47
1 Biotransformation of saponins to diosgenin for enhanced yield from Dioscorea sp using indigenous fungal strains FINAL TECHNICAL REPORT BACK TO LAB PROGRAMME (08-34/BLP/WSD/KSCSTE/ 2016-17) Dr. Reji. S. R, Post Doctoral Researcher Division of Microbiology Jawaharlal Nehru Tropical Botanic Garden and Research Institute Palode, Thiruvanathapuram, India, 695562

Biotransformation of saponins to diosgenin for enhanced

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Biotransformation of saponins to diosgenin for enhanced

1

Biotransformation of saponins to diosgenin for enhanced yield from Dioscorea sp using

indigenous fungal strains

FINAL TECHNICAL REPORT

BACK TO LAB PROGRAMME

(08-34/BLP/WSD/KSCSTE/ 2016-17)

Dr. Reji. S. R,

Post Doctoral Researcher

Division of Microbiology

Jawaharlal Nehru Tropical Botanic Garden and Research Institute

Palode, Thiruvanathapuram, India, 695562

Page 2: Biotransformation of saponins to diosgenin for enhanced

2

CONTENT

Titles Page no

Authorization 3

Acknowledgement 4

Abstract 5

Introduction and review of literature 6-10

Objectives 11

Materials and methods 12- 18

Results and discussion 19 – 40

Summary 41

Scope of future work 42

Outcomes of the project 43 – 46

References 45 -47

Page 3: Biotransformation of saponins to diosgenin for enhanced

3

AUTHORIZATION

The project entitled “Biotransformation of saponins to diosgenin for enhanced yield from

Dioscorea sp using indigenous fungal strains” by Reji. S.R, was carried out under the “Back to

lab programme” of Women Scientists Division, Kerala State Council for Science Technology

and Environment, Govt. of Kerala. The work was carried out at Microbiology Division,

KSCSTE- Jawaharlal Nehru Tropical Botanic Garden and Research Institute, Palode under the

mentorship of Dr. N. S. Pradeep. The project was initiated on 15th march 2016 with sanction

No: 08-34/BLP/WSD/KSCSTE/ 2016-17 and scheduled completion by 14th August 2020 with

a financial expenditure of Rs. 1,847,630 lakhs.

Page 4: Biotransformation of saponins to diosgenin for enhanced

4

ACKNOWLEDGEMENT

This work was carried out during the year 2017– 2020 at Jawaharlal Nehru Tropical Botanical

Garden at the Department of Microbiology. I owe my deepest gratitude to the Director

JNTBGRI for providing the infrastructure for the implementing the project.

I would like to express my sincere gratitude to Dr. N. S. Pradeep (senior scientist, JNTBGRI)

for the guidance and encouragement to realize this assignment. My sincere thanks to all

scientific and technical staff of the department for their valuable advice and support.

This work was supported by funds from Back to Lab program of KSCSTE. So i express my

gratitude to Women Scientist Division and KSCSTE for the financial support.

I want to express my gratitude to the revisors of the project proposal for giving such a

wonderful opportunity to carry out 3 years of Post-Doctoral research. I humbly extend my

thanks to all concerned persons who co-operated with me in this regard.

Finally I thank God for the wisdom and perseverance that he has bestowed upon me during

this project and indeed through-out my life.

Page 5: Biotransformation of saponins to diosgenin for enhanced

5

ABSTRACT

Diosgenin is a hydrolysate of dioscin found in the rootstock of yam (Dioscorea) and

exists widely in the natural plant in the form of glucoside. It is the major base chemical for

several steroid hormones and an active ingredient in the oral contraceptive pill. The most

promising source of diosgenin is Dioscorea sp. In the Initial stage of our studies we selected 3

dioscorea sp (Dioscorea composita, floribunda and esculanta) for diosgenin production. From

this only one sp Dioscoria floribunda was screened for the further studies due to the high

concentration of diosgenin in the tuber. Enzymatically treated floribunda tubers were

employed for the studies. Multienzyme producing fungal strains were employed for the

production of diosgenin from the treated tubers. During project period we have isolated 32

multienzyme producing fungal strains. From which the most productive strain was selected

through primary and secondary screening. The strain was identified as pencilium chrysogenum

and is deposited in NCBI with accession number MH201392 and was employed for the

diosgenin production from Dioscorea tuber. Different fermentation factors and parameters for

diosgenin production using this fungal strain also studied using RSM technique. This study has

demonstrated that treatment with multienzyme producing pencilium chrysogenum is a very

effective and eco-friendly approach for the cleaner production of diosgenin from the tubers of

Dioscorea floribunda. The results show that the novel method enhances product yield and also

reduces the usages of water, acid and organic solvents.

Page 6: Biotransformation of saponins to diosgenin for enhanced

6

CHAPTER -1

INTRODUCTION & REVIEW OF LITERATURE

Traditional knowledge of medicine has long been used since ages for curing various

human ailments. About 60-80% of world populations still rely on plant-based medicines.

Though the traditional Indian system of medicine has a long history of use, yet they lack

adequate scientific documentation, particularly in light of modern scientific knowledge. The

medicinal value of plant lies in the bioactive phytochemical constituents of the plant and which

shows various physiological effects on human body. So through phytochemical screening one

could detect the various important compounds which could be used as the base of modern drugs

for curing various diseases. Keeping this in view, the plant Dioscorea commonly known as

yam has been taken for phytochemical screening.

Dioscorea, a pan-continental genus belonging to the family Dioscoreaceae, is found in

Africa, India, Southeast Asia, Australia and tropical America, with about 630 scientifically

described taxa. Prain and Burkill (1936) reported the occurrence of about 50 different

Dioscorea in India, largely in the west, east and northeast regions. In addition to the importance

of many Dioscorea species (yams) as starchy staple food, some representatives are used as a

source for the steroidal saponin diosgenin. Diosgenin occurs in plants in the form of saponins

attaching glucose or rhamnose to aglycone by glyco-sidic bonds at C-3 and C-26 (Qian et al.,

2006). Diosgenin has been reported to significantly used a precursor for partial synthesis of

oral contraceptives, sex hormones, and other steroids. It also shows pharmacological activities

such as antilipoperoxidative (Jayachandran et al., 2009) and antiskin aging (Yayoi et al., 2009)

effects. Preparation of diosgenin from saponins mainly depends on hydrolyzation of sugars at

these two positions. In industry, sulfuric acid and solvent extraction method are usually applied

in hydrolyzing raw herb to produce diosgenin. This method, however, is associated with many

environmental problems due to the high concentration of chemical oxygen demand and acid in

wastewater (Zhao et al., 2008; Cheng et al., 2009). Efforts have been made by many researchers

to solve this problem by focusing on clean methods to produce diosgenin. If microorganisms

could be applied in trans-forming saponins from the treated tubers, the cost and environmental

pollution of the biological process would be significantly reduced.

Out of the total steroid drug precursors, diosgenin accounts for 60% of the steroidal products

in the world. The current global demand for diosgenin is approximately between 50,000 and

80,000 kg/annum. About 10,000 tons of Dioscorea tubers per annum is the current requirement

for diosgenin in pharmaceutical industry. In India, commercial production of steroidal drugs in

Page 7: Biotransformation of saponins to diosgenin for enhanced

7

pharmaceutical industry is totally based on diosgenin which is about 450 tones. So this project

may help to increase the production of diosgenin through microbial transformation and may

boost up the industrial production of steroidal drugs. The current annual production of

diosgenin is 30 tones which is well short of global requirements (150 ton) and therefore relies

on production of new plant species and new production methods, including biotechnological

approaches. Emphasizes should be given to develop techniques for enhancing diosgenin

production which can overcome the deficit of the current techniques applied for its synthesis.

Dioscorea zingiberensis C. H. Wright is the dominant source for diosgenin production

in China. However, overexploitation of natural D. zingiberensis has led to a rapid decrease of

this plant resource and a sharp shortage of diosgenin for pharmaceutical synthesis. Plant cell

culture has been considered as an efficient and convenient alternative for the production of

diosgenin, but the low yield of diosgenin obtained in suspension cultures makes a barrier for

its commercialization. Therefore great efforts have been made seeking strategies for

improvement of secondary metabolite production, such as selection of cell lines with high

productivity, optimization of medium and culture conditions, application of genetic

engineering and biotransformation, use of immobilization and permeabilization of cell cultures,

and enhancement of secondary metabolite production by using elicitors. In the specific project

we employed fungal preparations as elicitor in batch fermentation technique and become one

of the most important and successful method to enhance secondary metabolite production in

Dioscorea sp.

Medicinal yam

Dioscorea is the largest genus among the monocotyledons of over 850 species in the world of

angiosperms and was first described by Robert Brown in 1810 (Arackal et al., 2015). The roots

generally known as yam, furnish to the basis of the staple starchy food and employed in the

third position as the food crop in the world next to cereals and pulses. So has an immense value

during the period of scarcity of food and was mainly cultivated in South East Asia, Africa and

South America. In addition to the importance of many yams as starchy staple food, some

representatives are used as a source for the steroidal saponin diosgenin, which is the starting

material of industrial interest in the synthesis of many steroids which are on the market as anti-

inflammatory, androgenic, estrogenic, and contraceptive drugs (Djerassi, 1992; Sautour et al.,

2007). In addition to sapogenins, alkaloids, steroid derivatives, phenolic compounds are also

found in yam so few varieties were used in traditional hunting and fishing, and other traditional

practices (Onwueme, 1978; Osagie 1992; Degras, 1993; IITA, 1995). Diosgenin play an

important role among the tribal communities as a traditional medicine to cure different diseases

Page 8: Biotransformation of saponins to diosgenin for enhanced

8

like hypercholesterolemia, hypertriacylglycerolemia, diabetes and hyperglycemia (Chen et al.,

2011). Many South Asians use syrup of the root, Powdered tubers, plant juice etc to treat

different illness like cholera, constipation, piles, skin diseases obesity etc (Kumar et al., 2013).

Modern medicine also extensively studied, diosgenin as a therapeutic agent for different illness

like cancer (Sethi et al., 2018), osteoporosis (Chiang et al., 2011), cardiovascular diseases

(Kalailingam et al.,2014), atherosclerosis (Lv et al., 2015), diabetes mellitus (Hua et al., 2016),

and skin diseases (Kim et al.,2016).

Chemical synthesis of diosgenin is not attain successes therefore Dioscorea species are

the only source for diosgenin. Edible Dioscorea species lacks exceeding amounts of steroidal

drug diosgenin. Therefore wild Dioscorea are cultivated in different countries like Mexico,

China, India, Europe etc. About 15 species of Dioscorea genus known to contains diosgenin in

which the highest concentrations (6- 8%) of diosgenin has been found in D. floribunda Mart.

& Gal. from Mexico (Correll, et al., 1955). D. floribunda is suitable for cultivation in Kerala,

Karnataka, West Bengal, Assam and Thamilnadu and was introduced by Indian Institute of

Horticultural Research. Tubers of D. floribunda were elongated growing horizondaly upto 15-

50cm long, skin white with brown bark, semi woody, somewhat hairy, heavily wrinkled or

rectaculate with amorphous establishment. Dioscorea floribunda can be propagated by tuber

pieces, single node stem cuttings or seed. Commercial planting is normally established by tuber

pieces only. During the dry season, the plants are inclined to dormancy and vine dieback.

Diosgenin (C27H42O3) belongs to the family of spirostanol steroidal compounds with

molecular mass 414.62 (Cai et al., 2020). Diosegenin is relatively stable to light and

temperature exposure. It is highly soluble in most nonpolar organic solvents such as

chloroform, dichloroethane, propanol, ethyl acetate etc and partially in polar solvents such as

acetone, methanol, and anhydrous ethanol (Cai et al., 2020). The oral bioavailability of

diosgenin is very low due to poor aqueous solubility and strong hydrophobicity. In plants it

noticed in the form of saponins attaching glucose or rhamnose to aglycone by glyco-sidic bonds

at C-3 and C-26 (Qian et al., 2006). Preparation of diosgenin from saponins mainly depends

on hydrolyzation of sugars at these two positions. In industry, sulfuric acid and solvent

extraction method are usually applied in hydrolyzing raw herb to produce diosgenin. This

method, however, is associated with many environmental problems due to the high

concentration of chemical oxygen demand and acid in wastewater (Zhao et al., 2008; Cheng et

al., 2009).

Medicinal Significance of Diosgenin

Page 9: Biotransformation of saponins to diosgenin for enhanced

9

Diosgenin, one of the most important secondary metabolites present in Dioscorea tuber

and is successfully exploited in a number of commercial applications in food, cosmetics,

agriculture and pharmaceutical sectors. Pharmaceutical applications of saponins include as raw

materials for production of hormones (Blunden et al., 1975), immunological adjuvants (Kensil

et al., 2004), treatment of cognitive impairment (Chuang et al., 2011) and as drugs. It is a

precursor of sex hormones (progesterone), corticosteroids (corticosone) and contraceptives

(Onwueme, 1978; Coursey, 1967). Diosgenin induces apoptosis in cancerous cells and in HeLa

cells by different pathways (Huo et al., 2004). Diosgenyl saponins induce apoptosis and mitotic

arrest in human leukemia cell lines (Ming-Jie, 2004). Diosgenin has both antioxidant property

and anticholesterolomic activity. Cholesterol-lowering activity of saponins, which was

demonstrated in animal (Matsuura, 2001), and human trials were attributed to inhibition of the

absorption of cholesterol from the small intestine, or the re-absorption of bile acids (Oakenfull

and Sidhu, 1990).

In cosmetics, saponins are being utilized as natural surfactants in cleansing products in

the personal care sector such as shower gels, shampoos, foam baths, hair conditioners and

lotions, bath/shower detergents, liquid soaps, baby care products, mouth washes, and

toothpastes (Indena, 2005; Brand and Brand, 2004; Olmstead, 2002). Saponins and sapogenins

are also marketed as bioactive ingredients in cosmetic formulations with claims to delay the

aging process of the skin (Yoo et al., 2003; Bonte et al., 1998) and prevent acne (Bombardelli

et al., 2001).

Extraction techniques of Diosgenin

Because of the biological activities, numerous researches on separation and purification

of diosgenin from herb have widely been explored through the traditional separation techniques

such as liquid-liquid extraction (LLE) and solid-phase extraction (SPE), which can meet the

separating requirements of purity. The first step in the processing of saponins involves their

extraction from the plant matrix. Factors that determine extraction efficiency and the characters

of the end product are the extraction solvent, extraction conditions and sample pretreatments.

The sample processing methods and temperatures used for drying have considerable effect on

the quality of the medicinal plant materials. Shade drying or drying at lower temperatures are

the suitable method. Lower temperature can maintain loss of color of the plant material and

loss of volatile substances in the plant materials (Ibanez et al., 2003, Bartram, 1995).

A few analytical methods for the Diosgenin estimation from plant material were

mentioned as follows. These protocols refer to use pulversied plant material of approximately

8.0 gm of fresh tubers/whole plantlet. The hydrolysed sample for 4 h with hydrochloric acid

Page 10: Biotransformation of saponins to diosgenin for enhanced

10

were filtered using Qualigen filter paper No. 615 and washed with distilled water until the

residue was acid free. The washed residue was extracted with petroleum ether (Boiling point:

60-80°C) in a soxhlet extractor for 4-6 h. The solvent was evaporated and the residue dissolved

in HPLC grade light petroleum ether and isopropanol (12:1). It was then filtered into a

measuring flask using a sample clarification kit (Millipore, Bedford, MA) consisting of a 10

mL syringe, filter holder and Millipore filters (0. 5 mM) (Dixit et al., 2003). Another general

method consist of using fresh tubers, which were cleaned under running tap water and dried by

wiping with clean cloth/tissue. The whole plantlet/ tubers were chopped and dried. The dried

tubers/whole plantlet were powdered and mixed with 50 mL of distilled water with

simultaneous stirring for 10 minutes in round bottom flask. To the slurry add distilled water

and concentrated hydrochloric acid in accord to maintain 5% of acid concentration (w/v). The

flask fixed with condenser was refluxed on a boiling water bath for 2 hour 30 minutes to 3.0

hour to complete the hydrolysis. After the hydrolysis, this slurry was allowed to attain room

temperature and filtered in a Buchner funnel under vacuum. The residue was washed with

distilled water till the filtrate is free from acid. The acid free residue was transferred to Petri

dish and dried in an oven at 100°C at 6 hours. The dried residue was extracted with n-hexane

in a soxhlet apparatus for 8 hours. The extracted solvent containing Diosgenin was

concentrated, chilled on ice (0°C) and filtered. The mother liquor obtained after filtering was

again concentrated, chilled on ice and re-extracted.

Diosgenin obtained from extractions were pooled and weighed after drying (for 2 hours at

80°C) temperature and values were expressed on dry weight basis (Nandi, 1980).

Application of microbes for the extraction of microbes

Microorganisms can secrete enzymes rapidly responding to the ambient environment and

reduce product inhibition effects through metabolism, microbial treatment is suggested to be

more effective than direct enzymatic treatment in enzymatic hydrolysis. In addition, the high

cost of enzymes can be reduced when microbial treatment is employed in place of enzymatic

treatment. Therefore, it is of great interest to screen microorganisms which can efficiently

secrete related enzymes that can disintegrate the tuber compositions, and to apply them in

microbial treatment to promote the release of saponins. After recovering the starch from dried

tuber, Trichoderma reesei exhibited a significant effect on biotransformation of saponion.

Moreover, Trichoderma harzianum and Aspergillus oryzae were also found can convert

saponin into diosgenin through biotransformation (Dong et al., 2009, Liu et al., 2010). Inspired

by the above said successful research, Pencillium strain had been selected as fermentation

strain for biotransformation after a series of screen work.

Page 11: Biotransformation of saponins to diosgenin for enhanced

11

OBJECTIVES:

Collection and cultivation of Dioscorea composita, Dioscorea floribunda and

Dioscorea esculanta

Isolation and identification of efficient cellulase and amylaseproducing fungal

strains for diosgenin production.

To investigate the effects of different pretreatment methods for fungal growth

and enzyme production in microbial transformation procedure.

Optimization diosgenin production using most prominent strain by response

surface methodology.

Purity analysis of diosgenin using HPLC studies

Development of an effective microbial system for diosgenin production using

fungal strains.

Page 12: Biotransformation of saponins to diosgenin for enhanced

12

CHAPTER -2

MATERIALS AND METHODS

Plant collection and taxonomic studies

Extensive survey has been conducted in Western Ghats for collecting different species of

Dioscorea. Specimens were collected in flowering and fruiting conditions preferably in

quadupilcate and relavant field notes on them were recorded in the field book then and there.

Special attention was paid to gather data pertaining to note habit, habitat and other features like

colour of the flowers, fragrance etc. which cannot be deduced from the examinations of the

herbarium specimens. The photographs of the specimens were also taken. The taxonamic

identities of the collected materials were confirmed with the help of various regional and

adjacent countries floras and also by consulting with authentic specimens depositries in various

National Herberium like CAL, BLAT, BSI, BSD, BSIM, NBG, MH, TBGT, CALI etc.

In the laboratory, the specimens were carefully examined under zoom stereomicroscope on the

morphological characters of both aerial and underground parts. micromorphological characters

were observed under a compound research microscope. Observations on venation pattern were

carried out after clearing the leaves. Clearing was done by immersing leaves in 10% KOH in a

petridish for about 12 hours at 60 C and washing in running water. The specimens were stained

with safranin. Excess safranin was removed by washing again in water. The specimens were

then dried and mounted on butter paper and examined with zoom steriomicroscope and

illustration was made.

Comparative analysis of diosgenin content in Dioscorea esculanta, floribunda and

composita

Diosgenin extraction was carried out according to (Drapeau et al., 1986) dried and powdered

tubers (10g each) were refluxed with10 ml 2N HCL for 2 hours to hydrolyse the saponins to

sapogenins and sugars. The residue seperarated by filtration was washed with 50ml of water,

dried at 650C for 2 hours and extracted with chloroform in a soxhlet apparatus. The extracts

were filtered through nylon 0.45µm membrane filters (PAL Gelman Laboratory, India) and

concentrated to dryness. The dried residue was then dissolved in 1ml of 100% (v/v) methanol.

The diosgenin concentration was determined by high pressure liquid chromatography (HPLC)

according to the method described by Huang et al., 2010.

Soil collection and isolation of fungal culture

Soil samples were randomly collected from 4-5 cm depth with help of sterile spatula from the

various locations in Western Ghats. Isolation of fungal colony was performed by serial dilution

and spread plate method. One gram of soil sample was serially diluted in sterilized distilled

Page 13: Biotransformation of saponins to diosgenin for enhanced

13

water to get a concentration range from 10-1 to 10-6. A volume of 0.1 ml of each dilution was

transferred aseptically to Potato dextrose agar plates. The sample was spreaded uniformly using

a glass rod. The plates were incubated at 28 °C for 72 hr.

The selected fungal isolates were screened for amylolytic activity by starch hydrolysis test on

starch agar plate. The selected fungal isolates were streaked on the starch agar plate and

incubated at 37°C for 2-3 days. After incubation iodine solution was flooded with dropper for

30 seconds on the starch agar plate (Malik et al., 2017).

Screening of potent cellulase producing fungi

The isolated fungus was grown on carboxymethylcellulose agar medium. The pure cultures

were inoculated in the centre with almost equal amounts and incubated at 30 ± 2°C until

substantial growth was recorded. The Petri plates were flooded with Congo red solution (0.1%),

and after 5min the Congo red solution was discarded, and the plates were washed with 1N NaCl

solution, allowed to stand for 15 - 20 minutes (Reddy et al., 2014).

Determination of soil pH

Soil pH was determined in 1: 3.0 soil/water ratio by a combination glass electrode HI98129,

Hanna Instruments.

Determination of soil organic carbon and soil organic matter

The soil organic C (SOC) content was estimated by dichromate oxidation method in which the

oxidation of K2Cr2O7 in a concentrated H2SO4 medium and the excess dichromate was

measured using (NH4)2Fe(SO4)2 (Yeomans and Bremner, 1989). Soil organic matter (SOM)

were determined according to Pribyl, 2010.

Quantitative enzyme assays

Amylase assay

Enzyme production medium

Production medium contained (g/l) peptone- 26.7g; dipottasiumorthophosphate – 2.7g; tween

80 – 7.3ml; soluble starch10%. 100 ml of medium was taken in a 250 ml conical flask. The

flasks were sterilized in autoclave at 1210C for 15 min and after cooling the flask was

inoculated with fungal cultures. The inoculated medium was incubated at 270C in shaker

incubator for different incubation time. At the end of the fermentation period, the culture

medium was centrifuged at 5000rpm for 15 min to obtain the crude extract, which served as

enzyme source. The enzyme activity was assayed following the method of using 3, 5-

dinitrosalicylic acid.

Amylase activity was determined by measuring the release of reducing sugar from starch by

DNS method (Miller 1959). The reaction mixture contains 0.2ml of crude enzyme and 0.8ml

Page 14: Biotransformation of saponins to diosgenin for enhanced

14

of 100mM phosphate buffer (pH 7) containing 1% (W/V) of soluble starch. The mixture was

indubated at 550C for the reaction was stopped by adding 2ml of DNSA (3, 5dinitrosalicyclic

acid). The content were boiled exactly for 5 minute in water bath and cooled for 20- 25 minutes

after which 1ml of 40% Rochelle salt (sodium pottassium tartarate) was added. Finally the

colour developed was read at 540nm in a spectrophotometer. The amount of reducing sugar

released in the mixture was determined.

Cellulase assay

The positive fungal strains were used to know their potential for cellulase production and

activities. A volume of 100 ml of Czapek-Dox broth medium amended with 1% cellulose was

distributed into separate 250 ml conical flasks. The pH of the medium was adjusted to 5. After

autoclaving at 121°C and 15 lb. pressure, the fungal spore suspensions were inoculated into the

conical flasks. The flasks were incubated at 27 °C on a rotary shaker at 120 rpm for 3 days.

After 3 days, culture filtrate was collected, centrifuged at 6000 rpm for 15 min and supernatant

was used to the estimation of cellulase source.

Activity of Cellulase in the culture filtrates was determined and quantified by carboxy-methyl

cellulase method (Ghosh 1987). The reaction mixture with 1.0 ml of 1% carboxymethyl

cellulose in 0.2 M acetate buffer (pH 5.0) was pre-incubated at 50°C in a water bath for 20

minutes. An aliquot of 0.5 ml of culture filtrate with appropriate dilution was added to the

reaction mixture and incubated at 50 °C in water bath for one h. Appropriate control without

enzyme was simultaneously run. The reducing sugar produced in the reaction mixture was

determined by dinitro- salicylic acid (DNS) method (Miller 1959). 3, 5-dinitro-salicylic acid

reagent was added to aliquots of the reaction mixture and the color developed was read at

wavelength 510 nm.

Morphological & microscopic identification of selected fungal strain

Morphological characters were studied by inoculating the fungal isolate onto Czapek Solution

Agar (CZA) which contained (g/L): Sucrose, 30.0; NaNO3, 2.0; KH2PO4, 1.0; MgSO4.7H2O,

0.5; KCl, 0.5; FeSO4.7H2O, 0.01; Agar, 15.0; pH 7.3 ± 0.2 and incubated for 5 to 7 days. Every

24 h plates were examined and the colony characteristics like surface and reverse colony

colour, colony margins, elevations, growth rate etc were noted.

Micro-morphological characters were studied by staining the 5-day old fungal colonies with

lactophenol cotton blue. One loopful of culture was aseptically transferred onto a clean glass

slide with the help of sterile inoculating needle. The slide was placed on a staining tray, flooded

with lactophenol cotton blue and left it for 1 min. A clean cover slip was placed onto it with

the help of a needle and excess stain was blotted with bibulous paper and examined under low,

Page 15: Biotransformation of saponins to diosgenin for enhanced

15

high & oil immersion objectives. The selected isolates were maintained on Sabouraud Dextrose

Agar (SDA) slants and stored at 4 °C for further study.

Molecular characterization of selected strains

Molecular characterization was performed by isolating genomic DNA followed by PCR

analysis and sequencing.

DNA isolation

DNA was extracted by modified CTAB method described by Möller et al. (1992). 50 mg of

mycelia was scraped from 10 day old fungal cultures. This was manually ground in 1.5 mL

micro-centrifuge tubes with a micro-pestle adding 500 µL of pre-warmed (60°C) TES lysis

buffer (100 mM Tris pH 8.0; 10 mM EDTA; pH 8.0; 2% SDS). 50 µg of proteinase K were

added to the ground material and incubated at 60 °C for 60 min. To the suspension 140 µL of

5 M NaCl and 64 µL of 10% (w/v) CTAB were added and incubated at 65 °C for 10 min. DNA

was extracted by adding equal amount of phenol: chloroform: isoamyl alcohol (25:24:1) and

centrifuged at 14000 g for 10 min. The supernatant was collected and equal amount of

chloroform: isoamyl alcohol (24:1) was added and centrifuged at 14000 g for 10 min. DNA

was precipitated by adding 0.6 volume of cold isopropanol and 0.1 volume of 3 M sodium

acetate, pH 5.2 and maintained at -20 °C overnight. The DNA was pelleted out by

centrifugation at 12000 rpm for 10 min at 4 °C and washed twice with 70% ethanol and

suspended in 50 µL TE buffer. RNA was digested by adding 10 mg/mL of RNase and incubated

at 37 °C for 45 min and stored at -20 °C for further use.

PCR amplification of ITS region

PCR amplification was carried out in 25 μl reaction mixture containing 2.5 μl of 10X

amplification buffer (100 mM Tris HCl, pH-8 at 25 °C, 15 mM MgCl2, 500 mM KCl and 10%

Triton X-100), 0.2 μl of 25 mM dNTP mixture, 0.74 U of Taq polymerase (Finzyme, Finland),

1μl each of the primer pair ITS4 (5’TCCTCCGCTTATTGATATGC-3’) and ITS5 (5’-

GGAAGTAAAAGTCGT AAC-3’) (Integrated DNA Technologies, Inc., USA) and 40 ng of

genomic DNA.

Bio-rad thermal cycler (S 1000TM) was used for amplification with the following PCR profile:

an initial denaturation for 5 min at 97 °C, followed by 40 cycles of 1 min at 97 °C, 1 min at 48

°C and 2 min at 72 °C and a final extension at 72 °C for 5 min. The amplified products were

resolved in 1.2% agarose gel containing 0.5 mg/mL ethidium bromide.

Sequencing using bigdye terminator v3.1

Page 16: Biotransformation of saponins to diosgenin for enhanced

16

Sequencing reaction was done in a PCR thermal cycler (GeneAmp PCR System 9700, Applied

Biosystems) using the BigDye Terminator v3.1 Cycle sequencing Kit (Applied Biosystems,

USA) following manufactures protocol.

The PCR mix consisted of the following components:

PCR Product (ExoSAP treated) - 10-20 ng Primer - 3.2 pM (either Forward or Reverse)

Sequencing Mix - 0.28 µL 5x Reaction buffer - 1.86 µL Sterile distilled water - make up

to 10 µL. The sequencing PCR temperature profile consisted of a 1st cycle at 96 °C for 2 min

followed by 30 cycles at 96 °C for 30 sec, 50 °C for 40 sec and 60 °C for 4 min for all the

primers.

Sequence analysis

The sequence quality was checked using Sequence Scanner Software v1 (Applied Biosystems).

Sequence alignment and required editing of the obtained sequences were carried out using

Geneious Pro v5.1 (Drummond et al., 2010). The nucleotide sequences obtained were

compared with already available in the databank of the NCBI, using BLAST search tool

(Altschul et al.,1990) for the identification of the 5 isolates. The identification of the species

was determined based on the best score.

Phylogenetic analysis

Homology search of the ITS sequence obtained was performed using BLAST search algorithm.

Alignment of similar sequences was done using CLUSTAL W multiple alignment software

and the phylogenetic tree was constructed using MEGA4 software. Distance estimation was

done following maximum composite likelihood method by Tamura et al. (2007). The stability

of relationship was assessed from bootstrap analysis of the neighbour-joining data.

Different pretreatment methods for fungal growth and enzyme production in microbial

transformation

Physical treatment (P1)

100g of dried tuber were emerged in 3L water for 24 hours and is then cut into slices, grounded

for 40S. the wet powder was suspended and partitioned in a large amount of water to form 3

layers. The top layer is fiber, the middle and bottom layers were combined grounded and

suspended again to remove fiber. The residue was suspended and partitioned. the bottom layer

was starch, the top and middle layers were combined, centrifuged and dried at 60oC and

grounded to pass through a 60-mesh screen thus physically treated tuber was obtained.

Catalytic solvent extraction (P2)

100g of dried tuber were grounded. The dried tuber was mixed with 1.2 L of 60% EtOH, 0.15g

of NaHCO3 and 0.3g of NaOH was then added. The mixture was stirred at 700C for 2 h. the

Page 17: Biotransformation of saponins to diosgenin for enhanced

17

slurry was then filtered. The residue was extracted with 50% EtOH again. the two filtrates

were combined and concentrated.

Enzymatic treatment (P3)

100g of dried tuber were ground for 2 minutes the dried powder was mixed with 600 ml of

water and boiled for 1h. After cooling, the slurry was incubated with 2g amylase at 700C and

p H 6.5 for 1h. The hydrolysate was then centrifuged and washed with water. The residue was

dried at 600C and grounded to pass through a 60- meshscreen.

Complex enzymatic treatment (P4)

100g of dried tuber were grounded for 2 min. the dried powder was mixed with 600ml water

and then incubated with 1.5g of cellulase and 1.5g of amylase at 550C and pH 4.0 for 6 h. the

enzymatic hydrolysate was centrifuged and washed with water. the residue was dried at 600C

and grounded to pass through a 60-mesh screen.

Optimization diosgenin production using most prominent strain by response surface

methodology

The conventional one factor at a time optimization approach is time consuming and not

available to evaluate the effects of independent variables and interactions between different

factors. Therefore RSM has been widely used in the optimization of microbial transformation

process by building models, designing experiments, evaluate the effect of variables and

optimization conditions.

Microbial transformation

Microbial transformation experiments was taken out with pretreated (P4) substrate (10%) and

fermentation medium containing peptone, K2HPO4 and tween 80 with pH 5.8 and incubated

with 300ml of the sub-cultured fungal spore suspension with an agitation rate of 300 rpm for

168h. Optimization of fermentation conditions was carried out using response surface

methodology (RSM). A three factor and five level central composite design, consisting of 20

experimental trials was employed. The design contained three independent variables –

peptone, K2HPO4 and tween 80. The response variable (y)was diosgenin yield (%) resulting

from Pencillium chrysogenum. The experimental data from 20 runs were analyzed with RSM

algorithm of design expert 10 and fitted to a second order polynominal equation.

Y=β0+∑ 𝛽𝑛𝑖=1 iXi+∑ 𝛽𝑛

𝑖=1 ii Xi2+ ∑ ∑ 𝛽𝑛

𝑗=1𝑛𝑖=1 ijXiXj

where Y is the predicted response, β0 is an offset term, βi is the linear effect, βj the quadratic

effect, βij is the interaction effect, and Xi, Xj are the levels of the independent variables.

Design-Expert 10 (Stat- Ease, Inc., Minneapolis, MN, USA), was used for the regression

Page 18: Biotransformation of saponins to diosgenin for enhanced

18

analysis and plotting of graphs. The statistical significance of the model equation and the

model terms were evaluated via the Fisher’s test. The coefficient of determination (R2) and

adjusted R2 determines the quality of fit of the model using a second order regression equation.

The polynomial equation is further expressed in the form of three-dimensional response

surface plots inorder for a better understanding of the relationship between the response of

various levels of each variable in the trial. The combinations of all significant variables which

gave the highest response were also determined from the model.

Experimental Validation of the Response model

In order to validate the response surface model, a random set of experiments were setup

according to conditions predicted by the model. The validation experiments were done for all

the significant variables in the design space. A random set of six combinations of variables

were prepared and analyzed for protease production. The experimental values should be in

close agreement with the statistically predicted ones to confirm the authenticity and

applicability of the statistical model (RSM) for the optimization of process variables.

Diosgenin yield

Sample medium (10ml) in each flask from optimization tests was dried, extracted with 10.0ml

CHCL3 by ultrasonic treatment for 30 min and filtrated. Diosgenin content in filtrate was

analyzed by HPLC and diosgenin yield was calculated as

𝑍 =𝑐𝑚 − 𝑐0𝑐𝑎 − 𝑐0

× 100

Where Z is diosgenin yield (%), Cm and C a are contents of diosgenin in 0.5g substrate after

microbial and acid treatment, Co is content of diosgenin in substrate.

Page 19: Biotransformation of saponins to diosgenin for enhanced

19

CHAPTER -3

RESULTS AND DISCUSSIONS

Morphological description of Dioscorea sp

Dioscorea floribunda

Tubers elongated growing horizondaly upto 15- 50cm long, skin white with brown bark, semi

woody, some what hairy, heavily wrinkled or rectaculate with amorphous establishment. Stems

unarmed but with short stings, twines to the right, terete. Leaves 7.5 × 1.4 × 4-9cm long

triangular ovate or ovate lanceolate to slightly sagitate, exstipulate, 7-9 nerved, the base

truncate to deeply and widely cordate, apex acute to accuminate petiole 4.5 – 8.5cm long.

Pulvinous slightly pinkish spiny. Flowers 44-50 per spike. Flowers in auxillary panicles to

9cm long pedicles 0.5- 1mm long, bracts1.5 – 4mm long, purplish or brown, perianthtube 0.5-

3mm long lobes ovate withwhite or cream margin. Stamens 6, didynamous,3 long ca 1.5mm,

3 short ca 1mm long inserted almost in the center of the disc. Female flower auxillary clusters

30- 35cm long, rachis sharply angled, bracts 1.5- 3mm long purple or dark brown, bracteoles

0.5 – 2mm, style ca 1mmlong, bifid, terete, thin, capsule 21-25 × 17 × 20mm oblanceolate to

obvoate or subquadrate, membraneous, brown to reddish, the base acute to rounded to

emarginated. Seeds 2 per locules 10 -13mm × 7- 9mm oblong to elliptical, winged peripherally,

reddish brown smooth.

Fig: 1 Dioscorea floribunda

Dioscorea composita

Tubers 50- 70cm long with whitish cream, yellow or pik. Stem robust, terete, twins to right.

Leaves alternate ovate or ovate lanceolate or subordicular bloched with white patches, rarely

Page 20: Biotransformation of saponins to diosgenin for enhanced

20

small and ovate – triangular, estipulate and very prominent. Base cordate apex abruptly acute

or shortly acuminate. Nerves 7 -9 , petioles 0.5 -1mm long slightly pulvinous. Flowers 2- 4

spike, pedicle 0.5- 1mm long, bracts 2-3mm brown or brown to purple bracteoles 1.5mm long

ovate, lobed. Stamens 6, inserted in the center of the torus, 3 long eac ca 0.2mm long, triangular

to conical inconspicuous. Female flowers with one clusters ca 17-40cm long, axillary, bracts

1.5- 2mm long brown or purple, bractioles 1mm long, staminode 0.2mm long, triangular,

inserted at the periphery of the torus semi errect. Ovary globose ca 2mm long, stylar column,

1mm long, bifid, terete slender. Capsules 25- 37 × 18 -22mm, oblong to elliptic, semi ligneous,

brown base rounded to oblique, apex rounded. Seeds two per locule 5- 7mm long, oblong,

winged perpherally smooth, reddish brown. Male flower axillary or terminal panicles, 1-2

clusters 30- 70cm long rachis grooved glabrous.

Fig: 2 Dioscorea composita

Dioscorea esculanta

Tubers several, ca-40 in numbers each upto 12cm long and 10cm diameter, weight upto 3-4kg.

the underground parts consisting of a hard knot, feeding roots, which arising from it, protective

thorns, laxs or close bunch of storage tubers arising from the woody base of the stem lying just

beneath the soil surface, shortly stalked tubers oblong ellipsoidor cylindrical, flattened and

lobed covered with root fibers, some raises bearing spines, skin brown to greenish brown, very

thin, flesh soft white edible varying slightly in sweetness. Stem usually one rarely morethan

one arising from the woody knot, not terete at the base but terete in the upper parts. Sometimes

tinged with purple colour towards the base, but usually green, twines to the left, pubescent but

Page 21: Biotransformation of saponins to diosgenin for enhanced

21

glabrescent with age, prickles seen often densely at the base, upto 3mm long, straight or slightly

deflected downwards. Bulbils absent. Leaves alternate cordate, sometimes broadly ovate, blade

cordiform with an obtusesinus, membraneous, acuminate, glabrescentabove, pubescent

beneath, nerves 9-13, the inner 5 veins reaching apex, the outer most pair super basal ,

secondary veins almost parallel margin distinct, petioles almost as long as blade, 4-15cm long

pubescent with a small prickles upon them, stipules prickle like, larger than internodal prickles

upto 7mm long pubscent, directed downwards. Staminate inflorescence solitary axillary

spikes, the rachis firm and ascending, ca 10-14cm long, some what angled, pubscent. Flowers

sterile, 70- 100 per spike covered with thin wightish hairs, opening irregularly along the axis,

2.5-3.5× 3.5 – 5mm shortly pedicelled, bracts ovate, acuminate scarious 1.5 -3.2 × 1- 1.5mm

long, bracteoles similar to bractsouter tepals 0.8- 1.5× 1.5- 3mm, the lobes oblong to ovate,

acute, 1-1.7 × 1- 1.2mm. stames 6, inserted at the rim or half way up the perinath tube, the

filament 0.2 – 0.6mm long with the lobes closely held by a narrow connective, pistillode

conical. Pistillate inflorescence very rare arranged axillary, solitary, spike like racemens, the

axis upto 40- 45cm long, pendent, perbescent. Flowers up to 50-60 per spike, shortly

pedicelled, bracts ovate, acuminate at apex ca. 2mm long, pubescent scarious at the margins,

bracteoles 0.8- 1mm long. Sepals 6 arranged in two whorls of 3 each, each narrowly ovate,

acute, ca 1.5 × 0.5mm, pubscent outside, glabrous within. Slaminodes 6, ovary densely

pubscent, style connate the stimas terminally reflexed as 3 pairs of reflexed hooks. Capsule

oblong, obovoid, basically rounded to turnate, apically retuse, ca 27 × 2.5cm glabrascent,

refluxed upwards. Seeds broadly winged all rouned.

Fig : 3 Dioscorea esculanta

Comparative analysis of diosgenin content in dioscorea esculanta, floribunda and

composite

Page 22: Biotransformation of saponins to diosgenin for enhanced

22

Page 23: Biotransformation of saponins to diosgenin for enhanced

23

Page 24: Biotransformation of saponins to diosgenin for enhanced

24

Page 25: Biotransformation of saponins to diosgenin for enhanced

25

Page 26: Biotransformation of saponins to diosgenin for enhanced

26

Page 27: Biotransformation of saponins to diosgenin for enhanced

27

Soil collection and isolation of fungal culture

In this investigation, we have navigated amylolytic and cellulolytic fungi from different

locations of south Western Ghat mountain rain forest (Ponmudi, Kallar, Kulathupuzha,

Menmutty and Marayoor) and the evergreen forest of Wayanad (Fig:4), which marks the

transition zone between the north and southern ecological region of Western Ghats. The

southern ecological regions are more wetter and species rich and that’s why we have selected

these south Western Ghat regions. pH and EC are the most significant parameters for measuring

soil quality and soil microbial biomass. Soil samples collected from six high land areas were

showed slight variations in PH ranging from 5.6 to 8 (Table :1). Strains from different land

areas were showed significantly wide ranges in cellulase and amylase activity and were more

prominent in regions having neutral pH and electrical conductivity in the range of 43 - 49µs.

Highest EC and pH was recorded in Ponmudi and Wayanad, fungal strains isolated from

Ponmudi and wayanad were not able to produce both cellulase and amylase. However the

number of organisms isolated from both wayanad and Ponmudi were competable with other

sites. Strains isolated from locations with neutral pH and less electrical conductivity (Kallar,

Meenmutty, Kulathupuzha and Marayoor) were able to produce both cellulase and amylase but

the microbial biomass was less.

Page 28: Biotransformation of saponins to diosgenin for enhanced

28

Fig:4 sampling regions

Primary screening of potent amylase and cellulase producing fungi

A total of thirty-two fungal isolates were scraped up from different regions of Western Ghats.

All the selected isolates were primary screened for production of amylase and cellulase using

starch agar plate method and carboxymethyl cellulose plate method. Freshly prepared single

spore cultures of fungal strains were point inoculated on the centre of the plates and incubated

at 300 C for 3 days. In case of amylase producing strains hydrolysis of starch around the

colonies were visualized by flooding the plates with Gram’s iodine solution (Fig: 13). The zone

formation around the colony was due to the hydrolysis of starch by amylolytic enzymes

produced by the strains. For detecting cellulolytic activity CMC agar plates were flooded with

0.1% (w/v) Congo red solution for 15 minutes followed by destaining with 1M NaCl solution

for 15 minutes. Congo red clearing zone assay is suitable for qualitative display of cellulase

activity (Fig: 14). The clearing zone of enzymatic activity will be visible around the batch of

growth. The NaCl solution elutes the dye in the clearing zone where the cellulose has been

degraded into simple sugars by the enzymatic activity. Only eight fungal isolates (Fig: 2 – 12)

were found to be positive for both amylase and cellulase production, as determined by

measuring the width of the clear zone (zone of hydrolysis) formed around the fungal colonies

on starch agar (SA) medium and carboxymethyl cellulose (CMC) agar medium. The fungal

isolate TBGRI-7 isolated from Marayoor showed maximum zone of hydrolysis i.e. 1.7cm and

1.0 cm in starch agar and carboxymethyl cellulose agar medium respectively. Followed by

TBGRI- 4 (1.2 cm, 1.0 cm), TBGRI -1 (1.1 cm, 0.8 cm), TBGRI- 2, 5 & 16 (1.0 cm, 0.72 cm),

TBGRI – 12 (0.8 cm, 1.2 cm) and TBGRI - 14 (0.5 cm, 0.8 cm), respectively in SA and CMC

agar plates respectively.

Page 29: Biotransformation of saponins to diosgenin for enhanced

29

Fig: 5 TBGRI 1 Fig: 6 TBGRI 2

Fig: 7 TBGRI 3 Fig: 8 TBGRI 4

Fig: 9 TBGRI 5 Fig : 10 TBGRI 10

Page 30: Biotransformation of saponins to diosgenin for enhanced

30

Fig: 11 TBGRI 7 Fig: 12 TBGRI 8

Fig:13 Amylolytic activity in starch agar medium Fig: 14 Cellulolytic activity in CMC

medium

Si

no

Sample

collection sites

Number

of fungal

strains

collected

Number and

name of

cellulase and

amylase

positive

strains

PH Organic

carbon

Soil

organic

matter

1 Wayanad 7 0 5.6 3.09% 5.32

2 Ponmudi 6 0 8 1.68% 2.88

3 Kallar 4 3 (TBGRI -5

14 & 16)

6.9 10.02% 17.23

Page 31: Biotransformation of saponins to diosgenin for enhanced

31

4 Kulathupuzha 4 1 (TBGRI-1) 7 3% 5.16

5 Meenmutty 5 2 (TBGRI 2

& 4)

7.11 0.99% 1.70

6 Marayoor 6 2 (TBGRI-, 7

and 12)

7.20 2% 3.44

Table: 1 Soil physiological characters

Secondary screening

Cellulase and amylase assay

Batch fermentation was carried out in triplicate flasks and enzymatic activity was estimated at

regular intervals. The differences among the mean values data to the activity obtained at at

different hours were statistically tested using one way ANOVA. Pre-induced fungal spores

(1×107 spores/mL) were inoculated onto sterilized media and incubated. The culture filtrate

was centrifuged at 12,000 rpm for 30 min at 4 °C and assayed for enzyme activity. The isolate

TBG RI - 14 showed maximum amylase activity of about 56.43 U/mL on 5th day of incubation

followed by TBGRI – 4 (52.25 U/mL), TBGRI-5 (50.69 U/mL), TBGRI – 7 (46.5 U/mL),

TBGRI – 2 (45.35 U/mL), TBGRI – 16 (40.02 U/mL), TBGRI – 12 (37.47 U/mL) and TBGRI

- 1 (35.30 U/mL) (Fig : 11). TBGRI – 1and 14 were the strains showing maximum activity at

5th day the other six strains showing their maximum activity at 4th day. The isolate TBG RI -

5 showed maximum cellulase activity of about 380.19 U/mL on 4th day of incubation followed

by TBGRI – 4 (363.05 U/mL), TBGRI-16 (343.44 U/mL), TBGRI – 14 (336.71 U/mL),

TBGRI – 7 (335 U/mL), TBGRI – 1 (300 U/mL), TBGRI – 12 (273.72 U/mL) and TBGRI -

2 (266.93 U/mL) (Fig: 12). All the strains showed maximum cellulose production on 4th day.

0

10

20

30

40

50

60

70

TBGRI 1 TBGRI 2 TBGRI 4 TBGRI 5 TBGRI 7 TBGRI 12 TBGRI 14 TBGRI 16

Enzy

me

acti

vity

(U/m

l)

DAY 3

DAY 4

DAY 5

DAY 6

DAY 7

Page 32: Biotransformation of saponins to diosgenin for enhanced

32

Fig: 15 Amylase production by different fungal strains

Fig: 16 Cellulase production by different fungal strains

Morphological studies

Colonies on Czapek solution agar attaining 50 mm diameter after seven days, colony

color was initially white becoming deep green to black with conidial production, reverse mostly

pale brown with entire margins and rapid growth

Fig: 17 colony morphology of selected fungal strain

Microscopic studies

Conidia were subglobose, smooth, with length 2.8µm and 2.5µm width. Stipes was smooth

with 258µm length and 3.4 µm width. Phiallides were in flask shaped and following quarte

verticillate pattern.

0

50

100

150

200

250

300

350

400

450

TBGRI -1 TBGRI - 2 TBGRI - 4 TBGRI - 5 TBGRI - 7 TBGRI- 12 TBGRI - 14 TBGRI - 16

DAY -3

DAY -4

DAY- 5

DAY - 6

DAY -7

Page 33: Biotransformation of saponins to diosgenin for enhanced

33

Fig: 18 Microscopic characteristic

Molecular characterization

Molecular characterization of the pencillium was done by extracting genomic DNA followed

by amplification of ITS regions and sequencing. The DNA was extracted by modified CTAB

method (Moller et al., 1992) and the OD260/OD280 ratio was found to be between 1.8 and 2.

The extracted genomic DNA was resolved in 0.8% agarose gel containing 0.5 mg/mL ethidium

bromide.

Amplification of ITS1-5.8S-ITS2 rDNA fragments were done using the primer pair ITS4 and

ITS5 and the molecular size of the product were found to be 821 bp (Figure: 19).

Fig: 19 The amplified product was sequenced using the BigDye Terminator v3.1 Cycle

sequencing Kit (Applied Biosystems, USA) following manufactures protocol and the sequence

obtained as follows.

TBGRI -14

Page 34: Biotransformation of saponins to diosgenin for enhanced

34

CCCGTTAGGGGGGCCCCCCGAAACAACAAGGTAAATTAAAACAAGGGGGGAGTT

GGACCC

AGAGGGCCCTCACTCGGTAATTCCTCCGCTTATTGATATGCTTAAGTTCAGCGGG

TAAAT

CCATACCTGATCCGAGGTCAACCTGGATAAAAATTTGGGTTGATCGGCAAGCGC

CGGCCG

GGCCTACAGAGCGGGTGACAAAGCCCCATACGCTCGAGGACCGGACGCGGTGCC

GCCGCT

GCCTTTCGGGCCCGTCCCCCGGGATCGGAGGACGGGGCCCAACACACAAGCCGT

GCTTGA

GGGCAGAAATGACGCTCGGACAGGCATGCCCCCCGGAATACCAGGGGGCGCAAT

GTGCGT

TCAAAGACTCGATGATTCACTGAATTTGCAATTCACATTACGTATCGCATTTCGC

TGCGT

TCTTCATCGATGCCGGAACCAAGAGATCCGTTGTTGAAAGTTTTAAATAATTTAT

ATTTT

CACTCAGACTACAATCTTCAGACAGAGTTCGAGGGTGTCTTCGGCGGGCGCGGG

CCCGGG

GGCGTAAGCCCCCCGGCGGCCAGTTAAGGCGGGCCCGCCGAAGCAACAAGGTAA

AATAAA

CACGGGTGGGAGGTTGGACCCAGAGGGACCACCTCCCCCACTAACGGGGGAGAC

GAGATG

ATCCTTCCGTAACAGGTTCATCGATCAAATGCGGAAGTACCGAGTGAGGGCCCTC

TGGGT

CCACCTCCACCCTGTTTATTTTACTTTGGTGGCTTCGGCGGGCCGGCTAAATGGCC

CGGG

GGGGCTTAAGACCCGGGCCGCACCCAAAAACCCCGAATTTT

Phylogentic Analysis

Page 35: Biotransformation of saponins to diosgenin for enhanced

35

Different pretreatment methods for fungal growth and enzyme production in microbial

transformation

Pretreatments recover the starch and fiber molecules from tuber this releases saponins from

the network of cell wall and may obliging for the production of diosgenin. The effects of

different pretreatment methods on the properties of dried tuber were showed in table :1

Chemical

composition

of Dioscorea

floribunda

Acid

hydrolysis

Physical

treatment

(P1)

Catalytic

solvent

extraction

(P2)

Enzymatic

treatment

(P3)

Complex

enzymatic

treatment

(P4)

Saponin 3.4% 1.3% 1.5% 2.01% 2.8%

Optimization diosgenin production using most prominent strain by response surface

methodology

Optimization of different nutritional fermentation parameters (Tweem 80, K2HPO4 and

peptone were analyzed by using RSM to exploit the production. The addition of peptone offers

the nitrogen source for fungal growth and is the main component of proteins and nucleic acids

(Zhu et al., 2010). According to Doppelbauer et al., 1987 addition of peptone from 0.05 to

0.2% in the medium increased β-glucosidase production. Another factor optimized was the

buffering agent, K2HPO4. It boosts the diosgenin production by supplementing phosphate and

potassium. Phosphate in K2HPO4 plays a title role in electron transport and energy cycle during

oxidative phosphorylation. Pottasium is a key ion involved in the enzyme stereo-inversion

Page 36: Biotransformation of saponins to diosgenin for enhanced

36

reaction (Zhu et al., 2010). Wen et al., 2005 reported that T.reesi reduces the cellulase

production during the purging of phosphate from the medium. Tween 80 is the most

significant component of the medium it provides the transfer of oxygen and nutrients in the

medium to promote fungal growth it alters the membrane surface property and increase the

enzyme production. There by rises the diosgenin production (Zhu et al., 2010).

From the CCD experiments of 20 runs was carried out to optimize medium composition

and the results were presented in table: 2. The design matrix and the corresponding results of

RSM experiments to determine the effect of three independent variables are shown in table: 3

along with mean, predicted values. The NOVA analysis of the study indicated that the model

terms A and C were significant. The effect of Tween 80 (c) was more significant than the other

factors. The effect of peptone (A) was also significant in comparison with effect of K2HPO4

(B). The model F value was 7.40, the value of lack of fit was 2.46 and was not significant.

The higher F value and non-significant lack of fit proves the model to be appropriate.

By applying multiple regression analysis on the experimental coded data and a second order

polynominal equation for diosgenin yield was obtained

Diosgenin yield = 23.48 +1.08*A + 0.50*B + 1.38*C

Where A is peptone concentration, B is Dipottasium hydrogen orthophosphate concentration,

C is Tween 80.

The regression equation obtained from ANOVA showed that R2 (Multiple Correlation

Coefficient) was 0.5169. For a good statistical model, the R2 value should be in range of 0-1.0

and the near to 1.0 value is more fit model is deemed to be. The predicted R2 value is 0.2946

and is not close to the adjacent R2 (0.5160). The difference is more than 0.2. this may indicate

a large block effect. The adequate precision value was 9.781. a ratio grater than 4 is desirable.

So the model an adequate signal and can be used to navigate the design.

Factor 1 Factor 2 Factor 3 Response 1

Std Block Run A:Peptone conc B:K2HPO4 C:Tween 80 Diosgenin yield

% % % %

3 Block 1 1 0.2 1 0.1 21

2 Block 1 2 4 0.1 0.1 23.7

8 Block 1 3 4 1 1 30

11 Block 1 4 2.1 0.55 0.55 22

4 Block 1 5 4 1 0.1 25.3

Page 37: Biotransformation of saponins to diosgenin for enhanced

37

1 Block 1 6 0.2 0.1 0.1 23

10 Block 1 7 2.1 0.55 0.55 22.8

6 Block 1 8 4 0.1 1 27

9 Block 1 9 2.1 0.55 0.55 22.4

7 Block 1 10 0.2 1 1 25

5 Block 1 11 0.2 0.1 1 25.3

12 Block 1 12 2.1 0.55 0.55 24.5

20 Block 2 13 2.1 0.55 0.55 23

18 Block 2 14 2.1 0.55 1.30681 23.7

14 Block 2 15 5.29541 0.55 0.55 23.8

16 Block 2 16 2.1 1.30681 0.55 24

17 Block 2 17 2.1 0.55 -0.206807 21

13 Block 2 18 -1.09541 0.55 0.55 22

19 Block 2 19 2.1 0.55 0.55 22.2

15 Block 2 20 2.1 -0.206807 0.55 21.3

Table :2 central composite design matrix for peptone, K2HPO4 and tween 80 and experimental

designs

Source Sum of

squares

Degree of

freedom

Mean

square

F-value P-value>F

Block 14.01 1 14.01

Model 45.30 3 15.10 7.40 0.0029 significant

A- Peptone

conc 15.88 1 15.88 7.78 0.0138

B-K2HPO4 3.43 1 3.43 1.68 0.2147

C-Tween 80 25.99 1 25.99 12.73 0.0028

Residual 30.62 15 2.04

Lack of fit 26.67 11 2.42 2.46 0.2000 not

significant

Pure error 3.95 4 0.99

Cor.Total 89.93 19

Page 38: Biotransformation of saponins to diosgenin for enhanced

38

Table :3 Box Behnken design matrix for the experimental design and predicted responses for

diosgenin production

R2 = 0.5967; CV = 6.04%; Adj R2 = 0.5160; Pred R2 = 0.2946

Fig:1 Graph showing the predicted vs actual values of the design matrix

Fig:2 Three-dimensional response surface plot for diosgenin production showing the

interactive effects of the K2HPO4 and peptone concentration

Design-Expert® SoftwareDiosgenin yield

Color points by value ofDiosgenin yield:

30

21

Actual

Pred

icted

Predicted vs. Actual

20

22

24

26

28

30

20 22 24 26 28 30

Design-Expert® SoftwareFactor Coding: ActualDiosgenin yield (%)

30

21

X1 = A: Peptone concX2 = B: K2HPO4

Actual FactorC: Tween 80 = 0.306757

0.1 0.2

0.3 0.4

0.5 0.6

0.7 0.8

0.9 1

0.2

1.15

2.1

3.05

4

18

20

22

24

26

28

30

Dio

sgen

in y

ield

(%

)

A: Peptone conc (%)B: K2HPO4 (%)

Page 39: Biotransformation of saponins to diosgenin for enhanced

39

Fig: 3 Three-dimensional response surface plot for diosgenin production showing the

interactive effects of the peptone concentration and tween 80

Fig:4 Three-dimensional response surface plot for diosgenin production showing the

interactive effects of the tween 80 and K2HPO4

validation studies

Design-Expert® SoftwareFactor Coding: ActualDiosgenin yield (%)

Design points above predicted value30

21

X1 = C: Tween 80X2 = A: Peptone conc

Actual FactorB: K2HPO4 = 0.1

0.2

1.15

2.1

3.05

4

0.1 0.2

0.3 0.4

0.5 0.6

0.7 0.8

0.9 1

18

20

22

24

26

28

30

Dio

sge

nin

yie

ld (

%)

C: Tween 80 (%)A: Peptone conc (%)

Design-Expert® SoftwareFactor Coding: ActualDiosgenin yield (%)

Design points above predicted valueDesign points below predicted value30

21

X1 = B: K2HPO4X2 = C: Tween 80

Actual FactorA: Peptone conc = 2.1

0.1 0.2

0.3 0.4

0.5 0.6

0.7 0.8

0.9 1

0.1 0.2

0.3 0.4

0.5 0.6

0.7 0.8

0.9 1

18

20

22

24

26

28

30

Dio

sge

nin

yie

ld (

%)

B: K2HPO4 (%)C: Tween 80 (%)

Page 40: Biotransformation of saponins to diosgenin for enhanced

40

y = 0.9998xR² = 0.9796

25.4

25.5

25.6

25.7

25.8

25.9

26

26.1

26.2

26.3

25.4 25.5 25.6 25.7 25.8 25.9 26 26.1 26.2 26.3 26.4

Chart Title

Page 41: Biotransformation of saponins to diosgenin for enhanced

41

SUMMARY

Diosgenin, obtained from Dioscorea tubers, is the major base chemical for several

steroid hormones and an active ingredient in the oral contraceptive pill. The most promising

source of diosgenin is Dioscorea sp. In the Initial stage of our studies we selected 3 dioscorea

sp (Dioscorea composita, floribunda and esculanta) for diosgenin production. From this only

one sp Dioscoria floribunda was screened for the further studies due to the high concentration

of diosgenin in the tuber. Enzymatically treated floribunda tubers were employed for the

studies. Multienzyme producing fungal strains were for the production of diosgenin from the

treated tubers. During project period we have isolated 32 multienzyme producing fungal

strains. From which the most productive strain was selected through primary and secondary

screening. The strain was identified as pencilium chrysogenum and is deposited in NCBI with

accession number MH201392 and was employed for the diosgenin production from Dioscorea

tuber. This study has demonstrated that treatment with multienzyme producing pencilium

chrysogenum is a very effective and eco-friendly approach for the cleaner production of

diosgenin from the tubers of Dioscorea floribunda. The results show that the novel method

enhances product yield and also reduces the usages of water, acid and organic solvents.

Page 42: Biotransformation of saponins to diosgenin for enhanced

42

SCOPE OF FUTURE WORK

Microorganisms are capable of producing a great variety of enzymes within a short

period of time due to their high rate of cell multiplication. Therefore, microbial treatment has

been studied for centuries for the development of a compound through an environmentally

friendly approach. In our studies it was proven that a multienzyme producing pencilium

chrysogenum is able to produce the diosgenin from Dioscrea floribunda tubers via an

ecofriendly method. In this sense, a reasonable number of compounds of various biological

interests can be furnished by the help of microorganisms- an eco-friendly transformation of

natural products.

Page 43: Biotransformation of saponins to diosgenin for enhanced

43

OUT COMES OF THE PROJECTS

SALIENT FINDINGS:

Isolated multienzyme producing fungal strains

Total of 32 fungal strains were isolated from Western Ghats area, in which 8 strains

were identified as multienzyme producers.

Identified potential fungal strains

Potent multienzyme producing strain (pencillium chrysogenum) was identified based

on the morphological, biochemical and molecular characterisation.

Pencillium chrysogenum was deposited in NCBI with accession number MH201392

Germplasm storage of Dioscoria floribunda and composite collected from Western

Ghats.

An eco-friendly diosgenin production was developed.

PUBLICATIONS:

International

S.R. Reji and N.S. Pradeep. (2019) “Isolation and Selection of Fungal Strains for

Multienzyme Production from Western Ghats” International Journal of Agriculture,

Environment and Biotechnology, Citation: IJAEB: 12(1): 23-32.

Lekshmi K Edison, Vipin Mohan Dan, Reji. S. R & N. S. Pradeep. (2020) “Insilico

Perceptions in Structural Elucidation of Exo-Beeta-1,3 Glucanase (endo 13) from

Streptomyces spp” -Accepted by the journal applied biology and biotechnology with

manuscript number - JABB – 2020-07-209.

CONFERENCES/WORKSHOPS/SEMINARS/ PROCEEDINGS etc..

International:

Reji.S.R and Pradeep N. S., 2017. “Exploring Western Ghats fungal diversity for

cellulase and amylase production”. International conference on frontiers in bioscience,

15 -16th November 2017 at SNGIST Arts and science college, North Paravur.

National :

Reji. S. R and Pradeep N. S, (2017) “Microbial diversity of cellulase and amylase

producing fungal strains from Western Ghats of India”. National symposium on future

of functional genomics, 13 -14th October 2017 at Transdisciplinary University,

Bengaluru.

Page 44: Biotransformation of saponins to diosgenin for enhanced

44

Reji. S. R and N. S. Pradeep, (2016) “Isolation and screening of fungal strains for

multienzyme production from Western Ghats” National seminar on insights into the

interdisciplinary perspectives of chemical and biosciences, 26 -28th February 2018 at

Government arts college, Thiruvananthapuram.

Page 45: Biotransformation of saponins to diosgenin for enhanced

45

REFERENCES

1. Cai B, Zhang Y, Wang Z, Xu D, Jia Y, Guan Y, Liao A, Liu G, Chun C, Li J (2020)

Therapeutic potential of diosgenin and its major derivatives against neurological

diseases: recent advances, Oxidative medicine and cellular longevity, Article ID

3153082, https://doi.org/10.1155/2020/3153082

2. Chen P S, Shih Y W, Huang H C and Cheng H W, Diosgenin – A steroidal sapogenin

inhibits migration and invasion of Human Prostrate cancer PC – 3 cells by reducing

matrix metalloproteinases expression. PloS One; 2011; 6; e20164. doi:

10.1371/journal.pone.oo20164.

3. Cheng P, Zhao H Z, Zhao B, Ni J R (2009) Pilot treatment of wastewater from

Dioscorea zingiberensis C.H. Wright production by anaerobic digestion combined with

a biological aerated filter, Bioresour Technol, 12:2918–2925.

4. Cheng P, Zhao H Z, Zhao B, Ni J R (2009) Pilot treatment of wastewater from

Dioscorea zingiberensis C.H. Wright production by anaerobic digestion combined with

a biological aerated filter, Bioresour Technol, 12:2918–2925.

5. Chiang S.S, Chang S. P and Pan T. M (2011) Osteoprotective effect of Monascus-

fermented Dioscorea in ovariectomized rat model of postmenopausal osteoporosis,

Journal of Agricultural and Food Chemistry, 59 (17) 9150–9157.

6. Correll D G, Schubert B G, Gentry H S, and Hawley W O (1955) The search for plant

precursors of cortisone, Econ. Bot 9, 305- 375.

7. Degras L M (1993) The yam: A tropical root crop. The Technical Centre for

Agricultural and Rural Cooperation (CTA). The McMillan Press, London. 408 pp.

8. Djerassi C. (1992), Steroid research at Syntex: ‘The pill’and cortisone. Steroids 57,

631D641.

9. Drapeau D, Sauvaire Y, Blanch H W, Wilke C R (1986) Improvement of diosgenin

yield from Dioscorea deltoidea plant cell culture by use of a non-traditional hydrolysis

method. Planta Med 6: 474-478.

10. Georg- Alexander Hoyer (1975) Diosgenin saponins from Dioscrea Floribunda,

Phytochemistry 14 (2) 539 -542.

11. Hua S, Li Y, Su L and Liu X (2016) Diosgenin ameliorates gestational diabetes

through inhibition of sterol regulatory element-binding protein-1,” Biomedicine &

Pharmacotherapy, 84, 1460–1465.

Page 46: Biotransformation of saponins to diosgenin for enhanced

46

12. Huang W, Zhao H Z, Ni J R, Zuo H, Qiu L L, Li H (2008) The best utilization of D.

zingiberensis C. H. Wright by an eco-friendly process, Bioresour Technol, l99:7407–

7411

13. Hui Li, Xiangdong Wang, Yang Ma, Nannan Yang, Xiaojuan Zhang, Zhenghong Xu

(2016) Purification and characterization of a glycosidase with hydrolyzing multi 3-0

glycosidase of spirostanol saponins activity from Gibberella intermedia 128: 46- 51.

14. International Institute of Tropical Agriculture (IITA). 1995. Annual Report. Ibadan,

Nigeria. IITA.

15. Jayachandran K S, Vasanthi H R, Rajamanickam G V (2009) Antilipoperoxidative and

membrane stabilizing effect of diosgenin, in experimentally induced myocardial

infarction, Mol Cell Biochem, 327:203–210.

16. Kalailingam P, Kannaian B, Tamilmani E, and Kaliaperumal R (2014) Efficacy of

natural diosgenin on cardiovascular risk, insulin secretion, and beta cells in

streptozotocin (STZ)-induced diabetic rats, Phytomedicine, 21(10) 1154–1161.

17. Kim J E, Go J, Koh E K (2016) Diosgenin effectively suppresses skin inflammation

induced by phthalic anhydride in IL-4/Luc/CNS-1 transgenic mice, Bioscience,

Biotechnology and Biochemistry, 80 (5) 891–901.

18. Kumar S, Anuo Kumar Parida and Padan Kumar Jena (2013) Ethano -Medico-Biology

of Ban- Alu (Dioscorea species): A neglected tuber crops of Odisha, India, International

Journal of Pharmacy and life science, 4 (12) 3143- 3150.

19. Lv Y C, Yang J and Yao F (2015) Diosgenin inhibits atherosclerosis of ATP-binding

cassette transporter A1, Atherosclerosis, 240 (1) 80–89.

20. Ofelia Espejo, Jesus Campos Llavot, Helgi Jung, Francisco Giral (1982) Sprirostanic

diosgenin precursors from Dioscorea composite tubers, Phytochemistry, 21(2) 413-

416.

21. Onwueme I C (1978) Yams, cassava, sweet potato and cocoyams. In: The Tropical

Tuber Crops. John Wiley and Sons, New York, USA 234pp.

22. Osagie A U (1992) The yam tuber in storage, Post-harvest Research Unit. University

of Benin, Benin City, Nigeria. 247 pp.

23. Prain D, Burkill IH. 1936. An account of the Genus Dioscorea in the East. Annals of

the Royal Botanical Gardens, Culcutta. Longman, London , UK . 57

24. Qian S H, Yuan L H, Yang N Y, Ou Yang P K (2006) Study on steroidal compounds

from Dioscorea zingiberensis, Chinese Traditional Herbal Drugs, 29:1174–1176.

Page 47: Biotransformation of saponins to diosgenin for enhanced

47

25. Qian S H, Yuan L H, Yang N Y, Ou Yang P K (2006) Study on steroidal compounds

from Dioscorea zingiberensis, Chinese Traditional Herbal Drugs, 29:1174–1176.

26. Rytas Vigalys and Mark Hester (1990) Rapid genetic identification and mapping of

enzymatically amplified ribosomal DNA from several Cryptococcus Sp, Journal of

bacteriology 172 (8) 4238- 4246.

27. Sethi G, Shanmugam M and Warrier S (2018) Pro-apoptotic and anti-cancer properties

of diosgenin: a comprehensive and critical review, Nutrients,10 (5) 645 -.

28. Yayoi T, Naoko K, Akinori H, Megumi T, Hideyo U, Shinichi W (2009) Novel effects

of diosgenin on skin aging, Steroids, 74:504–511.

29. Yu – Linh Zhu, Wen Huang, Jin- Ren Ni, Wei Liu (2010) Production diosgenin from

Dioscorea zingiberensis tubers through enzymatic saccharification and microbial trans

formation 85: 1409- 1416.

30. Yu- Ling Zhu, Wen Huang, Jin- Ren Ni, Wei Liu, Hui Li, (2010) Production of

diosgenin from Dioscorea zingiberensis tubers through enzymatic saccharification and

microbial transformation, Appl microbial Biotechnol, 85: 1409- 1416.

31. Zhang Q, Lo C M, Ju L K (2007) Factors affecting foaming behavior in cellulase

fermentation by Trichoderma reesei Rut C-30, Bioresour Technol, 98:753–760.

32. Zhao H Z, Cheng P, Zhao B, Ni J R (2008) Yellow ginger processing wastewater

treatment by a hybrid biological process, Process Biochem, 43:1427–1431.

33. Zhao H Z, Cheng P, Zhao B, Ni J R (2008) Yellow ginger processing wastewater

treatment by a hybrid biological process, Process Biochem, 43:1427–1431.

34. Zheng T X, Yu L D, Zhu Y L (2014) Evalution of different pretreatment on microbial

transformation of saponins in Dioscorea zingiberensis for diosgenin production,

Biotechnol Biotechno Equip 28(4) 740- 746.