Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing...

98
Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis Submitted in Partial Fulfillment of the Requirement for the Degree of Master of Sciences At University of Petra Faculty of Pharmacy and Medical Sciences Amman-Jordan December 13

Transcript of Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing...

Page 1: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Enhancing Oral Delivery of Glutathione Using Chitosan

Nanoparticles

By

Zainab A. Alobaidy

A Thesis Submitted in

Partial Fulfillment of the

Requirement for the Degree of

Master of Sciences

At

University of Petra

Faculty of Pharmacy and Medical Sciences

Amman-Jordan

December 13

Page 2: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

1

Chapter One

Introduction and Literature Review

Page 3: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

2

1 Introduction

In past years biotechnologically derived drugs, including peptides and proteins, have

become the focus of attention of drug development efforts (Tang et al., 2004). The

destiny of biotechnologically derived drugs after in vivo administration is usually

monitored by the standard pharmacokinetic factors namely; absorption, distribution,

metabolism, and elimination (ADME) (Barratt, 2003).

Having adequate bioavailability, which is defined as “the ratio of drug accumulation

at its site of action to the amount delivered to the body” is considered a significant

limitation in the use of protein or peptide biologics as treatments (Hamman, et al.,

2005).

Developing and finding a polypeptide drug delivery via the gastrointestinal tract has

been a continuous challenge attributed to their unfavorable physicochemical

properties, which includes poor absorption, enzymatic degradation, poor membrane

permeability in the gastrointestinal tract, unfolding of the protein, and their relatively

short half life post absorption (Vincent and Duncan, 2006). Beside a variety of

physiological and morphological factors in the gastrointestinal tract, proteolytic

enzymes in the gut lumen like pepsin, trypsin and chymotrypsin along with

proteolytic enzymes at the brush border membrane (endopeptidases); the bacterial gut

flora; the mucus layer and epithelial cell lining became barriers against protein or

peptide delivery (Lee et al., 1990).

Such barriers led to the necessity of developing special delivery systems that can

improve the oral bioavailability of therapeutic proteins from less than 1% to at least

30-50 % (Shaji and Patole, 2008). Despite various attempts, however, no clinically

efficient oral formulations have been developed yet (Park et al., 2011).

Page 4: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

3

Nanoparticles delivery systems have many advantages including the high in vivo

stability, long-term payload capacity release, and the ability of permeating through

small capillaries and cellular compartments (Yih and Al-Fandi, 2006). Moreover,

using nanosized particles could enhance the drug’s therapeutic index, modulate its

pharmacokinetic and biodistribution, and aid in forming sustained release reservoirs

(Herman et al., 1997).

The primary demands for a typical delivery of a nanoparticle include: be small sized

(50–200 nm), have high loading capacity, slow complex dissociation in vivo, and

target optimization to the desired tissue with limited absorption by other tissues. The

development of formulations that can integrate these characteristics while considering

the cost and the simplicity of the design is essential for effective delivery systems (De

Jong and Borm, 2008.‏).

1.1 Nanoparticles Delivery Systems

There has been a growing research attention in the field of drug delivery using

particulate delivery systems as carriers for small and large molecules. Particulate

systems, including nanoparticles, have been used successfully as a physical approach

to modify and ameliorate the pharmacokinetic and pharmacodynamic properties of

diverse types of drug molecules (Agnihotri, et al., 2004).

Different polymers have been used in the formulation of nanoparticles for drug

delivery research to improve therapeutic utility while minimizing side effects. The

drug could be dissolved, entrapped, encapsulated or attached to a nanoparticle matrix,

and relying on the method of preparation, nanoparticles, nanospheres or nanocapsules

could be gained (Agnihotri, et al., 2004).

Nanocapsules are systems in which the drug is reserved to a cavity encompassed by a

unique polymer membrane while nanospheres are matrix systems in which the drug is

Page 5: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

4

physically and equally dispersed (Mohanraj and Chen, 2006). In past years,

biodegradable polymeric nanoparticles, essentially those coated with hydrophilic

polymer, have been used as prospective drug delivery devices because of their ability

to circulate for a prolonged period time (Bhadra et al., 2002).

Despite these characteristics, nanoparticles do have some constraints for example,

their small size and large surface area can result in particles aggregation, making the

handling of nanoparticles difficult in liquid and dry forms; in addition to that, it can

also leads to restricted drug loading and burst release (Kommareddy et al., 2005).

Furthermore, some nanoparticles do not degrade or dissolve immediately in the body;

alternatively, they would accumulate in the biological systems and stay for prolonged

time, which could lead, to its interaction with the tissues (Grenha et al., 2005).

1.1.2 Nanoparticles Preparation Methods

Nanoparticles have been prepared mostly by three methods: dispersion of preformed

polymers; polymerization of monomers; and ionic gelation or coacervation of

hydrophilic polymers (Reverchon et al., 2006). Though, other methods such as

supercritical fluid technology and particle replication in non-wetting templates

(PRINT) are also mentioned in the literatures for nanoparticles production (Rolland et

al., 2005).

Dispersion of preformed polymers: Dispersion of preformed polymers is a prevalent

technique used to produce biodegradable nanoparticles from; poly (D, L-glycolide),

PLG; poly (D, L-lactide-co-glycolide) (PLGA), poly (lactic acid) (PLA) and poly

(cyanoacrylate) (PCA) (Kompella et al., 2001). This method can be used in several

ways including solvent evaporation spontaneous emulsification or solvent diffusion

techniques, where both can be used for hydrophobic or hydrophilic drugs (Kompella

et al., 2001).

Page 6: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

5

Polymerization method: In this method, monomers are polymerized to form

nanoparticles in an aqueous solution. Drug is inserted either by being dissolved in the

polymerization medium or by adsorption onto the nanoparticles after polymerization

is completed (Ravi et al., 2004).

Coacervation or ionic gelation method: Many research attempts have been

concentrating on using biodegradable hydrophilic polymers such as chitosan, gelatin

and sodium alginate. Among these a method developed for preparing hydrophilic

chitosan nanoparticles by ionic gelation (Calvo et al., 1997). This method involves a

mixture of two aqueous phases, one of which is the polymer (chitosan) and the other

is a polyanion (e.g. sodium tripolyphosphate). In this case, positively charged amino

group of chitosan interacts with the negatively charged tripolyphosphate to form

coacervates with a size in the range of nanometer (Calvo et al., 1997).

1.1.3 Drug Delivery Carriers

Although developing convenient carriers for peptides and proteins remains a

challenge; polymeric nanoparticles permit encapsulation of bioactive molecules and

protect them against enzymatic and hydrolytic degradation; for example, many

research attempts showed successful insulin delivery via the oral route by

incorporating insulin in nanosized systems (Elsayed et al., 2010).

One significant design to overcome the gastrointestinal barrier is to carry the drug in a

colloidal carrier system, which is capable of improving the interaction mechanisms

between the drug delivery system and the epithelial cells in the gastrointestinal tract

(Kramarenko et al., 2006).

Colloidal drug carrier systems, such as polymers, micellar solutions, vesicle and

liquid crystal dispersions, as well as nanoparticle dispersions consisting of small

particles of 10–400 nm diameters, show great promises as drug delivery systems

Page 7: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

6

(Kramarenko et al., 2006). In the development of these systems, the priority has been

focused to obtain formulations with optimized drug loading and release properties

with long shelf life and low toxicity. Nevertheless, during formulation the

incorporated drug participates in the microstructure of the system, and might even

influence the molecular interactions, especially if the drug possesses amphiphilic

and/or mesogenic properties (Kramarenko et al., 2006).

Chitosan has been the subject of interest to be utilized as a polymeric drug carrier

material in many designed dosage forms, due to its appealing properties such as

biocompatibility, biodegradability, low toxicity and its relatively low production cost

due to its abundant natural sources, however, one drawback of using chitosan in

modified release dosage forms for oral administration is the fast dissolution rates that

take place in the stomach (Hamman, 2010).

Since chitosan is positively charged at low pH values, it spontaneously associates with

negatively charged polyions in solutions to form polyelectrolyte complexes (PEC)

(Kean and Thanou, 2010). Such chitosan based polyelectrolyte complexes exhibit

favorable physicochemical properties with preservation of chitosan’s biocompatible

characteristics, therefore, these complexes are considered to be desirable candidate

excipient for the design of various kinds of dosage forms (Kean and Thanou, 2010).

Page 8: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

7

1.2 Chitin and Chitosan

Due to their salient proportion of nitrogen (6.89%), compared to artificial substituted

cellulose (1.25%), chitin and chitosan are of commercial concern leading to harness

chitin as a chelating agent. Natural polymers such as cellulose, chitin, chitosan and

their derivatives have much more biocompatibility and biodegradability than most of

the currently available synthetic polymers (Muzzarelli & Muzzarelli, 2005).

1.2.1 Chitin

Etymology the French word chitine, which mean, covering is the radix of the English

word "chitin"; first appeared in 1821 and derived from the Greek word χιτον (chiton)

(Chydyrov, 2007).

Existing in many places over the natural world, chitin is the basic constituent of

arthropods such as insects, crustaceans (e.g., crabs, lobsters and shrimps), the beaks

and internal shells of cephalopods, squid and octopuses, and in most invertebrates

(Fabritius et al., 2009). Structurally, chitin may be compared to the polysaccharide

cellulose in its high insolubility and its fiddling chemical reactivity. While

functionally, chitin may be compared to the protein keratin (Ravi, 2000).

Chitin in its unmodified form is a white, hard, translucent, pliable, and resilient

substance while in its "pure form", it has leathery texture. Albert Hofmann is the one

who solved the chitin structure in 1929 (Hofmann, 1929). Chemically, chitin is a

long-chain polymer of an N-acetylglucosamine (2-(acetylamino)-2-deoxy-D-glucose),

a derivative of glucose, and can be described as cellulose with one hydroxyl group on

each monomer substituted with an acetyl amine group.(Shur, 2007).

Chitin’s N-acetylglucosamine units form covalent β-1,4 linkages similar to the

linkages between glucose units forming cellulose. This allows increased hydrogen

Page 9: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

8

bonding between adjacent polymers and increases the strength of chitin-polymer

matrix. Chitin is frequently described as nitrogen rich polysaccharide (Jain et al.,

2007).

1.2.2 Chitosan

Chitosan, the N-deacetylated derivative of chitin, is a linear polysaccharide composed

of randomly distributed deacetylated units of β-(1-4)-linked D-glucosamine along

with acetylated units of N-acetyl-D-glucosamine. Chitosan is produced from the

treatment of chitin obtained from shrimps and other crustacean shells with the alkali

sodium hydroxide (Hadwiger, 2013‏).

The pKa value of chitosan is ~6.5, which allows protonation in acidic to neutral

solutions with a charge density that is dependent on pH and the % deacetylation value.

This causes chitosan to possess many attractive characteristics such as being soluble,

reactive and bioadhesive (Chenite et al., 2001).

Although chitosan is not approved by FDA for drug delivery; purified quantities of

chitosans can be commercially obtained for biomedical applications since chitosan

enhances the transport of polar drugs through epithelial surfaces (Chenite et al.,

2001).

1.2.3 Synthesis of Chitosan from Chitin

While chitin is obtained directly by physical and chemical treatment of crustacean's

shells, however, chitosan is synthesized from chitin. The deacetylation of chitin under

alkaline conditions is considered the common method for such synthesis, whereas the

most widely used alkali is sodium hydroxide (NaOH). The ambit of deacetylation is

mainly determined by the alkali concentration, temperature, and reaction’s time

(Khor, and Lim, 2003). Chitin is deacetylated in excess amount of a NaOH solution

Page 10: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

9

(40% wt/wt) as a reagent and water as a solvent at 1208 ˚C for 1–3 h in a closed

reactor and oven for heating. Such treatment produces 70% deacetylated chitosan

(Krajewska, 2004).

This reaction pathway, when it permitted to go to complete deacetylation, results with

up to 98% deacetylated chitosan (Hu et al., 2002). The deacetylation process and the

chemical modification of chitin and chitosan result in exposing the amino groups,

which makes the molecule soluble in a majority of diluted acids and improve their

solubility in organic solvents generally (Mourya & Inamdar, 2008). The structures of

cellulose, chitin and chitosan are shown in Figure 1.1.

Figure 1.1: The chemical structures of chitin, chitosan and cellulose.

Page 11: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

11

1.2.4 Chitosan Applications

As a natural polymer that has excellent properties such as biocompatibility,

biodegradability, non-toxicity, adsorption properties, etc., chitosan is recommended as

a suitable functional material in various fields. Much attention has been paid to

chitosan as a potential polysaccharide resource with many industrial and medical

applications (Gupta & Ravi, 2000). Chitosan is used currently in transdermal drug

delivery and it is a recommended carrier in drug delivery systems. Chitosan has an

antibacterial characteristic and found capable to inhibit the growth of Fusarium,

Alternaria, Escherichia coli and Helminthosporium (Wedmore et al., 2006). It is

likely that the cationic amine groups of chitosan binds to the microorganism’s anionic

cell membrane thus lead to inhibit their growth. Lately chitosan merit approval in the

United States and Europe for use in bandages and other hemostatic agents as it

displays a blood clotting properties (Wedmore et al., 2006).

Due to its polycationic nature, chitosan has been also used for clarification and

deacidification of fruit juices in food industry (Shahidi et al., 1999) and more recently

as a functional food for managing obesity (Qinna et al., 2013). As a natural

supplement, however, chitosan has been prescribed for weight loss and cholesterol

lowering for its ability to bind to free fatty acids and bile salts. In winemaking, it can

be used as a fining agent, also helping to prevent spoilage (Muzzarelli, 1996).

Chitosan has other commercial possible uses; in agriculture "for example" it was used

as a seed treatment and, biopesticide helping plants to fight off fungal infections. As a

polycationic carrier, chitosan is capable of binding to metal ions and "therefore" it has

been used in waste water purification (Crini, 2005). In paint industry, it was used as a

self-healing polyurethane paint coating (Rabea et al., 2003).

Page 12: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

11

1.2.5 Chitosan Properties:

1.2.5.1 Degree of Deacetylation (DDA)

DDA is the most influencing factor related to chitosan physicochemical

characteristics and consequently to its applications. DDA is defined as "the mole

fraction of deacetylated units in the polymer chain" (Hirai et al., 1991). The content of

free amino acids is identified by the degree of deacetylation. DDA is also use to

distinguish between chitosan and chitin. Conventionally, the presence of 50% amine

group is a boundary between chitin and chitosan, i.e. chitin has DDA value <50%, and

chitosan has DDA >50% (Lavertu et al., 2003).

In addition to its influence on the properties of chitosan, DDA controls the degree of

cross-linking in chitosan in the presence of any suitable cross-linker; and the

relationship between them is a direct proportional (Gupta & Jabrail, 2006).

1.2.5.2 Molecular Weight and Viscosity

The preparation method and the source of the used row material of chitosan influence

its molecular weight which is a very significant factor that impacts the

physicochemical properties and the antimicrobial activity of chitosan (Huang et al.,

2004).

Viscosity is another crucial parameter and considered to be one of the simplest factors

for measuring chitosan molecular weight using the Mark-Houwink equation (Imeri &

Knorr, 1988).

ŋ] = M

Where [ŋ] is the intrinsic viscosity, M is the molecular weight, K and ɑ are two

parameters that depend on the solvent and polymer.

Page 13: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

12

Chitosan solution viscosity is influenced by many factors such as the DDA, molecular

weight, concentration, ionic strength, pH, and temperature (Iium, 1998).

1.2.5.3 Solubility

An abundance number of factors affect the solvent and solution properties of chitosan.

Among these is the DDA, ionic concentration, pH, the distribution of the acetyl group,

and nature of the acid used in the protonation, and the condition of isolation and

drying of the polyschacaride (Ravi, 2000). Chitosan is a weak base that is insoluble in

water and organic solvents and is soluble in dilute aqueous acidic solutions (pH 2-

6.5). This is mainly due to the protonation of the amine group (-NH2) at C-2 D-

glucosamine units that converts chitosan into a polycation in acidic media (Marguerite

et al., 2006). Chitosan is poorly soluble in pH values above 7.0 due to inefficient

protonation and can be depolymrized in concentrated acidic solutions (Li et al., 2005).

The average value of DDA affects chitosan’s water solubility mostly by the

hydrophilic properties of the amine group, which is represented by the equilibrium

between the free amino and its ammonium salt form, and this is directly related to the

pKa value (Amiji, 1995).

Page 14: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

13

1.3 Reduced L-glutathione (GSH)

Reduced L-glutathione (GSH) is a significant, well-known, water-soluble tripeptide,

and the most plentiful low-molecular-weight thiol that exists in all eukaryotic cells,

including all mammalian tissues. GSH is composed of three amino acids namely, L-

glutamate, L- cysteine and glycine, with an unusual peptide linkage between the

amine group of cysteine and the carboxyl group of the glutamate side-chain. GSH

chemical nomenclature is N-(N-L-Ɣ-Glutamyl-L-cysteinyl) glycine (C₁₀H₁₇N₃O₆S)

and has a molecular weight of 307.3 g mol ¹ (Pompella, et al 2003)‏. The structure of

GSH is shown in Figure 1.2.

Figure 1.2 The structure of GSH.

1.3.1 Synthesis of Glutathione

GSH was first recognized in 1921 by Hopkins and his colleagues (Hopkins et al.,

1922). For commercial purposes, GSH is synthesized by the fermentation of yeast

followed by purification with manufacturing processes to get pure product free of

endotoxins (Pereira et al., 2007). In the body, it is endogenously synthesized in cells

from its constitutive amino acids (Allen and Bradley 2011). GSH also can be obtain

by dietary intake, its available in almost all foods, and normal diet would provide

Page 15: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

14

approximately 100-250 mg per day, many food such as asparagus, avocados, raw goat

milk and walnuts are rich sources of GSH (Pereira et al., 2007).

There are two consecutive ATP-dependent reactions that construct the pathway of

GSH biosynthesis. Initially, L-γ glutamylcysteine is synthesised from L-glutamate and

L-cysteine followed by the formation of GSH by the addition of glycine to the C-

terminal end of L-γ glutamylcysteine (Meister, 1988). It has been recognized that γ-

glutamylcysteine synthetase and glutathione synthetase (GS) are the catalyzers of this

reaction as illustrated in Figure 1.2 (Gandhi et al., 2006‏).

Figure 1.3 Synthesis of GSH in two steps enzymatic process in which: glutamine and

cysteine are covalently linked by the heterodimeric enzyme γ-glutamylcysteine

synthetase to form γ-glutamylcysteine, γ-glutamylcysteine is then bonded to glycine

to form a complete GSH molecule (Gandhi et al., 2006‏).

1.3.2 Glutathione Functions in Mammals

The most substantial group in GSH structure is the thiol group; which is a powerful

reducing agent making GSH the most important intracellular low molecule weight

Page 16: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

15

antioxidant in the body with up to millimolar concentrations in some tissues

(Anderson et al., 1998).

As a major endogenous antioxidant, GSH protects cells from toxins and pollutants

such as free radicals and peroxides while maintaining a normal balance between

oxidation and anti-oxidation (Townsend et al., 2003). Likewise, GSH leads an

important role in aging (Toba et al., 2000) as well as in the pathological path of many

diseases by upholding mitochondrial function and organizing many of the cell’s vital

functions such as the synthesis of proteins, the activation and regulation of enzymes

and the synthesis and repairing of DNA (Kerksick et al., 2005). Furthermore, GSH

was found capable of keeping vitamin C and vitamin E in their active state (Wu et al.,

2004). Therefore, GSH has essential roles in many systems and organs in the body;

especially the lungs, the immune system, the nervous system, and the gastrointestinal

tract (Gawryluk et al., 2011). In addition, GSH is essential for the activation of T-

lymphocytes and polymorphonuclear leukocytes as well as for cytokine production,

and therefore, for mounting successful immune responses when the host is

immunologically challenged (Townsend et al., 2003).

1.3.3 Glutathione Role in Mammals Health and Pathophysiology

As oxidative stress indicates the effects of endogenously produced toxins by normal

metabolism and exogenously from the environment, (e.g., free radicals, ROS/RNS

superoxide, hydrogen peroxide, and hydroxyl radical /reactive nitrogen species), it has

been evidenced that a lot of neurodegenerative diseases, such as Alzheimer’s disease

(AD) and Parkinson’s disease (PD), are started and develop due to the oxidative stress

(Dringen, et al., 2000). The piling up of surplus of ROS/RNS will oxidize the cell’s

components; including lipids, proteins, and DNA, causes functions disable and

eventually cell death. Furthermore, a significant number of studies on GSH and

Page 17: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

16

glutathione-dependent enzymes have demonstrated possible mechanisms and

consequences of the dysregulation of the GSH antioxidant network in

neurodegenerative diseases (Dringen, et al., 2000).

GSH along with the glutathione-dependent enzymes (glutathione peroxidases,

glutathione transferases, glutaredoxins, and glutathione reductases) and redox

regulation are all considered to be a main line of antioxidant to face neuron

degeneration (Miller et al., 2009).

Since GSH is recognized as the master free radical scavenger in the brain, its low

levels elevate cellular instability towards oxidative stress; marked by the

accumulation of reactive oxygen species and this explains how GSH deficiency is

correlated to the pathophysiology of bipolar disorder (BD), major depressive disorder

(MDD), and schizophrenia (SCZ) (Melo et al., 2011).

Much clinical diseases are associated with oxidative stress such as prostate and other

cancers, cataracts, chronic obstructive pulmonary disease, asthma, radiation

poisoning, malnutrition states, arduous physical stress, and aging. Nevertheless, GSH

deficiency is not limited to these conditions, it has been also associated with many

other conditions including HIV/AIDS, chemical and infectious hepatitis and myalgic

encephalomyelitis chronic fatigue syndrome ME/CFS (Floyd, 1999).

1.3.4 Glutathione as a Possible Drug

GSH critical roles in mammal's pathophysiology led to considering it as a possible

drug. Searching for a potential route of administration is still undergoing with a focus

its oral bioavailability. From previously published research attempts, it is obvious that

delivering oral GSH without a protective formulation to the blood stream seems

Page 18: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

17

difficult. Conversely, the bioavailability of intravenous dosing of GSH appears well

established (Aebi et al., 1991).

1.3.5 Previous Studies of Parenteral Administered Glutathione

A randomized double blinded placebo- controlled trial on 50 gastric cancer patients

was performed in order to find out whether the addition of GSH to the IV

administration of cisplatin can reduce neurotoxicity. The administered dose of GSH

was 1.5 g/M2 in 100 mL of normal saline over a 15 minutes period prior to cisplastin

administration, at day 2 to day 5, the patients received 600 mg intramuscular injection

of glutathione. At week 9, patients in the GSH group showed no neuropathy, while 16

of the 25 patients in the control group did (Cascinu et al., 1995). Later an analogous

randomized, double-blinded, placebo-controlled trial on colorectal cancer patients was

performed in order to find out whether the addition of GSH to the IV administered

oxaliplatin would reduce neurotoxicity; a significant reduction of neuropathy in the

GSH group was noted (Cascinu et al., 2002).

The administration of 2500 mg of GSH IV was also effective in reducing

neurotoxicity in 79 patients with ovarian cancer treated with cisplastin and

cyclophosphamide (Dire et al., 1993). In another study, the IV GSH treatment also

increased measures of hemoglobin, red blood cells, and hematocrit in uremic anemia

patients (Pirovano et al., 1992). Moreover, a randomized double-blinded placebo-

controlled trial done on patients suffering from peripheral artery disease indicated that

IV GSH significantly improved the walking distance and microcirculatory and

microcirculatory parameters (Arosio et al., 2002).

In another study, IV GSH administration improved total glucose uptake in normal and

diabetic patients (Mattia et al., 1998). Furthermore, when IV GSH was given to 28

patients undergoing hemodialysis, it was found that 60% of the patients showed

Page 19: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

18

hemoglobin and hematocrit improvement after about nine months (Usberti et al.,

1997).

A 600 mg twice daily of IV GSH for 30 days were given in another study to untreated

parkinson’s disease patients, a 42% disability decline was observed (Sechi et al.,

1996).

Finally, Aebi et al. have shown a raised the levels of plasma and urinary total GSH by

IV of 2 g/ml of GSH, however, the overall half-life of GSH was very short at 14.1 ±

9.2 min. (Aebi et al., 1991).

1.3.6 Previous Studies of Orally Administered Glutathione

Witschi and coworkers pointed out in a study of acute oral administration of a very

large dose of oral GSH (3 g) that "it is not possible to increase circulating GSH to a

clinically beneficial extent by the oral administration of a single dose of 3 g of

glutathione"(Witschi et al., 1992).

In a clinical trial to determine the efficacy of two commonly used GSH supplements

in subjects diagnosed with autism in increasing blood GSH levels, Janet et al. were

unclear if the increase could consider of clinical significance due to the small size of

changes in GSH levels (Janet et al., 2011).

Fukagawa and coworkers found that after the administration of glutathione, the

cysteine levels in blood increased by 61%, which equalize the rate of exogenous GSH

infusion. Nevertheless, they supposed that the increase in the sulfate, cysteine, and

taurine might be due to the breakdown of GSH and not a true improvement in the

transsulfuration pathway (Fukagawa et al., 1996).

Page 20: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

19

Recently, Jason et al. indicated that with all the biochemical plausibility, dose, and

timing optimization, and appropriate choice of outcome measures, after 4 weeks of

oral GSH supplementation in healthy adults there were no remarkable changes

observed in oxidative stress biomarkers or erythrocyte GSH concentrations (Jason et

al., 2011).

Hagen et al. reported that, plasma GSH levels, and ultimately the oxidative stress, was

improved by direct supplementation of oral GSH in rats (Hagen et al., 1990).

All of the previous studies showed almost certainly that it would be difficult to

administer GSH orally without a suitable delivery system.

1.3.7 Glutathione Risks and Side Effects

Depending on the available literature and previous studies, there seems no risk or side

effects observed from the oral administration of GSH except of some temporary

increase in flatulence after consistent use (Wu et al., 2004).

Another side effect that can be considered as a benefit is the lightening of the skin,

and many enjoying this side effect as it the safest alternative in compare to harsh

ingredients of skin whitening products such as hydroquinone and mercury, which are

hazardous and toxic to the body (Wu et al., 2004).

1.4 Reduced Glutathione-Chitosan Polyelectrolyte Complex (GC-PEC) and

Glutathione Loaded Nanoparticles (GRM-LN)

Chitosan is known to have significant ability to form complexes as a PECs (Yancheva

et al., 2007). In an aqueous condition, it is able to form electrostatic complexes with

acidic molecules. This includes the formation of an ion pair between the free

ammonium groups of low molecular weight chitosan and the carboxylate groups of

the acidic molecule. With regard to GSH; the presence of the two carboxylic acid

Page 21: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

21

groups, the thiol group, and the amino group, it has sixteen various charged species

with net charges ranging from to 3 at various pKa values. Thence, at pH range 5-

7, solution of GSH, consists entirely of ( , , 0, - ), (symbols in parentheses identify

the net charge ]from left to right[ of the terminal amino group, the glutamyl

carboxylate group, the thiol group of the cystine residue, and the terminal glycyl

carboxylate group) so the net charge is 1 (Wu, et al, 2009‏). Thus at pH 5.5 and in

liquid state, the ammonium group ( NH3⁺) of chitosan can interact with either the

glutamyl carboxylate group or the terminal glycyl carboxylate group ( ) of

GSH to form polyelectrolyte complex. Figure 3.1 describes the interaction between

the ammonium group ( NH₃⁺) of the protonated chitosan with either the glutamyl

carboxylate group or the terminal glycyl carboxylate group ( ) of GSH to form

the PEC.

Figure 1.4 epresentation of the ionic interaction in a ueous state between the

ammonium group ( NH₃⁺) of chitosan and carboxylate group ( ) of GSH at

pH 5.5 due to the ionization of the two molecules at this pH.

After the addition of PEC to an oily phase (oleic acid), nanoparticles are constructed

by the self-assembly or self-organization of surfactants (labrasol®

(PEG 8

caprylic/capric glycerides) and cosurfactant Plurol oleique® (polyglyceryl-6-

Page 22: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

21

dioleate)), that occur in the system, where the hydrophobic tails should form the core

of the aggregate, and the hydrophilic heads must be in contact with the surrounding

liquid. Figure 1.5.

Figure 1.5 Diagram of a nanoparticle of water in oil system. Water-soluble tails

project into the water, while the oil-soluble ends remain in contact with the oil phase.

1.5 Aims of the Current Research

In this research, two main aims were predetermined. The first aim associated with

studying and demonstrating the ion pair interaction between the ammonium group (

NH3⁺) of chitosan and the carboxylate group ( ) of GSH and to characterize

the formed PEC. For this purpose, three measuring techniques were used; FT-IR

spectroscopy, differential scanning calorimetry (DSC), and nuclear magnetic

resonance spectroscopy (1H NMR). On the other hand, the second aim was directed

towards delivering intact GSH to the blood stream using a nanosized particles loaded

with glutathione-chitosan PEC as an attempt to enhance the oral bioavailability of

glutathione. To achieve this aim, the nanoparticle were characterized by dynamic light

scattering for determination of particle size and zeta potential. Later, the

Page 23: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter One: Introduction

22

bioavailability of the oral formula was determined in vivo. Such assessment was

achieved utilizing liquid chromatography–mass spectrometry (HPLC-MS) for

analyzing and measuring GSH concentrations in the collected rat serum samples.

Page 24: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

Chapter Two

Experimental

Page 25: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

2 Materials and Methods

2.1. Materials

L-reduced Glutathione (98%), formic acid advanced gradient HPLC grade and sodium

hydroxide were obtained from Acros Organics, Geel, Belgium. Ellman’s Reagent (5,

5-dithiobis (2-nitrobenzoic acid) was purchased from Sigma Chemical Co., St. Louis,

MO. Free base chitosan with molecular weights of 1.3, 8, 13, 18, 30 kDa, were

kindely obtained from the Jordanian Pharmaceutical Manufacturing Co. (JPM), Naor,

Jordan. Highly pure oleic acid was purchased from Merck KGaA, Darmstadt,

Germany. Surfactant Labrasol® (PEG 8 caprylic/capric glycerides) and cosurfactant

Plurol oleique® (polyglyceryl-6-dioleate) were obtained from Gattefosse, Lyon,

France. Methanol advanced gradient HPLC grade and acetonitrile (TCA) were

purchased from Fisher Scientific, Pittsburgh, PA, USA. All chemicals were used as

received without further purification, water was deionized.

2.2 Preparation and Characterization of the GSH and Chitosan Polyelectrolyte

Complex (GC-PEC)

Five grades of chitosans with molecular weights of 1.3, 8, 13, 18, 30 kDa all having

99% DDA were used to prepare GC-PEC by dissolving 100 mg of GSH in deionized

water. The pH of GSH solution was adjusted to 5.5 using NaOH (2 M) added

dropwise under stirring, the volume was then completed up to1 mL. On the other

hand, chitosan solution was prepared by dissolving 71.5 mg of chitosan in deionized

water under magnetic stirring followed by adjusting the pH to 5.5 using NaOH (2 M)

added dropwise under stirring, the volume was then complete up to1 mL. Finally, the

two solutions were mixed together with gentle shaking at 1:1 ratio (v/v). For the

characterization of the prepared GC-PEC, all samples were freeze-dried into powder

(Hetopower dry PL 9000-90 HSC, Thermo Electron, Czech Republic). The physical

mixture of GSH and chitosan was prepared by mixing the freeze dried samples with

Page 26: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

1:1 ratio (wt/wt).

2.2.1 Fourier Transform Infrared Spectroscopy (FT-IR)

FT-IR analysis were recorded for all samples using Nicolet Avatar 360 FT-IR ESP

spectrometer (Nicolet, USA) connected to omnic software. Dried sample (5-10 mg)

were mixed and pressed to result in homogenous sample/KBr disk with 1:6 ratio. All

spectra were recorded at ambient temperature in the wave number region between

an m n all ases - R s e t a e e e e a m lat n at

least s ans t a es l t n m ¹.

2.2.2 Differential Scanning Calorimetry (DSC)

DSC thermograms were generated using thermal analysis system consisting of

STARe Software TGA/STDTA1600 oC DSC 822e/400 modular unit, DSC 827e High

Pressure modular unit (Mettler Toledo, 2915 Argentia Road, Unit 6, Mississauga L5N

8G6, Canada).

For analysis, 2 to 5 mg of each sample was placed in an aluminum pan. An empty

sample holder was used as a reference and the runs were performed by heating the

sam les m t 5 ˚C at a ate ˚C/m n A al at n as ne s ng t e

melting point of highly pure indium metal (99.99%) in relation to temperature. Indium

melting temperature is 56 6 ˚C

2.2.3 Hydrogen-1 Nuclear Magnetic Resonance Determinations (¹H-NMR)

¹H NMR spectra were generated on a Bruker ultrashield 500 MHz spectrometer

(Karlsruhe, Germany). Samples (15 mg) were dissolved in deuterium oxide (D₂O) in

5 mm NMR tubes followed by mixing the tubes using a vortex mixer.

Page 27: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

2.3 Preparation and Characterization of Glutathione Loaded Nanoparticles

Reversed Micelles (GLN-RM)

The prepared oral delivery system for glutathione namely, glutathione loaded

nanoparticles reversed micelles (GLN-RM), was developed by The Jordanian

Pharmaceutical Manufacturing Co. Plc, Amman, Jordan. Such a formula was

consisted of an oily phase and an aqueous phase. For preparing 20g of the oily phase,

2g of labrasol® (PEG 8 caprylic/capric glycerider) was weighted in a clean erlenmeyer

flask and mixed thoroughly with 2g of plurol oleique® (polyglyceryl-6-dioleate) by

vortexing. This produced a surfactant-cosurfactant (SCOS) mixture at a ratio of 1:1

(w/w). Later, accurately weighed amount of 16 g of oleic acid was then added to the

above mixture and mixed thoroughly by vortexing. The reverse micelle system was

finally produced by the addition of 2% (400 µL) of the previously prepared GC-PEC

(see section 2.3) to the oily phase drop wisely and vortexed for 30 seconds.

2.3.1 Physical Appearance of the Prepared Glutatione Loaded Nanoparticles

Reversed Micelles (GLN-RM)

All GLN-RM were placed under observation for 10 hr. The occurrence (presence or

absence) of precipitation of the GC-PEC were recorded every 30 min for each

preparation.

2.3.2 Dynamic Light Scattering (DLS) for Particle Size and Zeta Potential

Determination

The hydrodynamic diameter of the prepared nanoparticles was measured by photon

correlation spectroscopy using a Zetasizer (Model Nano- ZS, Malven Instrument,

Malven UK) at 5 ˚C t an angle ete t n eq als t 9 ˚ The samples were

measured in folded capillary cells integrated with gold electrodes. Three

measurements were conducted, and the number of runs in each measurement was

automatically determined by the software.

Page 28: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

2.3.3 Liquid Chromatography–Mass Spectrometry (LC-MS)

An API (Thermo, San Jose, CA) LC-MS was used to measure serum content of

glutathione. The components of the system were; a degasser (Agilent 1260), a solvent

delivery pump system (Agilent 1200), an autosampler (Agilent 1200), a thermostat

column compartment (Agilent 1200), a Mass Spectrometer (API 3200), ACE C18 (50

x 2.1 mm), 5 µm, simultaneously monitor ions with m/z of 273.1, 109 which

correspond to the protonated molecular ions of GSH and citalopram. For the analysis

of in vivo gluthathione levels in rats, a valid method published by Xiangming and

coworkers was adapted with some modifications (Xiangming et al., 2002). These

modifications included using formic acid and methanol as a mobile phase while using

citalopram as an internal standard with a retention time of around 1.8 min.

2.3.3.1 Solutions

Stock solutions of GSH (20 mg/mL) and of citalopram (20 mg/mL), as an internal standard

(IS), were freshly prepared in deionized water and stored immediately at - ˚C Standard

solutions were prepared from stock solutions by dilution with deionized water (Tables 2.1,

2.2). Citalopram solution (2 µg/mL) was prepared in TCA (precipitating agent) and a saturated

solution of Ellman’s eagent ( mM) as e a e ss lv ng 39 6 mg t e eagent n

10 mL of deionized water (Xiangming et al., 2002).

Page 29: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

Table 2.1 Preparation of GSH standard curve serial dilution and spiked serum from stock

solution of 20 mg/mL.

Solution No.

Volume

taken (mL)

from stock

solution

Total volume

(mL)

GSH concentration

(µg/mL)

S1 0.005 10 1.0

S2 0.010 10 2.0

S3 0.025 10 5.0

S4 0.100 10 20.0

S5 0.300 10 60.0

S6 0.600 10 021.1

S7 1.000 10 211.1

Samples of STD curve in serum were prepared by spiking 500 µL from serial solution into 9.5

mL of serum, using seven concentrations, not including zero to attain STD concentrations of 1,

2, 5, 20, 60, 120 and 200 µg /mL for GSH in serum.

Each concentration of the serum sample was divided to 0.050 mL in 1.5 mL eppendorf tube and

kept at (-20 ˚C) stan a sam les e e g ven a l t get e t t e q al t nt l sam les

Table 2.2 Preparation of GSH quality control (QC) serial dilution and spiked serum from stock

solution of 500 µg/mL.

Solution No

Volume

taken (mL)

from stock

solution

Total volume

(mL)

GSH concentration

(µg/mL)

S 8 0.061 10 3

S 9 2.000 10 001

S 10 3.211 10 060

Page 30: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

Samples of QC in serum were prepared by spiking 500 µL from serial solution into 9.5 mL of

serum, using three concentrations to attain QC concentrations of 3, 100 and 160 µg/mL for GSH

in serum. Each concentration of the serum sample was divided to 0.05 mL in 1.5 mL eppendorf

tube and kept at -20 ˚C stan a sam les were given together with the quality control samples.

Page 31: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

22

2.3.3.2 Chromatographic Conditions

Table 2.3 Summery table of the chromatographic conditions.

Column oven

temperature

Autosampler

temperature

Autosampler

injection volume

Pump flow

rate HPLC conditions

5˚C ˚C 5 Ul 1.0 mL/ min

0.1%

Formic Acid Methanol

Total

Time A% B%

0 100 0

0.5 100 0

0.51 0 100

1.5 0 100

1.51 100 0

3 100 0

Mobile phase Chromatography

ACE C18 Column (50 X 2.1 mm), 5µ Column type

Citalopram (IS) Glutathione Expected

retention times

(minutes)

1.8 1.5

2.4 Analytical Method Validation

The validation of the method was performed in three separate days. In each day, seven

standard calibration levels (not including zero) were prepared. The validation

parameters should not exceed the limits by the FDA Guidance for Industry.

2.4.1 Standard Calibration Curve and Linearity

The linearity was measured by plotting the peak area ratio (analyte peak area/IS peak

area) versus nominal concentrations. The concentrations of all samples were measured

by fitting the obtained data (area ratio data) to a straight line equation. Three

Page 32: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

23

calibration curves consisting of a blank, zero and seven non-zero standards prepared

in rat serum for each analyte were prepared.

The concentrations of calibration standards covered the range from lower limit of

quantification (LLOQ) to the highest expected concentration. The linearity was

evaluated by the linear regression (correlation coefficient, R²).

2.4.2 Intra-Day Accuracy and Precision

Intra-day accuracy and precision was measured by analyzing six replicates for each of

QC level (Low, Mid and High) within the batch for GSH.

2.4.3 Recovery

Serum samples containing concentration of QC Low, QC Mid and QC High analytes

were prepared in triplicate. The absolute peak areas obtained from injections of the

prepared serum standards were compared to the absolute peak areas of equivalent

standards, which were prepared by spiking GSH after precipitation, and same

treatment for the IS. The recovery extent of analytes and IS should be consistent,

precise, and reproducible.

Recovery was performed by preparing triplicates from each QC level of serum and

triplicates from each QC level prepared in the mobile phase (FDA, 2001).

2.5 In vivo Experimental Protocol

Adult male Sprague Dawley (S.D) rats (150-300 mg) were accommodated at the

Animal House of University of Petra, Amman, Jordan under a photoperiod schedule

of 12 hr light /12 hr dark cycles. The rats received standard chow and tap water three

weeks prior to experiments. All experiments on animals were carried out in

accordance with the guidelines of the Federation of European Laboratory Animal

Science Associations (FELASA). The study protocol was approved by the Committee

of High Education Council, Faculty of Pharmacy and Medical Sciences, University of

Petra, Amman, Jordan.

Rats were randomly assigned to five groups: a control (sham-treatment) group (n=4),

Page 33: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Two: Experimental

23

subcutaneously injected group with GSH at a 10 mg/kg dose (n=8), a group

administered a dose of 2.5 mg/kg of GLN-RM orally (n=9), a group administered a

dose of 10 mg/kg of GLN-RM orally (n=8), and group administered a dose of 20

mg/kg of GLN-RM orally (n=8). For the control group, the measurements were

repeated at 6 time intervals in order to established the basal GSH serum concentration

to a maximum of 4 hr. While for subcutaneously-injected rates, the measurements

were repeated at 12 time intervals, in order to established GSH serum concentration to

a maximum of 4 hr. As for the oral groups, the measurements were repeated at 8 time

intervals in order to established GSH serum concentration to a maximum of 8 hr.

2.6 Preparation and Derivatization of Biological Samples

Blood samples were collected from the tail of the rats in 1.5 mL eppendorf tube and

centrifuged for 1 min then serum (supernatant) were collected in 1.5 mL eppendorf and

kept at (- ˚C), and used for analysis. To 50 μL of serum sample, 3 μL Ellman’s

reagent (10 mM) were added along with 500 μL of the internal standard (2 µg/mL

citalopram in TCA) in a 1.50 mL eppendorf tube. The mixture was then vortexed for 30

seconds, then centrifuged for 15 min at 14000 rpm. (Eppendorf Centrifuge, model

5417C, Hamburg, Germany). The supernatants were transferred into the autosampler

vials.

Page 34: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Chapter Three

Results

Page 35: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3 Results

3.1 Preparation and Characterization of Glutathione-Chitosan Polyelectrolyte

Complex (GC-PEC)

T NH₃⁺) of the protonated chitosan

with either the glutamyl carboxylate gr

) of GSH to form the PEC was confirmed by characterizing the formed PEC

using different techniques as described in the below sections.

3.2 Characterization of the Polyelectrolyte Complex

3.2.1 Fourier Transform Infrared Spectroscopy (FT-IR) Spectroscopy Studies

Figure 3.1 displays the FT-IR spectrum of GSH, chitosan, GSH-chitosan physical

mixture (GC-physical mixture), and of GC-PEC. GSH FT-IR spectrum showed a

distinguish peak at 1710 ¹ due to the carbonyl groups (C=O). Chitosan spectrum

showed a characteristic band at 1250–1020 ¹ due to C-N stretching (Spectroscopy

Tutorial).

The spectra of GC-PEC are obviously distinguished from that of GC-physical

mixture ¹ in the spectrum of GSH, that

corresponds to the carbonyl groups, was shifted and the characteristic moderate band

at 1250–1020 ¹ in chitosan spectrum appeared sharper in GC-PEC spectrum.

Page 36: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Tran

smis

sion

/%

d

c

b

a

Wave number cm¯¹

Figure 3.1 Fourier Transform Infrared Spectroscopy (FTIR) of (a) GSH. (b) chitosan

(c) GC-physical mixture and (d) GC-PEC. The spectra show the changes at the band

at 1 – ¹ that corresponds to the stretching of C-N in chitosan, and the

changes at the band at ¹ that corresponds to the carbonyl groups (C=O) in

GSH. The spectra were measured by KBr-supported samples and over the frequency

rang of - ¹.

Page 37: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.2.2 Differential Scanning Calorimetry (DSC)

Figure 3.2 displays the thermograms of GSH, chitosan, GC-physical mixture and of

GC-PEC. In the thermogram of chitosan, two decomposition steps were observed. At

first, a broad endothermic peak appear between 30 and 130 ˚C; which can be, ascribed

to the evaporation of adsorbed water and water ligands. Later, chitosan exhibited no

endothermic peak owing to its amorphous form. The second decomposition step

occurred in the range of 200-25 ˚ .

The DSC curve of GSH has one sharp endothermic peak that corresponds to GSH

melting with peak temperature of 200-205 ˚ . The thermal analysis of GC-physical

mixture shows the peak of GSH melting due to the free GSH with few degree shifting

to 189 ˚C with changes in the decomposition steps of chitosan, which shows some

decrease in the sharpness.

With regard to GC-PEC, the thermal analysis showed a shift in GSH melting peak up

to 215 ˚C and strong remarkable reduction in the sharpness of the peak was observed.

This was accompanied with changes in the shape of the decomposition peak of

chitosan where there was no sharp exothermic peak in the case of complex, instead,

however, a gradual thermal degradation behavior was observed.

Page 38: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Temperature (˚C)

a

b

c

d

Hea

t fl

ow

Figure 3.2 Differential scanning calorimetry (DSC) of (a) GSH, (b), chitosan, (c) GC-

physical mixture and (d) GC-PEC, using an aluminum pan as sample holder with 2-5

mg of sample. An empty sample holder was used as reference. The runs were

performed by heating the samples from 40 up to 250 ˚C at a rate of 10 ˚C/min. The

spectra shows a shift in the GSH melting peak up to 215 ˚C and strong remarkable

reduction in the sharpness of s ’s peak with changes in the shape of the

decomposition.

Page 39: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.2.3 Nuclear Magnetic Resonance Spectroscopy ¹H-NMR Determination

Figure 3.3 shows the chemical structure of chitosan and GSH along with labeled

carbons indicating each proton involved in the ionic interaction. The collection of

peaks in the 3.18-4.88 ppm region as manifested get along with chitosan backbone

protons (i.e, single hydrogen atoms attached to Ca-Ce carbon atoms and the two

hydrogen atoms attached to Cf carbon atom). The highest proton is the hydrogen at

Cb (3.18 ppm). There are no peaks at the range corresponding to the protons of

acetyle group; hence, chitosan was described as fully deacetylated.

It is worth mentioning that protons of (-OH, -NH2, -SH) have no characteristic

chemical shift due to the disappearance of the peak when formation ed with D2O, as

deuterium will replace a protium atom. This method generally referred to as a D2O

shake. Acidic protons may also be penned when a solvent containing acidic deuterium

ions are used (e.g. methanol-d4) (Gottlieb et al., 1997), this method can not be utilized

due to the fact that the pH had to be maintained at 5.5.

In the case of GSH, however, ¹H NMR spectrum peak at 3.70 usually corresponds to

(Cb,i) (Sharad et al., 2000).

In the ¹H-NMR spectrum of GC-PEC, it can be noticed that the peak at (3.7 ppm)

corresponding to the Cb,i of GSH was shifted down filed with splitting the peak, as

well as the peak in chitosan ¹H NMR spectrum at (3.18 ppm) that corresponds to Cb

that was shifted up filed.

Page 40: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Ihitosan

Figure 3.3Shows the ¹H NMR spectra of (a) GSH (b) chitosan and(c) GC-PEC. The

spectra were recorded on a Bruker ultrashield 500 MHz spectrometer, sample (15 mg)

were dissolved in D₂O.

Page 41: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

3.3 Preparation and Characterization of the GSH Loaded Nanoparticles (GLN-

RM)

The nanoparticles was prepared by the addition of 2% of the GC-PEC to the oily

phase drop wisely. The unprotonated NH₂ in chitosan bind with oleic acid to form the

nanoparticles.

3.3.1 Dynamic Light Scattering (DLS) Particle Size Determination

The aggregation diameter of the GSH loaded nanoparticles prepared with different

low molecular weight chitosan (1.3, 8, 13, 18, 30 kDa) are shown in Table 3.1.

Each GLN-RM was measure in ten replicates to exclude the environmental factors,

the results of DLC gave the desired outcome for all the formulas except the 1.3 kDa

GLN-RM which, as expected from the previous studies on insulin, gave larger particle

size.

Page 42: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

Table 3.1 (a) The aggregation diameter of the GSH loaded nanoparticles, prepared

with different low molecular weight chitosan (1.3, 8, 13, 18, 30 kDa).

(b) Dynamic Light Scattering DLS plot of 30 kDa chitosan GC-PEC. DLS was done

by photon correlation spectroscopy at 25 ˚ A s DLS f s

existence of a single population of scatters.

a

chitosan molecular weight

(kDa) Aggregation diameter of (nm)

1.3 396±69.2

8 70.3±4.2

13 87.5±10.8

18 93.4±5.6

30 94±2.16

b

3.3.2 Physical Appearance of the Prepared Glutatione Loaded Nanoparticles

Reversed Micelles (GLN-RM)

By putting all the GLN-RM under observation it was noticed that the GLN-RM

prepared by 1.3, 8, and 13 kDa chitosan recorded shorter time to start precipitation

≈3 ) GLN-RM prepared using 18 and 30 kDa chitosan, which showed

relatively long time to start precipitation (up to 7 hr). After repetition with large

concentrations the 30 kDa GLN-RM chitosan showed consistency in comparing to

other formulas with no aggregation.

Based on the above 30 kDa GLN-RM was used for pharmacological activity

evaluation of GSH loaded nanoparticles oral formula.

Page 43: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

3.3.3 Zeta Potential Determination

Zeta potential of 30 kDa GLN-RM was determined to be 277.5 ±1.42, while the zeta

potential of placebo formula of chitosan alone is 56.83 ± 0.13(Elsayed, et al., 2011).

The significance of zeta potential is that its value can be related to the stability of

colloidal dispersions. Stability behavior of the colloid: from 0 to ±5, rapid coagulation

or flocculation, from ±10 to ±30, incipient instability, from ±30 to ±40, moderate

stability, from ±40 to ±60, good stability, more than ±61, excellent stability (Kirby

.(‏2010

Page 44: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.4 In vivo Evaluation of GLN-RM In Rats

For the pharmacological activity evaluation of GLN-RM and to see the extent of its

impact on GSH concentration in rats the valid method published by Xiangming and

coworkers were used, standard samples were given with every analysis together with

the quality control samples. Calibration curve of spiked serum samples with the

regression equation and their respective correlation coefficient (R²) of GSH is shown

in figure 3.4 (Xiangming et al., 2002). The relative peak area (ratio of peak area of the

drug to the peak area of the its IS) was yielded a linear correlation.

Y= 0.0545 x + 0.0563 (R² =0.9995)

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 50 100 150 200 250

GSH

AU

C /

IS A

UC

GSH concentration ug/mL

y = 0.0454 x + 0.0563r² = 0.9995

Figure 3.4 Calibration curve of GSH with citalopram as IS.

Page 45: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.4.1 Glutathione Base line Determinations and Estimation the Effect of GLN-

RM on Glutathione Serum Concentration

In order to evaluate the pharmacological activity of GLN-RM, rats serum GSH

baseline was established by measuring the concentration of GSH serum of fasting rats

(n=8). The measurements were repeated at 6 time intervals in order to established

GSH serum concentration to a maximum of 4 hours.

The serum concentration of SC administered GSH was evaluated on rates for sample

size of 8. For the SC experiment, 11 blood samples were withdrawn at different time

intervals.(0., 5., 10., 15., 20., 25., 30., 45 min., 1, 2, 3,4 hr.)

Then the serum concentration of GSH was evaluated on rats after administration of

2.5, 10, 20 mg/kg dose of GLN-RM orally on sample size of 8. The blood samples

were withdrawn at different time intervals (0, 30 min., 1, 2, 3, 4, 6, 8hr.). In order to

evaluate the effect of GLN-RM on GSH serum concentration to a maximum of 8

hours. Figures 3.5, 3.6 and 3.7.

Page 46: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

0

50

100

150

200

250

0 1 2 3 4 5

Co

nce

ntr

atio

n (u

g/m

l)

Time (hr)

SC 10mg/kg

Figure 3.5 Shows GSH serum concentrations after administration of subcutaneous

GSH of 10 mg/kg dose. Each point represents the mean ± SEM,

n=8.

0

50

100

150

200

250

0 1 2 3 4 5 6 7 8 9

Co

nce

ntr

atio

n (u

g/m

L)

Time (hr)

2.5 mg/kg oral

10 mg/kg oral

20 mg/kg oral

Figure 3.6 Shows GSH serum concentration after administration of 2.5, 10 and 20

mg/kg dose of GLN-RM orally. Each point represents the mean ± SEM, n=8.

Page 47: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

0

50

100

150

200

250

0 1 2 3 4 5 6 7 8 9

Seru

m G

SH (µ

g/m

L)

Time (hr)

2.5 mg/kg oral

10 mg/kg oral

20 mg/kg oral

10 mg/kg SC

placebo

Figure 3.7 Rats serum GSH baseline of fasting rats; the measurements were repeated

at 6 time intervals to a maximum of 4 hours. GSH serum concentration after

administration of 2.5, 10, 20 mg/kg dose of GLN-RM orally and 10 mg/kg dose

subcutaneously. Each point represents the mean ± SEM, n=8.

Page 48: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Pharmacokinetic data of orally and subcutaneously administered GSH were presented

as mean ± standard error of mean, Cmax increased as the oral dose increased,

however, the subcutaneously administered GSH showed the higher Cmax. Tmax rang

was 2.8-4.5 (±0.78-0.73). Area under the curve showed that it is a dose dependant

formula as the AUC increased with increasing the dose, Table 3.2.

Table 3.2 Pharmacokinetic data of orally and subcutaneously administered GSH.

Drug Cmax

(µg/mL)

Tmax

(hr)

AUC

(µg/mL*hr)

2.5 mg/kg

Dose (oral)

64.00

±6.10

3.17

±0.91

113.00

±37.66

10 mg/kg Dose

(oral)

151.23

±27.38

2.8

±0.78

466.88

±147.64

20 mg/kg Dose

(oral)

205.875

±41.19

4.5

±0.73

726.13

±256.72

10 mg/kg Dose

(SC)

344.60

±15.11

3.44

±0.06

343.96

±17.02

Data represents mean ± SEM

Page 49: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.5 Validation

Validation of this analytical HPLC-MS method was performed in order to be

evaluated in terms of recovery, linearity of response, precision, accuracy, and

sensitivity for quantification of GSH.

3.5.1 Validation of Day One, Two, and Three on Linearity, Accuracy, and

Precision Data for GSH Quantification

According to USFDA, the coefficient of determination (R²) should be equal or more

than 0.98 and for accuracy are 85.00-115.00% Except for the LLOQ is 80.00-120% to

be within the accepted criteria

Inter and intraday accuracy, precision and linear response for standard calibration

curve and QC samples of the three days of validation are explained in the following

tables and figures:

First day of validation: Table 3.3 represents the standard calibration curve and intra-

day accuracy data shows an accuracy range of 86.5% - 114%.

As shown in Figure 3.6, R² is 0.99845, which represents the strength of the

correlation; therefore, the correlation coefficient of standard calibration curve was

consistently greater than 0.99 during the validation course. Data of the standard curve

with regards to correlation, slope, R², and intercept for day one are showed in Table

3.4.

Therefore, first day of validation results passed the required criteria in terms of

linearity and accuracy.

Page 50: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Table 3.3 Standard calibration curve of the first day of validation, intraday accuracy

data for GSH.

Theoretical Conc.

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured**‏

Conc.

Accuracy%

1.0 3352.0 27365.0 0.122 1.1 111.0

2.0 4222.0 29467.0 0.143 2.2 114.0

5.0 5602.0 29350.0 0.191 4.94.0 98.7

20.0 8883.0 21384.0 0.415 17.5 87.5

60.0 26511.0 25717.0 1.031 51.9 86.5

120.0 57458.0 27657.0 2.078 110.0 92.0

200.0 120852.0 29954.0 4.035 220.0 110.0

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/ 010.0‏9 ) - (+ 0.1031).

Table 3.4 Data of the standard curve with regards to correlation, slope, R², and

intercept on the first day for GSH.

Correlation (R) Slope R² Intercept

0.999 010.09 0.9945 + 0.1031

Page 51: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

y = 0.0179x + 0.1031R² = 0.9945

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 50 100 150 200 250

An

alyt

e a

rea

/ I

S ar

ea

Analyte concentration / IS concentration

Figure 3.8 Plot of calibration curve levels against their analytical response and

regression linear equation on the first day of validation for GSH.

Second day of validation: Table 3.5 represents the standard calibration curve and

intra-day accuracy data shows an accuracy range of 83.9% - 113%.

As shown in Figure 3.7, R² is 0.9978. Therefore, the correlation coefficient of

standard calibration curve was consistently greater than 0.99 during the validation

course. Data of the standard curve with regards to correlation, slope, R², and intercept

for day two are showed in Table 3.6. Therefore, second day of validation results

passed the required criteria in terms of linearity and accuracy.

Page 52: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

Table 3.5 Standard calibration curve of the second day of validation, intraday

accuracy data for GSH.

Theoretical Conc.

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured‏

Conc.**

Accuracy%

1.0 16377.0 22356.0 0.733 0.8 83.9

2.0 19162.0 25150.0 0.762 1.9 99.0

5.0 21405.0 25152.0 0.851 5.4 109.0

20.0 35556.0 29237.0 1.216 19.7 98.3

60.0 70046.0 28560.0 2.453 67.8 113.0

120.0 108150.0 28016.0 3.86 123.0 102.0

200.0 170912.0 30574.0 5.5 190.0 94.9

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/0.0257) - (+ 0.711).

Table 3.6 Data of the standard curve with regards to correlation, slope, R², and

intercept on the second day for GSH.

Correlation (R) Slope R² Intercept

0.999 0.0257 0.9978 + 0.711

Page 53: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

y = 0.0257x + 0.711R² = 0.9978

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 50 100 150 200 250

An

alyt

e a

rea

/ I

S ar

ea

Analyte concentration / IS concentration

Figure 3.9 Plot of calibration curve levels against their analytical response and

regression linear equation on the second of validation for GSH.

Third day of validation: As seen in Table 3.7 which represents the standard

calibration curve and intra-day accuracy data shows an accuracy range of 82.4% -

111%.

As shown in Figure 3.8, R² is 0.9995. The correlation coefficient of standard

calibration curve was consistently greater than 0.99 during the validation course. Data

of the standard curve with regards to correlation, slope, R², and intercept for day three

are showed in Table 3.8.

Therefore, third day of validation results passed the required criteria in terms of

linearity and accuracy.

Page 54: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Table 3.7 Standard calibration curve of the third day of validation, intraday accuracy

data for GSH.

Theoretical Conc.

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured‏

Conc.**

Accuracy%

1.0 9002.0 14665.0 0.614 1.1 111.0

2.0 12287.0 18491.0 0.664 2.23.0 111.0

5.0 14436.0 19236.0 0.750 4.12.0 82.4

20.0 25348.0 17655.0 1.436 19.2.0 96.1

60.0 58310.0 17984.0 3.242 59.0 98.3

120.0 110475.0 18661.0 5.920 118.0 98.3

200.0 176801.0 17956.0 9.846 204.0 102.0

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/0.0454) - (+ 0.0563).

Table 3.8 Data of the standard curve with regards to correlation, slope, R², and

intercept on the third day for GSH.

Correlation (R) Slope R² Intercept

0.999 0.0454 0.9995 + 0.0563

Page 55: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

0

0.5

1

1.5

2

2.5

3

3.5

4

4.5

0 50 100 150 200 250

An

alyt

e ar

ea /

IS a

rea

Analyte concentration / IS concentration

y = 0.0454 x + 0.0563R² = 0.9995

Figure 3.10 Plot of calibration curve levels against their analytical response and

regression linear equation on the third day of validation for GSH.

The linearity and linear working range of three standard curves of GSH data based on

the measured concentrations (obtained from three days of validation) are showed in

Table 3.9.

Table 3.9 Linearity and linear working range of three standard curves of GSH data

based on the measured concentrations.

Calibratio

n curve

Measured Concentrations for each Standard Point (µg/ml)

1.0 2.0 5.0 20.0 60.0 120.0 200.0

1 1.10 2.28 4.94 17.5 51.9 110.0 110.0

2 0.83 1.98 5.45 19.7 67.8 123.0 190.0

3 1.11 2.23 4.12 19.2 59 118.0 204.0

Mean 1.01 2.16 4.83 18.8 63.4 117.0 168.0

STDV 0.15 0.16 0.67 1.15 7.96 6.55 50.71

CV% 14.85 7.04 13.87 6.11 12.55 5.59 3.01

Min 0.83 1.98 4.12 17.5 51.9 110.0 220.0

Max 1.11 2.28 5.45 19.7 67.8 123.0 204.0

Page 56: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

The linearity and linear working range of three standard curves of GSH data based on

the calculated area ratio (obtained from three days of validation) are showed in Table

3.10.

Table 3.10: Linearity and linear working range of three standard curves of GSH

data based on the calculated area ratio.

Calibration

curve

AUC Ratio for each Standard Point

1 0.122 0.143 0.191 0.415 1.031 2.078 4.0350

2 0.733 0.762 0.851 1.216 2.453 3.860 5.590

3 0.614 0.664 0.750 1.436 3.242 5.920 9.846

Mean 0.489 0.523 0.597 1.022 2.242 3.952 6.490

STDV 0.323 0.332 0.355 0.537 1.120 1.922 3.008

CV% 66.151 63.617 59.514 52.560 49.977 48.642 46.350

Min 0.122 0.143 0.191 0.415 1.031 2.078 4.035

Max 0.733 0.762 0.851 1.436 3.242 5.92 9.846

Data of three standard curves with regards to correlation, slope, R², and intercept are

showed in Table3.11.

Table 3.11: Data of three standard curves with regards to correlation, slope, R²,

and intercept for GSH.

Correlation (R) Slope R² Intercept

0.999 0.0343 0.9865 + 0.3517

The plot of linearity of calibration curve levels for GSH quantification against their

analytical response and regression linear equation that represents the all three days of

validation was done by plotting the calculated mean of the measured concentrations

Page 57: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

versus the calculated mean of the AUC ratio for each standard point showed in Figure

3.11.

y = 0.0300x + 0.4400R² = 0.9996

0

1

2

3

4

5

6

7

0 50 100 150 200 250

AU

C m

aen

rat

io

GSH concentration (µg/mL)

Figure 3.11: Plot of linearity of calibration curve levels for GSH quantification against

their analytical response and regression linear equation.

Page 58: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Table 3.12 represents intra-day precision and accuracy data for QC low samples based

on the standard calibration curve of the first day of validation. Accuracy range and

precision (CV %) for the six replicates of QC low samples was (93.5-114%) and

(8.17%), respectively. Six replicates QC mid samples of the first day of validation

with accuracy range and precision (89.6-113.3%) and (9.07%), respectively, are

represented in Table 3.13, while six replicates QC high samples of the same day with

accuracy range and precision (96.5-105.8%) and (7.12%), respectively, are

represented in Table 3.14.

Table 3.12 Intra-day precision and accuracy data for QC low samples of GSH based

on the standard calibration curve of the first day of validation.

QC Low

Concentration

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured

Conc.**

Accuracy%

3.0 2277.9 40677.0 0.05 2.9 98.9

2214.8 39550.0 0.05 3.1 105.0

2285.0 40088.0 0.05 3.4 114.0

2269.1 40520.0 0.05 3.3 113.0

2259.0 40341.0 0.05 3.0 100.0

2223.8 40433.0 0.05 2.8 93.5

Mean 2254.9 40268.0 0.05 3.1 104.0

STDV 29.0 402.8 0.00063 0.2 8.1

CV% 1.2 1.0 1.2 7.6 7.7

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/ 010.0‏9 ) - (+ 0.1031).

Page 59: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Table 3.13 Intra-day precision and accuracy data for QC mid samples of GSH based

on the standard calibration curve of the first day of validation.

QC Mid

Concentration

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured

Conc.**

Accuracy%

100.0 9574.5 39240.0 0.24 107.0 107.0

9889.0 39556.0 0.25 111.0 111.0

8982.2 40279.0 0.22 95.9 95.9

9551.6 39965.0 0.23 105.0 105

8321.8 39254.0 0.21 89.6 89.6

10022.0 39457.0 0.25 113.0 113.0

Mean 9390.2 39625.1 0.23 103.5 103.5

STDV 634.9 415.2 0.01 9.0 9.0

CV% 6.7 1.0 4.3 8.6 8.6

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/ 010.0‏9 ) - (+ 0.1031).

Table 3.14 Intra-day precision and accuracy data for QC high samples of GSH based

on the standard calibration curve of the first day of validation.

QC High

Concentration

(µg/ml)

AUC

Drug

AUC IS AUC

Ratio*

Measured

Conc.**

Accuracy%

160.0 13260.2 39465.0 0.336 158.0 98.8

12719.1 38660.0 0.329 154.0 96.5

14841.2 39789.0 0.373 179.0 112.0

14090.5 40030.0 0.352 167.0 104.0

14482.1 38619.0 0.375 180.0 112.0

14938.6 39943.0 0.374 179.0 112.0

Mean 14055.3 39417.6 0.350 169.5 105.8

STDV 894.3 632.9 0.020 11.5 7.1

CV% 6.3 1.6 5.7 6.7 6.7

*AUC Ratio=AUC Drug/AUC IS.

**Measured concentration= (AUC Ratio/ 010.0‏9 ) - (+ 0.1031).

Page 60: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

3.5.2 Recovery data for GSH quantification

Table 3.16 shows data of triplicate quality control samples of GSH spiked after

precipitation, while Table 3.17 shows data of triplicate quality control samples of

GSH spiked before precipitation.

Absolute recoveries for GSH and IS were calculated by dividing average peak area of

triplicate from each QC level of samples in Table 3.16 over the same set of QC

samples in Table 3.17 multiplied by 100%.

Table 3.18 shows recovery % of GSH and Table 3.19 shows recovery % of IS which

shows a high recovery % and acceptable at the studied concentration.

Table 3.16: Data of GSH and IS, prepared by spiking GSH after precipitation,

for the quality control samples.

concentration

µg/ml

AUC

GSH

AUC IS Mean

GSH

Mean IS

3.0

QC low

2690.0 40520.0

2729.3

40431.3

2724.0 40341.0

2774.0 40433.0

100.0

QC mid

16334.0 39965.0 16185.0

39558.6

16681.0 39254.0

15540.0 39457.0

160.0

QC high

26651.0 40030.0

25964.0

39530.6

24966.0 38619.0

26275.0 39943.0

Page 61: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

Table 3.17: Data of GSH and IS, prepared by spiking GSH before precipitation,

for the quality control samples.

concentration

µg/ml

AUC

CAM

AUC IS Mean

GSH

Mean IS

3.0

QC low

2486.0 40677.0

2570.6

40478.3

2619.0 40670.0

2607.0 40088.0

100.0

QC mid

15060.0 40666.0

15737.0

40167.0

15996.0 39556.0

16155.0 40279.0

160.0

QC mid

25958.0 38660.0

25679.3

39530.6

24829.0 39465.0

26251.0 39789.0

Table 3.18: Recovery % for GSH.

concentration

µg/ml

Mean

(before)

Mean (after)

Recovery

%

3.0 QC low 2570.6 40478.3 94.1

100.0 QC mid 15737.0 40167.0 97.2

160.0 QC high 25679.3 39530.6 98.9

Table 3.19: Recovery % for IS.

concentration

µg/ml Mean serum

Mean mobile

phase

Recovery

%

3.0 QC low 40478.3 40431.3 100.1

100.0 QC mid 40167.0 39558.6 101.5

160.0 QC high 39530.6 39530.6 99.4

Page 62: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

Figure 3.12: GSH placebo chromatogram.

Figure 3.13: GSH STD 1 chromatogram.

Page 63: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

Figure 3.14: GSH STD 2 chromatogram.

Figure 3.15: GSH STD 3 chromatogram.

Page 64: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Figure 3.16: GSH STD 4 chromatogram.

Figure 3.17: GSH STD 5 chromatogram.

Page 65: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Figure 3.18: GSH STD 6 chromatogram.

Figure 3.19: GSH STD 7 chromatogram.

Page 66: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Figure 3.20: GSH rat sample chromatogram at 1.00 hr measured as 53.00 µg/ml.

Figure 3.21: GSH rat sample chromatogram at 2.00 hr measured as 28.00 µg/ml.

Page 67: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Figure 3.22: GSH rat sample chromatogram at 3.00 hr measured as 29.00 µg/ml.

Figure 3.23: GSH rat sample chromatogram at 4.00 hr measured as 45.00 µg/ml.

Page 68: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

Figure 3.24: GSH rat sample chromatogram at 5.00 hr measured as 77.00 µg/ml.

Page 69: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

GSH IS

Figure 3.25: GSH QCL chromatogram.

Page 70: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

33

GSH IS

Figure 3.26: GSH QCM chromatogram.

Page 71: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Three: Results

34

GSH IS

Figure 3.27: GSH QCH chromatogram.

Page 72: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

55

Chapter Four

Discussion

Page 73: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

55

4. Discussion

It has been always acknowleged that the oral route is more favorable than the

subcutaneous route of drug administration and much better in respect patients’

compliance. Upto date, the currently marketed GSH oral products relies mainly on the

breakdown of GSH and the absorption of its constituent amino acids. Such products

have not been proven efficient or effective in the direct elevation of GSH

concentration to the optimal levels that the body needs.

GSH oral suplimentations remains a challenge due to its direct distruction in the

digestion process before it reaches the cells. Furhtermore, the extremely short half life

of the subcutaneously administered GSH encouraged an intensive research attempts to

develop an oral GSH system.

Without a protected system, many studies had demonstrated that direct oral GSH

suplimentation fail to raise its circulating levels. For example, Briviba and his co-

workers, indicated no presumed increase of GSH in humans following oral

administration of 3 g of unprotected GSH (Briviba et al., 1998). The same dose was

used by Witschi et al. were they stated that "it is not possible to increase circulating

glutathione to a clinically beneficial extent by the oral administration of a single dose

of 3 g of glutathione." (Witschi et al., 1992).

In another study, a 2.5-fold increase in GSH following an oral load of 15 mg/kg was

detected, but this increase was fugacious and represented only a small fraction of the

total GSH administered, however, a significant increase of GSH in tissue was found

only in the lung (Staack et al., 1998).

Page 74: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

55

Jason and Ryan in a randomized, double-blind, placebo-controlled trial in healthy

human adults concluded that, despite the high commitment of the volunteers to the

oral supplementation schecuals, the oral GSH supplementation did not result in

significant changes in glutathione status in participated healthy adults (Jason and

Ryan, 2011).

Nevertheless, in order to develop a protected oral GSH delivery system, the technique

applied should protect it against acidic and enzymatic actions in the stomach and

enhance the permeation of GSH to reach the blood system. Only few studies

published in recent years, state to find an effective way to deliver the GSH intact to

the circulatory system.

There has been a growing research attention in the field of drug delivery using

particulate delivery systems as carriers for small and large molecules. Particulate

systems, including nanoparticles, have been used successfully as a physical approach

to modify and ameliorate the pharmacokinetic and pharmacodynamic of drugs. The

important technological advantages of nanoparticles used as drug carriers are high

stability, high carrier capacity, and feasibility of incorporation of both hydrophilic and

hydrophobic substances. These properties of nanoparticles enable improvement of

drug bioavailability (Gelperina et al., 2005‏).

Chitosan has been the subject of interest to be utilized as a polymeric drug carrier

material in many designed dosage forms, due to its appealing properties such as

biocompatibility, biodegradability, low toxicity and its relatively low production cost

due to its abundant natural sources, (Hamman, 2010).

Chitosan is known to have significant ability to form complexes as a PECs

(Yancheva et al., 2007). T s at p an in i i state t e a ni

Page 75: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

55

p NH3⁺) of chitosan can interact with either t e ta a ate

p t e te ina a ate p ) of GSH to form

polyelectrolyte complex as illustrated in Figure 3.1 (Wu, et al, 2009‏). After

the addition of the formed PEC to an oily phase (oleic acid), nanoparticles

were constructed by the self-assembly or self-organization of the surfactant

(labrasol®) and the cosurfactant Plurol oleique

®,. in a manner where the

hydrophobic tails formed the core of the aggregate while the hydrophilic

heads were in contact with the surrounding liquid (Figure 1.5).

Many research atempts have examined the possibility of the interaction between

chitosan and GSH for a variety of reasons, most importantly, to increase the delivery

and bioavailability of GSH.

Kafedjiiski and coworker aimed to synthesize and characterize chitosan-glutathione

(GSH) conjugate, providing improved mucoadhesive and permeation-enhancing

properties. They found that the conjugate would be a promising multifunctional

excipient for various drug delivery systems (Kafedjiiski et al, 2005).

In a study Yousefpour and coworkers, synthesized chitosan-glutathione conjugates of

different thiolation degrees. They conclude "Cht-GSH seems to possess suitable

characteristics for the preparation of nanoparticulate oral drug delivery system"

(Yousefpour et al., 2011). However, no in vivo pharmacological testing has been

done.

Li and coworkers, developed a novel non-viral gene vector, based on

poly[poly(ethylene glycol) methacrylate] (PMPEG) and l-glutathione (GSH) grafted

Page 76: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

55

chitosan (CS). They indicate that, the non-viral vector is a promising candidate for

gene therapy in clinical application (Li et al, 2011).

In other work, Li and coworkers, developed thermosensitive chitosan chloride-

glutathione (CSCl-GSH) hydrogel, to suppress the oxidative stress injury in

cardiomyocytes (CMs). Their data show that CSCl-GSH conjugates in vitro could

effectively scavenge ROS. Thus, suppress the oxidative stress damage and apoptosis

in CMs, in the presence of high ROS (Li, et al, 2013).

In another research, Koo and coworkers studied the characteristics and oxidative

stability of chitosan-glutathione conjugate (CS-GSH) and CS-GSH nanoparticles (CS-

GSH NPs) for exploring the potentials of these nanoparticle systems for GSH

delivery. Thier results indicated that CS-GSH NP can be used to increase the

oxidative stability of GSH (Koo et al, 2011).

A gastrointestinal patch system containing chitosan-glutathione was developed and

investigated by Hoyer and coworkers in terms of water-absorbing capacity, adhesive

properties, in vitro release, unidirectional release and permeation enhancing effect.

This work indicated that, this system could be an interesting possibility for the

transport of macromolecules through the intestinal mucosa (Hoyer et al, 2007).

However Miri and coworkers successfully acquired two hybrid systems, GSH-MMT

(MMT: montmorillonite) hybrid and AEA-GSH-MMT hybrid (AEA: polyvinylacetal

diethylaminoacetate), by intercalating GSH into the interlayer spaces of MMT, and

they stated "the present GSH-MMT hybrids remarkably enhanced GSH concentration

in the plasma, heart, kidney, and liver, especially when AEA-GSH-MMT hybrid was

administered under GSH-deficient condition. Moreover, both hybrids did not induce

a te a t i it p t 2000  /k s estin t ei eat p tentia f

Page 77: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

56

pharmaceutical application", the highest concentration of GSH reached was 70-80

µg/mL (Miri et al., 2012).

The current research, however, includes two main courses, the first concerning with

characterizing the PEC and confirming the ion pair formation et een t e a ni

p NH3⁺) of chitosan an a ate p ) of GSH while the second

course of the investigation was directed towards the evaluation of the prepared GLN-

RM.

In terms of PEC formation, the variances observed in FT-IR spectrum of GC-PEC

from that of GSH, chitosan and GC-physical mixture indicate the formation of the

ionic interaction and subsequently the polyelectrolyte complex. Mainly the peak at

1710 ¹ corresponds to the carbonyl groups (C=O) in GSH FT-IR spectrum, is

shifted in the spectrum of GC-PEC, and the characteristic band at 1250–1020 ¹,

corresponds to C-N stretching in chitosan spectrum, emerged sharper in GC-PEC

spectrum (Figure 3.1).

DSC is consedered a powerful analytical tool. The thermodynamic parameters

obtained from DSC experiments are quite sensitive to the structural state of the

biomolecule. Any change in the conformation would affect the position, sharpness,

and shape of transition(s) in DSC scans as described by Pooria et al. in 2010.

Concerning our DSC readings (Figure 3.2), a shift in glutathione melting peak, down

to 170˚ was noted with a strong remarkable reduction in the sharpness of the peak.

As for the changes in the shape of the decomposition peak of chitosan, there was no

sharp exothermic peak seen while in the case of complex there was a gradual thermal

degradation behavior. This indicates the possible ionic interaction between the

carboxylate group of glutathione and the ammonium group of chitosan.

Page 78: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

56

The nuclear magnetic resonance spectroscopy (¹H-NMR) results, give virtual certainty

indication on the ionic exchange, in the ¹H-NMR spectrum of GC-PEC, the peak at

3.7 ppm, which, corresponding to Cb, i of GSH, was shifted down filed, with splitting

in the peak, as well as, the peak in chitosan spectrum, at (3.18 ppm), that correspond

to Cb of chitosan, was shifted up filed (william 1999).

Accordingly, all of the obtained characterization data serve as an evidence for the

complex formation between chitosan and GSH through ionic bonds; mainly

electrostatic.

The second course of the current investigation was directed towards the evaluation of

the prepared GLN-RM in relation to the nanoparticles size, zeta potential, and

assessing the pharmacological activity of GLN-RM and it is incidence to elevate GSH

in the blood.

It was noticed that GLN-RMs prepared by 1.3, 8 and 13 kDa chitosan recorded

shorter time to start precipitation (nearly 3 hr), than GLN-RM prepared using 18 and

30 kDa chitosan which showed relatively long time to start precipitation (up to 7 hr.).

After repetition with large concentrations, 30 kDa GLN-RM showed consistency

compared to other chitosan grades without and noted aggregation.

The results of DLC gave the desired outcome for all the formulas, except the 1.3 kDa

GLN-RM which, as expected from previous studies on insulin (Elsayed et al., 2011,

gave larger particle size. The reason behind this is the lack of enough amino groups to

bind with the whole amount of GSH to reduce it is size to a nano-scale. Such an

explanation further support the advantage of using 30 kDa chitosan that showed

longer time for precipitation and consistency with repetitions, especially at high

concentration, by providing enough chitosan to bind to all of the presented GSH.

The zeta potential of the prepared GLN-RM was found 277.5 ±1.42. For molecules

Page 79: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

56

and particles that are small enough, a high zeta potential will confer stability, i.e., the

solution or dispersion will resist aggregation. Thus, colloids with high zeta potential

(negative or positive) are electrically stabilized while colloids with low zeta potentials

tend to coagulate (Greenwood and Kendall 1999).

One of the biggest obstacles in the work was the high amount of the originally

existing GSH in rat's blood. This can be explained by the high stress of the rats

included in the experiment. Emotional stress significantly increases free radical load

on the body. As stress induces the release of hormones, the body mobilizes for action;

known as the fight and flight response. This creates biochemical changes which

increase free radical stress in the body (Liu et al., 1996 ‏). This led us to take certain

measures during the experimentation on rats. For example, the withdrawal of a blood

samples an hour before starting the experiment which showed a reduction in GSH

concentration to a great extent. Moreover, giving a high GSH dose in order to repeal

the significance of the GSH that already exists.

For the analysis of GSH in rat's serum, an HPLC/MS was used, the primary advantage

that HPLC/MS has is that it is capable of analyzing a wide range of components.

Compounds that exhibit high polarity or have a high molecular mass may all be

analyzed, including proteins, with high sensitivity and selectivity.

For the analysis of in vivo gluthathione levels in rats, a valid method published by

Xiangming and coworkers was adapted with some modifications (Xiangming et al.,

2002). These modifications included using formic acid and methanol as a mobile

phase while using citalopram as an internal standard with a retention time of around

1.8 min.

A derivatizing reagent, E an’s Rea ent -dithiobis (2-nitrobenzoic acid), was

used. This compound produces a measurable yellow-colored product when it reacts

Page 80: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

56

with sulfhydryls. While citalopram was chosen as an internal standard, because it has

the same method of extraction of GSH while having a RT close to GSH.

The validation of method in terms of linearity, accuracy and precision was acceptable

(Table 3.11). While Table 3.12 represents the intra-day precision and accuracy data

for QC low samples based on the standard calibration curve of the first day of

validation. Accuracy range and precision (CV %) for the six replicates of QC low

samples was (93.5-114%) and (8.17%), respectively. Six replicates QC mid samples

of the first day of validation with accuracy range and precision (89.6-113.3%) and

(9.07%), respectively, are represented in Table 3.13, while six replicates QC high

samples of the same day with accuracy range and precision (96.5-105.8%) and

(7.12%), respectively, are represented in Table 3.14. While, Table 3.1 and Table 3.19

shows recovery % of GSH and IS., respectively, which shows a high recovery % and

acceptable at the studied concentration.

According to the United States Food and Drug Administration (USFDA), the

coefficient of determination (R²) should be equal or more than 0.98 and for accuracy

and precision are (85.00-115.00%)(˂ 1 .00%), respectively, to be within the accepted

criteria. Thus, the evaluation results of our method of analysis are acceptable and

within the criteria.

The in vivo pharmacodynamics of GLN-RM, and by comparing the results obtained

from the different groups. It was clear the significant increase of the concentration in

groups of rats administered GLN-RM., comparing with GSH baseline in the rat's

plasma

The nanoparticles (GLN-RM), showed a rapid and prolonged release for the drug, by

comparing with, subcutaneously administered GSH. It also, a dose depends formula,

as the GSH concentration in rat's plasma increase, by increasing concentration of the

Page 81: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

Chapter Four: Discussion

56

formula. The drug releasing profile from nanoparticles indicated zero-order release,

these results was expected, due to the small particles size, the stability and equilibrate,

even in the maximum loading concentration of the PEC, oral GSH formulation

bioavailability and action was shown promising for the development of oral GSH

product.

It is obviously that the system successfully protected and delivered the drug to the

bloodstream as the groups received low doses there GSH blood level was to a large

extent close to the baseline while a remarkable increase in GSH levels occurred after

the administration of high doses.

Conclusion

This research investigates, the potentiality of formulating an oral delivery system for

GSH, and we have successfully incorporated the powerful antioxidant, into lipid-

based formulation; GSH was protected from gastric enzymes by merging the features

of nanoparticles and the use of an oily vehicle. The formula has been found to have a

modified particle size distribution with a mean diameter of 94±2.16 nm.

One of the most things that influenced the work adversely is the high presence of

internal GSH in the rat's blood, which can be avoided in future studies using models

with GSH deficiency.

Future studies are expected to aim for, studying the chemical and physical stability of

the entrapped GSH in the nanoparticle dispersion system, giving multiple dose to rats,

doing accelerated stability of GC-PEC with more stable formula, of higher loading

capacity and higher bioavailability, by enhancing the absorption of GSH, with

studying the mechanism by which GSH in vivo penetration occur.

Page 82: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

References

Page 83: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

5 References

Aebi, S., Assereto, R., & Lauterburg, B. H. (1991). High‐dose intravenous

glutathione in man. Pharmacokinetics and effects on cyst (e) in plasma and

urine. European Journal of Clinical Investigation, 21(1), 103-110.‏

Agnihotri, S. A., Mallikarjuna, N. N., & Aminabhavi, T. M. (2004). Recent

advances on chitosan-based micro-and nanoparticles in drug delivery. Journal

of Controlled Release, 100(1), 5-28.‏

Allen, J., & Bradley, R. D. (2011). Effects of oral glutathione supplementation

on systemic oxidative stress biomarkers in human volunteers. The Journal of

Alternative and Complementary Medicine, 17(9), 827-833.‏

Amiji, M. M. (1995). Pyrene fluorescence study of chitosan self-association in

aqueous solution. Carbohydrate Polymers, 26(3), 211-213.‏

Anderson, M. E. (1998). Glutathione: an overview of biosynthesis and

modulation. Chemico-Biological Interactions, 111, 1-14.‏

Arosio, E., De Marchi, S., Zannoni, M., Prior, M., & Lechi, A. (2002,

August). Effect of glutathione infusion on leg arterial circulation, cutaneous

microcirculation, and pain-free walking distance in patients with peripheral

obstructive arterial disease: a randomized, double-blind, placebo-controlled

trial. In Mayo Clinic Proceedings (Vol. 77, No. 8, pp. 754-759). Elsevier.‏

Barratt, G. (2003). Colloidal drug carriers: achievements and

perspectives.Cellular and Molecular Life Sciences CMLS, 60(1), 21-37.‏

Page 84: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

Baudouin P, Lagniel G, Kumar C, Huang ME, Labarre J. (2012) Glutathione

degradation is a key determinant of glutathione homeostasis. Journal of

Biologecal Chemistry, 287(7):4552-61

Bhadra, D., Bhadra, S., Jain, S., & Jain, N. K. (2003). A PEGylated dendritic

nanoparticulate carrier of fluorouracil. International Journal of

Pharmaceutics, 257(1), 111-124.‏

Briviba K., Kissner R., Koppenol WH, Sies H., 1998 Kinetic study of the

reaction of glutathione peroxidase with peroxynitrite. Chemical Research

in Toxicology, 11:1398-1401.

Calvo, P., Remunan-Lopez, C., Vila-Jato, J. L., & Alonso, M. J. (1997). Novel

hydrophilic chitosan-polyethylene oxide nanoparticles as protein carriers.

Journal of Applied Polymer Science, 63(1), 125-132.‏

Calvo, P., Remuñan-López, C., Vila-Jato, J. L., & Alonso, M. J. (1997).

Chitosan and chitosan/ethylene oxide-propylene oxide block copolymer

nanoparticles as novel carriers for proteins and vaccines. Pharmaceutical

Research, 14(10), 1431-1436.‏

Cascinu, S., Catalano, V., Cordella, L., Labianca, R., Giordani, P., Baldelli, A.

M., ... & Catalano, G. (2002). Neuroprotective effect of reduced glutathione on

oxaliplatin-based chemotherapy in advanced colorectal cancer: a randomized,

double-blind, placebo-controlled trial. Journal of Clinical Oncology, 20(16),

‏.3478-3483

Page 85: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

Chydyrov, P. R. (2007). New mite species of the cohort tarsonemina

(Trombidiformes, Acarina) of the fauna of Turkmenistan. Entomological

Review, 87(6), 767-775.‏

Crini, G. (2005). Recent developments in polysaccharide-based materials used

as adsorbents in wastewater treatment. Progress in Polymer Science, 30(1),

‏.38-70

DDS Medication Administration Recertification Manual. DDS Recertification

Review Manual. State of Connecticut Department of Developmental Services.

2006. Retrieved April 2, 2013.

De Jong, W. H., & Borm, P. J. (2008). Drug delivery and nanoparticles:

applications and hazards. International Journal of Nanomedicine, 3(2), 133

De Mattia, G., Bravi, M. C., Laurenti, O., Cassone-Faldetta, M., Armiento, A.,

Ferri, C., & Balsano, F. (1998). Influence of reduced glutathione infusion on

glucose metabolism in patients with non—insulin-dependent diabetes mellitus.

Metabolism, 47(8), 993-997.‏

Dire F., Bohm S., Oriana S., Spatti G., Pirovano C., Tedeschi M., Zunino F.

(1993) High-dose cisplatin and cyclophosphamide with glutathione in the

treatment of advanced ovarian cancer. Annals of Oncology, 4(1):55-61.

Dodane, V., Amin Khan, M., & Merwin, J. R. (1999). Effect of chitosan on

epithelial permeability and structure. International Journal of Pharmaceutics,

‏.21-32 ,(1)182

Dringen, R., Gutterer, J. M., & Hirrlinger, J. (2000). Glutathione metabolism

in brain. European Journal of Biochemistry, 267(16), 4912-4916.‏

Page 86: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

Ellman’s Reagent Number 22582 thermo scientific.

Elsayed, A., Al-Remawi, M., Qinna, N., Farouk, A., Al-Sou’od, K. A., &

Badwan, A. (2011). Chitosan–sodium lauryl sulfate nanoparticles as a carrier

system for the in vivo delivery of oral insulin. American Association of

Pharmaceutical Scientists, 12(3), 958-964.‏

Fabritius, H. O., Sachs, C., Triguero, P. R., & Raabe, D. (2009). Influence of

Structural Principles on the Mechanics of a Biological Fiber‐Based Composite

Material with Hierarchical Organization: The Exoskeleton of the Lobster

Homarus americanus. Advanced Materials, 21(4), 391-400.‏

Floyd, R. A. (1999). Antioxidants, oxidative stress, and degenerative

neurological disorders. Experimental Biology and Medicine, 222(3), 236-245.‏

Fukagawa N., Ajami A., Young V. (1996) Plasma methionine and cysteine

kinetics in response to an intravenous glutathione infusion in adult humans.

American Journal of Physiology, 270: 209-14.

Gandhi, T. K. B., Zhong, J., Mathivanan, S., Karthick, L., Chandrika, K. N.,

Mohan, S. S., & Pandey, A. (2006). Analysis of the human protein

interactome and comparison with yeast, worm and fly interaction datasets.

Nature Genetics, 38(3), 285-293.

Ganguli, D., Kumar, C., & Bachhawat, A. K. (2007). The alternative pathway

of glutathione degradation is mediated by a novel protein complex involving

three new genes in Saccharomyces cerevisiae. Genetics, 175(3), 1137-1151.‏

Ganguly, D., Srikanth, C. V., Kumar, C., Vats, P., & Bachhawat, A. K. (2003).

Why is glutathione (a tripeptide) synthesized by specific enzymes while TSH

Page 87: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

releasing hormone (TRH or thyroliberin), also a tripeptide, is produced as part

of a prohormone protein?. The International Union of Biochemistry and

Molecular Biology life, 55(9), 553.‏‏

Gawryluk, J. W., Wang, J. F., Andreazza, A. C., Shao, L., & Young, L. T.

(2011). Decreased levels of glutathione, the major brain antioxidant, in post-

mortem prefrontal cortex from patients with psychiatric disorders. The

International Journal of Neuropsychopharmacology, 14(1), 123.‏

Gelperina, S., Kisich, K., Iseman, M. D., & Heifets, L. (2005). The potential

advantages of nanoparticle drug delivery systems in chemotherapy of

tuberculosis. American Journal of Respiratory and Critical Care

Medicine, 172(12), 1487.

Gottlieb, H. E., Kotlyar, V., & Nudelman, A. (1997). NMR chemical shifts of

common laboratory solvents as trace impurities. The Journal of Organic

Chemistry, 62(21), 7512-7515.‏

Greenwood, R., & Kendall, K. (1999). Selection of suitable dispersants for

aqueous suspensions of zirconia and titania powders using

acoustophoresis.Journal of the European Ceramic Society, 19(4), 479-488.‏

Gupta K., & Jabrail F. (2006) Effects of degree of deacetylation and cross-

linking on physical characteristics, swelling and release behavior of chitosan

microspheres. Carbohydrate polymers, 66(1): 43-54.

Gupta, K. C., & Ravi K. (2000). An overview on chitin and chitosan

applications with an emphasis on controlled drug release formulations.Journal

of Macromolecular Science, Part C: Polymer Reviews, 40(4), 273-308.‏

Page 88: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

Hadwiger, L. A. (2013). Plant Science review: Multiple effects of chitosan on

plant systems: solid science or hype. Plant Science.

Hagen T., Wierzbicka G., Sillau A. (1990) Bioavailability of dietary

glutathione: effect on plasma concentration, American Journal of Physiology.

259:524-29.

Hamman, J. H. (2010). Chitosan based polyelectrolyte complexes as potential

carrier materials in drug delivery systems. Marine drugs, 8(4), 1305-1322.‏

Herman E., Zhang J., Hasinoff, P., Chadwick B., and Ferrans V. (1997)

Comparison of the protective effects against chronic doxorubicin

cardiotoxicity and the rates of iron (III) displacement reactions of ICRF-187

and other bisdiketopiperazines. Cancer Chemother. Pharmacol, 40(5):400-

408.

Hirai, A., Odani, H., & Nakajima, A. (1991). Determination of degree of

deacetylation of chitosan by 1H NMR spectroscopy. Polymer Bulletin, 26(1),

‏.87-94

Hofmann A., 1929 On the enzymatic degradation of chitin and Chitosan.

Ph.D. thesis, University of Zurich; Zurich, Switzerland.

Hopkins, F. G., & Dixon, M. (1922). On glutathione II. A thermostable

oxidation-reduction system. Journal of Biological Chemistry, 54(3), 527-563.‏.

Hoyer, H., Föger, F., Kafedjiiski, K., Loretz, B., & Bernkop-Schnürch, A.

(2007). Design and evaluation of a new gastrointestinal mucoadhesive patch

system containing chitosan-glutathione. Drug Development and Industrial

Pharmacy, 33(12), 1289-1296.‏

Page 89: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

11

Hu, Y., Jiang, X., Ding, Y., Ge, H., Yuan, Y., & Yang, C. (2002). Synthesis

and characterization of chitosan–poly (acrylic acid)

nanoparticles. Biomaterials, 23(15), 3193-3201.‏

Hughes, A. E., Cole, I. S., Muster, T. H., & Varley, R. J. (2010). Designing

green, self-healing coatings for metal protection. NPG Asia Materials, 2(4),

‏.143-151

Imeri A., & Knorr D., 1988 Effects of chitosan on yield and compositional

data of carrot and apple juice. Journal of Food Science, 53(6): 1707-1709.

Jain, A., Gupta, Y., & Jain, S. K. (2007). Perspectives of biodegradable natural

polysaccharides for site-specific drug delivery to the colon. The Journal

of Pharmaceutical Sciences, 10(1), 86-128.‏

James, S. J., Melnyk, S., Fuchs, G., Reid, T., Jernigan, S., Pavliv, O., ... &

Gaylor, D. W. (2009). Efficacy of methylcobalamin and folinic acid treatment

on glutathione redox status in children with autism. The American Journal of

Clinical Nutrition, 89(1), 425-430.‏

Janet K., David A., James B., Carolyn R., Mark R. (2011) A clinical trial of

glutathione supplementation in autism spectrum disorders. Medical Science

Monitor's, 17(12):CR677-682.

Jason A., and Ryan D. (2011) Effects of Oral Glutathione Supplementation on

Systemic Oxidative Stress Biomarkers in Human Volunteers. Journal of

Alternative and Complementary Medicine, 17 (9): 827-833.

Josias H. (2010) Chitosan Based Polyelectrolyte Complexes as Potential

Carrier Materials in Drug Delivery Systems. Marine Drug Journal, 8(4):

1305-1322.

Page 90: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

18

Kafedjiiski, K., Föger, F., Werle, M., & Bernkop-Schnürch, A. (2005).

Synthesis and in vitro evaluation of a novel chitosan–glutathione

conjugate.Pharmaceutical Research, 22(9), 1480-1488.‏

Kapoor, A., & Viraraghavan, T. (1995). Fungal biosorption—an alternative

treatment option for heavy metal bearing wastewaters: a review. Bioresource

Technology, 53(3), 195-206.‏

Kean, T., & Thanou, M. (2010). Biodegradation, biodistribution and toxicity

of chitosan. Advanced Drug Delivery Reviews, 62(1), 3-11.

Kerksick, C., & Willoughby, D. (2005). The antioxidant role of glutathione

and N-acetyl-cysteine supplements and exercise-induced oxidative

stress. International Society of Sports Nutrition 2(2), 38-44.‏

Khor, E., & Lim, L. Y. (2003). Implantable applications of chitin and chitosan.

Biomaterials, 24(13), 2339-2349.‏

Kidd, P. M. (1997). Glutathione: systemic protectant against oxidative and

free radical damage. Altern Med Rev, 1, 155-176.‏

Kirby, B. (2010). Micro-and Nanoscale Fluid Mechanics (Vol. 32). New

York: Cambridge University Press.‏

Kompella, U. B., Bandi, N., & Ayalasomayajula, S. P. (2003).

Subconjunctival nano-and microparticles sustain retinal delivery of

budesonide, a corticosteroid capable of inhibiting VEGF

expression. Investigative Ophthalmology & Visual Science, 44(3), 1192-1201.‏

Page 91: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

89

Koo, S. H., Lee, J. S., Kim, G. H., & Lee, H. G. (2011). Preparation,

characteristics, and stability of glutathione-loaded nanoparticles. Journal of

Agricultural and Food Chemistry, 59(20), 11264-11269.‏

Krajewska, B. (2004). Application of chitin-and chitosan-based materials for

enzyme immobilizations: a review. Enzyme and Microbial Technology, 35(2),

‏.126-139

Kramarenko E., Khokhlov A., Reineker P. (2006) Stoichiometric

polyelectrolyte complexes of ionic block copolymers and oppositely charged

polyions. Journal of Chemical Physics, 125:1-8.

Kumar M., Baboota S., Ahuja A., Hasan S. & Ali J. (2007) Recent advances in

protein and peptide drug delivery systems. Current Drug Delivery, 4(2): 141-

151.

Lavertu, M., Xia, Z., Serreqi, A. N., Berrada, M., Rodrigues, A., Wang, D., &

Gupta, A. (2003). A validated< sup> 1</sup> H NMR method for the

determination of the degree of deacetylation of chitosan. Journal of

Pharmaceutical and Biomedical Analysis, 32(6), 1149-1158.‏

Lee, V. H. L. (1995). Oral route of peptide and protein drug delivery. In

Absorption of Orally Administered Enzymes (pp. 39-46). Springer Berlin

Heidelberg.‏

Li, C., Guo, T., Zhou, D., Hu, Y., Zhou, H., Wang, S. & Zhang, Z. (2011). A

novel glutathione modified chitosan conjugate for efficient gene

delivery. Journal of Controlled Release, 154(2), 177-188.‏

Li, J., Shu, Y., Hao, T., Wang, Y., Qian, Y., Duan, C., ... & Wang, C. (2013).

A chitosan–glutathione based injectable hydrogel for suppression of oxidative

stress damage in cardiomyocytes. Biomaterials, 34(36), 9071-9081.‏

Page 92: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

Li, N., Bai, R., & Liu, C. (2005). Enhanced and selective adsorption of

mercury ions on chitosan beads grafted with polyacrylamide via surface-

initiated atom transfer radical polymerization. Langmuir, 21(25), 11780-

‏.11787

Liu, J., Wang, X., Shigenaga, K., Yeo, H., Mori, A., and Ames, B., (1996).

Immobilization stress causes oxidative damage to lipid, protein, and DNA in

the brain of rats. The Journal of Federation of American Societies for

Experimental Biology, 10(13), 1532-1538.

Lu, S. C. (2009). Regulation of glutathione synthesis. Molecular Aspects of

Medicine, 30(1), 42-59.‏

Marguerite R. (2006) Chitin and chitosan: Properties and applications.

Progress in Polymer Science, 48(2): 603-632.

Meister A. (1988). Glutathione metabolism and its selective modification.

Journal of Biological Chemistry, 263 (17): 205-8.

Melo, A., Monteiro, L., Lima, R. M., de Oliveira, D. M., de Cerqueira, M. D.,

& El-Bachá, R. S. (2011). Oxidative stress in neurodegenerative diseases:

mechanisms and therapeutic perspectives. Oxidative Medicine and Cellular

Longevity, 2011.‏

Michael C., Rachel L., Jovana P., Jill J., Cornelia M., and Frederik N. (2008).

A mathematical model of glutathione metabolism. Theoretical Biology and

Medical Modelling, 5:8.

Miller R., James K., Sun G., Sun A. (2009) Oxidative and inflammatory

pathways in Parkinson’s disease. Neurochem, (34): 55-65.

Page 93: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

Mourya V. & Inamdar N. (2008) Chitosan-modifications and applications:

opportunities galore. Reactive and Functional polymers, 68(6): 1013-1051.

Muzzarelli, R. A. (1996). Chitosan-based dietary foods. Carbohydrate

Polymers, 29(4), 309-316.‏

Muzzarelli, R. A. A., & Muzzarelli, C. (2005). Chitosan chemistry: Relevance

to the biomedical sciences. In Polysaccharides I (pp. 151-209). Springer

Berlin Heidelberg.‏

Obrador, E., Benlloch, M., Pellicer, J. A., Asensi, M., & Estrela, J. M. (2011).

Intertissue Flow of Glutathione (GSH) as a Tumor Growth-promoting

Mechanism Intertissue flow of glutathione (GSH) as a tumor growth-

promoting mechanism: interleukin 6 induces GSH release from hepatocytes in

metastatic B16 melanoma-bearing mice. . Journal of Biological

Chemistry, 286(18), 15716-15727.‏

Pereira-Rodrigues, N., Cofré, R., Zagal, J. H., & Bedioui, F. (2007).

Electrocatalytic activity of cobalt phthalocyanine CoPc adsorbed on a graphite

electrode for the oxidation of reduced l-glutathione (GSH) and the reduction of

its disulfide (GSSG) at physiological pH. Bioelectrochemistry, 70(1), 147-154.‏

Pirovano C., Balzarini A., Bohm S., Oriana S., Spatti G., Zunino F. (1992)

Peripheral neurotoxicity following high-dose cisplatin with glutathione:

clinical and neurophysiological assessment. Tumori, 78(4): 253-7.

Pompella, A., Visvikis, A., Paolicchi, A., Tata, V. D., & Casini, A. F. (2003).

The changing faces of glutathione, a cellular protagonist. Biochemical

Pharmacology, 66(8), 1499-1503.

Page 94: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

Pooria G., Tahereh T., Moghadam, and Bijan R. (2010) Differential Scanning

Calorimetry Techniques: Applications in Biology and Nanoscience Biomol

Tech. 21(4): 167-193.

Rabea E., Badawy M., Stevens C., Smagghe G. & Steurbaut W. (2003)

Chitosan as antimicrobial agent: applications and mode of action.

Biomacromolecules. 4(6): 1457-1465.‏

Ravi Kumar, M. N. (2000). A review of chitin and chitosan

applications. Reactive and Functional Polymers, 46(1), 1-27.‏

Ravi MN., Bakowsky U., Lehr CM. (2004) Preparation and characterization of

cationic PLGA nanospheres as DNA carriers. Biomaterials, (25): 1771-1777.

Reverchon E, Adami R. (2006) Nanomaterials and supercritical Fluids. The

Journal of Supercritical Fluids; 37:1-22.

Rolland JP., Maynor BW., Euliss LE., Exner AE., Denison GM., DeSimone

JM. (2005) Direct fabrication and harvesting of monodisperse shape-specific

nanobiomaterials. Journal of the American Chemical Society, 127(28):10097-

10100.

Routes for Drug Administration. Emergency Treatment Guidelines Appendix.

Manitoba Health. 2003. Retrieved April 2, 2013.

Sechi, G., Deledda, M. G., Bua, G., Satta, W. M., Deiana, G. A., Pes, G. M., &

Rosati, G. (1996). Reduced intravenous glutathione in the treatment of early

Parkinson's disease. Progress in Neuro-Psychopharmacology and Biological

Psychiatry, 20(7), 1159-1170.‏

Shahidi, F., Arachchi, J. K. V., & Jeon, Y. J. (1999). Food applications of

chitin and chitosans. Trends in Food Science & Technology, 10(2), 37-51.‏

Page 95: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

Shur B. (2007) Prepartion of chitosan and its use in defluorination of wate

(Doctoral dissertation).

Staack, R., Kingston, S., Wallig, M. A., & Jeffery, E. H. (1998). A comparison

of the individual and collective effects of four glucosinolate breakdown

products from brussels sprouts on induction of detoxification

enzymes. Toxicology and Applied Pharmacology, 149(1), 17-23.‏

Tang, L., Persky, A. M., Hochhaus, G., & Meibohm, B. (2004).

Pharmacokinetic aspects of biotechnology products. Journal of

Pharmaceutical Sciences, 93(9), 2184-2204.‏

The Administration of Medicines. Nursing Practice Clinical Zones:

Prescribing. NursingTimes.net. 2007. Retrieved April 2, 2013.

Toba, G., & Aigaki, T. (2000). Disruption of the Microsomal glutathione S-

transferase-like gene reduces life span of Drosophila

melanogaster.Gene, 253(2), 179-187.‏

Townsend, D. M., Tew, K. D. & Tapiero, H. (2003) The importance of

glutathione in human disease. Biomed. Pharmacother. 57: 145–155.

Townsend, D. M., Tew, K. D., & Tapiero, H. (2003). The importance of

glutathione in human disease. Biomedicine & Pharmacotherapy, 57(3), 145-

‏.155

Usberti M., Lima G., Arisi M., Bufano G., D'Avanzo L. (1997) Effect of

exogenous reduced glutathione on the survival of red blood cells in

hemodialyzed patients. Journal of Nephrology, 10(5): 261-265.

Vincent M. and Duncan R. (2006) Polymer conjugates: Nanosized conjugates

for treated cancer. Trends Biotechnol, 24(1): 39-47

Page 96: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

Vincenzini, M. T., Iantomasi, T., & Favilli, F. (1989). Glutathione transport

across intestinal brush-border membranes: effects of ions, pH, Δψ, and

inhibitors. Biochimica et Biophysica Acta (BBA)-Biomembranes, 987(1), 29-

‏.37

Witschi, A., Reddy, S., Stofer, B., & Lauterburg, B. H. (1992). The systemic

availability of oral glutathione. European Journal of Clinical

Pharmacology, 43(6), 667-669.‏

Wu G, Fang Y., Yang S., Lupton J., Turner N. (2004) Glutathione Metabolism

and Its Implications for Health. Journal of Nutrition, (134): 489-492.

Xiangming G.,Brianna H., Chandradhar D.,Duane P. (2002) A simultaneous

liquid chromatography/mass spectrometric assay of glutathione, cysteine,

homocysteine and their disulfides in biological samples. Journal of

Pharmaceutical and Biomedical Analysis, (31): 251-261.

Yancheva E., Paneva D., Maximova V., Mespouille L., Dubois P., Manolova

N. (2007) Polyelectrolyte complexes between (cross-linked) N-

carboxyethylchitosan and (quaternized) poly[2-(dimethylamino)ethyl

methacrylate]: preparation, characterization, and antibacterial properties.

Journal Biomacromolecule,. 8(3):976-84.

Yih, T. C., & Al‐Fandi, M. (2006). Engineered nanoparticles as precise drug

delivery systems. Journal of Cellular Biochemistry, 97(6), 1184-1190.‏

Yokoyama M., Fukushima S., Uehara R., Okamoto K., Kataoka K., Sakurai

Y., and Okano T. (2003) Characterization of physical entrapment and

chemical conjugation of adriamycin in polymeric micelles and their design for

Page 97: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

in vivo delivery to a solid tumor. Journal of Controlled Release, 50(1–3):79-

92.

Yousefpour, P., Atyabi, F., Dinarvand, R., & Vasheghani-Farahani, E. (2011).

Preparation and comparison of chitosan nanoparticles with different degrees of

glutathione thiolation. DARU: Journal of Faculty of Pharmacy, Tehran

University of Medical Sciences, 19(5), 367.‏

Page 98: Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles · 2015-11-02 · Enhancing Oral Delivery of Glutathione Using Chitosan Nanoparticles By Zainab A. Alobaidy A Thesis

References

88

ناوسيتكاالالنانووية تحفيز توصيل الجلوتاثيون عبر الفم باستخدام الجزيئات

الخالصة

‏التوافر‏ ‏وزيادة ‏المختزل ‏الجلوتاثيون ‏مع ‏الكايتوسان ‏من ‏المركب ‏تكون ‏دراسة ‏هو ‏البحث ‏هذا ‏الهدف‏من ان

‏.الجزيئات‏النانووية‏‏المحمولة‏في‏نظام‏الزيتي عن‏طريق‏الفم‏باستخدامللجلوتاثيون‏البيولوجي‏

‏متضاعف‏ا ‏الطيفيواستخدمت‏ثالث‏تقنيات‏لقياس‏لدراسة ‏التحليل ‏المسح‏‏و،((FT-IRلكتروليتي‏معقدة مسعر

الجزيئات‏النانووية‏‏بينما‏لتقييم‏(.‏NMR 1H)‏مطيافية‏الرنين‏المغناطيسي‏النووي‏للبروتينو،‏(DSC)‏التبايني

ت‏الجزيئاماية‏ح،‏ودرجة‏‏احتمالية‏زيتا و‏,قمنا‏بقياس‏حجم‏الجزيئات‏النانووية‏‏المحمولةجلوتاثيون‏المحمولة‏لل

‏ةالكتلة‏الطيفي-مستويات‏الجلوتاثيون،‏حقق‏هذا‏التقييم‏باستخدام‏السائل‏اللونيل‏مدى‏رفعهلجلوتاثيون‏والنانووية‏‏ل

(HPLC-MS‏)‏.الجلوتاثيون‏لتحليل

‏ا‏بين‏الكتروليتي‏متضاعف‏مجمع‏تكوين‏تم‏القياس‏انه‏نتائج‏أكدت ‏‏الشيتوزان‏ولجلوتاثيون ‏التجارب‏وكذلك.

في‏‏‏الجلوتاثيونعلى‏رفع‏مستوى‏‏الجزيئات‏النانووية‏‏المحمولة‏في‏نظام‏الزيتي‏درةان‏اكدت‏قعلى‏الفئر‏المجراة

‏.الدورة‏الدموية