evaluating the neuroprotective effects of fermented

121
EVALUATING THE NEUROPROTECTIVE EFFECTS OF FERMENTED ROOIBOS HERBAL TEA IN WISTAR RATS EXPOSED TO BISPHENOL-A DURING GESTATION AND LACTATION By BUSHRA KHALIFA GAMOUDI Student Number 3607196 A thesis submitted in fulfilment of the requirements for the degree of Magister Scientiae (MSc) Department of Medical Biosciences Faculty of Natural Sciences Supervisor Prof. Okobi Ekpo http://etd.uwc.ac.za/

Upload: others

Post on 05-Apr-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

EVALUATING THE NEUROPROTECTIVE EFFECTS OF

FERMENTED ROOIBOS HERBAL TEA IN WISTAR RATS

EXPOSED TO BISPHENOL-A DURING GESTATION AND

LACTATION

By

BUSHRA KHALIFA GAMOUDI

Student Number 3607196

A thesis submitted in fulfilment of the requirements for the degree of

Magister Scientiae (MSc)

Department of Medical Biosciences

Faculty of Natural Sciences

Supervisor

Prof. Okobi Ekpo

http://etd.uwc.ac.za/

ii

DECLARATION

I declare that the thesis titled: “Evaluating the neuroprotective effects of

fermented Rooibos Herbal Tea in Wistar rats exposed to Bisphenol-A during

gestation and lactation” is my work and is hereby submitted for the Master of

Science degree in Medical Biosciences at the University of the Western Cape,

South Africa. This work has not been submitted for any degree or examination in

any other university, and all sources used or quoted have been duly indicated and

acknowledged by complete references

Full name: Bushra Khalifa Gamoudi

Signed.......................................

http://etd.uwc.ac.za/

iii

DEDICATION

To my dearest family for their love, support, encouragement, and making my

dreams a reality.

The Prophet Muhammad (Peace Be upon Him) said, regarding knowledge:

“Acquire knowledge and impart it to the people...One who treads a path in search

of knowledge has his path to Paradise made easy by God” (Sunan Tirmidhi,

Hadith 107; Riyadh us-Saleheen, 245).

http://etd.uwc.ac.za/

iv

ACKNOWLEDGEMENTS

First of all, I would like to acknowledge my Allah for giving me the faith, strength,

good health, wisdom and perseverance, without you nothing is possible, and I

would not have completed my Masters!

This research would not have been completed without the generous help and

support received from different people. I want to express my sincere gratitude for

the valued assistance I received throughout my MSc programme from the

following people:

A special thanks to my husband who stood by me through the rough times

and my children whose presence always brings joy to my life.

My supervisor Prof. Okobi Ekpo for the knowledge gained,

encouragement, for his confidence in me, his patience, support and

motivation throughout the duration and completion of this research. I

acknowledge him for proofreading my thesis. THANK YOU!

I would also like to thank my parents and sisters for their love, care,

prayers, and support during my period of study.

Staff at the histology laboratory of the University of Stellenbosch, for their

help and the technical support.

To my Lab mates and colleagues at the Medical Bioscience department of

UWC, I say thank you for your kind and the friendly working relationship;

I enjoyed my times with you.

http://etd.uwc.ac.za/

v

Lastly, but by no means the least, big thanks to the Libyan government for

funding the project and my studies at the University of the Western Cape.

http://etd.uwc.ac.za/

vi

TABLE OF CONTENTS

DECLARATION ............................................................................................... ii

DEDICATION .................................................................................................. iii

ACKNOWLEDGEMENTS ............................................................................. iv

TABLE OF CONTENTS ....................................................................................... vi

LIST OF FIGURES ............................................................................................... xi

LIST OF TABLES ............................................................................................... xiii

LIST OF ABBREVIATIONS .............................................................................. xiv

ABSTRACT ......................................................................................................... xvi

CHAPTER 1 ........................................................................................................... 1

INTRODUCTION ............................................................................................. 1

1.1. RESEARCH HYPOTHESIS .................................................................... 5

1.2. RESEARCH AIM AND OBJECTIVES................................................... 5

1.2.1. AIM ........................................................................................................ 5

1.2.2. OBJECTIVES ........................................................................................ 5

CHAPTER 2 ........................................................................................................... 7

LITERATURE REVIEW ................................................................................. 7

2.1. THE CENTRAL NERVOUS SYSTEM (CNS) ....................................... 7

2.1.1. THE CEREBRUM ................................................................................. 8

2.1.1.1. CYTOARCHITECTURE OF THE CEREBRAL CORTEX ............. 9

2.1.1.2. FUNCTION OF THE CEREBRAL CORTEX ................................ 10

2.1.1.3. THE HIPPOCAMPUS ...................................................................... 11

http://etd.uwc.ac.za/

vii

2.1.2. THE CEREBELLUM .......................................................................... 13

2.1.2.1. CYTOARCHITECTURE OF THE CEREBELLAR CORTEX ...... 15

2.2. THE ENDOCRINE SYSTEM ................................................................ 16

2.2.1. ENDOCRINE DISRUPTOR COMPOUNDS (EDCs) ........................ 17

2.2.1.1. BISPHENOL A (BPA) ..................................................................... 21

2.2.1.1.1. PHYSICAL AND CHEMICAL PROPERTIES OF BPA ............. 22

2.2.1.2. RELEASE AND HUMAN EXPOSURE TO BPA .......................... 23

2.2.1.3. METABOLISM OF BPA ................................................................. 24

2.2.1.4. ACTIVITY OF BPA ......................................................................... 26

2.3. ROOIBOS (Aspalathus linearis) ............................................................ 28

2.3.1. CHEMICAL COMPOSITION OF ROOIBOS ................................... 29

2.3.1.1. POLYPHENOLS .............................................................................. 30

2.3.2. BIOLOGICAL PROPERTIES OF ROOIBOS TEA ........................... 32

2.3.4.1. NEUROPROTECTIVE EFFECT OF ROOIBOS ............................ 33

CHAPTER 3 ......................................................................................................... 36

MATERIALS AND METHODS .................................................................... 36

3.1. ETHICAL CONSIDERATION .............................................................. 36

3.2. MATERIALS.......................................................................................... 36

3.3. ANIMALS .............................................................................................. 37

3.4. TREATMENT PROTOCOL .................................................................. 38

3.5. PREPARATION OF REAGENTS ......................................................... 39

3.5.1. ROOIBOS HERBAL TEA PREPARATION ..................................... 39

3.5.2. BISPHENOL A (BPA) AND NORMAL SALINE ............................. 40

http://etd.uwc.ac.za/

viii

3.5.3. PREPARATION OF 4% FORMALDEHYDE SOLUTION .............. 40

3.5.4. PHOSPHATE BUFFERED SALINE (PBS) ....................................... 40

3.6. MOTOR FUNCTION TESTS (NEUROBEHAVIORAL TESTS) ........ 41

3.6.1. OPEN FIELD TEST (OFT) ................................................................. 41

3.6.2. NOVEL OBJECT RECOGNITION (NOR) TEST ............................. 43

3.7. ANIMAL SACRIFICE ........................................................................... 44

3.8. HISTOLOGICAL PREPARATION OF BRAIN SAMPLES ................ 44

3.8.1. AUTOMATED TISSUE PROCESSING ............................................ 44

3.8.2. EMBEDDING ..................................................................................... 45

3.8.3. SECTIONING ..................................................................................... 45

3.8.4. STAINING........................................................................................... 45

3.8.4.1. HAEMATOXYLIN AND EOSIN.................................................... 45

3.8.4.2. CRESYL VIOLET (CV) / NISSL STAINING ................................ 46

3.8.4.3. IMMUNOHISTOCHEMICAL (IHC) STAINING .......................... 47

3.9. NEUROCHEMICAL ASSAYS ............................................................. 47

3.9.1. HOMOGENIZATION OF TISSUES .................................................. 48

3.9.1.1. SUPEROXIDE DISMUTASE (SOD) .............................................. 48

3.9.1.2. CATALASE...................................................................................... 48

3.10. STATISTICAL ANALYSIS ................................................................ 49

CHAPTER 4 ......................................................................................................... 50

RESULTS ......................................................................................................... 50

4.1. INTRODUCTION .................................................................................. 50

4.2. EFFECT OF BPA AND FERMENTED ROOIBOS HERBAL TEA ON

BODY MASS ................................................................................................ 51

http://etd.uwc.ac.za/

ix

4.3. NEUROBEHAVIOURAL TESTS ......................................................... 52

4.3.1. THE OPEN FIELD TEST ................................................................... 52

4.3.1.1. THE FREQUENCY OF REARING EPISODES ............................. 52

4.3.1.2. TOTAL FREEZING TIME .............................................................. 53

4.3.1.3. TOTAL SELF-GROOMING TIME ................................................. 54

4.3.2. NOVEL OBJECT RECOGNITION (NOR) TEST ............................. 55

4.4. EFFECT OF BPA AND FERMENTED ROOIBOS HERBAL TEA ON

THE ACTIVITY OF ANTIOXIDANT ENZYMES ..................................... 57

4.4.1. SUPEROXIDE DISMUTASE ............................................................. 57

4.4.2. CATALASE......................................................................................... 59

4.5. HISTOLOGICAL STUDIES .................................................................. 60

4.5.1. LENGTH OF HIPPOCAMPAL CA1 REGION IN EXPERIMENTAL

RATS ............................................................................................................. 61

4.5.2. NUMBER OF PURKINJE CELLS IN PURKINJE CELL LAYER

(PCL) IN EXPERIMENTAL RATS ............................................................. 62

4.6. IMMUNOHISTOCHEMISTRY (IHC) .................................................. 64

4.6.1. GLIAL FIBRILLARY ACIDIC PROTEIN (GFAP) .......................... 64

CHAPTER 5 ......................................................................................................... 66

DISCUSSION ................................................................................................... 66

5.1. INTRODUCTION .................................................................................. 66

5.2. ROOIBOS TEA PREVENTED BPA-INDUCED GAIN IN BODY

MASS............................................................................................................. 67

5.3. ROOIBOS TEA MITIGATED BPA-INDUCED

NEUROBEHAVIOURAL DEFICITS .......................................................... 68

5.4. ROOIBOS TEA REGULATED THE ACTIVITY OF ANTIOXIDANT

ENZYMES..................................................................................................... 72

http://etd.uwc.ac.za/

x

5.5. ROOIBOS TEA ATTENUATED NEURODEGENERATION IN RATS

EXPOSED TO BPA ...................................................................................... 73

5.6. CONCLUSION .................................................................................................. 77

REFERENCES ...................................................................................................... 79

http://etd.uwc.ac.za/

xi

LIST OF FIGURES

Figure 2.1: The brain structure 7

Figure 2.2: The layers of the cerebral cortex 8

Figure 2.3: The lobes of the cerebral cortex 9

Figure 2.4: The structure of the Hippocampus 10

Figure 2.5: The layers of the hippocampus proper 11

Figure 2.6: The structure of the cerebellum A) Sagittal and B) Dorsal sections 12

Figure 2.7: The structure of the cerebellar cortex 13

Figure 2.8: Synthesis of Bisphenol A 19

Figure 2.9: Metabolism of BPA 22

Figure 2.10: Rooibos tea; (A) Unfermented (B) Fermented 25

Figure 2.11: Classification of polyphenols 27

Figure 2.12: Chemical structure of Aspalathin 28

Figure 3.1: Flow diagram showing the experimental design for the study 34

Figure 3.2: Open Field Test Apparatus 38

Figure 4.1: Average body mass of experimental rats on PND 42 47

Figure 4.2: Number of rearing episodes of experimental rats on PND 42 49

Figure 4.3: Total freezing time of experimental rats on PND 42 50

Figure 4.4: Total self-grooming time of experimental rats on PND 42 51

Figure 4.5: Recognition index of experimental rats on PND 42 52

Figure 4.6: Effects of different treatments on SOD enzyme activity in the

cerebral homogenate of experimental rats on PND 42 53

http://etd.uwc.ac.za/

xii

Figure 4.7: Effects of different treatments on SOD enzyme activity in the

cerebellar homogenate of experimental rats on PND 42 54

Figure 4.8: Effects of different treatments on CAT enzyme activity in the

cerebral homogenate of experimental rats on PND 42 55

Figure 4.9: Effects of different treatments on CAT enzyme activity in the

cerebellar homogenate of experimental rats on PND 42 55

Figure 4.10: Diagram of the hippocampus showing the landmarks used for

determining hippocampal length in the experimental rats. 57

Figure 4.11: CA1 hippocampal length in experimental rats on PND42 57

Figure 4.12: Number of Purkinje cells in experimental rats on PND42 58

Figure 4.13: Number of GFAP positive cells in experimental rats on PND42 60

http://etd.uwc.ac.za/

xiii

LIST OF TABLES

Table 2.1: Examples of EDCs and their endocrine disruptor effects 16

Table 2.2: Physical properties of BPA 19

Table 2.3: Quantification of the major flavonoids in aqueous rooibos extract 26

Table 3.1: Chemicals used in this study 32

Table 3.2: Equipment used in this study 33

Table 3.3: Tissue Processing Protocol 40

Table 3.4: IHC staining procedure 43

http://etd.uwc.ac.za/

xiv

LIST OF ABBREVIATIONS

•OH

Hydroxyl radical

ANOVA Analysis of variance

ATP Automated tissue processor

BPA Bisphenol A

CA Cornu ammonis

CAT Catalase

CNS Central nervous system

CV Cresyl Violet

DDT Dichlorodiphenyltrichloroethane

DG Dentate gyrus

EDCs Endocrine Disruptor compounds

FRHT Fermented rooibos herbal tea

GFAP Glial fibrillary acidic protein

H&E Haematoxylin and eosin

H2O2 Hydrogen peroxide

HCB Hexachlorobenzene

IHC Immunohistochemistry

NOR Novel Object Recognition

NS Normal saline

NTP National Toxicology Program

O−2 Superoxide

OFT Open Field Test

PAHs Polycyclic aromatic hydrocarbons

http://etd.uwc.ac.za/

xv

PBS Phosphate buffered saline

PCBs Polychlorinated biphenyls

PCDF Polychlorinated dibenzofurans

PCL Purkinje cell layer

PND Post-natal day

PNS Peripheral nervous system

RI Recognition index

ROS Reactive oxygen species

SOD Superoxide dismutase

UGT UDP-glucuronosyl transferase

http://etd.uwc.ac.za/

xvi

ABSTRACT

Exposure to endocrine-disrupting chemicals as bisphenol A (BPA) during

gestation and early postnatal life is known to disrupt normal developmental

processes and alter the body’s endocrine system leading to deleterious effects in

the developing central nervous system (CNS). BPA is an industrial synthetic

chemical commonly used in the production of a range of polymers and consumer

products, despite concerns about its safety. There is therefore the need to protect

the developing CNS from potential damage through the administration of

neuroprotective agents. Most medicinal plants are reported to possess significant

protective potential against tissue damage through different mechanisms that

prevent cell death, oxidative stress, inflammation, immunodeficiency, etc. In this

study, the protective effects of fermented rooibos (Aspalathus linearis) tea against

the deleterious effects of BPA were investigated. Rooibos is a herbal beverage

indigenous to South Africa with widely acclaimed health benefits often linked to

the bioactivity of its polyphenolic compounds, especially aspalathin. The anti-

allergic, cardiovascular, antioxidant and neuroprotective effects of this herb have

been previously reported hence, the present study aims to investigate if regular

consumption of rooibos tea during pregnancy and lactation could protect the

developing brain from the deleterious effects of BPA in a Wistar rat model. A

total of 40 three-month old adult female pregnant dams, with an average weight of

250g, were divided into four groups (n=10). Group 1 control rats received 9%

normal saline ad libitum; group 2 rats received 400µg/kg/day BPA only; group 3

rats received 20% fermented rooibos tea as well as 400µg/kg/day BPA, while

group 4 rats received ad libitum 20% fermented rooibos tea only. Offspring rats

were housed in the same cages as the dams and only separated after weaning on

postnatal day (PND) 21. Neurobehavioural assessment using the open field test

was done on postnatal day (PND) 42 after which the final body masses were taken

before the rats were decapitated under deep anaesthesia, and the desired CNS

parts carefully dissected out and processed for histological, biochemical and

immunohistochemical studies. The results obtained showed that there was

http://etd.uwc.ac.za/

xvii

significant impairment of neurobehavioural activity, decreased cerebral and

cerebellar antioxidant enzyme activity, reduced hippocampal CA1 length,

significant loss of cerebellar Purkinje cells and significant astrocyte activation

demonstrated by increased glial fibrillary acidic protein (GFAP) activity in

experimental rats exposed to BPA only. However, co-administration of rooibos

tea significantly attenuated the BPA-induced distortions. Taken together, these

findings suggest that rooibos could be a potent neuroprotective agent against

BPA-induced structural, functional and biochemical alterations in the developing

CNS.

Keywords: Endocrine disrupting chemicals, Bisphenol A, rooibos, antioxidants,

neurodegeneration, neuroprotection, neurobehavioural activity.

http://etd.uwc.ac.za/

1

CHAPTER 1

INTRODUCTION

Many chemical substances have been identified as Endocrine Disruptor

Compounds (EDCs) and humans become exposed to them either at the workplace,

through food or environment sources (air, water and soil). EDCs have been widely

reported to directly or indirectly affect the normal functioning of the endocrine,

nervous and reproductive systems (Colborn et al., 1993; Clotfelter, et al., 2004;

Kajta and Wójtowicz, 2013). Bisphenol A (BPA) is one of the most recognised

EDCs and is a widespread estrogenic chemical (Krishnan et al., 1993, Eladak et

al., 2015) and more than 8 billion pounds (4 billion kilograms) of BPA are

produced for the manufacturing of polycarbonate plastic food and beverage

containers per year (Rubin, 2011). BPA is also used in the resin lining of metal

cans, dental sealants, and is employed as an additive in a wide array of other

products (Vandenberg et al., 2007).

BPA has been reported to be capable of crossing the placental barrier during

pregnancy and is present in different human fluids including foetal and maternal

serum, as well as in full-term amniotic fluid (Ikezuki et al., 2002, Yamada et al.,

2002). In one study, BPA was identified in developing organs of mice including

the CNS (Mita et al., 2012). Other studies have also reported the effects of BPA

on the developing and adult brain (MacLusky et al., 2005, Zsarnovszky et al.,

2005, Rubin and Soto, 2009, Negri-Cesi, 2015, Gupta et al., 2018). The

hippocampus and cerebellum are some of the CNS structures which have been

http://etd.uwc.ac.za/

2

identified as susceptible targets of BPA-induced distortions. Hippocampal

apoptosis and neurodegeneration (Tiwari, et al., 2015; Xu et al., 2010a) as well as

alteration of early cerebellar morphology (Mathisen, et al., 2013) have been

reported in developing animals. Both these structures were the focus of the

present study.

The cerebellum is a component of the CNS situated at the posterior aspect of the

brainstem, inferior to the occipital and temporal lobes of the cerebral cortex. It is

mainly involved in maintaining balance, posture and the coordination of voluntary

movements through a stabilising control system which receives early warning

signals from each motor impulse (Fonnum and Lock, 2000). It is also involved in

motor, learning and cognitive functions (albeit via poorly understood

mechanisms). Although motor commands are mostly initiated in other CNS

regions, the cerebellum modifies them according to rate, range, force and

sequence of contractions by allowing more adaptive and accurate movements

(Fonnum and Lock, 2000).

The cerebellum belongs to a distributed sensorimotor coordination network that

includes the cerebral cortex, basal motor nuclei, thalamus and the reticular

formation (Doya, 1999). Therefore, any dysfunction of the cerebellum could

disrupt the entire sensorimotor coordination network and manifest as locomotor

dysfunctions. The cerebellum accounts for approximately 10% of the brain’s

volume and contains over 50% of the total number of neurons in the brain

(Herculano-Houzel, 2009). The remarkable diversity in its neuronal population

together with its unique post-natal development makes it an appropriate neural

http://etd.uwc.ac.za/

3

structure for studying the effects of different substances on cellular dynamics

during pre- and post-natal development. Whereas, granule and Purkinje cells are

the most important neurons in the cerebellum, Purkinje cells are very important

because they are the largest neurons in the brain and the sole output of the

cerebellum, thus implying that any distortions in their structure and function could

affect overall cerebellar function (Fonnum and Lock, 2000).

Neurodevelopmental deficits potentially related to cerebellar injury may be arise

from the effects of environmental substances leading to such impaired motor

functions as hypotonia, fine motor incoordination, ataxia and impaired motor

sequencing (Powls et al., 1995, Goyen et al., 1998). Cerebellar injury has also

been implicated in cognitive, social and behavioural dysfunction among older

patients (Berquin et al., 1998, Levisohn et al., 2000) and may contribute to the

long-term cognitive, language, and behavioural dysfunction seen among 25-50%

formerly pre-term infants (Limperopoulos et al., 2007, Messerschmidt et al.,

2008). Only a few studies have focused on the vulnerability of the cerebellum to

toxic agents (Bist and Bhatt, 2009, Sanders et al., 2009), mainly focused on the

effects of lead, polychlorinated biphenyl and mercury on the nervous system (Yun

and Hoyer, 2000, Mervis et al., 2002, Verstraeten et al., 2008).

The functions of the hippocampus in memory acquisition and storage as well as

spatial learning have been adequately described (Sutherland and Rudy, 1989).

Hippocampal CA1 neurons are the most sensitive and susceptible to selective

degeneration in response to such EDCs as BPA, ischemia and other toxic

substances (Kirino, 1982, Pulsinelli et al., 1982, Elsworth et al., 2013, Kimura et

http://etd.uwc.ac.za/

4

al., 2016). In addition, axons of CA1 have been considered the main output of the

hippocampus (Duvernoy, 2013) which informs the choice of this region for

investigation in the current study.

The use of herbal products for the treatment of diseases is as old as humanity, and

certain plants have been reported to contain active compounds which could

protect the brain against such EDCs as BPA. Herbal products are considered to be

natural, green, pure, and with minimal side effects (Prashant et al., 2014), often

representing a source of new compounds with potent antioxidant activity to

address oxidative stress (Auddy et al., 2003). Rooibos (Aspalathus linearis) a

herbal beverage indigenous to South Africa, is one such plant with high

antioxidant activity (McKay and Blumberg, 2007, Joubert et al., 2008). Rooibos

naturally grows in the Western Cape Fynbos region of Clanwilliam and is

naturally caffeine-free, with a low tannin content (Erickson, 2003). The highly

acclaimed antioxidant properties of rooibos could be due to its unique polyphenol

content, aspalathin, known to be capable of delaying, inhibiting, or preventing

oxidation through scavenging of free radicals to diminish oxidative stress (McKay

and Blumberg, 2007, Joubert et al., 2008).

It has been reported that long-term consumption of rooibos could protect rat

brains from age-related changes, possibly due to its ability to prevent the

accumulation of lipid peroxides in the brain (Inanami et al., 1995). Also, rooibos

herbal tea has been shown to decrease steroidal hormone levels, indicating the

possibility that it could protect against the effects of BPA (Schloms et al., 2012).

This study was therefore designed to investigate the potential neuroprotective

http://etd.uwc.ac.za/

5

activity of rooibos herbal tea through the amelioration of the effects of the

endocrine disruptor BPA in an in vivo model.

1.1. RESEARCH HYPOTHESIS

This study hypothesizes that regular consumption of fermented rooibos herbal tea

could protect the developing brain against the deleterious effects of BPA. It is

believed that the polyphenolic compounds in rooibos tea are responsible for its

biological and pharmacological activities.

1.2. RESEARCH AIM AND OBJECTIVES

1.2.1. AIM

This study aims to evaluate the potential neuroprotective effects of fermented

rooibos herbal tea on the development of some CNS regions in rats exposed to

BPA during pregnancy and lactation.

1.2.2. OBJECTIVES

The following objectives were set for this study:

To determine the effects of maternal exposure to rooibos herbal tea and BPA

on the body masses of offspring rats.

To determine the effects of maternal exposure to rooibos and BPA on

neurobehavioural activity in offspring rats.

To determine the effects of maternal exposure to rooibos and BPA on

antioxidant enzymatic activity in cerebri and cerebella of offspring rats.

To determine the effects of maternal exposure to rooibos and BPA on

hippocampal and cerebellar morphology in offspring rats.

http://etd.uwc.ac.za/

6

To determine the effects of rooibos and BPA exposure on the activation of

astrocytes in offspring rats.

http://etd.uwc.ac.za/

7

CHAPTER 2

LITERATURE REVIEW

2.1. THE CENTRAL NERVOUS SYSTEM (CNS)

The nervous system consists of two major parts or subdivisions, the central

nervous system (CNS) and the peripheral nervous system (PNS) (Walsh and

Marshall, 1957). The CNS is the control centre for the entire nervous system and

is comprised of the brain and spinal cord. The brain is further subdivided into

three major sections, cerebrum, brainstem and cerebellum (Figure 2.1). The

cerebrum is the largest part of the human brain and is responsible for the

processing of speech, learning, emotions, muscular contractions as well as the

interpretation of sensory data related to hearing, vision and touch (Abhang et al.,

2016). The brainstem consists of the midbrain, pons and medulla oblongata, and

connects the cerebrum and cerebellum to the spinal cord. The cerebellum lies

posterior to the cerebrum, has important functions in motor control and is

responsible for the maintenance of balance and equilibrium (Morton and Bastian,

2004).

http://etd.uwc.ac.za/

8

Source: https://www.ncbi.nlm.nih.gov/pubmedhealth/PMHT0024735/

Figure 2.2: The brain structure

2.1.1. THE CEREBRUM

The cerebrum is the largest subdivision of the brain, located superiorly and

anteriorly in relation to the brainstem. It consists of a pair of cerebral hemispheres

(right and left), separated by the midline longitudinal cerebral fissure (the falx

cerebri) (Patestas and Gartner 2016). The cerebral cortex forms the outer covering

of the cerebral hemispheres and consists of a central core of white matter and a

thin outer covering of grey matter. It is estimated to comprise approximately 85%

volume of the adult human brain (Stephan et al., 1981, Rilling and Insel, 1999),

and each hemisphere is further subdivided into four lobes: the frontal, parietal,

temporal, and occipital lobes. The cerebral cortex is few millimetres thick and is

http://etd.uwc.ac.za/

9

highly convoluted to permit a large surface area of the brain to fit inside skulls

(Hilgetag and Barbas, 2009).

2.1.1.1. CYTOARCHITECTURE OF THE CEREBRAL CORTEX

The cerebral cortex is organized into six layers, and each of these layers has

different roles.

Source: https://medicine.academic.ru/135149/layers_of_cerebral_cortex

Figure 2.2: The layers of the cerebral cortex

The layers of the cerebral cortex, from superficial to deep, are:

Molecular layer (Plexiform) contains scattered horizontal cells and the apical

dendrites of pyramidal neurons of the cerebral cortex

The external granular layer contains small neuronal cell bodies.

http://etd.uwc.ac.za/

10

The external pyramidal layer is composed of small and medium pyramidal

cells.

The internal granular layer is made up of densely packed small round

pyramidal cells.

Internal pyramidal layer (Ganglionic) contains larger pyramidal cells and

scattered non-pyramidal cells.

Fusiform or Multiform layer comprises of variably shaped cells.

2.1.1.2. FUNCTION OF THE CEREBRAL CORTEX

Different regions of the cerebral cortex are associated with certain functions. For

instance, the frontal lobe is associated with conscious thought, memory,

personality and control of voluntary muscle movements (Chayer and Freedman,

2001).

Source:https://www.wpclipart.com/medical/anatomy/brain/brain_3/four_lobes_of_the_cerebral_c

ortex.png.html

Figure 2.3: The lobes of the cerebral cortex

http://etd.uwc.ac.za/

11

The parietal lobe gives individuals perspective and to help them understand space,

touch, and volume; the occipital lobe processes visual information while temporal

lobe is involved in auditory reception, helps to understand speech and is involved

in retrieving visual as well as verbal memories (Aversi-Ferreira et al., 2010,

Kiernan, 2012). The process of speech involves two different areas of the

cerebrum, namely, the Broca’s area which is responsible for the coordination of

the muscles for speaking and translation of thought into speech (Fadiga et al.,

2009), and the Wernicke’s area which is involved in the ability to read,

understand, and speak written word (Ardila et al., 2016).

2.1.1.3. THE HIPPOCAMPUS

The hippocampus is a small neuronal curved structure located on the medial

aspect of the temporal lobe of each cerebral cortex and forms an important part of

the limbic system. The hippocampus plays an important role in long-term memory

formation and spatial navigation. The hippocampus consists of two interlocking

U-shaped grey matter structures, the hippocampus proper (cornu ammonis, CA;

Figure 2.4) which is differentiated into four regions, CA1-4, and the dentate gyrus

separated from each other by hippocampal sulcus (Lorente de Nó, 1934;

Duvernoy, 2005).

http://etd.uwc.ac.za/

12

Source: https://www.slideshare.net/amandahessborzacchini/hippocampus-13053807

Figure 2.4: The structure of the Hippocampus

The CA is divided into three primary layers, each defined by a particular feature

of the large pyramidal cells or their afferents (Figure 2.5). The polymorph layer is

found between the alveus and the pyramidal layer, and contains cell bodies of the

basket cells and the basal dendrites of the pyramidal cells, as well as afferents

from the septum; the cell bodies of the pyramids dominate the pyramidal layer

(stratum pyramidale), and the molecular layer contains pyramidal dendrites

(Bayer, 1985).

Source: https://www.slideshare.net/drpraveenktripathi/limbic-system-brain

Figure 2.5: The layers of the hippocampus proper

http://etd.uwc.ac.za/

13

2.1.2. THE CEREBELLUM

The cerebellum is located conspicuously in the hindbrain with pons and medulla

oblongata. Although it is only 10% of total brain mass, the cerebellum has about

75% as much surface area as either of the much larger cerebral hemispheres,

containing approximately 70-101 billion neurons in the brain (Snider, 1958,

Lange, 1975, Andersen et al., 1992). The cerebellum has a relatively longer

developmental period and is one of the first structures in the brain to differentiate

and one of the last to mature (Liu et al., 2011). It is derived from the rhombic lips

and thickenings along the margins of the embryonic hindbrain. By the second

trimester of pregnancy, the fissures of the cerebellar cortex have appeared (Liu et

al., 2011).

The cerebellum consists of two paired lateral extensions known as hemispheres

united by the midline part called the vermis (Figure 2.6). Typically, the

cerebellum is divided into the anterior, posterior and flocculonodular lobes as

defined by the major fissures.

http://etd.uwc.ac.za/

14

Source: http://what-when-how.com/neuroscience/the-cerebellum-motor-systems-part-1/

Figure 2.6: The structure of the cerebellum A) Sagittal and B) Dorsal sections

The cerebellum consists of an outer layer of grey matter called the cortex and an

inner core of white matter known as medulla, containing four paired deep nuclei

namely, dentate nucleus, globose nucleus, emboliform nucleus, and fastigial

nucleus (Snell, 2010). All cerebellar input goes to the cortex, and all output comes

from the deep nuclei. The cerebellum is connected to the brainstem by three pairs

of stalks called cerebellar peduncles. Whereas the inferior peduncles connect the

cerebellum to the medulla oblongata, the middle peduncles connect the

cerebellum to the pons and the superior peduncles connect the cerebellum to the

midbrain (Snell, 2010).

http://etd.uwc.ac.za/

15

2.1.2.1. CYTOARCHITECTURE OF THE CEREBELLAR CORTEX

The cerebellar cortex is made up of three layers: the molecular layer, the granule

cell layer and the Purkinje cell layer in the middle (Figure 2.7). The input to the

cerebellum comes via the mossy and climbing fibres. The molecular layer consists

of two main types of neurons: stellate cells and basket cells (Apps and Garwicz,

2005). The Purkinje layer is formed by a single row of large Purkinje cells and

their axons which provide the only efferent pathway to the deep cerebellar nuclei,

thus constituting the sole output of all motor coordination in the cerebellar cortex

(Apps and Garwicz, 2005). The granular layer is densely populated by small

granule cells with dark-staining nuclei and scanty cytoplasm (Apps and Garwicz,

2005).

Source: http://vanat.cvm.umn.edu/neurHistAtls/pages/cns9.html

Figure 2.7: The structure of the cerebellar cortex.

Early studies considered the cerebellum a motor structure because cerebellar

damage mostly leads to impairments in motor function. Although the cerebellum

does not initiate movement, it contributes to coordination, precision, and accurate

http://etd.uwc.ac.za/

16

timing of movements (Penhune et al., 1998, Salman, 2002). The cerebellum

receives input from the sensory system of the spinal cord and other parts of the

brain, and integrate these inputs to fine-tune motor activity (Fine et al., 2002).

Damage to the cerebellum may occur as a result of stroke, head injury, cancer,

cerebral palsy, viral infection, or neurodegenerative diseases and may

subsequently cause individuals to experience tremors, difficulties in maintaining

balance, lack of muscle tone, speech difficulties, lack of control over eye

movement, difficulty in standing upright, and inability to perform accurate

movements (Fine et al., 2002). In addition, cerebellar neurons may become

damaged resulting in the loss of muscle control or coordination of movement in a

condition known as ataxia.

Some studies have shown that ataxia may also be triggered by the exposure to

toxins such as alcohol, drugs, or heavy metals (Manto, 2012, Barsottini et al.,

2014).

2.2. THE ENDOCRINE SYSTEM

The physiological activity of the endocrine system is closely linked to that of the

nervous system, with both systems coordinating the activity of the other (Brück,

1983). However, the differentiating aspect of the endocrine system is that its

effect is deployed through hormones secreted by glands located throughout the

body, conveyed in the bloodstream to particular areas in the body to regulate an

array of activities such as digestion, growth, metabolism and blood pressure

(Hiller-Sturmhöfel and Bartke, 1998).

http://etd.uwc.ac.za/

17

The hypothalamus is the brain structure that connects the nervous and the

endocrine systems (Cleghorn, 1955). It contains a small group of nuclei found at

the base of the forebrain and is responsible for the control of behaviour and such

rudimentary needs as sleep, hunger, thirst and sex as well as emotional and stress

responses (Zha and Xu, 2015). The hypothalamus also regulates the secretion of

hormones from the pituitary glands and other endocrine glands in the body

(Charlton, 2008).

2.2.1. ENDOCRINE DISRUPTOR COMPOUNDS (EDCs)

The United States Environmental Protection Agency (EPA) defines EDCs as “an

exogenous agent that interferes with synthesis, secretion, transport, metabolism,

binding action, or elimination of natural blood-borne hormones that are present

in the body and are responsible for homeostasis, reproduction, and developmental

process” (Diamanti-Kandarakis et al., 2009). Reports show that EDCs alter and

modify natural endocrine function, thus emerging as a major public health

challenge (Diamanti-Kandarakis et al., 2009, Schug et al., 2011, Zoeller et al.,

2014). These deleterious effects are primarily due to their potentially disruptive

activity on physiological processes, particularly through direct interaction with

steroid hormone receptors which are expressed abundantly in the hypothalamus

and other brain areas involved in the regulation of neuroendocrine functions

(Thomas and Doughty, 2004, Tokumoto et al., 2007).

Humans can take up EDCs or its precursors through consumption of contaminated

food, drinking water, inhalation and direct dermal contact with contaminated

products (Caballero-Gallardo et al., 2016). Studies have linked EDCs to adverse

http://etd.uwc.ac.za/

18

biological effects in animals, giving rise to concerns that its exposure might cause

serious medical effects (Colborn et al., 1993). Examples of some common EDCs

and their effects are summarized in Table 2.1.

http://etd.uwc.ac.za/

19

Table 2.3: Examples of EDCs and their endocrine disruptor effects

Name Uses Effects References

Endrin Insecticide and

pesticide

Competitively binds to androgen receptors; Toxic to fish and the

CNS

(Coble et al., 1967, Bhattacharya

and Mukherjee, 1975, Lemaire et

al., 2004)

Polychlorinated dibenzofurans (PCDF)

Insulators and

lubricants

Toxic to infants and is associated with memory and attention

deficits, decreased verbal abilities, and adverse behavioural as

well as emotional effects in early childhood.

(Henretig, 2009, Kodavanti et al.,

2017)

Heptachlor Insecticide and

pesticide

Toxic to the liver and the central nervous system as well as

reproductive, hematopoietic, immune, and renal systems.

(Fendick et al., 1990)

Hexachlorobenzene (HCB) Pesticide In animals, it induces neurological symptoms such as paralysis,

tremors and convulsions. In humans, it causes damage to the

liver, thyroid, nervous system, bones, kidneys, blood, and

immune and endocrine systems

(Addae et al., 2013)

Mirex Insecticide Human carcinogen and high toxicity to fish and other aquatic

animals

(Sanders et al., 1981, Spinelli et

al., 2007)

Dichlorodiphenyltrichloroethane

(DDT)

Pesticide Carcinogenic, early pregnancy loss, toxic to the reproductive

and nervous systems.

(Longnecker et al., 1997, Salleh et

al., 2015)

Aldrin Pesticide Carcinogenic (de Jong et al., 1997, Hooker et al.,

2014)

Chlordane Insecticide Carcinogenic, toxic to the reproductive systems (Fry, 1995, Persson and

Magnusson, 2015, Xiong, 2017)

http://etd.uwc.ac.za/

20

The adverse effect of EDCs can be seen during critical developmental stages such

as intrauterine, perinatal and in puberty, during which neuroendocrine systems are

modulated by steroid and other hormones (Frye et al., 2012). EDCs cross the

blood-placental and blood-brain barriers to interfere with development and

function, thus affecting pregnant mothers and children, both categories of which

have been considered the most susceptible to the harmful effects of exposure to

EDCs (Perera and Herbstman, 2011, Schug et al., 2015). Various reports indicate

that foetuses and offspring animals are exposed to EDCs through the placenta and

breast milk respectively, with such resultant effects as learning difficulties,

infertility and increased vulnerability to cancer not becoming evident until much

later in life (Poongothai et al., 2007, Li et al., 2013b). The major adverse health

effects arising from exposure to EDCs in humans include a myriad of

reproductive problems such as reduced count, motility and quality of sperms,

decreased female fertility, longer time to conception, higher miscarriage rates

(Singleton and Khan, 2003, Crain et al., 2008, Campion et al., 2012), increased

occurrence of testicular, prostate and breast cancer (Manibusan and Touart, 2017,

Balabanič and Klemenčič, 2018) and premature puberty in females (Zama et al.,

2016).

Some nervous system disorders may have their origins in endocrine disruptions,

particularly if the hippocampus and hypothalamus are targeted resulting in such

conditions as schizophrenia, bipolar disorders and cognitive dysfunctions (Brown

Jr, 2008; Meeker, 2012). A typical EDC that has been implicated in organ

function disruption and several other nervous system pathologies is 2, 2-bis (4-

hydroxyphenyl) propane, commonly known as BPA (Brown Jr, 2008).

http://etd.uwc.ac.za/

21

2.2.1.1. BISPHENOL A (BPA)

BPA is an industrial synthetic chemical commonly used in the production of a

range of polymers such as polycarbonate plastics and epoxy resins, as well as in

the production of several indoor applications and consumer products such as

thermal printer paper, in the lining of cans presently used for food and beverages.

It is also used for electronic equipment, water pipes, dental sealants, sports

equipment, medical devices, tableware, among others (Geens et al., 2011, Ribeiro

et al., 2017)

BPA is reported to be one of the highest chemicals produced worldwide, and there

has been an exponential increase in its use during the last 30 years, thus making

its potential for food and environment contamination higher (vom Saal and Myers,

2008, Gao et al., 2015). Human exposure to BPA is recognized as a widespread

occurrence, especially in developed countries, with reports indicating that

approximately 100 tonnes of BPA are released into the atmosphere each year, and

analyses of human samples showed the presence of BPA in most tested subjects

(Geens et al., 2012). The consumption of BPA-contaminated food is anticipated to

contribute to the overall BPA environmental exposure by more than 90% for all

age groups and its presence in the air, waste and drinking water, as well as in dust

particles has been reported (Vandenberg et al., 2007, Rudel et al., 2011, vom Saal

and Welshons, 2014).

Structurally, BPA is a diphenyl compound composed of two hydroxyl groups in

the ‘‘para’’ position connected by a methyl bridge, with two methyl functional

groups attached to the bridge making it structurally similar to hormones such as

http://etd.uwc.ac.za/

22

synthetic oestrogen, diethylstilbestrol (Kang et al., 2006). BPA is synthesized by

the condensation of acetone with two equivalents of phenol, and the reaction is

catalyzed by strong acids like hydrochloric acid (Figure 2.8) or a sulfonated

polystyrene resin (Uglea and Negulescu, 1991).

Figure 2.8: Synthesis of Bisphenol A

Prenatal BPA exposure is associated with neural, behavioural and bipolar

disorders in neonates, infants and children (Brown Jr, 2008). Reports indicate that

BPA induces hyperinsulinemia relating to type 2 diabetes mellitus and obesity at

very low doses and also triggers a high risk of miscarriage in women (Sugiura-

Ogasawara et al., 2005, Alonso-Magdalena et al., 2010).

2.2.1.1.1. PHYSICAL AND CHEMICAL PROPERTIES OF BPA

The physical and chemical properties of BPA are listed below.

Table 2.4: Physical properties of BPA.

Properties

Boiling point 220 oC (4 mmHg)

Melting point 158-159 oC

Molecular formula (CH3)2C(C6H4OH)2

Molar Mass 228.29 g·mol−1

Water solubility 21.5 oC

Vapour Pressure 25 oC

Density 1.20 g/cm³

BPA Acetone

Phenol Phenol

http://etd.uwc.ac.za/

23

2.2.1.2. RELEASE AND HUMAN EXPOSURE TO BPA

BPA has received considerable attention in recent years due to widespread

sources for human exposure through such routes as the environment (soil, air, and

aquatic) and contaminated foods. As a result of the high quantities of BPA

produced and an increase in the number of products based on epoxy resins and

polycarbonate plastics, there is widespread environmental contamination and

well-documented human exposure to BPA (Le et al., 2008). Although BPA is a

colourless solid soluble in organic solvents and poorly soluble in water, studies

have shown that BPA leaches from baby bottles, tin cans, reusable plastic water

bottles, and polycarbonate plastic containers into water, beverages, drink solutions

and food liquors, mostly due to the exposure of these BPA-containing plastics to

high temperatures (Grumetto et al., 2008, Le et al., 2008, De Coensel et al., 2009).

For instance, boiling to sterilize infant feeding bottles or addition of very hot

water or beverages to drinking bottles was reported to increase the rate of BPA

leaching by up to 55-fold (Le et al., 2008). Also, the discolouration, scratching of

polycarbonate, abnormal increase in pH and prolonged storage of water, drinks or

food in BPA-containing products increases the chances of leaching (Munguia-

Lopez et al., 2005, Biedermann-Brem et al., 2008).

In most industrialized countries, BPA detection in human amniotic fluid, serum,

breast milk and urine using various evaluation techniques has been reported

(Vandenberg et al., 2007) leading to the observation that BPA concentration in

human serum ranges from 0.2–1.6 ng/mL or 0.88–7.0 nM (Sajiki et al., 1999,

Takeuchi and Tsutsumi, 2002). In the United States, BPA was found to be present

in 95% of urine samples of 394 adults with a median concentration of 1.28 μg/L

http://etd.uwc.ac.za/

24

(Calafat et al., 2004) and according to the U.S. Environmental Protection Agency,

a reference dose of BPA is 50 mg kg/day was set while the European Union set a

no-observed-adverse-effect level (NOAEL) of 5 mg kg/day (Moriyama et al.,

2002, Gao et al., 2015).

2.2.1.3. METABOLISM OF BPA

Following oral intake of BPA in humans and rodents, reports show that BPA is

absorbed from the gastrointestinal tract and is glucuronidated by UDP-

glucuronosyltransferase (UGT, Figure 2.9) as a first-pass metabolism in the liver

to its main metabolite, BPA-glucuronide (Pottenger et al., 2000, Pritchett et al.,

2002, Völkel et al., 2002). The glucuronidation of BPA is a detoxification reaction

by UGT to detoxify BPA, its metabolite BPA-glucuronide is quickly excreted in

the urine due to its half-life of less than 6 hours. Bisphenol A-sulphate on the

other hand is reported to be a minor urinary BPA metabolite in humans (Ye et al.,

2005, 2006).

Following the oral intake of BPA in humans, the first-pass metabolism in the liver

is reported to be a super-effective process that results in a tremendously reduced

systemic availability (Pottenger et al., 2000, Upmeier et al., 2000). Also, BPA-

glucuronide and BPA-sulphate do not hinder hormonal control of reproduction,

thus indicating that these reactions are strictly detoxification pathways (Snyder et

al., 2000, Shimizu et al., 2002, Willhite et al., 2008). In rats, BPA is also

predominantly glucuronidated, with sulphation representing a minor pathway

(Pottenger et al., 2000), however the BPA-glucuronide formed is excreted from

the liver via the bile into the gastrointestinal tract, cleaved back to BPA and

http://etd.uwc.ac.za/

25

reabsorbed into the blood. Thus it undergoes enterohepatic recirculation resulting

in a significantly slower elimination of BPA including its conjugate in rodents

when compared with humans (Vandenberg, 2010).

Source: (Aschberger et al., 2010)

Figure 2.9: Metabolism of BPA

Several studies have suggested that the absorption and distribution of BPA in

maternal organs and foetuses are extremely rapid and that BPA easily crosses the

placenta after oral administration to pregnant rats (Miyakoda et al., 1999,

Takahashi and Oishi, 2000). Currently, there is an increase in research on the

adverse effects of foetal BPA exposure due to the ease of movement across the

placenta and the detection of low concentrations of endogenous UGT in foetal

tissues (Nishikawa et al., 2010, Angle et al., 2013). BPA exposure alters

secondary sexual characteristics and triggers neuronal, behavioural, and immune

disorders in foetuses and young children (Gies and Soto, 2013, Pouzaud et al.,

2018). In the human foetal liver, studies have shown that UGT is present at a

concentration five-times lower than in the adult liver and the UGT detoxification

http://etd.uwc.ac.za/

26

activities are not detected in foetal rat liver, thus suggesting that foetuses may be

more susceptible to the deleterious effects of BPA as most of it could be

accumulating in the tissues with potential consequencies (Matsumoto et al., 2002,

Jalal et al., 2017).

2.2.1.4. ACTIVITY OF BPA

BPA possesses estrogenic properties and acts as an agonist for oestrogen receptors

(Matsushima et al., 2010, Shanle and Xu, 2010b). For instance, the E-SCREEN,

often regarded as the most sensitive assay for oestrogenicity based on its ability to

differentiate between partial and full agonists, has confirmed the estrogenic

properties of BPA (Matsushima et al., 2010). Prepubescent CD-1 mice treated

with BPA demonstrated estrogenic responses such as increased expression of the

oestrogen-inducible protein lactoferrin, improved uterine wet weight and luminal

epithelial height (Markey et al., 2001). In another study, ovariectomized rats

exposed to BPA treatment experienced triggered proliferation of the uterine and

vaginal epithelial cells and induced estrogenic responses in the mammary and

pituitary glands (Colerangle and Roy, 1997, Steinmetz et al., 1998).

There is evidence in literature which indicates that exposure to BPA during early

development may increase breast cancer risk (Murray et al., 2007). This assertion

is supported by studies showing that BPA activates genes involved in the growth

of mammary glands and promotes breast cancers in rats after exposure to known

carcinogens at a dose that would not cause cancer in BPA-untreated mice

(Betancourt et al., 2010, Wadia et al., 2013). In treated male rats, a reduction in

sperm count, testosterone and luteinizing hormone (LH) as well as in the weights

http://etd.uwc.ac.za/

27

of the testes, epididymis, seminal vesicle and prostate gland were reported

(Sakaue et al., 2001, Akingbemi et al., 2004, Nakamura et al., 2010).

A report by the National Institutes of Health in Norway indicates that BPA has

adverse effects on foetal and infant brain development and behaviour. In

agreement with the previous report, the U.S. National Toxicology Program (NTP)

expressed concerns on the effects of BPA on the prostate gland, brain and

behaviour in foetuses, infants, and children (Abdallah, 2016). In rodents and non-

human primates, it has been reported that BPA affects the brain even at relatively

low exposure levels (Leranth et al., 2008, Nakagami et al., 2009). In-vivo studies

demonstrate that perinatal or neonatal BPA exposure modifies brain sexual

differentiation (Patisaul et al., 2006, Rubin et al., 2006), and initiates aggression,

anxiety, cognitive deficits, and learning-memory impairment (Miyagawa et al.,

2007, Tian et al., 2010, Xu et al., 2010b). BPA also induces changes in the

differentiation of ectodermal tissues, including neural tissues in cynomolgus

monkeys (Yamamoto et al., 2007), and triggers apoptosis in the central neurons of

tadpoles leading to head, vertebral and abdominal developmental deficits (Oka et

al., 2003).

Whereas BPA exposure in male rats during lactation is linked to hyperactivity and

the loss of dopaminergic neurons (Ishido et al., 2007), exposure during gestation

and lactation led to significant changes in the Glutamate/Aspartate (G/A) ratio in

the hippocampus of the cerebrum in Sprague-Dawley rats, thus suggesting

neuronal and glial developmental alterations (Kunz et al., 2011).

http://etd.uwc.ac.za/

28

2.3. ROOIBOS (Aspalathus linearis)

Rooibos tea is prepared from a leguminous shrub, Aspalathus linearis which is

native to the Cederberg Mountains of the Western Cape region of South Africa

(Joubert et al., 2004) and is commonly used in the fermented (red; Figure 2.10) or

unfermented (green) form (Joubert et al., 2004). Fermentation is the enzymatic

oxidation of polyphenols and other components in the leaves of the plant, thus

converting the green grassy-smelling and malty tasting tea to the sweet-smelling

and fruity red tea (Krafczyk and Glomb, 2008).

(A) (B)

Source: http://www.carmientea.co.za/tea-variants/rooibos/ & http://amavida.com/learn/

Figure 2.10: Rooibos tea; (A) Unfermented (B) Fermented

Rooibos tea has become a popular and reputable herbal beverage due to its

characteristic taste, absence of alkaloids and low tannin content (Cheney and

Scholtz, 1963, Jaganyi and Wheeler, 2003). In 1968, Annetjie Theron discovered

that an infusion of rooibos alleviated prolonged restlessness, vomiting and

stomach cramps when administered to her colicky baby, thus leading to its

classification as a healthy beverage. This led to a broader consumer base, and

http://etd.uwc.ac.za/

29

several babies have since been reared with rooibos either added to their milk or

given as a weak brew (Joubert et al., 2008).

Other pharmacological reports suggest that rooibos relieves ingestion, heartburn

and nausea, improves appetite, decreases nervous tension, enhances sound sleep

and has anti-allergic effects (Morton, 1983, Van Wyk et al., 1997). Rooibos

formulations have also been reported to be capable of relieving such

dermatological complications as eczema, rashes and acne, leading to the

production of toiletries and cosmetic products, now sold through many

supermarkets and beauty shops (Joubert et al., 2008).

2.3.1. CHEMICAL COMPOSITION OF ROOIBOS

The leaf tannin content of fermented rooibos is between 3.2% to 4.4% (Reynecke

et al., 1949, Blommaert and Steenkamp, 1978) and traces of the alkaloid,

sparteine, flavonoids (aspalathin and aspalalinin) have been detected in this herbal

tea (Rabe et al., 1994, Shimamura et al., 2006). Other compounds detected in

rooibos include nothofagin, flavones (orientin, iso-orientin, vitexin, isovitexin,

chrysoeriol, luteolin and luteolin-7-O-glucoside), flavanones (dihydro-orientin,

dihydro-iso-orientin and hemiphlorin), flavonols (quercetin and its glycosides,

quercetin-3-robinobioside, hyperoside, isoquercitrin and rutin), phenolic acids,

lignans, esculetin, orientin, iso-orientin, and trace quantities of the aglycones,

luteolin, quercetin and chrysoeriol (Joubert et al., 2008).

http://etd.uwc.ac.za/

30

Table 2.3: Quantification of the major flavonoids in aqueous rooibos extract (mg/500 ml)

brewed with 500 ml of boiled water and 10 g of dried green rooibos tea leaves.

Phenolic compounds mg/500 ml µmol

Aspalathin 287 ± 0.1 636 ± 20

Nothofagin 34.4 ± 0.0 79 ± 3.1

Isoorientin 26 ± 0.0 58 ± 2.6

Orientin 17 ± 0.0 38 ± 1.8

Rutin 7.9 ± 0.0 13 ± 0.3

Hyperoside 0.8 ± 0.0 1.7 ± 0.1

Isoquercitrin 2.9 ± 0.0 6.3 ± 0.4

Vitexin 3.1 ± 0.0 7.1 ± 0.2

Isovitexin 2.8 ± 0.0 6.6 ± 0.3

Luteolin-O-galactoside 0.9 ± 0.0 1.9 ± 0.2

TOTAL 383 ± 0.1 848 ± 29

Source: (Breiter et al., 2011)

2.3.1.1. POLYPHENOLS

The potential health benefits and bioactivity of rooibos have been linked to its

naturally-occurring polyphenolic content (Joubert and Ferreira, 1996, McKay and

Blumberg, 2007, Joubert and de Beer, 2011). Polyphenols have received great

reviews in the past decade due to their abundance in diets as well as their

antioxidative action, regulation of several protein functions and the prevention of

such oxidative stress-related diseases as cancer and neurodegenerative disorders

(Joubert and Ferreira, 1996, Canda, 2012). Polyphenols scavenge for free radicals

and neutralize reactive oxygen species (ROS) by altering free radical chain

reactions as well as control the activities of enzymes and chelating metal ions that

partake in the formation of free radicals (Perron and Brumaghim, 2009, Fraga et

al., 2010).

http://etd.uwc.ac.za/

31

Polyphenols are subdivided into flavonoids and phenolic acids (Figure 2.11), and

may also be further classified as monomeric molecules (containing one unit) or

polymeric molecules (containing more than one unit) (Erickson, 2003).

Source: Modified from (Mentor, 2014)

Figure 2.11: Classification of polyphenols

Regardless of its considerable reduction during fermentation, aspalathin (Figure

2.12) is the main flavonoid of unfermented rooibos and is one of the major

components of the water extract of fermented rooibos (Joubert, 1996, Bramati et

al., 2002).

Figure 2.12: Chemical structure of Aspalathin

http://etd.uwc.ac.za/

32

2.3.2. BIOLOGICAL PROPERTIES OF ROOIBOS TEA

As a result of its high polyphenolic contents, rooibos contains antioxidants that

scavenge free radicals and thus prevent cellular oxidative damage (Joubert et al.,

2008). The antidiabetic potential of the rooibos tea extract was confirmed using a

glucose-lowering test of its flavonoids in streptozotocin-treated rats (Muller et al.,

2012). In this regard, aspalathin has been reported to have a beneficial effect on

glucose homoeostasis through the stimulation of glucose uptake in muscle tissues

and increased insulin secretion from pancreatic β-cell, thus preventing type II

diabetes in mice (Kawano et al., 2009).

The aqueous extract of rooibos tea extract also exhibited bronchodilatory,

antispasmodic and blood pressure lowering activities in rabbits, guinea-pigs and

rats through the activation of KATP with the selective bronchodilatory effect, thus

validating its therapeutic potential in the treatment of congestive respiratory

diseases (Khan and Gilani, 2006). In different studies, treatment of rats with

rooibos tea improved sperm concentration, motility and viability (Opuwari and

Monsees, 2014) while consumption of the fermented tea considerably improved

the lipid profiles and redox status in adults predisposed to the development of

cardiovascular diseases (Marnewick et al., 2011).

The natural antioxidant and hepatoprotective activity of rooibos tea extract were

demonstrated via its inhibition of increased concentrations of triacylglycerols,

cholesterol, malondialdehyde, aminotransferases, bilirubin and alkaline

phosphatase in rats treated with carbon tetrachloride (Bosek and Nakano, 2003).

Fermented rooibos tea was reported to prevent obesity through the inhibition of

http://etd.uwc.ac.za/

33

adipogenesis and adipocyte metabolism in 3T3-L1 adipocytes, thus highlighting

its potential in preventing obesity (Sanderson et al., 2014). In a different study,

rooibos increased the activity of cytosolic glutathione S-transferase alpha,

microsomal UGT and glutathione, in addition to the modulation of phase II drug

metabolizing enzymes and oxidative status in the liver of treated rats, thus

suggesting that it is important in protecting against the adverse effects related to

mutagenesis and oxidative damage (Marnewick et al., 2003). Similar

antimutagenic properties of fermented and unfermented rooibos tea in preventing

the transformation of a mutagenic compound into a mutagen was investigated

with the Salmonella typhimurium mutagenicity assay, and rooibos showed potent

antimutagenic activity against 2-acetylaminofluorene and aflatoxin B1-induced

mutagenesis (Van der Merwe et al., 2006).

2.3.4.1. NEUROPROTECTIVE EFFECTS OF ROOIBOS

The brain is one of the most sensitive targets of oxidative stress (Garbarino et al.,

2015). It accounts for approximately 2% of total body mass but consumes 15–

20% of the energy produced in the human body, and this increase in mass-specific

metabolic rate is ascribed to the high amount of omega-three polyunsaturated fatty

acids in brain tissue which are vulnerable to peroxidation (Hulbert et al., 2007). In

the brain tissue, there are high amounts of redox-active iron and copper, thereby

increasing its susceptibility to oxidative damage. Also, the brain is mostly unable

to replace impaired cells since it is composed generally of terminally

differentiated glia and neurons (Garbarino et al., 2015).

http://etd.uwc.ac.za/

34

When there is excessive production of ROS, endogenous defence mechanisms

against ROS may not be sufficient to suppress ROS-associated oxidative damage,

consequently, exogenous dietary antioxidants are needed to supplement and

protect neurons against oxidative damage (Liu et al., 2018). Several natural

beverages such as rooibos are reported to be good sources of antioxidants due to

its polyphenolic content and have been subsequently utilized against a variety of

oxidative stress-induced neurodegenerative disorders such as Alzheimer’s and

Parkinson’s disease (Hong et al., 2014). In one study, rooibos tea showed efficient

protection against immobilization-induced oxidative stress in rats through the

reduction of stress-related metabolites (5-HIAA and FFA), prevention of lipid

peroxidation, restoration of stress-induced protein degradation, regulation of

glutathione metabolism and modulation of such antioxidant enzymes as

superoxide dismutase and catalase (Hong et al., 2014). The administration of

rooibos was observed to suppress the age-related accumulation of lipid peroxides

in several regions of rat brains (Inanami et al., 1995). Also, prolonged

consumption of fermented rooibos tea was found to offer neuroprotection against

ischemic brain injury through the attenuation of brain oedema and neuronal

apoptosis as well as improving neurobehavioural outcomes in rats given rooibos

tea compared to the untreated rats (Akinrinmade et al., 2017).

Although there is evidence in literature on the protective activity of rooibos

against some oxidative stress-inducing agents, its neuroprotective activity on the

development and function of the developing CNS is not clearly known. In the

present study, the neuroprotective effects of rooibos on the developing Wistar rat

hippocampus and cerebellum exposed to BPA during gestation and lactation was

http://etd.uwc.ac.za/

35

investigated. Results from this study are expected to provide the first research

evidence on the benefits of rooibos tea as a remedy for ameliorating potential

BPA toxicity in the developing CNS using Wistar rats exposed to BPA.

http://etd.uwc.ac.za/

36

CHAPTER 3

MATERIALS AND METHODS

3.1. ETHICAL CONSIDERATION

Ethical clearance for this study was provided by the Faculty of Natural Science

Research Ethics Committee of the University of the Western Cape, Cape Town,

South Africa. Ethical and project registration numbers were assigned to the

research project before commencement (Ethics Registration No: 2013-0410).

3.2. MATERIALS

The materials and equipment used in this study are shown in Table 3.1 and 3.2.

Table 3.4: Chemicals used in this study

Chemicals Supplier

Bisphenol A (BPA) Sigma-Aldrich (USA)

Ethanol Sigma-Aldrich (USA)

Sodium Chloride (NaCl) Merck ( SA)

Formaldehyde Sigma-Aldrich (USA)

Sodium hydroxide (NaOH) Merck ( SA)

Di-sodium hydrogen phosphate (anhydrous) (Na2HPO4) Merck (Germany)

Sodium di-hydrogen orthophosphate (Anhydrous)

(NaH2PO4)

Associated chemical enterprises

(SA)

Potassium chloride (KCL) Merck ( SA)

Sodium pentobarbitone Norpham Medical (SA)

DPX Kimix (SA)

Paraffin wax Merck (Germany)

Haematoxylin and eosin (H&E) stain Kimix (SA)

Cresyl Violet (CV) stain Kimix (SA)

Perchloric acid (PCA) Sigma-Aldrich (USA)

http://etd.uwc.ac.za/

37

Table 3.5: Equipment used in this study

Product Supplier

Automatic tissue processor Duplex processor, Shandon Elliott (UK)

Autostainer machine Leica AutoStainer XL, (Germany)

Weighing Balance Adam, Keynes (USA) and Radwag Wagi (Poland)

Centrifuge Eppendorf centrifuge (Germany)

Embedding system Leica, EG1160 (Germany)

Liquid chromatographic system Waters Acquity (USA)

Microtome Leica, RM 2125 RT (Germany)

Water bath Leica SMM (Germany)

3.3. ANIMALS

Forty (40) adult female Wistar rats approximately three months old, with an

average weight of 250g were used in this study. Animals were procured from the

University of Stellenbosch animal facility, Cape Town, South Africa and

maintained under standard laboratory conditions at the University of the Western

Cape animal house. Daily body masses of rats were measured using a weighing

balance, and progressive weight changes relative to the initial weights were noted.

Mating was implemented by keeping one male rat with one female rat (1:1) in a

cage for seven days (Figure 3.1).

During mating, pregnancy and post-partum periods, animals were allowed free

access to standard rat chow and tap water. Successful mating was confirmed by

the presence of a vaginal plug or spermatozoa in the vaginal smear the following

morning (Ramírez-López et al., 2016). Pregnant dams were randomly assigned to

four treatment groups of ten dams each (n = 10; total = 40).

http://etd.uwc.ac.za/

38

Figure 3.3: Flow diagram showing the experimental design for the study (10 rats per

group; G1=control, G2= BPA only, G3= rooibos + BPA and G4= rooibos tea only

3.4. TREATMENT PROTOCOL

Throughout pregnancy, dams were randomly assigned to four different groups of

ten rats each (n=10) and housed in separate cages as summarized below:

G1 received 9 % normal saline ad libitum (NS)

G2 received 400µg/ kg/day BPA (BPA)

G3 received 20 % fermented rooibos tea and 400µg/ kg/day BPA (RB)

G4 received 20 % fermented rooibos tea ad libitum (RT)

After parturition, the pups remained with mothers in the respective cages

maintained at temperature 21–24ºC under a 12 hours’ light and 12 hours’ darkness

cycle. The treatment was stopped after delivery on PND 21 (weaning day) and the

pups were kept in separate cages and were allowed food and tap water ad libitum.

Female Rat

G1: NS G2: BPA G3: RB G4: RT

Male Rat

http://etd.uwc.ac.za/

39

The day of birth was considered as PND 0 and pups were sacrificed at the end of

the experiment on postnatal day (PND) 42. It is important to note that the

treatment was only applied to the pregnant dams in the groups and not the

offspring. The offspring rats were thus exposed to treatment only during

pregnancy (via the placenta) and lactation. A total of 12 pups per group were

sacrificed on each of the assigned days.

3.5. PREPARATION OF REAGENTS

3.5.1. ROOIBOS HERBAL TEA PREPARATION

The fermented rooibos herbal tea used in this study was a gift from Rooibos Ltd

(Clanwilliam, South Africa) to the research laboratory of Prof. Thomas Moonses

at the Department of Medical Biosciences, University of the Western Cape, South

Africa. A concentration of 0.02g/ml of fermented rooibos herbal tea reported to be

the routine amount used by rooibos consumers for tea-making purposes

(Marnewick et al., 2003, Pantsi et al., 2011) was used throughout this study.

Briefly, 1000ml freshly boiled tap water was added to 20g of fermented rooibos

leaves and stems for 5 minutes followed by filtration with cheesecloth and

Whatman’s filter papers (no 4 and 1 respectively). The aqueous extract was then

allowed to cool off to room temperature before administration to rats ad libitum.

Fresh tea was prepared daily while intake of rooibos tea and water was measured

throughout the experimentation period by subtracting the volume of the remaining

fluid from the initial volume. No major fluid leakage from water bottles was

observed, and the average consumption rate per cage was 40ml/day.

http://etd.uwc.ac.za/

40

3.5.2. BISPHENOL A (BPA) AND NORMAL SALINE

BPA was dissolved in 1% absolute ethanol, diluted in water, and administered

orally as the vehicle at a concentration of 400µg/ kg/day. This concentration was

selected based on the findings from a previous study (Ahmed and Eid, 2015). An

oral route of BPA administration to the pregnant and lactating female was chosen

throughout this study to mimic the most likely route of exposure of the compound

in humans and wildlife. Glass water bottles were used in this study to ensure that

related compounds did not leach from plastic water bottles to possibly confound

the results of this study. The preparation of 0.9% normal saline was done by

adding 9g of Sodium Chloride (NaCl) to a litre (1000ml) of distilled water.

3.5.3. PREPARATION OF 4% FORMALDEHYDE SOLUTION

The formaldehyde solution was needed to preserve the specimens for histological

and immunohistochemical studies. Briefly, a litre of 4% formaldehyde was

prepared by adding 40 g of paraformaldehyde powder into 700 ml of distilled

water at 60 °C. The paraformaldehyde solution was stirred on a burner, and two

drops of 2 M sodium hydroxide (NaOH) solution were added until the

paraformaldehyde solution was clear. The volume of solution was made up to 1 L

with distilled water. The solution was prepared in small amounts as needed to

ensure that only fresh solutions were used each time.

3.5.4. PHOSPHATE BUFFERED SALINE (PBS)

PBS was prepared by adding 1.44 g anhydrous di-sodium hydrogen phosphate

(Na2HPO4), 0.24 g anhydrous sodium di-hydrogen orthophosphate (NaH2PO4), 8

g of sodium chloride (NaCl) and 0.2 g of potassium chloride (KCL). The

http://etd.uwc.ac.za/

41

chemicals were dissolved in 800 ml until complete dissolution. The pH of the

solution was confirmed to be 7.4 by a pH meter and was thereafter made up to

1000 L.

3.6. MOTOR FUNCTION TESTS (NEUROBEHAVIORAL TESTS)

Neurobehavioral studies are commonly used to evaluate behavioural changes

induced by exposure to treatments of varying types, concentrations and dosages

(Seale et al., 2012). Common neurobehavioral parameters include locomotor

activity, cognitive function (e.g. memory deficit, etc.), gait, depression, anxiety,

equilibrium, etc. In this study, only the open field test (OFT) and the novel object

recognition (NOR) tests were done due to some constraints.

3.6.1. OPEN FIELD TEST (OFT)

The OFT is a commonly used neurobehavioral assessment tool that provides

simultaneous measurement of locomotion and anxiety in laboratory animals

(Kendigelen et al., 2012). On PND 42, the OFT was performed in the OFT

apparatus (Figure 3.2), a square plexiglass box (72×72 cm with 36cm walls) with

a video camera (Samsung HMX-F90, South Korea) positioned above the

apparatus to record each trial for subsequent analysis.

The open-field arena was divided into 16 equal squares, via a 4 × 4 grid, for ease

of data analysis. Animals were tested singly, each transported from the housing

room to the testing room and allowed to acclimatize in the OFT apparatus. Pre-

testing was done for two days to prepare the animals. On the third day, each rat

was placed in the centre zone of the OFT arena and observed for 5 minutes. This

was repeated twice after which the rat was returned into its home cage and the

http://etd.uwc.ac.za/

42

OFT box cleaned with 70% ethanol before testing the next rat. Video recording of

all tests was done using the overhead camera, and recordings were analysed using

the Smart video tracking software version 3.0, from Panlab Harvard Apparatus

(Massachusetts, USA) to measure the locomotor activity of each rat by extracting

the total distance travelled in the OFT arena. As a measure of anxiety, the total

distance travelled in the 12 squares near the walls was compared with the distance

travelled in the four squares at the centre of the arena. These assays were

performed my the researcher as well as two blind” observers, and the results were

averaged.

Source: http://www.neuralconnections.net/2011/10/stress-receptors-and-responses.html

Figure 3.4: Open Field Test Apparatus.

The activities evaluated include the frequency of rearing episodes (the number of

times the rat stood on its hind limbs with its forelimbs against the wall of the

http://etd.uwc.ac.za/

43

testing cage), total freezing time and total self-grooming activity (the duration that

a rat spends licking or scratching itself while remaining stationary).

3.6.2. NOVEL OBJECT RECOGNITION (NOR) TEST

The NOR test is widely used to evaluate cognition, particularly recognition

memory, in rodent models of CNS disorders. This test measures the ability of rats

to recognize a novel object in an otherwise familiar environment, thus reflecting

the use of learning and recognition memory (Ennaceur and Delacour, 1988,

Ennaceur, 2010). The apparatus for the NOR test included a square plastic/glass

box (40x40x25 cm), with a digital camera (Samsung HMX-F90, South Korea)

mounted directly above it. The object test arena was divided into four equal

squares via a 2 × 2 grid, and rats were tested singly.

The NOR test was conducted with two different kinds of objects which were

different in shape, colour, and texture. The task procedure consisted of three

phases: habituation, familiarization, and test phase. During habituation, the rats

were allowed to explore an empty arena. Twenty-four hours after habituation

(familiarization), the rats were exposed to the familiar arena with two identical

objects placed at an equal distance. The next day (test phase), the rats were

allowed to explore the arena in the presence of the familiar object (old object) and

a new object to test recognition memory. Recognition memory was evaluated

using a recognition index (RI) for each animal calculated using the formula: RI =

TN/ (TN + TF) where TN and TF represent the time an animal spends with the

novel object (TN) versus a familiar (old) one (TF) respectively. The testing box

was cleaned with 70% ethanol before testing the next rat.

http://etd.uwc.ac.za/

44

3.7. ANIMAL SACRIFICE

On PND 42, the final body masses of the animals were taken before injection of

150 mg/bw (i.p) sodium pentobarbital and subsequent decapitation under deep

anaesthesia. The neural specimens of interest were carefully dissected out and

either fixed in 4% paraformaldehyde solution (for histology and

immunohistochemical studies) or stored in cold phosphate buffered saline (PBS)

(mainly for biochemical studies).

3.8. HISTOLOGICAL PREPARATION OF BRAIN SAMPLES

3.8.1. AUTOMATED TISSUE PROCESSING

After two days in 4% paraformaldehyde solution, brain specimens were put in

appropriately labelled cassettes before processing in a Leica-2125 automated

tissue processor, ATP (Leica, Germany) for 7 hours in preparation for sectioning,

staining and microscopic analysis. The programme selected for the specimens

consisted of 4 hours of dehydration (through different grades of ethanol to remove

water), 1 hour 30 minutes of xylene clearing (to remove ethanol) and 1 hour 30

minutes of infiltration with molten paraffin wax (to displace xylene, Table 3.3).

Table 3.6: Tissue Processing Protocol

Step Solution Time (min) Temperature (ºC)

1 10% Formalin 30 Room Temperature

2 70% Ethanol 30 Room Temperature

3 96% Ethanol 30 Room Temperature

4 96% Ethanol 30 Room Temperature

5 99.9% Ethanol 30 Room Temperature

6 99.9% Ethanol 30 Room Temperature

7 99.9% Ethanol 30 Room Temperature

8 Xylene 30 Room Temperature

9 Xylene 30 Room Temperature

10 Paraffin 60 60

11 Paraffin 60 60

12 Paraffin 60 60

http://etd.uwc.ac.za/

45

3.8.2. EMBEDDING

The cassettes of CNS specimens were removed from the last change of paraffin

wax in the ATP (Leica, Germany), and samples were manually removed from

cassettes. After that, samples were carefully placed in steel moulds containing

liquid paraffin wax in the desired orientation, and more liquid wax was added

until the samples were completely covered by paraffin wax. The moulds were

then transferred to the refrigerating plate of an embedding machine to allow the

wax to solidify. Tissue blocks were later removed from the moulds and

refrigerated until sectioning.

3.8.3. SECTIONING

Tissue blocks were removed from refrigeration and allowed for two hours before

trimming to appropriate dimensions followed by sectioning at 5 microns using a

Leica TP-1020 microtome (Leica, Germany). Each section was placed in warm

water to allow for spreading before mounting on a labelled glass slide.

3.8.4. STAINING

Standard haematoxylin and eosin (H&E), cresyl violet as well as

immunohistochemical (GFAP) staining were done for morphometric studies.

3.8.4.1. HAEMATOXYLIN AND EOSIN

H & E staining was done with an autostainer machine (Leica AutoStainer XL) at

the Histology Laboratory of the University of Stellenbosch, South Africa using a

standardized protocol. Briefly, the glass slides were placed on a staining rack,

deparaffinized in two changes of xylene for 5 minutes, rehydrated by immersion

http://etd.uwc.ac.za/

46

in two changes of 99% ethanol for 2 minutes followed by 96% and 70% ethanol

for two minutes each. Thereafter the slides were rinsed in running tap water for at

least two minutes before staining in haematoxylin for 8 minutes. The slides were

then placed in running water at room temperature for 5 minutes, differentiated in

acid alcohol for 2-3 seconds before being rinsed in running water. Slides were

counterstained in eosin for 4 minutes, and thereafter rinsed in running tap water

for at least one minute followed by dehydration in three different baths of alcohol

(70%, 96%, and 99%) for 30 seconds, one minute and 30 seconds respectively and

finally cleared in xylene for one minute. Coverslips were then mounted on the

slides using DPX mounting medium and left to dry for 48hours (Cardiff et al.,

2014).

3.8.4.2. CRESYL VIOLET (CV) / NISSL STAINING

The CV staining technique is commonly used to study cellular cytoarchitecture in

neural tissues because it presents clearly delineated neuronal perikaryons in

comparison to the modest rendition of shape and size of cell bodies offered by

staining with haematoxylin and eosin (Akinrinmade et al., 2017). Briefly, sections

were deparaffinized in xylene twice for 10 minutes each and then hydrated in

100% ethanol twice for 5 minutes each, then in 95% ethanol for 3 minutes and

again in 70% ethanol for 3 minutes. Hydrated sections were then rinsed in tap

water and distilled water, stained in 0.1% Cresyl violet for 5 minutes and

thereafter rinsed quickly in distilled water. Sections were further differentiated in

95% ethanol for 2 minutes and then dehydrated in 100% ethanol for 10 minutes,

cleared in xylene for 10 minutes before coverslip mounting with DPX and drying

for 48 hours.

http://etd.uwc.ac.za/

47

3.8.4.3. IMMUNOHISTOCHEMICAL (IHC) STAINING

All immunohistochemical (IHC) staining procedures were done using the Leica

Bond Autostainer and the Bond Polymer at the Histology Laboratory of the

University of Stellenbosch, South Africa using a standardized protocol. The tissue

sections were then manually rehydrated in ethanol grades, cleared in xylene,

mounted with DPX solution and then left to dry for 48 hours. (Table 3.4).

Table 3.4: IHC staining procedure

Step Type Incubation

Time

Temperature Dispense Type

1 Peroxide Block 5 min Ambient selected vol.

2-4 Bond Wash Solution 0 min each Ambient selected vol.

5 Primary Antibody 15 min Ambient selected vol.

6-8 Bond Wash Solution 0 min each Ambient selected vol.

9 Post Primary 8 min Ambient selected vol.

10-12 Bond Wash Solution 2 min each Ambient selected vol.

13 Polymer 8 min Ambient selected vol.

14-15 Bond Wash Solution 2 min each Ambient selected vol.

16-17 Deionized Water 0 min each Ambient selected vol.

18 Mixed DAB Refine 10 min Ambient selected vol.

19-21 Deionized Water 0 min each Ambient selected vol.

22 Haematoxylin 5min Ambient selected vol.

23-25 Deionized Water 0 min Ambient selected vol.

Rehydration and clearing

Step Solution Duration

1 70% alcohol 5 dips

2-3 96% alcohol 5 dips each

4-5 99% alcohol 5 dips each

6-7 Xylene Dip for 1 min each

Mounting for observation was done using DPX and slide cover slips.

3.9. NEUROCHEMICAL ASSAYS

Neurochemical assays are often done to determine the quantities of biological

molecules and compounds of interest, ranging from neurotransmitters,

neuromodulators, reactive oxygen species, neurosteroids, ions, neurohormones,

neuro enzymes, immunogens, ingested drugs, neurotoxins etc. The neurochemical

http://etd.uwc.ac.za/

48

assays done in this study include superoxide dismutase (SOD) and catalase (CAT)

mainly to evaluate cerebral and cerebellar antioxidant enzyme activity.

3.9.1. HOMOGENIZATION OF TISSUES

Brain tissues were homogenized in 10 times (w/v) 0.1M PBS (pH 7.4) in a Teflon

glass homogenizer (IKA Laboratories, Germany) for two periods of 10 seconds

each. The homogenate obtained was then centrifuged at 15,000 rpm in a

microcentrifuge at 4°C for 10 minutes and the supernatant collected and

transferred into newly marked Eppendorf tubes for relevant neurochemical assays.

3.9.1.1. SUPEROXIDE DISMUTASE (SOD)

SOD activity assays measure the amount of enzyme that inhibited the oxidation of

pyrogallol, expressed as units per milligram of protein. In this study, the

supernatants were collected on ice and the SOD colorimetric activity kit (Life

Technologies, USA) was used to measure SOD activity according to the

manufacturer’s instructions. Briefly, 10μL of standards and samples, 50μL of 1X

substrate and 25µL of 1X xanthine oxidase were added to each well of a 96 well

half area plate and then incubated for 20 minutes at room temperature.

Absorbance was read using a Polarstar Omega plate reader (BMG Labtech, USA)

at 450nm and a curve-fitting software was used to generate the standard curve

using a four parameter algorithm.

3.9.1.2. CATALASE

The catalase colorimetric activity kit (Life Technologies, USA) was used to

measure the activity of catalase in the samples. Catalase is a highly conserved

http://etd.uwc.ac.za/

49

enzyme expressed in all mammalian tissues and catalyzes the hydrolysis of

hydrogen peroxide (H2O2) into H2O and O2, to prevent the potential harmful

effects of excessive levels of H2O2 (Nagata et al., 1999, Agati et al., 2012). The

supernatants were collected on ice and experiments were performed according to

manufacturer’s instructions. Briefly, 25μL of standards, 25μL of the sample and

25μL of hydrogen peroxide reagent were added into each well and incubated for

30 minutes at room temperature. Thereafter, 25 µL of the substrate and 25μL of

1X HRP solution were added to each well and further incubated for 15 minutes at

room temperature. Absorbance was read using a Polarstar Omega plate reader at

560nm, and a curve-fitting software was used to generate the standard curve using

a four parameter algorithm.

3.10. STATISTICAL ANALYSIS

The GraphPad Prism Software V7 was used for all statistical analyses

(www.graphpad.com/scientific-software/prism/) and data obtained was expressed

as mean ± standard error of mean (SEM). A one-way analysis of variance

(ANOVA) followed by Tukey's multiple comparison post-hoc test was performed

to determine statistical significance (p<0.05).

http://etd.uwc.ac.za/

50

CHAPTER 4

RESULTS

4.1. INTRODUCTION

In this study, the potential neuroprotective effects of fermented rooibos herbal tea

on the development of the cerebrum and cerebellum was evaluated in rats exposed

to BPA during pregnancy and lactation. Two groups of pregnant Wistar rat dams

(RB and RT) were administered fermented rooibos herbal tea ad libitum during

pregnancy and lactation while the third group of pregnant Wistar rat dams (BPA)

were administered with BPA only. The fourth group of dams (NS) was

administered normal saline ad libitum and offspring from this group served as

controls.

Daily consumption of rooibos tea and water was measured throughout the

experimentation period by deducting the volume of the remaining fluid from the

original volume; the average intake per cage was 40ml/day throughout the study

period. No major fluid leakage from the water bottles was detected. Rats were

allowed ad libitum access to tap water, normal saline and rooibos tea depending

on the treatment groups. Offspring rats from each group (n = 12) were subjected

to neurobehavioural assessment on PND 42, weighed and sacrificed for further

neurochemical, histological and immunohistochemical evaluation.

The control group (NS) results were used as non-pathological reference for all

other groups and a one-way ANOVA multiple comparisons test and Turkey’s

http://etd.uwc.ac.za/

51

multiple comparisons post-hoc test (GraphPad Prism version 7.0) were used to

compare differences between respective treatment groups for all parameters.

Findings from this study will provide information on the ability of natural

products to protect the developing brain against the deleterious effects of EDCs.

4.2. EFFECTS OF BPA AND FERMENTED ROOIBOS HERBAL TEA ON

BODY MASS

Previous studies have indicated that perinatal BPA exposure leads to body mass

increases in offspring rats (Rubin et al., 2001, Miyawaki et al., 2007). The effects

of rooibos tea and BPA treatments on changes in body mass in offspring Wistar

rats was investigated as part of the present study. Findings show that by PND 42,

BPA treatment led to a significant increase (p<0.0001, Figure 4.1) in body masses

when compared to the controls. The consumption of rooibos tea appeared to have

prevented the BPA-induced body mass increment as corroborated by a significant

reduction (p<0.0001) in the body masses of offspring rats in the RB group when

compared to the BPA group. There was no significant difference (p>0.05) in the

body masses of offspring rats in the RT group when compared to the control

group.

http://etd.uwc.ac.za/

52

NS

BP

RB

RT

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0

Av

era

ge

bo

dy

we

igh

t (g

)

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=12). P<0.0001 vs NS; фP<0.0001 vs BPA

Figure 4.2: Average body mass of experimental rats on PND 42

4.3. NEUROBEHAVIOURAL TESTS

4.3.1. THE OPEN FIELD TEST

The open field test is one of the most commonly used measures of animal

neurobehavioral deficits. In the present study, video recordings were analysed to

evaluate the frequency of rearing episodes, total freezing time and total self-

grooming time on PND 42. Three different observers captured the data and results

were averaged to obtain the final values which were analysed statistically for

comparisons between the respective groups.

4.3.1.1. THE FREQUENCY OF REARING EPISODES

Rearing episodes indicate the frequency at which the animal stands on hind limbs

or leans against the walls of the open field apparatus with the forelimb paws

(Davies et al., 2012). It is considered one of the measures of anxiety in rodents

http://etd.uwc.ac.za/

53

and has been reported to reveal exploratory behaviour triggered by new stimuli

(Van Abeelen, 1977, Alves et al., 2012). Findings show that BPA treatment led to

a significant reduction (p=0.0005, Figure 4.2) in rearing episodes when compared

to the control, however, in the RB group of rats, there was a significant increase

(p<0.0001) in rearing episodes when compared to BPA group of rats. Also, there

was no significant difference (p>0.05) between the RT group of rats and the NS

control group.

NS

BP

A

RB

RT

0

1 0

2 0

3 0

4 0

Re

ari

ng

ep

iso

de

s

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=6). P<0.001 vs NS; фP<0.0001 vs BPA

Figure 4.2: Number of rearing episodes of experimental rats on PND 42

4.3.1.2. TOTAL FREEZING TIME

Freezing (complete absence of body movements) is commonly used as an

indicator of anxiety and high-stress conditions (Walsh and Cummins, 1976, Díaz-

Morán et al., 2014). Total freezing time (seconds) was determined by scoring with

a stopwatch and reviewing video recordings of each rat during the test period.

Findings show that BPA treatment led to a significant increase (p<0.0001, Figure

http://etd.uwc.ac.za/

54

4.3) in total freezing time when compared to control. Conversely, rooibos

treatment significantly reduced (p=0.0343) the total freezing time in the RB group

of rats when compared to the BPA group of rats and there was significant increase

(p>0.0044) in total freezing time in the RT group of rats when compared to the

NS control group.

NS

BP

A

RB

RT

0

1 0

2 0

3 0

4 0

Imm

ob

ilit

y d

ura

tio

n (

se

c)

*

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=6). P<0.001 vs NS; фP<0.05 vs BPA; *P<0.005 vs

NS

Figure 4.3: Total freezing time of experimental rats on PND 42

4.3.1.3. TOTAL SELF-GROOMING TIME

Previous reports have demonstrated an increase in the total self-grooming time in

rodent models of anxiety and stress-exposed rats or mice (Kalueff and Tuohimaa,

2005, Kalueff et al., 2007). In this study, the cumulative duration that a rat spent

licking or scratching itself while remaining stationary was used as a measure of

self-grooming. Whereas BPA treatment led to a significant increase (p<0.0001,

http://etd.uwc.ac.za/

55

Figure 4.4) in total self-grooming time when compared to the control, rooibos

treatment significantly reduced (p<0.0001) the total self-grooming time as seen in

the RB group when compared to the BPA group. Also, there was no significant

difference (p>0.05) in total freezing time in the RT group of rats when compared

to the NS control group.

NS

BP

A

RB

RT

0

1 0

2 0

3 0

4 0

5 0

Gro

om

ing

tim

e (

se

c)

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=6). P<0.001 vs NS; фP<0.05 vs BPA.

Figure 4.4: Total self-grooming time of experimental rats on PND 42

4.3.2. NOVEL OBJECT RECOGNITION (NOR) TEST

The NOR test was used in this study to determine the ability of a rat to recognize

and spend longer time exploring a novel object introduced into its environment. In

this study, rats were exposed to both novel (N) and familiar (old, F) objects for 3

minutes, and the Recognition Index (RI) was calculated from the formula below:

http://etd.uwc.ac.za/

56

𝑅𝐼 = 𝑇𝑁

(𝑇𝑁 + 𝑇𝐹)… … … … … . 𝐹𝑜𝑟𝑚𝑢𝑙𝑎 1,

Where RI = Recognition Index, TN = Time spent with a novel object and TF = time spent

with a familiar object

Findings show that BPA treatment led to a reduction (p=0.6289, Figure 4.5) in RI

when compared to the control, however, in the RB group of rats, there was a

significant increase (p<0.0269) in RI when compared to the BPA group of rats.

Also, there was no significant difference (p>0.05) between the RT group of rats

and the NS control group.

NS

BP

A

RB

RT

0

5

1 0

1 5

2 0

2 5

Re

co

gn

itio

n I

nd

ex

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=6). фP<0.05 vs BPA.

Figure 4.5: Recognition index of experimental rats on PND 42

http://etd.uwc.ac.za/

57

4.4. EFFECT OF BPA AND FERMENTED ROOIBOS HERBAL TEA ON THE

ACTIVITY OF ANTIOXIDANT ENZYMES

4.4.1. SUPEROXIDE DISMUTASE

Reactive oxygen species (ROS) are generally regarded as cytotoxic agents that

induce oxidative damage by attacking cell membranes and DNA (Ozaydın et al.,

2018). The production of ROS may be triggered by inflammation, radiation,

drugs, aging and such chemical substances as BPA. However, antioxidant

enzymes such as SOD and CAT are known to inhibit the damage caused by ROS.

An increase in ROS generation or a reduction in cellular antioxidant levels results

in a condition known as oxidative stress (Hassan et al., 2012, Abdel-Wahab,

2014). BPA has been reported to cause damage in many organs such as the liver,

kidney and brain by lowering antioxidant activity, thus increasing oxidative stress

(Song et al., 2014, Moghaddam et al., 2015).

Superoxide dismutases (SOD) are enzymes that counteract oxidative damage by

catalyzing the dismutation of superoxide radical (O2-) into either molecular

oxygen O2 or H2O2 which are less toxic molecules (Hollman and Katan, 1999).

Findings from this study show that BPA treatment resulted in a significant

decrease (p<0.005, Figure 4.6-4.7) in cerebral and cerebellar SOD activity;

however, in the RB group of rats, there was a significant increase (p<0.05) in

SOD activity when compared to BPA group of rats.

There was no significant difference (p<0.05) in SOD activity in the RT group of

rats when compared to the NS control group.

http://etd.uwc.ac.za/

58

T re a tm e n t

SO

D a

cti

vit

y (

U/m

g p

ro

tein

)

NS

BP

AR

BR

T

0 .0 0

0 .0 5

0 .1 0

0 .1 5

0 .2 0

0 .2 5

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.005 vs NS; фP<0.005 vs BPA.

Figure 4.6: Effects of different treatments on SOD enzyme activity in the cerebral

homogenate of experimental rats on PND 42

T re a tm e n t

SO

D a

cti

vit

y (

U/m

g p

ro

tein

)

NS

BP

AR

BR

T

0 .0 0

0 .0 5

0 .1 0

0 .1 5

0 .2 0

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.05 vs NS; фP<0.05 vs BPA.

Figure 4.7: Effects of different treatments on SOD enzyme activity in the cerebellar

homogenate of experimental rats on PND 42

http://etd.uwc.ac.za/

59

4.4.2. CATALASE

CAT is involved in the detoxification of H2O2 through the conversion of two

molecules of H2O2 to two molecules of H2O and one molecule of O2 in tissues

(Nagata et al., 1999, Agati et al., 2012). The rate of H2O2 disintegration is directly

proportional to catalase concentration hence, higher CAT concentrations indicate

highly potent antioxidant effects and vice versa. Following treatment of rats with

BPA, there was a significant reduction in the cerebral and cerebellar catalase

activity (p<0.05, Figure 4.8-4.9). In the RB group, there was a significant increase

(p<0.05) in CAT activity when compared to BPA group. Also, there was no

significant difference (p<0.05) in CAT activity in the RT group of rats when

compared to the NS control group.

T re a tm e n t

CA

T a

cti

vit

y (

U/m

l)

NS

BP

AR

BR

T

0

2

4

6

8

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.05 vs NS; фP<0.05 vs BPA.

Figure 4.8: Effects of different treatments on CAT enzyme activity in the cerebral

homogenate of experimental rats on PND 42

http://etd.uwc.ac.za/

60

T re a tm e n t

CA

T a

cti

vit

y (

U/m

l)

NS

BP

AR

BR

T

0

2

4

6

8

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.05 vs NS; фP<0.05 vs BPA.

Figure 4.9: Effects of different treatments on CAT enzyme activity in the cerebellar

homogenate of experimental rats on PND 42

4.5. HISTOLOGICAL STUDIES

In this study, coronal sections of the cerebrum were obtained and stained to study

the potential neuroprotective effects of fermented rooibos herbal tea on the

development of CNS structures in rats exposed to BPA during pregnancy and

lactation. Coronal sections were cut and stained with H&E and Cresyl violet (CV)

for cell counting and histological analysis of the shapes and sizes of cell bodies in

the (CA1) region of the hippocampus while GFAP immunostaining was used for

analysis of damage responses in astrocytes. Similar analysis was done for the

Purkinje cells and the glial cells in the cerebellum.

http://etd.uwc.ac.za/

61

4.5.1. LENGTH OF HIPPOCAMPAL CA1 REGION IN EXPERIMENTAL

RATS

The H & E staining revealed very clear outlines of the pyramidal cell layer of the

hippocampal region of interest - the CA1 region. The length of this region was

determined by measurement along the dotted red landmark using an appropriate

Image-j tool as shown in Figure 4.10. In order to ensure that the sampling size

was sufficient for this analysis, specimens from six rats were assigned to each

group and three (3) separate staggered serial sections were used per animal, giving

a total of 18 sections per group. The singular CA1 region of each slide was

measured and average values were computed and plotted for each group using the

Graph Pad Prism software version 6.

CA - cornua ammonis (subregions - CA1, CA2 and CA3); DG – Dentate gyrus.

Figure 4.10: Diagram of the hippocampus showing the landmarks used for determining

the hippocampal length in experimental rats.

Findings show that on PND 42, there was a significant reduction (p<0.0001,

Figure 4.11) in the hippocampal CA1 length of rats treated with BPA when

http://etd.uwc.ac.za/

62

compared to the rats in the control group. In the RB group of rats, there was a

significant increase (p=0.0082) in the CA1 length when compared to the BPA

group. Findings also show that there was a significant reduction (p=0.0004) in the

RT group of rats when compared to the control rats.

T re a tm e n t

CA

1 l

en

gth

(

m)

NS

BP

AR

BR

T

0

2 0 0 0

4 0 0 0

6 0 0 0

*

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.0001 vs NS; фP<0.01 vs BPA; *P<0.0005

vs NS

Figure 4.11: CA1 hippocampal length in experimental rats on PND42

4.5.2. NUMBER OF PURKINJE CELLS IN THE PURKINJE CELL LAYER

(PCL) IN EXPERIMENTAL RATS

A loss or significant reduction in the Purkinje cells in the cerebellum has been

linked to motor disorders (Darmanto et al., 2000, Maloku et al., 2010, Celik et al.,

2018) and in the current study, the number of Purkinje cells significantly

decreased (p=0.0002, Figure 4.12) following treatment with BPA. However, in

rats treated with rooibos tea and BPA (RB), Purkinje cell numbers significantly

http://etd.uwc.ac.za/

63

increased (p<0.0001) when compared to rats treated with BPA only. In the RT

group, there was no significant difference (p=0.9849) in the number of Purkinje

cells when compared to rats in the NS control group.

T re a tm e n t

Nu

mb

er o

f P

urk

inje

ce

lls

NS

BP

AR

BR

T

0

2 0

4 0

6 0

8 0

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=4). P<0.0005 vs NS; фP<0.0001 vs BPA.

Figure 4.12: Number of Purkinje cells in experimental rats on PND42

http://etd.uwc.ac.za/

64

4.6. IMMUNOHISTOCHEMISTRY (IHC)

4.6.1. GLIAL FIBRILLARY ACIDIC PROTEIN (GFAP)

GFAP is an essential protein of intermediate glial filaments in astrocytes used for

the assessment of damage responses in astrocytes (Cikriklar et al., 2016). An

increase in the expression of GFAP is a key feature in astrocytic and CNS damage

(Hausmann et al., 2000, Yu et al., 2004). A total of three rats were assigned per

group, and tissue sections were processed for immunostaining to investigate

GFAP. The immunoreactive astrocytes for GFAP were determined in six counting

boxes (440 × 330 µm) per slide along the CA1 region, which surrounded the

polymorphic and molecular layers adjoining the pyramidal layer. The average

values were computed, plotted and compared between the different treatment

groups using the Graph Pad Prism software version 6.

Results showed that there were significantly more (p<0.0001, Figure 4.13) GFAP-

 immunoreactive astrocytes in the BPA group of rats when compared to control

while the rooibos (RB) group of rats had significantly fewer (p<0.0001) GFAP-

 immunoreactive astrocytes when compared to the BPA only -treated group. Also,

there was no significant difference (p>0.05) between the RT group of rats and the

rats in the NS control group.

http://etd.uwc.ac.za/

65

T re a tm e n t

No

. o

f G

FA

P p

os

itiv

e c

ell

s

NS

BP

AR

BR

T

0

5 0

1 0 0

1 5 0

2 0 0

2 5 0

NS: Normal saline, BPA: Bisphenol A, RB: Rooibos tea + Bisphenol A, RT: Rooibos tea.

Bars represent the mean + SEM (n=3). P<0.0001 vs NS; фP<0.0001 vs BPA.

Figure 4.13: Number of GFAP positive cells in experimental rats on PND42

http://etd.uwc.ac.za/

66

CHAPTER 5

DISCUSSION

5.1. GENERAL INTRODUCTION

Endocrine-disrupting chemicals (EDCs) are a wide range of compounds that

disrupt normal growth and development by halting or modifying the action of

hormones upon ingestion or absorption into the body (Schug et al., 2015).

Examples of EDCs include a broad group of natural and synthetic chemical

substances, such as polycyclic aromatic hydrocarbons (PAHs), polychlorinated

biphenyls (PCBs), pesticides, herbicides, fungicides, BPA and

dichlorodiphenyltrichloroethane (DDT). EDCs are abundant in our environs, diet,

and consumer products, which makes exposure to these toxins very frequent and

regular (Waring and Harris, 2005). Exposure to EDCs during initial

developmental stages is known to interrupt normal developmental processes,

thereby altering the body’s endocrine system to initiate deleterious effects in

various systems in the human body (Schug et al., 2015). For instance, EDCs

initiates neurotoxic effects that disturb the synthesis, transport, and release of such

neurotransmitters as glutamate, dopamine, serotonin, norepinephrine, which play

important roles in regulating behaviour, cognition, learning, and memory

(Dickerson and Gore, 2007, Rasier et al., 2007).

This study was designed to investigate if continuous consumption of fermented

rooibos herbal tea (FRHT) could offer diverse protection against potential damage

http://etd.uwc.ac.za/

67

to the developing CNS of offspring Wistar rats exposed to BPA, following ad

libitum maternal consumption during and after pregnancy as observed on PND 42.

The offspring were considered to have ‘formed’ and ‘developed’ in the presence

of ad libitum rooibos tea consumption including postnatal consumption through

breastfeeding. After the evaluation of neurobehavioural activity, rats were

sacrificed, and cerebral as well as cerebellar samples were examined for changes

in antioxidant enzyme activities and histomorphology. The results obtained from

this study are discussed in the following sections.

5.2. ROOIBOS TEA PREVENTED BPA-INDUCED GAIN IN BODY MASS

In this study, findings revealed an increase in the body mass of experimental rats

treated with BPA only. This is in line with previous reports demonstrating that

perinatal BPA exposure increases body mass when compared to controls (Rubin

et al., 2001, Miyawaki et al., 2007, Ezz et al., 2015). There are confirmed reports

which link BPA to weight gain and obesity (Newbold et al., 2009, Rubin and

Soto, 2009, Shankar et al., 2012). For instance, Li and colleagues reported that

high BPA exposure might promote childhood obesity and might advance global

obesity epidemic in the human population (Li et al., 2013a). In the same regard,

Harley and co-workers revealed that elevated urinary BPA concentrations in

children aged nine were linked to higher odds of obesity, increased waist

circumference, BMI z-score and percentage body fat (Harley et al., 2013). An

appraisal of available literature reveals other reports of body mass gain in

offspring of rodent mothers exposed to BPA during gestation and/or lactation, or

during the early postnatal period (Howdeshell et al., 1999). Also, postnatal

exposure of BPA in male rat pups to investigate the effects of BPA on anxiety

http://etd.uwc.ac.za/

68

revealed that exposed males substantially weighed more than the controls

(Patisaul and Bateman, 2008). Consequently, the increase in body mass gain in

this study highlights the ability of BPA to promote the progression of obesity

which could in turn further aggravate several metabolic, cardiovascular and

neurological disorders associated with BPA exposure.

Several mechanisms have been proposed to explain the association between BPA

exposure and body mass gain including the ability of BPA to enhance

differentiation of adipocytes and promote lipid accumulation in target cells

(Masuno et al., 2002, Wada et al., 2007), as well as increased gene expression of

adipogenic transcription factors (Phrakonkham et al., 2008). Others include the

ability of BPA to inhibit the release of adiponectin from human adipose tissue

explants (Hugo et al., 2008), dysregulate neuronal appetite controlling circuits

through its action on the hypothalamus (Wade and Schneider, 1992, Simerly,

2008), and inhibit the signalling of PPARgamma, which is essential for lipid

homeostasis and differentiation of brown and white adipocytes (Grun and

Blumberg, 2006, Grün and Blumberg, 2007). Findings from this study confirm

that although perinatal BPA exposure is a major challenge with respect to weight

gain and possibly childhood obesity, these effects could be attenuated by rooibos

through its protective activity against BPA.

5.3. ROOIBOS TEA MITIGATED BPA-INDUCED NEUROBEHAVIOURAL

DEFICITS

In this study, the open field test (OFT) was employed to investigate certain

neurobehavioral tests such as the frequency of rearing episodes, total freezing

http://etd.uwc.ac.za/

69

time, total self-grooming time and the novel object recognition (NOR) test. The

OFT, initially developed in 1934, is a routine test that evaluates emotionality,

exploratory behaviour and general activity in rodents (Hall, 1934). It is one of the

most commonly used measures of behaviour in animal psychology because it

offers a prompt and easy evaluation of precise behaviours that require little or no

training to the test subject and the person administering the test (Walsh and

Cummins, 1976, Prut and Belzung, 2003). Also, its popularity stems from the

psychological and physiological conceptions underlying the tests, which are

largely uncomplicated and easy to understand (Seibenhener and Wooten, 2015).

Although the primary outcome of interest is “movement”, it is sometimes

influenced by other factors such as motor output, exploratory drive, freezing, fear-

related behaviour and sickness (Gould et al., 2009). The OFT is also employed to

assess the toxic effects of compounds in rodents and thus evaluates different

facets of behaviour other than movement/locomotion (Gould et al., 2009).

The frequency of rearing episodes is regarded as a measure of exploratory activity

in the OFT and indicates the number of times rats stood on both hind paws in a

vertically upright position (Erdoğan et al., 2004, Ennaceur, 2014). Reports show

that a reduction in rearing episodes is indicative of increased anxiety (Costall et

al., 1989) and is further demonstrated under experimental conditions, such as

stress (Masood et al., 2003, Fan et al., 2011), drug treatment (van Lier et al.,

2004) and brain stimulation (Racine, 1972, Hannesson et al., 2001). In this study,

the reduction in the frequency of rearing episodes in the group of rats exposed to

BPA only could be indicative of increased anxiety and stress as demonstrated in

previous studies. However, following exposure to rooibos, it was observed that

http://etd.uwc.ac.za/

70

experimental rats exhibited an increase in rearing episodes, thus indicating that

rooibos was protective against BPA-induced reduction in the frequency of rearing,

possibly indicative of induced anxiety or stress.

Freezing activity, an important indicator of anxiety in rodents (Díaz-Morán et al.,

2014), is defined as the duration of the OFT within which the rodent was

stationary. It is conspicuous in conditions where stress levels are noticeably high

and occurs in response to apparent danger, environmental displacement, sudden

changes and fright stimuli (Walsh and Cummins, 1976, Brandao et al., 2008). In

line with earlier studies showing increased freezing activity in stress conditions

(Ranjbar et al., 2017) and in rats with impaired brain function (Saikhedkar et al.,

2014), total freezing time was significantly elevated in this study in the group of

rats exposed to BPA only. Conversely, we observed a significant decline in total

freezing time in the BPA-and-rooibos treated rats, thus confirming the protective

activity of rooibos against BPA in experimental rats.

Total self-grooming time is defined as the duration of time the experimental rats

expended in licking or scratching itself while motionless. It is a very important

behavioural activity in rodents and is involved in biologically essential processes,

such as thermoregulation, social communication and de-arousal (Spruijt et al.,

1988, Spruijt et al., 1992, Leonard et al., 2005). Previous reports indicate that self-

grooming is enhanced by exposure to stress (Jolles et al., 1979, Katz and Roth,

1979) and in rodent models of anxiety such as stress-induction in rats or mice

after exposure to anxiogenic drugs (Kalueff and Tuohimaa, 2005, Kalueff et al.,

2007).

http://etd.uwc.ac.za/

71

In this study, the increase in self-grooming observed in the BPA-only group could

be indicative of increased anxiety and stress. However, following exposure to

rooibos in the BPA-and-rooibos group, it was observed that the rats exhibited a

significant reduction in self-grooming, thus demonstrating that rooibos tea

consumption could have offered protection against BPA-induced anxiety-like

behaviour demonstrated as increase in self-grooming in this study.

The integrity of the medial temporal lobe is essential for memory processing and

this can be evaluated through the recognition index (RI), which is a measure of

the capability of a person or an animal to determine if a previously encountered

item is familiar or not (Squire et al., 2007). Tests that evaluate RI are becoming

gradually valuable tools for research designed to explore the neural basis of

memory (Winters et al., 2004). One such tests is the novel object recognition task

(NOR), studied for the first time by Ennaceur and Delacour to evaluate short-term

memory in rodents (Ennaceur and Delacour, 1988) which highlights the capacity

of the rodent to recall information for a short amount of time (Mathiasen and

DiCamillo, 2010). The NOR can be designed to evaluate working memory,

attention, anxiety, and preference for novelty in rodents (Silvers et al., 2007,

Goulart et al., 2010). It has also been utilized to investigate the effects of several

pharmacological treatments and brain impairment in rodents (Goulart et al.,

2010).

Findings from the present study showed that RI was significantly lowered in

BPA-only treated rats when compared to the NS controls. This is in line with

previous reports which show reduced RI in stress conditions (Nagata et al., 2009,

http://etd.uwc.ac.za/

72

Eagle et al., 2013). Conversely, when compared to rats exposed to BPA only, a

significant increase in RI was observed in the RB group thus suggesting a

neuroprotective role by rooibos tea against possible BPA-induced short-term

memory loss.

5.4. ROOIBOS TEA REGULATED THE ACTIVITY OF ANTIOXIDANT

ENZYMES

Reports show that exposure to BPA during pregnancy can result in a variety of

foetal malformations and growth retardations as well as early embryonic losses

(Bajaj et al., 1993, Friedler, 1996, Khattak et al., 1999), possibly via the

production of excessive ROS known to be toxic to cells (Nicol et al., 2000, Wells

et al., 2005). BPA-induced ROS production has been investigated in many cell

types and at different concentrations (Huc et al., 2012, Babu et al., 2013, Gassman

et al., 2015). For instance, nanomolar BPA concentration ranges were found to

result in short and transient elevations of ROS (Koong and Watson, 2015, Pfeifer

et al., 2015); nevertheless prolonged and increased levels of oxidative stress have

been reported (Huc et al., 2012, Porreca et al., 2016). BPA exposure in

micromolar concentration ranges have been reported to induce higher levels of

oxidative stress (Huc et al., 2012, Babu et al., 2013, Gassman et al., 2015, Leem et

al., 2017), and cytotoxicity via ROS-triggered DNA damage (Ooe et al., 2005,

Huc et al., 2012, Leem et al., 2017). Different in vivo studies have shown an

association between high urinary BPA levels and elevated concentrations of such

oxidative stress biomarkers as lipid peroxidation and malondialdehyde (Watkins

et al., 2015, Zhang et al., 2016), thus supporting the pro-oxidant activity of BPA.

http://etd.uwc.ac.za/

73

Cellular antioxidants mitigate the deleterious effects arising from interactions

between lipid, protein, DNA molecules and ROS (Hassan et al., 2012).

Irrespective of the existence of a potent antioxidant system, an imbalance in the

generation of ROS due to natural or synthetic chemicals may result in some

disorders. The concentration of free radical scavengers and the levels of such

antioxidant enzymes as SOD and CAT regulate the antioxidant capacity of cells;

thus numerous endpoints have been investigated following BPA exposure to

evaluate its effect on antioxidant capacity (Gassman, 2017).

The enzyme SOD is known to defend tissues from oxidative damage by

catalyzing the conversion of O2− to H2O2 (Fridovich, 1997), while catalase is

responsible for the detoxification of the H2O2 generated by SOD through the

conversion of two molecules of H2O2 to form two molecules of H2O and one

molecule of O2 (Nagata et al., 1999, Agati et al., 2012), thus providing protection

against ROS. Findings from the current study showed that whereas BPA induced

marked oxidative damage by inhibiting the activities of the antioxidant enzymes

SOD and CAT, the intake of rooibos tea could attenuate these effects of BPA.

These findings are in agreement with other studies that report a significant

decrease in SOD and CAT levels following BPA treatment (Eid et al., 2015,

Moghaddam et al., 2015, Ozaydın et al., 2018).

5.5. ROOIBOS TEA ATTENUATED NEURODEGENERATION IN RATS

EXPOSED TO BPA

The development of the brain involves a complex interplay between dendritic

growth and the production of neuronal connections to form a neural network

(McAllister et al., 1997, Wong and Ghosh, 2002). Different brain regions are

http://etd.uwc.ac.za/

74

tasked with different functions, and histological damage to these brain regions

hinders their function (Mcallister et al., 2004, Kimura et al., 2016). The

hippocampus is subdivided into two main parts which have been studied

comprehensively for spatial navigation, learning and memory functions; these are

the cornu ammonis (subfields: CA1, CA2, CA3, CA4) and the dentate gyrus

(Bannerman et al., 2014, Moser et al., 2014). Alterations in hippocampal

morphology are linked to changes in cognitive function (Van Praag et al., 2000)

and the hippocampal CA1 (H-CA1) neurons in rodents are reported to be essential

for the processing of hippocampus-dependent memory (Bartsch et al., 2011).

These neurons are also the most sensitive and most susceptible neurons to

selective degeneration in response to such EDCs as BPA, ischemia and other toxic

substances (Kirino, 1982, Pulsinelli et al., 1982, Elsworth et al., 2013, Kimura et

al., 2016).

Previous studies have shown that there was a reduction in the dendritic spines of

neurons in the H-CA1 region following exposure of mice to BPA (Eilam-Stock et

al., 2012, Kimura et al., 2016, Liu et al., 2016). The results obtained from the

present study are in agreement with the above findings as there was a significant

reduction in the average length of the H-CA1 region in rats exposed to BPA-only

when compared to the NS control group. However, a significant increase in the

average H-CA1 length was observed in rats treated with BPA-and-rooibos when

compared to BPA-only rats. These morphological findings indicate a protective

effect of rooibos in the CA1 region of the hippocampus in rats, thus attenuating

BPA-induced impairment of cognition in the rats.

http://etd.uwc.ac.za/

75

In certain human inherited disease conditions, such as ataxia telangiectasia,

paraneoplastic cerebellar degeneration and lysosomal storage diseases, a

significant loss of Purkinje cells has been reported (Gilman et al., 1981, Anderson

et al., 1987) as a noticeable component of the histopathological alterations

detected in cerebellar injury. Some of the clinical indicators of cerebellar

dysfunction frequently manifested in these conditions include ataxia and tremor

(Riedel et al., 1990). Purkinje cell loss is a significant observation in many

diseases but the exact cause is unknown.

In some in vivo studies, the significant reduction in Purkinje cell population is due

to the neurotoxic effect of EDCs on Purkinje cells (Kimura-Kuroda et al., 2007,

Ibhazehiebo et al., 2011) and the reduction in Purkinje cells observed in this study

is similar to findings from these previous studies, possibly indicating that the

deleterious effects of BPA on the cerebellum (Ahmed and Eid, 2015). The number

of Purkinje cells however significantly increased in rats treated with BPA-and-

rooibos tea when compared to the BPA-only rats, thus suggesting a

neuroprotective role for rooibos tea.

Apart from neuronal damage caused by most EDCs, it is plausible to also

investigate possible damage to the support cells of the CNS. Astrocytes (also

called astroglia) are the most abundant support cells in the CNS (Zuchero and

Barres, 2015) and contain GFAP which is an important protein of intermediate

glial filaments in astrocytes commonly used to investigate astrocytic damage

response (Cikriklar et al., 2016). In this study, there was an increase in the

expression of GFAP-positive astrocytes in BPA-exposed rats when compared to

http://etd.uwc.ac.za/

76

controls. Increased GFAP expression is prominent in conditions of CNS injury

and astrocyte damage (Hausmann et al., 2000, Yu et al., 2004, Cai et al., 2006)

previous studies have reported on the various mechanisms of astrocyte activation

possibly associated with BPA exposure. For instance, increased generation of

hydroxyl radicals has been reported in the adult mouse brain following BPA

exposure (Kabuto et al., 2003) and elevated concentrations of lipid peroxidation in

postnatal mouse brains was reported following BPA exposure during pregnancy

and lactation (Kabuto et al., 2004). Consequently, BPA-triggered oxidative stress

and peroxidation could be said to be responsible for the astroglial activation

reported in this study. However, rats exposed to rooibos tea had significantly

reduced GFAP levels when compared to BPA-only treated rats. Taken together,

findings from this study seem to indicate that rooibos tea is protective against the

deleterious effects of BPA in the developing brain.

http://etd.uwc.ac.za/

77

5.6. CONCLUSION

This study provides the first research evidence of the neuroprotective potential of

fermented rooibos herbal tea against BPA exposure on brain development in

Wistar rats. The protective activity observed is possibly due to the natural and

bioactive constituents of this herb, reported to be responsible for the regulation of

cellular antioxidants which inhibit BPA-induced oxidative damage. These

findings suggest that rooibos tea offers protection against BPA toxicity, and may

provide the basis for clinical trials and policy decisions aimed at regulating the

use of products containing such EDCs as BPA.

Limitations of this study include investigation of few immunohistochemical

markers in the brains of experimental rats, few neurobehavioural tests to confirm

BPA-induced impairment of cognitive function, and the evaluation of only few

oxidative stress markers to fully elucidate the impact of oxidative stress on BPA

exposure. These limitations are partly due to financial constraints and the need to

pitch the volume of research data at the MSc level.

Future recommendations

Isolation of the bioactive compounds in rooibos and investigation of their

neuroprotective activity against BPA.

Investigation of additional immunohistochemical, apoptotic, inflammatory and

oxidative stress biomarkers to corroborate findings from this study.

http://etd.uwc.ac.za/

78

Assessment of additional OFT activities and neurobehavioural tests (such as

T-maze test and water maze test) to offer more information on BPA-induced

impairment of locomotion, learning and memory.

http://etd.uwc.ac.za/

79

REFERENCES

Abdallah, M. a. M. 2016. Endocrine Disruptors as Pollutants in Marine Ecosystem: A

Case Study in Egypt. The Open Biotechnology Journal, 10.

Abdel-Wahab, W. M. 2014. Thymoquinone attenuates toxicity and oxidative stress

induced by bisphenol A in liver of male rats. Pak J Biol Sci, 17, 1152-1160.

Abhang, P. A., Gawali, B. W. & Mehrotra, S. C. 2016. Introduction to EEG-and Speech-

Based Emotion Recognition, Academic Press.

Addae, C., Cheng, H. & Martinez-Ceballos, E. 2013. Effect of the environmental

pollutant hexachlorobenzene (HCB) on the neuronal differentiation of mouse

embryonic stem cells. International journal of environmental research and public

health, 10, 5244-5256.

Agati, G., Azzarello, E., Pollastri, S. & Tattini, M. 2012. Flavonoids as antioxidants in

plants: location and functional significance. Plant Science, 196, 67-76.

Ahmed, M. a. a. S. & Eid, R. A. 2015. Effect of Bisphenol-A on the Post-Natal

Development and Structure of Rat Cerebellum. Int. J. Curr. Microbiol. App. Sci,

4, 14-35.

Akingbemi, B. T., Sottas, C. M., Koulova, A. I., Klinefelter, G. R. & Hardy, M. P. 2004.

Inhibition of testicular steroidogenesis by the xenoestrogen bisphenol A is

associated with reduced pituitary luteinizing hormone secretion and decreased

steroidogenic enzyme gene expression in rat Leydig cells. Endocrinology, 145,

592-603.

Akinrinmade, O., Omoruyi, S., Dietrich, D. & Ekpo, O. 2017. Long‑term consumption of

fermented rooibos herbal tea offers neuroprotection against ischemic brain injury

in rats. Acta Neurobiol Exp, 77, 94-105.

Alonso-Magdalena, P., Ropero, A. B., Soriano, S., Quesada, I. & Nadal, A. 2010.

Bisphenol-A: a new diabetogenic factor. Hormones (Athens), 9, 118-26.

Alves, R., De Carvalho, J. G. B. & Venditti, M. a. C. 2012. High-and low-rearing rats

differ in the brain excitability controlled by the allosteric benzodiazepine site in

the GABAA receptor. Journal of Behavioral and Brain Science, 2, 315.

Andersen, B. B., Korbo, L. & Pakkenberg, B. 1992. A quantitative study of the human

cerebellum with unbiased stereological techniques. Journal of Comparative

Neurology, 326, 549-560.

Anderson, N., Cunningham, J. & Posner, J. 1987. Autoimmune pathogenesis of

paraneoplastic neurological syndromes. Critical reviews in neurobiology, 3, 245-

299.

Angle, B. M., Do, R. P., Ponzi, D., Stahlhut, R. W., Drury, B. E., Nagel, S. C., Welshons,

W. V., Besch-Williford, C. L., Palanza, P. & Parmigiani, S. 2013. Metabolic

http://etd.uwc.ac.za/

80

disruption in male mice due to fetal exposure to low but not high doses of

bisphenol A (BPA): evidence for effects on body weight, food intake, adipocytes,

leptin, adiponectin, insulin and glucose regulation. Reproductive toxicology, 42,

256-268.

Apps, R. & Garwicz, M. 2005. Anatomical and physiological foundations of cerebellar

information processing. Nature Reviews Neuroscience, 6, 297.

Ardila, A., Bernal, B. & Rosselli, M. 2016. The role of Wernicke's area in language

comprehension. Psychology & Neuroscience, 9, 340.

Aschberger, K., Castello, P., Hoekstra, E., Karakitsios, S., Munn, S., Pakalin, S. &

Sarigiannis, D. 2010. Bisphenol A and baby bottles: challenges and perspectives.

Luxembourg: Publications Office of the European Union, 5-50.

Auddy, B., Ferreira, M., Blasina, F., Lafon, L., Arredondo, F., Dajas, F., Tripathi, P.,

Seal, T. & Mukherjee, B. 2003. Screening of antioxidant activity of three Indian

medicinal plants, traditionally used for the management of neurodegenerative

diseases. Journal of Ethnopharmacology, 84, 131-138.

Aversi-Ferreira, T. A., Araújo, M. F. P. D., Lopes, D. B. & Nishijo, H. 2010. History,

citoarchitecture and neurophysiology of human and non human primates' parietal

lobe: A review. Dementia & Neuropsychologia, 4, 173-180.

Babu, S., Uppu, S., Claville, M. O. & Uppu, R. M. 2013. Prooxidant actions of bisphenol

A (BPA) phenoxyl radicals: implications to BPA-related oxidative stress and

toxicity. Toxicology mechanisms and methods, 23, 273-280.

Bajaj, J., Misra, A., Rajalakshmi, M. & Madan, R. 1993. Environmental release of

chemicals and reproductive ecology. Environmental health perspectives, 101,

125-130.

Balabanič, D. & Klemenčič, A. K. 2018. Endocrine-disrupting chemicals and male

reproductive health: a review. Slovenian Medical Journal, 87.

Bannerman, D. M., Sprengel, R., Sanderson, D. J., Mchugh, S. B., Rawlins, J. N. P.,

Monyer, H. & Seeburg, P. H. 2014. Hippocampal synaptic plasticity, spatial

memory and anxiety. Nature reviews neuroscience, 15, 181.

Barsottini, O. G. P., Albuquerque, M. V. C. D., Braga Neto, P. & Pedroso, J. L. 2014.

Adult onset sporadic ataxias: a diagnostic challenge. Arquivos de neuro-

psiquiatria, 72, 232-240.

Bartsch, T., Döhring, J., Rohr, A., Jansen, O. & Deuschl, G. 2011. CA1 neurons in the

human hippocampus are critical for autobiographical memory, mental time travel,

and autonoetic consciousness. Proceedings of the National Academy of Sciences,

201110266.

Bayer, S. A. 1985. Hippocampal region. The rat nervous system, 1, 335-352.

http://etd.uwc.ac.za/

81

Berquin, P., Giedd, J., Jacobsen, L., Hamburger, S., Krain, A., Rapoport, J. &

Castellanos, F. 1998. Cerebellum in attention-deficit hyperactivity disorder A

morphometric MRI study. Neurology, 50, 1087-1093.

Betancourt, A. M., Eltoum, I. A., Desmond, R. A., Russo, J. & Lamartiniere, C. A. 2010.

In utero exposure to bisphenol A shifts the window of susceptibility for

mammary carcinogenesis in the rat. Environmental health perspectives, 118,

1614.

Bhattacharya, S. & Mukherjee, S. 1975. Toxic effects of endrin on hepatopancreas of the

teleost fish, Clarias batrachus (Linn.). Indian journal of experimental biology, 13,

185-186.

Biedermann-Brem, S., Grob, K. & Fjeldal, P. 2008. Release of bisphenol A from

polycarbonate baby bottles: mechanisms of formation and investigation of worst

case scenarios. European Food Research and Technology, 227, 1053-1060.

Bist, R. & Bhatt, D. K. 2009. The evaluation of effect of alpha-lipoic acid and vitamin E

on the lipid peroxidation, gamma-amino butyric acid and serotonin level in the

brain of mice (Mus musculus) acutely intoxicated with lindane. Journal of the

neurological sciences, 276, 99-102.

Blommaert, K. & Steenkamp, J. 1978. Tannien-en moontlike kafeieninhoud van

rooibostee, Aspalathus (subgen. nortieria) linearis (Burm. Fil.) R. Dahlgr.

Agroplantae.

Bosek, P. & Nakano, M. 2003. Hepatoprotective effect of rooibos tea (Aspalathus

linearis) on CCl 4-induced liver damage in rats. Physiological Research, 52, 461-

466.

Bramati, L., Minoggio, M., Gardana, C., Simonetti, P., Mauri, P. & Pietta, P. 2002.

Quantitative characterization of flavonoid compounds in rooibos tea (Aspalathus

linearis) by LC− UV/DAD. Journal of Agricultural and Food Chemistry, 50,

5513-5519.

Brandao, M. L., Zanoveli, J. M., Ruiz-Martinez, R. C., Oliveira, L. C. & Landeira-

Fernandez, J. 2008. Different patterns of freezing behavior organized in the

periaqueductal gray of rats: association with different types of anxiety.

Behavioural brain research, 188, 1-13.

Breiter, T., Laue, C., Kressel, G., Gröll, S., Engelhardt, U. H. & Hahn, A. 2011.

Bioavailability and antioxidant potential of rooibos flavonoids in humans

following the consumption of different rooibos formulations. Food chemistry,

128, 338-347.

Brown Jr, J. S. 2008. Effects of bisphenol-A and other endocrine disruptors compared

with abnormalities of schizophrenia: an endocrine-disruption theory of

schizophrenia. Schizophrenia bulletin, 35, 256-278.

Brück, K. 1983. Functions of the endocrine system. Human physiology. Springer.

http://etd.uwc.ac.za/

82

Caballero-Gallardo, K., Olivero-Verbel, J. & L Freeman, J. 2016. Toxicogenomics to

evaluate endocrine disrupting effects of environmental chemicals using the

zebrafish model. Current genomics, 17, 515-527.

Cai, Z., Lin, S., Fan, L.-W., Pang, Y. & Rhodes, P. 2006. Minocycline alleviates

hypoxic–ischemic injury to developing oligodendrocytes in the neonatal rat brain.

Neuroscience, 137, 425-435.

Calafat, A. M., Kuklenyik, Z., Reidy, J. A., Caudill, S. P., Ekong, J. & Needham, L. L.

2004. Urinary concentrations of bisphenol A and 4-nonylphenol in a human

reference population. Environmental health perspectives, 113, 391-395.

Campion, S., Catlin, N., Heger, N., Mcdonnell, E. V., Pacheco, S. E., Saffarini, C.,

Sandrof, M. A. & Boekelheide, K. 2012. Male reprotoxicity and endocrine

disruption. Molecular, Clinical and Environmental Toxicology. Springer.

Canda, B. D. 2012. Modulation of oxidative stress by rooibos (aspalathus linearis) herbal

tea, chinese green (camellia sinensis) tea and commercial tea supplements using

a rodent model. Cape Peninsula University of Technology.

Cardiff, R. D., Miller, C. H. & Munn, R. J. 2014. Manual hematoxylin and eosin staining

of mouse tissue sections. Cold Spring Harbor protocols, 2014, pdb. prot073411.

Celik, I., Seker, M. & Salbacak, A. 2018. Histological and histomorphometric studies on

the cerebellar cortex and silver stained nucleolus organizer regions of Purkinje

neurons in chronic morphine-treated rats. Veterinarski arhiv, 88, 75-88.

Charlton, H. 2008. Hypothalamic control of anterior pituitary function: a history. Journal

of neuroendocrinology, 20, 641-646.

Chayer, C. & Freedman, M. 2001. Frontal lobe functions. Current neurology and

neuroscience reports, 1, 547-552.

Cheney, R. H. & Scholtz, E. 1963. Rooibos tea, a South African contribution to world

beverages. Economic Botany, 17, 186-194.

Cikriklar, H. I., Uysal, O., Ekici, M. A., Ozbek, Z., Cosan, D. T., Yucel, M., Yurumez, Y.

& Baydemir, C. 2016. Effectiveness of GFAP in Determining Neuronal Damage

in Rats with Induced Head Trauma. Turkish neurosurgery, 26.

Cleghorn, R. 1955. The hypothalamic-endocrine system. Psychosomatic medicine, 17,

367-376.

Clotfelter, E. D., Bell, A. M., & Levering, K. R. (2004). The role of animal behaviour in

the study of endocrine-disrupting chemicals. Animal behaviour, 68(4), 665-676.

Coble, Y., Hildebrandt, P., Davis, J., Raasch, F. & Curley, A. 1967. Acute endrin

poisoning. JAMA, 202, 489-493.

http://etd.uwc.ac.za/

83

Colborn, T., Vom Saal, F. S. & Soto, A. M. 1993. Developmental effects of endocrine-

disrupting chemicals in wildlife and humans. Environmental health perspectives,

101, 378.

Colerangle, J. B. & Roy, D. 1997. Profound effects of the weak environmental estrogen-

like chemical bisphenol A on the growth of the mammary gland of Noble rats.

The Journal of steroid biochemistry and molecular biology, 60, 153-160.

Costall, B., Jones, B., Kelly, M., Naylor, R. J. & Tomkins, D. 1989. Exploration of mice

in a black and white test box: validation as a model of anxiety. Pharmacology

Biochemistry and Behavior, 32, 777-785.

Crain, D. A., Janssen, S. J., Edwards, T. M., Heindel, J., Ho, S.-M., Hunt, P., Iguchi, T.,

Juul, A., Mclachlan, J. A. & Schwartz, J. 2008. Female reproductive disorders:

the roles of endocrine-disrupting compounds and developmental timing. Fertility

and sterility, 90, 911-940.

Darmanto, W., Inouye, M., Takagishi, Y., Ogawa, M., Mikoshiba, K. & Murata, Y. 2000.

Derangement of Purkinje cells in the rat cerebellum following prenatal exposure

to X-irradiation: decreased Reelin level is a possible cause. Journal of

Neuropathology & Experimental Neurology, 59, 251-262.

Davies, K. G., Ekpenyong, C., Nwangwa, J. & Antai, A. B. 2012. Locomotor and

Exploratory Behaviour in Mice with Treated Oral Artemether Suspension.

Research in Neuroscience, 1, 17-24.

De Coensel, N., David, F. & Sandra, P. 2009. Study on the migration of bisphenol‐A

from baby bottles by stir bar sorptive extraction‐thermal desorption‐capillary GC‐

MS. Journal of separation science, 32, 3829-3836.

De Jong, G., Swaen, G. & Slangen, J. 1997. Mortality of workers exposed to dieldrin and

aldrin: a retrospective cohort study. Occupational and environmental medicine,

54, 702-707.

Diamanti-Kandarakis, E., Bourguignon, J.-P., Giudice, L. C., Hauser, R., Prins, G. S.,

Soto, A. M., Zoeller, R. T. & Gore, A. C. 2009. Endocrine-disrupting chemicals:

an Endocrine Society scientific statement. Endocrine reviews, 30, 293-342.

Díaz-Morán, S., Estanislau, C., Cañete, T., Blázquez, G., Ráez, A., Tobeña, A. &

Fernández-Teruel, A. 2014. Relationships of open-field behaviour with anxiety in

the elevated zero-maze test: focus on freezing and grooming. World Journal of

Neuroscience, 4, 1.

Dickerson, S. M. & Gore, A. C. 2007. Estrogenic environmental endocrine-disrupting

chemical effects on reproductive neuroendocrine function and dysfunction across

the life cycle. Reviews in Endocrine and Metabolic Disorders, 8, 143-159.

Doya, K. 1999. What are the computations of the cerebellum, the basal ganglia and the

cerebral cortex? Neural networks, 12, 961-974.

http://etd.uwc.ac.za/

84

Duvernoy, H.M., 2005. The human hippocampus: functional anatomy, vascularization

and serial sections with MRI. Springer Science & Business Media.

Duvernoy, H.M., 2013. The human hippocampus: an atlas of applied anatomy. JF

Bergmann-Verlag.

Eagle, A. L., Fitzpatrick, C. J. & Perrine, S. A. 2013. Single prolonged stress impairs

social and object novelty recognition in rats. Behavioural brain research, 256,

591-597.

Eid, J. I., Eissa, S. M. & El-Ghor, A. A. 2015. Bisphenol A induces oxidative stress and

DNA damage in hepatic tissue of female rat offspring. The Journal of Basic &

Applied Zoology, 71, 10-19.

Eilam-Stock, T., Serrano, P., Frankfurt, M. & Luine, V. 2012. Bisphenol-A impairs

memory and reduces dendritic spine density in adult male rats. Behavioral

neuroscience, 126, 175.

Eladak, S., Grisin, T., Moison, D., Guerquin, M.-J., N'tumba-Byn, T., Pozzi-Gaudin, S.,

Benachi, A., Livera, G., Rouiller-Fabre, V. & Habert, R. 2015. A new chapter in

the bisphenol A story: bisphenol S and bisphenol F are not safe alternatives to

this compound. Fertility and sterility, 103, 11-21.

Elsworth, J. D., Jentsch, J. D., Vandevoort, C. A., Roth, R. H., Redmond Jr, D. E. &

Leranth, C. 2013. Prenatal exposure to bisphenol A impacts midbrain dopamine

neurons and hippocampal spine synapses in non-human primates.

Neurotoxicology, 35, 113-120.

Ennaceur, A. 2010. One-trial object recognition in rats and mice: methodological and

theoretical issues. Behavioural brain research, 215, 244-254.

Ennaceur, A. 2014. Tests of unconditioned anxiety—pitfalls and disappointments.

Physiology & behavior, 135, 55-71.

Ennaceur, A. & Delacour, J. 1988. A new one-trial test for neurobiological studies of

memory in rats. 1: Behavioral data. Behavioural brain research, 31, 47-59.

Erdoğan, F., Gölgeli, A., Arman, F. & Ersoy, A. Ö. 2004. The effects of

pentylenetetrazole-induced status epilepticus on behavior, emotional memory,

and learning in rats. Epilepsy & Behavior, 5, 388-393.

Erickson, L. 2003. Rooibos tea: research into antioxidant and antimutagenic properties.

HerbalGram, 59, 34-45.

Ezz, H. S. A., Khadrawy, Y. A. & Mourad, I. M. 2015. The effect of bisphenol A on

some oxidative stress parameters and acetylcholinesterase activity in the heart of

male albino rats. Cytotechnology, 67, 145-155.

Fadiga, L., Craighero, L. & D’ausilio, A. 2009. Broca's area in language, action, and

music. Annals of the New York Academy of Sciences, 1169, 448-458.

http://etd.uwc.ac.za/

85

Fan, S. J., Jiang, H., Yang, L.-J., Liu, X., Song, J. & Pan, F. 2011. Effects of adrenergic

agents on stress-induced brain microstructural and immunochemical changes in

adult male Wistar rats. Annals of Anatomy-Anatomischer Anzeiger, 193, 418-424.

Fendick, E., Mather-Mihaich, E., Houck, K., Clair, M. S., Faust, J., Rockwell, C. &

Owens, M. 1990. Ecological toxicology and human health effects of heptachlor.

Reviews of environmental contamination and toxicology. Springer.

Fine, E. J., Ionita, C. C. & Lohr, L. 2002. The history of the development of the cerebellar

examination. Seminars in neurology, 22, 375-384.

Fonnum, F. & Lock, E. 2000. Cerebellum as a target for toxic substances. Toxicology

letters, 112, 9-16.

Fraga, C. G., Galleano, M., Verstraeten, S. V. & Oteiza, P. I. 2010. Basic biochemical

mechanisms behind the health benefits of polyphenols. Molecular aspects of

medicine, 31, 435-445.

Fridovich, I. 1997. Superoxide anion radical (O· 2), superoxide dismutases, and related

matters. Journal of Biological Chemistry, 272, 18515-18517.

Friedler, G. 1996. Paternal exposures: impact on reproductive and developmental

outcome. An overview. Pharmacology Biochemistry and Behavior, 55, 691-700.

Fry, D. M. 1995. Reproductive effects in birds exposed to pesticides and industrial

chemicals. Environmental Health Perspectives, 103, 165.

Frye, C., Bo, E., Calamandrei, G., Calza, L., Dessì‐Fulgheri, F., Fernández, M., Fusani,

L., Kah, O., Kajta, M. & Le Page, Y. 2012. Endocrine disrupters: a review of

some sources, effects, and mechanisms of actions on behaviour and

neuroendocrine systems. Journal of neuroendocrinology, 24, 144-159.

Gao, H., Yang, B.-J., Li, N., Feng, L.-M., Shi, X.-Y., Zhao, W.-H. & Liu, S.-J. 2015.

Bisphenol A and hormone-associated cancers: current progress and perspectives.

Medicine, 94.

Garbarino, V. R., Orr, M. E., Rodriguez, K. A. & Buffenstein, R. 2015. Mechanisms of

oxidative stress resistance in the brain: lessons learned from hypoxia tolerant

extremophilic vertebrates. Archives of biochemistry and biophysics, 576, 8-16.

Gassman, N. R. 2017. Induction of oxidative stress by bisphenol A and its pleiotropic

effects. Environmental and molecular mutagenesis, 58, 60-71.

Gassman, N. R., Coskun, E., Stefanick, D. F., Horton, J. K., Jaruga, P., Dizdaroglu, M. &

Wilson, S. H. 2015. Bisphenol a promotes cell survival following oxidative DNA

damage in mouse fibroblasts. PloS one, 10, e0118819.

Geens, T., Aerts, D., Berthot, C., Bourguignon, J.-P., Goeyens, L., Lecomte, P.,

Maghuin-Rogister, G., Pironnet, A.-M., Pussemier, L. & Scippo, M.-L. 2012. A

review of dietary and non-dietary exposure to bisphenol-A. Food and chemical

toxicology, 50, 3725-3740.

http://etd.uwc.ac.za/

86

Geens, T., Goeyens, L. & Covaci, A. 2011. Are potential sources for human exposure to

bisphenol-A overlooked? International journal of hygiene and environmental

health, 214, 339-347.

Gies, A. & Soto, A. M. 2013. 10 Bisphenol A: contested science, divergent safety

evaluations. Late lessons from early warnings: science, precaution, innovation,

20.

Gilman, S., Bloedel, J. & Lechtenberg, R. 1981. Disorders of the Cerebellum:

Contemporary Neurology Series. Philadelphia, Davis.

Goulart, B., De Lima, M., De Farias, C., Reolon, G., Almeida, V., Quevedo, J.,

Kapczinski, F., Schröder, N. & Roesler, R. 2010. Ketamine impairs recognition

memory consolidation and prevents learning-induced increase in hippocampal

brain-derived neurotrophic factor levels. Neuroscience, 167, 969-973.

Gould, T. D., Dao, D. T. & Kovacsics, C. E. 2009. The open field test. Mood and anxiety

related phenotypes in mice. Springer.

Goyen, T. A., Lui, K. & Woods, R. 1998. Visual‐motor, visual‐perceptual, and fine motor

outcomes in very‐low‐birthweight children at 5 years. Developmental Medicine

& Child Neurology, 40, 76-81.

Grumetto, L., Montesano, D., Seccia, S., Albrizio, S. & Barbato, F. 2008. Determination

of bisphenol A and bisphenol B residues in canned peeled tomatoes by reversed-

phase liquid chromatography. Journal of agricultural and food chemistry, 56,

10633-10637.

Grün, F. & Blumberg, B. 2007. Perturbed nuclear receptor signaling by environmental

obesogens as emerging factors in the obesity crisis. Reviews in Endocrine and

Metabolic Disorders, 8, 161-171.

GruN, F. & Blumberg, B. 2006. Environmental obesogens: organotins and endocrine

disruption via nuclear receptor signaling. Endocrinology, 147, s50-s55.

Gupta, S., Guha, P., Majumder, S., Pal, P., Sen, K., Chowdhury, P., Chakraborty, A.,

Panigrahi, A. K. & Mukherjee, D. 2018. Effects of bisphenol A (BPA) on brain-

specific expression of cyp19a1b gene in swim-up fry of Labeo rohita.

Comparative Biochemistry and Physiology Part C: Toxicology & Pharmacology,

209, 63-71.

Hall, C. S. 1934. Emotional behavior in the rat. I. Defecation and urination as measures of

individual differences in emotionality. Journal of Comparative psychology, 18,

385.

Hannesson, D. K., Howland, J., Pollock, M., Mohapel, P., Wallace, A. E. & Corcoran, M.

E. 2001. Dorsal hippocampal kindling produces a selective and enduring

disruption of hippocampally mediated behavior. Journal of Neuroscience, 21,

4443-4450.

http://etd.uwc.ac.za/

87

Harley, K. G., Schall, R. A., Chevrier, J., Tyler, K., Aguirre, H., Bradman, A., Holland,

N. T., Lustig, R. H., Calafat, A. M. & Eskenazi, B. 2013. Prenatal and postnatal

bisphenol A exposure and body mass index in childhood in the CHAMACOS

cohort. Environmental health perspectives, 121, 514.

Hassan, Z. K., Elobeid, M. A., Virk, P., Omer, S. A., Elamin, M., Daghestani, M. H. &

Alolayan, E. M. 2012. Bisphenol A induces hepatotoxicity through oxidative

stress in rat model. Oxidative Medicine and Cellular Longevity, 2012.

Hausmann, R., Riess, R., Fieguth, A. & Betz, P. 2000. Immunohistochemical

investigations on the course of astroglial GFAP expression following human

brain injury. International journal of legal medicine, 113, 70-75.

Henretig, F. M. 2009. Toxins. Developmental-Behavioral Pediatrics (Fourth Edition).

Elsevier.

Herculano-Houzel, S. 2009. The human brain in numbers: a linearly scaled-up primate

brain. Frontiers in human neuroscience, 3, 31.

Hilgetag, C. C. & Barbas, H. 2009. Sculpting the brain. Scientific American, 300, 66-71.

Hiller-Sturmhöfel, S. & Bartke, A. 1998. The Endocrine System. Alcohol Health Res.

World, 22, 153-164.

Hollman, P. C. H. & Katan, M. B. 1999. Dietary flavonoids: intake, health effects and

bioavailability. Food and Chemical Toxicology, 37, 937-942.

Hong, I.-S., Lee, H.-Y. & Kim, H.-P. 2014. Anti-oxidative effects of Rooibos tea

(Aspalathus linearis) on immobilization-induced oxidative stress in rat brain.

PloS one, 9, e87061.

Hooker, E. P., Fulcher, K. G. & Gibb, H. J. 2014. Aldrin and Dieldrin: A Reevaluation of

the Cancer and Noncancer Dose‐Response Assessments. Risk Analysis, 34, 865-

878.

Howdeshell, K. L., Hotchkiss, A. K., Thayer, K. A., Vandenbergh, J. G. & Vom Saal, F.

S. 1999. Environmental toxins: exposure to bisphenol A advances puberty.

Nature, 401, 763.

Huc, L., Lemarié, A., Guéraud, F. & Héliès-Toussaint, C. 2012. Low concentrations of

bisphenol A induce lipid accumulation mediated by the production of reactive

oxygen species in the mitochondria of HepG2 cells. Toxicology in vitro, 26, 709-

717.

Hugo, E. R., Brandebourg, T. D., Woo, J. G., Loftus, J., Alexander, J. W. & Ben-

Jonathan, N. 2008. Bisphenol A at environmentally relevant doses inhibits

adiponectin release from human adipose tissue explants and adipocytes.

Environmental health perspectives, 116, 1642.

http://etd.uwc.ac.za/

88

Hulbert, A., Pamplona, R., Buffenstein, R. & Buttemer, W. 2007. Life and death:

metabolic rate, membrane composition, and life span of animals. Physiological

reviews, 87, 1175-1213.

Ibhazehiebo, K., Iwasaki, T., Shimokawa, N. & Koibuchi, N. 2011. 1, 2, 5, 6, 9, 10-

αHexabromocyclododecane (HBCD) impairs thyroid hormone-induced dendrite

arborization of Purkinje cells and suppresses thyroid hormone receptor-mediated

transcription. The Cerebellum, 10, 22-31.

Ikezuki, Y., Tsutsumi, O., Takai, Y., Kamei, Y. & Taketani, Y. 2002. Determination of

bisphenol A concentrations in human biological fluids reveals significant early

prenatal exposure. Human reproduction, 17, 2839-2841.

Inanami, O., Asanuma, T., Inukai, N., Jin, T., Shimokawa, S., Kasai, N., Nakano, M.,

Sato, F. & Kuwabara, M. 1995. The suppression of age-related accumulation of

lipid peroxides in rat brain by administration of Rooibos tea (Aspalathus linearis).

Neuroscience letters, 196, 85-88.

Ishido, M., Yonemoto, J. & Morita, M. 2007. Mesencephalic neurodegeneration in the

orally administered bisphenol A-caused hyperactive rats. Toxicology letters, 173,

66-72.

Jaganyi, D. & Wheeler, P. J. 2003. Rooibos tea: equilibrium and extraction kinetics of

aspalathin. Food Chemistry, 83, 121-126.

Jalal, N., Surendranath, A. R., Pathak, J. L., Yu, S. & Chung, C. Y. 2017. Bisphenol A

(BPA) the mighty and the mutagenic. Toxicology reports.

Jolles, J., Rompa-Barendregt, J. & Gispen, W. 1979. Novelty and grooming behavior in

the rat. Behavioral and neural biology, 25, 563-572.

Joubert, E. 1996. HPLC quantification of the dihydrochalcones, aspalathin and

nothofagin in rooibos tea (Aspalathus linearis) as affected by processing. Food

Chemistry, 55, 403-411.

Joubert, E. & De Beer, D. 2011. Rooibos (Aspalathus linearis) beyond the farm gate:

From herbal tea to potential phytopharmaceutical. South African Journal of

Botany, 77, 869-886.

Joubert, E. & Ferreira, D. 1996. Antioxidants of Rooibos tea-a possible explanation for its

health promoting properties? SA Journal of Food Science and Nutrition, 8, 79-84.

Joubert, E., Gelderblom, W., Louw, A. & De Beer, D. 2008. South African herbal teas:

Aspalathus linearis, Cyclopia spp. and Athrixia phylicoides—a review. Journal

of ethnopharmacology, 119, 376-412.

Joubert, E., Winterton, P., Britz, T. J. & Ferreira, D. 2004. Superoxide anion and α, α-

diphenyl-β-picrylhydrazyl radical scavenging capacity of rooibos (Aspalathus

linearis) aqueous extracts, crude phenolic fractions, tannin and flavonoids. Food

Research International, 37, 133-138.

http://etd.uwc.ac.za/

89

Kabuto, H., Amakawa, M. & Shishibori, T. 2004. Exposure to bisphenol A during

embryonic/fetal life and infancy increases oxidative injury and causes

underdevelopment of the brain and testis in mice. Life sciences, 74, 2931-2940.

Kabuto, H., Hasuike, S., Minagawa, N. & Shishibori, T. 2003. Effects of bisphenol A on

the metabolisms of active oxygen species in mouse tissues. Environmental

research, 93, 31-35.

Kajta, M. & Wójtowicz, A. K. 2013. Impact of endocrine-disrupting chemicals on neural

development and the onset of neurological disorders. Pharmacological Reports,

65, 1632-1639.

Kalueff, A. V., Aldridge, J. W., Laporte, J. L., Murphy, D. L. & Tuohimaa, P. 2007.

Analyzing grooming microstructure in neurobehavioral experiments. Nature

protocols, 2, 2538.

Kalueff, A. V. & Tuohimaa, P. 2005. Mouse grooming microstructure is a reliable

anxiety marker bidirectionally sensitive to GABAergic drugs. European journal

of pharmacology, 508, 147-153.

Kang, J., Kondo, F. & Katayama, Y. 2006. Human exposure to bisphenol A Toxicology

226 (2–3): 79–89. Find this article online.

Katz, R. J. & Roth, K. A. 1979. Stress induced grooming in the rat--an endorphin

mediated syndrome. Neuroscience Letters, 13, 209-212.

Kawano, A., Nakamura, H., Hata, S.-I., Minakawa, M., Miura, Y. & Yagasaki, K. 2009.

Hypoglycemic effect of aspalathin, a rooibos tea component from Aspalathus

linearis, in type 2 diabetic model db/db mice. Phytomedicine, 16, 437-443.

Kendigelen, P., Sun, D. & Cengiz, P. 2012. Neurobehavioral assessments of global

cerebral ischemia. Animal Models of Acute Neurological Injuries II. Springer.

Khan, A.-U. & Gilani, A. H. 2006. Selective bronchodilatory effect of Rooibos tea

(Aspalathus linearis) and its flavonoid, chrysoeriol. European journal of

nutrition, 45, 463.

Khattak, S., Guiti, K., Mcmartin, K., Barrera, M., Kennedy, D. & Koren, G. 1999.

Pregnancy outcome following gestational exposure to organic solvents: a

prospective controlled study. Jama, 281, 1106-1109.

Kiernan, J. 2012. Anatomy of the temporal lobe. Epilepsy research and treatment, 2012.

Kimura-Kuroda, J., Nagata, I. & Kuroda, Y. 2007. Disrupting effects of hydroxy-

polychlorinated biphenyl (PCB) congeners on neuronal development of

cerebellar Purkinje cells: a possible causal factor for developmental brain

disorders? Chemosphere, 67, S412-S420.

Kimura, E., Matsuyoshi, C., Miyazaki, W., Benner, S., Hosokawa, M., Yokoyama, K.,

Kakeyama, M. & Tohyama, C. 2016. Prenatal exposure to bisphenol A impacts

http://etd.uwc.ac.za/

90

neuronal morphology in the hippocampal CA1 region in developing and aged

mice. Archives of toxicology, 90, 691-700.

Kirino, T. 1982. Delayed neuronal death in the gerbil hippocampus following ischemia.

Brain research, 239, 57-69.

Kodavanti, P. R. S., Valdez, J., Yang, J.-H., Curras-Collazo, M. & Loganathan, B. G.

2017. Polychlorinated Biphenyls, Polybrominated Biphenyls, Polychlorinated

Dibenzo-p-dioxins, and Polychlorinated Dibenzofurans. Reproductive and

Developmental Toxicology (Second Edition). Elsevier.

Koong, L. Y. & Watson, C. S. 2015. Rapid, nongenomic signaling effects of several

xenoestrogens involved in early-vs. late-stage prostate cancer cell proliferation.

Endocrine Disruptors, 3, e995003.

Krafczyk, N. & Glomb, M. A. 2008. Characterization of phenolic compounds in rooibos

tea. Journal of Agricultural and Food Chemistry, 56, 3368-3376.

Krishnan, A. V., Stathis, P., Permuth, S. F., Tokes, L. & Feldman, D. 1993. Bisphenol-A:

an estrogenic substance is released from polycarbonate flasks during autoclaving.

Endocrinology, 132, 2279-2286.

Kunz, N., Camm, E. J., Somm, E., Lodygensky, G., Darbre, S., Aubert, M. L., Hüppi, P.

S., Sizonenko, S. V. & Gruetter, R. 2011. Developmental and metabolic brain

alterations in rats exposed to bisphenol A during gestation and lactation.

International Journal of Developmental Neuroscience, 29, 37-43.

Lange, W. 1975. Cell number and cell density in the cerebellar cortex of man and some

other mammals. Cell and tissue research, 157, 115-124.

Le, H. H., Carlson, E. M., Chua, J. P. & Belcher, S. M. 2008. Bisphenol A is released

from polycarbonate drinking bottles and mimics the neurotoxic actions of

estrogen in developing cerebellar neurons. Toxicology letters, 176, 149-156.

Leem, Y. H., Oh, S., Kang, H. J., Kim, J. H., Yoon, J. & Chang, J. S. 2017. BPA‐toxicity

via superoxide anion overload and a deficit in β‐catenin signaling in human bone

mesenchymal stem cells. Environmental toxicology, 32, 344-352.

Lemaire, G., Terouanne, B., Mauvais, P., Michel, S. & Rahmani, R. 2004. Effect of

organochlorine pesticides on human androgen receptor activation in vitro.

Toxicology and applied pharmacology, 196, 235-246.

Leonard, S. T., Alizadeh-Naderi, R., Stokes, K. & Ferkin, M. H. 2005. The role of

prolactin and testosterone in mediating seasonal differences in the self-grooming

behavior of male meadow voles, Microtus pennsylvanicus. Physiology &

behavior, 85, 461-468.

Leranth, C., Hajszan, T., Szigeti-Buck, K., Bober, J. & Maclusky, N. J. 2008. Bisphenol

A prevents the synaptogenic response to estradiol in hippocampus and prefrontal

cortex of ovariectomized nonhuman primates. Proceedings of the National

Academy of Sciences.

http://etd.uwc.ac.za/

91

Levisohn, L., Cronin-Golomb, A. & Schmahmann, J. D. 2000. Neuropsychological

consequences of cerebellar tumour resection in children: cerebellar cognitive

affective syndrome in a paediatric population. Brain, 123, 1041-1050.

Li, D.-K., Miao, M., Zhou, Z., Wu, C., Shi, H., Liu, X., Wang, S. & Yuan, W. 2013a.

Urine bisphenol-A level in relation to obesity and overweight in school-age

children. PloS one, 8, e65399.

Li, L.-X., Chen, L., Meng, X.-Z., Chen, B.-H., Chen, S.-Q., Zhao, Y., Zhao, L.-F., Liang,

Y. & Zhang, Y.-H. 2013b. Exposure levels of environmental endocrine disruptors

in mother-newborn pairs in China and their placental transfer characteristics.

PloS one, 8, e62526.

Limperopoulos, C., Bassan, H., Gauvreau, K., Robertson, R. L., Sullivan, N. R., Benson,

C. B., Avery, L., Stewart, J., Soul, J. S. & Ringer, S. A. 2007. Does cerebellar

injury in premature infants contribute to the high prevalence of long-term

cognitive, learning, and behavioral disability in survivors? Pediatrics, 120, 584-

593.

Liu, F., Zhang, Z., Lin, X., Teng, G., Meng, H., Yu, T., Fang, F., Zang, F., Li, Z. & Liu,

S. 2011. Development of the human fetal cerebellum in the second trimester: a

post mortem magnetic resonance imaging evaluation. Journal of anatomy, 219,

582-588.

Liu, Z.-H., Ding, J.-J., Yang, Q.-Q., Song, H.-Z., Chen, X.-T., Xu, Y., Xiao, G.-R. &

Wang, H.-L. 2016. Early developmental bisphenol-A exposure sex-independently

impairs spatial memory by remodeling hippocampal dendritic architecture and

synaptic transmission in rats. Scientific reports, 6, 32492.

Liu, Z., Ren, Z., Zhang, J., Chuang, C.-C., Kandaswamy, E., Zhou, T. & Zuo, L. 2018.

Role of ROS and Nutritional Antioxidants in Human Diseases. Frontiers in

physiology, 9.

Longnecker, M. P., Rogan, W. J. & Lucier, G. 1997. The human health effects of DDT

(dichlorodiphenyltrichloroethane) and PCBS (polychlorinated biphenyls) and an

overview of organochlorines in public health. Annual review of public health, 18,

211-244.

Maclusky, N. J., Hajszan, T. & Leranth, C. 2005. The environmental estrogen bisphenol

A inhibits estradiol-induced hippocampal synaptogenesis. Environmental health

perspectives, 113, 675.

Maloku, E., Covelo, I. R., Hanbauer, I., Guidotti, A., Kadriu, B., Hu, Q., Davis, J. M. &

Costa, E. 2010. Lower number of cerebellar Purkinje neurons in psychosis is

associated with reduced reelin expression. Proceedings of the National Academy

of Sciences, 107, 4407-4411.

Manibusan, M. & Touart, L. 2017. A comprehensive review of regulatory test methods

for endocrine adverse health effects. Critical reviews in toxicology, 47, 440-488.

http://etd.uwc.ac.za/

92

Manto, M. 2012. Toxic agents causing cerebellar ataxias. Handbook of clinical

neurology. Elsevier.

Markey, C. M., Michaelson, C. L., Veson, E. C., Sonnenschein, C. & Soto, A. M. 2001.

The mouse uterotrophic assay: a reevaluation of its validity in assessing the

estrogenicity of bisphenol A. Environmental health perspectives, 109, 55.

Marnewick, J. L., Joubert, E., Swart, P., Van Der Westhuizen, F. & Gelderblom, W. C.

2003. Modulation of hepatic drug metabolizing enzymes and oxidative status by

rooibos (Aspalathus linearis) and honeybush (Cyclopia intermedia), green and

black (Camellia sinensis) teas in rats. Journal of agricultural and food chemistry,

51, 8113-8119.

Marnewick, J. L., Rautenbach, F., Venter, I., Neethling, H., Blackhurst, D. M.,

Wolmarans, P. & Macharia, M. 2011. Effects of rooibos (Aspalathus linearis) on

oxidative stress and biochemical parameters in adults at risk for cardiovascular

disease. Journal of ethnopharmacology, 133, 46-52.

Masood, A., Banerjee, B., Vijayan, V. & Ray, A. 2003. Modulation of stress-induced

neurobehavioral changes by nitric oxide in rats. European journal of

pharmacology, 458, 135-139.

Masuno, H., Kidani, T., Sekiya, K., Sakayama, K., Shiosaka, T., Yamamoto, H. &

Honda, K. 2002. Bisphenol A in combination with insulin can accelerate the

conversion of 3T3-L1 fibroblasts to adipocytes. Journal of lipid research, 43,

676-684.

Mathiasen, J. R. & Dicamillo, A. 2010. Novel object recognition in the rat: a facile assay

for cognitive function. Current protocols in pharmacology, 49 (1), 5-59.

Mathisen, G.H., Yazdani, M., Rakkestad, K.E., Aden, P.K., Bodin, J., Samuelsen, M.,

Nygaard, U.C., Goverud, I.L., Gaarder, M., Løberg, E.M. and Bølling, A.K.,

2013. Prenatal exposure to bisphenol A interferes with the development of

cerebellar granule neurons in mice and chicken. International Journal of

Developmental Neuroscience, 31(8), 762-769.

Matsumoto, J., Yokota, H. & Yuasa, A. 2002. Developmental increases in rat hepatic

microsomal UDP-glucuronosyltransferase activities toward xenoestrogens and

decreases during pregnancy. Environmental health perspectives, 110, 193.

Matsushima, A., Liu, X., Okada, H., Shimohigashi, M. & Shimohigashi, Y. 2010.

Bisphenol AF is a full agonist for the estrogen receptor ERα but a highly specific

antagonist for ERβ. Environmental health perspectives, 118, 1267.

Mcallister, A. K., Katz, L. C. & Lo, D. C. 1997. Opposing roles for endogenous BDNF

and NT-3 in regulating cortical dendritic growth. Neuron, 18, 767-778.

Mcallister, T. W., Flashman, L. A., Sparling, M. B. & Saykin, A. J. 2004. Working

memory deficits after traumatic brain injury: catecholaminergic mechanisms and

prospects for treatment—a review. Brain Injury, 18, 331-350.

http://etd.uwc.ac.za/

93

Mckay, D. L. & Blumberg, J. B. 2007. A review of the bioactivity of South African

herbal teas: rooibos (Aspalathus linearis) and honeybush (Cyclopia intermedia).

Phytotherapy Research: An International Journal Devoted to Pharmacological

and Toxicological Evaluation of Natural Product Derivatives, 21, 1-16.

Meeker, J. D. 2012. Exposure to environmental endocrine disruptors and child

development. Archives of pediatrics & adolescent medicine, 166, 952-958.

Mentor, S. 2014. In vitro modulatory effects of fermented rooibos extract (Aspalathus

linearis) against ethanol-induced effects on the mouse blood-brain barrier. Msc,

University of the Western Cape.

Mervis, R., Bachstetter, A., Harry, G., Tilson, H. & Kodavanti, P. 2002. Long-lasting

neurostructural consequences in the rat hippocampus by developmental exposure

to a mixture of polychlorinated biphenyls. The Toxicologist, 133.

Messerschmidt, A., Fuiko, R., Prayer, D., Brugger, P. C., Boltshauser, E., Zoder, G.,

Sterniste, W., Weber, M. & Birnbacher, R. 2008. Disrupted cerebellar

development in preterm infants is associated with impaired neurodevelopmental

outcome. European journal of pediatrics, 167, 1141-1147.

Mita, L., Baldi, A., Diano, N., Viggiano, E., Portaccio, M., Nicolucci, C., Grumiro, L.,

Menale, C., Mita, D.G., Spugnini, E.P. and Viceconte, R., 2012. Differential

accumulation of BPA in some tissues of offspring of Balb-C mice exposed to

different BPA doses. Environmental toxicology and pharmacology, 33(1), pp.9-

15.

Miyagawa, K., Narita, M., Narita, M., Akama, H. & Suzuki, T. 2007. Memory

impairment associated with a dysfunction of the hippocampal cholinergic system

induced by prenatal and neonatal exposures to bisphenol-A. Neuroscience letters,

418, 236-241.

Miyakoda, H., Tabata, M., Onodera, S. & Tkeda, K. 1999. Passage of bisphenol A into

the fetus of the pregnant rat. Journal of health science, 45, 318-323.

Miyawaki, J., Sakayama, K., Kato, H., Yamamoto, H. & Masuno, H. 2007. Perinatal and

postnatal exposure to bisphenol a increases adipose tissue mass and serum

cholesterol level in mice. Journal of atherosclerosis and thrombosis, 14, 245-

252.

Moghaddam, H. S., Samarghandian, S. & Farkhondeh, T. 2015. Effect of bisphenol A on

blood glucose, lipid profile and oxidative stress indices in adult male mice.

Toxicology mechanisms and methods, 25, 507-513.

Moriyama, K., Tagami, T., Akamizu, T., Usui, T., Saijo, M., Kanamoto, N., Hataya, Y.,

Shimatsu, A., Kuzuya, H. & Nakao, K. 2002. Thyroid hormone action is

disrupted by bisphenol A as an antagonist. The Journal of Clinical Endocrinology

& Metabolism, 87, 5185-5190.

Morton, J. F. 1983. Rooibos tea, Aspalathus linearis, a caffeineless, low-tannin beverage.

Economic Botany, 37, 164-173.

http://etd.uwc.ac.za/

94

Morton, S. M. & Bastian, A. J. 2004. Cerebellar control of balance and locomotion. The

Neuroscientist, 10, 247-259.

Moser, E. I., Roudi, Y., Witter, M. P., Kentros, C., Bonhoeffer, T. & Moser, M.-B. 2014.

Grid cells and cortical representation. Nature Reviews Neuroscience, 15, 466.

Muller, C., Joubert, E., De Beer, D., Sanderson, M., Malherbe, C., Fey, S. & Louw, J.

2012. Acute assessment of an aspalathin-enriched green rooibos (Aspalathus

linearis) extract with hypoglycemic potential. Phytomedicine, 20, 32-39.

Munguia-Lopez, E., Gerardo-Lugo, S., Peralta, E., Bolumen, S. & Soto-Valdez, H. 2005.

Migration of bisphenol A (BPA) from can coatings into a fatty-food simulant and

tuna fish. Food additives and contaminants, 22, 892-898.

Murray, T. J., Maffini, M. V., Ucci, A. A., Sonnenschein, C. & Soto, A. M. 2007.

Induction of mammary gland ductal hyperplasias and carcinoma in situ following

fetal bisphenol A exposure. Reproductive toxicology, 23, 383-390.

Nagata, H., Takekoshi, S., Takagi, T., Honma, T. & Watanabe, K. 1999. Antioxidative

action of flavonoids, quercetin and catechin, mediated by the activation of

glutathione peroxidase. Tokai Journal of Experimental and Clinical Medicine,

24, 1-11.

Nagata, K., Nakashima-Kamimura, N., Mikami, T., Ohsawa, I. & Ohta, S. 2009.

Consumption of molecular hydrogen prevents the stress-induced impairments in

hippocampus-dependent learning tasks during chronic physical restraint in mice.

Neuropsychopharmacology, 34, 501.

Nakagami, A., Negishi, T., Kawasaki, K., Imai, N., Nishida, Y., Ihara, T., Kuroda, Y.,

Yoshikawa, Y. & Koyama, T. 2009. Alterations in male infant behaviors towards

its mother by prenatal exposure to bisphenol A in cynomolgus monkeys (Macaca

fascicularis) during early suckling period. Psychoneuroendocrinology, 34, 1189-

1197.

Nakamura, D., Yanagiba, Y., Duan, Z., Ito, Y., Okamura, A., Asaeda, N., Tagawa, Y., Li,

C., Taya, K. & Zhang, S.-Y. 2010. Bisphenol A may cause testosterone reduction

by adversely affecting both testis and pituitary systems similar to estradiol.

Toxicology letters, 194, 16-25.

Negri-Cesi, P. 2015. Bisphenol A interaction with brain development and functions.

Dose-Response, 13, 1559325815590394.

Newbold, R. R., Padilla-Banks, E. & Jefferson, W. N. 2009. Environmental estrogens and

obesity. Molecular and cellular endocrinology, 304, 84-89.

Nicol, C. J., Zielenski, J., Tsui, L.-C. & Wells, P. G. 2000. An embryoprotective role for

glucose-6-phosphate dehydrogenase in developmental oxidative stress and

chemical teratogenesis. The FASEB Journal, 14, 111-127.

http://etd.uwc.ac.za/

95

Nishikawa, M., Iwano, H., Yanagisawa, R., Koike, N., Inoue, H. & Yokota, H. 2010.

Placental transfer of conjugated bisphenol A and subsequent reactivation in the

rat fetus. Environmental health perspectives, 118, 1196.

Oka, T., Adati, N., Shinkai, T., Sakuma, K., Nishimura, T. & Kurose, K. 2003. Bisphenol

A induces apoptosis in central neural cells during early development of Xenopus

laevis. Biochemical and biophysical research communications, 312, 877-882.

Ooe, H., Taira, T., Iguchi-Ariga, S. M. & Ariga, H. 2005. Induction of reactive oxygen

species by bisphenol A and abrogation of bisphenol A-induced cell injury by DJ-

1. Toxicological Sciences, 88, 114-126.

Opuwari, C. & Monsees, T. 2014. In vivo effects of A spalathus linearis (rooibos) on

male rat reproductive functions. Andrologia, 46, 867-877.

Ozaydın, T., Oznurlu, Y., Sur, E., Celik, I., Uluısık, D. & Dayan, M. 2018. Effects of

bisphenol A on antioxidant system and lipid profile in rats. Biotechnic &

Histochemistry, 1-8.

Pantsi, W., Marnewick, J., Esterhuyse, A., Rautenbach, F. & Van Rooyen, J. 2011.

Rooibos (Aspalathus linearis) offers cardiac protection against

ischaemia/reperfusion in the isolated perfused rat heart. Phytomedicine, 18, 1220-

1228.

Patisaul, H. B. & Bateman, H. L. 2008. Neonatal exposure to endocrine active

compounds or an ERβ agonist increases adult anxiety and aggression in

gonadally intact male rats. Hormones and behavior, 53, 580-588.

Patisaul, H. B., Fortino, A. E. & Polston, E. K. 2006. Neonatal genistein or bisphenol-A

exposure alters sexual differentiation of the AVPV. Neurotoxicology and

teratology, 28, 111-118.

Penhune, V. B., Zatorre, R. J. & Evans, A. C. 1998. Cerebellar contributions to motor

timing: a PET study of auditory and visual rhythm reproduction. Journal of

cognitive neuroscience, 10, 752-765.

Perera, F. & Herbstman, J. 2011. Prenatal environmental exposures, epigenetics, and

disease. Reproductive toxicology, 31, 363-373.

Perron, N. R. & Brumaghim, J. L. 2009. A review of the antioxidant mechanisms of

polyphenol compounds related to iron binding. Cell biochemistry and biophysics,

53, 75-100.

Persson, S. & Magnusson, U. 2015. Environmental pollutants and alterations in the

reproductive system in wild male mink (Neovison vison) from Sweden.

Chemosphere, 120, 237-245.

Pfeifer, D., Chung, Y. M. & Hu, M. C. 2015. Effects of low-dose bisphenol A on DNA

damage and proliferation of breast cells: the role of c-Myc. Environmental health

perspectives, 123, 1271.

http://etd.uwc.ac.za/

96

Phrakonkham, P., Viengchareun, S., Belloir, C., Lombes, M., Artur, Y. & Canivenc-

Lavier, M.-C. 2008. Dietary xenoestrogens differentially impair 3T3-L1

preadipocyte differentiation and persistently affect leptin synthesis. The Journal

of steroid biochemistry and molecular biology, 110, 95-103.

Poongothai, S., Ravikrishnan, R. & Murthy, P. 2007. Endocrine disruption and

perspective human health implications: a review. The Internet Journal of

Toxicology, 4.

Porreca, I., Severino, L. U., D’angelo, F., Cuomo, D., Ceccarelli, M., Altucci, L.,

Amendola, E., Nebbioso, A., Mallardo, M. & De Felice, M. 2016. “Stockpile” of

slight transcriptomic changes determines the indirect genotoxicity of low-dose

BPA in thyroid cells. PloS one, 11, e0151618.

Pottenger, L. H., Domoradzki, J. Y., Markham, D. A., Hansen, S. C., Cagen, S. Z. &

Waechter Jr, J. M. 2000. The relative bioavailability and metabolism of bisphenol

A in rats is dependent upon the route of administration. Toxicological Sciences,

54, 3-18.

Pouzaud, F., Thierry-Mieg, M., Burga, K. & Vérines-Jouin, L. 2018. Concerns related to

ED-mediated effects of Bisphenol A and their regulatory consideration.

Molecular and Cellular Endocrinology, 475, 92-106.

Powls, A., Botting, N., Cooke, R. W. & Marlow, N. 1995. Motor impairment in children

12 to 13 years old with a birthweight of less than 1250 g. Archives of Disease in

Childhood-Fetal and Neonatal Edition, 73, F62-F66.

Prashant, T., Dheeraj, A. & Shubhangi, D. 2014. Impact of Plants having Potential Action

on CNS: An Overview. Asian Journal of Pharmacy and Technology, 4, 100-105.

Pritchett, J., Kuester, R. & Sipes, I. 2002. Metabolism of bisphenol a in primary cultured

hepatocytes from mice, rats, and humans. Drug Metabolism and Disposition, 30,

1180-1185.

Prut, L. & Belzung, C. 2003. The open field as a paradigm to measure the effects of drugs

on anxiety-like behaviors: a review. European journal of pharmacology, 463, 3-

33.

Pulsinelli, W. A., Brierley, J. B. & Plum, F. 1982. Temporal profile of neuronal damage

in a model of transient forebrain ischemia. Annals of Neurology: Official Journal

of the American Neurological Association and the Child Neurology Society, 11,

491-498.

Rabe, C., Steenkamp, J. A., Joubert, E., Burger, J. F. & Ferreira, D. 1994. Phenolic

metabolites from rooibos tea (Aspalathus linearis). Phytochemistry, 35, 1559-

1565.

Racine, R. J. 1972. Modification of seizure activity by electrical stimulation: II. Motor

seizure. Electroencephalography and clinical neurophysiology, 32, 281-294.

http://etd.uwc.ac.za/

97

Ramírez-López, M. T., Arco, R., Decara, J., Vázquez, M., Blanco, R. N., Alén, F.,

Suárez, J., De Heras, R. G. & De Fonseca, F. R. 2016. Exposure to a highly

caloric palatable diet during the perinatal period affects the expression of the

endogenous cannabinoid system in the brain, liver and adipose tissue of adult rat

offspring. PloS one, 11, e0165432.

Ranjbar, H., Radahmadi, M., Reisi, P. & Alaei, H. 2017. Effects of electrical lesion of

basolateral amygdala nucleus on rat anxiety‐like behaviour under acute, sub‐

chronic, and chronic stresses. Clinical and Experimental Pharmacology and

Physiology, 44, 470-479.

Rasier, G., Parent, A.-S., Gérard, A., Denooz, R., Lebrethon, M.-C., Charlier, C. &

Bourguignon, J.-P. 2007. Mechanisms of interaction of endocrine-disrupting

chemicals with glutamate-evoked secretion of gonadotropin-releasing hormone.

Toxicological Sciences, 102, 33-41.

Reynecke, J., Coetzee, W. & Bester, J. 1949. Rooibos tea. A preliminary report on the

composition. Farming in South Africa, 24, 397-398.

Ribeiro, E., Ladeira, C. & Viegas, S. 2017. Occupational exposure to Bisphenol A (BPA):

a reality that still needs to be unveiled. Toxics, 5, 22.

Riedel, C. J., Muraszko, K. M. & Youle, R. J. 1990. Diphtheria toxin mutant selectively

kills cerebellar Purkinje neurons. Proceedings of the National Academy of

Sciences, 87, 5051-5055.

Rilling, J. K. & Insel, T. R. 1999. The primate neocortex in comparative perspective

using magnetic resonance imaging. Journal of human evolution, 37, 191-223.

Rubin, B. S. 2011. Bisphenol A: an endocrine disruptor with widespread exposure and

multiple effects. The Journal of steroid biochemistry and molecular biology, 127,

27-34.

Rubin, B. S., Lenkowski, J. R., Schaeberle, C. M., Vandenberg, L. N., Ronsheim, P. M.

& Soto, A. M. 2006. Evidence of altered brain sexual differentiation in mice

exposed perinatally to low, environmentally relevant levels of bisphenol A.

Endocrinology, 147, 3681-3691.

Rubin, B. S., Murray, M. K., Damassa, D. A., King, J. C. & Soto, A. M. 2001. Perinatal

exposure to low doses of bisphenol A affects body weight, patterns of estrous

cyclicity, and plasma LH levels. Environmental health perspectives, 109, 675.

Rubin, B. S. & Soto, A. M. 2009. Bisphenol A: perinatal exposure and body weight.

Molecular and cellular endocrinology, 304, 55-62.

Rudel, R. A., Gray, J. M., Engel, C. L., Rawsthorne, T. W., Dodson, R. E., Ackerman, J.

M., Rizzo, J., Nudelman, J. L. & Brody, J. G. 2011. Food packaging and

bisphenol A and bis (2-ethyhexyl) phthalate exposure: findings from a dietary

intervention. Environmental health perspectives, 119, 914.

http://etd.uwc.ac.za/

98

Saikhedkar, N., Bhatnagar, M., Jain, A., Sukhwal, P., Sharma, C. & Jaiswal, N. 2014.

Effects of mobile phone radiation (900 MHz radiofrequency) on structure and

functions of rat brain. Neurological research, 36, 1072-1079.

Sajiki, J., Takahashi, K. & Yonekubo, J. 1999. Sensitive method for the determination of

bisphenol-A in serum using two systems of high-performance liquid

chromatography. Journal of Chromatography B: Biomedical Sciences and

Applications, 736, 255-261.

Sakaue, M., Ohsako, S., Ishimura, R., Kurosawa, S., Kurohmaru, M., Hayashi, Y., Aoki,

Y., Yonemoto, J. & Tohyama, C. 2001. Bisphenol-A affects spermatogenesis in

the adult rat even at a low dose. Journal of occupational health, 43, 185-190.

Salleh, N., Giribabu, N., Feng, A. O. M. & Myint, K. 2015. Bisphenol A,

Dichlorodiphenyltrichloroethane (DDT) and Vinclozolin Affect ex-vivo Uterine

Contraction in Rats via Uterotonin (Prostaglandin F2α, Acetylcholine and

Oxytocin) Related Pathways. International journal of medical sciences, 12, 914.

Salman, M. S. 2002. Topical review: the cerebellum: it's about time! but timing is not

everything-new insights into the role of the cerebellum in timing motor and

cognitive tasks. Journal of Child Neurology, 17, 1-9.

Sanders, H. O., Huckins, J., Johnson, B. T. & Skaar, D. 1981. Biological effects of

kepone and mirex in freshwater invertebrates. Archives of environmental

contamination and toxicology, 10, 531-539.

Sanders, T., Liu, Y., Buchner, V. & Tchounwou, P. B. 2009. Neurotoxic effects and

biomarkers of lead exposure: a review. Reviews on environmental health, 24, 15-

46.

Schloms, L., Storbeck, K.-H., Swart, P., Gelderblom, W. C. & Swart, A. C. 2012. The

influence of Aspalathus linearis (Rooibos) and dihydrochalcones on adrenal

steroidogenesis: Quantification of steroid intermediates and end products in

H295R cells. The Journal of steroid biochemistry and molecular biology, 128,

128-138.

Schug, T. T., Blawas, A. M., Gray, K., Heindel, J. J. & Lawler, C. P. 2015. Elucidating

the links between endocrine disruptors and neurodevelopment. Endocrinology,

156, 1941-1951.

Schug, T. T., Janesick, A., Blumberg, B. & Heindel, J. J. 2011. Endocrine disrupting

chemicals and disease susceptibility. The Journal of steroid biochemistry and

molecular biology, 127, 204-215.

Seale, S. M., Feng, Q., Agarwal, A. K. & El-Alfy, A. T. 2012. Neurobehavioral and

transcriptional effects of acrylamide in juvenile rats. Pharmacology Biochemistry

and Behavior, 101, 77-84.

Seibenhener, M. L. & Wooten, M. C. 2015. Use of the open field maze to measure

locomotor and anxiety-like behavior in mice. Journal of visualized experiments:

JoVE.

http://etd.uwc.ac.za/

99

Shankar, A., Teppala, S. & Sabanayagam, C. 2012. Urinary bisphenol a levels and

measures of obesity: results from the national health and nutrition examination

survey 2003–2008. ISRN endocrinology, 2012.

Shanle, E. K. & Xu, W. 2010. Endocrine disrupting chemicals targeting estrogen receptor

signaling: identification and mechanisms of action. Chemical research in

toxicology, 24, 6-19.

Shimamura, N., Miyase, T., Umehara, K., Warashina, T. & Fujii, S. 2006. Phytoestrogens

from Aspalathus linearis. Biological and Pharmaceutical Bulletin, 29, 1271-

1274.

Shimizu, M., Ohta, K., Matsumoto, Y., Fukuoka, M., Ohno, Y. & Ozawa, S. 2002.

Sulfation of bisphenol A abolished its estrogenicity based on proliferation and

gene expression in human breast cancer MCF-7 cells. Toxicology in vitro, 16,

549-556.

Silvers, J. M., Harrod, S. B., Mactutus, C. F. & Booze, R. M. 2007. Automation of the

novel object recognition task for use in adolescent rats. Journal of neuroscience

methods, 166, 99-103.

Simerly, R. B. 2008. Hypothalamic substrates of metabolic imprinting. Physiology &

behavior, 94, 79-89.

Singleton, D. W. & Khan, S. A. 2003. Xenoestrogen exposure and mechanisms of

endocrine disruption. Front Biosci, 8, s110-s118.

Snell, R. S. 2010. Clinical neuroanatomy, Lippincott Williams & Wilkins.

Snider, R. S. 1958. The cerebellum. Scientific American, 199, 84-91.

Snyder, R. W., Maness, S. C., Gaido, K. W., Welsch, F., Sumner, S. C. & Fennell, T. R.

2000. Metabolism and disposition of bisphenol A in female rats. Toxicology and

applied pharmacology, 168, 225-234.

Song, S., Zhang, L., Zhang, H., Wei, W. & Jia, L. 2014. Perinatal BPA exposure induces

hyperglycemia, oxidative stress and decreased adiponectin production in later life

of male rat offspring. International journal of environmental research and public

health, 11, 3728-3742.

Spinelli, J. J., Ng, C. H., Weber, J. P., Connors, J. M., Gascoyne, R. D., Lai, A. S.,

Brooks‐Wilson, A. R., Le, N. D., Berry, B. R. & Gallagher, R. P. 2007.

Organochlorines and risk of non‐Hodgkin lymphoma. International journal of

cancer, 121, 2767-2775.

Spruijt, B. M., Welbergen, P., Brakkee, J. & Gispen, W. H. 1988. An ethological analysis

of excessive grooming in young and aged rats. Annals of the New York Academy

of Sciences, 525, 89-100.

Spruijt, B. V., Van Hooff, J. & Gispen, W. 1992. Ethology and neurobiology of grooming

behavior. Physiological reviews, 72, 825-852.

http://etd.uwc.ac.za/

100

Squire, L. R., Wixted, J. T. & Clark, R. E. 2007. Recognition memory and the medial

temporal lobe: a new perspective. Nature Reviews Neuroscience, 8, 872.

Steinmetz, R., Mitchner, N. A., Grant, A., Allen, D. L., Bigsby, R. M. & Ben-Jonathan,

N. 1998. The xenoestrogen bisphenol A induces growth, differentiation, and c-fos

gene expression in the female reproductive tract. Endocrinology, 139, 2741-2747.

Stephan, H., Frahm, H. & Baron, G. 1981. New and revised data on volumes of brain

structures in insectivores and primates. Folia primatologica, 35, 1-29.

Sugiura-Ogasawara, M., Ozaki, Y., Sonta, S.-I., Makino, T. & Suzumori, K. 2005.

Exposure to bisphenol A is associated with recurrent miscarriage. Human

reproduction, 20, 2325-2329.

Sutherland, R.J. and Rudy, J.W., 1989. Configural association theory: The role of the

hippocampal formation in learning, memory, and amnesia. Psychobiology, 17(2),

129-144.

Takahashi, O. & Oishi, S. 2000. Disposition of orally administered 2, 2-Bis (4-

hydroxyphenyl) propane (Bisphenol A) in pregnant rats and the placental transfer

to fetuses. Environmental Health Perspectives, 108, 931.

Takeuchi, T. & Tsutsumi, O. 2002. Serum bisphenol A concentrations showed gender

differences, possibly linked to androgen levels. Biochemical and biophysical

research communications, 291, 76-78.

Thomas, P. & Doughty, K. 2004. Disruption of rapid, nongenomic steroid actions by

environmental chemicals: interference with progestin stimulation of sperm

motility in Atlantic croaker. Environmental science & technology, 38, 6328-6332.

Tian, Y. H., Baek, J. H., Lee, S. Y. & Jang, C. G. 2010. Prenatal and postnatal exposure

to bisphenol a induces anxiolytic behaviors and cognitive deficits in mice.

Synapse, 64, 432-439.

Tiwari, S. K., Agarwal, S., Seth, B., Yadav, A., Ray, R. S., Mishra, V. N., & Chaturvedi,

R. K. (2015). Inhibitory effects of bisphenol-A on neural stem cells proliferation

and differentiation in the rat brain are dependent on Wnt/β-catenin pathway.

Molecular neurobiology, 52(3), 1735-1757.

Tokumoto, T., Tokumoto, M. & Thomas, P. 2007. Interactions of diethylstilbestrol (DES)

and DES analogs with membrane progestin receptor-α and the correlation with

their nongenomic progestin activities. Endocrinology, 148, 3459-3467.

Uglea, C. V. & Negulescu, I. I. 1991. Synthesis and characterization of oligomers, CRC

Press.

Upmeier, A., Degen, G. H., Diel, P., Michna, H. & Bolt, H. M. 2000. Toxicokinetics of

bisphenol A in female DA/Han rats after a single iv and oral administration.

Archives of toxicology, 74, 431-436.

http://etd.uwc.ac.za/

101

Van Abeelen, J. 1977. Rearing responses and locomotor activity in mice: Single-locus

control. Behavioral biology, 19, 401-404.

Van Der Merwe, J., Joubert, E., Richards, E., Manley, M., Snijman, P., Marnewick, J. &

Gelderblom, W. 2006. A comparative study on the antimutagenic properties of

aqueous extracts of Aspalathus linearis (rooibos), different Cyclopia

spp.(honeybush) and Camellia sinensis teas. Mutation Research/Genetic

Toxicology and Environmental Mutagenesis, 611, 42-53.

Van Lier, H., Drinkenburg, W. H., Van Eeten, Y. J. & Coenen, A. M. 2004. Effects of

diazepam and zolpidem on EEG beta frequencies are behavior-specific in rats.

Neuropharmacology, 47, 163-174.

Van Praag, H., Kempermann, G. & Gage, F. H. 2000. Neural consequences of

enviromental enrichment. Nature Reviews Neuroscience, 1, 191.

Van Wyk, B.-E., Oudtshoorn, B. V. & Gericke, N. 1997. Medicinal Plants of South

Africa, Briza.

Vandenberg, L.N., Chahoud, I., Padmanabhan, V., Paumgartten, F.J. and Schoenfelder,

G., 2010. Biomonitoring studies should be used by regulatory agencies to assess

human exposure levels and safety of bisphenol A. Environmental health

perspectives, 118(8): 1051.

Vandenberg, L. N., Hauser, R., Marcus, M., Olea, N. & Welshons, W. V. 2007. Human

exposure to bisphenol A (BPA). Reproductive toxicology, 24, 139-177.

Verstraeten, S. V., Aimo, L. & Oteiza, P. I. 2008. Aluminium and lead: molecular

mechanisms of brain toxicity. Archives of toxicology, 82, 789-802.

Völkel, W., Colnot, T., Csanády, G. A., Filser, J. G. & Dekant, W. 2002. Metabolism and

kinetics of bisphenol A in humans at low doses following oral administration.

Chemical research in toxicology, 15, 1281-1287.

Vom Saal, F. S. & Myers, J. P. 2008. Bisphenol A and risk of metabolic disorders. Jama,

300, 1353-1355.

Vom Saal, F. S. & Welshons, W. V. 2014. Evidence that bisphenol A (BPA) can be

accurately measured without contamination in human serum and urine, and that

BPA causes numerous hazards from multiple routes of exposure. Molecular and

cellular endocrinology, 398, 101-113.

Wada, K., Sakamoto, H., Nishikawa, K., Sakuma, S., Nakajima, A., Fujimoto, Y. &

Kamisaki, Y. 2007. Life style-related diseases of the digestive system: endocrine

disruptors stimulate lipid accumulation in target cells related to metabolic

syndrome. Journal of pharmacological sciences, 105, 133-137.

Wade, G. N. & Schneider, J. E. 1992. Metabolic fuels and reproduction in female

mammals. Neuroscience & Biobehavioral Reviews, 16, 235-272.

http://etd.uwc.ac.za/

102

Wadia, P. R., Cabaton, N. J., Borrero, M. D., Rubin, B. S., Sonnenschein, C., Shioda, T.

& Soto, A. M. 2013. Low-dose BPA exposure alters the mesenchymal and

epithelial transcriptomes of the mouse fetal mammary gland. PloS one, 8,

e63902.

Walsh, E. G. & Marshall, J. 1957. Physiology of the nervous system, Longmans, Green.

Walsh, R. N. & Cummins, R. A. 1976. The open-field test: a critical review.

Psychological bulletin, 83, 482.

Waring, R. & Harris, R. 2005. Endocrine disrupters: a human risk? Molecular and

cellular endocrinology, 244, 2-9.

Watkins, D. J., Ferguson, K. K., Del Toro, L. V. A., Alshawabkeh, A. N., Cordero, J. F.

& Meeker, J. D. 2015. Associations between urinary phenol and paraben

concentrations and markers of oxidative stress and inflammation among pregnant

women in Puerto Rico. International journal of hygiene and environmental

health, 218, 212-219.

Wells, P. G., Bhuller, Y., Chen, C. S., Jeng, W., Kasapinovic, S., Kennedy, J. C., Kim, P.

M., Laposa, R. R., Mccallum, G. P. & Nicol, C. J. 2005. Molecular and

biochemical mechanisms in teratogenesis involving reactive oxygen species.

Toxicology and applied pharmacology, 207, 354-366.

Willhite, C. C., Ball, G. L. & Mclellan, C. J. 2008. Derivation of a bisphenol A oral

reference dose (RfD) and drinking-water equivalent concentration. Journal of

Toxicology and Environmental Health, Part B, 11, 69-146.

Winters, B. D., Forwood, S. E., Cowell, R. A., Saksida, L. M. & Bussey, T. J. 2004.

Double dissociation between the effects of peri-postrhinal cortex and

hippocampal lesions on tests of object recognition and spatial memory:

heterogeneity of function within the temporal lobe. Journal of Neuroscience, 24,

5901-5908.

Wong, R. O. & Ghosh, A. 2002. Activity-dependent regulation of dendritic growth and

patterning. Nature reviews neuroscience, 3, 803.

Xiong, J. 2017. Toxicity of chlordane at early developmental stage of zebrafish. bioRxiv,

119248.

Xu, X.H., Wang, Y.M., Zhang, J., Luo, Q.Q., Ye, Y.P. and Ruan, Q., 2010. Perinatal

exposure to bisphenol‐A changes N‐methyl‐D‐aspartate receptor expression in

the hippocampus of male rat offspring. Environmental toxicology and chemistry,

29(1), 176-181.

Xu, X.-H., Zhang, J., Wang, Y.-M., Ye, Y.-P. & Luo, Q.-Q. 2010. Perinatal exposure to

bisphenol-A impairs learning-memory by concomitant down-regulation of N-

methyl-D-aspartate receptors of hippocampus in male offspring mice. Hormones

and behavior, 58, 326-333.

http://etd.uwc.ac.za/

103

Yamada, H., Furuta, I., Kato, E. H., Kataoka, S., Usuki, Y., Kobashi, G., Sata, F., Kishi,

R. & Fujimoto, S. 2002. Maternal serum and amniotic fluid bisphenol A

concentrations in the early second trimester. Reproductive toxicology, 16, 735-

739.

Yamamoto, S., Onoe, H., Tsukada, H. & Watanabe, Y. 2007. Effects of increased

endogenous serotonin on the in vivo binding of [11C] DASB to serotonin

transporters in conscious monkey brain. Synapse, 61, 724-731.

Ye, X., Kuklenyik, Z., Needham, L. L. & Calafat, A. M. 2005. Quantification of urinary

conjugates of bisphenol A, 2, 5-dichlorophenol, and 2-hydroxy-4-

methoxybenzophenone in humans by online solid phase extraction–high

performance liquid chromatography–tandem mass spectrometry. Analytical and

bioanalytical chemistry, 383, 638-644.

Ye, X., Kuklenyik, Z., Needham, L. L. & Calafat, A. M. 2006. Measuring environmental

phenols and chlorinated organic chemicals in breast milk using automated on-line

column-switching–high performance liquid chromatography–isotope dilution

tandem mass spectrometry. Journal of Chromatography B, 831, 110-115.

Yu, H. M., Yuan, T. M., Gu, W. Z. & Li, J. P. 2004. Expression of glial fibrillary acidic

protein in developing rat brain after intrauterine infection. Neuropathology, 24,

136-143.

Yun, S. & Hoyer, S. 2000. Effects of low-level lead on glycolytic enzymes and pyruvate

dehydrogenase of rat brain in vitro: relevance to sporadic Alzheimer's disease?

Journal of neural transmission, 107, 355-368.

Zama, A. M., Bhurke, A. & Uzumcu, M. 2016. Effects of Endocrine-disrupting

Chemicals on Female Reproductive Health. The Open Biotechnology Journal, 10.

Zha, X. & Xu, X. 2015. Dissecting the hypothalamic pathways that underlie innate

behaviors. Neuroscience bulletin, 31, 629-648.

Zhang, T., Xue, J., Gao, C.-Z., Qiu, R.-L., Li, Y.-X., Li, X., Huang, M.-Z. & Kannan, K.

2016. Urinary concentrations of bisphenols and their association with biomarkers

of oxidative stress in people living near e-waste recycling facilities in China.

Environmental science & technology, 50, 4045-4053.

Zoeller, R. T., Bergman, Å., Becher, G., Bjerregaard, P., Bornman, R., Brandt, I., Iguchi,

T., Jobling, S., Kidd, K. A. & Kortenkamp, A. 2014. A path forward in the debate

over health impacts of endocrine disrupting chemicals. Environmental Health,

13, 118.

Zsarnovszky, A., Le, H. H., Wang, H.-S. & Belcher, S. M. 2005. Ontogeny of rapid

estrogen-mediated extracellular signal-regulated kinase signaling in the rat

cerebellar cortex: potent nongenomic agonist and endocrine disrupting activity of

the xenoestrogen bisphenol A. Endocrinology, 146, 5388-5396.

Zuchero, J. B., & Barres, B. A. 2015. Glia in mammalian development and disease.

Development, 142(22), 3805-3809.

http://etd.uwc.ac.za/

104

Electronic

http://vanat.cvm.umn.edu/neurHistAtls/pages/cns9.html accessed 28th September 2018

http://what-when-how.com/neuroscience/the-cerebellum-motor-systems-part-1/ accessed 16th September 2018

https://medicine.academic.ru/135149/layers_of_cerebral_cortex accessed 15th

August 2018

https://www.ncbi.nlm.nih.gov/pubmedhealth/PMHT0024735/ accessed 8th August

2018

https://www.slideshare.net/amandahessborzacchini/hippocampus-13053807

accessed 30th September 2018

https://www.slideshare.net/drpraveenktripathi/limbic-system-brain accessed 7th

August 2018

https://www.wpclipart.com/medical/anatomy/brain/brain_3/four_lobes_of_the_ce

rebral_cortex.png.html accessed 12th September 2018

http://etd.uwc.ac.za/