production and characterization of pectinase …

95
PRODUCTION AND CHARACTERIZATION OF PECTINASE INDUCED FROM ASPERGILLUS NIGER USING PECTIN EXTRACTED FROM PINEAPPLE PEELS AS CARBON SOURCE BY IBEAWUCHI, ADIELE NORA PG/M.Sc/10/57599 DEPARTMENT OF BIOCHEMISTRY UNIVERSITY OF NIGERIA NSUKKA SEPTEMBER, 2012.

Upload: others

Post on 02-Dec-2021

3 views

Category:

Documents


0 download

TRANSCRIPT

PRODUCTION AND CHARACTERIZATION OF PECTINASE INDUCED

FROM ASPERGILLUS NIGER USING PECTIN EXTRACTED FROM

PINEAPPLE PEELS AS CARBON SOURCE

BY

IBEAWUCHI, ADIELE NORA

PG/M.Sc/10/57599

DEPARTMENT OF BIOCHEMISTRY

UNIVERSITY OF NIGERIA NSUKKA

SEPTEMBER, 2012.

TITLE PAGE

PRODUCTION AND CHARACTERIZATION OF PECTINASE INDUCED FROM

ASPERGILLUS NIGER USING PECTIN EXTRACTED FROM PINEAPPLE PEELS AS

CARBON SOURCE.

A PROJECT WORK SUBMITTED IN PARTIAL FULFILMENT OF THE REQUIREMENT

FOR THE AWARD OF DEGREE OF MASTER OF SCIENCE (M.Sc) IN BIOCHEMISTRY,

UNIVERSITY OF NIGERIA NSUKKA.

BY

IBEAWUCHI, ADIELE NORA

PG/M.Sc/10/57599

DEPARTMENT OF BIOCHEMISTRY

UNIVERSITY OF NIGERIA NSUKKA

SUPERVISORS: PROF. F.C. CHILAKA AND DR. S.O.O. EZE

SEPTEMBER, 2012

CERTIFICATION

Ibeawuchi, Adiele Nora, a post-graduate student with registration number PG/M.Sc/10/57599 in

the Department of Biochemistry has satisfactorily completed the requirement for the course work

and research for the Master Degree of Science (M.Sc) in Biochemistry with Industrial

Biochemistry and Biotechnology as an option. This entire work in this report is original and has

not been submitted in part or full for any other diploma or degree in this or any other university.

__________________________ _______________________

Prof. F.C. Chilaka Dr S.O.O. Eze

(Chief Supervisor) (Co-Supervisor)

____________________________ ________________________

Prof. L.U.S. Ezeanyika External Examiner

DEDICATION

This work is dedicated to the members of my family, for their endless support and prayers. To

my parents Mr & Mrs D.E. Ibeawuchi and siblings; Ijeoma Ibeawuchi, Chinyere Nwanya,

Chinwe Osuagwu, Uloma Ibeawuchi and Victor Ibeawuchi; I say thank you.

ACKNOWLEDGEMENT

I would like to use this means to appreciate my wonderful supervisors; Prof F.C. Chilaka and Dr.

S.O.O. Eze, for their relentless effort in bringing this work to completion. I admire their patience,

hardwork and dedication. To Dr C.U. Anyanwu, I appreciate your generosity by allowing me

carry-out the microbiological aspect of this research in your laboratory.

To the HOD, Prof. L.U.S. Ezeanyika, I thank you for being a man of great honor and living an

exemplary life. To all my lecturers, thank you for imparting knowledge unto me from your

individual wells of experience and understanding. Dr J. Parker, you were always there to correct

and give me guidance whenever I fell short; you are appreciated.

Without a doubt, I thank Dr P. Mounmbegna, Dr F.M Awah and Dr S. Onuoha for being of

assistance when I considered enrolling for this programme. My colleagues; Nsude Nonso, Ezike

Tobechukwu, Ezugwu Arinze, Nwamaka Odu, Agu Chidozie and Okonkwo Chukwudi were

supportive and I thank them for their intelligent input in this work. My friends; Amaka Nwoye,

Onyedikachi Aruma, Preye Wokoro and Olufemi, you gave me a reason to smile every day. The

entire Biochemistry Post-Graduate class of 2010, we sure had an experience that made us better

people today.

I also would love to acknowledge my parents, siblings and their spouses for believing in me and

encouraging me throughout the entire M.Sc programme. My nephews; Chinomso and Chidozie

Osuagwu, Obinna Nwanya and Lewechi Ibeawuchi, you understood when I was always absent

on those special days. I love you all.

I ultimately want to thank God for HIS grace and mercies upon my life. He has been more than

faithful even when tough times prevailed and no silver lining was in sight; You still gladdened

my heart.

ABSTRACT

The peels of pineapple (Ananas cosmosus) generated as agricultural wastes represent about 35%

of the entire fruit. However, these wastes can be converted to various forms or more useful

products such as pectin and dietary fibres. Pectin was extracted from pineapple peels with a

percentage yield of 8.33% at pH 2.2 and temperature of 70ºC. Three pectinolytic fungi:

Aspergillus niger, Aspergillus fumigatus and Aspergillus flavus were isolated from natural

sources and tested for their pectinolytic activity. Aspergillus niger showed relatively higher

pectinolytic activity and was therefore used for further studies. Aspergillus niger was inoculated

into a broth containing pineapple pectin under submerged fermentation system. The enzyme was

harvested after four days of fermentation, the biomass was filtered off and 80% ammonium

sulphate precipitation was carried out on the crude enzyme. The crude enzyme was desalted

through dialysis and yielded a 4- fold increase in specific activity. The pH and temperature

optima were found to be 5.5 and 55ºC respectively. The Vmax and Km were also found to be

5000U and 89.5mg/ml respectively. Thus, pineapple peels can be successfully employed in the

production of pectinase.

TABLE OF CONTENTS

Title Page-------------------------------------------------------------------------------------I

Certification----------------------------------------------------------------------------------II

Dedication------------------------------------------------------------------------------------III

Acknowledgement--------------------------------------------------------------------------IV

Abstract---------------------------------------------------------------------------------------V

Table of Contents---------------------------------------------------------------------------VI

List of Figures-------------------------------------------------------------------------------VII

List of Tables -------------------------------------------------------------------------------XIV

List of Abbreviations-----------------------------------------------------------------------XV

CHAPTER ONE: INTRODUCTION

1.1 Pineapple- scientific classification------------------------------------------------2

1.1.1 History and description of pineapple----------------------------------------------3

1.1.2 Plant cell wall------------------------------------------------------------------------6

1.1.2.1 The middle lamella of the fruit cell------------------------------------------------6

1.2 Pectic substances----------------------------------------------------------------------7

1.2.1 Structure of pectic substances------------------------------------------------------8

1.2.2 Classification of pectic substances-------------------------------------------------9

1.3 Pectin----------------------------------------------------------------------------------10

1.3.1 Structural types of pectin-----------------------------------------------------------11

1.3.2 The primary cell wall pectic network---------------------------------------------13

1.3.3 Types of pectin-----------------------------------------------------------------------14

1.3.4 General properties of pectins-------------------------------------------------------15

1.3.5 Applications of pectin---------------------------------------------------------------15

1.3.5.1 Muco-adhesive polymer-------------------------------------------------------------15

1.3.5.2 Gelling agent, thickener &water binding-----------------------------------------15

1.3.5.3 Pectin in medicine and pharmaceutical industry--------------------------------16

1.4 Role of microbes in pectinase production----------------------------------------16

1.5 Pectinases-----------------------------------------------------------------------------17

1.5.1 Protopectinases----------------------------------------------------------------------18

1.5.2 Polygalacturonases------------------------------------------------------------------21

1.5.3 Lyases---------------------------------------------------------------------------------22

1.5.4 Pectinesterases-----------------------------------------------------------------------24

1.6 Biotechnological application of microbial pectinases--------------------------26

1.6.1 Fruit juice extraction----------------------------------------------------------------27

1.6.2 Coffee and tea fermentation-------------------------------------------------------27

1.6.3 Textile processing and bioscouring of cotton fibers----------------------------27

1.6.4 Degumming of plant bast fibers---------------------------------------------------27

1.6.5 Waste water treatment--------------------------------------------------------------28

1.6.6 Paper and pulp industry------------------------------------------------------------28

1.6.7 Animal feed--------------------------------------------------------------------------28

1.6.8 Purification of plant viruses--------------------------------------------------------28

1.6.9 Improvement of chromaticity and stability of red wines-----------------------28

1.7 Substrates for the production of pectinases--------------------------------------29

1.8 Fermentation conditions------------------------------------------------------------29

1.8.1 Types of fermentation--------------------------------------------------------------30

1.9 Micro-organisms commonly used in submerged and solid--------------------31

state fermentation for pectinases production

1.10 Aim and objectives of the study--------------------------------------------------32

1.10.1 Aim of the study---------------------------------------------------------------------32

1.10.2 Specific objectives of the study----------------------------------------------------32

CHAPTER TWO: MATERIALS AND METHODS

2.1 Materials-------------------------------------------------------------------------------33

2.1.1 Chemicals / Reagents----------------------------------------------------------------33

2.1.2 Apparatus / Equipment--------------------------------------------------------------33

2.1.3 Collection of pineapple samples---------------------------------------------------33

2.1.4 Collection of micro-organisms-----------------------------------------------------34

2.2 Methods-------------------------------------------------------------------------------34

2.2.1 Preparation of reagents--------------------------------------------------------------34

2.2.1.1 Preparation of 3N HCl--------------------------------------------------------------34

2.2.1.2 Preparation of ethanol-HCl solution----------------------------------------------34

2.2.1.3 Preparation of buffers---------------------------------------------------------------35

2.2.1.4 Preparation of 2mg/ml bovine serum albumin (BSA)--------------------------35

standard protein

2.2.1.5 Preparation of dinitrosalicylic acid (DNS) reagent-----------------------------35

2.2.1.6 Preparation of 20mM galacturonic acid------------------------------------------36

2.2.1.7 Preparation of component reagents for protein determination---------------36

2.2.2 Preparation of ground pineapple peels-------------------------------------------36

2.2.3 Extraction of pectin from pineapple peels---------------------------------------36

2.2.4 Isolation of pectinolytic fungi-----------------------------------------------------37

2.2.4.1 Collection of soil samples----------------------------------------------------------37

2.2.4.2 Preparation of soil sample extracts for microbial isolation-------------------37

2.2.4.3 Preparation of the solid medium--------------------------------------------------37

2.2.4.4 Sub-culturing onto solid medium-------------------------------------------------38

2.2.4.5 Storage of micro-organism on potato dextrose agar (PDA)------------------38

2.2.4.6 Microscopic features of the isolated fungi--------------------------------------38

2.2.4.7 Fungal identifications--------------------------------------------------------------38

2.2.5 Fermentation experiments---------------------------------------------------------38

2.2.5.1 Preparation of the fermentation medium----------------------------------------38

2.2.5.2 Inoculating with Aspergillus niger-----------------------------------------------39

2.2.5.3 Harvesting the crude enzyme-----------------------------------------------------39

2.2.6 Galacturonic acid standard curve-------------------------------------------------39

2.2.7 Polygalacturonase assay-----------------------------------------------------------40

2.2.7.1 Procedure for polygalacturonase assay------------------------------------------40

2.2.8 Protein determination--------------------------------------------------------------40

2.2.8.1 Procedure for protein determination---------------------------------------------40

2.2.9 Partial purification of protein-----------------------------------------------------41

2.2.9.1 Ammonium sulphate precipitation profile---------------------------------------41

2.2.9.2 Ammonium sulphate precipitation------------------------------------------------41

2.2.9.3 Dialysis-------------------------------------------------------------------------------41

2.2.10 Studies on partially purified enzymes--------------------------------------------42

2.2.10.1 Effect of pH change on pectinase activity---------------------------------------42

2.2.10.2 Effect of temperature change on pectinase assay-------------------------------42

2.2.10.3 Effect of substrate concentration on pectinase assay---------------------------42

2.2.10.4 Further studies with partially purified enzyme----------------------------------42

CHAPTER THRE: RESULTS

3.1 Pineapple pectin extraction--------------------------------------------------------43

3.1.1 Pineapple pectin extraction yield-------------------------------------------------43

3.1.2 Photograph of pineapple pectin extract------------------------------------------43

3.2 Micro-organisms--------------------------------------------------------------------44

3.2.1 Selection of pectinolytic fungi----------------------------------------------------44

3.2.2 Macroscopic and microscopic examination of fungal isolates----------------45

3.3 Pectinases production undersubmerged fermentation system----------------46

3.4 Ammonium sulphate precipitation-----------------------------------------------47

3.5 Assays carried out on the pectinases obtained----------------------------------48

3.5.1 Protein concentration of the crude, precipitated--------------------------------48

and dialyzed enzymes

3.5.2 Activity of pectinase in the crude, precipitated---------------------------------49

and dialyzed forms

3.5.3 Total protein content of the crude, precipitated---------------------------------50

and dialyzed pectinases

3.5.4 Total activity of the crude, precipitated------------------------------------------51

and dialyzed pectinase

3.5.5 Specific activity of the crude, precipitated--------------------------------------52

and dialyzed pectinases

3.5.6 Purification folds of the partially purified enzymes---------------------------53

3.6 Characterization of pectinase-----------------------------------------------------55

3.6.1 Effect of change in pH on pectinase activity------------------------------------55

3.6.2 Effect of change in temperature on pectinase activity-------------------------56

3.6.3 Kinetic parameters------------------------------------------------------------------57

CHAPTER FOUR: DISCUSSION

4.1 Discussion--------------------------------------------------------------------------------59

4.2 Conclusion-------------------------------------------------------------------------------62

4.3 Suggestions for further studies--------------------------------------------------------63

References------------------------------------------------------------------------------------64

Appendices-----------------------------------------------------------------------------------79

LIST OF FIGURES

Fig.1: Parts of a pineapple fruit-------------------------------------------------------------------4

Fig.2: Structure of the plant cell wall------------------------------------------------------------6

Fig. 3: Primary structure of pectic substances---------------------------------------------------9

Fig. 4: Structure of galacturonic acid------------------------------------------------------------10

Fig. 5: Schematic representative structures of the constituent--------------------------------11

polysaccharides of pectin

Fig. 6: HM pectin formula-------------------------------------------------------------------------14

Fig. 7: LM pectin formula-------------------------------------------------------------------------14

Fig. 8: Amidiated pectin formula-----------------------------------------------------------------14

Fig. 9: Enzymatic mode of action of some of the most frequent------------------------------20

depolymerases on the pectin molecule

Fig. 10: Photograph of pineapple pectin---------------------------------------------------------43

Fig. 11: Pure culture of Aspergillus niger-------------------------------------------------------45

Fig. 12: Ammonium sulphate precipitation profiling of pectinase----------------------------47

obtained from Aspergillus niger

Fig. 13: Comparison in the protein concentration of the crude,-------------------------------48

precipitated and dialyzed pectinases

Fig. 14: Comparison in the activities obtained from the crude,-------------------------------49

precipitated and dialyzed pectinases

Fig. 15: Comparison in the total protein content of the crude,---------------------------------50

precipitated and dialyzed

Fig.16: Comparison in the total activity of the crude,------------------------------------------51

Precipitated and dialyzed pectinases

Fig. 17: Comparison in the specific activities of the crude,------------------------------------52

precipitated and dialyzed pectinases

Fig. 18: Purification folds of the crude, precipitated and dialyzed pectinases--------------53

Fig. 19: Effect of change in pH on pectinase activity------------------------------------------55

Fig. 20: Effect of change in temperature on pectinase activity-------------------------------56

Fig. 21: Lineweaver-Burk plot of pectinases from Aspergillus niger----------------------57

LIST OF TABLES

Table 1: Nutritional values per 100g of pineapple------------------------------------------5

Table 2: Composition of pectic substances in different------------------------------------8

fruits and vegetables

Table 3: Extensive classification of pectinolytic enzymes--------------------------------19

Table 4: Biochemical and physicochemical properties of some-------------------------22

polygalacturonases

Table 5: Biochemical and physicochemical properties of--------------------------------24

some lyases

Table 6: Biochemical and physicochemical properties of --------------------------------26

some pectinesterases

Table 7: Comparison of solid state and submerged-----------------------------------------32

Fermentation for pectinases production

Table 8: Summary of the parameters determined from the crude------------------------54

precipitated and dialyzed pectinases

Table 9: Summary of pectinase characterization-------------------------------------------58

LIST OF ABBREVIATIONS

AG- Arabinogalactan

Ala- Alanine

Api- Apiogalacturonan

Ara- Arabinan

BSA- Bovine Serum Albumin

GRAS- Generally Regarded as Safe

HGA- homogalacturonan

HM- High Methylester

LM- Low Methylester

PDA- Potato Dextrose Agar

PE- Pectinesterase

PG- Polygalacturonase

PGase- Polygalacturonase

PGL- PolygalacturonateLyase

PME- Pectin methylesterase

PMG- polymethylgalacturonase

PMGE- Polymethylgalacturonate Esterase

PMGL- PolymethylgalacturonateLyase

Ppase- Protopectinase

Pro- Proline

RG- Rhamnogalacturonan

Rha- Rhamnose

Ser- Serine

SmF- Submerged fermentation

SSF- Solid state fermentation

Thr- Threonine

UDP-D- Uridinediphosphate

XGA- Xylogalacturonan

Xyl- Xylose

β-Gal- β-Galactosidase

CHAPTER ONE

INTRODUCTION

Pineapple (Ananas cosmosus) is the common name for a tropical plant and its edible fruits which

is actually a multiple fruit consisting of coalesced berries (Purseglove, 1972; Bartholomew et al.,

2003). Pineapple is by far the most economically important plant in the Bromeliaceae family.

Besides being produced for consumption, it can be grown as an ornamental or house plant

(Bartholomew et al., 2003). Pineapple can be consumed fresh, canned or juiced and can be used

in a variety of ways. It is popularly used in salads, jams, yoghurts, ice-cream, etc (Rohrbach et

al., 2003). However, processing and utilization of pineapple into various products leads to

generation of waste in form of peels and pomace. Pineapple waste can be conventionally bio-

transformed anaerobically into humus; although valuable by-products can be produced from the

rich waste. In other words, wealth can be derived from this waste by value addition and products

such as pectin, dietary fibers and predominantly pectinases can be easily harnessed.

Pectin was first isolated and described in 1825 by Henri Braconnot (Braconnot and Keppler,

1825). Pectin is one of the major components of the primary cellular walls in the middle lamella

of plant tissues. The pectic matrix provides an environment for deposition, slippage and

extension of the cellulosic-glycan network, and is the major adhesive materials between cells

(Willats et al., 2001). Pectin degradation leads to disassembly of the cellulose and hemicellulose

network and plays an important role in fruit ripening (Lohani et al., 2004; Sañudo-Barajas et al.,

2009). Wall degrading enzymes were the major factors to initiate disassembly of cellular walls of

harvested fruits (Miller and Fry, 2001). Involvement of pectic enzymes, such as

polygalacturonase (PG), pectin methyl esterase (PME) and β-galactosidase (β-Gal), in enzymatic

disassembly of cellular walls has been widely reported (Prasanna et al., 2007; Nikolić and

Mojovic, 2007; Rugkong et al., 2010; Wei et al., 2010; Almeida and Huber, 2011). In the food

sector, pectins are primarily used as a gelling agent in the production of jam; and as stabilizers in

the production of yoghurt (Koubala et al., 2006). In the pharmaceutical industry, these

polysaccharides are used as drug delivery systems, which can also reduce the toxicity of the

drugs (Morris et al., 2010; Pilnik et al., 1970; Schols et al., 2009; Thakur et al., 1997).

Pectinases are a group of enzymes, which cause degradation of pectin that, are chain molecules

with a rhamnogalacturonan backbone; associated with other polymers and carbohydrates. These

pectinases have wide applications in fruit juice industry and wine industry. In fruit juice industry,

it is used for clarification; reduction in viscosity is caused which ultimately leads to formation of

clear juice. They increase the yield of juices by enzymatic liquefaction of pulps; these pectinases

also helps in formation of pulpy products by macerating the organized tissue into suspension of

intact cells. In wine industry pectinases are mainly used for decreasing astringency by

solubilizing anthocyanins without leaching out procyadin polyphenols, and pectinases also

increase pigmentation by extracting more anthocyanins (Tucker and Woods, 1991). The

increasing energy demand has been focused on the utilization of renewable agricultural and

industrial wastes (Martin et al., 2004).

Pectinases can be produced by both submerged and solid state fermentation (SSF). Submerged

fermentation is cultivation of microorganisms on liquid broth. It requires high volumes of water,

continuous agitation and generates lot of effluents. SSF incorporates microbial growth and

product formation on or within particles of a solid substrate (Mudgett, 1986) under aerobic

conditions, in the absence or near absence of free water, and does not generally require aseptic.

1.1 Pineapple- Scientific Classification

Kingdom Plantae

Phylum Pteridofitae

Class Angiosperm

Sub-class Monocotyledoneal

Order Farinosae

Family Bromaliaceae

Sub-family Bromelioideae

Genus Ananas

Species Cosmosus

Binomial name Ananas cosmosus

Source: Py and Tisseau, 1969)

1.1.1 History and Description of Pineapple

Pineapple (Ananas comosus) is the common name for a tropical plant and its edible fruit, which

is actually a multiple fruit consisting of coalesced berries. It was given the name pineapple due to

its resemblance to a pine cone. The pineapple is by far the most economically important plant in

the Bromeliaceae family. (Coppens d‟Eeckenbrugge and Leal, 2003). Besides being produced for

consumption, it can be grown as an ornamental or houseplant, obtained from the crown of a

supermarket fruit.

Some sources say that the plant will flower after about 24 months and produce a fruit during the

following six months (Purseglove, 1972), while others indicate a 20-month timetable. Pineapple

can be consumed fresh, canned or juiced and can be used in a variety of ways. It is popularly

used in desserts, salads (usually tropical fruit salads, but it can vary), jams, yogurts, ice creams,

various candies, as a complement to meat dishes and in fruit cocktail. (Rohrbach et al., 2003).

The popularity of the pineapple is due to its sweet-sour taste. The core of the pineapple is

continuous with the stem supporting the fruit and with the crown, a feature unique among

cultivated fruits.

Pineapple contains the proteolytic enzyme bromelain which is used as a meat tenderizing agent

and for medicinal purposes. It has been reported to have valuable biological properties such as

interfering with the growth of malignant cells, inhibiting platelet aggregation, fibrinolytic and

anti-inflammatory actions. (Gailhofer et al., 1998). Pineapple leaf juice is used as a purgative

(agent that cleanses the bowel), emmenagogue (agent that induces menstrual bleeding) and

vermifuge (agent that expels intestinal worms). (Leal and Coppens d‟Eeckenbrugge, 1996).

The stems and leaves of the pineapple plant are sources of fiber, which can be processed into

paper and cloth. The cloth made from pineapple fiber is known as „pina cloth‟ and was in use as

early as 1571. Parts of the pineapple plant (Fig. 1) are used as silage and hay for cattle feed such

as the processed wastes in the form of pomace or centrifuged solids from juice production

(Wikipedia, 2011)

Fig. 1: Parts of a pineapple fruit (Elfick, 2007)

The word “pineapple” in English was first recorded in 1398, when it was originally used to

describe the reproductive organs of conifer trees (now termed pine cones). The term pine cone

for the reproductive organ of conifer trees was first recorded in 1694. When European explorers

discovered this tropical fruit, they called them pineapples (term first recorded in that sense in

1664 because of their resemblance to what is now known as the pine cone (Wikipedia, 2011)

Table 1: Nutritional Value per 100g of Pineapple Fruits

Nutrient Nutritional Value per 100g (3.5oz)

Energy 202kJ (48kcal)

Carbohydrate 12.63g

Sugar 9.26g

Dietary Fiber 1.4g

Fat 0.12g

Protein 0.54g

Thiamine (Vit B1) 0.079mg (3%)

Riboflavin (Vit B2) 0.03mg (3%)

Niacin (B3) 0.489mg (3%)

Panthothenic Acid( B5) 0.205

Vitamin B6 0.11mg (8%)

Folate (B9) 15µg (4%)

Vitamin C 36.2mg (44%)

Calcium 13mg (1%)

Iron 0.28mg (2%)

Magnesium 12mg

Source: Wikipedia, 2011

1.1.2 Plant Cell Wall

Plant cell walls consist of plant middle lamella, primary cell wall and secondary cell wall as can

be seen in Fig. 2. The primary walls of enlarging plant cells are composed of approximately 30%

cellulose, 30% hemicellulose and 35% pectin with about 1-5% structural protein (glycoprotein)

on a dry weight basis (Cosgrove, 1997).

Fig. 2: Structure of the Plant Cell Wall (Carpita and Gibeaut, 1993)

1.1.2.1 The Middle Lamella of the Fruit Cell

The middle lamella is the first layer formed during cell division, and can also be seen as the

space between the cell walls, and as the connecting region between adjacent cells, binding cells

together. The highest concentrations of pectin are found in the middle lamella of cell walls, with

a gradual decrease as one passes through the primary wall toward the plasma membrane

(Kertesz, 1951).

In the process of the middle lamella formation, the methyl-esterified homogalacturonan (HGA)-

rich carbon brushes or pectin with virtually no arabinan or galactan are deposited first in the cell

plate of two daughter cells. Subsequently, more layers of pectin molecules are deposited. There

may be de-esterification of HGA hairs by the activity of intact pectin methylesterase (PME). The

unesterified regions of HGAs from different cells become sensitive to Ca2+

and can form gel

(Vicken et al., 2003). Cosgrove (1997) reported the localization of PME and Ca2+

-pectate in the

areas of cell-cell contact such as the middle lamella. Hence, the middle lamella is a Ca2+

-pectate

gel formed by the cross-linking of first layer pectin (HGA-rich molecular brushes) to HGA-

containing molecular brushes of the primary cell wall of two neighboring cells (Vicken et al.,

2003).

1.2 Pectic Substances

Pectic substance is the generic name used for the compounds that are acted upon by the

pectinolytic enzymes. They are high molecular weight, negatively charged, acidic, complex

glycosidic macromolecules (polysaccharides) that are present in the plant kingdom. They are

present as the major components of middle lamella between the cells in the form of calcium

pectate and magnesium pectate (Rastogi, 1998). The synthesis of pectic substances occurs in the

Golgi apparatus from UDP-D-galcturonic acid during early stages of growth in young enlarging

cell walls (Sakai et al., 1993).

Compared with young, actively growing tissues, lignified tissues have a low content of pectic

substances. The content of the pectic substances is very low in higher plants usually less than

1%. They are mainly found in fruits and vegetables, constitute a large part of some algal biomass

(up to 30%) and occur in low concentration in forestry or agricultural residues (Table 2). Pectic

substances account for 0.5–4.0% of the fresh weight of plant material (Kashyap et al., 2001;

Sakai et al., 1993). Contrary to the proteins, lipids and nucleic acids, which are polysaccharides,

pectic substances do not have defined molecular masses.

Table 2: Composition of pectic substances in different fruits and vegetables

Fruit/vegetable Tissue Pectic substance (%)

Apple Fresh 0.5–1.6

Banana Fresh 0.7–1.2

Peaches Fresh 0.1–0.9

Strawberries Fresh 0.6–0.7

Cherries Fresh 0.2–0.5

Peas Fresh 0.9–1.4

Carrots Dry matter 6.9–18.6

Orange pulp Dry matter 12.4–28.0

Potatoes Dry matter 1.8–3.3

Tomatoes Dry matter 2.4–4.6

Sugar beet pulp Dry matter 10.0–30.0

Source: Kashyap et al., 2001.

1.2.1 Structure of Pectic Substances

Pectic substances mainly consist of galacturonans and rhamnogalacturonans in which the C-6

carbon of galacturonic acids are oxidized to carboxyl groups, the arabinans and the

arabinogalactans as seen in Fig. 3 (Whitaker, 1990). These substances are a group of complex

colloidal polymeric materials, composed largely of a backbone of anhydrogalacturonic acid units

(Cho et al., 2001; Codner, 2001). The carboxyl groups of galacturonic acids are partially

esterified by methyl groups and partially or completely neutralized by sodium, potassium or

ammonium ions (Kashyap et al., 2001). The primary chain consists of D-galacturonic acid units

linked α-(1-4), with 2–4% of L-rhamnose units linked β-(1-2)and β-(1-4) to the galacturonic acid

units (Whitaker, 1990). The side chains of arabinan, galactan, arabinogalactan, xylose or fructose

are connected to the main chain through their C-1 and C-2 atoms (Blanco et al., 1999;

Sathyanarayama and Panda, 2003; Van der Vlugt-Bergmans et al., 2000). The above description

indicates that the pectic substances are present in various forms in plant cells and this is the

probable reason for the existence of various forms of pectinolytic enzymes.

Fig. 3: Primary Structure of Pectic Substances (Pilnik and Voragen, 1993)

1.2.2 Classification of Pectic Substances

According to Alkorta et al., 1998, the American Chemical Society classified pectic substances as

follows:

i) Protopectin: is the water insoluble pectic substance present in intact tissue. Protopectin on

restricted hydrolysis yields pectin or pectic acids.

ii) Pectic acid: is the soluble polymer of galacturonans that contains negligible amount of

methoxyl groups. Normal or acid salts of pectic acid are called pectates.

iii) Pectinic acids: is the polygalacturonan chain that contains >0 and <75% methylated

galacturonate units. Normal or acid salts of pectinic acid are referred to as pectinates.

iv) Pectin (Polymethyl galacturonate): is the polymeric material in which, at least, 75% of the

carboxyl groups of the galacturonate units are esterified with methanol. It confers rigidity on cell

wall when it is bound to cellulose in the cell wall.

1.3 Pectin

Through various studies, it has been brought in notice that the structure of pectin is difficult to

determine because pectin subunit composition can change during isolation from plants, storage

and processing of plant material (Novosd‟skaya, 2002). Pectin was first isolated and described in

1825 by Henri Braconnot (Braconnot and Keppler., 1825). At present, pectin is thought to

consist mainly of D-galacturonic acid (Gal A) units (Sriamornsak, 2002), joined in chains by

means of α (1-4) glycosidic linkage (Fig. 4). These uronic acids have carboxyl groups which are

naturally present as methyl esters and others which are commercially treated with ammonia to

produce carboxiamide group (Sriamornsak., 1998; Yujaroen et al., 2008).

Fig. 4: Structure of Galacturonic Acid (Pilnik and Voragen, 1993)

Pectin is composed of as many as 17 different monosaccharides (Ridley et al., 2001; Voragen et

al., 2003). These monosaccharides are organized in a number of distinct polysaccharides, the

structures of which are schematically shown in Fig. 5a→g. Together, these polymers form the

pectin network (Visser and Voragen, 1996; Ridley et al., 2001; Voragen et al., 2003).

Fig. 5(a→g): Schematic representative structures of the constituent polysaccharides of pectin.

The symbols for the various monosaccharide building units are explained in the accompanying

legend. (Vincken et al., 2003)

1.3.1 Structural Types of Pectin

Pectins are made up of a backbone of 1,4- linked α-D Galp A residues and are referred to as

galacturonans. Three types of galacturonan can be distinguished in the plant cell wall: the

unsubstituted homogalacturonan (HG), rhamnogalacturonan II (RG-II) and xylogalacturonan

(XGA) (Schols et al., 1995; Visser and Voragen, 1996). Addition of side chains bring about

other polysaccharides such as rhamnogalacturonan I (RG-I), arabinogalactan I and II (AG-I &

AG-II).

The Gal A residues of HG can be methyl-esterified at C-6 and carry acetyl groups on O- 2 and

O-3. This however affects to a large extent, the industrial applicability of pectins. Blocks of more

than 10 unesterified Gal A residues generally yield pectin molecules which are sensitive to Ca 2+

-

cross-linking (Daas et al., 2001). Two unesterified HG chains can engage in a complex

a)

d) e) f)

g)

b) c)

sometimes referred to as “egg boxes”, in which the carboxyl groups of two GalA residues form a

negatively charged pocket that can accommodate a Ca2+

cation (Braccini et al., 1999; Willats et

al., 2001a).

RG-II is not made of a rhamnogalacturonan backbone as the name implies, rather the rhamnose

residues are present in the side chains (Ridley et al., 2001). RG-II are formed when HG contain

clusters of four different side chains with very peculiar sugar residues such as Api, Ace A, Dha

and Kdo). These side chains together with the approximately nine galacturonyl residues to which

they are connected are referred to as RG II (O‟Neil et al., 2001; Ridley et al., 2001). Also the

order in which they follow on the backbone is not known with a certainty (du Penhoat et al.,

1999; Glushka et al., 2003; Perez et al., 2003)

XGA is a branched galacturonan with β-D-xylp-(1→3) side chains (Schols et al., 1995; Visser

and Voragen, 1996).

There is another constituent polysaccharide of pectin known as rhamnogalacturonan I (RG-I). It

is composed of a repeating disaccharide unit [→2),α-L-Rhap-(1→4)-α-D-Galp A-(1→]n, where

n can be larger than 100 (McNeil et al., 1980; Visser and Voragen, 1996). The galacturonyl

residues can carry acetyl groups on O-2 and O-3 while the rhamnosyl residues can be substituted

at O-4 with neutral sugars (McNeil et al., 1980; Lau et al., 1987). These neutral sugars are

mainly D- galactose, L- arabinose and D- xylose (Shembekar and Dohtre, 2009). The side chains

of RG-I can be single units (β-D-Galp-(1→4) or polymeric such as arabinogalactan I (AG-I)

which is composed of 1,4- linked β-D-Galp backbone or arabinan which consists of 1,5- linked

α-L-Araf backbone (Ridley et al., 2001).

Complexes of RG-I, AG-I and arabinan are often referred to as pectic hairy regions (HR) in

which AG-I and arabinan are the “hairs”. However, arabinogalactan II is mainly associated with

proteins (arabinogalactan proteins or AGPs) and it is still unclear whether this polysaccharide is

part of the pectin complex. Pectin and AG-II often seem to co-extract and are subsequently

difficult to separate from each other, suggesting that they can be covalently linked. AG-II is

composed of 1,3- linked β-D-Galp backbone and short side chains of α-L-Araf-(1→6)-[β-D-

Galp-(1→6)]n where n=1, 2 or 3 (Ridley et al., 2001). The major moiety of AGp (> 90%)

consists of polysaccharides while the protein moiety is rich in Pro, Ala, Ser and Thr (Gaspar et

al., 2001).

1.3.2 The primary cell wall pectic network

The covalent cross-linking of the pectic polysaccharides HG, RG-I, and RG-II has been

demonstrated repeatedly in the literature by the EPGase-dependent release of pectic

polysaccharides from the wall (O‟Neil et al., 1996). The available data suggest that the RG-I and

RG-II backbones are continuous with the HG backbone, not that of RG-I side chains, as

suggested by Vincken et al. (2003). If the backbones of the pectins are continuous, the pectic

network may be thought of as a macromolecular structure having specific domains of HG, RG-I,

and RG II, however, the arrangement of these domains in vivo is not known. The linkage of HG,

RG-I, and RG-II through backbone glycosidic linkages is just one possible way in which the

pectins are cross-linked.

The pectic network is based on multiple levels of cross-linking that include, but are not limited

to, backbone glycosidic linkages, calcium cross-linking, borate ester cross-linking, and covalent

linkages to phenolic and possibly other compounds. The HG domains of pectin may self-

associate depending on the degree of methylesterification and thus the affinity of HG for calcium

ions. RG-I has a unique backbone of alternating 2-linked Rhap and 4-linked GalpA residues.

Some rhamnose residues are branched by arabinan, galactan, and/or AG side chains (McNeil et

al., 1980) that may be cross-linked to other wall components such as xylans, xyloglucans,

proteins, and lignins. RG-II domains form cross-links to other RG-II molecules via borate di-

ester linkages, to form RG-II dimmers that contribute to wall strength and that affect pore size

and flexibility of the pectic network (Ishii and Matsunaga, 1996; Fleisher et al., 1999). Greater

than 95% of RG-II molecules participate in dimer complexes of RG-II (O‟Neil et al., 2004).

The linkages that pectic polysaccharides make to other pectins, as well as to other wall

molecules, combine to assemble the pectic network of the plant cell wall. The complexity of the

pectic network structure and the modulation of the pectic cross-links contribute strength,

flexibility, and functionality to the pectic network, and thus, to the primary cell wall.

1.3.3 Types of Pectin

The percentage of ester groups is called degree of esterification. High methyl ester pectins are

classified in groups according to their gelling temperature as rapid set to slow set pectins.

Pectin as extracted normally has more than 50% of the acid units esterified, and is classified as

"high methyl ester (HM) pectin" as shown in Fig. 6.

Fig. 6: HM pectin formula (IPPA, 2001)

Modification of the extraction process, or continued acid treatment, will yield ”low methyl ester

LM pectin" with less than 50% methyl ester groups as observed in Fig. 7.

Fig. 7: LM pectin formula (IPPA, 2001)

Some pectins are treated during manufacture with ammonia to produce amidated pectins, which

have particular advantages in some applications.

Fig. 8: Amidated pectin formula (IPPA, 2001)

1.3.4 General Properties of Pectins

Pectin is soluble in pure water as monovalent cation (alkali metal) salts of pectinic and pectic

acids; are usually soluble in water unlike di- and trivalent cation salts that are weakly soluble or

insoluble (Sriamornsak, 1998). Dilute pectin solutions are Newtonian but at a moderate

concentration, they exhibit the non-Newtonian, pseudo plastic behavior characteristics.

Viscosity, solubility and gelation are generally related. For example, factors that increases gel

strength will increase viscosity and vice versa (Marshal and Chow, 2007). As such, monovalent

cation salts of pectins are highly ionized in solution and the distribution of ionic charges along

the molecule tends to keep it in extended form by reason of coulombic repulsion (Paolettis,

1986). Dissolved pectins are decomposed spontaneously by de-esterification as well as by

depolymerization. Hence the rate of decomposition depends on pH, water activity and

temperature (Sriamornsak, 1998).

1.3.5 Applications of Pectin

1.3.5.1 Mucoadhesive Polymer

Different types of pectin were characterized for gastrointestinal (GI) mucoadhesion

(Sriamornsak, 2002). The mucoadhesive process involved in the formation of bioadhesive bonds

has been described in 3 steps;

i) Wetting and swelling of polymers to permit intimate contact with biological tissues.

ii) Interpenetration of bioadhesive polymer chain and entanglement of polymer and mucin chains

and

iii) Formation of weak chemical bonds between entangled chains (Malviya et al., 2010;

Srivastava et al., 2010).

1.3.5.2 Gelling agent, Thickener and Water Binder

Pectins are mainly used as gelling agents, but can also act as thickner, water binder and

stabilizer. Low methoxyl pectins (< 50% esterified) form thermo-reversible gels in the presence

of calcium ions and at low pH (3 – 4.5) whereas high methoxyl pectins rapidly form thermally

irreversible gels in the presence of sufficient sugars such as sucrose and at low pH (<3.5); the

lower the methoxyl content, the slower the set (Kohn, 1982).

1.3.5.3 Pectin in Medicine and Pharmaceutical Industry

i) As dietary fibers

Ruminant nutritionists recommend that the digestibility and energy concentration in forages can

be improved by increasing pectin concentration in the forage (Helene et al., 2005).

ii) In site specific targeting

Pectin has a promising pharmaceutical use and it is presently considered as a carrier material in

colon-specific drug delivery system (Sriamornsak and Nunthanid, 1998), for the treatment of

diseases such as ulcerative colitis, colon carcinomas. The rationale for this is that pectin and

calcium pectinate will be degraded by colonic pectinolytic enzymes (Malviya et al., 2010), but

will slow down the drug release in the upper gastrointestinal tract due to its insolubility and

because it is not degraded by gastric or intestinal enzymes. Therefore it reduces the toxicity of

the drugs and makes their activity longer lasting without altering their therapeutic effects (Morris

et al., 2010; Pilnik et al., 1970; Schols et al., 2009; Thakur et al., 1997).

1.4 Role of Microbes in Pectinase Production

Pectolysis is one of the most important processes for plant, as it plays a role in cell elongation

and growth as well as in fruit ripening. Pectolytic enzymes are wide spread in nature and are

produced by Bacteria, Fungi, Yeast, Insects, Nematodes and Protozoa. For example Bacteria

such as Bacillus species, Clostridium species; Fungi such as Aspergillus species, Penicillum

species; Yeast such as Saccharomyces and Candida. Microbial pectolysis is important in plant

pathogenesis, symbiosis and decomposition of plant deposits (Lang and Dornenburg, 2000).

Thus by breaking down pectin polymer for nutritional purposes, microbial pectolytic enzymes

play an important role in nature. The enzymes are inducible i.e. produced only when needed and

they contribute to the natural carbon cycle.

Microbial pectinolytic enzymes are not only enzymes available to attack plant polysaccharides.

However, pathogenic attack on plant tissue in normally initiated by pectic enzymes because

pectic substances are most readily accessible. Aspergilli are used by industries for the production

of primary metabolites (organic acids, vitamins, fatty acids and amino acids) and enzymes as

well as bioconversion processes. Two species, Aspergillus niger and Aspergillus oryzae prevail

as enzyme producers (Oxenboll, 1994). While Aspergillus niger is mainly exploited in the area

of food (bakery, starch industry) and beverages (wine and juice, brewery, distilling industry),

Aspergillus oryzae is applied in the fermentation of oriental sauces such as soya sauce. Hence

many of their products obtained the GRAS (Generally Regarded As Safe) status.

1.5 Pectinases

These are a group of related enzymes and may be divided in three broader groups as follows

(Sakai, 1992; Palomaki and Saarilahti, 1997):

i) Protopectinases: degrade the insoluble protopectin and give rise to highly polymerized soluble

pectin.

ii) Esterases: catalyze the de-esterification of pectin by the removal of methoxy esters.

iii) Depolymerases: catalyze the hydrolytic cleavage of the α (1,4) -glycosidic bonds in the D-

galacturonic acid moieties of the pectic substances. Depolymerases act on pectic substances by

two different mechanisms; hydrolysis, in which they catalyze the hydrolytic cleavage with the

introduction of water across the oxygen bridge and trans-elimination lysis, in which they break

the glycosidic bond by a trans-elimination reaction without any participation of water molecule

(Codner, 2001; Albersheim et al., 1960). Depolymerases can be subdivided into four different

categories, depending on the preference of enzyme for the substrate, the mechanism of cleavage

and the splitting of the glycosidic bonds (Rexova-Benkova and Markovic, 1976).

Polygalacturonase and polymethylgalacturonase breakdown pectate and pectin, respectively by

the mechanism of hydrolysis. However, polygalacturonate lyase and polymethylgalacturonate

lyase breakdown pectate and pectin by β-elimination, respectively. Depending upon the pattern

of action, i.e. random or terminal, these enzymes are termed as Endo or Exo enzymes,

respectively.

1.5.1 Protopectinases

This enzyme catalyzes the solubilization of protopectin. Pectinosinase is also synonymous with

protopectinase (PPase). Protopectinase catalyzes the following reaction:

Protopectin + H20 Pectin

(insoluble) (soluble)

Protopectinases are classified into 2 types based on their mechanism of action: A-type

protopectinases react with the inner site, i.e. the polygalaturonic acid region of protopectin

whereas B-type protopectinases react on the outer site i.e. on the polysaccharide chains that may

connect the polygalacturonic acid chain and cell wall constituents (Sakamoto et al., 1994).

The A-type protopectinases are found in the culture filtrates of yeast and yeast-like fungi. Some

have been isolated from Kluyveromyces fragilis IFO 0288, Galactomyces reesei L. and

Trichosporon penicillatum SNO 3 referred to as protopectinase -F, -L and –S respectively

(Whitaker, 1990).

Subsequently, B-type protopectinases have been discovered in Bacillus subtilis IFO 12113,

Bacillus subtilis IFO 3134 and Trametes sp. These are referred to as protopectinase –B, -C and –

T respectively (Sakai and Ozaki, 1988).

All 3 A-type protopectinases are similar in biological properties and have similar molecular

weight of 30 kDa. Protopectinases -F is an acidic protein while protopectinases –L and –S are

basic proteins. The protopectinases -B, -C and –T have molecular weights of 45, 30 and 55kDa

respectively. However, these 3 types act on protopectin from various citrus fruits and other plant

tissues, releasing pectin (Sakai, 1992).

Table 3. Extensive Classification of Pectinolytic Enzyme

Source: Kashyap et al., (2001)

Enzyme E.C No Action

Mechanism

Action

Pattern

Primary

Substrate

Product

ESTERASE 1. Pectin

Methylesterase

3.1.1.11

Hydrolysis

Random

Pectin

Pectic acid +

Methanol

DEPOLYMERASE

a) Hydrolase 1. Protopectinase

Hydrolysis

Random

Protopectin

Pectin

2. Endo-

polygalacturonase

3.2.1.15 Hydrolysis Random Pectic Acid Oligogalacturona

tes

3. Exo-

polygalacturonase

3.2.1.67 Hydrolysis Terminal Pectic Acid Monogalacturon

ates

4. Exo-

polygalacturonan di-

galacturonohydrolase

3.2.1.82 Hydrolysis Penultimate

bonds

Pectic Acid Digalacturonates

5.Oligo-galacturonate

hydrolase

Hydrolysis Terminal Trigalacturonat

e

Monogalacturon

ates

6. Δ4:5 Unsaturated

Oligo-galacturonate

hydrolases

Hydrolysis Terminal Δ4:5

(Galacturonate)

n

Unsaturated

monogalacturona

tes & Saturated

(n-1)

7.Endo-polymethyl-

galacturonase

Hydrolysis Random Highly

Esterified

Pectin

Oligomethylgala

cturonates

8.Endo-polymethyl-

galacturonase

Hydrolysis Terminal Highly

Esterified

Pectin

Oligogalacturona

tes

b) Lyases

1. Endo-

polygalacturonase lyase

4.2.2.2

Trans-

elimination

Random

Pectic Acid

Unsat Oligo-

galacturonates

2. Exo-

polygalacturonase lyase

4.2.2.9 Trans-

elimination

Penultimate

bond

Pectic Acid Unsat Oligo-

galacturonates

3. Oligo-D-

galacturonate lyase

4.2.2.6 Trans-

elimination

Terminal Unsat

digalacturonates

Unsat Mono-

galcturonates

4. Endo-polymethyl-D-

galacturonate lyase

4.2.2.10 Trans-

elimination

Random Unsat poly-

(methyl-D-

galacturonates)

Unsat methyl

Oligogalacturona

tes

5. Exo-polymethyl-D-

galacturonate lyase

Trans-

elimination

Terminal Unsat poly-

(methyl-D-

galacturonates)

Unsat methyl

Monogalacturon

ates.

Fig. 9: Enzymatic mode of action of the most frequent depolymerases on the pectin molecule.

PMGL: Polymethylgalacturonate lyase (pectin lyase); PMG: Polymethylgalacturonase (pectin

hydrolase); PMGE: Polymethylgalacturonate esterase (pectinesterase); PGL: Polygalacturonate

lyase (pectate lyase); PG: Polygalacturonase (pectate hydrolase). (Serra et al., 1992)

1.5.2 Polygalacturonases

These are enzymes that catalyze the hydrolytic cleavage of the polygalacturonic acid chain with

the introduction of water across the oxygen bridge. They can be subdivided into Exo and Endo-

PGase.

Endo-PGases are found in fungi, bacteria and many types of yeast (Luh and Phaff, 1951) also in

some higher plants and some plant parasitic nematodes (Sakai et al., 1993). In contrast, exo-

PGases occur less frequently as they have been reported in Erwina carotovora and Bacillus sp.

(Koboyashi et al., 2001)).

Exo-PGases can exist in 2 forms; as fungal exo-PGases which produces mono-galacturonic acid

as the main end product and as bacterial exo-PGases, which produces di-galacturonic acid as the

main end product (Sakai et al., 1993).

PGases isolated from different microbial sources differ markedly from each other with respect to

their physicochemical and biological properties as well as their mode of action. Table 4

summarizes the biochemical and physicochemical properties of PGases obtained from various

sources. Most of the PGases obtained from different microbial sources have an optimal pH range

of 3.5 - 5.5 and optimal temperature range of 30-50 ºC; but 2 endo-PGases (PG I & PG II)

isolated from Aspergillus niger have optimal pH range of 3.8 - 4.3 and 3.0 - 4.6 respectively

(Singh and Rao, 2002).

TABLE 4: Biochemical and Physicochemical Properties of Some Polygalacturonases

Source of

PGase

Nature Molecular

Weight

(kDa)

pI Specific

Activity

Km Optimum

Temp

(ºC)

Optimum

pH

Temp

Stability

pH

Stabil

ity

Aspergillus

japonicas

Endo

Endo

38 (PG I)

65 (PG II)

5.6

3.3

-

-

-

-

30

30

4.0-5.5

4.0-5.5

-

-

-

-

Mucor

flavus

- 40 8.3 - - 45 3.5-5.5 40 2.5-

6.0

Thermococc

us

aurantiacus

Endo 35 5.9 5890 0.13 55 5.0 60 4.0-

6.5

Aspergillus

niger

Endo

Endo

61 (PG I)

38 (PG II)

-

-

982

3750

0.12

0.72

43

45

3.8-4.3

3.0-4.6

50

51

-

-

Bacillus sp.

KSM- P410

Exo 45 5.8 54 1.3 60 7.0 50 7.0-

12.0

Penicillum

frequentus

Exo

Exo

63

79

-

-

2571

185

1.6

0.059

50

50

5.0

5.8

-

-

-

-

Bacillus

lichenformis

Exo 38 - 209 - 69 11.0 - 7.0-

11.0

Saccharomy

ces

cerevisiae

IMI-8b

- 43 - - - 45 4.5 - -

Kluyveromy

ces

marxianus

Endo

Endo

Endo

Endo

496 (PG I)

496 (PG II)

496 (PG III)

496 (PG IV)

6.3

6.0

6.3

5.7

102.6

102

107.8

97.6

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

-

Source: (Jayani et al., 2005)

1.5.3 Lyases

Lyases (or transeliminases) perform non-hydrolytic breakdown of pectates or pectinates,

characterized by a trans-eliminative split of the pectic polymer (Sakai et al., 1993). The lyases

break the glycosidic linkages at C-4 and simultaneously eliminate H from C-5, producing a D 4:5

unsaturated products (Codner, 2001).

Lyases can be classified into the following types based on their mechanism of action and the

substrates they act on:

i). Endo-polygalaturonate lyase (endo-PGL)

ii). Exo-polygalacturonate lyase (exo-PGL)

iii). Endo-polymethylgalacturonate lyase (endo-PMGL)

iv).Exo-polymaethylgalacturonate lyases (exo-PMGL)

Polygalacturonate lyases, otherwise known as pectate lyases or PGLs are produced by many

bacteria and some pathogenic fungi with endo-PGLs being more abundant than exo-PGLs. PGLs

have been isolated from bacteria and fungi associated with food spoilage and soft rot.

Erwina carotovora (Kotoujansky, 1987), Collectotrichum lindemuthionum (Wijesundera et al.,

1984) have been reported to produce PGL while PMGLs have been reported to be produced by

Aspergillus japonicas (Ishii and Yokosuka, 1975) and Aspergillus sp (Sunnotel and Nigam,

2002).

In bacteria, lyases are the largest group of pectinolytic enzymes and are directly involved in plant

pathogenicity (Dixit et al., 2004). PGLs have an absolute requirement for Ca2+

ions (Margo et

al., 1994) and hence chelating agents such as EDTA act as their inhibitors whereas PMGLs do

not have an absolute requirement of cations but are stimulated by Ca2+

and other cations (Szajer

and Szajer, 1984).Properties of some of the lyases have been shown on Table 5.

Interestingly, endo-PMGL is the only enzyme known to be able to cleave the α 1,4-glycosidic

bonds of highly esterified pectins without the prior action of other enzymes (Sakai et al., 1993;

Alana et al., 1990).

Table 5: Biochemical and Physicochemical Properties of Some Lyases

Source: (Jayani et al., 2005)

1.5.4 Pectinesterases

Pectinesterase (PE) often referred to as pectin methylesterase, is a carboxylic acid esterase and

belongs to the hydrolase group of enzymes (Whitaker, 1984). It catalyzes the de-esterification of

methyl ester linkages of galacturonan backbone of pectic substances to release acidic pectins and

Source of

Lyases

Nature Molecular

Weight

(kDa)

pI Km Optimum

Temp

(ºC)

Optimum

pH

Temp

Stability

pH

Stability

Erwinia

cantovora

PGL(PL I)

PGL (PL II)

36-38

36-38

10.7

10.1

0.12

1.1

50

60

10.0

10.0

-

Bacteroides

thetaiotaomicr

on

PGL 74 7.5 0.04-

0.07

- 8.7

Aureobasidium

pullulans LV-

10

PMGL (L I)

PMGL(L II)

89

55

-

-

-

-

40

40

5.0

7.5

50

40

3.5

5.9

Penicillum

italicum

PMGL 22 8.6 3.2 40 6.0- 7.0 50 8.0

Aspergillus

japonicas

PMGL - 7.7 0.16 55 6.0 -

Bacillus sp PGL 38 - - 69 11.0 60 7.0-

11.0

Bacillus sp

TS44

PGL 50 5.3 - 70 8.0 70 11.0

Thermoascus

auratniacus

PMGL - - - 65 10.5-11.0 70 4.0

methanol (Cosgrove, 1997). The resulting pectin is then acted upon by polygalacturonases and

lyases (Sakai et al., 1993; Prade et al., 1999).

The mode of action of PE varies according to its origin (Micheli, 2001). Fungal PEs act by a

multi-chain mechanism, removing the methyl groups at random. In contrast, plants PEs tend to

act either at the non-reducing end or next to a free carboxyl group, and proceed along the

molecule by a single chain mechanism. This reaction catalyzed by PE can be represented in the

reaction below

Pectin + nH2O Pectate + nC2H5OH

Pectinesterase activity is implicated in cell wall metabolism including cell growth and ripening

of fruits (Gaffe et al., 1997; Dorokhov et al., 1999). Commercially, PE can be used for protecting

and improving the texture and firmness of several fruits and vegetables as well as in the

extraction and clarification of fruit juices (Gailing et al., 2000). Pectinesterase is found in plants,

pathogenic bacteria and fungi (Hasunuma et al., 2003) such as Saccharomyces cerevisiae

(Gainvors et al., 1994), Aspergillus niger (Maldonaldo et al., 1994; Maldonaldo and Saad, 1998),

E. chrysanthemi 3604 (Laurent et al., 2000). Also there are many reports of occurrence of PE in

plants such as Carica papaya (Fayyaz et al.1993; Innocenzo and Lajalo, 2001), Vitis vinifera

(Corredig et al., 2000), Citrus sp. (Arias and Burns, 2002) and Pouteria sapota (Arenas-Ocampo

et al., 2003). Pectinesterase shows highest activity on 65-75% methylated pectin since the

enzyme is thought to act on methoxyl groups adjacent to free carboxyl groups (Whitaker, 1984).

PE are highly specific enzymes in that, some PE attack only at the reducing chains while others

attack the non-reducing end (Sakai et al., 1993). Most PE are active at a pH range from 4.0-8.0,

however fungal PE have lower pH optimum than those from bacterial sources. Table 6 shows

properties of some pectinesterases.

Table 6: Biochemical and Physicochemical Properties of Some Pectinesterases

Source of

PE

Molecular

Weight

(kDa)

pI Km Optimum

Temp

(ºC)

Optimu

m pH

Temp

Stability

(ºC)

pH

Stability

Rhodotorul

a sp

- - - 40 6.0 50 4.9-9.0

Erwina

chrysanthe

mi 3341

37 9.6-9.9 - 50 5.0-9.0 - -

Aspergillus

niger

- - 1.01 45 5.0 - -

ApplePE 36 9.0 0.12

3

60 7.0 75 -

E.

chrysanthe

mi 3604

37 - 0.03 50 8.0-9.0 - -

Aspergillus

japonicas

46 (PE I)

47 (PE II)

3.8

3.8

-

-

-

-

4.0-5.5

4.0-5.5

50

50

-

-

Source: (Jayani et al., 2005)

1.6 Biotechnological Applications of Microbial Pectinases

Over the years, pectinases have been used in several conventional industrial processes, such as

textile, plant fiber processing, oil extraction, treatment of industrial wastewater, containing

petinacious material, etc. They have also been reported to work on purification of viruses

(Salazar and Jayasinghe, 1999) and in making of paper (Reid and Richard, 2004; Viikari et al.,

2001). However they are yet to be commercialized.

1.6.1 Fruit Juice Extraction

The largest industrial application of pectinases is in fruit juice extraction and clarification. A

mixture of pectinases and amylases is used to clarify fruit juices and thus decreasing the filtration

time up to 50% (Blanco et al., 1999). Treatment of fruit pulps with pectinases also showed an

increase in fruit juice volume from banana, grapes and apples (Kaur et al., 2004). Pectinases in

combination with other enzymes such as cellulases, arabinases and xylanases, have been used to

increase the pressing efficiency of the fruits for juice extraction (Gailing et al., 2000). Vacuum

infusion of pectinases has a commercial application to soften the peel of citrus fruits for removal.

This technique may expand in future to replace hand cutting for the production of canned

segments (Baker and Wicker, 1996).

1.6.2 Coffee and Tea Fermentation

Pectinase treatment accelerates tea fermentation and also destroys the foam forming property of

instant tea powders by destroying the pectins (Carr, 1985). Pectinolytic microorganisms are used

in the fermentation of coffee to remove the mucilaginous coat from the coffee beans.

1.6.3 Textile Processing and Bioscouring of Cotton Fibers

Pectinases have been used in conjunction with amylases, lipases, cellulases and hemicellulases to

remove sizing agents from cotton in a safe and eco-friendly manner, replacing toxic caustic soda

used for the purpose earlier (Hoondal et al., 2000). Bio-scouring is a novel process for removal

of non-cellulosic impurities from the fiber with specific enzymes. According to Hoondal et al.,

(2000) pectinases have been used for this purpose without any negative side effect on cellulose

degradation.

1.6.4 Degumming of Plant Bast Fibers

Bast fibers are the soft fibers formed in groups outside the xylem, phloem or pericycle. For

example Ramie and sunn hemp. The fibers contain gum, which must be removed before its use

for textile making (Hoondal et al., 2000). The chemical degumming treatment is polluting, toxic

and non-biodegradable. Biotechnological degumming using pectinases in combination with

xylanases presents an eco-friendly and economic alternative to the above problem (Kapoor et al.,

2001).

1.6.5 Waste Water Treatment

Vegetable food processing industries release pectin, containing wastewaters as by-product.

Pretreatment of these wastewaters with pectinolytic enzymes facilitates removal of pectinaceous

material and renders it suitable for decomposition by activated sludge treatment (Hoondal et al.,

2000).

1.6.6 Paper and Pulp Industry

During papermaking, pectinase can deploymerize pectins and subsequently lower the cationic

demand of pectin solutions and the filtrate from peroxide bleaching (Reid and Richard, 2004;

Viikari et al., 2001).

1.6.7 Animal Feed

Pectinases are used in the enzyme cocktail, used for the production of animal feeds. This reduces

the feed viscosity, which increases absorption of nutrients, liberates nutrients, either by

hydrolysis of non-biodegradable fibers or by liberating nutrients blocked by these fibers, and

reduces the amount of faeces (Hoondal et al., 2000).

1.6.8 Purification of Plant Viruses

In cases where the virus particle is restricted to phloem, alkaline pectinases and cellulases can be

used to liberate the virus from the tissues to give very pure preparations of the virus (Salazar and

Jayasinghe, 1999).

1.6.9 Improvement of Chromaticity and Stability of Red Wines

Pectinolytic enzymes added to macerated fruits before the addition of wine yeast in the process

of producing red wine resulted in improved visual characteristics (color and turbidity) as

compared to the untreated wines. Enzymatically treated red wines presented chromatic

characteristics, which are considered better than the control wines. These wines also showed

greater stability as compared to the control (Revilla and Ganzalez-Sanjose, 2003).

1.7 Substrates for the Production of Pectinases

Substrates that are employed in the production of enzyme should be solid as solid substrate can

encourage the growing cells. Substrates should provide all needed nutrients to the

microorganisms for its growth.

Other factors like particle size, moisture levels are also to be taken for consideration. Generally

agro-industrial wastes are employed for the pectinase production. Various substrates that are

being used are sugarcane bagasse, wheat bran, rice bran, wheat straw, rice straw, sawdust, corn

cobs, coconut coir pith, banana waste, tea waste, sugar beet pulp, apple pomace, orange peel,

pineapple peels etc (Pilar et al., 1999).

1.8 Fermentation Conditions

Pectinases are constitutive or inducible enzymes that can be produced either by submerged

(Aquilar and Huitron, 1999) or solid state fermentation (Acuna-arguelles et al., 1995).Various

factors related to environment affect the production of pectinase. Some of them are concentration

of nutrients, pH, temperature, moisture content, influence of extraction parameters on recovery

of pectinases and the effects played by the inducers. Both carbon and nitrogen sources show

overall effect on the productivity of pectinases (Catarina et al., 2003; Almeida and Huber, 2011).

Pectin, glucose and sucrose when added to the media in higher concentration have a repression

effect on the studied enzyme activity (Maria et al., 2000) of the various nitrogenous matters that

can be used. Optimum sources are (NH4)2SO4, yeast extract, soya bean pulp powder, soya

peptone.

Temperature and pH are also important parameters to be taken note of. The pH is regulated using

a mixture of sources of nitrogen as when Aspergillus niger is being used, pH turns to be acidic.

Besides the nature of the substance also plays a vital role in the pH maintenance. Moisture

content in the substrate also plays a significant role (Martin et al., 2004). The previous studies

show that it was generally maintained around 50-55% for the production of pectinases by

microbial means (Leda et al., 2000).

Two types of fermentations can be carried out for pectinase production, they are solid state

fermentation and submerged fermentation. The growth of organisms is very high with large

quantities of enzyme being produced in solid- state fermentation (Ramanujam and Saritha,

2008). However in the production of extracellular pectinases, submerged fermentation is

preferable as the extracellular pectinases are easier and cheaper to use in great quantities. Most

important applications of these enzymes are in juice and wine making, and in the processing of

vegetables. Submerged or solid state mediums are used for producing of the pectinolitic enzymes

by fungi (Bali, 2003).

1.8.1 Types of Fermentation

i) Solid State Fermentation (SSF)

ii) Submerged Fermentation (SmF)

Solid state fermentation is defined as the cultivation of microorganisms on moist solid supports,

either on inert carriers or on insoluble substrates that can be used as carbon and energy source.

This process occurs in the absence or near absence of free water in the space between substrate

particles. In this system, water is present in the solid substrate whose capacity for liquid retention

varies with the type of material (Lonsane et al., 1985; Pandey et al., 2001).

Submerged liquid fermentation is the cultivation of microorganisms in liquid nutrient broth.

Industrial enzymes can be produced using this process. This involves growing carefully selected

microorganisms (bacteria and fungi) in closed vessels containing a rich broth of nutrients (the

fermentation medium) and a high concentration of oxygen (Grigelmo-Migeul and Martin-

Belloso, 1998).

SSF has the following advantages over SmF:

i) Low water demand which leads to less waste water production.

ii) Utilization of solid substrates that enhances the concentration of the growth substrates.

iii) Utilization of otherwise unusable carbon sources that is abundant in supply.

However, there are also several disadvantages of SSF which have discouraged the use of this

technique for industrial production and therefore have made SmF more applicable in the

production of enzymes. These include: the buildup of gradients of temperature, pH, moisture,

substrate concentration or CO2 during cultivation which are difficult to control under limited

water availability (Holker et al., 2004).

1.9 Microorganisms Commonly Used in Submerged and Solid State Fermentation for

Pectinases Production

Microorganisms are currently the primary source of industrial enzymes: 50% originate from

fungi and yeast; 35% from bacteria, while the remaining 15% are either of plant or animal origin.

Filamentous microorganisms are most widely used in submerged and solid-state fermentation for

pectinase production. Ability of such microbes to colonize the substrate by apical growth and

penetration gives them a considerable ecological advantage over non-motile bacteria and yeast,

which are less able to multiply and colonize on low moisture substrate (Smith and Aidoo,

1988). Among filamentous fungi three classes have gained the most practical importance in SSF;

the phycomycetes such as genera Mucor, the ascomycetes genera Aspergillus and

Basidiomycetes especially the white and rot fungi (Young et al., 1983). Bacteria and yeasts

usually grow on solid substrates at the 40%to70% moisture levels (Young et al., 1983). Common

bacteria in use are (Bacillus licheniformis, Aeromonas cavi and Lactobacillus) and common

yeasts in use are Saccharomyces and Candida. Pectinase production by Aspergillus strains has

been observed to be higher in solid-state fermentation than in submerged process (Solis et al.,

1996).

TABLE 7: Comparison of Solid State and Submerged Fermentation

Factor Submerged Fermentation

(SmF)

Solid State Fermentation (SSF)

Substrate Soluble Substrate (sugars) Polymer insoluble substrate:

Starch, Cellulose, Pectins

Aseptic Conditions Heat sterilization and aseptic

Control

Vapor treatment, non-sterile

conditions

Water High volumes of water

consumed and effluents

discarded

Limited consumption of water

Metabolic Heating Easy control of temperature Low heat transfer capacity

pH Control pH control Buffered solid substrate

Source: (Raimbault, 1998)

1.10 Aim and Objectives of the Study

1.10.1 Aim of the Study

To extract pectin from pineapple peels

To isolate Aspergillus niger from soil containing decomposing pineapple peels.

To produce extracellular pectinase by inducing Aspergillus niger in submerged fermentation

with pectin extracts from pineapple peels.

To partially purify extracellular pectinase by carrying out ammonium sulphate precipitation

and dialysis.

To characterize the partially purified enzyme

1.10.2 Specific Objectives of the Study

To determine the effect of change in pH on pectinase activity.

To determine the effect of change in temperature on pectinase activity.

To determine the Km and Vmax of the enzyme from the Lineweaver-Burk plot.

CHAPTER TWO

MATERIALS AND METHODS

2.1 MATERIALS

2.1.1 Chemicals/ Reagents

All the chemicals used in this research work were of analytical grade and were obtained from

Sigma Chemical Company, USA; Sigma-Aldrich, USA; Bio Rad Laboratories, India; Merck,

Germany; BDH Chemical Ltd, England; May and Baker Ltd, England; Riedel-De Haen

Hannaves, Germany; Hopkins and Williams Essex, England and Lab Tech. Chemicals Avighkar,

India.

2.1.2 Apparatus/ Equipment

Autoclave: UDAY BURDON‟s Patent Autoclave, made in India.

Centrifuge: Finland Nigeria 80-2B.

Glass wares: Pyrex

Incubator: B and T Trimline incubator.

Magnetic stirrer: AM-3250B Surgi Friend Medicals, England.

Microscope: WESO microscope.

Milling machine: Thomas Willey laboratory Mill Model 4, Anthor H (Thomas

Company, Philadelphia, USA).

Oven: Gallenkamp Hotbox, made in England.

pH meter: Ecosan pH meter, made in Singapore.

UV/ spectrophotometer: Jenway 6405

Water bath: Model DK.

Weighing balance: B2404-5 mettler Toledo, made in Switzerland.

Weighing balance: Ohaus Dial-O-Gram, Ohaus Co-operation, N.J. USA.

2.1.3 Collection of Pineapple Samples

Pineapple (Ananas cosmosus) peels were obtained from Ogige market, Enugu State, Nigeria.

2.1.4 Collection of Micro-organisms

Mixed colonies of microorganisms were obtained from a dump containing decaying pineapple

peels and Aspergillus niger was isolated using morphological characteristics.

2.2 METHODS

2.2.1 Preparation of Reagents

2.2.1.1 Preparation of 3N HCl

Normality of the stock HCl in Winchester bottle was calculated using the formulae outlined

below:

Normality (N) = % assay x 1000 x specific gravity

100 x Equivalent weight

For HCl, % assay = 37, specific gravity = 1.19 and equivalent weight (in this case) = 36.5. The

volume of the stock solution required to prepare 3N HCl was calculated using the formula

N1V1 = N2V2

Where: N1 = Normality of stock HCl,

V1 = volume of stock HCl required for the preparation,

N2 = required normalty (3N),

V2 = working volume (1000ml).

N.B: V1 obtained from the calculation was diluted in 1000ml of diluent (water) to make the

required 3N HCl.

2.2.1.2 Preparation of Ethanol-HCl Solution

The ethanol-HCl solution contained 0.5M HCL prepared using similar formula as that described

above except that molecular weight was used instead of equivalent weight in order to obtain the

molarity of stock HCl. The volume of the stock solution required to prepare 0.5M HCl was

calculated using the formula:

C1V1 = C2V2;

where C1= Molarity of stock HCl,

V1 = volume of stock HCl required for the preparation,

C2 = required molarity (0.5M),

V2 = working volume (1000ml).

N.B: V1 obtained from the calculation was diluted in 1000ml of diluent (ethanol) to make the

required ethanol-HCl solution.

2.2.1.3 Preparation of Buffers

The standard buffers used in this study were pH 4.0 and pH 7.0. These buffers were used to

standardize the pH meter. The working buffers were prepared by this procedure.

Sodium acetate buffer of 0.05M and Tris-HCl buffer of 0.05M were prepared by dissolving

4.10g sodium acetate salt and 6.01g Tris base, respectively in 1000ml of distilled water and

stirred with a magnetic stirrer till a homogenous solution was formed. The solutions were titrated

against acetic acid and HCl, respectively till the required pH was obtained.

Phosphate buffer of 0.05M was prepared by dissolving 7.10g disodium hydrogen phosphate salt

in 1000ml stirred as well with a magnetic stirrer and then titrated against the solution of its

conjugate acid which is sodium di-hydrogen phosphate till the required pH was obtained.

2.2.1.4 Preparation of 2mg/ml Bovine Serum Albumin (BSA) Standard Protein

An amount of 0.2g of BSA was dissolved in 100ml of distilled water and then used as a protein

stock solution.

2.2.1.5 Preparation of Dinitrosalicylic Acid (DNS) Reagent

A modification of DNS reagent method of Miller (1959) as contained in Wang et al. (1997) was

used in the assay. The reagent contains 44mM dinitrosalicylic acid, 4mM sodium sulphite, and

375mM sodium hydroxide.

2.2.1.6 Preparation of 20mM Galacturonic Acid

An amount of 0.42g D-(+)-Galaturonic acidmonohydrate (molecular weight 212.15g/mole) was

dissolved in 100ml 0.05M sodium acetate buffer stirred over a magnetic stirrer until a

homogenous stock solution was obtained.

2.2.1.7 Preparation of the Component Reagents for Protein Determination

Solution A: An alkaline sodium carbonate (Na2CO3) was prepared by dissolving 2g of Na2CO3

in 100ml of 0.1M NaOH (0.4g of sodium hydroxide pellets were dissolved in 100ml of distilled

water).

Solution B: A copper tetraoxosulphate VI - sodium potassium tartarate solution was prepared by

dissolving 0.5g of CuSO4 in 1g of sodium potassium tatarate, all in 100ml of distilled water. It

was prepared fresh by mixing stock solution, and so was done whenever required.

Solution C: Folin-Ciocalteau phenol reagent was made by diluting the commercial reagent with

water in a ratio of 1:1 on the day of use.

Solution D: Standard protein (Bovine Serum Albumin, BSA) solution.

Solution E: Freshly prepared alkaline solution was made by mixing 50ml of solutions A and 1ml

of solution B.

2.2.2 Preparation of Ground Pineapple Peels

The pineapple peels were washed and cut into small bits and then treated with hot 96% ethanol

in order to reduce the microbial load. The ethanol treated peels were washed with water and sun-

dried for 7 days. The dried peels were then ground to powder using a milling machine.

2.2.3 Extraction of Pectin from Pineapple Peels

Pectin was extracted by the method of McCready, (1970). 100g of ground pineapple peels was

poured into a 2000ml beaker containing 800ml of distilled water, then 12g of freshly ground

sodium hexa-metaphosphate was added to the mixture and the initial pH was adjusted with 3N

HCL to 2.2 ± 0. The mixture was heated up in a water bath at 70 ºC for 1 hr and stirred

continuously using a propeller typed stirrer.

The pH was checked at an interval of 15 minutes. The water lost by evaporation was replaced

except in the last 20 minutes of the extraction time. The extract was vacuum-filtered using a

muslin cloth and the residue was washed with 200ml of distilled water. The washings were

however added to the filtrate and then concentrated by evaporation on a hot plate to

approximately one-fifth of its initial volume.

The concentrated pectin mixture was cooled to 50 ºC and a volume of ethanol containing 0.5M

HCL in the ratio of 3:1 to the pectin mixture was added, it was stirred continuously for 30

minutes and allowed to stand for 1 hour. The precipitate was vacuum filtered and washed with

acetone in order to remove traces of HCL and ethanol. The extract was dried in an oven at 40 ºC

for a few hours and ground to powder.

2.2.4 Isolation of Pectinolytic Fungi

2.2.4.1 Collection of Soil Samples

Soil samples were collected from site containing decomposing pineapple peels. This compost

site was located within the school premises of the University of Nigeria Nsukka, Enugu state.

The soil samples were collected in a clean dry plastic container.

2.2.4.2 Preparation of Soil Sample Extracts for Microbial Isolation.

Soil samples from site of decomposing pineapple peels were homogenized in sterile medium

containing 1% pineapple pectin, 0.14% of (NH4)2SO4, 0.2% of K2HPO4, 0.02% of MgSO4.7H2O

& 0.1% of nutrient solution containing 5mg/L of FeSO4.7H2O,1.6mg/L of MnSO4.H2O, 1.4mg/L

of ZnSO4.7H2O, 2.0mg/L of CoCl2.The mixture was incubated at 30ºC for 24hrs.

2.2.4.3 Preparation of the Solid Medium

The mixture contained 1% pineapple pectin, 0.14% of (NH4)2SO4, 0.2% of K2HPO4, 0.02% of

MgSO4.7H2O & 0.1% of nutrient solution containing 5mg/L of FeSO4.7H2O,1.6mg/L of

MnSO4.H2O, 1.4mg/L of ZnSO4.7H2O, 2.0mg/L of CoCl2 and 3% agar-agar (the gelling agent)

(w/v).

The medium was autoclaved at 121oC for 15min. It was allowed to cool to about 45

oC and then

poured into Petri dishes and allowed to gel. The plates were then incubated in a B & T Trimline

incubator at 37oC overnight to check for sterility.

2.2.4.4 Sub-culturing into Solid Medium

A loop of homogenized extract from the sample extracts was streaked onto the solid medium

under the flame of Bunsen burner. The plates were incubated at 35oC (room temperature) till

visible colonies were observed. All morphological contrasting colonies were purified by repeated

streaking and sub-culturing on separate plates. This process was continued till pure fungal

cultures were obtained.

2.2.4.5 Storage of Micro-organisms on Potato Dextrose Agar (PDA)

Pure fungal isolates were maintained on PDA slants as stock cultures. The PDA medium was

prepared according to the manufacturer‟s instructions.

2.2.4.6 Microscopic Features of the Isolated Fungi

Three days old pure cultures were examined; the color, texture, nature of mycelia or spores and

growth patterns were also observed. Photographs of the culture were also taken.

2.2.4.7 Fungal Identification

Three days old cultures were used in preparing microscopic slides. A tuft of the mycelia was

dropped on the slide and a drop of lacto-phenol blue was added to it. A cover slip was placed

over it and viewed under a light microscope at ×400 magnification. Identification was carried out

by relating the microscopic features and the micrographs to „Atlas of Mycology‟ by Barnett and

Hunter (1972).

2.2.5 Fermentation Experiments

2.2.5.1 Preparation of the fermentation medium

The substrate for fermentation consisted of ground pineapple pectin extracts. Submerged

fermentation was carried out using 10 250ml Erlenmeyer flask containing 200 ml of the sterile

cultivation medium. The medium was optimized for pectinase production with 0.1% NH4NO3,

0.1% NH4H2PO4, 0.1% MgSO4.7H2O and 20g of powdered pineapple pectin extracts. The flask

was covered with aluminium foil and autoclaved at 121ºC for 20mins.

2.2.5.2 Innoculating with Aspergillus niger

In every sterile flask, two disc of Aspergillus niger obtained from the freshly prepared plates are

added using a cork borer of diameter 10mm under sterile conditions. The flasks are plugged

firmly and incubated for four days at room temperature.

2.2.5.3 Harvesting the crude enzyme

At the end of day 4 on which the highest enzyme activity has been detected, the mycelia biomass

was filtered using filter paper. 2.0 litres of filtrate recovered was used as the crude enzyme, while

the residue was treated with lime and discarded properly.

2.2.6 Galacturonic Acid Standard Curve

The reaction mixture contained 0.0-1.0ml of galacturonic acid stock solution in test tubes

arranged in triplicates. Each test tube was made up to 1ml using freshly prepared 0.05M sodium

acetate buffer of pH 5.0. 1ml of DNS reagent was added to each of the test tubes and placed in a

boiling water bath for 10min. 1ml of 1.4M Rochelle salt (sodium potassium tartarate) was added

to the test tube immediately after heating and the total volume of the solution was adjusted to

4ml with distilled water. The mixture was cooled to room temperature and the absorbance read at

575nm. The concentration of reducing sugar in each of the tubes was calculated using the

formula

C1 V1 = .C2 V2

Where: C1 = initial concentration of reducing sugar (mM)

C2 = final concentration of reducing sugar (mM)

V1 = initial volume of 20mM galacturonic acid preparation measured into the tube

V2 = final volume of the preparation.

Using the values obtained from above the calculations, the plot of optical density was

constructed and the concentration of galacturonic acid released at a given absorbance was

extrapolated (Appendix Two).

2.2.7 Polygalacturonase Assay

Polygalacturonase activity was determined by measuring the release of reducing sugars from the

pineapple pectin using the 3,5- dinitrosalicyclic acid reagent assay as described by Miller,

(1959).

2.2.7.1 Procedure for Polygalacturonase Assay

This was carried out by the method described by Miller, (1959) as contained in Wang et al.,

(1997) with slight modifications. The reaction mixture contained 0.5ml of 0.5% pineapple pectin

in 0.05M sodium acetate buffer pH 5.0 and 0.5ml of the crude enzyme solution. After one hour

incubation time, 1ml of DNS reagent was added and the reaction was stopped by boiling for

10mins at 70ºC.

The total volume was brought up to 4ml by adding 1ml of rochelles salt and 1ml of distilled

water. The reaction mixture was allowed to cool and then absorbance was read at 575nm. One

unit of enzyme was defined as the amount of enzyme that catalyzes the formation of 1

micromole of galacturonic acid per minute.

2.2.8 Protein Determination

Protein content of the enzyme was determined by the method of Lowry et al. (1951), using

Bovine Serum Albumin as standard.

2.2.8.1 Procedure for Protein Determination

For the reaction mixture, test tubes were arranged in duplicates containing 0.0- 1.0 ml of 0.2mg

of protein stock solution (2mg/ml BSA) and brought up to 1ml with distilled water. For the test

mixture, 0.5ml of sodium acetate buffer pH 5.5 was added to 0.5ml of the crude enzyme.

To both the reaction and test mixture, 5 ml of solution D was added soon after and the mixture

was allowed to stand for 10 mins. 0.5ml of Solution C (dilute Folin-ciocalteau reagent) was

added and then the solution was mixed thoroughly and allowed to stand for 30mins under room

temperature. The absorbance was read at 750nm and the protein concentration was determined

thereafter.

2.2.9 Partial Purification of Protein

2.2.9.1 Ammonium Sulphate Precipitation Profile

This procedure is carried out in order to know the percentage of ammonium sulphate

concentration is suitable to precipitate the most protein from the crude enzyme. Nine test tubes

were used containing 10ml of the crude enzyme and the enzyme was precipitated from 20% -

100% saturation of solid ammonium sulphate at 10% interval in each test tube.

The contents of the tubes were mixed thoroughly to ensure that the salts were dissolved and then

allowed to stand for 30hrs at 4ºC. The test tubes were centrifuged at 3500 rpm for 30 mins and

the filtrates were decanted while the pellets were re-dissolved in equal volume of 0.05M sodium

acetate buffer pH 5.0. Pectinase activity was determined on the contents of each tube.

2.2.9.2 Ammonium Sulphate Precipitation

After determining the percentage saturation of ammonium sulphate salts that gave the highest

activity, the equivalent amount of salt for 1 litre of crude enzyme is added. The salt is allowed to

dissolve completely and the mixture is allowed to stand for 30 hrs at 4ºC. It is then centrifuged at

3500 rpm for 30mins. The pellets are collected and stored in a cool place for further studies.

2.2.9.3 Dialysis

Dialysis tubes stored in 90% ethanol were used. However the tubes were rinsed thoroughly with

distilled water and finally with 0.05M sodium acetate buffer in order to remove traces of ethanol.

An amount of the precipitated enzyme is poured into the dialysis tubes and placed in a beaker

containing 0.05 M sodium acetate buffer. The beaker is placed on a magnetic stirrer which

allows for a homogenous environment. The dialysis is carried out according to Dixon and Webb

(1964) for 12 hours and the buffer is changed after 6 hours which allows for the exchange of low

molecular weight substances and left over ammonium sulphate salts that may interfere with the

activity. After dialysis, the partially purified enzyme is stored frozen at -24ºC.

2.2.10. Studies on Partially Purified Enzyme

2.2.10.1 Effect of pH Change on Pectinase Activity

The effect of pH on enzyme activity was determined using 0.05M sodium acetate buffer pH 3.5

- 5.5, phosphate buffer pH 6.0 - 7.5 and Tris-HCl buffer pH 8.0 - 10.0 at intervals of 0.5. 0.1%

pectin solution was prepared by dissolving 0.1g pectin in 100ml of 0.05M of the respective

buffers. Also 0.5 ml of the partially purified enzymes was added to 0.5 ml of each of the buffers.

Then ultimately, 0.5ml of each of the enzyme-buffer solution was mixed with 0.5ml pectin

solution at the corresponding pH for pectinase assays as described previously.

2.2.10.2 Effect of Temperature Change on Pectinase Activity

The optimum temperature was determined by incubating the enzyme with pectin solution at 25-

70oC interval of 5

oC for 1hour and at the pH with the highest activity. The activity was then

determined as described in previously.

2.2.10.3 Effect of Substrate Concentration on Pectinase Activity.

The effect of substrate concentration on the activity of pectinase was determined by incubating

the enzyme with 0.5 up to 6.0mg/ml of pineapple pectin at an interval of 0.5 using the buffer at

the pH with highest activity and the temperature at which highest activity was determined. The

Vmax and Km values of the enzyme were determined using the double reciprocal plot.

2.2.10.4 Further Studies with Partially Purified Enzyme

Pectinase activity and total protein concentration of partially purified enzymes were determined

using the methods described in sections 2.2.7.1 and 2.2.8.1 respectively.

CHAPTER THREE

RESULTS

3.1 Pineapple Pectin Extraction

3.1.1 Percentage Extraction Yield of Pineapple Pectin

Pectin extraction yield was found to be 8.33% at pH 2.2, temperature of 70oC and extraction time

of 1hr.

3.1.2 Photograph of Pineapple Pectin Extract

Fig. 10 shows a photograph of the pineapple pectin after extraction.

Fig. 10: Photograph of Pineapple Pectin

3.2 Microorganism

3.2.1 Selection of Pectinolytic Fungi

Three species of fungi namely: Aspergillus niger, Aspergillus fumigatus and Aspergillus flavus

were isolated from natural source of soil containing decaying pineapple peels. These organisms

were qualitatively screened for pectinolytic activity on selective media and their isolation was

based on the similarities of their morphological features in both test cultures containing

pineapple pectin and the standard culture containing apple pectin as carbon respectively.

3.2.2 Macroscopic and Microscopic Features of Fungal Isolates

Genus identification was by examining both macroscopic and microscopic features of a three day

old pure culture. Color, texture, nature of mycelia and/or spores produced, growth pattern in

addition to microscopic features such as separation and spore shapes were examined. Based on

these characteristics, Aspergillus niger, Aspergillus fumigatus and Aspergillus flavus were

confirmed as the three pectinolytic fungal isolates, respectively. However, Aspergillus niger

showed relatively higher pectinase activity and was selected for further studies. Fig. 11 shows a

photograph of the pure culture of Aspergillus niger.

Fig. 11: Pure Culture of Aspergillus niger

3.3 Pectinase Production under Submerged Fermentation System

A volume of 2 litres of crude enzyme was harvested after 4 days of submerged fermentation

using Aspergillus niger.

3.4 Ammonium Sulphate Precipitation

Crude enzyme obtained from Aspergillus niger was precipitated within the range of 20-100% at

an interval of 10%. At 80% ammonium sulphate saturation, the highest activity was obtained at

81.62U/ml as shown in Fig. 12; thus, the percentage was used for the precipitation of pectinases.

Fig. 12: Ammonium sulphate precipitation profiling of pectinases obtained from Aspergillus

niger.

3.5 Assays Carried Out on Pectinase Obtained

3.5.1 Protein Concentration of the Crude, Precipitated and Dialyzed Pectinases

In Fig 13, initial protein concentration of pectinase in the crude state was 10.42mg/ml but after

ammonium sulphate precipitation and dialysis, the concentration increased to 15.82mg/ml and

later decreased to 12.84mg/ml.

Figure 13: Comparison in the protein concentration of the crude, precipitated and dialyzed

pectinases

3.5.2 Activity of Pectinase in the Crude, Precipitated and Dialyzed Enzyme

The activities of the enzymes increased progressively from one purification step to the other with

the corresponding values of 116.12U/ml, 272.98U/ml and 634.56U/ml respectively as shown in

Fig. 14.

Figure 14: Comparison in the activities obtained from the crude, precipitated and dialyzed

pectinases

3.5.3 Total Protein Content of the Crude, Precipitated and Dialyzed Enzymes

The total protein content of the crude enzyme reduced from the crude to the precipitated and also

to the dialyzed enzyme with the respective values of 10420mg, 1012.48mg and 48226.56mg as

seen in Fig. 15.

Figure 15: Comparison in the total protein content of the crude, precipitated and dialyzed

pectinases

3.5.4 Total Activity of the Crude, Precipitated and Dialyzed Enzymes

Fig. 16 shows activity-dependent decrease from the crude enzyme across precipitated enzyme to

after dialysed sample. The crude enzyme had the highest total enzyme activity of 116120U with

after dialysed sample having the least enzyme activity of 4822.56U.

Fig. 16: Comparison in the total activity of the crude, precipitated and dialyzed pectinases

3.5.5 Specific Activity Determined on the Crude, Precipitated and Dialyzed Enzymes

Specific activity-dependent increase was observed in Fig. 17 with crude enzymes having the

lowest activity and after, dialyzed sample had the highest enzyme activity with values of

11.14U/mg and 49.42U/mg respectively. This further explains that the enzyme was in a purer

state after dialysis than after ammonium sulphate precipitation.

Fig. 17: Comparison in the Specific Activities of the crude, precipitated and dialyzed pectinases

3.5.6 Purification Folds of the Partially Purified Enzymes

As shown in Fig. 18, after dialysis and ammonium sulphate precipitation, the purification fold

increased from 1.55 after ammonium sulphate precipitation and further increased to 4.43 after

dialysis which infers that the enzyme had undergone a 4.43 purification fold.

Fig. 18: Purification folds of the pectinase after precipitation and dialysis

Table 8: Summary of the Parameters Determined From the Crude, Precipitated and Dialyzed

Pectinases

Purification

Step

Volume

(ml)

Protein

Conc.

(mg/ml)

Activity

(U/ml)

Total

Protein

(mg)

Total

Activity

(U)

Specific

Activity

(U/mg)

Purification

fold

Crude

Enzyme

1000 10.42 116.12 10420 116120 11.14 1

80%

ammonium

sulphate

precipitation

64

15.82

272.98

1012.48

17470.72

17.26

1.55

Dialysed

Enzyme

76 12.84 634.56 975.84 48226.56 49.42 4.43

µmole/min= Unit (U)

According to Table 8, the crude state of pectinase had the least value for activity of 116.12U/ml

unlike the precipitated and dialyzed, which were 272.98U/ml and 634.56U/ml respectively. Also,

the protein concentration increased after ammonium sulphate precipitation due to the precipitated

proteins. However, the protein concentration reduced after dialysis to 12.84mg/ml. Thus it can be

said that protein concentration is inversely relative to activity.

3.6. Characterization of Pectinase

3.6.1 Effect of pH Change on Pectinase Activity

Fig. 19 shows that the highest pectinase activity was recorded at pH 5.5. Also, observed in Fig.

19 was a decline in activity when pH was increased or decreased beyond 5.5.

Figure 19: Showing the effect of change in pH on pectinase activity

3.6.2 Effect of Change in Temperature on Pectinase Activity

Fig. 20 shows gradual decline of the enzyme activity at a temperarture of 40˚C with subsequent

rise at temperature of 45 and 50˚C. The peak of the enzyme activities was observed at a

temperature of 55˚C accompanied with a sharp drop of the enzyme activity with corresponding

rise in temperature.

Figure 20: Effect of Change in Temperature on Pectinase Activity

3.6.3 Kinetic Parameters

The kinetic parameters such as Km and Vmax of the enzyme were obtained from the Lineweaver

Burk plot below and the values obtained were 89.5mg/ml and 5000U respectively.

Figure 21: Lineweaver-Burk plot of pectinases from Aspergillus niger

Table 9: Pectinase characterization

Properties Aspergillus niger

pH 5.5

Temperature (°C) 55°C

Vmax(U/ml) 5000U/ml

Km(mg/ml) 89.5mg/ml

Table 9 shows the characterization of the pectinase obtained from Aspergillus niger with the pH

and temperature optima being 5.5 and 55˚C respectively. The Km and Vmax were 89.5mg/ml and

5000U/ml respectively.

CHAPTER FOUR

DISCUSSION

4.1 Discussion

Pineapple (Ananas cosmosus) peels as agricultural wastes represent about 35% of the fruit mass.

During extraction of pectin from pineapple peels, the yield of pectin extracted was 8.33% at pH

2.2, temperature of 70ºC and extraction time of 1hr using the method as described by McCready

(1970). The yield could be affected by the pH of the extraction medium and extraction time.

Three fungal species isolated from natural waste source were selected which include Aspergillus

niger, Aspergillus fumigatus and Aspergillus flavus. However, Aspergillus niger (pure culture as

shown in Fig. 11) was used for the fermentation process because it showed the highest pectinase

activity after 4 days of fermentation after which a decline in activity was observed. This may be

attributed to the depletion of the nutrients and pectin in the fermentation medium which could

result in decrease in enzyme or protein synthesis over time (Haq et al., 2005) since Aspergillus

niger produces extracellular enzymes.

In the present study, accumulation of maximal extracellular pectinase was observed after 96hour

of fermentation. The period of fermentation depends on the nature of medium, fermenting

organisms, concentration of nutrients and the process physiological conditions (Patil and

Dayanand, 2006). Substrates should provide all needed nutrients to the micro-organism for its

growth. Various substrates being used are sugarcane bagasse, rice bran, banana wastes, tea

waste, pineapple peels, apple pomace, etc, (Pilar et al., 1999). Although an increased level in the

production of pectinases was noticed when the agro wastes were supplemented with additional

carbon and nitrogen sources and supplementation of sucrose was more effective than glucose in

solid state fermentation (Patil and Dayanand, 2006). A 96hour incubation time was also obtained

in the poly-galacturonase production in solid state fermentation (Martin et al., 2004).

Ramanujam et al. (2008) recorded an 84hour incubation time for the optimum production of

pectin lyase by Aspergillus niger in solid state fermentation which is comparable to what was

obtained.

In this research, submerged fermentation technique was applied due to the fact that temperature,

pH, moisture, substrate concentration or CO2 during cultivation would have been difficult to

control under limited water availability. (Holker et al., 2004). Submerged liquid fermentation is

the cultivation of microorganisms in liquid nutrient broth. Industrial enzymes can be produced

using this process. Submerged fermentation involves growing carefully selected microorganisms

(bacteria and fungi) in closed vessels containing a rich broth of nutrients (the fermentation

medium) and a high concentration of oxygen (Grigelmo-Migeul andMartin-Belloso, 1998).

Ammonium sulphate precipitation profile was carried out on the crude enzyme and the activities

of the pellets obtained from the different percentages of saturation from 20%-100% was

determined. Fig. 12 shows that 80% had the highest activity and was therefore used for the actual

precipitation process of the enzyme. Buga et al. (2010) reported 70% ammonium sulphate

saturation for pectinase from Aspergillus. niger (SA6) while Adejuwon and Olutiola, 2007

reported 90% ammonium sulphate saturation for pectinase from Lasidioplodia theobromae.

After the protein was precipitated, one way to remove this excess salt is to dialyze the protein.

Also it changes the buffer composition of solutions of biomolecules too large to pass through the

membrane (Rosenberg, 2004). It was observed that there was an increase in volume of the

enzyme after dialysis which was done for 12 hours; this may be due to the buffer that entered

from the dialyzing medium during the process.

The protein concentration increased from 10.42 to 15.82 mg/ml after ammonium sulphate

precipitation and then there was a decrease in the concentration to 12.84mg/ml after dialysis as

seen in Fig. 13. This may be due to the removal of other proteins of lower molecular weight

during dialysis that was not the protein of choice.

Fig. 14 shows the pectinase activity which increased from 116.12 to 272.98 U/ml after

precipitation and further increased to 634.56 U/ml after dialysis. This may be due to the removal

of impurities during dialysis such as other proteins which may have affected the enzyme activity

negatively (Lukong et al., 2007).

The specific activity of the crude, precipitated and dialyzed were determined and it showed that

there was an increase after each purification step as shown in Table 8 from 11.14 U/mg in the

crude to 17.26 U/mg in the precipitated and then to 49.42 U/mg in the dialyzed. This is in

agreement with the report by Lukong et al. (2007) that for a purification procedure to be

successful, the specific activity of the desired enzyme must be greater after the purification

procedure than as it was before. The increase in specific activity is a measure of purification

achieved thus the enzyme had a 4.43 purification fold as purification fold increased from 1.54

after ammonium sulphate precipitation to 4.43 after dialysis as observed in Fig. 18.

The partially purified enzyme was characterized based on the effect of change in pH,

temperature and substrate concentration. The maximal activity was observed at pH 5.5 as seen in

Fig. 19 for pectinase from Aspergillus niger using pineapple peels as substrate which is

comparable to the optimum pH for pectinase activity from thermo-tolerant Aspergillus sp N12

(Freitas et al., 2006). Changes in pH can change the shape of the active site in an enzyme.

Extremely high or low pH concentrations usually result in complete loss of enzyme activity due

to denaturation (Helms et al., 1998). At extremely high and low pH values, the tertiary structure

of the protein (enzyme) may be disrupted and the protein denatured; even at moderate pH values,

where the tertiary structure is not disrupted, enzyme activity may depend on the alteration of

ionisation states of: certain amino acid side chains responsible for substrate binding, certain side

chains involved in catalysis and certain groups on the substrate (Lukong et al., 2007).

Temperature has a complex effect on enzyme activity. The "optimum temperature" of an enzyme

is the temperature in which the enzyme functions most efficient. An increase in temperature

below the optimum results in an increase in the kinetic energy of enzymes. This leads to higher

efficiency of enzyme-substrate complex formation; therefore a higher rate of reaction. It differs

in each enzyme, depending on its nature and structure. At extreme temperatures above the

optimum, the increased kinetic energy disrupts the bonds holding the active site; the enzyme is

unstable and the shape of its active site changes. This means that the enzyme is less efficient and

successful at enzyme-substrate complex formation. When the enzyme is said to be denatured; it

has lost its ability to catalyze reactions. Fig. 20 shows that 55˚C was the temperature at which

pectinase from Aspergillus niger exhibited the highest activity, which falls within the range as

exo-polygalacturonase from Monascus and Aspergillus sp. (Freitas et al., 2006) which exhibited

maximum activity at 60 and 50ºC respectively. It was also reported that polygalacturonase by

Aspergillus kawachii (Esquivel and Voget, 2004) and P. frequentans (Chellagatti et al., 2002)

exhibited maximum activity at 60ºC. At a higher temperature, the activity dropped and this could

be as a result of the denaturation of the enzyme. It could be that enzymes are proteins and are

heat sensitive, once temperatures have reached extreme heat, the enzymes denature, thereby

exposing the protein structure which leads to inactivation. All enzymes have an optimal

temperature at which reaction rates go fastest without denaturing the enzyme (Campbell and

Reece, 2002).

The Km and Vmax were calculated from the double reciprocal plot as shown in Fig. 21 and Km

was 89.5mg/ml while the Vmax was 5000U/ml. According to Anosike (2001), Km values provide

a parameter for comparing enzymes from different organisms. With the knowledge of Michaelis

Menten, it is known that a small Km indicates that the enzyme requires only a small amount of

substrate to become saturated; hence, the maximum velocity is reached at relatively low substrate

concentration while a large Km indicates the need for high substrate concentrations to achieve

reaction velocity. On the other hand, the Vmax or maximum velocity gives information on the

turnover number of an enzyme (Anosike, 2001). The turnover number of an enzyme is the

number of moles of substrate converted into product per active site of the enzyme per second,

when the enzyme is fully saturated with substrate which gives information on how efficient a

given enzyme is as a catalyst.

4.2 Conclusion

From this work, it was discovered that although pineapple peels have low pectin content of about

8.33%, it can be successfully used to induce the production of pectinase under submerged

fermentation system. The enzymes obtained can be industrially used in the production of fruit

juice, paper making, retting of plant fibres, etc. Ultimately, the rationale behind this research was

the conversion of waste to wealth which could increase the revenue base of any establishment or

country obtained and also geared towards a cleaner and safer environment.

4.3 Suggestions for Further Studies

The following suggestions for further studies are made based on the results obtained;

1) The organism used can be modified genetically in order to produce more enzymes and

probably on less number of days.

2) The biotechnological application of this process should be encouraged by individuals and the

governments since the advantages have been addressed.

3) More research works should be done on the production of pectinases from Aspergillus niger

using pineapple peels as carbon source. This inevitably makes their characterization more

available.

REFERENCES

Acuna-Arguelles, M.E., M.Gutierrez-Rojas, G.,Vinigra-Gonz-alez, E. and Favela-Torres. (1995).

Production and properties of three pectinoloytic activities produced by Aspergillus niger in

submerged and solid state fermentation. Applied microbiology and biotechnology, 43: 808-

814.

Adejuwon, A.O. and Olutiola, P.O. (2007). Pectin lyase activity in culture medium of

Lasidioplodia theobramae.Journal of Plant Sciences,2 (3): 334-340.

Alana, A., Alkorta, I., Dominguez, J.B., Llama, M.J. and Serra, J.L. (1990). Pectin lyase activity

in a Penicillium italicum strain. Applied Environmental Microbiology, 56: 3755–3759.

Albersheim, P., Neukom, H. and Deuel, H. (1960). Splitting of pectin chain molecules in neutral

solution. Archeology, Biochemistry and Biophysics, 90: 46–51.

Almeida, D.P.F. and Huber, D.J. (2011). Autolysis of cell walls from polygalacturonase-

antisense tomato fruit in simulated apoplastic solutions, Plant Physiologyof Biochemistry,

10: 1016

Alkorta, I., Garbisu, C., Llama, M.J. and Serra, J.L. (1998). Industrial applications of pectic

enzymes. A review: Process Biochemistry, 33: 21–28.

Anosike, E.O. (2001). Basic Enzymology. University of Port Harcourt Press, Pp 11-87.

Aquiler, G. and C.Huitron. (1999). Constitute exopectinase produced by Aspergillus sps ch-y-

043 on different carbon sources. Biotechnology, 12: 655-660.

Arenas-Ocampo, M.L., Evangelista-Lozano, S., Arana-Errasquin, R., Jime´nez-Aparicio, A. and

Da´vila-Ortíz, G. (2003). Softening and biochemical changes of sapote mamey fruit

(Pouteria sapota) at different development and ripening stages. Journal in Food Chemistry,

27: 91–95.

Arias, C. and Burns, J. (2002). A pectin methylesterase gene associated with a heat stable extract

from citrus. Journal in Agriculture and Food Chemistry, 50: 3465–3472.

Baker, R.A. and Wicker, L. (1996). Current and potential application of enzyme infusion in the

food industry. Trends in Food Science Technology, 7: 279– 284.

Bali, R. (2003). Isolation of extracellular fungal pectinoytic enzymes and extraction of pectin

using kinnow waste as substrate.MSC thesis. Thaper institute of engineering technology

(Deemed University) Punjab, India.

Barnett, H. L., and Hunter, B.B. (1972). Illustrated general of imperfect fungi.3rd

Edn. Burgess,

Minneapolis, Minnesota, USA. pp 20-130.

Barthelomew, D. P., Paul, R. E. and Rohrbach, K. G. (eds). (2003). The Pineapple: Botany,

Production and Uses. CABI Publishing, Wallingford UK. Pp. 1-301.

Blanco, P., Sieiro, C. and Villa, T.G. (1991).Production of pectic enzymes in yeasts.

Microbiological Letters, 175:1–9.

Blanco, P., Sieiro, C. and Villa, T.G. (1999).Production of pectic enzymes in yeasts.

Microbiology Letters, 175:1–9.

Braccini, I., Grasso, R.P. and Pérez, S. (1999). Carbohydrate Resources, 317: 119–130.

Braconnot, H. and Keppler, F. (1825). Methane emission from terrestrial plant under aerobic

conditions.Nature, 439:187-190.

Buga, M.L., Sani, I. and Adrew, J.W. (2010). Partially purified polygalacturonase from

Aspergillus niger (SA6). African Journal of Biotechnology, 9(52): 8944-8954.

Campbell, N, and Reece, I. (2002). Biology, Don O‟Neal (ed.), 6th Edn. Benjamin Cummings

Publishing, Menlo Park, California. Pp. 300-340.

Carpita, N.C. and Gibeaut, D.M. (1993). Structural models of primary cell walls in flowering

plants-consistency of molecular structures with the physical properties of the wall during

growth. Plant Journal, 3:1-30.

Carr, J.G. (1985). Tea, coffee and cocoa. In: Microbiology of fermented foods (ed. Wood,

B.J.B), London: Elsevier Science Ltd, 2:133–154.

Catarina, A., Tomas, B., Pedro, M. and Jose. (2003). Teixeir continuous production of pectinase

by immobilized yeast cell on srent grains. Journal of Bioscience and Bioengineering, 96:

513-551.

Chellegati, M.A.S.C., Fonseca, M.J.V. and Said, S. (2002). Purification and partial

characterization of exo-polygalacturonase from Mucor rouxii NRRL 1894. Enzyme

Microbiology and Technology, 41: 800-805.

Cho, S.W., Lee, S. and Shin, W. (2001). The X-ray structure of Aspergillus aculeatus

polygalacturonases and a modeled structure of the polygalacturonase–octagalacturonate

complex. Journal in Molecular Biology, 314: 863–878.

Codner, R.C. (2001). Pectinolytic and cellulolytic enzymes in the microbial modification of

plant tissues. Journal in Applied Bacteriology, 84: 147–60.

Coppens d'Eeckenbrugge, G; Leal, F. (2003). “Morphology, Anatomy, and Taxonomy". In: D.P

Bartholomew, R.E. Paull, and K.G. Rohrbach. The Pineapple: Botany, Production, and

Uses. Wallingford, UK: CABI Publishing. p. 21.

Corredig, M., Kerr, W. and Wicker, L. (2000). Separation of thermostable pectin methylesterase

from marsh grapefruit pulp. Journal in Agriculture and Food Chemistry, 48: 4918–4923.

Cosgrove, D.J. (1997). Assembly and enlargement of the primary cell wall in plants. Annual

Review on Cell Development and Biology, 13:171–201.

Daas, P.J.H., Boxma, B., Hopman, A.M.C.P., Voragen, A.G.J. and Schols, H.A. (2001).

Biopolymers, 58: 1–8.

Dixit, V.S., Kumar, A.R., Pant, A. and Khan, M.I. (2004). Low molecular mass pectate lyase

from Fusarium moniliforme: similar modes of chemical and thermal denaturation.

Biochemistry, Biophysics Resource Communique, 315: 477–84.

Dixon, M. and Webb, E.C. (1964). Enzymes. 2nd

ed. Longmans, New York. Pp 116-166.

Dorokhov, Y.L., Makinen, K., Frolova, O.Y., Merits, A., Saarinen, J. Kalkkinen, N., Atabekov,

J.G. and Saarma, M. (1999). A novel function for a ubiquitous plant enzyme pectin

methylesterase: the host-cell receptor for the tobacco mosaic virus movement protein.

Federation of European Biochemical Societies, 461: 223–228.

du Penhoat, C.H., Gey, C., Pellerin, P. and Perez, S. (1999). An NMR solution study of the mega

oligosaccharide, rhamnogalacturonan II. Journal of Biomolecules. NMR, 14: 253-271.

Elfick,J. (2007). Pineapple Project.

http://www.uq.ed.au/_School_Science_Lessons/PineProj.html#Lesson2. Retrieved:2-8-2011

Esquivel, J.C.C and Voget, C.E. (2004). Purification and partial characterization of an acidic

polygalacturonase from Aspergillus kawachii. Journal in Biotechnology, 110: 21-28.

Fayyaz, A., Asbi, B.A., Ghazali, H.M., CheMan, T.B. and Jinap, S. (1993). Pectinesterase

extraction from papaya. Food Chemistry, 47: 183–185.

Fleisher, A., O‟Neill, M.A., Ehwald, R. (1999). Plant Physiology, 121: 829-838.

Freitas, P.M., Martin, N., Silva, D., Silva, R. and Gomes, E. (2006). Production and partial

characterization of polygalacturonase production by Thermophilic Monascus sp. N8 and by

Thermotolerant Aspergillus sp. N12 on solid state fermentation. Brazillian Journal in

Microbiology, 37: 32-306.

Gaffe J., Tizando, M.E. and Handa, A.K. (1997). Characterization and functional expression of a

ubiquitously expressed tomato pectin methylesterase. Plant Physiology, 114: 1547–1556.

Gailhofer, G., Wilders-Truschnig, M., Smolle, J., Ludvan, M. (1998). Asthma caused by

bromelain: an occupational allergy. Clinical Allergy 18: 445-450.

Gailing, M.F., Guibert, A. and Combes, D. (2000). Fractional factorial designs applied to

enzymatic sugar beet pulps pressing improvement. Bioprocess Engineering, 22: 69–74.

Gainvors, A., Frezier, V., Lemaresquier, H., Lequart, C., Aigle, M. and Belarbi, A. (1994).

Detection of polygalacturonase, pectin lyase and pectinesterase activities in Saccharomyces

cerevisiae strain. Yeast, 10: 1311–1319.

Gaspar, Y., Johnson, K.L., McKenna, J.A., Bacic, A. and Schultz, C.J. (2001).Plant Molecular

Biology, 47: 161–176.

Glushka, J.N., Terrel, M., York, W.S., O‟Neil, M.A., Gucwa, A., Darvill, A.G., Albersheim, P.

and Prestegard, J.H. (2003). Primary structure of the 2-O-methyl-α-fructose containing side

chain of the pectic polysaccharide, rhamnogalacturonan II. Carbohydrate Residues, 338:

341-352.

Grigelmo-Migeul, N. and Martin-Belloso, O. (1998). Characterization of dietary fiber from

orange juice extraction. Food Resources International, 31: 355-361.

Haq, I., Javed, M.M., Khan, T.S. and Siddiq, Z. (2005). Cotton saccharifying acvtivity of

cellulases produced by co-culture of Aspergillus niger and Trichoderma viride. Research

Journal of Agriculture and Biological Science, 1: 241-245.

Hasunuma, T., Fukusaki, E.I. and Kobayashi, A. (2003). Methanol production is enhanced by

expression of an Aspergillus niger pectin methylesterase in tobacco cells. Journal in

Biotechnology, 106: 45–52.

Helene, M., Canteri-Schenim, Heloísa, C.R.F., Nina, W., Wosiacki, G. (2005).Extraction of

pectin from apple pomace.Brazillian Archeology of Biological Technology. Curitiba, 48: 2-

3.

Helms, D., Helms, C., Kosinski, R. and Cummings, J. (1998). Biology in the Laboratory, 3rd Ed.

Judith Wilson (ed.), Freeman Publishing, New York, New York. Pp. 9: 1-8.

Holker, U., Hofer, M. and Lenz, J.(2004).Biotechnological advantages of laboratory-scale solid

state fermentation with fungi. Applied Microbiology and Biotechnology, 64: 175-186.

Hoondal, G.S., Tiwari, R.P., Tiwari, R., Dahiya, N. and Beg, Q.K. (2000). Microbial alkaline

pectinases and their applications: A review: Applied Microbiology in Biotechnology, 59:

409–418.

Innocenzo, M.D. and Lajalo, F.M. (2001). Effect of gamma irradiation on softening changes and

enzyme activities during ripening of papaya fruit. Journal in Food Biochemistry, 25: 19–27.

IPPA (2001). Types of pectin. Retrieved from http://www.ippa.info/types_of_pectin. htm.

Downloaded on June 5, 2011.

Ishii, S. and Yokotsuka, T. (1975).Purification and properties of pectin lyase from Aspergillus

japonicus.Agriculture, Biology and Chemistry, 39: 313–21.

Ishii, T., Matsunaga, T. (1996). Carbohydrate Residues, 284: 1-9.

Jayani, R.S., Saxena, S. and Gupta, R. (2005). Microbial pectinolytic enzymes: A review.

Process Biochemistry, 40(99): 2931- 2944.

Kapoor, M., Beg, Q.K., Bhushan, B., Singh, K., Dadich, K.S. and Hoondal, G.S. (2001).

Application of alkaline and thermostable polygalacturonase from Bacillus sp. MG-cp-2 in

degumming of ramie (Boehmeria nivea) and sunn hemp (Crotolaria juncia) bast fibers.

Process Biochemistry, 36: 803–807.

Kashyap, D.R., Vohra, P.K., Chopra, S. and Tewari R. (2001). Applications of pectinases in

commercial sector: a review. Bioresource in Technology, 77:215–27.

Kaur, G., Kumar, S. and Satyanarayana, T. (2004). Production, characterization and application

of a thermostable polygalacturonase of a thermophilic mould Sporotrichum thermophile

Apinis. Bioresource in Technology, 94: 239–243.

Kertesz, Z, J. (1951). The Pectic Substance. Interscience Publishers, New York.p 68.

Koboyashi, T., Higaki, N., Yajima, N., Suzumatsu, A., Haghihara, H. and Kawai, S. (2001).

Purification and properties of a galacturonic acid-releasing exo-polygalacturonase from a

strain of Bacillus. Bioscience, Biotechnology and Biochemistry, 65: 842–847.

Kohn, R. (1982). Binding of toxic cations to pectin, it‟s oligomeric fragment and plant tissues,

Carbohydrate Polymers, 2: 273-275.

Kotoujansky, A. (1987). Molecular genetics of pathogenesis by soft-rot Erwinia. Annual Review

in Phytopathology, 25: 405–430.

Koubala, B.B., Kansci, G., Mbome, M.J., Crepeau, J.F., Thibault, J.F. and Ralet, M.C. (2006).

Mango peels as a source of pectin and the effect of extraction method. Food is life- 13th

World Congress of IUFoST (International Union of Food Science and Technology). Nantes

(FRA: 2006/09/17-21).

Lang, C. and Dornenburg, H. (2000).Perspectives in the biological function and the

technological application of polygalacturonases.Applied Microbiology in Biotechnology,

53:366–75.

Lau, J.M., McNeil, M., Darvill, A.G. and Albersheim, P. (1987). Carbohydrate Resources, 168:

245–274.

Laurent, F., Kotoujansky, A. and Bertheau, Y. (2000). Overproduction in Escherichia coli of the

pectin methylesterase A from Erwinia chrysanthemi 3937: one-step purification,

biochemical characterization, and production of polyclonal antibodies. Canadian Journal in

Microbiology, 46: 474–480.

Leal, F., Coppens d'Eeckenbrugge, G. (1996).Fruit Breeding. Leal, F., Coppens d'Eeckenbrugge,

G. (eds.). In. John Wiley and Sons, New York. pp 565-606.

Leda, R., Castilho, A., Ricardo, M. and Tito. L.M. (2000). Alves production and extraction of

pectinase obtained by solid state fermentation of agro industrial residues with Aspergillus

niger. Bioresource Technology, 71: 45-50.

Lohani, S., Trivedi, P.K. and Nath, P. (2004). Changes in activities of cell wall hydrolases during

ethylene-induced ripening in banana: Effect of 1-MCP, ABA and IAA. Postharvest

Biological Technology, 31: 119-126.

Lonsane, B.K., Ghildyal, N.P., Budiatman, S. and Ramakrishna, S.V.(1985). Engineering aspects

of solid state fermentation. Enzyme Microbiology and Technology, 7: 258-265.

Lowry, O.H., Rosenbrough, P.J., Fass, A.L. and Randall, R.J. (1951). Protein measurement with

Folin-phenol reagent. Journal of Biochemistry, 193: 265-275.

Luh, B.S. and Phaff, H.J. (1951). Studies on polygalacturonase of certain yeasts. Archeology,

Biochemistry and Biophysics, 33: 212–227.

Lukong, B.C., Awah, F. M., Nwuke, C.P. and Fobellah, A.D. (2007). Protein Isolation,

Purification and Estimation. In: Introduction to Protein Science. OSPREY Publication

Centre, Owerri, Pp 55-56.

Maldonado, M.C, Saad, A.M.S. and Callieri, D.A.S. (1994). Purification and characterization of

pectinesterase produced by a strain of Aspergillus niger. Curriculum in Microbiology,

24:193–196.

Maldonaldo, M.S. and Saad, A.M.S. (1998). Production of pectinesterase and polygalacturonase

by Aspergillus niger in submerged and solid state systems. Journal of Industrial

Microbiology in Biotechnology, 20: 34–38.

Malviya, R., Srivastava, P., Bansal, M. and Sharma, P.K. (2010). Preparation and evaluation of

disintegrating properties of Cucurbita maxima pulp powder. International Journal in

Pharmaceutical Science, 2(1): 395-399.

Margo, P., Varvaro, L., Chilosi, G., Avanzo, C. and Balestra, G.M. (1994). Pectinolytic

enzymes produced by Pseudomonas syringaepv. Glycinea. Microbiology Letters,117:1–6.

Maria, F.S., Jose, L., Lime, F. and Nelson, D. (2000). Carbon sources effect on pectinase

production from Aspergillus japonicus. Brazillian Journal of Microbiology, 31: 286-290.

Marshal, C. and Chow, K. (2007).Pectin extraction from pectin and sugar beets. Agricultural

Resources, 2: 16-17.

Martin, N., de Souza, S.R., da Silva, R. and Gomes, E. (2004).Pectinase production of fungal

strains in solid-state fermentation using agro-industrial bioproduct. Brazilian Archives of

Biology and Technology, 47(5):813-819.

McCready, R.M. (1970). Methods on Food Analysis. Academic Press, New York and London,

Vol. 2. p 365.

McNeil, M., Darvill, A.G. and Albersheim, P. (1980). Plant Physiology, 66: 1128–1134.

Micheli, F. (2001). Pectin methylesterase: cell wall enzymes with important roles in plant

physiology. Trends in Plant Science, 6: 414–9.

Miller, G.L. (1959). Use of Dinitrosalicylic acid reagent for determination of reducing sugars.

Analytical Chemistry, 31: 426-428.

Miller, J.G. and Fry, S.C. (2001). Characteristics of xyloglucan after attack by hydroxyl radicals.

Carbohydrate Resources, 332: 389-403.

Morris, G., Kӧk, S., Harding, S. and Adams, G. (2010). Polysaccharide drug delivery system

based on pectin and chitosan. Biotechnology & Genetic Engineering Review, 27: 257-284.

Mudgett, A.E. (1986). Solid state fermentations. In: Demain, A.L. and Solomon, N.A.(eds.)

Manual of Industrial Microbiology and Biotechnology,American Society for Microbiology

Washington, D.C., 66-83.

Nikolić, M.V. and Mojovic, L. (2007).Hydrolysis of apple pectin by the coordinated activity of

pectic enzymes. Food Chemistry, 101: 1-9.

Novosel‟skaya, I.L. (2002). Trends in the science and application of pectins.Chemical National

Compilation.36: 1-10.

O‟Neil, M.A., Warrenfiltz, D., Kates, K., Pellerin, P., Doco, T., Darvil, A.G. and Albersheim, P.

(1996). Journal in Biology & Chemistry, 271: 22923-22930.

O‟Neil, M.A., Eberhard, S., Albersheim, P. and Darvill, A.G. (2001). Science 294: 846–849.

O‟Neill, M.A., Ishi, T., Albersheim, P. and Darvill, A. G. (2004). Annual Review of Plant

Physiology and Plant Molecular Biology, 55: 109–139.

Oxenboll, K. (1994). Aspergillus enzymes and industrial uses. In the genus Aspergillus: From

Taxonomy and Genetics to Industrial Application (eds. Powel, K.A., Renwick, A. and

Pederby, J.F). Plenum Press, London. Pp 147-154.

Palomaki, T. and Saarilahti, H.T. (1997). Isolation and characterization of new C-terminal

substitution mutation affecting secretion of polygalacturonases in Erwinia carotovora ssp.

Carotovora, 400: 122–6.

Pandey, A., Soccol, C.R., Rodriguez-Leon, J.A. and Nigam, P.(2001). Solid-State Fermentation

in Biotechnology: Fundamentals and Applications. 1st Edn., Asiatech Publishers Inc., New

Delhi. Pp: 221.

Paoletti, S.(1986). In: Chemistry and function of pectins (eds. Fishman, M.L. and Jen, J.J).

Washington DC: American Chemical Society, pp. 73-87.

Patil, S.R. and Dayanand A. (2006). Production of pectinase from deseeded sunflower head by

Aspergillus niger in submerged and solid-state conditions. Bioresources Technology, 97:

2054-2058.

Perez, S., Rodriguez-Carvajal, M. and Doco, T. (2003). A complex plant cell wall

polysaccharide: rhamnogalacturonan II. A structure in quest of a function. Biochemistry, 85:

109-121.

Pilar, B., Carman, S. and Tmaes, G. (1999). Villa production of pectic enzymes in yeast.

Microbiology Letters.175: 1-9.

Pilnik, N., Voragen, A. G. J. and Hulme A. C. (ed.). (1970). Pectic substances and other

uronides. The biotechnology of Fruits and their products, 1: 53-87.

Pilnik, W. and Voragen, A.G.J. (1993). Pectic enzymes in Fruit and Vegetable Juice

Manufacture. Enzymes in Food Processing,Academic Press Limited, London,Third Edition.

Pp 363-392.

Prade, R.A., Zohan, D., Ayoubi, P. and Mort, A.J. (1999).Pectins, pectinases and plant-microbe

interactions. Biotechnology in Genetic Engineering Review, 16: 361–391.

Prasanna, V., Prabha, T.N. and Tharanatha, R.N. (2007).Fruit ripening phenomena– An

overview. Critical Review in Food Science and Nutrition, 47: 1-19.

Purseglove, J. W. (1972). Tropical crops. Monocotyledons. Longman, London. Pp. 71-91.

Py.C. and Tisseau, M. (1969). La Pina tropical. Ed. Blumé. Barcelona, p. 278.

Raimbault, M. (1998). General and Microbiological Aspects of Solid Substrate

Fermentation.Electronic Journal Of Biotechnology, p 1,3.

Ramanujam, N. and Saritha, P.S. (2008). Production of pectinase by solid-state fermentation of

sugarcane bagasse using Aspergillus niger. Advanced Biotechnology. Pp. 30-33.

Rastogi, G. (1998). Vishal‟s objective botany. Meerut, India: Vishal Publishers. Pp. 23-25.

Reid, I. and Richard, M. (2004). Purified pectinase lowers cationic demand in peroxide-bleached

mechanical pulp. Enzyme in Microbial Technology, 34: 499–504.

Revilla, I. and Ganzalez-san jose, M.L. (2003). Addition of pectolytic enzymes: an enological

practice which improves the chromaticity and stability of red wines. International Journal

on Food Science and Technology, 38: 29–36.

Rexová-Benková, L. and Marković, O. (1976). Pectic enzymes. Advanced Carbohydrate

Chemistry, 33:323–85.

Ridley, B.L., O‟Neil, M.A. and Mohnen, D. (2001). Phytochemistry 57: 929–967.

Rohrbach, K.G., Bathelomew, D. P. and Rohbach, K. G. (2003). Pests, Disease and Weeds.

CABI, New York Pp 203-252.

Rosenberg, I.M. (2004). Protein Analysis and Purification: Benchtop Technique, 2nd

Edn.

Springer, Birkhauser, p 520.

Rugkong, A., Rose, J.K.C., Lee, S.G., Giovannoni, J.J. O‟Neill, M.A. and Watkins, C.B. (2010).

Cell wall metabolism in cold-stored tomato fruit. Postharvest Biological Technology, 57:

106-113.

Sakai, T. and Ozaki, Y. (1988). Protopectin solubilizing enzyme that does not catalyze the

degradation of polygalacturonic acid. Agriculture, Biology and Chemistry, 52: 1091–1093.

Sakai, T. (1992).Degradation of pectins. In: Microbial degradation of natural products. (ed.

Winkelmann, G.). Weinheim. Pp. 57–81.

Sakai, T., Sakamoto, T., Hallaert, J. and Vandamme, E.J. (1993). Pectin, pectinase and

protopectinase: Production, properties and applications. Advanced Application of

Microbiology, 39: 231–294.

Sakamoto, T., Hours, R.A. and Sakai, T. (1994).Purification, characterization and production of

two pectic-transeliminases with protopectinase activity from Bacillus subtilis.Bioscience in

Biotechnology and Biochemistry, 58: 353–358.

Salazar, L. and Jayasinghe, U. (1999).Fundamentals of purification of plant viruses. In:

Techniques in plant, virology, CIP. International Potato Centre, Peru. Pp. 1–10.

Sañudo-Barajas, J.A., Labavitch, J., Greve, C., Osuna-Enciso, T., Muy-Rangel, D. and Siller-

Cepeda, J. (2009). Cell wall disassembly during papaya softening: Role of ethylene in

changes in composition, pectin-derived oligomers (PDOs) production and wall hydrolases.

Postharvest Biological Technology, 51: 158-167.

Sathyanarayana, N.G. and Panda, T. (2003).Purification and biochemical properties of microbial

pectinases. A review: Process Biochemistry, 38: 987–996.

Serra, J.L., Alkorta, I., Llama, M.J. and Alaiia, A.(1992).Aplicacion industrial de 1os enzimas

pecticos.Production, purification, inmovilizacion y algunas propiedades de la pectina liasa

de Penicillium italicum.Alimentacion Equiposy Technology, 11: 127-134.

Schols, H.A., Bakx, E.J., Schipper, D. and Voragen, A.G.J. (1995).Carbohydrate Resources,

279: 265–279.

Schols, H. A. Visser, R. G. F. and Voragen, A. G. J. (2009).Pectins and pectinases. Wegenigen

Academic Publisher, The Netherlands. Pp. 980-990.

Shembekar, V.S. and Dhotre, A. (2009).Studies of pectin degrading microorganisms from

soil.Journal of Microbial World 11(2): 216-222.

Singh, S.A. and Rao, A.G.A. (2002). A simple fractionation protocol for, and a comprehensive

study of the molecular properties of two major endopolygalacturonases from Aspergillus

niger. Biotechnology in Applied Biochemistry, 35: 115–123.

Smith, J.E. and Aidoo, K.E. (1988). Growth of fungi on Solid Substrates. Physiology of

Industrial Fungi, Blackwell, Oxford, England. Pp 249-269.

Solis-Pereyra, S., Favela-Torres, E., Gutierrez–Rojas, M., Roussos, S., Saucedo, C.G. and

Viniegra, G.G.1996. Production ofpectinases by Aspergillus nigerin solid state fermentation

at highglucose concentrations. World Journal of Microbiology and Biotechnology, 12: 275-

260.

Sriamornsak, P. (1998). Chemistry of Pectin and it‟s pharmaceutical uses: A review, Journal of

Pharmacology in Phamaceutical Science, 44: 207-228.

Sriamornsak, P. and Nunthanid, J. (1998). Calcium pectinate gel beads for controlled release

drug delivery: In Preparation and in vitro release studies, International Journal in

Pharmacy, 160: 207-212.

Sriamornsak, P. (2002). Analysis of selected physicochemical properties of pectins and alginate

gels intended for drug delivery. PhD thesis, Charles Sturt University.

Srivastava, P., Malviya, R, Gupta, S. and Sharma, P.K. (2010). Preparation and evaluation of

matrix based tablet using natural polymers as release modifiers, International Journal in

Pharmaceutical Science, 2(1): 411-417.

Sunnotel, O. and Nigam, P. (2002). Pectinolytic activity of bacteria isolated from soil and two

fungal strains during submerged fermentation. World Journal in Microbiology and

Biotechnology, 18: 835–839.

Szajer, I. and Szajer, C. (1984). Pectin lyase of Penicillium paxilli. Biotechnology Letters, 4:

549–52.

Thakur, B. R., Singh, R. K. and Handa A. K. (1997). Chemistry and uses of pectin: a review.

Critical Review in Food science and nutrition, 37: 47-73.

Thakur, A., Pahwa, R., Singh, S. and Gupta, R. (2010). Production, Purification, and

characterization of polygalacturonase from Mucor circinelloides ITCC 6025. Enzyme

Research,(170549), 1-7.

Tucker, G.A. and Woods, L.F.J. (1991). Enzymes in Production of Beverages and Fruit juices.

Enzyme in Food Processing, Blackie, New York, Pp. 201-203.

van der Vlugt-Bergmans, C.J.B., Meeuwsen, P.J.A., Voragen, A.G.J. and van Oogen, A.J.J.

(2000). Endo-xylogalacturonan hydrolase, a novel pectinolytic enzyme. Applied

Environmental Microbiology, 66: 36–41.

Vicken, J.P., Schols, H.A., Oomen, R.J.F.J., McCann, M., Ulvskov, P., Voragen, A.G.J., Visser,

R.G.F. (2003). Plant Physiology, 132: 1781-1789.

Viikari, L., Tenakanen, M. and Suurnakki, A. (2001). Biotechnology in the pulp and paper

industry. In: Biotechnology, Rehm, H.J. (Ed). VCH-Wiley, Pp. 523–546.

Visser, J., Voragen, A.G.J. (1996) Progress in Biotechnology: Pectins and Pectinases. Elsevier,

Amsterdam. Pp. 14-15.

Voragen, A.G.J., Schols, H.A. and Visser, R.G.F. (2003).Advances in Pectin and Pectinase

Research. Kluwer Academic Publishers, Dordrecht, The Netherlands. Pp. 203-205.

Wang, G., Michailides, T.J. and Bostock, R. M. (1997). Improved detection of polygalacturonase

activity due to Mucor piriformis with a modified dinitrosalicylic acid

reagent.Phytopathology, 87: 161-163.

Wei J., Ma,F., Shi, S., Qi, X., Zhu, X. and Yuan, J. (2010). Changes and postharvest regulation

of activity and gene expression of enzymes related to cell wall degradation in ripening apple

fruit. Postharvest Biological Technology, 56: 147-154.

Whitaker, J.R. (1984). Pectic substances, pectic enzymes and haze formation in fruit juices.

Enzyme Microbial Technology, 6: 341–347.

Whitaker, J.R. (1990). Microbial pectinolytic enzymes. In: Microbial enzymes and

biotechnology (eds. Fogarty, W.M. and Kelly, C.T.), 2nd ed. London: Elsevier Science Ltd.

Pp. 133–76.

Wijesundera, P.L.C., Bailey, J.A. and Byrde, R.J.W. (1984). Production of pectic lyase by

Colletotricum lindemuthionum in culture and in infected bean (Phaseolus vulgaris) tissue.

Journal in General Microbiology., 130: 285-290.

Wikpedia.(2011). The Pineapple fruit. Retrieved from http://en.wikipedia.org/wiki/pineapple on

19/06/2011.

Willats, W.G.T., McCartney, L., MacKie, W. and Knox, J.P. (2001a). Plant Molecular Biology

47: 9–27.

Young, M.M.; Moriera, A.R.and Tengerdy, R.P. (1983).Principles of Solid State Fermentation

in Smith J.E.; Berry, D. R. and Kristiansen, B. (eds). Filamentous fungi Fungal

Technology, Arnold, E. London. Pp. 117-144.

Yujaroen, P., Supjaroenkul, U., Rungrodnimitchai, S. (2008). Extraction of pectin from sugar

palm meal. Thamm International Journal in Science and Technology (special edition), 13:

44-47.

APPENDIX ONE

Appendix 1: Protein Standard Curve, Using 2mg/ml Bovine Serum Albumin (BSA)

APPENDIX TWO

Appendix 2: Galaturonic Acid Standard Curve, Using 20mM D-(+)-Galacturonic Acid

Monohydrate