isolation, characterization and cosmeceutical application

117
Isolation, Characterization and Cosmeceutical Application of Bioactive Secondary Metabolites from Marine Fungi By Shivankar Agrawal B. Pharm. (Ayurveda), M.Sc. (Biochemistry) Submitted in fulfillment of the requirements for the degree of Doctor of Philosophy Deakin University September, 2017

Upload: others

Post on 24-Dec-2021

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Isolation, Characterization and Cosmeceutical Application

Isolation, Characterization and Cosmeceutical

Application of Bioactive Secondary Metabolites

from Marine Fungi

By

Shivankar Agrawal

B. Pharm. (Ayurveda), M.Sc. (Biochemistry)

Submitted in fulfillment of the requirements for the degree of

Doctor of Philosophy

Deakin University

September, 2017

Page 2: Isolation, Characterization and Cosmeceutical Application
sfol
Retracted Stamp
Page 3: Isolation, Characterization and Cosmeceutical Application
sfol
Retracted Stamp
Page 4: Isolation, Characterization and Cosmeceutical Application

iii

I dedicate this thesis to God Almighty and my family…..

Page 5: Isolation, Characterization and Cosmeceutical Application

iv

Acknowledgements

“Don’t take rest after your first victory because if you fail in second,

more lips are waiting to say that your first victory was just luck.”

- A.P.J Abdul Kalam

I would like to express my sincere gratitude and admiration to my Principal

supervisor Prof. Colin J. Barrow for his expert guidance, support and unflinching

encouragement throughout my Ph.D. journey. I would like to thank him for providing

excellent scientific advice through my entire thesis report with immense patience and

providing his valuable suggestions, even after his demanding schedule.

I put on record to express my deepest gratitude and profound indebtedness towards my TERI

lead supervisor, Dr. Sunil Kumar Deshmukh, Fellow and Area Convenor, Nano

Biotechnology Centre, TERI for his immense support and guidance. I would like to

thank him for his valuable discussions towards the designing of the experiments and

publications of the work.

I am extremely thankful to my associate supervisor Dr. Alok Adholeya, Director,

Nano Biotechnology Centre, TERI for giving me the opportunity to carry out this

research. Dr. Alok was the main source of inspiration behind this project. Additionally, Dr.

Alok provided me with every opportunity for success during my Ph.D. career while

allowing me the freedom to individually develop my own scientific approach to

problems. He has always been there to support and provide an insightful answer to all

my queries. His positive attitude, scientific temperament and words like “There is

nothing like No Results in research” have always been a great motivation.

I would like to take this opportunity to pay sincere gratitude to my former Associate

supervisor, Dr. Nisha Aggarwal (not working with TERI anymore) for her support in

the first two years of my Ph.D. program. She has always been a very caring and

understanding person.

I sincerely acknowledge to Dr. Debabrata Acharya, Dr. Pannalal Dey, all Fellow and

Associate Fellow of Teri-Deakin nanotechnology research center for their caring and

valuable help and support.

Page 6: Isolation, Characterization and Cosmeceutical Application

v

I would also like to thank Mr. Chandrakant Tripathi and technical staff for their

expert assistance and help in handling and working on various instruments.

I am thankful to my seniors for their friendly, but valuable suggestions and guidance

throughout.

I would like to thank all my lab mates for their support. Extending, this thanks to all

the lab attendants for their help in my daily experiments.

Finally, very special thanks to my very good friends Ranjit Singh, Sreeparna

Samantha, Sandeep Sure, Amritpreet Kaur Minhas, Rita Choudhary, Ankita Bedi for

their friendship, encouragement, moral and emotional support and great company.

I would also like to thank the administrative staff both at TERI and Deakin

International Office, New Delhi and Deakin University, Australia for their help and

support.

I would like to acknowledge the financial, academic and technical support of Deakin

University, Australia and TERI, India for pursuing my Ph.D. in the field of Nano

biotechnology

Last but not least, I would like to express my gratitude to my parents and my brothers

for their support and helping me to pray.

Shivankar Agrawal

Page 7: Isolation, Characterization and Cosmeceutical Application

vi

List of Publications

Published

1. Agrawal, S., Adholeya, A., & Deshmukh, S. K. (2016). The Pharmacological

Potential of Non-ribosomal Peptides from Marine Sponge and Tunicates. Frontiers in

pharmacology, 7 DOI: 10.3389/fphar.2016.00333

Communicated

1. Shivankar Agrawal, Alok Adholeya, Colin J. Barrow, Sunil Kumar Deshmukh.

“Screening of marine fungi for potential tyrosinase inhibitor and antioxidants for

cosmeceutical application”. Submitted in BMC complementary and alternative

medicine.

2. Shivankar Agrawal, Alok Adholeya, Colin J. Barrow, Sunil Kumar

Deshmukh. “In-vitro evaluation of marine derived fungi against

Propionibacterium acnes”. Submitted in Anaerobe.

3. Shivankar Agrawal, Colin J. Barrow, Sunil Kumar Deshmukh, Alok Adholeya.

“Marine fungi: An untapped Bioresource for future cosmeceutical”. Submitted in

Phytochemistry Letters.

Poster presentations

1. Shivankar Agrawal, Nisha Aggarwal, Mandira Kochar, Alok Adholeya*, Colin

Barrow. “Isolation and characterization of nonribosomal peptides from marine fungi

and their application in cosmetics”. At DIRI symposium 2012 organized by Deakin

University in association with TERI held at Gurgaon, Haryana on 27- 30, November

2012.

2. Shivankar Agrawal, Nisha Aggarwal, Mandira Kochar, Alok Adholeya, Colin

Barrow*. “Marine fungi as sources of nonribosomal peptides for cosmeceutical

applications”. Australia-India Strategic Research funded work shop on

Bio/nanotechnology applications to improve plant productivity and ecosystems

services during 5-7 March, 2014 at Geelong, Australia.

3. Shivankar Agrawal, Sunil Kumar Desmukh, Colin Barrow, Alok Adholeya*.

Cosmecutical potential of marine fungi isolated from west coast and Andaman

Page 8: Isolation, Characterization and Cosmeceutical Application

List of Publications

vii

Island, India. At DIRI symposium 2015 organized by Deakin University in

association with TERI held at Gurgaon, Haryana on 27-30, November 2015.

Oral presentations

1. “Screening for antibacterial activity of fungi from Indian marine environments:

A possible alternative for new antibiotics for the treatment of skin microbial

infections” in ICEP2017 19th

international conference on Ethnopharmacology and

Pharmacology at Singapore, 08-09, January 2017 and secured best presentation

award.

Page 9: Isolation, Characterization and Cosmeceutical Application

viii

Table of Contents

Chapter 1 ....................................................................................................................... 1

Introduction and literature review .................................................................................... 1

1. Introduction ................................................................................................................ 2

1.1. Marine natural products ........................................................................................ 2

1.2. Natural products from marine fungi ....................................................................... 5

1.2.1. Rate limiting factors for secondary metabolites biosynthesis in marine fungi ..... 7

1.3. Marine cosmeceuticals .......................................................................................... 7

1.3.1. Current status of cosmetics-cosmeceuticals...................................................... 8

1.4. Cosmeceuticals from marine fungi ........................................................................10

1.4.1. Marine fungi for prevention of skin aging and cancer ......................................10

1.4.2. Marine fungi for skin whitening .....................................................................12

1.4.3. Marine fungi for acne vulgaris .......................................................................15

2. Conclusion.................................................................................................................16

3. Aim of this Research ..................................................................................................17

Chapter 2 ......................................................................................................................18

Isolation and purification of marine fungi from a range of marine environment in India .....18

2.1. Introduction.............................................................................................................19

2.2. Materials and methods .............................................................................................20

2.2.1. Chemicals used .................................................................................................20

2.2.2. Study area ........................................................................................................20

2.2.3. Collection of samples ........................................................................................21

2.2.4. Isolation of marine fungi ...................................................................................22

2.2.5. Growth assessment of marine-derived fungal strains using different media conditions ..................................................................................................................23

2.3. Results and Discussion.............................................................................................23

2.3.2. Effect of nutrient concentration on fungal growth................................................30

2.4. Conclusion ..............................................................................................................33

Chapter 3 .......................................................................................................................34

Screening for antimicrobial activity of marine fungi against bacteria causing skin and wound infections in humans .......................................................................................................34

3.1. Introduction.............................................................................................................35

3.2. Materials and methods .............................................................................................38

3.2.1.. Chemicals used ................................................................................................38

3.2.2. Test organisms..................................................................................................38

3.2.2.1. Marine fungi ..............................................................................................38

3.2.2.2. Bacterial strains .........................................................................................38

3.2.2.3. Bacterial inoculum .....................................................................................38

Page 10: Isolation, Characterization and Cosmeceutical Application

Table of Contents

ix

3.2.3. Broth fermentation and extraction ......................................................................39

3.2.4. Antimicrobial assay in vitro ...............................................................................40

3.2.5. Determination of minimum inhibitory concentration (MIC) .................................40

3.2.6. Determination of the antibacterial mechanism.....................................................41

3.2.7. Morphological and molecular characterization of potential marine fungi strain .....41

3.2.7.1. Scanning electron microscopy (SEM) of fungi .............................................41

3.2.7.2. DNA extraction, PCR amplification and sequencing .....................................42

3.3. Results and discussion: ............................................................................................43

3.3.1. Antimicrobial screening ....................................................................................43

3.3.2. Determination of minimum inhibitory concentration (MIC) .................................46

3.3.3. Mechanism of action (SEM analysis) .................................................................47

3.3.4. Identification of marine fungi with strong antibacterial activity ............................51

3.4. Discussion...............................................................................................................55

3.5. Conclusion ..............................................................................................................57

Chapter 4 ......................................................................................................................58

Screening of marine fungi for potential tyrosinase inhibitor and antioxidants.....................58

4.1. Introduction.............................................................................................................59

4.2. Materials and methods .............................................................................................60

4.2.1. Chemicals used .................................................................................................60

4.2.2. Test organisms..................................................................................................60

4.2.2.1 Marine fungi ...............................................................................................60

4.2.3. Cosmeceutical activity ......................................................................................60

4.2.3.1 .Culture extracts ..........................................................................................60

4.2.3.2. Anti tyrosinase assay (MBTH assay) ...........................................................61

4.2.3.3. Antioxidant assay .......................................................................................61

4.2.3.4 Morphological and molecular characterization of potential fungal strains........62

4.3. Results ....................................................................................................................62

4.3.1. In vitro depigmenting activity ............................................................................62

4.3.2. Antioxidant activity of fungal extracts ................................................................66

4.3.3. Identification of promising marine fungal strains.................................................66

4.4. Discussion...............................................................................................................68

4.5. Conclusion ..............................................................................................................68

Chapter 5 ......................................................................................................................69

Bioassay-guided isolation of antibacterial compounds from marine fungus Simplicillum lamellicola.....................................................................................................................69

5.1. Introduction.............................................................................................................70

5.2. Method and materials ...............................................................................................71

5.2.1. Chemicals used .................................................................................................71

5.2.2. Large scale fermentation and extraction..............................................................71

Page 11: Isolation, Characterization and Cosmeceutical Application

Table of Contents

x

5.2.3. Detection of antibacterial compounds by thin layer chromatography with direct bioautography assay ...................................................................................................71

5.2.4. Bioassay-guided fractionation of S. lamellicola methanol extract .........................72

5.2.5. Identification of isolated antibacterial compound ................................................75

5.2.5.1. HPLC analysis ...........................................................................................75

5.2.5.2. UPLC-mass spectrometry analysis ..............................................................75

5.3. Results and discussions ............................................................................................76

5.3.1. Characterization of substances from the culture broth of marine S. lamellicola ......76

5.3.2. Antibacterial activity of pure active compound ...................................................80

5.4. Conclusions.............................................................................................................80

Chapter 6 .......................................................................................................................81

Summary and future work ...............................................................................................81

References .....................................................................................................................85

Page 12: Isolation, Characterization and Cosmeceutical Application

xi

List of Tables

Chapter 1

Table 1.1: Marine natural product that are FDA-approved agents or used in clinical trial ... 2

Table 1.2: Fungi from aquatic habitats from around the world.......................................... 6

Table 1.3: Examples of some cosmeceutical ingredients from marine sources ................... 9

Table 1.4: Tyrosinase inhibitors from marine fungi ...................................................... 14

Chapter 2

Table 2.1: Samples collected from different parts of India ............................................. 24

Table 2.2: Pure fungal isolates obtained from the samples collected from different habitats

............................................................................................................... ...26

Table 2.3: Fungal mycelial growth on media A .........................................................30-31

Table 2.4: Fungal mycelial growth on media B .........................................................31-32

Chapter 3

Table 3.1: Antibacterial activity of crude fungal extracts against pathogens involves in

complicated skin infections by agar well-diffusion method ........................45-46

Table 3.2: Phylogenetic affiliations of selected fungi with strong antibacterial

activity ....................................................................................................... 51

Chapter 4

Table 4.1: Phylogenetic affiliations of selected fungi with strong activity ...................... 67

Page 13: Isolation, Characterization and Cosmeceutical Application

xii

List of Figures

Chapter 1

Figure.1.1: Cosmetics and their relative branches ............................................................. 8

Figure.1.2: Worldwide major skin problems and their probable solutions ......................... 10

Figure.1.3: Telomerase inhibitors from marine sources ................................................... 12

Figure.1.4: Synthesis of melanin ................................................................................... 13

Figure.1.5: Tyrosinase inhibitors from marine fungi ....................................................... 15

Figure.1.6: Structure of Trichodin A-B .......................................................................... 16

Chapter 2

Figure.2.1: Sites of sample collection in India (Goa, Gujarat and Andaman Islands) ......... 21

Figure.2.2: Different locations where sampling was done and material collected. ............. 25

Figure.2.3: Plates showing the diversity of culturable fungal isolates from different parts of

India.......................................................................................................26-29

Figure.2.4: Fungal growth differences with nutrient avability on solid culture medium ..... 32

Chapter 3

Figure.3.1: Causative pathogens involves in complicated skin infections.......................... 36

Figure.3.2: Fungal secondary metabolites extraction .......................................................................... 39

Figure.3.3 (A and B): MIC of active fungal crude extracts against respective bacteria........ 47

Figure.3.4: Effect of fungal crude extract on Staphylococcus aureus................................ 48

Figure.3.5: Effect of fungal crude extract on Staphylococcus epidermidis ........................ 49

Figure.3.6: Effect of fungal crude extract on Bacillus megaterium ................................. 49

Figure.3.7: Effect of fungal crude extract on Pseudomonas aeruginosa and

Escherichia coli .......................................................................................... 50

Figure.3.8: Effect of fungal crude extract on Propionibacterium acnes ............................ 50

Figure.3.9: SEM morphological analysis of most active sporulating fungi ....................52-53

Figure.3.10: Phylogenetic tree of partial ITS-rDNA sequences of marine fungal strains ..... 54

Page 14: Isolation, Characterization and Cosmeceutical Application

List of Figureures

xiii

Chapter 4

Figure.4.1 (A): Anti tyrosinase activity of Pladi and Dwarka marine fungal crude extracts . 63

Figure.4.1 (B): Anti tyrosinase activity of Andaman and Goa marine fungal crude extracts.64

Figure.4.2: Anti tyrosinase activity of eleven selected fungal crude extracts at different

concentrations............................................................................................. 64

Figure.4.3 (A, B and C): Dose and time dependent anti tyrosinase activity of eleven selected

fungal crude extracts ................................................................................... 65

Figure.4.4: Scavenging activity of crude extracts of selected fungi against DPPH ............. 66

Figure.4.5: Phylogenetic tree of partial ITS-rDNA sequences of fungal strains ................. 67

Figure.4.6: SEM morphological analysis of sporulating D4 strain.................................... 67

Chapter 5

Figure.5.1: Schematic representations of isolation of metabolites from fungi “D5” crude

extract ........................................................................................................ 74

Figure.5.2: Schematic representations of isolation of active compound from active fraction

1.3.4 of “D5” crude extract .......................................................................... 74

Figure.5.3: TLC bioautography of fraction 1.3.4 of “D5” crude extract . . . . . . . . . . . . . . . . . . . . . . 76

Figure.5.4: HPLC chromatogram of minor active fraction 5A .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77

Figure.5.5: HPLC chromatogram of purified active compound from fraction 6A5 ............ 77

Figure.5.6: UPLC-MS chromatograms of minor active fraction 5A ................................. 78

Figure.5.7: UPLC-MS chromatograms of active fraction 6A5 ......................................... 78

Figure.5.8: Structures of compounds identified from marine S. lamellicola ...................... 79

Figure.5.9: Fraction 6A5 with moderate antibacterial activity against S. epidermidis. .... 80

Page 15: Isolation, Characterization and Cosmeceutical Application

xiv

List of Abbreviations

< - Smaller than

% - Percentage

~ - Approximately

µg/ml

- Microgram per millilitre

µl - Microlitre

µM - Micro molar

µm - Micrometre

0C - Degree Celsius

FDA - Food and Drug Administration

Pre-NDA - New Drug Application

ASW - Artificial sea water

BHIB - Brain heart infusion broth

BHIA - Brain heart infusion agar

cm - Centimetre

Cm-1 - per centimetre

CFU - colony-forming unit

CPD - Critical point drying

DMSO - Dimethyl sulfoxide

DPPH - 2,2-diphenyl-1-picrylhydrazylv

gm/ L - Grams per litre

gm - Grams

GC - Gas chromatography

HPLC - High Pressure Liquid Chromatography

hr - Hour

HRTEM - High Resolution TEM

IC50 - Half maximal inhibitory concentration

ITS - Internal transcribed spacer

kg - Kilograms

LC - Liquid chromatography

M - Molar

mbar - Millibars

mg - Milligrams

mM - Millimolar

min - Minutes

Page 16: Isolation, Characterization and Cosmeceutical Application

List of Abbreviations

xv

mL - Millilitre

mm - Millimetre

mg/ml - Milligrams per millilitre

MBTH - 3-methyl-bezothiazolinonehydrazone

MQ - Milli Q

MEB - Malt extract broth

MS - Mass spectroscopy

NCBI - National Centre for Biotechnology Information

ng - Nanogram

NA - Nutrient agar

NMR - Nuclear magnetic resonance spectroscopy

PDA - Potato dextrose agar

PDB - Potato dextrose broth

ppm - parts per million

psi - pounds per square inch

RP - Reverse primer

rpm - Revolutions per minute

RT - Room temperature

Sec - Seconds

SD - Standard deviation

SWM - Sea water media

SAR - Structure activity relationship

SEM - Scanning Electron Microscopy

sp. - Species

TEM - Transmission Electron Microscopy

TLC - Thin layer chromatography

U - Enzyme Unit

UV - Ultraviolet

UV-Vis - UV-Visible

v/v - Volume by volume

UPLC - Ultra-high pressure liquid chromatography

w/v - Weight by volume

w/w - Weight by weight

α - alpha

Page 17: Isolation, Characterization and Cosmeceutical Application

xvi

Abstract

Natural products and their derivatives have historically been an extremely valuable

source for new entities in agriculture, pharmaceuticals and cosmeceuticals, to deliver

a unique variety of structural templates for product development. Historically,

terrestrial plants and microorganisms such as fungi and bacteria were the most

important sources for biologically active natural products. The development of plant

based products has some unique problems associated with the process, which can be

best described as the "problem of supply" due to factors such as deforestation,

environmental pollution, global warming and inability to synthesize due to structural

complexity. The rediscovery of large numbers of previously reported compounds

from terrestrial microbes contributes to the difficulties in discovering new structures.

These problems necessitate the continuous search for novel and alternative bio

resources, particularly those that can be produced at scale such as microorganisms.

Since the middle of the last century, marine species and microorganisms have been

explored as sources for the new chemical entity (NCEs). An advance in isolation and

structure determination methods and equipment over the last thirty years has seen a

rapid increase in the number of new structures identified. Nevertheless, marine

chemical ecology and biodiversity are still relatively unexplored, with respect to the

discovery of natural products, compared with terrestrial sources. Some

researchers have specifically compared natural products from terrestrial and marine

sources. They found that compounds from marine organisms generally exhibited a

higher chemical novelty. In this Ph.D. the research aimed to isolate and characterize

marine fungal secondary metabolite and evaluating their potential use in future

cosmetic skin care products.

The study begins with the collection of marine samples for the isolation of marine

fungi. A range of marine samples were collected from diverse and unexplored marine

habitats in India, which included beaches of Indian west coast and Andaman Island.

All collected samples were subjected to isolation of marine fungi using a variety of

techniques and culture conditions, as described in chapter 2. Seventy fungal strains

were isolated from 60 marine samples. Among these 70 only 35 morphologically

different stains were used in this research work. These 35 strains were analyzed for

their optimum growth condition toward maximizing the production of secondary

Page 18: Isolation, Characterization and Cosmeceutical Application

Abstract

xvii

metabolites using, by varying media conditions (salinity, nutrient strength, etc.). Of

the result, full strength PDA media in artificial sea water was found to be best for

90% of the fungal strains, in terms of their growth and production of secondary

metabolites.

All 35 strains were subjected to screening for antibacterial activity against eight

human pathogenic bacteria, including Escherichia coli (MTCC-443), Pseudomonas

aeruginosa (MTCC-424), Staphylococcus aureus (MTCC-96), Staphylococcus

epidermidis (MTCC-3615), Micrococcus luteus (MTCC-106), Bacillus subtilis

(MTCC-121), Bacillus megaterium (MTCC 1684) and Propionibacterium acnes

(MTCC-1951). The crude extracts, prepared from 20 day fungal fermentation broth,

were evaluated using agar well-diffusion and microdilution assays. Eight fungi were

found to be most active and successfully classified as Simplicillium lamellicola (D 5),

Emericellopsis minima (D 6), Leptosphaerulina sp. (AN 7), Penicillium citrinum

(AN 10), Aspergillus aculeatus (AN 11), P. chrysogenum (AN 12), A. oryzae (G 3),

A. sydowii (G 10). The relevance of this screening was also confirmed by SEM

analysis of the targeted bacteria before and after treatment with crude fungal extracts.

Results show that the presence of active molecules in crude extracts causes damage

to bacterial cell membranes. Marine isolate S. lamellicola (D 5) was the most active

extract against both Gram-negative and Gram-positive bacteria, with a MIC value of

1mg/ml. This promising strain was further exploited for the bioassay-guided

fractionation and isolation of bioactive compounds.

The aim of the next part of our investigation was the screening of marine fungi to

search for potential tyrosinase inhibitors and antioxidants to control

hyperpigmentation and photo aging of the skin. Fungal crude extracts were subjected

for MBTH and DPPH radical scavenging assays for depigmentation and antioxidant

potential, respectively. “Fungus P 2” was shown to be related to Talaromyces

stipitatus and “Fungus D 4” was grouped with Aspergillus terreus. The PDA extracts

of P2 and D4 showed antityrosinase activity (45%, 43%, respectively) at the lowest

concentration 0.125 mg/mL. They also exhibited 94%, 97%, antioxidant activity,

respectively, at a concentration of 0.500 mg/mL. Due to their bioactivity, these two

fungi were selected for the future study. This is the first report of antityrosinase

activity from T. stipitatus.

Page 19: Isolation, Characterization and Cosmeceutical Application

Abstract

xviii

The final research aim of this study was the isolation and characterization of active

antibacterial molecules present in the crude extract of marine fungi S. lamellicola (D

5). Bioassay guided isolation was performed using TLC direct bioautography against

S. epidermidis. One bioactive compound, 5, 6-dihydro-6-pentyl-5, 6-dihydropyran-2-

one (1), was isolated from the crude extract by chromatographic methods and

characterized using LC-MS/MS analysis. Two additional compounds were

tentatively identified as simplifungin (2) and a new derivative of halymecin G (3), by

comparing LC-MS/MS data with that of reported metabolites from Simplicillium sp.

The antibacterial activity of compound (1) has not previously been reported.

Through this work, we have identified a marine fungal strain S. lamellicola (D 5) that

is an efficient producer of antibacterial compounds against S. epidermidis. We also

found that the marine fungi P 2 (Talaromyces stipitatus) and D 4 (Aspergillus

terreus) are the potent producer of bioactive metabolites with antityrosinase and

antioxidants activity.

Page 20: Isolation, Characterization and Cosmeceutical Application

1

Chapter 1

Introduction and literature review

Page 21: Isolation, Characterization and Cosmeceutical Application

Chapter 1

2

1. Introduction

Nature provides a diversity of pharmacologically active biomolecules. These natural

biomolecules are important for the development of novel pharmaceuticals as well as

cosmeceuticals. Between 1989-1995 around 60% of Food and Drug Administration

(FDA) approved drugs and pre-NDA (New Drug Application) candidates were

obtained from the natural environment (1). Natural products are obtained from both

terrestrial and marine environments. Approximately 270,000 natural products have

been discovered and some of them are in the market, either as medical drugs or in

clinical use (2, 3).

1.1. Marine natural products

The marine ecosystem covers about 70% of the earth surface and is extraordinarily

rich in biological diversity, particularly in tropical environments. According to the

Global Biodiversity Assessment by the United Nations Environment Programme,

oceans consist of 178,000 marine species across 34 phyla. Marine organisms

comprise approximately half of the total biodiversity on earth and have shown to

produce novel biomolecules (4, 5). In the past 50 years, exploration of marine

bioresources for their unique natural products has been an important area of research.

Of the estimated 270,000 known natural products, 30,000 compounds have been

obtained from marine organisms (Blunt et al., 2015). Out of these, 9 are approved as

medical drugs and 12 are undergoing clinical trials (Table 1.1) (6).

Table 1.1: Marine natural products that are FDA-approved agents or used in clinical trial (6)

Clinical

Status

Compound

Name

Source

Organism

Structural class Molecular

Target

Disease Area

FDA

approved

Cytarabine (Ara-

C)

Sponge Nucleoside DNA

polymerase

Cancer

FDA

approved

Vidarabine (Ara-

A)

Sponge Nucleoside Viral DNA

polymerase I

Antiviral

FDA

approved

Ziconotide Cone Snail Cysteine Knot

Peptide

N-type

Ca2+

channel

Pain

FDA

approved

Eribulin

Mesylate

(E7389)

Sponge Complex

Polyketide

Microtubules Cancer

FDA

approved

Omega-3-acid

ethyl esters

Fish Omega-3 fatty

acids

Triglyceride-

synthesizing

enzymes

Hypertriglyceridemia

Page 22: Isolation, Characterization and Cosmeceutical Application

Chapter 1

3

FDA

approved

Trabectedin (ET-

743) (EU

registered only)

Tunicate NRPS-derived

Alkaloid

Minor groove

of DNA

Cancer

FDA

approved

Brentuximab

vedotin (SGN-

35)

Mollusk Antibody drug

conjugate (MM

auristatin E) –

Linear

NRPS/PKS

CD30 and

microtubules

Cancer

Phase III Plitidepsin

(Aplidine)

Tunicate Cyclic

Depsipeptide

Rac1 and JNK

activation

Cancer

Phase II DMXBA (GTS-

21)

Worm Alkaloid α7 nicotinic

acetylcholine

receptor

Cognition,

Schizophrenia

Phase III Plinabulin (NPI

2358)

Fungus Diketopiperazine Microtubules

and JNK stress

protein

Cancer

Phase II Elisidepsin Mollusk Cyclic

Depsipeptide

Plasma

membrane

fluidity

Cancer

Phase II Zalypsis

(PM00104)

Nudibranch Alkaloid DNA binding Cancer

Phase II Glembatumumab

vedotin (CDX-

011)

Mollusk Antibody drug

conjugate (MM

auristatin E) –

Linear

NRPS/PKS

Glycoprotein

NMB &

microtubules

Cancer

Phase I Marizomib

(Salinosporamide

A; NPI-0052)

Bacterium NRPS with β-

lactone & γ-

lactam

20S

proteasome

Cancer

Phase I Trabectedin

analog

(PM01183)

Tunicate NRPS Alkaloid Minor groove

of

DNA<comma>

Nucleotide

Excision

Repair

Cancer

Phase I SGN-75 Mollusk Antibody drug

conjugate (MM

auristatin F) –

Linear

NRPS/PKS

CD70 and

microtubules

Cancer

Phase I ASG-5ME Mollusk Antibody drug

conjugate (MM

auristatin E) -

linear

NRPS/PKS

ASG-5 and

microtubules

Cancer

Phase I Hemiasterlin

derivative

(E7974)

Sponge Modified linear

tripeptide

(NRPS-PKS)

Microtubules Cancer

Phase I Bryostatin 1 Bryozoan Polyketide Protein kinase

C

Cancer, Alzheimer's

Phase I Pseudopterosins Soft Coral Diterpene

glycoside

Eicosanoid

metabolism

Wound healing

Page 23: Isolation, Characterization and Cosmeceutical Application

Chapter 1

4

An extreme physical and chemical condition in the marine environment create stress

that contributes to the production of a unique variety of biomolecules by marine

organisms (7). Sponges, corals and other marine invertebrates have been an ongoing

source of novel chemical structures with many of these compounds exhibiting

interesting biological activities. However, the applicability of many promising

substances was hampered due to the presence of these organisms in relatively low

amounts and the complex structures making chemical synthesis at scale difficult.

Low recovery rates (less than 1 gm of substances such as halichondrin, ecteinascidin

or bryostatin obtained from a ton of sponges, ascidia or bryozoa, respectively) as well

as problems associated with mariculture of most marine macroorganisms made it

extremely difficult to produce substances in amounts sufficient for further studies (8,

9). Thus, marine microbial bioresources such as cyanobacteria, fungi and diverse

groups of marine eubacteria were of interest since many of these organisms can be

produced through fermentation at large scale. In fact, some compounds previously

isolated from macro-organisms, such as sponges and tunicates, were actually

metabolic products of associated microbes (10).

Approximately 3000 natural products have been identified from marine micro-

organisms by the end of 2008 (11). Some marine bacteria and fungi have provided

promising new lead structures for drug discovery (12, 13). An important example is

the discovery of Cephalosporin C (a non-ribosomal peptide secondary metabolite)

from the marine fungi Cephalosporium sp. (14). Over the past decade, more than

10,000 marine natural products have been isolated from marine organisms including

marine fungi (2, 7). During this period, approximately 1000 new natural products

have been isolated from marine fungi, including terpenoids, alkaloids, shikimate-

derived metabolites, lipids and most importantly polyketides and non-ribosomal

peptides (NRPs) (14). These compounds have a potential as pharmaceuticals,

nutraceuticals and cosmeceuticals.

Page 24: Isolation, Characterization and Cosmeceutical Application

Chapter 1

5

1.2. Natural products from marine fungi

Terrestrial fungi have been more widely investigated for bioactive secondary

compound production than marine fungi. Marine fungi are more difficult to collect

and grow and sometime require high salt levels, causing fermentor corrosion. These

fungi are divided into two groups on the basis of their ability to grow in marine

conditions, as obligate and facultative marine fungi (15). Obligate marine fungi grow

fast and sporulate exclusively in a marine or estuarine habitat, while facultative

marine fungi are generally obtained from fresh water or terrestrial environment that

can grow (and possibly sporulate) in the marine environment (15). Marine fungi are

mostly found in algae, corals and detritus of marine macrophytes. Over 1500 species

of marine fungi including about 530 species of obligate marine fungi are known. At

present, it is difficult to ascertain the obligate or facultative nature of fungi and

therefore, a more general expression “marine-derived fungi” is used (16). Durieu and

Montagne (1869) discovered the first obligate marine fungus on the rhizomes of the

sea grass, Posidonia oceanica. The first facultative marine fungus, Phaeosphaeria

typharum, was identified by Desmazieres in 1849 (15).

Marine fungi are major decomposers of woody and herbaceous substrates in marine

ecosystems and they also degrade dead animal (17). A broad variety of substrates are

available in marine and estuarine environments for fungal growth. These include

seaweeds, decaying leaves, mangroves, dead animals, algae and shells of various

mollusks (14). Most of these fungi grow on lignocellulosic material in the coastal as

well as deep sea environments (18). Coral reefs are also a rich source of marine fungi

that produce novel secondary metabolites due to the presence of other biotic

components present in the reefs (19). Till date, 3047 species of fungi have been

reported from aquatic habitats around the world (Table 1.2). Worldwide, nearly

100000 fungal species are known and about 27500 species are reported in India. The

higher filamentous marine fungi include 530 species in 321 genera, which includes

Ascomycota (424 sp. 251 genera), Mitosporic fungi (94 sp. 61 genera) and

Basidiomycota (12 sp. 9 genera) (20, 21).

Page 25: Isolation, Characterization and Cosmeceutical Application

Chapter 1

6

Table 1.2: Fungi from aquatic habitats from around the world Adapted from (22)

Marine fungi may have developed specific metabolic pathways that are not seen in

terrestrial fungi (23, 24). Marine mangroves and algicolous fungi are significant

sources of new bioactive compounds (16). The production of secondary metabolites

seems highly dependent on the culture conditions and the origin of strains (25). The

use of seawater for isolation and growth of marine-derived fungal species can

enhance the recovery of fungi that yield secondary metabolites, as compared to the

use of media prepared in distilled water (26-30). To produce these metabolites and to

maximize the potential chemical diversity, they need to be grown in various nutrient

limited media by varying carbon and nitrogen concentration.

Fungi can be cultivated for the isolation of a number of metabolites such as

glucoamlyase, amylase, α-glucosidase, and proteases. For example, Penicillium sp.

grown under limited carbon has shown to produce penicillins, whereas same fungi

grown under phosphorus and nitrogen limitation produce cephalosphorin,

vancomycin, and carbapenems (31). It is impossible to estimate how many marine

derived fungi have been screened so far for bioactive compounds. But the number of

new bioactive compounds reported from marine fungi has increased steadily over the

years: 1 (1981), 100 (2002), 272 (2004) with l690 new bioactive compounds reported

from 2006 – 2010 (14,32,33). A broad array of pharmacological activities, ranging

Taxonomic group Number of species

Chytridiomycota 576

Freshwater meiosporic ascomycota 450

Mangrove meiosporic ascomycota 612

Marine meiosporic and mitosporic ascomycota 465

Indoldian meiosporic fungi 290

Aeroaquatic meiosporic fungi 90

Miscellaneous meiosporic fungi 405

Oomycota (Saprolegniales) 138

Basidiomycota from freshwater habitats 11

Basidiomycota from brackish and marine habitats 10

Total number 3047

Page 26: Isolation, Characterization and Cosmeceutical Application

Chapter 1

7

from antimicrobial to anti-cancerous has been found in fungal metabolites (14),

although only a few marine fungal derived drugs are currently in the market (34).

1.2.1. Rate limiting factors for secondary metabolites biosynthesis in marine fungi

Secondary metabolites are synthesized mainly under stress conditions due to either

nutrition limitation or excess of carbon sources. The five main sources for the

synthesis of secondary metabolites are (1) amino acids, (2) shikimic acid pathway,

(3) polyketide biosynthesis pathway from acetyl-CoA, (4) mevalonic acid pathway

from acetyl-CoA (5) polysaccharides and peptidopolysaccharides. The production of

secondary metabolites by marine fungi varies due to several factors such as media

composition, salt concentration and other fermentation processes. The effect of sea

water concentration on hyphal growth and anti-microbial metabolite production was

studied in various marine fungal strains and was found to increase the hyphal as well

as metabolite production with increased sea water concentration (26, 27).

The antibiotic lipoxazolidinones produced by the marine fungal strain NPS8920 and

levels increase in natural sea water based media (29). Miao et al., (30), also estimated

the effect of salinity and nutrition on the growth and bioactivity of marine fungi

Arthrinium c.f. saccharicola. This study concluded that higher salt concentration

decreased fungal growth but increased bioactivity. Also, the higher concentration of

peptone and malt extract in growth media enhances fungal growth but decreases

antibacterial activity, whereas the opposite occurred at higher glucose concentrations.

Wang et al., (28), studied secondary metabolites production in the marine fungi,

Spicaria elegans at 3% and 10% salinity. Five new compounds were produced at

10% salinity, which was absent at 3% salinity. Thus it can be concluded that

regulation of physicochemical conditions is important for optimizing secondary

metabolite production in marine fungi.

1.3. Marine cosmeceuticals

The word cosmeceutical has been derived from blending of the terms „cosmetic‟ and

„pharmaceuticals‟ by Abbert Kligman in 1984 (35). Cosmeceuticals are topical or

oral cosmetic pharmaceutical hybrids intended to enhance beauty through bioactive

ingredient having a drug like benefits (36, 37). Therefore cosmetics have been placed

between non-prescribed and prescribed products (38). Cosmetics are defined as

Page 27: Isolation, Characterization and Cosmeceutical Application

Chapter 1

8

substance intended to be applied to the human body for cleaning, beautifying,

promoting attractiveness without affecting its structure and function (Figure 1.1). It

is anticipated that the global cosmeceuticals market will reach US$ 61 Billion by

2020. (http://www.researchandmarkets.com/research/246mph/global)

Figure 1.1: Cosmetics and their relative branches

1.3.1. Current status of cosmetics-cosmeceuticals

A variety of lifestyle and environmental factors cause various cosmetics and

dermatological problems (37). Natural cosmeceutical products that are safe and

efficacious are important for overcoming these problems. Plant derived ingredients

have some limitations because plants can contain toxic metabolites, grow too slow or

composition varies by seasonal changes (39). Marine flora and fauna produce

chemically different biomolecules in comparison to terrestrial sources and so are of

interest, particularly where organisms can be grown in large quantities, such as

macro- and microalgae. Examples include the Greek company Apivita that is using

sea fennel in sun care products, whilst Italy-based Lacote has a comprehensive range

of anti-cellulite skin care products formulated with Guam seaweed. Sea algae, rich in

vitamins and minerals, are becoming a common source of anti-ageing actives. Other

brands are using sea minerals in their formulations, looking to emulate the success of

Dead Sea mineral-based products.

Page 28: Isolation, Characterization and Cosmeceutical Application

Chapter 1

9

The popularity of marine ingredients is leading to concerns that large-scale sourcing,

or non-sustainable production methods, could disrupt marine ecosystems already

under strain. Marine microorganisms can be grown outside of the ocean in fermenters

and so are sustainable. Many novel bioactive molecules have been isolated from

marine plants, animals and microbes for cosmeceutical applications, including

phlorotannins, polysaccharides, carotenoid pigments, collagen, chitooligosaccharide

(COS) derivatives, enzymes, peptides and other natural materials. For example,

Phlorotanins from marine algae Ecklonia cava have anti-wrinkle (40) and anti

tyrosinase activity (41). Some of these biomolecules are produced commercially by

culturing specific marine organism which is used in cosmetics e.g. Myrothenones A

and B (Cyclopentenone) from Myrothecium sp. (marine fungi) and Homothallin-II

from Trichoderma viride H1-7 have skin whitening activity (42, 43). Moreover,

various marine natural products including nutritional supplement ingredients are

being developed into cosmeceutical (Table 1.3).

Table 1.3: Examples of some cosmeceutical ingredients from marine sources Adapted

and modified from (37)

Cosmetic Ingredients Marine Resources Applications/Activities

Mussel glycogen Sepia subaculeate Give smoothness brightness and luster

to skin

Aluminum silicate Sea mud Anticaking agent, Anti- psoriasis

Shell powder Oysters Skin whitening, scrubs

Turtle oil Chelonia mydas Smoothing and moisturizing agent

Phycoerythrin pigments

protein

Red algae Pigment

Proteoglycans Brown algae Boost skin collagen , improve skin

density

Omega-3-fatty acids Algae, animals Tissue repair, increase skin collagen

production

Peptide Sea weeds, sea food By-

product

Increase skin collagen production

Phytoplankton Sea animals Cellular regeneration

Collagen Marine fish Anti-wrinkle, sunscreen

However, the potential of secondary metabolites from marine fungi as a

cosmeceutical ingredient has only been partially evaluated. Only a few compounds

have been isolated and utilized as a cosmetic ingredient from marine fungi.

Therefore, there is considerable potential in screening marine fungal isolates and

their secondary metabolites for their cosmeceutical efficacy.

Page 29: Isolation, Characterization and Cosmeceutical Application

Chapter 1

10

1.4. Cosmeceuticals from marine fungi

Skin is the largest organ in our body and it plays an extremely important role in

protecting our body from external environmental stress and pathogens. Skin

cosmeceuticals were developed after research on common skin problems like

hyperpigmentation, skin cancer, skin microbial infections, wound healing, and

wrinkles associated with sun damage and aging (Figure 1.2).

Figure 1.2: Worldwide major skin problems and their probable solutions

1.4.1. Marine fungi for prevention of skin aging and cancer

According to Euromonitor International, anti-aging skin care is a large and dynamic

business, covering 22% of the global skin care market, worth US$ 66 billion in 2007

and is supposed to reach US$ 131 billion by 2019 (44, 45). The United States

population of individuals over the age of 65 represented 13% in 2000 and it is

expected to increase to 20% by 2030 (46). This data indicates an upcoming

requirement for increasing efforts to prevent aging, including developing safe and

effective cosmeceuticals.

Aging is a complex, multifactorial biological process that occurs in all living

creatures at variable rates. Various surgical and topical methods have been applied to

alter the aging process (47). There are two types of skin aging processes, intrinsic

and extrinsic. The intrinsic or innate aging refers to the natural programmed cellular

aging. It is characterized by a decrease in collagen synthesis, degeneration of elastic

fiber networks, and loss of hydration. On the other hand, extrinsic aging (premature

cutaneous aging) occurs as the result of external factors such as smoking, excessive

Page 30: Isolation, Characterization and Cosmeceutical Application

Chapter 1

11

alcohol consumption, poor nutrition, pollution, and particularly solar exposure. Solar

exposure mostly causes skin wrinkling and undesired pigmentation. Extrinsic aging,

also known as photo aging, has been studied extensively (48). Skin aging is governed

by intrinsic genetic factors and mediated by extrinsic influences. Telomere shortening

and metabolic oxidative damage generates reactive oxygen species (ROS), play a

critical role in the aging process. Healthy telomeres are essential for a long life of

cells but it shortens with aging. Telomerase (eukaryotic ribonucleoprotein (RNP)

complex, contains an essential RNA and a protein reverse transcriptase subunit) is an

enzyme which is responsible for maintaining the length of telomeres by an addition

of guanine rich repetitive sequences (49). Thus, regulating telomerase activity delay

the aging process.

Current research has proven that telomeres and telomerase have a strong correlation

to aging and cancer (50, 51). Telomerase (eukaryotic ribonucleoprotein, RNP)

maintains telomere length stability in almost all cancer cells. Until now there has

been no report on any topical skin care product, systemic drugs, or any other

treatment options that can target telomerase for aging and cancer therapeutic

approaches (inhibit telomerase with the intention of imparting anti-proliferative and

apoptosis inducing effects (52). Only a few biomolecules have been isolated from

various terrestrial as well as marine sources as telomerase inhibitors from marine

sources. Example: Dictyodendrins A–E, five new alkaloids were isolated from the

marine sponge Dictyodendrilla verongiformis, which inhibited telomerase

completely at a concentration of 50µg/ml (53). Marine sponge Axinella infundibula

were the source of Axinelloside A (highly sulfated lipopolysaccharide), Meridine and

ascididemin. All of these compounds strongly inhibit human telomerase at IC50

values of 2.0 µg/ml, 11 and ≥80µl respectively (54, 55) (Figure 1.3). Until now no

such activities has been reported with marine fungal secondary metabolites. Hence

exploring marine fungi for such targets may provide new molecules to treat these

problems.

Page 31: Isolation, Characterization and Cosmeceutical Application

Chapter 1

12

Figure 1.3: Telomerase inhibitors from marine sources

1.4.2. Marine fungi for skin whitening

Human skin color ranges from the darkest brown to the lightest pinkish-white due to

the presence of melanin. Melanin is the major skin pigment that protects skin from

harmful UV radiation. Longer exposure to ultraviolet (UV) radiation increases skin

melanogenesis and usually results in darkening of the skin. However, it may also

occasionally lead to the development of hyperpigmentation.

The biosynthesis pathway for melanin formation was first elucidated by Raper in

1928 and later modified by Cooksey in 1997. Tyrosinase (EC 1.14.18.1), a copper-

containing glycoprotein, is located in the membrane of melanosome and produced

only by melanocytic (56). Melanogenesis takes place in the melanosomes by three

specific enzymes namely, tyrosinase, tyrosinase-related protein (TRP)-1, and TRP-2.

There are two types of melanin which are synthesized within melanosomes:

Page 32: Isolation, Characterization and Cosmeceutical Application

Chapter 1

13

eumelanin (a dark brown-black insoluble polymer) and pheomelanin (light red-

yellow sulphur-containing polymer). L-tyrosine amino acid is the building stone

which acts as a substrate for the enzyme and is generally transported into the

melanosome by facilitated diffusion. The two proteins TRP-1 and TRP-2 are

structurally related to tyrosinase and share approximately 40% amino acid homology.

TRP-1 increases the ratio of eumelanin to pheomelanin and hence increases

tyrosinase stability (57, 58). Tyrosinase, catalyzes the first two steps of melanin

synthesis, that is, hydroxylation of L-tyrosine to dopaquinone that is subsequently

converted into L-dihydroxyphenylalanine (L-DOPA) through auto-oxidation

followed by oxidation of DOPA is converted to dopaquinone (59, 60). The remaining

steps of the melanogenic pathway are shown in Figure 1.4 which includes the

synthesis of eumelanin and pheomelanin.

Tyrosinase inhibitors have been proposed to be clinically useful for the treatment of

some dermatological diseases associated with melanin synthesis like

hyperpigmentation, melasma, cafe´aulait spot and solar lentigo, and are important in

cosmetics applications such as skin lightening (61). A large number of in vitro

tyrosinase inhibitors have been found, although only a few have shown to exhibit any

significant effects in clinical trials (62, 63).

Figure 1.4: Synthesis of melanin (62)

Page 33: Isolation, Characterization and Cosmeceutical Application

Chapter 1

14

Table 1.4 and Figure 1.5 describe in vitro tyrosinase inhibitors from marine fungi.

Two new 3-amino-5-ethenylcyclopentenones, myrothenones A and B have been

isolated from fungus of genus of Myrothecium. Only myrothenones A exhibited

tyrosinase inhibitory activity, with an IC50 value of 0.8 muM. This compound is more

active than kojic acid (IC50 value 7.7 muM)(43), which is currently used in various

skin whitening products. The marine fungus Botrytis sp. was the source of a new α-

Pyrone derivative, 6-[(E)-Hept-1-enyl]-α-pyrone. This compound also exhibited

greater tyrosinase inhibitory activity than kojic acid (IC50 value 4.5, 15.5 μM

respectively)(64). A competitive inhibitor of mushroom tyrosinase homothallin-II

have been isolated from marine derive T. viride (42). Two new sesquiterpenes, 1β,

5α, 6α, 14-tetraacetoxy-9α-benzoyloxy-7β H-eudesman-2β, 11-diol and 4α, 5α-

diacetoxy-9α-benzoyloxy-7βH-eudesman-1β, 2β, 11, 14-tetraol, were produced as

stress metabolites in the cultured mycelia of Pestalotiopsis sp. Z233 isolated from the

algae Sargassum horneri. These compounds showed tyrosinase inhibitory activities

with an IC50 value of 14.8 μM and 22.3 μM, respectively(65).

Table 1.4: Tyrosinase inhibitors from marine fungi

Sr. No. Source Biomolecule Cosmetic

activity

References

1. Myrothecium sp.

(Marine fungi)

Myrothenones A and B

(Cyclopentenone)

Tyrosinase

Inhibitory

(43)

2. Botrytis sp.

(Marine fungi)

6-[(E)-Hept-1-enyl]-α-pyrone

(α-Pyrone Derivative)

Tyrosinase

Inhibitory

(64)

3. T. viride H1-7

(Marine fungi)

Homothallin-II Tyrosinase

Inhibitory

(42)

4. Pestalotiopsis sp.

Z233 (Fungi)

1β,5α,6α,14-tetraacetoxy-9α-benzoyloxy-

7β H-eudesman-2β 11-diol and

4α,5α-diacetoxy-9α-benzoyloxy-7βH-

eudesman-1β,2β,11, 14-tetraol

Tyrosinase

Inhibitory

(65)

Page 34: Isolation, Characterization and Cosmeceutical Application

Chapter 1

15

HO

HN

O

OAbs

OH

O

NH

H

Myrothenone BMyrothenone A

OO

6-[(E)-Hept-1-enyl]-a-pyrone

HO

O

N

Homothallin II

O

O

O

HO

OO

OH

O

O

OO

O

OH

HO

OOH

OH

O

O

4a,5a-diacetoxy-9a-benzoyloxy-7ßH-eudesman-1ß,2ß,11, 14-tetraol

1ß,5a,6a,14-tetraacetoxy-9a-benzoyloxy-7ß H-eudesman-2ß 11-diol

Figure 1.5: Structures of tyrosinase inhibitors from marine fungi

1.4.3. Marine fungi for acne vulgaris

Acne vulgaris (commonly known as acne or pimples) is the most common skin

disorder caused by Propionibacterium acnes. It affects approximately 50 million

people in US, and many more throughout the world. Globally more than 80% of the

population suffers from acne at some stage in their life. Acne can be extremely

painful and cause lasting marks or scars as well as lead to psychosocial suffering.

Economically, it is estimated that US consumers spend more than 1.2 billion dollars

each year for the treatment of acne (66).

The prevalence of skin colonization by antibiotic-resistant P. acnes in acne patients

over a 10-year period (67) showed that the proportion of patients with strains

resistant to one or more commonly used anti-acne antibiotics is increasing.

Therefore, there is a need to discover new biomolecules against P. acnes. Recently,

Sargafuran obtained from marine brown algae, Sargassum macrocarpum has shown

potent anti-acne activity (68). Two unusual pyridones, trichodin A and trichodin B,

were extracted from mycelia and culture broth of the marine fungus, Trichoderma sp.

strain MF106 isolated from the Greenland Seas (Figure 1.6). The trichodin A

showed antibiotic activities against the clinically relevant microorganism,

Staphylococcus epidermidis, with IC50 values of 24 μM and 4 μM, respectively (69).

Page 35: Isolation, Characterization and Cosmeceutical Application

Chapter 1

16

NH

ORO

O

H

H

Trichodin A R=H

Trichodin B R=O

OHHO

HO

Figure 1.6: Structure of Trichodin A-B

2. Conclusion

Marine research has and will continue to offer novel marine-based lead compounds

for industrial applications. However, the immense microbial diversity of the marine

environment is still only partially explored. Marine fungi are rich-sources of

structurally diverse and pharmacologically active metabolites, with great industrial

potential and accessibility, and thus they have attracted attention for health and

cosmetic applications. The above literature presents research on marine fungal

secondary metabolites and their application to skin aging, as well as for

depigmentation and antimicrobial applications in the cosmetic industry. However, the

use of fungal secondary metabolites for cosmeceutical application remains relatively

unexplored, particularly for marine fungi.

Page 36: Isolation, Characterization and Cosmeceutical Application

Chapter 1

17

3. Aim of this Research

Natural products are important sources of drug and lead to drug discovery in a wide

variety of therapeutic indications. The primary aim of this study was to investigate

Indian west coast and Islands as a possible source for novel marine fungal strains that

produce secondary metabolites with cosmeceutical potential.

My research objectives to established suitable methods for isolating marine fungi

from various marine samples by plating methods and different culture conditions,

and to establish artificial marine liquid culture media for fungal fermentation.

Objectives for this project also included (a) setting up an assay model for

cosmeceutical activity screening, (b) identification of the taxonomy of isolated active

fungal strains, (c) extraction of secondary metabolites from active fungal culture

broth using bioassay guided isolation methods and (d) identification and structure

elucidation of isolated active pure compounds.

Page 37: Isolation, Characterization and Cosmeceutical Application

18

Chapter 2

Isolation and purification of marine fungi from a

range of marine environment in India

Page 38: Isolation, Characterization and Cosmeceutical Application

Chapter 2

19

2.1. Introduction

The marine ecosystem is one of the most extreme, complex and the largest aquatic

system on earth, which makes it difficult to fully investigate, including accessing its

biodiversity. It includes oceans, intertidal ecology, salt marsh, lagoons, estuaries,

coral reefs, mangroves, deep sea and sea floor. Marine habitats are generally divided

into two types, coastal and open ocean habitats. Most marine life is found in coastal

habitats (70). According to the Primordial Soup Theory, “the origin of life occurred

in a body of water, possibly a pond or ocean, by the combination of chemicals and

some form of energy which gave the building blocks like amino acids”, that may

have led to the evolution of the newer species (71). The marine ecosystem has a vast

variety of organisms that are different in their physiology and adaptations. According

to the Global Biodiversity Assessment by the United Nations Environment Program,

oceans consist of 178,000 marine species in 34 phyla. It is estimated that 102 fungi,

103 bacteria, and 107 viruses are likely to exist in one milliliter of seawater (72).

Marine fungi are extensively distributed in marine environments, are found on a wide

variety of substrata (73) and at varying depths (74). Most of these fungi grow on

lignocellulosic material in the coastal as well as deep sea environment (18). Coral

reefs, decaying seaweeds, sand grains and the east skins of marine invertebrates are

also a rich source of marine fungi (75). However, each substratum supports a specific

mycota, but some species may also be found on other substrata. Marine fungi are an

ecological group of organism and are to be found in all oceans of the world. Some

fungi are restricted to tropical or temperate waters while others are cosmopolitan in

their distribution (Jones, 1988). New species are still being described, especially

from new sea areas and substrata which not previously examined for example

mangroves (14, 21, 76).

In India, the west coast and the Andaman Islands are one of the most important

marine environments for its biogeological characteristics and has been not well

studied with respect to marine fungi. Marine fungi are an important class of marine

organisms that are responsible for recycling of complex organic material (77, 78) and

biodeterioration of materials in the sea (79, 80). In addition to this, they are a very

promising bioresource for discovering natural products that may be of economic

importance (14, 81). In this chapter, we present data on the diversity and occurrence

Page 39: Isolation, Characterization and Cosmeceutical Application

Chapter 2

20

of culturable marine fungi on various substrata (sea grasses, wood and seaweed)

collected at various locations in the west coast and the Andaman Islands.

2.2. Materials and methods

2.2.1. Chemicals used

All analytical grade chemicals were purchased from Fischer Scientific (Mumbai,

India) and used without further purification. Potato dextrose agar (PDA), Malt extract

and Mycological peptone were procured from HiMedia (Mumbai, India) and were

sterilized by autoclaving at 1200C for 15 mins before use.

2.2.2. Study area

The study area covered the Western Coastal Plains that lies between Western Ghats

and the Arabian Sea. The plains begin at Gujarat in the north and end at Kerala in the

south. It also includes the states of Maharashtra, Goa and Karnataka. The rivers that

flow through this region are Narmada, Tapi, Zuari and Mandovi. This vast area has a

variety of niches such as the Gulf of Kutch, Gulf of Khambhat and Salsette Island.

The Gulf of Kutch is an inlet of the Arabian Sea along the west coast of India, in the

state of Gujarat. The maximum depth of Gulf of Kutch is 401 feet (122 m). The first

national marine park of India is also situated in it. There are 42 islands on the

Jamnagar coast in the Marine National Park, most of them surrounded by reefs. The

Andaman Islands form an archipelago in the Bay of Bengal between India, to the

west, and Myanmar, to the north and east.

Sample collection was done from different beaches of the north and south Goa,

which have very limited tourist activities like Arambol, Querim and Agonda beaches,

and also from the marine estuary Chapora River. Samples have been collected from

low tidal areas having high biodiversity. From Dwarka, samples have been collected

from a small pit of sea water near the sea which was rich in marine algae and

sediment. Various marine samples were also collected from Havelock beach in the

Andaman Islands of India (Figure 2.1).

Page 40: Isolation, Characterization and Cosmeceutical Application

Chapter 2

21

Figure 2.1: Sites of sample collection in India (Goa, Gujarat and Andaman Islands)

2.2.3. Collection of samples

Sample collection was done after the rainy season during no moon days and low-tide

periods in November to December, since in the rainy season very high amount of

terrestrial water have been mixed with sea water causing contamination with

terrestrial organisms. Materials from the deep intertidal zone (depth ~5-10 meters)

were sampled. Marine algae, sea sediments/ estuary sediments, sea plants, mangrove

plants (wood, leaves, and roots), dead animals and sea water were collected in sterile

polypropylene bags and screw cap bottles, respectively. The collected samples were

brought to the laboratory for isolation and purification of marine fungi.

Marine Algae: - Algae samples (Caulerpa sp., Halimeda sp., Padina sp.,

Sargassum sp. and unidentified alga) were collected from different sites that

are associated with coral, mangrove and woods along with sea water.

Sea sediments: - Sediments were collected from the inner part of the sea (2-

3 km from beach shore) along with sea water, using a large spatula (12”

length).

Page 41: Isolation, Characterization and Cosmeceutical Application

Chapter 2

22

Sea plants: - Sea plants were collected when they come up during low tide,

in a plastic bottle along with sea water.

Mangrove plants: - Mangrove plant species (Avicenna marina and

Rhizophora sp.) were collected and stored in a sterile zip bag.

Dead animals: - Dead Sea animals like sea mussels and sea shell were

collected sedimentation sites.

Sea Water: - Stratified samples of marine water were collected in pre sterile

screw cap plastic 5 L bottle from six different depths, which varied from 5 to

200 centimetres in depth.

2.2.4. Isolation of marine fungi

The sediment samples were diluted at 10 and 100 fold with autoclaved aged seawater

(Sea water collected from Indian Ocean was stored at room temperature for more

than 5 years and this was provided by Dr. Sandeep Garg from Goa University, India)

and sea water was used directly as inoculum. 100 µl of each sample was spread onto

PDA media plates (39 gm potato dextrose agar in 1,000 ml of naturally aged

seawater, pH 7.5 ± 0.2). The antibiotics, streptomycin and penicillin, at 100 and 50

mg/L concentrations respectively, were added to each agar plate to inhibit the growth

of bacteria. Algal samples were rinsed with sterile aged seawater several times until

sea sand was removed and then plated on media plates. Likewise, plant material and

dead animals were sterilized with 70 % ethanol for 10 seconds to kill the residual

epiphytes and washed with sterile seawater to remove ethanol, and dried with sterile

tissue paper. Subsequently, the samples were cut into pieces using a sterilized knife

and pressed onto the media to inoculate endophytic fungi. Two replicate media plates

were used for each sample. All plates were incubated at room temperature (28 ± 2

°C) for 21 days. After 2 days of incubation, media plates were examined daily for the

presence of developing fungal growth. Distinct fungal colonies on the media plates

were then transferred to new media plates with artificial sea water (ASW) (82),

instead of natural sea water, for further purification and maintenance (83).

Page 42: Isolation, Characterization and Cosmeceutical Application

Chapter 2

23

2.2.5. Growth assessment of marine-derived fungal strains using different media

conditions

Radial growth experiments were performed to assess the growth and development of

isolated marine fungi to investigate the importance of specific nutrients. The

experiments were performed on the following media:

1. Media A (PDA in 100% sea water with pH 7.5 (High nutrient)

2. Media B (Sea water media (SWM), 1.0% glucose, 0.5% peptone, 0.1% yeast

extract and 1.7% agar, final pH of 7.5) (Low nutrient)

The agar plates (90mm) were inoculated at a central insertion point and the colony

diameters were measured after 2, 5, 7, 10, 15 and 20 days of cultivation in an

incubator at 28±2°C. Radial growth of a fungal colony measured by Vernier caliper,

when the growth was not round the average of length and width was taken into the

account. Each experiment was carried out in triplicate.

2.3. Results and Discussion

A total of 70 fungal isolates were isolated from marine samples collected from

various locations on the west coast and Islands of India (Figure 2.2, Table 2.1).

Coral reefs are considered an excellent source for the isolation of marine fungi and

other organisms having bioactivity (75). 28 Fungal strains were isolated from

Dwarka (22.2377N x 68.9673E, Gujarat) and 13 strains from Paldi (23.0121N x

72.5589E, Gujarat). A total of twelve marine fungi were isolated from sediments

collected from havelock beach Andaman Islands (11.7400N x 92.6586E). 17 fungal

strains were isolated from marine samples collected from different beaches in Goa

(28.38N x 72.12E) (Table 2.2, Figure 2.3). However, several of these fungal isolates

did not show any growth on lab prepared artificial media.

Page 43: Isolation, Characterization and Cosmeceutical Application

Chapter 2

24

Table 2.1: Samples collected from different parts of India (21.7679° N, 78.8718° E)

Sr.

No

Name of

the Place

Coordinates of

the Place

Types of

sample

collected

Total

Number

of sample

collected

Conditions/ Date

Goa§, India (28º38' N x 72°12' E)

1. Cacra

Beach,

North Goa

15° 26' 52" N x

73° 50' 19" E

Sea

sediments,

sea weeds,

sea animals,

sea water

10 Two day previous to NMD*

Time:- 2-4 PM Low tide (-0.06),

13/11/2012

2. Arambol

Beach,

North Goa

15° 41' 14" N x

73° 42' 11" E

Sea

sediments,

sea weeds,

sea animals,

sea water

10 One day previous to NMD

Time:- 3-5 PM Low tide (-0.01),

13/11/2012

3. Querim

Beach,

North Goa

15° 42' 57" N x

73° 41' 25" E

Sea

sediments,

sea weeds,

sea animals,

sea water

15 NMD Time:- 4-5 PM

Low tide (-0.12), 14/11/2012

4. Agonda

Beach,

South Goa

15° 2' 41" N x

73° 59' 8" E

Sea

sediments,

sea weeds,

sea animals,

sea water

12 One day after NMD

Time:- 3:30-5 PM Low tide (-

0.11), 14/11/2012

5. Chapora

River, North

Goa

15° 36' 21" N x

73° 44' 26" E

Sediments,

water.

2 Two day after NMD

Time:- 4:30-5:30 PM Low tide (-

0.11), 15/11/2012

Gujarat, India (23º00' N x 72°00' E)

6. Dwarkanath

Temple,

Dwarka

22.23' 77"N x

68.96' 73"E

Sea

sediments,

algae, sea

water

10 One day previous to NMD

Time:- 1-3 PM Low tide (-0.02),

20/12/2012

7. Paldi 23.01' 21"N x

72.55' 89"E

Sea

sediments,

algae, sea

water

5 One day previous to NMD

Time:- 4-5 PM Low tide (-0.02),

20/12/2012

Andaman and Nicobar, India (11.7400N x 92.6586 E)

8. Havelock

island

11.58N x

93.00E

Sea

sediments,

algae, sea

water, wood.

5 One day previous to NMD

Time:- 3-5 PM , 29/12/2012

Note: - * NMD: - No moon day

§:- These beaches are relatively less explored by tourists and less contaminated

Page 44: Isolation, Characterization and Cosmeceutical Application

Chapter 2

25

Figure 2.2: Different locations where sampling was done and material collected

Page 45: Isolation, Characterization and Cosmeceutical Application

Chapter 2

26

Table 2.2: Pure fungal isolates obtained from the samples collected from different

habitats

Sr.

No.

Place Coordinates of the

Place

No of

fungal

isolates

Culturable

fungal

isolates

Culture

conditions

for isolation

A. Dwarka (Gujarat) 22.2377N x 68.9673E 28 10

PDA media

in sea water,

28ºC

B. Paldi (Gujarat) 23.0121N x 72.5589E 13 06

C. Andaman & Nicobar 11.7400N x 92.6586E 12 11

D. Goa 28.38N x 72.12E 17 08

Page 46: Isolation, Characterization and Cosmeceutical Application

Chapter 2

27

Page 47: Isolation, Characterization and Cosmeceutical Application

Chapter 2

28

Page 48: Isolation, Characterization and Cosmeceutical Application

Chapter 2

29

Figure 2.3: Plates showing the diversity of culturable fungal isolates from different

parts of India (D= Dwarka, P= Paldi, AN= Andaman & Nicobar, G=Goa)

Page 49: Isolation, Characterization and Cosmeceutical Application

Chapter 2

30

2.3.2. Effect of nutrient concentration on fungal growth

Mycelial growth of the isolated marine fungi in different nutrient concentrations

(media A and media B) was investigated after incubation at 28°C and the results are

summarized in Table 2.3 and 2.4. The nutrient concentration greatly affected the

growth of fungi. The fungus grew two times faster in high-nutrient media (medium

A) than in low-nutrient media (medium B). The density and appearance of mycelia

was greater in media A, however, sporulation occurred earlier in media B (Figure 4).

Of the 35 fungi isolates, 3 strains grew more abundantly in media B, while the

growth of remaining strains was suppressed. These findings will be utilized in later

work to optimize bioactive metabolite production in these fungi.

Table 2.3: Fungal mycelial growth on medium A

Fungal code Days Radial growth diameter (in mm)

2nd 5th 7th 10th 15th 20th

P2 12 35 65 FPG FPG -

P3/2 GI 15 30 40 60 FPG

P4 10 15 19 30 40 55

P8 24 50 FPG FPG FPG -

P9 GI 30 65 FPG FPG -

P10 16 28 40 50 66 FPG

D2 GI 12 20 29 35 43

D3 00 GI 10 24 40 66

D4 22 55 FPG FPG - -

D5 GI GI 10 20 25 30

D6 13 24 40 50 60 FPG

D8 GI 18 29 40 50 FPG

D15 GI 14 24 40 52 60

D25 25 28 FPG FPG - -

D28 00 22 50 65 FPG -

D34 15 28 55 70 FPG -

AN1 35 FPG FPG FPG FPG -

AN2 00 10 19 25 30 39

AN4 GI 20 35 45 52 70

AN5 10 25 40 50 65 FPG

AN6/1 20 28 40 46 53 FPG

AN6/2 40 FPG FPG FPG FPG -

AN7 GI 22 35 50 70 FPG

AN9 15 22 30 40 56 FPG

AN10 10 20 35 50 65 FPG

AN11 30 60 FPG FPG FPG FPG

AN12 30 FPG FPG FPG FPG FPG

G2 00 GI 40 70 FPG FPG

G3 25 FPG FPG FPG FPG FPG

G4 10 16 29 35 52 FPG

G7 16 40 70 FPG FPG FPG

G8 50 7 FPG FPG FPG FPG

G10 GI 25 45 60 FPG -

Page 50: Isolation, Characterization and Cosmeceutical Application

Chapter 2

31

G14 GI 10 30 45 50 62

G15 GI 10 35 45 49 56

Table 2.4: Fungal mycelial growth on medium B

Fungal code Days Radial growth diameter (in mm)

2nd 5th 7th 10th 15th 20th

P2 12 26 55 FPG FPG FPG

P3/2 GI 13 25 35 55 FPG

P4 9 13 20 28 33 39

P8 14 40 FPG FPG FPG FPG

P9 GI 15 65 FPG FPG FPG

P10 7 20 35 55 65 FPG

D2 GI 10 16 25 32 40

D3 00 GI 30 45 55 FPG

D4 16 35 FPG FPG FPG FPG

D5 GI GI 9 20 25 30

D6 12 25 40 60 67 FPG

D8 GI 18 30 40 60 FPG

D15 GI 10 2 25 40 49

D25 15 40 FPG FPG FPG FPG

D28 00 20 45 65 FPG FPG

D34 12 22 40 50 57 FPG

AN1 24 55 FPG FPG FPG FPG

AN2 GI 9 15 20 28 34

AN4 GI 25 35 55 63 70

AN5 GI 25 36 50 70 FPG

AN6/1 15 20 32 41 50 FPG

AN6/2 33 60 FPG FPG FPG FPG

AN7 GI 20 30 55 70 FPG

AN9 13 22 28 45 50 FPG

AN10 GI 10 30 50 60 FPG

AN11 00 30 FPG FPG FPG FPG

AN12 25 FPG FPG FPG FPG FPG

G2 GI 13 42 50 62 FPG

G3 20 FPG FPG FPG FPG FPG

G4 10 15 19 25 29 35

G7 10 25 70 FPG FPG FPG

G8 258 60 FPG FPG FPG FPG

G10 GI 18 20 30 68 FPG

G14 GI 5 20 30 40 44

Page 51: Isolation, Characterization and Cosmeceutical Application

Chapter 2

32

G15 GI 5 25 25 45 49

Note: GI= Growth initiate, FPG= Full Plate Growth, D= Dwarka, P= Paldi, AN= Andaman &

Nicobar, G=Goa

Figure 2.4: Fungal growth differences with nutrient avability on solid culture medium

Page 52: Isolation, Characterization and Cosmeceutical Application

Chapter 2

33

2.4. Conclusion

70 marine fungi were isolated from various samples collected from the west coast

and Andaman Island of India. Of these, only 35 strains were culturable. Radial

growth experiments on different solid media plate showing a clear difference in

fungal mycelia growth and spore formation. Higher mycelial production was

observed in nutrient rich media and earlier spore formation was observed in nutrient

deficient media.

Page 53: Isolation, Characterization and Cosmeceutical Application

34

Chapter 3

Screening for antimicrobial activity of marine fungi

against bacteria causing skin and wound infections in

humans

Page 54: Isolation, Characterization and Cosmeceutical Application

Chapter 3

35

3.1. Introduction

Millions of people worldwide are affected by infectious diseases caused by bacteria

and fungi (84). Skin and skin structure infections (SSSI) represent a significant

category of infectious disease. Acute bacterial skin and skin-structure infection

(ABSSSI) are among the most prevalent in the world and the 28th most common

infections diagnosis in hospitalized patients (85, 86). According to U.S. Food and

Drug Administration (FDA), ABSSSI can be divided into two categories,

uncomplicated and complicated. Uncomplicated includes simple abscesses,

impetiginous lesions, furuncles, and cellulitis, mainly caused by Staphylococcus

aureus and Streptococcus pyogenes. Many traumatic wound infections can also be

considered uncomplicated when they are the result of common skin colonizers that

have entered through a break in the skin‟s natural defenses. These infections are

typically monomicrobial and caused by Gram positive organisms (87). Complicated

SSSIs characteristically involve deeper soft tissue or require significant surgical

intervention, such as infected ulcers, burns, and major abscesses or a significant

underlying disease state that complicates the response to treatment (88). The

complicated category includes infections either involving superficial infections or

abscesses in an anatomical site, such as the rectal area, where the risk of anaerobic or

Gram-negative pathogen involvement is higher and should be considered

complicated infections (89) (Figure 3.1). Antibiotics are among the most commonly

used drugs. Since the discovery of penicillin by Sir Alexander Fleming in 1928,

antibiotics have protected billions of lives and played an important role in human

history. Unexpectedly, many pathogens have developed resistant towards current

antibiotics and this problem has become more and more serious (90). Additionally,

the revitalization of almost vanished pathogens like Mycobacterium tuberculosis and

the emergence of some new infectious diseases, such as cryptococcal meningitis,

toxoplasmosis, H1N1, Ebola and severe acute respiratory syndrome (SARS) (90, 91).

All these new problems necessitate the continuous search for novel and alternative

antibiotics and antifungals.

Page 55: Isolation, Characterization and Cosmeceutical Application

Chapter 3

36

Figure 3.1: Causative pathogens involves in complicated skin infections (92)

Natural products have a wide range of structural diversity and a history of discovery

as new and novel antibiotics and chemotherapeutic agents. Terrestrial bacteria and

fungi have been wisely screened as sources of valuable bioactive metabolites with

novel structures for antibiotic purposes (93-95). However, the rediscovery of high

numbers of previously reported compounds has significantly diminished the study of

the terrestrial microbial resource (95). Recently, less explored habitats and ecological

niches such as the oceans and extreme conditions are being investigated. Recent

reviews demonstrate that marine fungi have become promising sources of bioactive

compounds (14, 96, 97). Due to unique physiochemical conditions in the marine

environments, marine-derived fungi have produced a variety of novel bioactive

compounds that have a potential for pharmaceutical and industrial uses (98, 99).

However, limited numbers of novel marine fungi have been isolated from marine

habitats as compared to their estimated high biodiversity (100). Research on marine

fungi has been relatively neglected, although the fungi are extremely potent

producers of secondary metabolites and bioactive substances.

In the last decade, many novel bioactive natural products from marine fungi have

been discovered that possess cytotoxic, anticancer, antiviral, antibacterial or

antifungal activities (32, 97). Marine fungal produced a wide range of secondary

metabolites belonging to the alkaloids, macrolides, terpenoids, peptide derivatives

Page 56: Isolation, Characterization and Cosmeceutical Application

Chapter 3

37

and other structure classes (101). These marine fungal derived compounds have

provided new drug candidates to fight the infectious diseases (81). Although the most

famous instance was the discovery of cephalosporins (non-ribosomal peptide) with

cephalosporin C from a marine strain of Acremonium chrysogenum in 1945 by G.

Brotzu (102), there are also more recent promising examples which include, halovir

and its naturally occurring analogs, which are potent inhibitors of Herpes simplex

viruses 1 and 2 from the marine-derived fungus of the genus Scytalidium (103). The

sponge-derived fungus Penicillium chrysogenum provides a new alkaloid

sorbicillactone A, with strong activity against leukemia cells and low cytotoxicity

(104). The marine fungus Pestalotia sp. was the source of the new chlorinated

benzophenone compound pestalone, which showed potent antibiotic activity against

Methicillin-resistant Staphylococcus aureus (MRSA, MIC=37 ng/mL) and

vancomycin-resistant Enterococcus faecium (VREF, MIC=78 ng/mL), revealing its

potential as a new antibiotic (105).

The aim of this study was to evaluate collected marine fungal strains from west coast

and Islands of India for their ability to prevent the growth of causative pathogens

involves in complicated skin infections. All 35 fungal strains that were able to be

grown in media were screened for antibacterial activity and then the most active

isolates were subjected to microscopic analysis and molecular identification using

molecular biological techniques. Scanning electron microscopy (SEM) was also used

to study the possible effects of extracts on targeted bacterial cells.

Page 57: Isolation, Characterization and Cosmeceutical Application

Chapter 3

38

3.2. Materials and methods

3.2.1.. Chemicals used

All analytical grade chemicals were purchased from Fischer Scientific (Mumbai,

India) and were used without further purification. Nutrient agar (NA), Nutrient broth

(NB), Brain heart infusion agar (BHIA), Brain heart infusion broth (BHIB) and

Müller-Hinton agar were procured from HiMedia (Mumbai, India) and were

sterilized by autoclaving at 1200C for 15min at 15psi before use.

3.2.2. Test organisms

3.2.2.1. Marine fungi

35 fungal strains used in this work were isolated from various marine samples

collected from west coast and Islands of India (Chapter 2, Table 2.2 and Figure 2.3 in

Section 2.3.1)

3.2.2.2. Bacterial strains

The following microorganisms were used for antimicrobial screening: Escherichia

coli (MTCC-443), Pseudomonas aeruginosa (MTCC-424), Staphylococcus aureus

(MTCC-96), Staphylococcus epidermidis (MTCC-3615), Micrococcus luteus

(MTCC-106), Bacillus subtilis (MTCC-121), Bacillus megaterium (MTCC 1684)

and Propionibacterium acnes (MTCC-1951). These organisms were procured from

Institute of Microbial Technology (IMTECH) Chandigarh, India.

3.2.2.3. Bacterial inoculum

A loopful of pure colonies of the all tested microorganisms, except P. acnes, was

inoculated into 20 ml nutrient broth and incubated at 37°C for 4 h. P. acnes was

inoculated into 20 ml of brain heart infusion media broth with 1% glucose for 72 h

under anaerobic conditions. Anaerobic condition during media preparation was

maintained as described by Singh et al., 2014 (106). The turbidity of all actively

growing bacterial suspension was adjusted to match the turbidity standard of 0.5

McFarland standard [(1.5 x 108 colony-forming unit, CFU/ml), prepared by mixing

0.5 ml of 1.75% (w/v) barium chloride dihydrate to 99.5 ml of 0.18 M (v/v)]

sulphuric acid with continuous mixing. The bacterial suspension so prepared was

used for testing their sensitivity to the samples under investigation (107).

Page 58: Isolation, Characterization and Cosmeceutical Application

Chapter 3

39

3.2.3. Broth fermentation and extraction

All fungal strains were cultured in 500 ml Erlenmeyer flasks containing 200 ml of

potato dextrose broth (PDB, HiMedia) and malt extract broth (malt extract 17gm/L,

mycological peptone 3gm/L) in (ASW), separately at 28 ± 2°C for 15-20 days under

shaking conditions at 140 rpm. After 20 days fermentation (Initially fungal growth

curve study was performed using ergosterol assay to evaluation optimum growth in

liquid broth), broths were separated from the mycelium by filtration. The supernatant

was mixed with 10% activated Diaion HP-20 resin (Mitsubishi Chemical Co.) and

the mixture was shaken for 30 min on a shaker. Then the mixture was packed in a

glass column, washed with distilled water until the eluent was no longer cloudy and

eluted with 50 ml methanol. Methanol was evaporated using a rotary evaporator

(BUCHI Rota vapor R-200), and crude extract was collected and stored in the cold

(108). The fungal mycelia were also soaked in methanol for 24 hours. The methanol

layer was collected and evaporated to dryness to give the cell methanol extract. The

broth and mycelial extracts were compared on TLC and when they were found same

both the extracts were pooled (Figure 3.2). The dry crude extracts were dissolved in

autoclaved Milli-Q water to prepare stock solutions of 5 mg/ml and kept for

biological testing.

Figure 3.2: Fungal secondary metabolites extraction

Page 59: Isolation, Characterization and Cosmeceutical Application

Chapter 3

40

3.2.4. Antimicrobial assay in vitro

An agar well-diffusion method described by (109) was used to determine the

antimicrobial activity. Mueller Hinton agar no. 2 (HiMedia, India) was prepared and

cooled to 40 - 45ºC and the bacterial inoculum (1.5×108 CFU/mL, 0.5 McFarland)

prepared above was then added aseptically to the molten agar and poured into sterile

petri dishes to give a solid plate. Wells (6 mm diameter) were made in each of these

plates using sterile cork borer. The wells were filled with 50 µl of test compound (5

mg/ml). The plates were first incubated at 4 ºC for 15 min and then at 37ºC for 18-24

h under aerobic and anaerobic conditions, respectively. The antimicrobial spectrum

of the extract was determined for the bacterial species in terms of zone sizes around

each well. The diameters of zones of inhibition (ZOI) produced by the test were

recorded and compared with those produced by positive control antibiotic,

tetracycline (5 mg/mL, Sigma-Aldrich). For negative control, pure solvent

(autoclaved Milli-Q water) were used instead of the extract. The experiment was

performed two times in triplicates to minimize error and to check the reproducibility

of results. The average values were recorded.

The % antimicrobial activity was calculated by applying the expression:

3.2.5. Determination of minimum inhibitory concentration (MIC)

The MIC of active extracts were determined by a microdilution assay in 96-well

microtiter plates according to the broth microdilution guideline of the Clinical and

Laboratory Standards Institute (CLSI)(110). The fresh cultures were prepared at 24 h

and 72 h broth cultures of E. coli, P. aeruginosa, S. aureus, S. epidermidis, M. luteus,

B. subtilis, B. megaterium and P. acnes and OD was set at 0.5 McFarland standard

(1.5×108 CFU/mL) at 600 nm with respective media broth. The bacterial suspension

and the serially diluted (1, 0.5, 0.25, 0.07 mg/mL) fungal crude extracts were added

to 96-well plates to obtain the final volume of 200 µl. The microplates were

incubated at 37˚C with continuous shaking. After 18 h and 48 h (for P. acne), the

optical density at 600 nm was measured with a microplate reader. Tetracycline was

% Inhibition= 100 - (ZOI positive control − ZOI sample) / ZOI positive control ×100

Page 60: Isolation, Characterization and Cosmeceutical Application

Chapter 3

41

used as positive controls. The MIC values were determined at the zero optical density

concentration. All experiments were repeated three times.

3.2.6. Determination of the antibacterial mechanism

Extracts with strong antimicrobial activity were further investigated for their possible

effects on targeted cells by scanning electron microscopic analysis. All test

organisms were grown overnight to mid-logarithmic phase or at a fixed OD of 1.5 at

600 nm. All test organisms in the present study were treated with a fixed

concentration of 2 mg/mL of crude fungal extracts and incubated at 37˚C with

continuous shaking. After 18 h and 48 h (for P. acne) the culture was centrifuged at

5,000 rpm for 5 min; the supernatant was removed and washed three times with

startle water. Test organisms were then fixed in 2.5% glutraldehyde in 25mm

phosphate buffer solution (PBS) for 5-6 hrs at 4°C, then washed with PBS 3 times for

10 min. Then the sample was dehydrated with an ethanol series (30%, 50%, 70%,

90% and 100% ethanol). Samples were dried using a critical point dryer (Emitech

K850, Berkshire, U.K.) were sputtered coated with gold and palladium coating at

120mA for 15 mins using sputter coater (Quorum Technologies SC7620, Berkshire,

U.K). The samples were scanned under SEM (Model EVMA10, ZEISS, Germany) at

TERI-Deakin Nano Biotechnology Centre.

3.2.7. Morphological and molecular characterization of potential marine fungi strain

The bioactive fungal isolates were identified initially by morphological

characterization by scanning electron microscopy (SEM, Model EVMA10, ZEISS,

Germany). Subsequently, they were identified by molecular techniques based on the

analyses of their nuclear ribosomal internal transcribed spacer (ITS) regions.

3.2.7.1. Scanning electron microscopy (SEM) of fungi

Morphological features of selective fungal isolates were observed through SEM (Carl

Zeiss, Oberkochen, Germany). Fungal isolates were cultured on PDA sea water

media and incubated at 280C ± 2 for 4-5 days. After that, fungal growth was taken

and then immersed in a fixative solution (Modified Karnovsky‟s fixative 2.5%

glutaraldehyde- 2.5% paraformaldehyde, 0.05M cacodilate buffer, 0.001M CaCl2) at

pH 7.2, followed by washing with Cacodilate buffer (thrice for 10mins each). Then

post-fixation was done by 1% osmium tetraoxide solution and water for 1hr (111).

Page 61: Isolation, Characterization and Cosmeceutical Application

Chapter 3

42

The samples were then washed with sterile distilled water three times and then

subjected to dehydration in acetone (25%, 50%, 75%, 90% and 100%) for 10 min

each followed by critical point drying (CPD) (Emitech K850, Berkshire, U.K.). The

samples were assembled on double-sided carbon tape placed on aluminum stubs. The

samples were then coated with gold-palladium in a sputter coater (Quorum

Technologies SC7620, Berkshire, U.K.) and viewed in SEM at an accelerating

voltage of 10kV.

3.2.7.2. DNA extraction, PCR amplification and sequencing

Fungi were cultured in potato dextrose broth (PDB) in ASW for one week. Fungal

genomic DNA was performed using the DNeasy Plant Mini Kit (Qiagen) following

manufacturer‟s instruction. Primer pairs ITS1/ITS4 (5‟-TCC GTA GGT GAA CCT

GCG G-3‟/ 5‟-TCC TCC GCT TAT TGA TAT GC-3‟) and

ITS1F/ITS4B(5‟CTTGGTCATTTAGAGGAAGTAA3‟/5‟CAGGAGACTTGTACA

CGGTCCAG-3‟) were used for the PCR reactions (112). The final PCR reaction mix

was of 25 µl reaction volumes containing 2.5 µl of 10x buffer, 1.5 µl of MgCl2 (25

mM), 0.5 µl of dNTP (10 mM), 1 µl of ITS1 primer (5 pm), 1 µl of ITS4 primer (5

pm), 0.2 µl of Taq Polymerase (2.5 U), 3 µl of DNA sample (5 µg/ ml), and

remaining volume was made up using sterile Milli-Q Water. The PCR reaction was

carried out in Veriti 96-well Thermal Cycler (Applied Biosystems, Massachusetts,

U.S.A) with conditions as follows: denaturation for 5 min at 94ᵒC, 33 elongation

cycles (50 s at 95˚C, 50 s at 56˚C, 1 min at 72ᵒC) final extension for 10 min at 72˚C

and last at 4˚C. Negative controls were used to confirm the absence of contamination.

The final products were analyzed by electrophoresis on 1% Agarose (Sigma-Aldrich,

St. Louis, USA). The amplified DNA was cut from the gel using Gel extraction kit

(Qiagen, Germany). The purified PCR-DNA products were sequenced by Xcelrislabs

Ltd., an Indian Biotechnology Company using the same primers. The sequences were

analyzed by Finch TV 1.4.0 (113). The sequences were then compared with the

NCBI GenBank database by the BLASTN program. Phylogenetic relationships were

estimated using MEGA7.0.18 (114).

Page 62: Isolation, Characterization and Cosmeceutical Application

Chapter 3

43

3.3. Results and discussion:

3.3.1. Antimicrobial screening

Organic extracts of fermentation broth and mycelia of the 35 isolated marine fungi

strains were screened for antibacterial activities at 5 mg/mL concentration against

eight human pathogenic bacteria (E. coli, P. aeruginosa, S. aureus, S. epidermidis,

M. luteus, B. subtilis, B. megaterium and P. acnes). As showed in Table 3.1, 18

fungal strains (41.8 %) displayed different levels of antibacterial activities against the

tested pathogenic bacteria. Among those, the malt extracts of fungal strains Dwarka

5, Andaman 12, Goa 10 showed the strongest inhibition (93%, 86% and 83%,

respectively) activity S. aureus. PDA extract of Goa 10 and malt extracts of

Andaman 12 and Goa 3 showed the strongest inhibitory activity against M. luteus,

with 77%, 86%, 75%, respectively. Similarly, the growth of B. subtilis was strongly

inhibited by malt extracts of Dwarka 5 and Andaman 10 and PDA extract of

Andaman 10. PDB extracts of fungal strains Andaman 10 and Andaman 11 showed

79%, 66% inhibition activity, respectively, against B. megaterium. The extract of

Dwarka 5 showed higher inhibitory activity then did the positive control. The Growth

of anaerobic bacteria P. acnes was strongly inhibited by PDA extracts of Dwarka 5

and Goa 10 and malt extract of Andaman 12 (60%, 60% and 70%, respectively).

PDA extracts of Dwarka 5 and Andaman 7 and malt extract of Andaman 10 all

showed higher inhibition then the positive control against S. epidermidis. Growth of

gram negative bacteria P. aeruginosa was effectively inhibited by PDA extracts of

Dwarka 5 and Dwarka 6 (87%, 50%, respectively). Only PDA extract of Andaman

10 showed inhibition against E. coli.

In the present study, we found that crude extracts of some fungi had significant

antibacterial activities. This indicates that different fungi can produce intracellular

bioactive metabolites or secrete extracellular bioactive compounds. In addition, the

results indicated that the fungal extracts exhibited higher inhibition activities to

Gram-positive bacteria, such as P. acnes, S. aureus, B. subtilis and B. megaterium

than to Gram-negative bacteria, which might be a result of different cell wall

compositions of Gram-positive and Gram-negative bacteria. Many diseases are

known to be caused by gram-positive bacteria, such as infections of the respiratory

tract and skin. The gram- positive bacteria can produce exotoxin which can infect

humans. The fungi with higher bioactivity toward gram-positive bacteria are a

Page 63: Isolation, Characterization and Cosmeceutical Application

Chapter 3

44

possible source of new drug leads for development to eventually treat these diseases.

Our results indicate that the marine-derived fungi isolated in this study are a

promising source of natural product candidates for isolation and structural studies.

Dwarka 5, Andaman 7, 10 and 12 were particularly promising and exhibited a broad

range of activity. Subsequent experiments were conducted to determine minimum

inhibitory concentrations and mode of action of selected fungal extracts.

Page 64: Isolation, Characterization and Cosmeceutical Application

Chapter 3

45

Table 3.1: Antibacterial activity of crude fungal extracts against pathogens involves in

complicated skin infections by agar well-diffusion method

Zone of inhibition

No

.

Marin

e

Fungi

Code

Media

Extract

Gram (+) bacteria Gram (-)

bacteria

S.

aureu

s

M.

luteu

s

B.

subtili

s

B.

megaterium

P.

acnes

S.

epidermidi

s

E.

col

i

P.

aeruginos

a

Diameter ( in mm) Inclusive of 6 mm of well

1. P 2 PDA - - 12 12 - - - -

MALT - - - - - - - -

2. P 3/2 PDA - - - - - - - -

MALT - - - - - - - -

3. P 4 PDA 15 - - - - 16 - -

MALT - - - - - - - -

4. P 8 PDA 11 20 - - 15 - -

MALT - - - - - - - -

5. P 9 PDA - - - - - - - -

MALT - 10 - - - - -

6. P 10 PDA - - - - - - - -

MALT - - - - - - - -

7. D 2 PDA - - - - - - - -

MALT - - - - - - - -

8. D 3 PDA - - - - - - - -

MALT - - - - - - - -

9. D 4

PDA - - - - - - - -

MALT - - - - - - - -

10. D 5

PDA 25 20 22 25 30 27 - 35

MALT 28 22 23 25 30 12 Hz - 35

11. D 6

PDA 15 17 13 12 15 11 Hz - 20

MALT 16 17 12 12 17 16 - 20

12. D 8

PDA - - - - - - - -

MALT - - - - 16 - - -

13. D 15

PDA 11 - - - 10 - - -

MALT - - - - - - - -

14. D 25 PDA - - - - - - - -

MALT - - - - - - - -

15. D 28 PDA - - - - - - - -

MALT - - - - - - - -

16. D 34 PDA - - - - - - - -

MALT - - - - - - - -

17. G 2 PDA - - - - - - - -

MALT - - - - - - - -

18. G 3 PDA - - - - - - - -

MALT 23 34 11 12 30 - - 15

19. G 4

PDA 16 16 15 15 10 15 Hz - 15 Hz

MALT

- - - - - - - -

20. G 7

PDA - - - - - - - -

MALT - - - - - - - -

21. G 8 PDA - 20 15 - 18 16 - -

MALT - 16 14 - 16 - -

22. G 10 PDA 21 35 - - 30 - - 15

MALT 25 32 - - 30 - - 12

Page 65: Isolation, Characterization and Cosmeceutical Application

Chapter 3

46

Note: Hz = Hazy, D= Dwarka, P= Paldi, AN= Andaman & Nicobar, G=Goa

3.3.2. Determination of minimum inhibitory concentration (MIC)

The MIC of all active crude extracts was determined using a colorimetric micro

dilution method. We have found that all active crude extract have shown 90-99 %

inhibition at 1 mg/ml concentration against their respective bacterial strains (Figure

3.3 A, B). We can conclude that the MIC values of the best active crude extracts

ranged from 1 to 1.5 mg/ml. However this is the MIC value of crude extract and after

getting bioactive compound one can decide the dose. It will also be decided after

checking the toxicity dose of active principle. Further purification and identification

of active compounds are described later in this thesis.

23. G 14 PDA - - - - - - - -

MALT - - - - - - - -

24. G 15 PDA - - - - 10 - - -

MALT - - - - - - - -

25. AN1 PDA - - - - - - - -

MALT 7 - - - - - - -

26. AN2 PDA - - - - - - - -

MALT - - - - 15 - - -

27. AN4 PDA - - - - - - - -

MALT - - - - - - - -

28. AN5 PDA - - - - - - - -

MALT - - - - - - - -

29. AN6/

1

PDA - - - - - - - -

MALT - - - - - - - -

30. AN6/

2

PDA - - - - - - - -

MALT - - - - - - - -

31. AN7 PDA - 11 7 - 20 22 - 16

MALT - - - - - - - -

32. AN9 PDA - - - - - - - -

MALT - - - - - - - -

33. AN10 PDA 17 17

Hz

20 19 10 16 15 16

MALT 20 20 22 20 25 20 - 17

34. AN11 PDA 10 20 16 16 17 16 - -

MALT - 11 7 - - - - -

35. AN12 PDA 15 19 - - - - - -

MALT 26 39 14 14 35 - - 15

36. Tetra WATER 30 45 25 24 50 16 35 40

Page 66: Isolation, Characterization and Cosmeceutical Application

Chapter 3

47

Figure 3.3 A, B: MIC of active fungal crude extracts against respective bacteria

3.3.3. Mechanism of action (SEM analysis)

The effects of the most active crude extracts against their susceptible test

microorganisms were investigated by SEM. SEM images of the targeted 8 bacterial

cells revealed ultrastructural changes due to the active extracts. Control bacterial

cells had well defined, intact shapes and were smooth. After treatment with crude

extract (2mg/mL) for 18 hr. the cells showed considerable morphological alterations,

including deformation, shrinkage, collapsed and broken cells. Cell wall shrinkage

Page 67: Isolation, Characterization and Cosmeceutical Application

Chapter 3

48

and breakage were observed in S. aureus treated with D5, AN12 and G10 fungal

extracts (Figure 3.4). Cell wall shrinkage and breakage were also observed in S.

epidermidis treated with D5, AN7 and AN10 (Figure 3.5). Membrane invagination

was observed in B. megaterium when treated with D5, AN10 and AN11 (Figure 3.6).

Similarly, cell surface deformation was noticed in the cell surface of P. aeruginosa

and E.coli cells treated with D5, D6 and AN10 (Figure 3.7). Cell wall shrinkage and

breakage were observed in P. acnes treated with D5, AN12 and G10 fungal extracts

(Figure 3.8).

Figure 3.4: Effect of fungal crude extract on Staphylococcus aureus

Page 68: Isolation, Characterization and Cosmeceutical Application

Chapter 3

49

Figure 3.5: Effect of fungal crude extract on Staphylococcus epidermidis

Figure 3.6: Effect of fungal crude extract on Bacillus megaterium

Page 69: Isolation, Characterization and Cosmeceutical Application

Chapter 3

50

Figure 3.7: Effect of fungal crude extract on Pseudomonas aeruginosa and E. coli

Figure 3.8: Effect of fungal crude extract on Propionibacterium acnes

Page 70: Isolation, Characterization and Cosmeceutical Application

Chapter 3

51

3.3.4. Identification of marine fungi with strong antibacterial activity

A total of 35 isolates were screened for antibacterial activity. Consequently, eight

strongly active marine fungal isolates (D5, D6, AN 7, AN 10, AN 11, AN 12, G 3,

and G10) were selected for SEM, sequencing and identification based on ITS

sequences (Figure 3.9, 3.10). According to NCBI nucleotide blast analysis, among

the 8 identified fungi, all belonged to the phylum Ascomycota including genera

Penicillium, Aspergillus, Emericellopsis, Simplicillium and Leptosphaerulina. These

identified fungi and their best matches in the NCBI database are summarized in

Table 3.2. Six isolates matched their closest relatives with 99 % similarity except for

AN 7 (95%). It should be noted that the strain AN 7 was unsuccessfully identified on

fungal ITS sequences.

Table 3.2: Phylogenetic affiliations of selected fungi with strong antibacterial activity

Strain Morphological

identification

Closest identified relative Access. No. Identity

(% )

D 5 Simplicillium sp. Simplicillium lamellicola KC894712.1 99

D 6 Emericellopsis sp Emericellopsis minima KT290876.1 99

AN 7 Leptosphaerulina sp. Leptosphaerulina sp. JN850998.1 95

AN 10 Penicillium sp. Penicillium citrinum KT898717.1 99

AN 11 Aspergillus sp. Aspergillus aculeatus JX501355.1 99

AN 12 Penicillium sp. Penicillium chrysogenum KU925907.1 99

G 3 Aspergillus sp. Aspergillus oryzae HM145964.1 99

G 10 Aspergillus sp. Aspergillus sydowii JN851052.1 100

Page 71: Isolation, Characterization and Cosmeceutical Application

Chapter 3

52

Page 72: Isolation, Characterization and Cosmeceutical Application

Chapter 3

53

Figure 3.9: SEM morphological analysis of most active sporulating fungi

Page 73: Isolation, Characterization and Cosmeceutical Application

Chapter 3

54

Figure 3.10: Phylogenetic tree of partial ITS-rDNA sequences of marine fungal strains

The evolutionary history was inferred us ing the Neighbor-Joining method. The optimal tree with the

sum of branch length = 1.39517377 is shown. The percentage of replicate trees in which the associated

taxa clustered together in the bootstrap test (1000 replicates) are shown next to the branches. The tree is

drawn to scale, with branch lengths in the same units as those of the evolutionary distances used to

infer the phylogenetic tree. The evolutionary distances were computed us ing the Maximum Composite

Likelihood method and are in the units of the number of base substitutions per site. The analysis

involved 43 nucleotide sequences. All positions containing gaps and missing data were eliminated.

There were a total of 362 positions in the final dataset. Evolutionary analyses were conducted in

MEGA7.

Page 74: Isolation, Characterization and Cosmeceutical Application

Chapter 3

55

3.4. Discussion

Thirty five fungal strains were selected which were isolated from range of marine

environment in India. 8 isolates exhibited biological activity, representing 5 genera of

fungi. Fungus “D5” was grouped with S. lamellicola (Figure 3.10), which is known

to produce antibacterial mannosyl lipids, halymecins F and G and (3R,5R)-3-O-b-

Dmannosyl-3,5-dihydrodecanoic acid (115). In a recent study by (116), a S.

lamellicola isolated from Antarctica marine sediments exhibited both antibacterial

and antifungal activity.

Fungus “D6” was grouped with E. minima (Figure 3.10) which is known to produce

the sesquiterpenes (5E)-2-methyl-5-[(1'R*, 5'R*)-2-methylidene-7-oxobicyclo [3.2.1]

oct-6-ylidene]-4-oxopentanoic acid and dihydroisocoumarin cis-(3R, 4R)-4-

hydroxymellein. Both compounds were found to show neither antimicrobial nor in

vitro growth inhibitory activities on three human tumor cell lines (117). However, in

our study, we found that the crude extract of our marine fungal strain D6 E. minima

showed significant antibacterial activity. This could be because of the different hosts

or culture conditions resulting in different production of secondary metabolites.

Fungus “AN7” related to Leptosphaerulina sp. (Figure 3.10) was however not tested

for any of these activities. Recently (118), discovered new 6-methylpyridinone

derivatives from the marine-derived fungus Leptosphaerulina sp. using 1H NMR

prescreening and tracing the diagnostic signals.

Out of 8 selected fungi only 2, “AN 10” and “AN 12”, were related to Penicillium sp.

P. citrinum and P. chrysogenum (Figure 3.10). Both fungal strains displayed

antimicrobial activity against both gram positive and negative bacteria. In a study by

(119), two structurally unique steroids, isocyclocitrinol A and 22-

acetylisocyclocitrinol A, were isolated from a saltwater culture of P. citrinum

(Axinella sp., Papua New Guinea). Both compounds exhibited weak antibacterial

activity against S. epidermidis and Enterococcus durans. Two new diterpene, JBIR–

59 and 124, were obtained from P. citrinum SpI080624G1f01 (Ishigaki Island,

Okinawa,Japan) (120, 121). Two anthraquinone-citrinin derivatives penicillanthranin

A and B, isolated from P. citrinum PSU–F51 (Gorgonian, Annella sp., Similan

Islands, Thailand) were found active against both the wild and methicillin-resistant S.

aureus with a MIC value of 16 μg/ml (122). Penicitrinols G and H and 1,7-

Page 75: Isolation, Characterization and Cosmeceutical Application

Chapter 3

56

dihydroxy-8-(methoxycarbonyl)-9-oxo-9H-xanthene-3-carboxylic acid three

polyketides were isolated from P. citrinum SCSGAF 0167 (Echinogorgia aurantiaca,

South China Sea, China)(123). P. citrinum (Gastrointestine of the parrot fish Scalus

ovifrons, Okinawa Island, Japan) produced scalusamides A–C in which only

scalusamide A displayed antibacterial activity against M. luteus with a MIC value of

33.3 μg/ml (124).

Similarly chrysotriazoles A and B were obtained from endophytic P. chrysogenum

(brown alga Sargassum palladium, Fujian, China)(125). The diketopiperazine 365

was obtained from endophytic P. chrysogenum (mangrove leaves Porteresia

coarctata, Chorao Is., Goa, India) and displayed antibacterial activity comparable to

that of streptomycin (126). Recently (127), found that a crude extract of P.

chrysogenum DSOA associated with Indian Ocean marine sponge (Tedania

anhelans) exhibited antimycobacterial activity.

Aspergillus species are mostly known for their role as a pathogen. However, they

also play important roles in industry and our environment. Our last three fungi

belong to same group. Fungus “AN 11” was grouped with A. aculeatus (Figure

3.10), which is known to produce useful multi polysaccharide degrading enzymes for

industrial applications (128). We did not assess enzymatic properties in our study.

Several bioactive compounds have been isolated from the marine strain A. aculeatus

in the last 10 years; however these strains have not been tested for any of these

activities. Fungus “G 3” was grouped with A. oryzae (Figure 3.10) which is known

to produce the indoloditerpene derivatives Asporyzins A–C. Only asporyzin C

exhibited strong activity against E. coli (129). Asporyergosterol was isolated from an

endophytic A. oryzae (H. japonica, Yantai, China)(130).

Fungus “G 10” was grouped with A. sydowii (Figure 3.10) which is known to

produce M. tuberculosis protein tyrosine phosphatase A (PTPA) inhibitors sydowiol

A–C (131). The endophytic fungus A. sydowii, isolated from the red alga

Acanthophora spicifera (collected from Rameswaram, south India), produced two

new chlorinated 2,5-diarylcyclopentenones, sydowins A and B (132).

Page 76: Isolation, Characterization and Cosmeceutical Application

Chapter 3

57

3.5. Conclusion

In this study, 35 marine fungi were screened for antibacterial activity. Eight fungi

were successfully classified at the genus level based on ITS region sequences with

relatives in the NCBI database, except for one isolate (AN 7). They were distributed

among five genera (Penicillium, Aspergillus, Emericellopsis, Simplicillium and

Leptosphaerulina). Crude extracts of these eight fungi displayed different levels of

antibacterial activities to pathogenic bacteria, and some strains of the genera

Penicillium, Emericellopsis and Simplicillium showed strong growth inhibition to S.

aureus, S. epidermidis and M. luteus and P. acne. The combined data gathered from

this investigation contributes to the knowledge of fungal diversity and antibacterial

bioactivities associated with Indian marine environments. In conclusion, “D5” S.

lamellicola was found to be the most potent fungi isolated in this study and is

investigated in more detail, as described later in this thesis.

Page 77: Isolation, Characterization and Cosmeceutical Application

58

Chapter 4

Screening of marine fungi for potential tyrosinase

inhibitor and antioxidants

Page 78: Isolation, Characterization and Cosmeceutical Application

Chapter 4

59

4.1. Introduction

The skin is an essential organ of the body. It protects our body from adverse effects

caused by direct contact with the outside environment. Ultraviolet (UV) irradiation is

the most common and pernicious environmental factor that injures the skin (133).

Melanin is a mixture of heterogeneous biopolymers, responsible for the color of

human skin, hair, and eyes and protecting skin against radiation. The process of

melanin synthesis is called melanogenesis; which occurs within melanosomes,

membrane-bound granules, from melanocyte and is then transferred to keratinocytes

(134). But the accumulation of abnormal melanin induces pigmentation disorders,

such as melasma, freckles, ephelide, and senile lentigines (135). This process is

regulated by a variety of environmental, hormonal, and genetic factors, such as

ultraviolet (UV) exposure, α-melanocyte-stimulating hormone (α-MSH),

melanocortin 1 receptor (MC1R), and agouti-related protein (136-138). Tyrosinase, a

copper-containing monooxygenase, is a key enzyme that catalyzes melanin synthesis

in melanocytes (139). Therefore, tyrosinase inhibitors may be clinically useful for the

treatment of some dermatological diseases associated with melanin

hyperpigmentation and important in cosmetics for skin whitening (140).

In last decade, a large number of natural and synthetic compounds have been

discovered or developed as tyrosinase inhibitors (62, 141). Many of them like kojic

acid (142), arbutin, ascorbic acid derivatives, retinoic acid, azealic acid, ellagic acid,

chlorophorin, norartocarpanone, hydroquinone, catechins, aloesin, resveratrol.

oxyresveratrol, and 4,4′-dihydroxybiphenyl and other natural products are now in the

market (63, 140). However, there are still serious health concerns with some of these

ingredients, such as irreversible cutaneous damage, ochronosis, inflammation of

synthetic compounds and less effectiveness of available natural products due to their

adverse effects, poor skin penetrations, and low formulation stabilities (143, 144).

More research is needed toward the discovery and development of new cosmetic

ingredients, such as effective and safe tyrosinase inhibitors.

An imbalance state between intracellular antioxidants and intracellular reactive

oxygen species (ROS) or the so-called state of oxidative stress is a known

contributing factor for a wide range of human diseases. For example, cancers,

neurological disorders (Alzheimer and Parkinson), aging, atherosclerosis, diabetes,

Page 79: Isolation, Characterization and Cosmeceutical Application

Chapter 4

60

rheumatoid arthritis, inflammation, and other oxygen related diseases (145, 146).

Antioxidants are vital substances, playing an important role in the prevention of

oxidative damage of biomolecules and cells and ROS-induced diseases by reacting

with free radicals, scavenging free radicals and chelating free catalytic metals (147).

Due to increasing safety concerns involved with consumption of synthetic

antioxidants, exploitation of cheaper and safer sources of antioxidants from natural

origins, and especially from marine sources, is of interest. The strong antioxidant

power of some marine fungal species was demonstrated in several reviews (2, 148,

149). Marine resources have are good sources of bioactive compounds for the

development of new drugs and health foods. Marine fungi are known to be rich

producer of a variety of natural products. Hence, there is the possibility to identify

novel bioactive compounds with antityrosinase and antioxidant potential from marine

fungi obtained from west coast and Islands of India. In this study, we investigated

skin depigmenting and antioxidant activity of extracts from some recently collected

marine fungal.

4.2. Materials and methods

4.2.1. Chemicals used

All analytical grade chemicals were purchased from Fischer Scientific (Mumbai,

India) and used as such without further purification. Mushroom tyrosinase, kojic acid

and L-DOPA were purchased from Sigma, India.

4.2.2. Test organisms

4.2.2.1 Marine fungi

35 fungal strains used in this work were isolated from various marine samples

collected from west coast and Islands of India (Table 2.2 and Figure 2.3 in Section

2.3.1)

4.2.3. Cosmeceutical activity

4.2.3.1 .Culture extracts

The method for secondary metabolite extraction of 35 isolated marine fungi was

described in Chapter 3, Figure 3.2 in Section 3.2.3.

Page 80: Isolation, Characterization and Cosmeceutical Application

Chapter 4

61

4.2.3.2. Anti tyrosinase assay (MBTH assay)

The DOPA oxidase activity of tyrosinase was measured using a colorimetric assay,

which relies on the oxidation of L-DOPA to dopaquinone, which can be reacted with

3-methyl-bezothiazolinonehydrazone (MBTH) to form a pink product with peak

absorbance at 508 nm (Winder, 1994). Briefly, A typical reaction mixture of total

volume 200 µl contained 120 µl of assay buffer (100 mM potassium phosphate (pH

6.8), 2% (v/v) N, N‟-dimethylformamide, 5 mM MBTH), 40 µl of mushroom

tyrosinase (10 unit) and 40 µl of 1.5 mM L-DOPA solution. To study the inhibitory

effect of fungal crude extracts, 40 µl of crude (5mg/ml) replaced the assay buffer.

Reaction mixture without enzyme was first incubated at 37°C for 5 min. Enzyme was

added and the mixture was incubated at 37°C for 10 min; then absorbance was

measured at 505 nm using a microtiter plate-reader (BioTek, Synergy™ H1). The

absorbance of the mixture without tyrosinase was used as the control. Kojic acid was

used as positive controls. The optical density of the inhibition in the control was

considered to represent 100%. The data are expressed as mean percentages and the

results were repeated in triplicate.

The % antityrosinase activity was calculated by applying the expression:

4.2.3.3. Antioxidant assay

Antioxidant activity of the crude extracts was determined by DPPH free radical

scavenging assay as described by (110) with slight modification. 20 μL of crude

extract in water was mixed with 180 μL of DPPH in methanol (0.1 mM) in wells of a

96-well plate. The plate was kept in the dark for 30 min, after which the absorbance

of the solution was measured at 517 nm in a microtiter plate-reader (BioTek,

Synergy™ H1). Appropriate blanks (water) and standards (quercetin solutions in

water) were run simultaneously. Extracts were first tested at a single concentration of

5 mg/mL, and those showing good evidence of antioxidant activity were tested over a

range of concentrations.

% Inhibition= (A control − A sample) / A control ×100

Where A is absorbance at 508 nm

Page 81: Isolation, Characterization and Cosmeceutical Application

Chapter 4

62

The data are expressed as mean percentages and the results were repeated in

triplicate.

Inhibition of the DPPH free radical in percentage (I %) was calculated as:

4.2.3.4 Morphological and molecular characterization of potential fungal strains

The method described in Section 3.2.7.

4.3. Results

4.3.1. In vitro depigmenting activity

In present study, we used the MBTH assay to evaluate the depigmenting activity of

marine fungal extracts. The data shown in Figure 4.1 indicated that the mushroom

tyrosinase activity was inhibited by fungal extracts. We observed that at fixed

concentration (1mg/mL), six strains have shown more than 40% inhibition and five

strains showed more than 20% inhibition. It was also observed that PDA extracts had

higher activity then malt extracts. We have selected the eleven most active strains to

check their activity at different concentrations and found that PDA extracts of P2 and

D4 strains displayed the highest antityrosinase activity (45%, 43%, respectively) at

the lowest concentration (0.125mg/mL, Figure 4.2).

Dose and time dependent anti-tyrosinase activity of crude extracts of the selected

eleven most active strains were also evaluated. The results indicated that anti-

tyrosinase activity of extracts increased with increasing concentration; however this

activity decreased over time (Figure 4.3 A, B, C). We hypothesized that either

enzyme present in the extract degrades the active compound, or crude extract acts

noncompetitively. The tyrosinase inhibition ability might depend on the hydroxyl

groups of the phenolic compounds of the fungal extracts that could form a hydrogen

I % = [(A control − A sample) / A control] ×100

Where A control is the absorbance of the control reaction (full reaction,

without the tested extract or quercetin) and A sample was the absorbance

in the presence of the sample. DPPH solution was prepared freshly.

Decreased absorbance of the reaction mixture indicates stronger DPPH

free radical-scavenging activity.

Page 82: Isolation, Characterization and Cosmeceutical Application

Chapter 4

63

bond to the active site of the enzyme, leading to a lower enzymatic activity. Some

tyrosinase inhibitors act through hydroxyl groups that bind to the active site on

tyrosinase, resulting in steric hindrance or changed conformation (62). The

antioxidant activity may also be an important mechanisms for tyrosinase inhibitory

activity, and so this was also been evaluated.

Page 83: Isolation, Characterization and Cosmeceutical Application

Chapter 4

64

Figure 4.1: (A) Anti tyrosinase activity of Pladi and Dwarka marine fungal crude

extracts (B) Anti tyrosinase activity of Andaman and Goa marine fungal crude extracts

Figure 4.2: Anti tyrosinase activity of eleven selected fungal crude extracts at different

concentrations

0

10

20

30

40

50

60

70

80

90

100

P2

P

P2

M

P4

P

P8

P

P8

M

D4P

D4M

D6P

D25

P

A6/2

P

A6/2

M

A10

P

A10

M

G8P

G10

P

G15

P

koji

c a

cid

% In

hib

itio

n

Fungal code

Anti tyrosinase activity of selected fungal crude extracts at

different concentrations

0.5 mg/ml

1 mg/ml

2 mg/ ml

Page 84: Isolation, Characterization and Cosmeceutical Application

Chapter 4

65

Figure 4.3: Dose and time dependent anti tyrosinase activity of eleven selected fungal crude extracts

0

20

40

60

80

100

% In

hib

itio

n

Fungal code

Anti tyrosinase activity of fungal crude extracts at final

concentration of 0.5 mg/mL

10MIN

20MIN

30MIN

A

0

20

40

60

80

100

P2

P

P2

M

P4

P

P8

P

P8

M

D4P

D4M

D6P

D25

P

A6/2

P

A6/2

M

A10

P

A10

M

G8P

G10

P

G15

P

koji

c a

cid

% In

hib

itio

n

Fungal code

Anti tyrosinase activity of fungal crude extracts at final

concentration of 1 mg/mL

10 MIN

20 MIN

30 MIN

B

0

20

40

60

80

100

% In

hib

itio

n

Fungal code

Anti tyrosinase activity of fungal crude extracts at final

concentration of 2mg/mL

10MIN

20MIN

30MIN

C

Page 85: Isolation, Characterization and Cosmeceutical Application

Chapter 4

66

4.3.2. Antioxidant activity of fungal extracts

The scavenging effects of eleven fungal extracts on DPPH radicals increased with

increasing concentration. At 0.125, 0.25 and 0.50 mg/mL, the scavenging activities

of extracts on DPPH radical were 55–79, 62–83 and 86–97%, respectively (Figure

4.4). The results show that the PDA extract of P2 and D4 exhibited the highest

activity (94%, 97%, respectively) at 0.500 mg/mL, however at 0.125–0.500 mg/mL,

quercetin showed an excellent scavenging activity of 99–100 percent.

Figure 4.4: Scavenging activity of crude extracts of selected fungi against DPPH

4.3.3. Identification of promising marine fungal strains

A total of 35 fungal strains were isolated and screened for anti-tyrosinase and

antioxidant activity. The eleven most strongly active marine fungal isolates (P2, P4,

P8, D4, D6, D25, AN 6/2, AN 10, G 8, G 10, and G15) were selected for further

study. Of the eleven selected fungal strains the two best (P2 and D4) were selected

for SEM, sequencing and identification based on ITS sequences. These fungi were

identified as Talaromyces sp. and Aspergillus sp. respectively (Figure 4.7). Their

best matches in the NCBI database are shown in Table 4.1, Figure 4.5 and 4.6.

0

10

20

30

40

50

60

70

80

90

100

P2

P

P2

M

P4

P

P8

P

P8

M

D4

P

D4

M

D6

P

D2

5P

A6/

2P

A6/

2M

A10

P

A10

M

G8

P

G1

0P

G1

5P

Qu

erc

eti

n

Scav

en

gin

g a

cti

vit

y (

%)

Fingal Code

Scavenging effect on DPPH

500 mg/mL

250 mg/mL

125 mg/mL

Page 86: Isolation, Characterization and Cosmeceutical Application

Chapter 4

67

Table 4.1: Phylogenetic affiliations of selected fungi with strong activity

Strain Morphological

identification

Closest identified relative Access. No. Identity

(% )

P2 Talaromyces sp. Talaromyces stipitatus KJ413383 99

D4 Aspergillus sp Aspergillus terreus KJ958375 99

Figure 4.5: Phylogenetic tree of partial ITS-rDNA sequences of fungal strains

The evolutionary history was inferred using the Neighbor-Joining method. The optimal tree with the

sum of branch length = 0.45289971 is shown. The percentage of replicate trees in which the associated

taxa clustered together in the bootstrap test (1000 replicates) are shown next to the branches. The tree

is drawn to scale, with branch lengths in the same units as those of the evolutionary distances used to

infer the phylogenetic tree. The evolutionary distances were computed using the Maximum

Composite Likelihood method and are in the units of the number of base substitutions per site. The

analysis involved 9 nucleotide sequences. Codon positions included were 1st+2nd+3rd+Noncoding.

All positions containing gaps and missing data were eliminated. There were a total of 328 positions in

the final dataset. Evolutionary analyses were conducted in MEGA7.

Figure 4.6: SEM morphological analysis of sporulating D4 strain

Page 87: Isolation, Characterization and Cosmeceutical Application

Chapter 4

68

4.4. Discussion

Thirty five (35) fungal strains were selected for this study. Only 2 out of 35 fungi,

“Fungus P 2” related to T. stipitatus and “Fungus D 4” grouped with A. terreus,

displayed both strong antityrosinase and antioxidant activities at the lowest

concentration. Therefore, these two fungi were selected for further study. This is the

first report of antityrosinase activity from T. stipitatus.

A. terreus was found to produce an extracellular polysaccharide, YSS, which had

good antioxidant activity, as determined by its scavenging ability on DPPH radicals

(150). Two new rubrolides, rubrolides R and S were previously isolated from the

fermentation broth from A. terreus OUCMDZ-1925. Rubrolide R showed

comparable antioxidation against 2,20-azino-di(3-ethylbenzthiazoline-6-sulfonic

acid) (ABTS) radicals to that of trolox and ascorbic acid, with an IC50 value of 1.33

mM (151). A novel UV-A protecting dipyrroloquinone, terreusinone has been

isolated from the marine algicolous fungus A. terreus (152). Many other potential

compounds like sesterterpenoids, terretonins H and I (153), Lumazine containing

peptides Terrelumamides A and B (154) and terrein (155) were isolated from marine

A. terreus. This is the first report of antityrosinase activity from A. terreus.

4.5. Conclusion

Inhibitor of tyrosinase is an important class of bioactive compounds, particularly for

cosmetic use. This study is the first report showing combined antioxidant and

antityrosinase activities from the marine fungi Talaromyces stipitatus and Aspergillus

terreus. The results obtained in this study show that compounds present in the

extracts of Fungus “P2” and “D4” act as inhibitors of mushroom tyrosinase.

However, they showed lower inhibitory activity than Kojic acid and Quercetin. The

reducing power of the extracts increased in a dose-dependent manner. Furthermore,

the fungal culture filtrate extracts inhibited mushroom tyrosinase and decreased

melanin synthesis in a dose-dependent manner, which may be mediated through its

antioxidant activity. These extracts and their isolated active compounds have

potential as and skin whiteners for future cosmeceutical use.

Page 88: Isolation, Characterization and Cosmeceutical Application

69

Chapter 5

Bioassay-guided isolation of antibacterial compounds

from marine fungus Simplicillum lamellicola

Page 89: Isolation, Characterization and Cosmeceutical Application

Chapter 5

70

5.1. Introduction

Strain “D5” is a white cottony fungal strain isolated from a marine algal sample

collected from Dwarka, India. This strain was found to be active against

Staphylococcus epidermidis bacteria that cause skin microbial infection in humans,

as reported in chapter 3. This fungal strain was identified as Simplicillium lamellicola

by morphological and molecular methods. The genus Simplicillium is known to

produce a variety of compounds but has not been fully chemically characterized.

A deep sea derived fungal strain Simplicillium obclavatum EIODSF 020 was the

source of eight new linear peptides, simplicilliumtides A–H. None of the compounds

showed growth inhibition towards Staphylococcus aureus, Pseudomonas aeruginosa,

Bacillus amyloliquefaciens, and Bacillus stearothermophilus (156). Chemical

investigation of the crude extract of a sponge-derived fungus Simplicillium sp. YZ-11

gave a new minor diketopiperazine alkaloid cyclo-(2-hydroxy-Pro-Gly) and a natural

lactone (S)-dihydro-5-[(S)- hydroxyphenylmethyl]-2(3H)-furanone. However, no

activity has been reported for these compounds (157). Three novel mannosyl lipids,

halymecins F, G and (3R,5R)-3-O-β-D-mannosyl-3,5-dihydrodecanoic acid were

isolated from fungus S. lamellicola. These three novel compounds inhibited the

growth of phytopathogenic bacteria, with halymecin F displaying the strongest

antibacterial activity (115). A new pyrrolidine alkaloid, Preussin B, was isolated

from the crude extract of the fungus Simplicillium lanosoniveum TAMA 173. This

compound showed weak antifungal activity against Saccharomyces cerevisiae with

an IC50 value of 25 μg/mL (158). A sample of the fungus Simplicillium sp. FKI-5985

collected from Aogashima, Tokyo, Japan, was the source of Aogacillins A and B.

Both compounds were displayed antibacterial activity against methicillin-resistant

Staphylococcus aureus (MRSA) at MIC values of 2.0 μg/mL (159). To the best of

our knowledge, this is the first report of S. lamellicola from a marine environment.

This chapter reports bioassay guided isolation of the active component of S.

lamellicola, using TLC bio autography assay for antibacterial activity. The successful

isolation of one active biomolecule and establishment of general isolation protocol

for active metabolites are presented. Identification of the isolated metabolites was

done using UPLC-MS/MS.

Page 90: Isolation, Characterization and Cosmeceutical Application

Chapter 5

71

5.2. Method and materials

5.2.1. Chemicals used

All analytical grade chemicals were purchased from Fischer Scientific (Mumbai,

India) and used without further purification. PDB and Müller-Hinton agar were

procured from HiMedia (Mumbai, India) and were sterilized by autoclaving before

use.

5.2.2. Large scale fermentation and extraction

S. lamellicola (D5) was maintained on PDA artificial sea water (ASW) at 28°C ± 2,

pH 7.5. Agar plugs were used to inoculate eight 500 mL Erlenmeyer flasks

containing 250 mL of PDB in ASW pH 7.5. Flasks were incubated at 28°C ± 1 on a

rotary shaker at 140 rpm for five days and used as seed culture. Eight 5 L Erlenmeyer

flasks, each containing 2500 mL of liquid medium, prepared as above, were

individually inoculated with 10 % (250 mL) of seed culture and then incubated at

28°C ± 1 on a rotary shaker at 140 rpm for twenty days. The fermented broth was

filtered through cheesecloth to separate the supernatant and mycelia (160). The

supernatant was passed through an activated Diaion HP-20 resin column (Mitsubishi

Chemical Co.). Then the column was washed with 500 ml of distilled water and

finally eluted with 100% methanol until no activity was observed in the collected

fraction (108). The collected methanol fractions with activity were pooled and

evaporated, using a Rotary evaporator (BUCHI Rota vapor R-200). The fungal

mycelia were also soaked in methanol for 24 hours. The methanol layer was

collected and evaporated to dryness to give a methanol extract of fungal cell biomass,

which was combined with the above crude extract. The crude extract (6gm) was

collected and stored in cold condition.

5.2.3. Detection of antibacterial compounds by thin layer chromatography with direct

bioautography assay

TLC was performed on aluminium-backed TLC plates (Merck, silica gel 60 F254). A

suitable mobile phase was standardized based on the separation of antimicrobial

compounds. TLC was carefully marked and 5 µL of crude extract (5mg/mL in

methanol) was spotted onto the plate using a glass capillary. A chloroform: methanol

(9:1) solvent system was used for the separation of active principles. The

Page 91: Isolation, Characterization and Cosmeceutical Application

Chapter 5

72

chromatographic chamber was first saturated with system solvent followed by

spotting TLC plates for chromatographic separation. TLC strips were dried to

completely remove traces of the eluting solvents; TLC plates were then observed in

visible as well as ultraviolet light (365 and 254 nm). The color of resolved bands and

Rf values were recorded and marked. TLC plates were prepared in duplicate for

analyzing band separation and bioautography, respectively (161).

A contact bioautography against S. epidermidis was used (162). Mueller Hinton agar

no. 2 (HiMedia, India) was prepared and cooled to 40 - 45ºC and bacterial inoculum

(1.5×108 CFU/mL, 0.5 McFarland), prepared as described in chapter 3, 3.1.2.3, was

then added aseptically to the molten agar and poured into sterile petri dishes to give a

solid plate. Dried and developed TLC plates were then placed on to the media and

were left for a period of refrigerated for one hour for diffusion. Thereafter, TLC

plates were removed from the surface with the help of sterile forceps and the plates

were incubated for 24 hours. The areas of inhibition were marked and checked with

reference TLC to determine which spot was active. For clear observation and

photography of inhibition spots dishes were stained with methylthiazolyldiphenyl-

tetrazolium bromide (MTT) (5 mg/mL) solution in water.

5.2.4. Bioassay-guided fractionation of S. lamellicola methanol extract

Six grams of the dried methanol extract of S. lamellicola D 5 was suspended in 100

ml of distilled water and partitioned three times with 100 ml of each organic solvent

in increasing polarity (petroleum ether, hexane, chloroform and ethyl acetate

(EtOAc). The organic layer was concentrated to yield a solid residue. All organic

extracts were subjected to TLC bioautography assay against S. epidermidis bacteria.

The bioactive chloroform fraction 1.3 (500 mg) (Figure 5.1) was then adsorbed onto

silica gel (5 g) and subjected to column chromatography on silica gel (60 X 20 cm,

100 g, 60–120 mesh). Elution was achieved using stepwise gradients of 400 mL

aliquots of each of petroleum ether (100%); – hexane (100%); – hexane:chloroform

(50: 50) ;– hexane:chloroform (20:80), respectively (Figure 5.1). Collected tubes

were subjected to thin layer chromatography (TLC) on silica gel 60F254 plates using

solvent system chloroform:methanol (80:20). Aliquots with similar TLC profiles

were pooled to obtain fractions. TLC based antibacterial assay as described above

showed fraction 1.3.4 was active. Fraction 1.3.4 (180mg) was further subjected to

Page 92: Isolation, Characterization and Cosmeceutical Application

Chapter 5

73

column chromatography on silica gel (60 X 20 cm, 100 g, 200–400 mesh). Elution

was achieved using solvent gradients of 250 mL aliquots of each of chloroform

(100%), – chloroform:methanol (99.5:0.5), – chloroform:methanol (99:01), –

chloroform:methanol (98:02), – chloroform:methanol (95:05) and methanol (100%),

using a stepwise gradient elution. Aliquots were collected and observed on TLC.

Aliquots with similar TLC profiles were polled to obtained 7 fractions (1A-7A). All

were subjected to TLC based antibacterial assay. Fraction 5A showed partial activity

and 6A showed significant antibacterial activity. Fraction 6A (20mg) was further

purified using column chromatography on silica gel (60 X 20 cm, 100 g, 200–400

mesh). Elution was achieved using solvent gradients of 200 mL aliquots of each of

chloroform (100%), – chloroform:methanol (99.5:0.5), – chloroform:methanol

(99:01), – chloroform:methanol (98:02), – chloroform:methanol (95:05) and

methanol (100%) using a stepwise gradient elution. Aliquots were collected and

observed on TLC. Aliquots with similar TLC profiles were polled to obtain 7

fractions (6A1-6A7) (Figure 5.2). All were subjected to TLC based antibacterial

assay. Only fraction 6A5 showed antibacterial activity.

Isolated pure compound 6A5 and fraction 5A obtained from the previous column

were subjected to MPLC and UPLC-MS/MS for fingerprinting and identification of

metabolites.

Page 93: Isolation, Characterization and Cosmeceutical Application

Chapter 5

74

Figure 5.1: Schematic representation of isolation of metabolites from fungi “D5” crude

extract

Figure 5.2: Schematic representation of isolation of active compound from active

fraction 1.3.4 of “D5” crude extract

Page 94: Isolation, Characterization and Cosmeceutical Application

Chapter 5

75

5.2.5. Identification of isolated antibacterial compound

5.2.5.1. HPLC analysis

Identification of active compounds from fraction 5A and 6A5 was done using high-

performance liquid chromatography (HPLC) and LC-MS analysis. Chromatographic

analysis was performed on a HPLC system (Shimadzu, Kyoto, Japan) equipped with

a controller (CBM - 20A), quaternary pump (LC- 20AT), solvent degasser system

(DEU- 20 A5), auto sampler (SIL- 20A) and photo diode array detector (SPDM-

20A). Inbuilt software (Shimadzu, LC solution) was used to control the HPLC pump

and acquire the data form the photodiode array. Initial Separations were performed

with C-18 Phenomenex reverse phase 250 mm column, 5 μm spherical particles and

10 μl injects at 30 minute intervals was: flow rate 1 ml/min. An isocratic ratio 20:80

of solvent A (HPLC grade water) and B (HPLC grade C2H3N), respectively, was

used to elute compounds from the column. The flow rate was 1.0 mL/min, and the

eluent was monitored at 215 nm.

5.2.5.2. UPLC-mass spectrometry analysis

Sample 5A and 6A5 was prepared in concentration of 1 mg/ml and subjected to

UPLC-MS/MS (At Advanced Instrumentation Research Facility (AIRF, JNU, New

Delhi). UPLC analyses were performed on a Waters Acquity Ultra Performance

Liquid Chromatographic system with quadrupole mass spectrometer (Waters,

Milford, MA, USA). To achieve chromatograms with better resolution in a short

analysis time, the chromatographic conditions were optimized. UPLC separation was

achieved on an Acquity UPLC BEH C18 column using the mobile phase 0.1%

formic acid in water (solvent A) and acetonitrile (solvent B) at a flow rate of 0.300

ml/min; a gradient elution was performed. The total run time was 20 min. The

injection volume was 5.0 µl. The UPLC system was interfaced with quadrupole mass

spectrometer (Waters, USA). The same column, elution gradient and flow rate were

used during the UPLC-MS/MS analysis. Both positive and negative ion modes were

investigated, and the results showed that higher sensitivities and more information

were obtained in negative mode. Mass spectrometry was therefore operated in the

negative ion electrospray mode.

Page 95: Isolation, Characterization and Cosmeceutical Application

Chapter 5

76

5.3. Results and discussions

This study focused on the identification of antibacterial metabolites produced by the

marine S. lamellicola and the in vitro activities of these compounds against human

pathogenic bacteria to identify either novel or lead compounds to synthesize

derivatives with enhanced activities. The Aogacillins A and B were previously

identified from a culture broth of terrestrial Simplicillium sp. FKI-5985 with

antibacterial activity against methicillin-resistant S. aureus (MRSA)(159). However,

we isolated the strain S. lamellicola from marine habitat. In this study, we have

characterized previously unidentified antibacterial metabolites produced by marine S.

lamellicola.

5.3.1. Characterization of substances from the culture broth of marine S. lamellicola

To reveal the antimicrobial constituents by bioautography, 5 mg/ml of D5 crude

extract was spotted on TLC. The anti- S. epidermice bioautography of this TLC

clearly showed that there was at least one or two antibacterial component. The crude

extract was further fractionated with various solvents. We found that the chloroform

layer significantly inhibited the growth of S. epidermice and so the material was

patriated by repeated silica gel column chromatography to provide 7 fractions. Using

bioassay-guided fractionation, the antibacterial metabolites were isolated from the 6th

fraction by like normal phase Colum chromatography at atmospheric pressure.

Figure 5.3: TLC bioautography of fraction 1.3.4 of “D5” crude extract

Page 96: Isolation, Characterization and Cosmeceutical Application

Chapter 5

77

Figure 5.4 HPLC chromatogram of minor active fraction 5A

Figure 5.5: HPLC chromatogram of purified active compound from fraction 6A5

Page 97: Isolation, Characterization and Cosmeceutical Application

Chapter 5

78

Figure 5.6: UPLC-MS chromatograms of minor active fraction 5A

Figure 5.7: UPLC-MS chromatograms of active fraction 6A5

Page 98: Isolation, Characterization and Cosmeceutical Application

Chapter 5

79

The present study identifies three compounds which were previously reported the be

isolated from Simplicillium sp. One compound was identified as 5, 6-dihydro-6-

pentyl-5, 6-dihydropyran-2-one, with the molecular formula is C10H18O3 as

determined by ESI-MS m/z ([M + Cl] -168). Compound 2 identified as simplifungin,

with molecular formula C18H37NO4 as determined by ESI-MS m/z ([M + Cl] - 331).

Compound 3 was identified as a derivative of halymecin G. Its molecular formula is

C20H38O6 as determined ESI-MS m/z ([M + Cl] -374) (Figure 5.8). The structure

confirmation of these three compounds by NMR is under investigation and will be

reported in due course.

O

O CH3

5,6-Dihydro-6-pentyl-5,6-dihydropyran-2-one (1) MW=168

NH2

H3C

OH COOH

OH

Simplifungin (2) MW=331

O O O

OR O OH O OH O OH

OH

O

O

O

HO

OH

HO

OHHalymecin G (R)= OAc, MW=779

O OH

O OH OORCompound 3 (R)=OH, MW=374

Figure 5.8: Structures of compounds identified from marine S. lamellicola

Page 99: Isolation, Characterization and Cosmeceutical Application

Chapter 5

80

5.3.2. Antibacterial activity of pure active compound

The in vitro antibacterial activity of purified compound 6A5 was tested by direct

bioautography and caused growth inhibition of S. epidermidis bacteria. This

compound was identified as 5, 6-dihydro-6-pentyl-5, 6-dihydropyran-2-one.

Figure 5.9: Fraction 6A5 with moderate antibacterial activity against S. epidermidis

5.4. Conclusions

The active constituent of marine fungus S. lamellicola was isolated and reported in

this chapter. Fungi “D5” produced one active antibacterial compound, which was

showed moderate active. Antibacterial bioassay-directed fractionation was used to

isolate 5, 6-dihydro-6-pentyl-5, 6-dihydropyran-2-one from 6A5 fraction. The

bioassay of fraction 5A showed minor inhibition. When analyzed the same, a peak at

Rt 1.198 (of the bioactive compound) was present in the fraction, which could be the

probable reason of its activity. UPLC-MS/MS data of 5A revealed presence of

compound “simplifungin” (m/z = 331, [M + Cl] - =368-37). We also propose a

derivative of previously reported compound “halymecin G” with m/z =374.

Page 100: Isolation, Characterization and Cosmeceutical Application

81

Chapter 6

Summary and future work

Page 101: Isolation, Characterization and Cosmeceutical Application

Chapter 6

82

Widespread application of natural products in many different fields, including

medicine, food, cosmetics and agriculture has increased the demand for new and

novel bioactive natural chemical entities. The marine world is a vast resource

containing structurally complex and novel chemical entities with unique

pharmacology. The complexity of the marine environment results in the evolution of

diverse genetically encoded biomolecules with a variety of ecological and metabolic

roles in marine organisms. The most promising bioactive marine natural products

have been derived from microbial bioresources such as bacteria, cyanobacteria and

fungi, particularly as these organisms can be used as production systems for these

often structurally complex compounds. However marine fungi remain a relatively

untapped bioresource for bioactive molecules, compared with terrestrial fungi.

The main goal of the research work described in this thesis was to discover novel

natural products with cosmeceutical potential by employing targeted

pharmacological methods. To achieve this goal, the present study describes the

isolation and identification of some new marine fungi, screening of these fungi for

bioactive compounds, the isolation and structure elucidation of some bioactive

secondary metabolites, and some assessment of the pharmacological activity of the

isolated compounds. Specifically, the antibacterial, antityrosinase and antioxidant

activities of the marine fungi extracts and purified compounds were assessed.

Chapter 2 describes the occurrence, distribution and diversity of culturable marine

fungi on various substrata (sea grasses, wood and seaweed) collected at various

locations in the Indian west coast and Andaman Islands. Effects of nutrient and salt

concentration on the fungal growth for secondary metabolites production were

investigated using a range of media conditions. The isolated fungal strains (70

strains) were maintained on potato dextrose agar in artificial sea water incubated at

28°C ± 2 for 7-10 days. Individual fungal colonies were picked and checked for

further purity by sub culturing on potato dextrose agar in artificial sea. Of these, only

35 strains were culturable and subjected for growth assessment experiments on

different solid media plate (Media A (PDA in 100% sea water with pH 7.5 (High

nutrient), Media B (Sea water media (SWM), 1.0% glucose, 0.5% peptone, 0.1%

yeast extract and 1.7% agar, final pH of 7.5) (Low nutrient), showing a clear

difference in fungal mycelia growth and spore formation. Higher mycelial production

was observed in nutrient rich media and earlier spore formation was observed in

Page 102: Isolation, Characterization and Cosmeceutical Application

Chapter 6

83

nutrient deficient media. The results described in this study will be useful for

selection of growth media and culture conditions for new active fungi for optimised

production of secondary metabolites.

In chapter 3, the primary aim was to evaluate antibacterial activity of all isolated

marine fungal strains. Selected strains were further characterized using microscopic

technique including phase contrast and SEM. A common protocol for secondary

relatives in the NCBI database, except for one isolates (AN 7). These were

Simplicillium lamellicola (D 5), Emericellopsis minima (D 6), Leptosphaerulina sp.;

(AN 7), Penicillium citrinum (AN 10), Aspergillus aculeatus (AN 11), P.

chrysogenum (AN 12), A. oryzae (G 3) and A. sydowii (G 10). SEM analysis of the

targeted bacteria before and after treatment with crude fungal extracts showed that

the presence of secondary metabolites in the extracts caused damage to bacterial cell

membrane. Isolate Simplicillium lamellicola (D 5) was the most active extract against

both Gram-negative and Gram-positive bacteria. This promising strain was

investigated further using bioassay-guided fractionation for the isolation of bioactive

compounds.

Chapter 4 describes the effect of selected marine fungi on the pigmentation and

photoaging of skin. Extracellular secondary metabolites of fungal fermentation were

concentrated by using Diaion HP20 resin. Here, the first target was inhibition of the

melanin forming enzyme tyrosinase by the crude extract of each fungal strain. This

was analyzed using a MBTH assay. The activity of the eleven most active strains was

investigated over a range of concentrations (0.5-2.0 mg/ml). These extracts were also

tested for antioxidant activity. Two of the isolates, Talaromyces stipitatus (P2) and

Aspergillus terreus (D4) were found to have potent activity, comparable with that of

the positive controls Quercetin and Kojic acid. To date, reports concerning

antioxidant and antityrosinase metabolites from marine fungi are rare. This study

demonstrates that marine fungi are a potentially an excellent source for the discovery

of novel cosmeceutical agents.

Chapter 5 describes the purification and identification of antibacterial bioactive

molecules from S. lamellicola extract, via bioassay-guided fractionation using TLC

bioautography assay. From 20 lit of fermented broth, 6 gm of crude extract was

prepared by using Diaon HP 20 extraction. Contact bioautography of the extract

Page 103: Isolation, Characterization and Cosmeceutical Application

Chapter 6

84

showed one spot inhibiting activity against Staphylococcus epidermidis bacteria.

Silica gel column chromatography was performed to purify the active compound

present in the extract. We isolated one pure active compound, identified as 5, 6-

dihydro-6-pentyl-5, 6-dihydropyran-2-one. One the basis of UPLC-MS/MS data we

also proposed the presence of the compound “simplifungin” (m/z = 331, [M + Cl] -

=368-37) and a new derivative of the previously reported compound “halymecin G”,

with m/z =374.

This thesis is the first report of the isolation of marine S. lamellicola from an Indian

marine environment and its evaluation for antibacterial activity against a range of

human pathogenic bacteria. The antityrosinase activity of Talaromyces stipitatus is

reported for the first time. Both of these fungi are promising strains for further

investigation. Future work will target the isolation and identification of antityrosinase

compounds from T. stipitatus and modification of the identified antibacterial

compound from S. lamellicola to increase its biological activity.

Page 104: Isolation, Characterization and Cosmeceutical Application

85

References

1. Grabley S, Thiericke R. Drug discovery from nature: Springer Science &

Business Media; 1998.

2. Blunt JW, Copp BR, Keyzers RA, Munro M, Prinsep MR. Marine Natural

Products. Natural product reports. 2015;32(2):116-211.

3. Katz L, Baltz RH. Natural product discovery: past, present, and future.

Journal of industrial microbiology & biotechnology. 2016:1-22.

4. Jimeno J, Faircloth G, Sousa-Faro J, Scheuer P, Rinehart K. New Marine

Derived Anticancer Therapeutics─ A Journey from the Sea to Clinical Trials. Marine

drugs. 2004;2(1):14-29.

5. Vignesh S, Raja A, James RA. Marine drugs: Implication and future studies.

International Journal of Pharmacology. 2011;7:22-30.

6. Gerwick WH, Moore BS. Lessons from the past and charting the future of

marine natural products drug discovery and chemical biology. Chemistry & biology.

2012;19(1):85-98.

7. Kijjoa A, Sawangwong P. Drugs and cosmetics from the sea. Marine drugs.

2004;2(2):73-82.

8. Molinski TF, Dalisay DS, Lievens SL, Saludes JP. Drug development from

marine natural products. Nature Reviews Drug Discovery. 2009;8(1):69-85.

9. Mayer AM, Glaser KB, Cuevas C, Jacobs RS, Kem W, Little RD, et al. The

odyssey of marine pharmaceuticals: a current pipeline perspective. Trends in

Pharmacological Sciences. 2010;31(6):255-65.

10. Piel J. Metabolites from symbiotic bacteria. Natural product reports.

2009;26(3):338-62.

11. Laatsch H. Antibase 2012: The natural compound identifier: Wiley-Vch;

2012.

12. Murphy B, Jensen P, Fenical W. The Chemistry of Marine Bacteria. In:

Fattorusso E, Gerwick WH, Taglialatela-Scafati O, editors. Handbook of Marine

Natural Products: Springer Netherlands; 2012. p. 153-90.

13. Waters AL, Hill RT, Place AR, Hamann MT. The expanding role of marine

microbes in pharmaceutical development. Current Opinion in Biotechnology.

2010;21(6):780-6.

Page 105: Isolation, Characterization and Cosmeceutical Application

References

86

14. Rateb ME, Ebel R. Secondary metabolites of fungi from marine habitats.

Natural product reports. 2011;28(2):290-344.

15. Borse B, Bhat D, Borse K, Tuwar N, Pawar N. Marine fungi of India

(Monograph): Broadway Publishing House; 2012.

16. Bugni TS, Ireland CM. Marine-derived fungi: a chemically and biologically

diverse group of microorganisms. Natural product reports. 2004;21(1):143-63.

17. Hyde KD, Jones EG, Leaño E, Pointing SB, Poonyth AD, Vrijmoed LL. Role

of fungi in marine ecosystems. Biodiversity and Conservation. 1998;7(9):1147-61.

18. Jones EBG, Sakayaroj J, Suetrong S, Somrithipol S, Pang K. Classification of

marine Ascomycota, anamorphic taxa and Basidiomycota. Fungal Diversity.

2009;35(1):187.

19. Thomas TR, Kavlekar DP, LokaBharathi PA. Marine drugs from sponge-

microbe association-a review. Marine drugs. 2010;8(4):1417-68.

20. Jones EG. Fifty years of marine mycology. Fungal Diversity. 2011;50(1):73-

112.

21. Jones E. Are there more marine fungi to be described? Botanica Marina.

2011;54(4):343-54.

22. Shearer C, Descals E, Kohlmeyer B, Kohlmeyer J, Marvanová L, Padgett D,

et al. Fungal biodiversity in aquatic habitats. Biodiversity and Conservation.

2007;16(1):49-67.

23. Liberra K, Lindequist U. Marine fungi-a prolific resource of biologically

active natural products? Die Pharmazie. 1995;50(9):583-8.

24. Abdel-Lateff A. Chaetominedione, a new tyrosine kinase inhibitor isolated

from the algicolous marine fungus Chaetomium sp. Tetrahedron Letters.

2008;49(45):6398-400.

25. Kossuga MH, Romminger S, Xavier C, Milanetto MC, Valle MZd, Pimenta

EF, et al. Evaluating methods for the isolation of marine-derived fungal strains and

production of bioactive secondary metabolites. Revista Brasileira de Farmacognosia.

2012;22(2):257-67.

26. Masuma R, Yamaguchi Y, Noumi M, Ōmura S, Namikoshi M. Effect of sea

water concentration on hyphal growth and antimicrobial metabolite production in

marine fungi. Mycoscience. 2001;42(5):455-9.

Page 106: Isolation, Characterization and Cosmeceutical Application

References

87

27. Huang J, Lu C, Qian X, Huang Y, Zheng Z, Shen Y. Effect of salinity on the

growth, biological activity and secondary metabolites of some marine fungi. Acta

Oceanologica Sinica. 2011;30(3):118-23.

28. Wang Y, Lu Z, Sun K, Zhu W. Effects of high salt stress on secondary

metabolite production in the marine-derived fungus Spicaria elegans. Marine drugs.

2011;9(4):535-42.

29. Sunga MJ, Teisan S, Tsueng G, Macherla VR, Lam KS. Seawater

requirement for the production of lipoxazolidinones by marine actinomycete strain

NPS8920. Journal of industrial microbiology & biotechnology. 2008;35(7):761-5.

30. Miao L, Kwong TF, Qian P-Y. Effect of culture conditions on mycelial

growth, antibacterial activity, and metabolite profiles of the marine-derived fungus

Arthrinium cf saccharicola. Applied Microbiology and Biotechnology.

2006;72(5):1063-73.

31. Lawrence RN. Rediscovering natural product biodiversity. Drug Discovery

Today. 1999;4(10):449-51.

32. Saleem M, Ali MS, Hussain S, Jabbar A, Ashraf M, Lee YS. Marine natural

products of fungal origin. Natural product reports. 2007;24(5):1142-52.

33. Duarte K, Rocha-Santos TA, Freitas AC, Duarte AC. Analytical techniques

for discovery of bioactive compounds from marine fungi. Trends in Analytical

Chemistry. 2012;34:97-110.

34. Kiuru P, D‟Auria MV, Muller CD, Tammela P, Vuorela H, Yli-Kauhaluoma

J. Exploring marine resources for bioactive compounds. Planta Medica.

2014;80(14):1234-46.

35. Draelos ZD. The future of cosmeceuticals: an interview with Albert Kligman.

Dermatologic Surgery. 2005;31(7):890.

36. Dureja H, Kaushik D, Gupta M, Kumar V, Lather V. Cosmeceuticals: An

emerging concept. Indian Journal of Pharmacology. 2005;37(3):155.

37. Kim S-K. Marine cosmeceuticals: trends and prospects: CRC Press; 2011.

38. Amer M, Maged M. Cosmeceuticals versus pharmaceuticals. Clinics in

dermatology. 2009;27(5):428-30.

39. Chermahini SH, Majid FAA, Sarmidi MR. Cosmeceutical value of herbal

extracts as natural ingredients and novel technologies in anti-aging. Journal of

Medicinal Plants Research. 2011;5:3074-7.

Page 107: Isolation, Characterization and Cosmeceutical Application

References

88

40. Athukorala Y, Kim K-N, Jeon Y-J. Antiproliferative and antioxidant

properties of an enzymatic hydrolysate from brown alga, Ecklonia cava. Food and

Chemical Toxicology. 2006;44(7):1065-74.

41. Yoon NY, Eom T-K, Kim M-M, Kim S-K. Inhibitory effect of phlorotannins

isolated from Ecklonia cava on mushroom tyrosinase activity and melanin formation

in mouse B16F10 melanoma cells. Journal of Agricultural and Food Chemistry.

2009;57(10):4124-9.

42. Tsuchiya T, Yamada K, Minoura K, Miyamoto K, Usami Y, Kobayashi T, et

al. Purification and determination of the chemical structure of the tyrosinase inhibitor

produced by Trichoderma viride strain H1-7 from a marine environment. Biological

and Pharmaceutical Bulletin. 2008;31(8):1618-20.

43. Li X, Kim MK, Lee U, Kim S-K, Kang JS, Choi HD, et al. Myrothenones A

and B, cyclopentenone derivatives with tyrosinase inhibitory activity from the

marine-derived fungus Myrothecium sp. Chemical and Pharmaceutical Bulletin.

2005;53(4):453-5.

44. Gasco-Buisson MC. Key trends driving anti-aging skin care in 2009 and

beyond. Textbook of aging skin: Springer; 2010. p. 1197-205.

45. Bronaugh RL, Katz LM. Cosmetics and aging skin. Textbook of Aging Skin:

Springer; 2010. p. 1063-8.

46. Kosmadaki MG, Gilchrest BA. The demographics of aging in the United

States: implications for dermatology. Archives of Dermatology. 2002;138(11):1427-

8.

47. Gorouhi F, Maibach H. Role of topical peptides in preventing or treating aged

skin. International journal of cosmetic science. 2009;31(5):327-45.

48. Baumann L. Skin ageing and its treatment. The Journal of Pathology.

2007;211(2):241-51.

49. Yaar M, Eller MS. Mechanisms of aging. Archives of Dermatology.

2002;138(11):1429.

50. Geserick C, Blasco MA. Novel roles for telomerase in aging. Mechanisms of

Ageing and Development. 2006;127(6):579-83.

51. Martínez P, Blasco MA. Telomeric and extra-telomeric roles for telomerase

and the telomere-binding proteins. Nature Reviews Cancer. 2011;11(3):161-76.

Page 108: Isolation, Characterization and Cosmeceutical Application

References

89

52. Dikmen ZG, Gellert GC, Jackson S, Gryaznov S, Tressler R, Dogan P, et al.

In vivo inhibition of lung cancer by GRN163L: a novel human telomerase inhibitor.

Cancer Research. 2005;65(17):7866-73.

53. Warabi K, Matsunaga S, van Soest RW, Fusetani N. Dictyodendrins AE, the

First Telomerase-Inhibitory Marine Natural Products from the Sponge

Dictyodendrilla v erongiformis 1. The Journal of Organic Chemistry.

2003;68(7):2765-70.

54. Guittat L, De Cian A, Rosu F, Gabelica V, De Pauw E, Delfourne E, et al.

Ascididemin and meridine stabilise G-quadruplexes and inhibit telomerase in vitro.

Biochimica et Biophysica Acta (BBA)-General Subjects. 2005;1724(3):375-84.

55. Warabi K, Hamada T, Nakao Y, Matsunaga S, Hirota H, Van Soest RW, et al.

Axinelloside A, an Unprecedented Highly Sulfated Lipopolysaccharide Inhibiting

Telomerase, from the Marine Sponge, Axinella infundibula 1. Journal of the

American Chemical Society. 2005;127(38):13262-70.

56. Jeon S, Kim K, Koh J, Kong K. Inhibitory effects on L-dopa oxidation of

tyrosinase by skin-whitening agents. Bulletin Korean Chemical Society.

2005;26(7):1135.

57. Lee H-S, Shin K-H, Ryu G-S, Chi G-Y, Cho I-S, Kim H-Y. Synthesis of

small molecule-peptide conjugates as potential whitening agents. Bulletin Korean

Chemical Society. 2012;33(9):3004-8.

58. Abu Ubeid A, M Hantash B. Minireview: peptide analogs and short sequence

oligopeptides as modulators of skin pigmentation. Current topics in Medicinal

Chemistry. 2014;14(12):1418-24.

59. Solano F, Briganti S, Picardo M, Ghanem G. Hypopigmenting agents: an

updated review on biological, chemical and clinical aspects. Pigment Cell Research.

2006;19(6):550-71.

60. Fais A, Corda M, Era B, Fadda MB, Matos MJ, Santana L, et al. Tyrosinase

inhibitor activity of coumarin-resveratrol hybrids. Molecules. 2009;14(7):2514-20.

61. Reszko AE, Berson D, Lupo MP. Cosmeceuticals: practical applications.

Dermatologic clinics. 2009;27(4):401-16.

62. Chang T-S. An updated review of tyrosinase inhibitors. International journal

of molecular sciences. 2009;10(6):2440-75.

63. Wu B. Tyrosinase inhibitors from terrestrial and marine resources. Current

topics in Medicinal Chemistry. 2014;14(12):1425-49.

Page 109: Isolation, Characterization and Cosmeceutical Application

References

90

64. Zhang D, Li X, Kang JS, Choi HD, Son BW. A New α-Pyrone Derivative, 6-

[(E)-Hept-1-enyl]-α-pyrone, with Tyrosinase Inhibitor Activity from a Marine Isolate

of the Fungus Botrytis. Bull Korean Chem Soc. 2007;28(5):887.

65. Wu B, Wu X, Sun M, Li M. Two novel tyrosinase inhibitory sesquiterpenes

induced by CuCl2 from a marine-derived fungus Pestalotiopsis sp. Z233. Marine

drugs. 2013;11(8):2713-21.

66. Lee DJ, Van Dyke GS, Kim J. Update on pathogenesis and treatment of acne.

Current Opinion in Pediatrics. 2003;15(4):405-10.

67. Coates P, Vyakrnam S, Eady E, Jones C, Cove J, Cunliffe W. Prevalence of

antibiotic‐resistant propionibacteria on the skin of acne patients: 10‐year surveillance

data and snapshot distribution study. British Journal of Dermatology.

2002;146(5):840-8.

68. Kamei Y, Sueyoshi M, Hayashi K-i, Terada R, Nozaki H. The novel anti-

Propionibacterium acnes compound, Sargafuran, found in the marine brown alga

Sargassum macrocarpum. The Journal of antibiotics. 2009;62(5):259-63.

69. Wu B, Oesker V, Wiese J, Schmaljohann R, Imhoff JF. Two New Antibiotic

Pyridones Produced by a Marine Fungus, Trichoderma sp. Strain MF106. Marine

drugs. 2014;12(3):1208-19.

70. Hedgepeth J. Sandy beaches: Geological Society of America; 1957. 587-608

p.

71. Bhatnagar I, Kim SK. Immense essence of excellence: marine microbial

bioactive compounds. Marine drugs. 2010;8(10):2673-701.

72. Kubanek J, Jensen PR, Keifer PA, Sullards MC, Collins DO, Fenical W.

Seaweed resistance to microbial attack: a targeted chemical defense against marine

fungi. Proceedings of the National Academy of Sciences. 2003;100(12):6916-21.

73. Jones EG, Byrne P. Physiology of the higher marine fungi. Recent Advances

in Aquatic Mycology EB Gareth Jones ed. 1976.

74. Battley EH. Marine Mycology: The Higher Fungi. Jan Kohlmeyer , Erika

Kohlmeyer. The Quarterly Review of Biology. 1980;55(2):185-.

75. Thomas TRA, Kavlekar DP, LokaBharathi PA. Marine drugs from sponge-

microbe association-A review. Marine drugs. 2010;8(4):1417-68.

76. Hyde K, Jones E. Marine mangrove fungi. Marine ecology. 1988;9(1):15-33.

77. Hooley P, Whitehead M. The genetics and molecular biology of marine fungi.

Mycologist. 2006;20(4):144-51.

Page 110: Isolation, Characterization and Cosmeceutical Application

References

91

78. Cuomo V, Jones EG, Grasso S. Occurrence and distribution of marine fungi

along the coast of the Mediterranean Sea. Progress in Oceanography.

1988;21(2):189-200.

79. Jones E. The distribution of marine fungi on wood submerged in the sea.

Biodeterioration of materials: Elsevier publishing co.; 1968. p. 460-85.

80. Jones E, Irvine J, editors. The role of marine fungi in the biodeterioration of

materials. Biodeterioration of materials Proceedings of the 2nd international

biodeterioration symposium Lunteren, the netherlands, 13th-18th september 1971;

1972: Applied Science.

81. Xu L, Meng W, Cao C, Wang J, Shan W, Wang Q. Antibacterial and

Antifungal Compounds from Marine Fungi. Marine drugs. 2015;13(6):3479-513.

82. Keller MD, Selvin RC, Claus W, Guillard RR. Media for the culture of

oceanic ultraphytoplankton1, 2. Journal of Phycology. 1987;23(4):633-8.

83. Rajasekar T, Balaji S, Kumaran S, Deivasigamani B, Pugzhavendhan S.

Isolation and characterization of Marine fungal metabolites against clinical

pathogens. Asian Pacific Journal of Tropical Disease. 2012;2:S387-S92.

84. Persidis A. Antibacterial and antifungal drug discovery. Nature

biotechnology. 1999;17(11):1141-2.

85. Elixhauser A, Steiner C. Most Common Diagnoses and Procedures in US

Community Hospitals, 1996. Healthcare Cost and Utilization Project, HCUP

Research Note Rockville, MD Agency for Health Care Policy and Research 1999:99-

0046.1999.

86. Russo A, Concia E, Cristini F, De Rosa F, Esposito S, Menichetti F, et al.

Current and future trends in antibiotic therapy of acute bacterial skin and skin-

structure infections. Clinical Microbiology and Infection. 2016;22:S27-S36.

87. DiNubile MJ, Lipsky BA. Complicated infections of skin and skin structures:

when the infection is more than skin deep. Journal of Antimicrobial Chemotherapy.

2004;53(suppl 2):ii37- ii50.

88. Nichols RL. Optimal treatment of complicated skin and skin structure

infections. Journal of Antimicrobial Chemotherapy. 1999;44(suppl 1):19-23.

89. Shah M, Shah HD. Acute bacterial skin and skin structure infections: current

perspective. Indian Journal of Dermatology. 2011;56(5):510.

Page 111: Isolation, Characterization and Cosmeceutical Application

References

92

90. Cassell GH, Mekalanos J. Development of antimicrobial agents in the era of

new and reemerging infectious diseases and increasing antibiotic resistance. Jama.

2001;285(5):601-5.

91. Daszak P, Cunningham AA, Hyatt AD. Emerging infectious diseases of

wildlife--threats to biodiversity and human health. Science. 2000;287(5452):443-9.

92. Giordano P, Weber K, Gesin G, Kubert J. Skin and skin structure infections:

treatment with newer generation fluoroquinolones. Therapeutics and clinical risk

management. 2007;3(2):309.

93. Radjasa OK. Marine Fungi: The Untapped Diversity of Marine

Microorganisms. Journal of Coastal Zone Management. 2015;2015.

94. Kelecom A. Secondary metabolites from marine microorganisms. Anais da

Academia Brasileira de Ciencias. 2002;74(1):151-70.

95. Jensen PR, Fenical W. Marine microorganisms and drug discovery: current

status and future potential. In Drugs from the Sea 2000: 6-29.

96. Bhadury P, Mohammad BT, Wright PC. The current status of natural

products from marine fungi and their potential as anti-infective agents. Journal of

industrial microbiology & biotechnology. 2006;33(5):325-37.

97. Bugni TS, Ireland CM. Marine-derived fungi: a chemically and biologically

diverse group of microorganisms. Natural product reports. 2004;21(1):143-63.

98. Jones E. Marine fungi: some factors influencing biodiversity. Fungal

Diversity. 2000;4(193):53-73.

99. Raghukumar C. Marine fungal biotechnology: an ecological perspective.

Fungal Diversity. 2008;31:19-35.

100. Richards TA, Jones MD, Leonard G, Bass D. Marine fungi: their ecology and

molecular diversity. Annual review of marine science. 2012;4:495-522.

101. Ebada SS, Proksch P. Marine-Derived Fungal Metabolites. Hb25_Springer

Handbook of Marine Biotechnology: Springer; 2015. p. 759-88.

102. Newton GG, Abraham E. Cephalosporin C, a new antibiotic containing

sulphur and D-α-aminoadipic acid. Nature. 1955;175(4456):548-.

103. Rowley DC, Kelly S, Kauffman CA, Jensen PR, Fenical W. Halovirs A–E,

new antiviral agents from a marine-derived fungus of the genus Scytalidium.

Bioorganic & medicinal chemistry. 2003;11(19):4263-74.

104. Bringmann G, Lang G, Gulder TA, Tsuruta H, Mühlbacher J, Maksimenka K,

et al. The first sorbicillinoid alkaloids, the antileukemic sorbicillactones A and B,

Page 112: Isolation, Characterization and Cosmeceutical Application

References

93

from a sponge-derived Penicillium chrysogenum strain. Tetrahedron.

2005;61(30):7252-65.

105. Cueto M, Jensen PR, Kauffman C, Fenical W, Lobkovsky E, Clardy J.

Pestalone, a new antibiotic produced by a marine fungus in response to bacterial

challenge. Journal of natural products. 2001;64(11):1444-6.

106. Singla A, Verma D, Lal B, Sarma PM. Enrichment and optimization of

anaerobic bacterial mixed culture for conversion of syngas to ethanol. Bioresource

technology. 2014;172:41-9.

107. Arora DS, Kaur GJ. Antibacterial activity of some Indian medicinal plants.

Journal of Natural Medicines. 2007;61(3):313-7.

108. Pandey S, Sree A, Sethi DP, Kumar CG, Kakollu S, Chowdhury L, et al. A

marine sponge associated strain of Bacillus subtilis and other marine bacteria can

produce anticholinesterase compounds. Microbial Cell Factories. 2014;13(1):1.

109. Perez C, Pauli M, Bazerque P. An antibiotic assay by the agar well diffusion

method. Acta Biol Med Exp. 1990;15:113-5.

110. Clarke G, Ting KN, Wiart C, Fry J. High correlation of 2, 2-diphenyl-1-

picrylhydrazyl (DPPH) radical scavenging, ferric reducing activity potential and total

phenolics content indicates redundancy in use of all three assays to screen for

antioxidant activity of extracts of plants from the malaysian rainforest. Antioxidants.

2013;2(1):1-10.

111. Silva DM, Batista LR, Rezende EF, Fungaro MHP, Sartori D, Alves E.

Identification of fungi of the genus Aspergillus section Nigri using polyphasic

taxonomy. Brazilian Journal of Microbiology. 2011;42(2):761-73.

112. White TJ, Bruns T, Lee S, Taylor J. Amplification and direct sequencing of

fungal ribosomal RNA genes for phylogenetics. PCR protocols: a guide to methods

and applications. 1990;18(1):315-22.

113. Mishra KN, Aaggarwal A, Abdelhadi E, Srivastava D. An efficient horizontal

and vertical method for online dna sequence compression. International Journal of

Computer Applications. 2010;3(1):39-46.

114. Kumar S, Tamura K, Nei M. MEGA: molecular evolutionary genetics

analysis software for microcomputers. Computer Applications in the Biosciences.

1994;10(2):189-91.

115. Le Dang Q, Shin TS, Park MS, Choi YH, Choi GJ, Jang KS, et al.

Antimicrobial activities of novel mannosyl lipids isolated from the biocontrol fungus

Page 113: Isolation, Characterization and Cosmeceutical Application

References

94

Simplicillium lamellicola BCP against phytopathogenic bacteria. Journal of

Agricultural and Food Chemistry. 2014;62(15):3363-70.

116. Gonçalves VN, Carvalho CR, Johann S, Mendes G, Alves TM, Zani CL, et

al. Antibacterial, antifungal and antiprotozoal activities of fungal communities

present in different substrates from Antarctica. Polar Biology. 2015;38(8):1143-52.

117. Pinheiro Â, Dethoup T, Bessa J, Silva AM, Kijjoa A. A new bicyclic

sesquiterpene from the marine sponge associated fungus Emericellopsis minima.

Phytochemistry Letters. 2012;5(1):68-70.

118. Chen T, Lam C-K, Chen W-D, Chen X-H, Feng G-K, Zhu X-F, et al. NMR

screening approach for discovery of new 6-methylpyridinone derivatives from the

marine-derived fungus Leptosphaerulina sp. Arabian Journal of Chemistry. 2015.

119. Amagata T, Amagata A, Tenney K, Valeriote FA, Lobkovsky E, Clardy J, et

al. Unusual C25 Steroids Produced by a Sponge-Derived Penicillium citrinum.

Organic Letters. 2003;5(23):4393-6.

120. Ueda J-y, Hashimoto J, Inaba S, Takagi M, Shin-ya K. JBIR-59, a new

sorbicillinoid, from a marine-derived fungus Penicillium citrinum SpI080624G1f01.

Journal of Antibiotics. 2010;63(4):203.

121. Kawahara T, Takagi M, Shin-ya K. JBIR-124: a novel antioxidative agent

from a marine sponge-derived fungus Penicillium citrinum SpI080624G1f01. Journal

of Antibiotics. 2012;65(1):45.

122. Khamthong N, Rukachaisirikul V, Phongpaichit S, Preedanon S, Sakayaroj J.

Bioactive polyketides from the sea fan-derived fungus Penicillium citrinum PSU-

F51. Tetrahedron. 2012;68(39):8245-50.

123. Sun Y-L, Zhang X-Y, Zheng Z-H, Xu X-Y, Qi S-H. Three new polyketides

from marine-derived fungus Penicillium citrinum SCSGAF 0167. Natural product

research. 2014;28(4):239-44.

124. Tsuda M, Sasaki M, Mugishima T, Komatsu K, Sone T, Tanaka M, et al.

Scalusamides AC, New Pyrrolidine Alkaloids from the Marine-Derived Fungus

Penicillium citrinum. Journal of natural products. 2005;68(2):273-6.

125. An CY, Li XM, Li CS, Gao SS, Shang Z, Wang BG. Triazoles and Other N‐

Containing Metabolites from the Marine‐Derived Endophytic Fungus Penicillium

chrysogenum EN‐118. Helvetica Chimica Acta. 2013;96(4):682-7.

Page 114: Isolation, Characterization and Cosmeceutical Application

References

95

126. Devi P, Rodrigues C, Naik C, D‟souza L. Isolation and characterization of

antibacterial compound from a mangrove-endophytic fungus, Penicillium

chrysogenum MTCC 5108. Indian journal of microbiology. 2012;52(4):617-23.

127. Visamsetti A, Ramachandran SS, Kandasamy D. Penicillium chrysogenum

DSOA associated with marine sponge (Tedania anhelans) exhibit antimycobacterial

activity. Microbiological research. 2016;185:55-60.

128. Suwannarangsee S, Arnthong J, Eurwilaichitr L, Champreda V. Production

and characterization of multipolysaccharide degrading enzymes from Aspergillus

aculeatus BCC199 for saccharification of agricultural residues. Journal of

Microbiology and Biotechnology. 2014;24(10):1427-37.

129. Qiao M-F, Ji N-Y, Liu X-H, Li K, Zhu Q-M, Xue Q-Z. Indoloditerpenes from

an algicolous isolate of Aspergillus oryzae. Bioorganic & medicinal chemistry letters.

2010;20(19):5677-80.

130. Qiao M-F, Ji N-Y, Liu X-H, Li F, Xue Q-Z. Asporyergosterol, a new steroid

from an algicolous isolate of Aspergillus oryzae. Natural Product Communications.

2010;5(10):1575-8.

131. Liu X, Song F, Ma L, Chen C, Xiao X, Ren B, et al. Sydowiols A–C:

Mycobacterium tuberculosis protein tyrosine phosphatase inhibitors from an East

China Sea marine-derived fungus, Aspergillus sydowii. Tetrahedron Letters.

2013;54(45):6081-3.

132. Teuscher F, Lin W, Wray V, Edrada RA, Padmakumar K, Proksch P, et al.

Two new cyclopentanoids from the endophytic fungus Aspergillus sydowii associated

with the marine alga Acanthophora spicifera. Natural Product Communications.

2006;1(11):927-33.

133. Matsumura Y, Ananthaswamy HN. Toxic effects of ultraviolet radiation on

the skin. Toxicology and Applied Pharmacology. 2004;195(3):298-308.

134. Simon JD, Peles D, Wakamatsu K, Ito S. Current challenges in understanding

melanogenesis: bridging chemistry, biological control, morphology, and function.

Pigment Cell & Melanoma Research. 2009;22(5):563-79.

135. Briganti S, Camera E, Picardo M. Chemical and instrumental approaches to

treat hyperpigmentation. Pigment Cell Research. 2003;16(2):101-10.

136. Petit L, Pierard G. Skin‐lightening products revisited. International journal of

cosmetic science. 2003;25(4):169-81.

Page 115: Isolation, Characterization and Cosmeceutical Application

References

96

137. Scott MC, Suzuki I, Abdel‐Malek ZA. Regulation of the human melanocortin

1 receptor expression in epidermal melanocytes by paracrine and endocrine factors

and by ultraviolet radiation. Pigment Cell Research. 2002;15(6):433-9.

138. Hwang J-H, Lee BM. Inhibitory effects of plant extracts on tyrosinase, L-

DOPA oxidation, and melanin synthesis. Journal of Toxicology and Environmental

Health, Part A. 2007;70(5):393-407.

139. Karioti A, Protopappa A, Megoulas N, Skaltsa H. Identification of tyrosinase

inhibitors from Marrubium velutinum and Marrubium cylleneum. Bioorganic &

Medicinal Chemistry. 2007;15(7):2708-14.

140. Parvez S, Kang M, Chung HS, Bae H. Naturally occurring tyrosinase

inhibitors: mechanism and applications in skin health, cosmetics and agriculture

industries. Phytotherapy Research. 2007;21(9):805-16.

141. Kim Y-J, Uyama H. Tyrosinase inhibitors from natural and synthetic sources:

structure, inhibition mechanism and perspective for the future. Cellular and

Molecular Life Sciences 2005;62(15):1707-23.

142. Battaini G, Monzani E, Casella L, Santagostini L, Pagliarin R. Inhibition of

the catecholase activity of biomimetic dinuclear copper complexes by kojic acid.

Journal of Biological Inorganic Chemistry. 2000;5(2):262-8.

143. Nakagawa M, Kawai K, Kawai K. Contact allergy to kojic acid in skin care

products. Contact Dermatitis. 1995;32(1):9-13.

144. Takizawa T, Mitsumori K, Tamura T, Nasu M, Ueda M, Imai T, et al.

Hepatocellular tumor induction in heterozygous p53-deficient CBA mice by a 26-

week dietary administration of kojic acid. Toxicological Sciences. 2003;73(2):287-

93.

145. Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M, Telser J. Free

radicals and antioxidants in normal physiological functions and human disease. The

International Journal of Biochemistry & Cell Biology. 2007;39(1):44-84.

146. Hekimi S, Lapointe J, Wen Y. Taking a “good” look at free radicals in the

aging process. Trends in Cell Biology. 2011;21(10):569-76.

147. Tiwari AK. Imbalance in antioxidant defence and human diseases: Multiple

approach of natural antioxidants therapy. CURRENT SCIENCE-BANGALORE-.

2001;81(9):1179-87.

148. Blunt JW, Copp BR, Keyzers RA, Munro MH, Prinsep MR. Marine natural

products. Natural product reports. 2014;31(2):160-258.

Page 116: Isolation, Characterization and Cosmeceutical Application

References

97

149. Mayer A, Rodríguez AD, Taglialatela-Scafati O, Fusetani N. Marine

pharmacology in 2009–2011: Marine compounds with antibacterial, antidiabetic,

antifungal, anti-inflammatory, antiprotozoal, antituberculosis, and antiviral activities;

affecting the immune and nervous systems, and other miscellaneous mechanisms of

action. Marine drugs. 2013;11(7):2510-73.

150. Wang C, Mao W, Chen Z, Zhu W, Chen Y, Zhao C, et al. Purification,

structural characterization and antioxidant property of an extracellular polysaccharide

from Aspergillus terreus. Process Biochemistry. 2013;48(9):1395-401.

151. Zhu T, Chen Z, Liu P, Wang Y, Xin Z, Zhu W. New rubrolides from the

marine-derived fungus Aspergillus terreus OUCMDZ-1925. Journal of Antibiotics.

2014;67(4):315.

152. Lee SM, Li XF, Jiang H, Cheng JG, Seong S, Choi HD, et al. Terreusinone, a

novel UV-A protecting dipyrroloquinone from the marine algicolous fungus

Aspergillus terreus. Tetrahedron Letters. 2003;44(42):7707-10.

153. Oleinikova GK, Denisenko VA, Berdyshev DV, Pushilin MA, Kirichuk NN,

Menzorova NI, et al. Two new sesterterpenoids, terretonins H and I, from the marine-

derived fungus Aspergillus ustus. Phytochemistry Letters. 2016;17:135-9.

154. You M, Liao L, Hong SH, Park W, Kwon DI, Lee J, et al. Lumazine Peptides

from the Marine-Derived Fungus Aspergillus terreus. Marine drugs.

2015;13(3):1290-303.

155. Yin Y, Gao Q, Zhang F, Li Z. Medium optimization for the high yield

production of single (+)-terrein by Aspergillus terreus strain PF26 derived from

marine sponge Phakellia fusca. Process Biochemistry. 2012;47(5):887-91.

156. Liang X, Zhang X-Y, Nong X-H, Wang J, Huang Z-H, Qi S-H. Eight linear

peptides from the deep-sea-derived fungus Simplicillium obclavatum EIODSF 020.

Tetrahedron. 2016;72(22):3092-7.

157. Yan B-F, Fang S-T, Li W-Z, Liu S-J, Wang J-H, Xia C-H. A new minor

diketopiperazine from the sponge-derived fungus Simplicillium sp. YZ-11. Natural

product research. 2015;29(21):2013-7.

158. Fukuda T, Sudoh Y, Tsuchiya Y, Okuda T, Igarashi Y. Isolation and

biosynthesis of preussin B, a pyrrolidine alkaloid from Simplicillium lanosoniveum.

Journal of natural products. 2014;77(4):813-7.

Page 117: Isolation, Characterization and Cosmeceutical Application

References

98

159. Takata K, Iwatsuki M, Yamamoto T, Shirahata T, Nonaka K, Masuma R, et

al. Aogacillins A and B produced by Simplicillium sp. FKI-5985: new circumventors

of arbekacin resistance in MRSA. Organic Letters. 2013;15(18):4678-81.

160. Chen Z, Song Y, Chen Y, Huang H, Zhang W, Ju J. Cyclic Heptapeptides,

Cordyheptapeptides C–E, from the Marine-Derived Fungus Acremonium persicinum

SCSIO 115 and Their Cytotoxic Activities. Journal of natural products.

2012;75(6):1215-9.

161. Bharti P, Anand V, Chander J, Singh IP, Singh TV, Tewari R. Heat stable

antimicrobial activity of Burkholderia gladioli OR1 against clinical drug resistant

isolates. The Indian Journal of Medical Research. 2012;135(5):666.

162. Khurram M, Khan MA, Hameed A, Abbas N, Qayum A, Inayat H.

Antibacterial activities of Dodonaea viscosa using contact bioautography technique.

Molecules. 2009;14(3):1332-41.