuber-regium
(Fr) Sing in developing tropical countries. Mushroom Growers Newsletter 4:2-4.
Isikhuemhen OS, Moncalvo JM, Nerud F, Vilgalys R (2000a).Mating compatibility and psycho geography in
Pleurotus tuberregium.Mycological Research 104:732-737.
Isikhuemhen OS, Nerud F, Vilgalys R (2000b). Cultivation studies on wild and hybrid strains of Pleurotus
tuberregium (Fr) Sing on wheat straw substrate. World Journal of Microbiology and
Biotechnology 16:431-435.
Jacqueline P, Bryony C. (2001). Immunology.The Lancet357:1777-89.
cxxxiii
Jacques B, Ralph M (1998). Dendritic cells and the control of immunity.Nature Vol 392; Macmillan
Publishers Ltd 245-252.
Janeway CA, Walport M, Travers P (2005). Immunobiology: the immune system in health and disease, 6th
Edition, Garland Science Publishing, ISBN 0815341016, p. 1-848.
Jin M, Jeon H, Hund HJ, Kim B, Shin SS, Choi JJ, lee JK, Kang CY, Kim S(2003). Enhancement of
repopulation and hematopoiesis of bone marrow cells in irradiated mice by oral administration of
PG101,
a
water-soluble
extract
from
Lentinus
leoideus.
Experimental
Biology
andMedicine228:759-66.
Jones K. (1998). Maitake: A Potent medicinal food. Alternate and ComplementaryTherapy4(6): 420-429
Jong SC, Birmingham JM (1992). Medicinal benefits of the mushroom ganoderma. Advancesin Applied
Microbiology 37:101-134.
Kadiri M, Fasidi IO (1990). Studies on enzyme activities of Pleurotus tuberregium (Fr.) Singer and
Tricholoma lobayensis Heim at various fruit body stages. Food/Nahrung34(8):695-699.
Kang NS, Park SY, Lee KR, Lee SM, Lee BG, Shin DH (2002). Modulation of macrophage function activity by
ethanolic extract of larvae of Holotrichia diomphalia.Journal ofEthnopharmacology79:89-94
Khanittha P, Christopher P, Usanee V (2004). Thai medicinal plant modulate nitric oxide and tumour
necrosis factor – α in J774.2 mouse macrophage. Journal of Ethnopharmacology 95(2-3): 183 189
cxxxiv
Kilbourn RG, Klostergaard J, Lopez -Berestein G (1984). Activatedmacrophages secrete a soluble factor
that inhibits mitochondrial respiration of tumor cells. Journal ofImmunology133:2577-81
Kim AJ, Kim YO, Shim JS, Hwang JK (2007). Immunostimulating activity of crude polysaccharide extract
isolated from Curcuma xanthorrhiza Roxb. Bioscience Biotechnology Biochemistry71:1428-38.
Kim GY, Choi GS, Lee SH, Park YM (2004). Acidic polysaccharide isolated from phellinus linteus enhances
through the up regulation of nitric oxide and tumor necrosis factor-alpha from peritoneal
macrophages. Journal of Ethnopharmacology 95:69-76.
Kim, YO, Han SB, Lee HW, Ahn HJ, Yoon YD, Jung JK, Kim HM, Shin CS. (2005). Immuno-stimulating effect
of the endo-polysaccharide produced by submerged culture of Inonotus obliquus. Life Science 77:
2438-2456.
Kodama N, Asakawa A, Inuia, Masuda Y, Nanba H (2005). Enhancement of cytotoxicity of NK cells by dfraction, a polysaccharide from Grifolia frondosa. Oncology Reports 13:497-502.
Kodama N, Komuta K, Sakai N, Nanba H (2002). Effects of D-fraction a polysacchrides from Grifolia
frondosa on tumor growth involve activation of nk cells. Biologyand Pharmaceutical Bulletin:
25:1647-50.
Korhonen R, Lahti A, Kankaanranta H, Malanen E (2005).Nitric oxide production and signaling in
inflammation.Current Drug Targets – Inflammation and. Allergy 4:471-479.
cxxxv
Kovacs B, Maus M, Riley J, Derimanov G, Koretzky G, June C, Finkel T (2002). Human CD8 + T cells do not
require the polarization of lipid raffs for activation and proliferation. Proceedings National
Academy of Science USA 99(23): 15006-11. PMID 12419850.
Lavigne LM, Albina JE, Reichner JS (2006). Beta-glucan is a fungal determinant for adhesion dependent
human neutrophil functions. Journal ofImmunology177:8667-867.
Leblanc BW, Albina JE, Reichner JS (2006). The Effect of PGG-(Beta)-glucan on neutrophil chemotaxis
invivo. Journal of Leukocyte Biology 76:667-75.
Lee BR, Lee YP, Kim DW, Song HY, Yoo KY, Won MH, Kang TC, Lee KJ, Kim KH, Joo JH, Ham HJ, Hur JH, Cho
SW, Han KH, Lee KS, Park J, Eum WS, Choi SY (2010). Amelioration of streptozotocin-induced
diabetes by agrocybe chaxingu polysaccharide.Molecules and Cells 29(4):349-354.
Lee YS, Nakahara K, Pham JW, Kim K, He Z, Sontheimer EJ, Carthew RW (2004).Distinct roles for
drosophilia dicer-1 and dicer-2 in the SI RNA/MI RNA silencing pathways.Cells117 (1): 69-81
Levay PF, Viljoen M (1995). Lactoferrin: A general review. Haematological80(3):252-67.
Lin K, kao Y, Kuo H, Yang W, Chou A, Lin H, Yu AL, Wong C (2006). Reishi polysaccharide induce
immunoglobulin productionthrough the TLR4/TLR2-mediated induction of transcription factor
blimp-I. Journal ofBiological Chemistry281(34):24111-23
Liszewski MK, Atkinson JP (1993). The complement system: In Paul WE, Ed. Fundamental Immunology, 3rd
Edition, Ramen Press Ltd., New York, P. 917-939.
cxxxvi
Lorke D (1983). A New Approach to Practical Acute Toxicity testing. Archives of Toxicology54(4): 275-87
Lui C, Lu S, Ji MR (1992). Effects of Cordyceps sinensis on invitro Natural Killer Cells.Zhongguo Zhong Xi Yi
Jie He Za Zhi12(5): 267-269
Lui G (1993). Pharmacology and clinical uses of ganoderma from mushroom biology and mushroom
products. Chang S. (eds.). The Chinese University Press, Hong Kong, 267-273
Liu KC, Phounsavan SF, Huang RL, Liao C, Hsu SY, Wang KJ.(1998). Pharmacological and liver
functional studies on mycelium of Ganoderma lucidumChinese Pharmaceutical Journal.
40: 21–9.
Male D, Brostoff J, Roth D, Roitt, I (2007). Immunology. (7th ed.). Philadelphia: Elsevier Ltd.
Matzinger P (1994). Tolerance, danger and the extended family.Annual Review in Immunology12:9911045.
Matzinger P (1998). An innate sense of danger.Seminars in Immunology10:399-415.
Miller J (1993). Self-non self discrimination and tolerance in T and B lymphocytes. Immunologic
Research12 (2): 115-30.
Min AD, Jones JL, Esposito S, Lebovics E, Jacobson IM, Klion FM, Goldman IS, Geders JM,Tobias H, Bodian
C, Bodenheimer HC (2001). Efficacy of high dose interferon in combination with ribavirin in
cxxxvii
patients
with
chronic
hepatatis
c
resistant
to
interferon.American
Journal
ofGastroenteroogyl96:1143-1149
Mitra SK, Gupta M, Sarma DN (1999). Immunomodulatory effect of IM-133.Phytotherapy Research
13:341-3.
Miyazaki T, Nishijima M (1981).Studies on fungal polysaccharides. XXVII. structural examination of a
water soluble, antitumor polysaccharide of Ganoderma lucidum. Chemical and. Pharmaceutical
Bulletin. 29:3611-3616
Mosser DM, Edwards JP (2008).Exploring the full spectrum of macrophage activation.Nature Reviews
Immunology 8(12):958-69.
Mossman T (1983).Rapid colorimetric assay for cellular and survival: Application to proliferation and
cytotoxicity assays Journal of ImmunologicalMethods 65: 55-63
Nagler AC (2001). Man the Barrier! strategic defenses in the intestinal Mucosa. Nature Reviews
Immunology1:59-67.
Narui T, Takahashi K, Kobayashi M, Shibata S (1980). Polysaccharide produced by laboratory cultivation of
Poria cocos Wolf. Carbohydrate Research 87: 161-163
Naved T, Siddiqui JI, Ansari SH, Ansari AA, Mukhtar HM (2005). Immunomodulatory activity of Magnifera
indica L, fruits (cv Neelam). Journal of Naural Remedies 5 (2):137-140.
cxxxviii
Ngai PH, Ng TB (2006). A Hemolysin from the mushroom Pleurotus eryngii.Applied Microbiology
Biotechnology 75: 1185-1191
Nworu CN, Akah, P, Okoye BC, Onwuakagba J, Okorafor UU, Esimone C (2012). Supplementation with
aqueous leaf extract of Morinda lucida enhances immunorestoration and up regulates the
expression of cytokines and immunostimulatory markers.Immunological Investigations41(8):
799–819
Nworu CS, Peter A, Festus BC, Donatien KT, Judith Udeh, Charles E (2011). The leaf extract of Spondias
mombin L. displays an anti-inflammatory effect and suppresses inducible formation of tumor
necreosis factor-alpha and nitric oxide. Journal of Immunotoxicology 8(1):10-16.
Nworu CS, Temchura V, Okoye FB, Akah PA, Esimone CO, Uberla K (2010). Activation of murine
lymphocytes and modulation of macrophage functions by fractions of Alchornea cordifolia
(Euphorbiaceae) leaf extract. Immunopharmacology Immunotoxicology.32(1):28-36.
Nworu CS, Ihim SA, Ugwu LE, Laiyemo KA, Akah, PA. (2014). Hepato- and-Nephroprotective Activities of a
Nigerian Local King Tuber Oyster Mushroom, Pleurotus tuberregium (Higher Basidiomycetes), in
Chemically-Induced Organ Toxicities in Rats. InternationalJournal of Medicinal Mushroom16(4):
305-318
Okhuoya JA, Okogbo FO (1990). Induction of edible sclerotia of Pleurotus tuber-regium (Fr) Sing in the
Laboratory.Annals ofApplied Biology117:295-298.
Okhuoya J, Okogbo F (1991). Cultivation of Pleurotus tuber-regium (Fr) Sing on Various Farm
Wastes.Proceedings of the Oklahoma Academy ofScience71:1-3.
cxxxix
Okhuoya JA, Isikhuemhen Os, Evue GA (1998). Pleurotus tuber-regium (Fr.) Sing, Sclerotia and
Sporophore yield during the cultivation on sawdust of different woody plants. International
Journal of Mushroom Sciences2:41-46.
Oso BA (1977). Pleurotus tuber-regium from Nigeria. Mycologia 69(2):271-279.
Page RC, Davies P, Allison AC (1978). The macrophage as a secretory cell.International Review of
Cytology52: 119-123
Pancer Z, Cooper M (2006). The evolution of adaptive immunity. Annual Reviews in Immunology24:497518. PMID 16551257.
Park K, Bowers W (2010). Tumor necrosis factor-alpha mediated signaling in neuronal homeostasis and
dysfunction. Cell Signal22: 977-983.
Park BW, Shin JW, Cho JH, Son CG, Lee YW, Yoo HS, et al., (2004). Effects of Armillaria mellea extract on
macrophage and NK cell activity. Korean Journal of Oriental Medicine25:161-70
Peakman M, Vergani D (1997). Basic and clinical immunology, Churchill Livingstone, London, ISBN
9780443046728.
Pier GB, Lyczak JB, Wetzler LM (2004).Immunology, Infection and Immunity. Asm Press. ISBN 1-55581246-5
cxl
Ponkshe C, Indap MM (2002).Invivo and invitro evaluation for immunomodulatory activity of three
marine animal extracts with reference to phagocytosis.Indian Journal of Experimental Biology
40:1399-402
Pugh N, Ross SA, ElSohly MA, Pasco DS (2001).Characterization of aloeride, a new high-molecular-weight
polysaccharide from Aloe vera with potent immunostimulatory activity.Journal of Agricultural
and Food Chemistry49:1030–1034.
Radoja S, Frey A, Vukmanovic S (2006). T-cell receptor signaling events triggering granule
exocytosis.Critical Reviews inImmunology26(3):265-90.
Rahar S, Swami G, Nagpal N, Nagpal MA, Singh GS.(2011). Preparation, characterization, and biological
properties of β-glucans.Journal of Advanced Pharmaceutical Technology and Research 2:94-103.
Robyt JF (1998). Essentials of carbohydrate chemistry. Springer verlag Inc: 3(5):1-2
Roitt IM, Delves PJ (2001). Roitt’s essential immunology, 10th Edition, ISBN 0632059028, Blackwell
Publishing, p. 1-1496.
Rothenberg M, Hogan S (2006). The Eosinophil. Annual Reviews inImmunology24(1):147-74.
Roitt I, Brostoff J, Male D (1993).In :Immunology 3rd Edition Musby Year Book HD., London.
Roitt I, Brostoff J, Male D (2001). Edinburgh; 6thed.
cxli
Rus H, Cudrici C, Niculescu F (2003). The role of the complement system in Innate Immunity.Immunology
Research 33(2): 103-12.
Sacca R, Cuff C, Ruddle N (1997). Mediators of inflammation.Current.Opinion in Immunology.9: 851-857.
Sangle V, Darp M, Nadkarni S (2004).Evaluation of immunomodulatory activity of Suvarnamalini vasant, a
generc Ayurvedic herbominereal formulation. Indian Journal Experimental Biology42: 115-6
Sato K, Iwabuchi I, Nagaoka Y, Adachi N, Ohno H, Tamura H, Seyama K, Fukuchi Y, NakayamaH, Yoshizaki
F, Takamori K, Ogawa H (2006). Induction of human neutrophil chemotaxis by Candida albicansderived (beta)-1, 6 - long glycoside side chain –branched (beta)-glucan. Journal of
LeukocyteBiology80:204-211.
Schmid F, Stone BA, McDougall BM., Bacic A, Martin KL, Brownlee
RTC, Chai E,
RJ.(2001). Structure of epiglucan, a highly side- chain/branched(1→3;1→6)fungus Epicoccum nigrum
Ehrenb. ex Schlecht. Carbohydrate
Seviour
β-glucan from the micro
Research331: 163-171.
Sharma ML, Singh B, Chandan BK, Khajuria A, Kaul A, Bani S, Banerjeee SK, Gambhir SS (1996). Actions of
some flavonoids on specific and non-specific Immune Mechanism.Phytomedicine 3(2):191-195.
Sim R, Tsiftsoglou S (2004). Proteases of the complement system.Biochemical Society Transactions32 (Pt
1): 21-7.
Stimpel M, Proksch A, Wanger H, Lohmann-Matthes ML (1984).Macrophage activation and induction of
macrophage cytotoxicity by purified polysaccharide fractions from the plant Echinacea
purpurea.Journal of Infection and Immunity46:845-9
cxlii
Surendra S, Saurabh C, Gokul S (2010). Studies on immunomodulatory activity of Capparis zeylanica leaf
extract. International Journal of Pharmaceutical Sciences and Nanotechnology3(1):887-892
Suzuki M (1990). Induction of endogenous lymphokine activated killer activity by combined
administration of Lentinan and interleukin 2. International Journal Immunopharmacology 12:613623
Swain SL, Bradley LM, Croft M et al., (1991). Helper T-cell subset: phenotype function and the role of
lymphokines in their development. Immunology Review 123:115-44.
Tao, Y, Zhang L, Cheung PC (2006). Physicochemical properties and antitumour activities of water- soluble
native and sulfated hyper branched mushroom polysaccharides.Carbohydrate Research
341(13):2261-2269.
Thakur M, Bhargava S, Dixit VK, (2007). Immunomodulatory activity of Chlorophytum borivilianum sant. E
CAM 4(4):419-423
Thao D, Roger M, Susan V, Carl W (2008). Lippincott’s Illustrated reviews: Immunology. New Delhi:
Wolterskulwar Ltd.
Tokunaka K, Ohno N, Adachi Y, Tanaka S, Tamura H, Yadomae T (2000). Immunopharmacological and
immunotoxicological activities of a water soluble (1-3) beta-D-glucan, CSBS from Candida Spp.
International Journal ofImmunopharmacology 22:383-94.
Urban CF, Richard U, Brinkman V, Zychlinsky A (2006). A cell Microbial 8(4): 668-76.
cxliii
USC, (2006).Microbiology and immunology e-textbook of University of South Carolina school of medicine
(Richard Hunt, Webmaster); Accessed on June, 2nd, 2013.
Vaghasiya YK, Shukla VJ, Chanda SV (2010). Acute oral toxicity study of Pluchea arguta boiss extract in
mice. Journal of Pharmacology and Toxicology.6: 113-23.
Wahi V (2010). Pharmacological Study of Tinospora Cordifolia as an Immunomodulator.Internationa
Journal of Current Pharmaceutical Research2(4): 52 5
Wang JH, Chung YS, Cho YW, Kim DY, YI JS, Bae JS, Shim MJ (2010). Palatine tonsil size in obese
overweight and normal weight children with sleep disorder breathing.Official Journal of American
Academy of Otolaryngology – Head and Neck Surgery 142(4): 516-9.
Wasser SP, Weis AL, (1999a). Medicinal properties of substances occurring in higher basidiomycete
mushrooms: current perspectives (review). International Journal of MedicinalMushrooms, 1: 3162.
Wasser SP, Weis AL (1999b). Therapeutic effects of substances occurring in higher basidomycetes
mushrooms: A modern perspective. Critical Reviews in Immunology.19: 65-96.
Wasser SP (2002). Medicinal mushrooms as a source of antitumor and immunomodulating
polysaccharides.Applied Microbiology Biotechnology. 60(3): 258-74
cxliv
Wasterlund E, Andersson R, Aman P (1993). Isolation and chemical characterization of water-soluble
mixed linked β-glucan and arabinoxylans in oat milling fractions. Carbohydrate Polymers, 20: 115123.
Wendy WL, Li YQ, Lee TL, Li N, You MM, Chang S. (2004). Medicinal mushroom extracts inhibit RasInduced cell transformation and the inhibiting effect requires the presence of normal cells.
Carcinogenesis 25:1177-83.
Willard T (1990). Resishi Mushroom. Herb of spiritual potency and medicinal wonder, Issaquah:Sylvan
press.
William JE (2001). Review of antiviral and immunodulating properties of plants of the Peruvian rainforest.
Alternative Medicine. Review .6(6): 567-579.
Wong KH, Lai CM, Cheung Pc (2011). Immunomodulatory activities of mushroom Sclerotial
polysaccharides. Food hydrocolloids 25(2):150-158.
Xle OW, Cho DS, Calaycay J, Mumford RA, Swiderek K, Lee TD, Ding A, Torso T, Nathan C (1992). Cloning
and characterization of indicible nitric oxide synthase from mouse macrophages.Science 256.
Yang Q, Jong S (1989).Medicinal Mushrooms in China. Mushroom Science XII (Part 1): 631-64
Ying J, Mao X, Ma Q, Zong Y, Wen H (1987). Icons of Medicinal Fungi from China. Translated by Xu
Yuehan, Science Press, Beijing.
cxlv
Yoon YD, Han SB, Kang JS, Lee CW, Park SK, Lee HS, el at., (2003). Toll-like receptor 4-dependent
activation of macrophages by polysaccharide isolated from the radix of Platycodon grandiflorum.
International Immunopharmacology3:1873-82
Yu MY, Xu XY, Qing Y, Luo X, Yang ZR, Zheng LY (2008). Isolation of an anti-tumor polysaccharide from
Auricularia polytricha (jew’s ear) and its effects on macrophage activation. European Food
Research and Technology, 228: 477-485.
Zhao LY, Dong YH, Chen GT, Hu QH (2010).Extraction, purification, characterization and antitumor activity
of polysaccharides from Ganoderma lucidum.Carbohydrate Polymers, 80: 783-789.
Zhang X, Li J, Wong DKW, Wagner TE, Wei Y. (2009). Fermented Noni
directly stimulate B-lymphocyte proliferation and
exudatestreateddendritic
cells
differentiation.
OncologyReports21:1147–1152.
Zhang L, Fan C, Liu S, Zang Z, Jiao L, Zhang L. (2011). Chemical composition and anti-tumor
activity
of polysaccharide from Inonotus obliquus. Journal of Medicin Plants Research 5:1251-1260
Zhou H, Chen Q, Wang S (1989). Antiaging effect of the polysaccharides from auricularia auricular and
Tremella fuciformis. Zhongguo Yaoke Daxue Xuebao, 20: 303-306
Zhu M., Chang Q, Wong LK, Chong FS, Li R C. (1999).Triterperne anti-oxidants from
lucidum.Phytotherapy Research13:529-531.
Zoberi MH (1973). Some Edible Mushrooms from Nigerian. Nigerian Field, 38:81-90.
Ganoderma
cxlvi
cxlvii
APPENDICES
(Tabular summary of some of the key results presented in charts in chapter 3)
cxlviii
Appendix 1: The Effect of Hot Aqueous Extract of Pleurotus tuber-regium (PT) on Phagocytic Activity in
Swiss Albino Mice.
Phagocytic Index (Mean titre±SEM)
Phagocytic Index
Phagocytic Index (% increase
Normalized to
Treatment
Dose (mg/kg)
in Parenthesis)
Control
PT
100
0.011±0.005(647.9)
7.479
200
0.016± 0.010(907.5)
400
0.012± 0.002(670.0)
7.701
Noni®
100
0.021±0.008(1296.0)
13.968
Negative Control
___
0.002±0.001
___
10.075
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
cxlix
Appendix 2: The Effect of Hot Aqueous Extract of P. tuber-regium on the Relative Spleen Weight of
Mice.
Relative Spleen Weight (mean titre ± SEM) in grams
Relative Spleen Weight (%
Treatment
Dose
increase in parenthesis)
PT
100
0.494±0.012(6.87)
200
0.611± 0.026*(32.2)
400
0.568± 0.036(22.76)
Noni®
100
0.503±0.013(8.79)
Negative Control
___
0.463±0.047
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
cl
*= level of significance
cli
Appendix 3: The Effect of Hot Aqueous Extract of P. tuber-regium on Delayed Type Hypersensitivity
Response in Mice
Delayed Type Hypersensitivity (mean titre ± SEM) in mm
Treatment
Dose(mg/kg)
DTH (mm)
% stimulation of DTHR
PT
100
0.288± 0.049
1.41
200
0.420±0.032
47.88
400
0.402±0.101
40.84
Noni®
100
0.474 ±0.022
65.49
Negative Control
___
0.284 ± 0.102
___
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clii
Appendix 4: The Effect of BGPon Delayed Type Hypersensitivity Response in Mice
Delayed Type Hypersensitivity (mean titre ± SEM) in mm
Treatment
Dose(mg/kg)
DTH (mm)
% stimulation of DTHR
BGP
100
0.618±0.084
36.72
200
0.658±0.052*
45.57
Noni®
100
0.500±0.132
10.61
Negative Control
___
0.452±0.154
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
___
cliii
Appendix 5: The Effect of Hot AqueousExtract of Pleurotus tuber-regium(PT) on Primary and
SecondaryIgG Humoral Antibody Response to Ovalbumin Mice
Humoral antibody response (mean titre±SEM)
Dose(mg
Secondary response (7-
Secondary Response
Treatment
/kg)
Primary Response
days Poost boost)
(14-days Poost boost)
PT
100
0.399±0.049(44.7)
1.105±0.139(41.9)
1.163±0.100(31.2)
200
1.052±0.196***(281.6)
1.437±0.106***(84.6)
1.301±0.106*(46.7)
400
0.760±0.037**(175.8)
1.282±0.115**(64.6)
1.200±0.139(35.4)
100
0.686±0.137*(148.7)
1.266±0.058**(62.6)
1.305±0.149*(8.6)
Noni®
cliv
Negative
Control
___
0.275±0.030
0.778±0.073
0.886±0.090
Naïve
___
0.154±0.003 (-43.9)
0.154±0.004***(-80.1)
0.154±0.003***(-82.5)
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group *= level of significance
Appendix 6: The Effects of Hot Aqueous Extract of Pleurotus tuber-regium(PT) on Primary and
Secondary IgG1 Humoral Antibody Response to Ovalbumin in Mice
Humoral antibody response (mean titre±SEM)
Dose(m
Secondary response (7-
Secondary Response
Treatment
g/kg)
Primary Response
days Poost boost)
(14-days Poost boost)
PT
100
1.446±0.176*(77.2)
2.756±0.146***(50.8)
2.557±0.162(16.3)
200
1.922±0.126***(135.5)
2.987±0.005***(63.4)
2.810±0.079**(27.7)
clv
400
1.597±0.214*(95.7)
2.753±0.222***(50.7)
2.795±0.234**(27.2)
100
1.903±0.256***(133.2)
3.035±0.099***(66.1)
2.878±0.068**(30.8)
Control
___
0.816±0.091
1.827±0.202
2.199±0.022
Naïve
___
0.239±0.005(-70.7)
0.239±0.006***(-86.8)
0.239±0.006***(89.)
Noni®
Negative
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
Appendix 7: The Effects of Hot Aqueous Extract of Pleurotus tuber-regium(PT) on Primary and
Secondary IgG2a Humoral Antibody Response to Ovalbumin in Mice
Humoral antibody response (mean titre±SEM)
clvi
Dose(m
Secondary response (7-
Secondary Response
Treatment
g/kg)
Primary Response
days Poost boost)
(14-days Poost boost)
PT
100
0.780±0.093(60.5)
1.130±0.198(82.1)
1.036±0.099(56.8)
200
1.483±0.153***(205.1)
1.753±0.159***(182.5)
1.611±0.115**(143.8)
400
0.937±0.039*(92.8)
1.174±0.198(89.2)
1.493±0.218**(126.0)
100
0.840±0.120(72.8)
1.358±0.159**(118.9)
1.172±0.131*(77.3)
Control
___
0.486±0.154
0.620±0.102
0.661±0.039
Naïve
___
0.232±0.021(52.3)
0.232±0.021(62.6)
0.232±0.021(64.9)
Noni®
Negative
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clvii
Appendix 8: The Effects of ΒGP on Primary and Secondary Total IgG Humoral Antibody Response to
ovalbumin in Mice
Humoral antibody response (mean titre±SEM)
Treatments
Dose (mg/kg)
Primary response
Secondary Response
BGP
100
0.268±0.019*(40.88)
0.623±0.046(9.12)
200
0.371±0.022***(93.98)
1.082±0.099***(89.49)
Noni®
100
0.281±0.014**(47.55)
0.723±0.074(26.61)
Negative Control
___
0.190±0.013
0.571±0.041
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clviii
Appendix 9: The Effects of BGP on Primary and Secondary IgG1 Humoral Antibody Response to
Ovalbumin in Mice
Humoral antibody response (mean titre±SEM)
Treatments
Dose (mg/kg)
Primary response
Secondary Response
BGP
100
0.576±0.054(58.6)
2.350±0.195(27.6)
200
0.816±0.069**(124.6)
2.965±0.286*(61.0)
Noni®
100
1.158±0.148***(218.6)
3.004±0.266**(63.2)
Negative Control
___
0.363±0.037
1.841±0.251
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clix
Appendix 10: The Effects of BGP on Primary and Secondary IgG2a Humoral Antibody Response to
Ovalbumin in Mice
Humoral antibody response (mean titre±SEM)
Treatments
Dose (mg/kg)
Primary response
Secondary Response
BGP
100
0.666±0.028***(59.0)
1.403±0.151*(75.6)
200
0.860±0.055***(105.5)
2.534±0.162***(217.1)
Noni®
100
0.635±0.041**(51.6)
1.284±0.088**(60.7)
Negative Control
___
0.419±0.021
0.799±0.078
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clx
Appendix 11: The Effect of BGP on Spleenocytes Proliferation
Spleenocyte proliferation (titre±SEM)
Spleenocyte
Treatment
Dose(µg/ml)
Proliferation index
% Proliferation
BGP
5
1.444±0.020***
44.4
25
1.573±0.012***
57.3
50
1.902±0.051***
90.2
100
2.251 ±0.146***
25.1
250
1.531±0.070***
53.1
1000
1.284±0.105*
28.4
2
1.861±0.033***
86.1
Con A
clxi
Control
___
1.000 ± 0.028
___
Stimulation, n=8 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
Appendix 12: The Effect of BGP on TNF-α Production by RAW264.7
TNF-α production(titre±SEM)
% increase in TNF-α
Treatment
Dose (µg/ml)
TNF-α production
Production
BGP
5
1.180±0.029***
544.8
25
1.580±0.017***
763.3
50
2.064± 0.088***
1027.8
clxii
100
1.987±0.022***
985.7
LPS
1
1.822±0.030***
895.6
Control
__
0.183±0.010
___
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clxiii
Appendix 13: The Effect of BGP on Nitric Oxide Production by RAW264.7
Nitric Oxide production(titre±SEM)
% Increase in Nitric
Treatment
Dose (µg/ml)
Nitric Oxide production
Oxide Release
BGP
5
6.933±0.088***
1055.5
25
9.767±0.218***
1527.8
50
16.200± 0.230***
2600.0
100
19.670±0.433***
3178.3
LPS
1
17.770±0.751***
2861.6
Control
__
0.600±0.057
___
Stimulation, n=6 ; *P<0.05, **P<0.001, ***P<0.0001
Where n= number of animals per group
*= level of significance
clxiv
Appendix 14: The Effect ofBGP onRate of Phagocytosis RAW264.7
Rate of Phagocytosis (titre±SEM)
Treatment
Dose (µg/ml)
Rate of Phagocytosis
% Rate of Phagocytosis
BGP
5
1.119±0.011***
11.9
25
1.165±0.016***
16.5
50
1.181± 0.001***
18.1
100
1.083±0.010***
8.3
LPS
1
1.149±0.011***
14.9
Control
__
1.000±0.013
___
Stimulation, n=6 ; *p<0.05, **p<0.001, ***p<0.0001
Where n= number of animals per group
*= level of significance
clxv