You are on page 1of 182

Ratan Singh Ray · Chandana Haldar

Ashish Dwivedi · Neeraj Agarwal


Jyoti Singh Editors

Photocarcinogenesis
& Photoprotection
Photocarcinogenesis & Photoprotection
Ratan Singh Ray
Chandana Haldar • Ashish Dwivedi
Neeraj Agarwal • Jyoti Singh
Editors

Photocarcinogenesis &
Photoprotection
Editors
Ratan Singh Ray Chandana Haldar
Indian Institute of Toxicology Research Department of Zoology
Lucknow, Uttar Pradesh, India Banaras Hindu University
Varanasi, Uttar Pradesh, India
Ashish Dwivedi
Department of Zoology Neeraj Agarwal
Banaras Hindu University Anschutz Medical Campus
Varanasi, Uttar Pradesh, India University of Colorado Denver
Aurora, CO, USA
Jyoti Singh
Photobiology Division
Indian Institute of Toxicology Research
Lucknow, Uttar Pradesh, India

ISBN 978-981-10-5492-1    ISBN 978-981-10-5493-8 (eBook)


https://doi.org/10.1007/978-981-10-5493-8

Library of Congress Control Number: 2018959229

© Springer Nature Singapore Pte Ltd. 2018


This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of
the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation,
broadcasting, reproduction on microfilms or in any other physical way, and transmission or information
storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology
now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the
editors give a warranty, express or implied, with respect to the material contained herein or for any errors
or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims
in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Preface

It is our privilege and honor to write the first edition of book on unique subject of
Photocarcinogenesis and Photoprotection.
Incidences of skin cancer are increasing globally which is caused mainly by
repeated and increased exposure to ultraviolet or sunlight through increased outdoor
activities, changes in clothing style, increased longevity, ozone depletion, genetics
and immune suppression. This prompt us to educate others by putting together
information on ultraviolet radiation exposure, how it initiates cancer, mechanisms
involved, potential therapy, and what protective measures can be taken. This book
also covers how commonly used therapeutic drugs, personal care products, and
other chemicals when consumed or applied to the body can be toxic under exposure
to UV radiation.
Although there is an increasing awareness about harmful effects of UV radiation,
exposure to UVR is still unavoidable, and people need to be educated on how UV
radiation causes its harmful effects and what protective measures can be taken.
All the authors in this book are highly skilled researchers with thorough and up-­
to-­date knowledge in the field and are actively doing research in the relevant fields.
For all the chapters, we put our best efforts to provide collated information from
research articles published so far related to the subject and arranged them in subsec-
tions to make them easy to follow. We have tried our best to use the simplified lan-
guage so that it can be understandable to all levels of readers.
We hope that after reading the book, the reader will have more knowledge,
understanding, and awareness about the harmful effects of repetitive or chronic UV
radiation exposure as well as its interactions with other chemicals and drugs on the
skin or inside the body.

Lucknow, Uttar Pradesh, India Ratan Singh Ray


Varanasi, Uttar Pradesh, India Chandana Haldar
Varanasi, Uttar Pradesh, India Ashish Dwivedi
Aurora, CO, USA Neeraj Agarwal
Lucknow, Uttar Pradesh, India Jyoti Singh

v
Acknowledgments

I am heartily thankful to all the authors for investing time and efforts in writing the
chapters for the book. I highly appreciate that they took extra stress besides their
other commitments to provide detailed information based on their experience and
from literature to make all the chapters in this book entitled Photocarcinogenesis
and Photoprotection up-to-date and more valuable for its readers. We also like to
take this opportunity to convey our special thanks to Prof. Alok Dhawan, Director
of CSIR-IITR, Lucknow, for his support, guidance, and valuable suggestions
throughout the preparation of this new edition.

vii
Contents

1 Ultraviolet Radiation (UVR): An Introduction ��������������������������������������   1


Ashish Dwivedi, Amit Kumar Tripathi, Jyoti Singh,
and Manish Kumar Pal
2 UVR-Induced Epigenetic Regulation and Photocarcinogenesis������������   9
Neera Yadav, Amit Kumar Tripathi, and Monisha Banerjee
3 Molecular and Genetic Response of Human Skin Under
Ultraviolet Radiation �������������������������������������������������������������������������������� 15
Neera Yadav and Monisha Banerjee
4 Photocarcinogenesis and Molecular Mechanism������������������������������������ 29
Neeraj Agarwal
5 Immunomodulation and Photocarcinogenesis���������������������������������������� 45
Neeraj Agarwal
6 Epidemiological Aspects of Photocarcinogenesis������������������������������������ 53
Neera Yadav and Monisha Banerjee
7 Photoaging�������������������������������������������������������������������������������������������������� 65
Jyoti Singh, Deepti Chopra, Ashish Dwivedi, and Ratan Singh Ray
8 Drug-Induced Phototoxic Response �������������������������������������������������������� 77
Syed Faiz Mujtaba, Ajeet K. Srivastav, Shikha Agnihotry,
and Mohammad Anas
9 PAHs and Phototoxicity���������������������������������������������������������������������������� 85
Ajeet K. Srivastav, Shikha Agnihotry, Syed Faiz Mujtaba,
Sandeep Negi, Ankit Verma, and Ratan Singh Ray
10 Phototoxicity of Hair Dyes: Challenge for Tropical Countries�������������� 101
Shruti Goyal, Ajeet Kumar Srivastav, Saroj K. Amar,
Shikha Agnihotry, and Ratan Singh Ray

ix
x Contents

11 Role of Personal Care Products and Phototoxicity�������������������������������� 109


Syed Faiz Mujtaba, Ajeet K. Srivastav, Shikha Agnihotry,
Sandeep Negi, Jaya Upadhayay, and Ratan S. Ray
12 Protective Role of Phytochemicals Against UVR������������������������������������ 129
Deepti Chopra, Jyoti Singh, Ajeet Kumar Srivastav, Divya Dubey,
Ratan Singh Ray, and Kailash Chand Gupta
13 Role of Nanotechnology in Skin Remedies���������������������������������������������� 141
Lipika Ray and K. C. Gupta
14 Role of Photodynamic Therapy in Cancer Treatment���������������������������� 159
Shikha Agnihotry, Mohammad Anas, Ajeet K. Srivastav,
Deepti Chopra, Jaya Upadhayay, and Syed Faiz Mujtaba
Contributors

Neeraj Agarwal Urology Division, Department of Surgery, Anschutz Medical


Campus, University of Colorado Denver, Aurora, CO, USA
Shikha Agnihotry Department of Biomedical-informatics, Sanjay Gandhi Post
Graduate Institute, Lucknow, India
Saroj K. Amar Department of Forensic Science, School of Bioengineering and
Biosciences, Lovely Professional University, Phagwara, Punjab, India
Mohammad Anas Photobiology Division, CSIR-Indian Institute of Toxicology
Research, Lucknow, Uttar Pradesh, India
Monisha Banerjee Molecular and Human Genetics Laboratory, Department of
Zoology, University of Lucknow, Lucknow, Uttar Pradesh, India
Deepti Chopra Photobiology Laboratory, Systems Toxicology and Health Risk
Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow,
Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
Divya Dubey Photobiology Laboratory, System Toxicology and Health Risk
Assessment Group, CSIR- Indian Institute of Toxicology Research, Lucknow, Uttar
Pradesh, India
Babu Banarasi Das University, Lucknow, India
Ashish Dwivedi Pineal Research Lab, Department of Zoology, Institute of Science,
Banaras Hindu University, Varanasi, Uttar Pradesh, India
Shruti Goyal Photobiology Laboratory, System Toxicology and Health Risk
Assessment Group, CSIR- Indian Institute of Toxicology Research (IITR), Lucknow,
Uttar Pradesh, India
Kailash Chand Gupta CSIR-Institute of Genomics and Integrative Biology,
Delhi, India
Department of Biological Sciences and Bioengineering (BSBE) and Centre for
Environmental Science and Engineering (CESE), Indian Institute of Technology,
Kanpur, India

xi
xii Contributors

Syed Faiz Mujtaba Department of Zoology, Faculty of Science, Shia P.G. College,
University of Lucknow, Lucknow, Uttar Pradesh, India
Sandeep Negi Photobiology Laboratory, System Toxicology and Health Risk
Assessment Group, CSIR- Indian Institute of Toxicology Research (IITR), Lucknow,
Uttar Pradesh, India
Manish Kumar Pal Department of Obstetrics and Gynecology, King George’s
Medical University, Lucknow, Uttar Pradesh, India
Lipika Ray Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug
Research Institute, Lucknow, Uttar Pradesh, India
Ratan Singh Ray Photobiology Division, CSIR- Indian Institute of Toxicology
Research (IITR), Lucknow, Uttar Pradesh, India
Department of Zoology, Faculty of Science, Shia P.G. College, Lucknow, India
CSIR-Institute of Genomics and Integrative Biology, Delhi, India
Department of Biological Sciences and Bioengineering (BSBE) and Centre for
Environmental Science and Engineering (CESE), Indian Institute of Technology,
Kanpur, India
Jyoti Singh Photobiology Laboratory, Systems Toxicology and Health Risk
Assessments Group, Indian Institute of Toxicology Research, Lucknow,
Uttar Pradesh, India
Academy of Scientific and Innovative Research, New Delhi, India
Ajeet Kumar Srivastav Photobiology Division, System Toxicology and Health
Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow,
Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, India
Amit Kumar Tripathi Electrophysiology Lab, School of Biomedical Engineering,
IIT (BHU), Varanasi, Uttar Pradesh, India
Jaya Upadhayay Babu Banarasi Das University, Lucknow, India
Ankit Verma Photobiology Laboratory, System Toxicology and Health Risk
Assessment Group, CSIR- Indian Institute of Toxicology Research (IITR), Lucknow,
Uttar Pradesh, India
Neera Yadav Molecular and Human Genetics Laboratory, Department of Zoology,
University of Lucknow, Lucknow, Uttar Pradesh, India
About the Editors

Prof. Ratan Singh Ray is a Senior Principal Scientist and Head of Photobiology
Division, CSIR-Indian Institute of Toxicology Research, Lucknow. He is also serv-
ing as a Professor in AcSIR, CSIR. He had joined CSIR-IITR in 1991 as a Scientist.
He has 26 years of research experience in the area of photosciences. Presently, he is
working on photosafety issues of different nanomaterials used in personal care
products and identifying novel nanotized phytochemicals for use in cosmetics, hair
dyes, as well as sunscreen for photosafety against UVR. He is an active member of
various scientific societies including the Bureau of Indian Standards (Cosmetics
Sectional Committee PCD 19), India; American Society for Photobiology, USA;
Indian Photobiology Society, India; and Society of Toxicology, India. He has pub-
lished many research articles in reputed journals of Photochemistry and
Photobiology, Toxicology, Biomaterials, The Journal of Hazardous Materials, etc.

Prof. Chandana Haldar is the Head of the Department of Zoology, Institute of


Science, Banaras Hindu University, Varanasi, India. She has 30 years of research
experience in photosciences. She has edited many books and published more than
200 research papers in the reputed journals of Photosciences. She has been twice
awarded Alexander von Humboldt Fellowship of Germany to work on the steroid
receptors and the pineal gland and immune function (1986–1987 and 1990–1991).
Besides three decades of training, research, and teaching experience, she has also
served as Chairperson and member of various societies, academic councils, and
administrative bodies in and outside Banaras Hindu University.

Dr. Ashish Dwivedi is a Postdoctoral Scientist in the Pineal Research Lab,


Department of Zoology, BHU, Varanasi. He has done his doctoral research from the
Photobiology Division, Indian Institute of Toxicology Research (IITR), Lucknow. His
doctoral research work was focused on phototoxicity assessment of different thera-
peutic drugs and environmental pollutants. He has 10 years of research experience in
photosciences and published many papers in reputed journals of Photochemistry and
Photobiology, Toxicology, Biomaterials, The Journal of Hazardous Materials, etc.

xiii
xiv About the Editors

Dr. Neeraj Agarwal is working as a Research Instructor in NCI-designated


University of Colorado Comprehensive Cancer Center, Aurora, CO, USA. Previously,
he has completed his Ph.D. (2001 to 2007) in photobiology Division of Indian
Institute of Toxicology Research (IITR), Lucknow. His work was focused on deter-
mining the phototoxic potential of commonly used therapeutic drugs and their
mechanism of action. He has published seven research papers and presented his
work at various conferences. He has received Best Poster Award in 2002 for the
work related to ciprofloxacin phototoxicity. In 2007, he joined the Louisiana State
University Health Sciences Center (LSUHSC), New Orleans, USA, as a Postdoctoral
Researcher and worked for 5 years toward understating the role of MTBP protein in
osteosarcoma metastasis and also on identifying and characterizing the cancer stem
cells in osteosarcoma. He has received Scientific Excellence Award and was invited
to present his work in the Cancer Center Retreat in 2010. Till now, he has published
18 research articles in reputed international journals with high impact factors,
including Cancer Research, Oncogene, Cell Death & Differentiation, Clinical
Cancer Research, and Photochemistry and Photobiology.

Jyoti Singh is presently pursuing her doctoral research from the CSIR-Indian
Toxicology Research Institute, Lucknow, under the supervision of Dr. Ratan Singh
Ray, Head of Photobiology Division. She did her master’s degree in forensic sci-
ence from Bundelkhand University, Jhansi. She is a member of the Indian
Photobiology Society, India. She has published a book chapter and research papers
in reputed journals like The International Journal of Biochemistry & Cell Biology,
Toxicology Letters, Biomaterials, The Journal of Hazardous Materials, etc.
Abbreviations

UVR Ultraviolet radiation


EMR Electromagnetic radiations
MED Minimal erythema dose
SED Standard erythemal dose
MBD Methyl-CpG-binding domain proteins
HDACs Histone deacetylases
5mC 5-Methylcytosine
5hmC 5-Hydroxymethylcytosine
TDG Thymine DNA glycosylase
PAs Proanthocyanidins
GTP Green tea polyphenols
EGCG Epigallocatechin-3-gallate
α-MSH α-Melanocyte-stimulating hormone
CREB cAMP response element-binding protein
8-OHdG 8-Hydroxy-2’-deoxyguanosine
CPD Cyclobutane pyrimidine dimers
NBCC Nevoid basal-cell carcinoma
LC Langerhans cells
CHS Contact hypersensitivity
TLR Toll-like receptor
ODS Ozone depleting substances
FDA Food and Drug Administration
MMPs Matrix metalloproteinases
ECM Extracellular matrix
NSAIDs Nonsteroidal anti-inflammatory drugs
PAHs Polycyclic aromatic hydrocarbons
IARC International Agency for Research on Cancer
PDT Photodynamic therapy

xv
Ultraviolet Radiation (UVR):
An Introduction 1
Ashish Dwivedi, Amit Kumar Tripathi, Jyoti Singh,
and Manish Kumar Pal

Abstract
Radiant energy of sun is essential to perform metabolic processes of all flora and
fauna on the earth. The electromagnetic radiations (EMR) emitted by sun extend
from very long wavelength radiation, such as radiowaves (A ″′ 3 × 108 m), to very
short wavelength radiation, such as cosmic rays (A ″′ 3 × 10−19 m). The EMR
reaching at the earth surface contains wavelength from 290 to 4000 nm. However,
the UV portion covers from 200 to 400 nm. The range from 200 to 400 nm is
often arbitrarily categorized into UVA, UVB, and UVC radiation. Solar radiation
less than 290 nm does not reach at the earth’s surface due to the presence of O3
layer in stratospheric zone. But, last from few decades due to anthropogenic
activities, the concentration of ozone layer decreases on stratospheric zone. As a
consequence of that, UVB radiation levels are rising to 1% a year. Thus, the del-
eterious health effects on human beings (skin aging, cataracts, skin cancer, and
immune suppression) are enhanced by UVR.

A. Dwivedi (*)
Pineal Research Lab, Department of Zoology, Institute of Science, Banaras Hindu University,
Varanasi, Uttar Pradesh, India
A. K. Tripathi
Electrophysiology Lab, School of Biomedical Engineering, IIT (BHU),
Varanasi, Uttar Pradesh, India
J. Singh
Photobiology Laboratory, Systems Toxicology and Health Risk Assessments Group
CSIR-Indian Institute of Toxicology Research (CSIR-IITR), MG Marg,
Lucknow, Uttar Pradesh, India
M. K. Pal
Department of Obstetrics and Gynecology, King George’s Medical University,
Lucknow, Uttar Pradesh, India

© Springer Nature Singapore Pte Ltd. 2018 1


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_1
2 A. Dwivedi et al.

Keywords
UVR · EMR · Ozone · Skin cancer · Skin aging

1.1 Introduction

Terrestrial life depends on radiant energy of sun which is essential for metabolic
processes of all the living systems. Without sunlight, the surface of the earth would
be cold and completely lifeless. From the beginning of life, light was an essential
component of man’s life. The word “radiation” originated from the Egyptian sun
god “Atom Ra.” It is depicted as the rays of the sun ending in hands holding the
symbol of life. Sir Isaac Newton, in 1669, found that white light gets separated into
different colors when passing through crude glass prism. Johann Wilhelm Ritter
from Jena in Germany discovered UVR [1]. After 200 years of the discovery of UV
rays, nobody would question the importance of Ritter’s discovery and the conse-
quences of it in photodermatology and photomedicine. In 1845, Bonnet used sun-
light to treat tuberculosis arthritis [2]. Jewish physicians in Arabia recommended
sunbaths for health.
Niels Finsen work established the branch of modern photobiology. He fixed the
role of ultraviolet radiation (UVR) in sunburn [3]. Finsen is the pioneer of helio-
therapy and actinotherapy throughout Europe and the USA. Saleeby in 1926 dis-
cussed the importance of heliotherapy and treatment of surgical tuberculosis by
natural sunlight [4].

1.1.1 Electromagnetic Radiation (EMR)

The interaction of EMR with matter can be described by viewing it either as a con-
tinuous wave (wave description) or as a series of packets containing energy (particle
description). Transmission through space, scattering, and diffraction can be under-
stood using the wave description. EMR is a form of energy and can be characterized
as a continuous wave of regular oscillations of electric and magnetic fields. These
fields are perpendicular to each other and to the direction of propagation. However,
the absorption of EMR by molecule and the photoelectric effect are best understood
when the particle description of EMR is used. In the particle description, EMR is
contained in discrete packets, called photons.
The energy of EMR is directly proportional to the frequency of the oscillation of
the two fields:
E = hv (1.1)
−34
where E = energy of a photon, h = Planck’s constant (6.63 × 10 J.s), and
v = frequency.
1 Ultraviolet Radiation (UVR): An Introduction 3

The product of the frequency and the wavelength is equal to a constant, the speed
of light in a vacuum:
vl = c (1.2)
where λ = wavelength and c = speed of light in vacuum.
Therefore, the energy of EMR is inversely proportional to its wavelength:

hc
E= (1.3)
l
The unit most often used for wavelength in the UV and visible range is the nanome-
ter, which is 1 × 10−9 m. The EMR extends from very long wavelength radiation
(low energy), such as radiowaves (A ″′ 3 × 108 m), to a very short wavelength radia-
tion (high energy), such as cosmic rays (A ″′ 3 × 10−19 m). The EMR reaching at the
earth surface from the sun contains wavelength from 290 to 4000 nm. The radiation
is described as UV, visible, or infrared, depending upon the wavelength. The UV
portion of the EMR covers from 200 to 400 nm. Solar radiation shorter than 290 nm
is absorbed by O3 in the stratosphere and does not reach at the earth’s surface. The
range from 200 to 400 nm is often arbitrarily divided into UVA, UVB, and UVC
radiation. It is based on skin reactions in human. The UVA portion (320–400 nm) is
longer-wavelength UVR; it is not strongly absorbed by proteins and nucleic acids.
It does not cause erythema in normal skin at moderate doses in the absence of pho-
tosensitizing chemicals. This range is also called black light and near-UVR. UVB
radiation (290–320 nm) is erythemogenic and is present in the terrestrial solar spec-
trum. It is also referred as sunburn radiation and midrange UVR. UVC radiation
(200–290 nm) is biologically active but does not reach at the earth’s surface.
However, the 254 nm wavelength in low-pressure mercury lamps (germicidal
lamps) is frequently used in experiments as a source of UVR.
The spectral distribution of solar irradiance, as measured above the earth’s sur-
face, is the infrared (>760 nm) and visible regions accounting for the great majority
of the sun’s emission 52.8% and 30.9%, respectively. Among invisible radiation,
UVA, UVB, and UVC represent 6.3%, 1.5%, and 0.5% of the total solar energy
output received on the earth’s surface, respectively [5]. On earth, the solar spectrum
is truncated in the UV waveband at approximately 290 nm after the absorption of
stratospheric O3. In this region, the middle wave UVR is of particular interest
because of its potential to damage organisms [6]. The biological effectiveness of
UVR increases logarithmically with decreasing wavelength. UVR shorter than
290 nm is absorbed by O2, O3, and water vapor in the upper atmosphere and does
not reach at the earth’s surface.

1.1.2 Nature of Ultraviolet Radiation

On the basis of biological effects, UV spectrum is further subdivided into three


regions. This division of UVR was first performed in the meeting of the Second
4 A. Dwivedi et al.

300 nm 400 nm
100 nm
280nm 315nm

UV-C UV-B UV-A


Shortwave Midwave Longwave

Fig. 1.1 UV regions of solar spectrum

International Congress on Light held during August 1932 at Copenhagen (Fig. 1.1).
It was proposed those three spectral regions as follows:

UVA 400–315 nm
UVB 315–280 nm
UVC 280–100 nm

1.1.3 UV Regions of Solar Spectrum

However few photodermatologists and environmentalists normally define the wave-


length regions of UVR as follows:

UVA 400–320 nm
UVB 320–290 nm
UVC 290–200 nm

1.1.4 Nomenclature and Units

The International Commission on Illumination (CIE) is an organization of 40 coun-


tries. It is devoted to international cooperation and exchange of information among
on all matters relating to light, illumination, color, and color spaces. CIE was estab-
lished in 1913 as a successor to the Commission Internationale de Photométrie. It is
based in Vienna, Austria.
Radiometric units are used to characterize the sources of UVR. The terms dose
(J/m2) and dose rate (W/m2) pertain to the energy and power, respectively [6]. The
radiant energy delivered to a given area in a given time is also referred to as “flu-
ence,” “exposure dose,” and “dose.”
Radiometric calculation: The most frequently radiometric calculation is to deter-
mine the time for which a subject who has been prescribed a certain dose (J/cm2)
1 Ultraviolet Radiation (UVR): An Introduction 5

should be exposed when the radiometer indicates an irradiance in watt/m2. The rela-
tion between three quantities (time, dose, and irradiance) is simplified:

Exposure time ( min ) =


(
Prescribed dose J / m 2 )
(
60 ´ measured irradiance w / m 2 )

1.1.5 Minimum Erythemal Dose

The minimum erythemal dose (MED, J/m2) is defined as the defined as the threshold
dose that produces a noticeable erythema on a previously unexposed skin of an
individual. This is determined by reddening of the skin and depends on many vari-
ables including skin pigmentation thickness of skin and exposure site.

1.1.6 Standard Erythemal Dose

The standard erythemal dose (SED, J/m2) is equivalent to an erythemal radiant


exposure of 100 J/m2. The SED is independent of skin type, and a particular expo-
sure dose in SED may cause erythema in fair skin but none in darker skin. The
global solar UV index was developed to promote public awareness of the risks of
UVR exposure.

1.1.7 UV Index

The UV index is used to aware the general public about UVR intensity. It was
developed by joint effort of World Health Organization, the United Nations
Environment Program, and International Commission on Non-Ionizing Radiation
Protection. Further, it was established by ISO/CIE. The UVI is an international
standard index which depicts the level of solar UV radiation at the earth’s surface.
It ranges from 0 to 11+, and the values are divided into various exposure catego-
ries. Higher index values represent greater potential for harmful effects to the
human skin and eyes (Fig. 1.2).

1.1.8 Sources and Exposure of Solar UVR

Optical radiation from the sun is modified substantially, and two-thirds of the energy
penetrates to ground level. Solar UVR from its path through the earth’s surface is
absorbed and scattered by various constituents of the atmosphere. Air molecules,
particularly oxygen and nitrogen, by aerosol and dust particles and atmospheric pol-
lutants absorb and scattered the UVR. Total solar irradiance varies with altitude.
Clouds attenuate infrared radiation more than UVR.
6 A. Dwivedi et al.

UV-Index
1 2 3 4 5 6 7 8 9 10 11
No
Protection Protection Required Extra Protection Required
Required

Avoid being outside during


Seek Shade During midday hours
midday hours

You can Slip on a shirt ,slop on sunscreen & slap on a


Safely stay Make sure you seek shade
hat
outdoor

Shirt, sunscreen & hat are


must

Fig. 1.2 UV index chart

Table 1.1 Thickness of O3 Position Thickness (Dobson) Thickness (mm)


layer in stratosphere at At poles 310–430 3.10–4.30
normal temperature and
At equatorial line 240–260 2.40–2.60
pressure
Near equatorial 150–200 1.50–2.00
line
At sea level 10–50 0.10–0.50

Stratospheric ozone layer acts like umbrella to control the intensity of UVR on
earth surface. Interestingly, stratosphere O3 layer varies reasonably in thickness
between 2.4 and 2.6 mm at equator and 3.1 and 4.3 mm at poles (Table 1.1). At the
sea level, it is present only in traces (0.02 ppb). This has been progressively depleted
as a result of accumulation of ozone-destroying chemicals in the earth’s atmosphere,
mostly chlorofluorocarbons (CFCs) and hydrochlorofluorocarbons (HCFCs). These
chemicals are mainly used in refrigeration and air conditioning. The Nobel Prize in
Chemistry was awarded in 1995 to the scientists for sounding the alarm about
depletion of the earth’s protective O3 layer. It was won jointly by Mario J. Molina
(MIT, USA), F.S. Rowland (University of California, USA), and Paul Curtzen
(Max-­Planck Institute, Germany).

1.2 Ozone Depletion

CFCs have been released into the atmosphere in sufficient quantities to damage the
O3 layer. The compounds carry chlorine to the stratosphere that acts as a catalyst to
decompose O3 by photolysis. O3 depletion promotes to an increase in UVB on earth
surface [5]. The half-life of the CFCs is between 50 and 400 years in stratosphere.
1 Ultraviolet Radiation (UVR): An Introduction 7

1.2.1 Production of O3 in Upper Atmosphere

O3 is produced in the upper atmosphere due to the reaction between O2 and O. O2


upon absorption of far UVR forms O3 as shown in reactions (i) and (ii). O3 can also
decompose to O2 and O (iii) and maintain a steady-state concentration in the upper
atmosphere. O3 is a remarkably minor but significant constituent of the upper atmo-
sphere. It provides a shield against damaging UVR by absorbing solar radiation. It
becomes an energy reservoir of the upper atmosphere which is responsible for cli-
matic regulation.

UV C (150 nm )
O2 O+O
¾¾¾¾¾¾¾ ® ( i)

O2 + O ¾¾¾ ¾O3¾¾¾® (ii)


UV C ( 260 nm )

O3 O2 + O
¾¾¾¾¾¾¾ ® (iii)

1.2.2 Consequences of O3 Depletion

Ozone layer acts as an umbrella and thereby protects us from the lethal solar UV
radiation. Even a small depletion in stratospheric O3 can pose a major threat to all
living systems. The UVA and visible radiation levels would remain much the
same as at present. A major depletion in O3 thickness will increase the intensity of
UVB and some UVC on the earth’s surface, leading to toxic effects [7]. It has
been observed that UVB radiation levels are rising up to 1% a year [8]. Therefore,
the continuous depletion of the O3 layer has been the driving force behind more
intense research into the effects of UVR on the body’s immune defenses and on
the diseases [9].
Stratospheric O3 absorbs most of the biologically harmful UVR in the wave-
length range below 320 nm [10]. The doses of UVR that reach the surface of the
earth are as follows: 0–90% UVA, 1–10% UVB, and a small fraction (unmeasur-
able) of UVC. Deleterious health effects on human beings are skin aging, cataracts,
immune suppression, and caner [11]. The incidence of non-melanoma skin cancer
would increase by 3% [12] and cataract prevalence by 0.25–0.6% (EPA, 1987) by
1% depletion in the O3 layer.
8 A. Dwivedi et al.

1.2.3 Detection of UVR

Techniques for the measurement of UVR fall into three classes: physical, chemical,
and biological. In general, physical devices measure power, whereas chemical and
biological systems measure energy. Chemical methods generally measure the
chemical changes produced by the radiation, and it is called actinometry. Biological
techniques use viruses and microorganisms for measurement.

1.3 Conclusion

Last from few decades due to ozone depletion and use of artificial tanning, the expo-
sure of UVR enhanced on human beings. Thus, the problem of skin aging, cataracts,
immune suppression, and skin cancer increases day by day. Avoiding the peak hour
exposure and application of sunscreen will be a step to protect against harmful
effect of UVR.

References
1. Frercks J, Weber H, Wiesenfeldt G (2009) Reception and discovery: the nature of Johann
Wilhelm Ritter’s invisible rays. Stud Hist Philos Sci A 40(2):143–156
2. Bonnet A (1845) Traité des maladies des articulations accompagné d'un atlas de 16 planches.
JB & G. Baillière
3. Roelandts R (2005) A new light on Niels Finsen, a century after his nobel prize. Photodermatol
Photoimmunol Photomed 21(3):115–117
4. Saleeby CW (1923) Sunlight and health. Nisbet & Company, Limited
5. Frederick JE, Snell HE, Haywood EK (1989) Solar ultraviolet radiation at the earth’s surface.
Photochem Photobiol 50(4):443–450
6. Jagger J Solar-UV actions on living cells
7. Voytek MA (1990) Addressing the biological effects of decreased ozone on the Antarctic envi-
ronment. Ambio 1:52–61
8. Blumthaler M, Ambach W (1990) Indication of increasing solar ultraviolet-B radiation flux in
alpine regions. Science 248(4952):206–208
9. Coldiron BM (1996) Ozone depletion update. Dermatol Surg 22(3):296–299
10. Niu X, Frederick JE, Stein ML, Tiao GC (1992) Trends in column ozone based on TOMS data:
dependence on month, latitude, and longitude. J Geophys Res Atmos 97(D13):14661–14669
11. Urbach F (1989) Potential effects of altered solar ultraviolet radiation on human skin cancer.
Photochem Photobiol 50(4):507–513
12. Van der Leun JC, Takizawa Y, Longstreth JD (1989) Environmental effects panel report.
United Nation Environmental Program, Nairobi, 64pp
UVR-Induced Epigenetic Regulation
and Photocarcinogenesis 2
Neera Yadav, Amit Kumar Tripathi, and Monisha Banerjee

Abstract
The human skin remains under constant exposure of solar radiation and is vul-
nerable to epigenetic modifications that later may develop skin cancer. The
effects of chronic UVR exposure to skin can alter epigenetic information in epi-
dermal cells, leading to epigenetic mosaicism. These epigenetic changes include
DNA methylation, chromatin modifications, and posttranscriptional modifica-
tions of noncoding RNAs. UVR-induced epigenetic changes are not irreversible.
Certain phytochemicals can potentially inhibit cancer signaling pathways which
are usually deregulated by epigenetic mechanisms. Indeed, recent therapeutic
strategies are directing toward phytochemical-based reversal of epigenetic modi-
fications to combat UVR-induced skin cancers. This chapter provides detailed
insights of different epigenetic alterations, their role in photocarcinogenesis, and
phytochemical-based reversal of epigenetic changes occurring in UVR-irradiated
skin.

Keywords
Ultraviolet · MicroRNA · Histone · DNA methylation · Phytochemicals

N. Yadav · M. Banerjee (*)


Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow,
Lucknow, Uttar Pradesh, India
e-mail: banerjee_m@lkouniv.ac.in
A. K. Tripathi
Electrophysiology Lab, School of Biomedical Engineering, IIT (BHU),
Varanasi, Uttar Pradesh, India

© Springer Nature Singapore Pte Ltd. 2018 9


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_2
10 N. Yadav et al.

2.1 Introduction

Prolonged exposure of UV radiation is a well-recognized etiological factor for skin


cancer development [1]. Extensive studies have been conducted on solar radiation-­
induced photocarcinogenesis of the human skin. However, the mechanisms under-
lying UVR-induced epigenetic changes are not well understood. Epigenetic
responses play a critical role in the process of photocarcinogenesis in the human
skin. UV irradiation leads to DNA damage, induction of oxidative stress, inflamma-
tory responses, and suppression of immune response and induces the epigenetic
modification such as DNA methylation, histone modification, and miRNA deregu-
lation. UVR-mediated chronic inflammation accelerates DNA methylation, an
important epigenetic modification. The DNA methylation is inherited unaltered,
and DNA nucleotide sequence remains unchanged. There are two types of DNA
methylation, i.e., hypomethylation and hypermethylation. These events may either
silence the tumor suppressor genes responsible for carcinogenesis [2] or upregulate
the oncogene expressions or decrease in genomic stability [3–5]. Modification of
histones is another important epigenetic alteration including acetylation, methyla-
tion, phosphorylation, and ubiquitination. Collectively, modifications at the level of
DNA and histones, both play a pivotal role in the silencing of fundamental tumor
suppressor genes that are responsible for the initiation and progression of skin can-
cers [3, 6]. However, epigenetic changes are reversible and can be reversed effec-
tively, by potent phytochemicals.

2.2 Mechanisms of Epigenetic Modification

There are three main mechanisms associated with epigenetic modifications: (1)
DNA methylation, (2) histone modifications, and (3) noncoding RNA-induced post-
transcriptional modification. These mechanisms are vital to normal development
and cell growth.

2.2.1 DNA Methylation

DNA methylation is the most studied epigenetic event occurring at CpG islands [7].
DNA methylation is a process by which methyl moiety is transferred to cytosine
bases in CpG dinucleotides at CpG islands. The process is mediated by DNA meth-
yltransferases (DNMTs) which catalyze the transfer of a methyl group from
S-adenosyl-methionine (SAM) to cytosine to form 5-methylcytosine at CpG islands.
However, in undifferentiated normal cells, most of the CpG islands usually remain
unmethylated. These unmethylated CpG islands have an open structure and coordi-
nation with the adjacent transcriptional promoter, leading to the transcriptional acti-
vation of genes. Three different DNMTs (DNMT1, DNMT3A, and DNMT3B) are
needed for DNA methylation to occur. DNMT1 is known as maintenance methylase
and is responsible for the harmony of established ornament of DNA methylation,
2 UVR-Induced Epigenetic Regulation and Photocarcinogenesis 11

while DNMT3A and DNMT3B are known as de novo methylases and are respon-
sible for beginning of new or de novo DNA methylation [8, 9].
The DNA hypermethylation at CpG islands contributes to silencing of tumor
suppressor genes in UV-exposed skin that may later develop skin cancer.
Hypermethylation of several tumor suppressor genes including P16INK4a,
RASSF1A, and CDH1 has been found downregulated in UV-exposed skin [10].

2.3 Histone Modifications

The chromatin structure can be regulated through histone modifications after UV


irradiation, which provides different levels of accessibility to transcription factors
[11]. Histone acetylation and methylation are well-characterized epigenetic markers
[12]. Histone modifications primarily occur at arginine, lysine, and serine residues
of the amino-terminal tails by posttranscriptional modifications such as methyla-
tion, acetylation, ubiquitination, phosphorylation, and sumoylation [12, 13].
Deacetylation and methylation of H3-Lys9 are the most common histone modifica-
tions that lead to epigenetic repression of genes [14]. Each modification is catalyzed
by different enzymes, including histone acetyltransferases (HATs), deacetylation by
histone deacetylases (HDACs), methylation of lysine and arginine by histone meth-
yltransferases (HMTs), demethylation of lysine residues by histone demethylases
(DMTs), and phosphorylation of specific serine groups by histone kinases (HKs)
[15]. Abnormal augmentation of HDAC and HAT activities may trigger carcinogen-
esis. Methylation of histone is known to be associated with transcriptionally active
chromatin [16]. Tri-methylation at H3-K4, H3-K36, or H3-K79 results into opening
of chromatin organization which stimulates a high level of histone acetylation.
These epigenetic markers can be removed by histone deacetylases leading to tran-
scriptional repression of chromatin. UVR-induced ubiquitination of histone H2A is
dependent on the H3–H4 chaperone CAF1 (chromatin assembly factor-1) and
requires ATR (ataxia telangiectasia and Rad3-related), NER (nucleotide excision
repair), and several other DDR (DNA damage response) factors. UVR-induced
DNA damage triggers CHK1 kinase (checkpoint kinase 1) dissociation from chro-
matin, and dephosphorylation of H3T11P resulting into repression of cyclin B1 and
Cdk1 genes [17].

2.4 Noncoding RNAs in Skin Cancer

The noncoding RNAs (ncRNAs) are involved in regulation of epigenetic mRNA


expressions associated with development of cancer [18]. Based on size, ncRNAs are
of two main types, i.e., small noncoding RNA (ncRNA) (200 bp) and long noncod-
ing RNA (lncRNA) (>200 bp). Small ncRNAs are further categorized into
microRNA (miRNA), Piwi-interacting RNA (piRNA), and small nucleolar RNA
(snoRNA). miRNA expression plays a critical role in cancer and some other dis-
eases like diabetes and neurodegenerative diseases. However, UVR-mediated
12 N. Yadav et al.

Fig. 2.1 UV-induced epigenetic alterations

expression of ncRNAs in carcinogenesis is not well characterized. The expression


of several miRNAs is altered in UV-irradiated human keratinocytes [19]. UV irra-
diation downregulates miRNA in melanocytes susceptible to melanoma, which may
lead to tumor progression [20]. miR-203 plays an important role in regulation of
c-jun signaling in rat model of cerebral ischemia and reperfusion injury. It is inter-
esting to know that UVR-induced miRNA antagomirs have been developed that can
restore the miRNA inhibitory activity on brief UV exposure [21] (Fig. 2.1).

2.5 Conclusion

Solar UV radiation is an important factor for epigenetic alterations that lead to skin
carcinogenesis. Epigenetic alterations including DNA methylation, histone modifi-
cations, and ncRNAs are crucial to photocarcinogenesis. UVR-induced epigenetic
alterations are reversible and can be effectively reversed by using small bioactive
dietary phytochemicals.

References
1. O’Shaughnessy JA, Kelloff GJ, Gordon GB, Dannenberg AJ, Hong WK, Fabian CJ, Sigman
CC, Bertagnolli MM, Stratton SP, Lam S, Nelson WG (2002) Treatment and prevention of
intraepithelial neoplasia: an important target for accelerated new agent development: recom-
mendations of the American Association for Cancer Research Task Force on the Treatment and
Prevention of Intraepithelial Neoplasia. Clin Cancer Res 8(2):314–346
2. Jones PA, Baylin SB (2002) The fundamental role of epigenetic events in cancer. Nat Rev
Genet 3(6):415
2 UVR-Induced Epigenetic Regulation and Photocarcinogenesis 13

3. Laird PW, Jaenisch R (1996) The role of DNA methylation in cancer genetics and epigenetics.
Annu Rev Genet 30(1):441–464
4. Baylin SB, Herman JG (2000) DNA hypermethylation in tumorigenesis: epigenetics joins
genetics. Trends Genet 16(4):168–174
5. Goodman JI, Watson RE (2002) Altered DNA methylation: a secondary mechanism involved
in carcinogenesis. Annu Rev Pharmacol Toxicol 42(1):501–525
6. Housman TS, Feldman SR, Williford PM, Fleischer AB, Goldman ND, Acostamadiedo JM,
Chen GJ (2003) Skin cancer is among the most costly of all cancers to treat for the Medicare
population. J Am Acad Dermatol 48(3):425–429
7. Bird AP (1986) CpG-rich islands and the function of DNA methylation. Nature
321(6067):209–213
8. Egger G, Liang G, Aparicio A, Jones PA (2004) Epigenetics in human disease and prospects
for epigenetic therapy. Nature 429(6990):457
9. Jones PA, Laird PW (1999) Cancer-epigenetics comes of age. Nat Genet 21(2):163
10. Nandakumar V, Vaid M, Katiyar SK (2011) (−)-Epigallocatechin-3-gallate reactivates silenced
tumor suppressor genes, Cip1/p21 and p 16 INK4a, by reducing DNA methylation and increas-
ing histones acetylation in human skin cancer cells. Carcinogenesis 32(4):537–544
11. Ma H, Baumann CT, Li H, Strahl BD, Rice R, Jelinek MA, Aswad DW, Allis CD, Hager GL,
Stallcup MR (2001) Hormone-dependent, CARM1-directed, arginine-specific methylation of
histone H3 on a steroid-regulated promoter. Curr Biol 11(24):1981–1985
12. Berger JR (2007) Progressive multifocal leukoencephalopathy. Handb Clin Neurol 85:169–183
13. Kouzarides T (2007) Histone-modifying enzymes. Cell 128:693
14. Druesne N, Pagniez A, Mayeur C, Thomas M, Cherbuy C, DuÈe PH, Martel P, Chaumontet C
(2004) Diallyl disulfide (DADS) increases histone acetylation and p21 waf1/cip1 expression
in human colon tumor cell lines. Carcinogenesis 25(7):1227–1236
15. Choudhuri S, Cui Y, Klaassen CD (2010) Molecular targets of epigenetic regulation and effec-
tors of environmental influences. Toxicol Appl Pharmacol 245(3):378–393
16. Nakayama JI, Rice JC, Strahl BD, Allis CD, Grewal SI (2001) Role of histone H3 lysine 9
methylation in epigenetic control of heterochromatin assembly. Science 292(5514):110–113
17. Shimada M, Haruta M, Niida H, Sawamoto K, Nakanishi M (2010) Protein phosphatase 1γ
is responsible for dephosphorylation of histone H3 at Thr 11 after DNA damage. EMBO Rep
11(11):883–889
18. Hauptman N, Glavac D (2013) MicroRNAs and long non-coding RNAs: prospects in diagnos-
tics and therapy of cancer. Radiol Oncol 47(4):311–318
19. Dziunycz P, Iotzova-Weiss G, Eloranta JJ, Läuchli S, Hafner J, French LE, Hofbauer GF
(2010) Squamous cell carcinoma of the skin shows a distinct microRNA profile modulated by
UV radiation. J Investig Dermatol 130(11):2686
20. Sha J, Gastman BR, Morris N, Mesinkovska NA, Baron ED, Cooper KD, McCormick T,
Arbesman J, Harter ML (2016) The response of microRNAs to solar UVR in skin-resident mela-
nocytes differs between melanoma patients and healthy persons. PLoS One 11(5):e0154915
21. Connelly CM, Thomas M, Deiters A (2012) High-throughput luciferase reporter assay for
small-molecule inhibitors of microRNA function. J Biomol Screen 17(6):822–828
Molecular and Genetic Response
of Human Skin Under Ultraviolet 3
Radiation

Neera Yadav and Monisha Banerjee

Abstract
Ultraviolet (UV) radiation is recognized as an essential risk factor due to its dual
role of affecting the human skin. Primarily, it is required for natural vitamin D
synthesis in the skin which is indispensable for human health in many construc-
tive ways. On the other hand, UV radiation acts as a non-specific damaging agent
and a mutagen as well. UV radiation has potential to cause both cancer initiation
and progression. Excessive and repeated exposure to UV is associated with
health risks, including pigment changes, wrinkle formation, atrophy, and malig-
nancy. Epidemiologically and molecularly UV is linked to DNA damage, either
directly or indirectly via oxidative injury resulting in various types of skin can-
cer. Genetic factors also stimulate threat of UV-mediated skin anomalies. This
chapter emphasizes on genetic and molecular mechanisms of pigmentation, tan-
ning, DNA damage and repair, Melanocortin 1 receptor (MC1R) gene expres-
sion, photoproduct formation, and p53 mutation.

Keywords
UV radiation · Pigmentation · MC1R · DNA damage · Photoproduct

3.1 Introduction

The skin is considered as the largest organ of the body and comprises roughly 16%
of body mass. It protects underlying structures from mechanical damage. It is made
up of two tissue layers: epidermis and dermis. The epidermis is the superficial

N. Yadav · M. Banerjee (*)


Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow,
Lucknow, Uttar Pradesh, India
e-mail: banerjee_m@lkouniv.ac.in

© Springer Nature Singapore Pte Ltd. 2018 15


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_3
16 N. Yadav and M. Banerjee

layer of the skin; it consists of epithelial, mesenchymal, glandular, and neurovascu-


lar tissues. The epidermis is the outermost layer of the body and directly interacts
with environment including UV radiation, chemical agents, infectious pathogens,
and many more. Epidermal layer is separated from the dermis by a basement mem-
brane and is composed of stratified squamous epithelium. The epidermis is a thin
layer. There are no blood vessels in the epidermis, and hence, nourishment is gained
by diffusion from capillaries entrenched in the dermis. Major proportion of the epi-
dermis is composed of keratinocyte cells. These cells are so named because they
produce a protein mixture called keratin. Keratinocytes make the epidermal cells
hard and also contribute to ability of the epidermis to reduce water loss and resist
abrasion (Fig. 3.1).
Besides keratinocytes, certain other types of cells play vital roles in the epider-
mis. These cells include melanocytes contributing to color of the skin, Langerhans
cells which are part of the immune system, and specialized epidermal cells with
nerve endings called Merkel cells for sensing touch, light, and superficial
pressure.
The human skin, hairs, and eyes can be recognized by different shades in popula-
tions. The color of skin, hair, and eyes is determined by a group of pigments called
melanin. Some parts of the body such as the skin, nipples, freckles, areolae of the
breasts, genitalia, and moles contain large amounts of melanin, whereas other body
parts such as palms of hands, lips, and soles of feet contain less melanin. Though
melanin is found in abundance in epidermal keratinocytes, it is not synthesized in
these cells. Melanin is synthesized in specific cells called melanocytes present in
the basal layer of the epidermis. The process of melanin synthesis is known as
melanogenesis. Melanogenesis proceeds via oxidation and subsequent cyclization
of amino acid tyrosine [1]. Melanogenesis plays a significant regulatory role in the

Fig. 3.1 The skin


3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 17

skin. Synthesized melanin is first deposited in membrane-bound organelles mela-


nosomes and subsequently transported to adjacent keratinocytes through melano-
cytic dendrites. Chemically, melanin occurs in two forms eumelanin and
pheomelanin. The former is a dark brown pigment, while the latter is a light-­colored
pigment responsible for dark color and fair color of individuals, respectively. The
proportion of eumelanin and pheomelanin depends mainly on the duration of our
skin’s exposure to ultraviolet radiation. Indeed, eumelanin synthesis increases with
seasonal UVB [2]. On the one hand, the amount and type of melanin in the epider-
mis determine skin complexion, while on the other hand, melanin absorbs UV radi-
ation and protects underlying tissues and organs from harmful effects of the sun.
Melanin acts as a natural sunscreen of skin and protects it against damaging effects
of UV photons. Fair-skinned people with less eumelanin are much more sensitive to
UV radiation. Therefore, the fairer the skin, the more will be the damage due to UV
exposure. Fair-skinned people have high risk of skin cancer than dark-skinned peo-
ple. Indeed, the levels of pheomelanin are comparable in both dark-skinned and
light-skinned individuals. However, skin color, UV sensitivity, and cancer risk
depend on eumelanin content of the epidermis [3]. Melanocytes are the merely
source of skin pigmentation; therefore, any genetic defect in melanocytes leads to
inherited pigmentary defects like albinism. Melanin also regulates epidermal
homeostasis, free radical scavenging, and possibly antimicrobial activity.
Since the formation of earth, sun has contributed vital role in the evolution of
life, and for most humans, solar energy is a mixed blessing. Although UV radiation
is only a small fraction of solar radiation, it is known for majority of its biological
activity on earth. It can be divided into three main components based on wavelength
range: UVC (100–280 nm), UVB (280–320 nm), and UVA (320–400 nm). The
component of UV light that reaches surface of the earth consists of 90–95% UVA
and 5–10% UVB. Although UVB radiation is about 20-folds less plentiful than
UVA, it is absorbed more efficiently by the biomolecules of the cellular system and
is able to induce damages at considerably lower doses than UVA (Fig. 3.2). UVC is
absorbed by the ozone layer of stratosphere and does not reach the earth’s surface
[4]. Usually, UV dose experienced on the earth varies in different regions of the
atmosphere through which it traverses. UV doses are higher toward equator since
sunlight strikes the earth with greatest intensity at equator. People living at equato-
rial places usually experience ample ambient UV doses as compared to people liv-
ing at other places. These people perform more occupational outdoor activities,
have more chances of interaction with ambient sunlight, and need less clothing to
wear [5]. It must be noted that besides natural source, i.e., solar light, there are
countless artificial sources of UVA and UVB radiation. Artificially, UVC is gener-
ated by lasers, germicidal lamps, and welding lamps [6]. Furthermore, UVA pene-
trates deeper into the skin, whereas UVC do not enter to deeper layers of the skin.
UVB can damage DNA directly, since it is primarily absorbed by nucleic acid chro-
mophores DNA and RNA (maximum absorption at 260 nm).
18 N. Yadav and M. Banerjee

UVC

UVB

UVA
Atmosphere

Keratinocyte

Melanocyte Epidermis

Fibroblast
Dermis

Effect of UV radiation on skin


UVB UVA
- Sunburn - Premature Aging
- Inflammation - Indirect DNA
- Direct DNA Damage
Damage - Oxidative Stress
- Eye Damage - Skin Cancer

Fig. 3.2 Effects of UV radiation on the human skin

3.2 Effects of Ultraviolet Radiation on Skin

UVR has both beneficial and damaging effects. The biologically relevant wave-
length consists of visible and UV radiation. The UV part of electromagnetic radia-
tion (EMR) ranges from 200 to 400 nm. UVR is essentially required for the
production of vitamin D in the human skin. Natural selection also promoted vitamin
D synthesis depending on seasonally variable UVB levels. UV effects physiological
functions of our skin and its components with some acute and delayed conse-
quences. UV radiation depending on dose and wavelength can affect cellular
homeostasis, expression of growth factors/cytokines as well as their receptors. It
can also affect DNA integrity, mutations in tumor suppressor genes, and oncogenes
[7]. UVB may induce various inflammatory responses in the skin by activation of
cascade of cytokines and certain neuroactive and vasoactive mediators which may
result in sunburn. UVR accelerates skin aging (i.e., photoaging) and photocarcino-
genesis. UV radiation can eventually cause various types of UV-induced skin can-
cers, carcinogenesis, and melanomas on repeated exposure to highly intense sunlight
[8]. Keratinocytes activate apoptotic pathways and ultimately die when the dose of
UV radiation exceeds a threshold level. Like all apoptotic cells, apoptotic
3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 19

keratinocytes contain pyknotic nuclei. These cells are known as sunburn cells [9,
10]. Other UV effects include p53 activation, cell cycle arrest, activation of DNA
repair mechanism, and apoptosis induction. It also exerts many other effects on the
skin, including pigmentation, tanning, and immunomodulation. Indeed, pigmenta-
tion of the skin is correlated with UV radiation in particular geographic areas. As a
result, natural selection favored dark-skinned human populations to be protected
against damaging UV radiation in tropics, where bright sunlight is observed
throughout the year. Interestingly, UVB and UVA have various medical applica-
tions. UVA is called black light or near-UVR. UVA is longer wavelength radiation
and is poorly absorbed by proteins and nucleic acids of the skin cells. Hence, it does
not cause erythema in normal skin at moderate doses. UVB radiation is erythemo-
genic and is referred as sunburn radiation or mid-range UVR.

3.2.1 Ultraviolet Radiation and Vitamin D Synthesis in Skin

Vitamin D regulates many processes including bone formation and remodeling,


bone metabolism, cell proliferation and differentiation, innate immune response,
intestinal calcium absorption, fertility, and normal functioning of many organs such
as the pancreas, brain, and heart [11]. Vitamin D3 (1, 25-dihydroxyvitamin D3) is
synthesized when UVB (wavelengths of 290–310 nm) is absorbed by 7-DHC
(7-dehydrocholesterol) present in the skin and converted into pre-vit D. Subsequently,
pre-vit D undergoes isomerization, yielding vit D. In the liver vit D is converted to
an active agent 1,25(OH)2 vit D by hydroxylation. The amount of pigment, the
thickness of skin, and the angle between vertical and the sun (known as solar zenith
angle; changes with time of the day, season, and latitude) are important factors for
biosynthesis of vitamin D3 in the skin [12]. When concentration of pre-vit D reaches
maximum, it is converted into inactive photoproducts, lumisterol and tachysterol. It
is very interesting to know that vitamin D is beneficial in various malignancies [13].

3.2.2 Ultraviolet Radiation and Skin Pigmentation

Skin pigmentation has direct relationship with UV radiation received by an indi-


vidual. Skin complexion is related with UV sensitivity of a person’s body and skin
cancer risk experienced during his/her lifetime. The amount of UV received in par-
ticular time and duration is different for each individual which can be determined in
the form of MED (minimal erythematous dose). MED is a quantitative method
used to determine the amount of UV required to cause sunburn in 24–48 h after
exposure in the form of edema and erythema as endpoints. Edema and erythema
describe swelling and redness, respectively, of the area exposed to UV radiation.
Severity of sunburn also depends on type of skin and its melanin content. Depending
on the amount of melanin, skin color has been divided into six phototypes. These
phototypes are determined through Fitzpatrick scale which is a semiquantitative
method. It is described by basal complexion, inflammatory response, melanin levels
20 N. Yadav and M. Banerjee

Table 3.1 The Fitzpatrick scale of UV sensitivity


Fitzpatrick skin Epidermal MED (mJ/ Cancer
phototype eumelanin Skin response to UV cm2) risk
I +/− Burns constantly, peels but 15–30 ++++
never tans
II + Burns easily, peels but tans 25–40 +++/++++
minimally
III ++ Burns moderately but tans 30–50 +++
marginally
IV +++ Marginal burning, easy 40–60 ++
tanning
V ++++ Tans easily, rarely but 60–90 +
substantial burn
VI +++++ Do not burn; fast and profuse 90–150 +/−
tanning occurs

to UV radiation, and cancer risk [14]. According to Fitzpatrick scale, higher dose
of UV radiation is required to burn eumelanin-rich skin. As a result, MED is highest
in dark-skinned and lowest in fair-skinned individuals (Table 3.1).

3.2.2.1 Molecular Mechanisms of Tanning


Tanning of skin in response to repeated UV exposure is a very common phenome-
non and usually elicited by most individuals [15] although it may vary from person
to person. It depends upon the amount and type of melanocyte-specific markers
which are modulated at different levels via different mechanisms induced by UV
[16]. UVA and UVB play synergistic effect on melanogenesis. UVB stimulates
expression of many pigment-related genes, such as tyrosinase-related protein 1,
TYR and DCT (DOPAchrome tautomerase) enzyme, as well as MITF
(microphthalmia-­associated transcription factor). UV radiation also causes damage
to DNA and other cellular macromolecules of keratinocytes present in the skin.
These damages result in production and secretion of α-MSH (melanocyte-­
stimulating hormone) which is encoded by POMC (pro-opiomelanocortin) gene.
However, the expression of POMC is upregulated by UV radiation for same pur-
pose. MC1R are UV-sensitive receptors present on the surface of melanocytes of the
basal epidermis. α-MSH binds to MC1R receptors and generates cAMP as a second
messenger in the presence of adenylyl cyclase enzyme. It leads to activation of PKA
(protein kinase A) and transcription factors CREB (cAMP-responsive binding ele-
ment) and Mitf (microphthalmia). CREB and Mitf, in turn, activate tyrosinase and
other melanin biosynthetic enzymes which result into increased levels of melanin
production. On the contrary, MC1R signaling also induces DNA repair pathways
and thereby upsurges UV-mediated resistance of melanocytes.
Melanin produced in melanocytes is transferred and accumulated in epidermal
keratinocytes. UVA causes oxidative effects which may result into immediate and
persistent pigment darkening of the skin. Though melanocytes are relatively lesser
in number than other cells in the skin, they predominantly express most of the
UV-responsive genes to protect from UV damage. Beside activation of genes
3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 21

involved in pigmentation pathway, UV radiation also upregulates genes involved in


cellular communication, motility, adhesion, immune responses, and growth.
Melanocyte activation and function are firmly regulated by numerous autocrine and
paracrine factors of melanocytes, keratinocytes, and fibroblasts of the skin [17].

3.3 Genetic Responses to UV Radiation

Nucleic acids are highly vulnerable to oxidative injury by reactive oxygen species
(ROS) since nucleic acids absorb UV radiation maximally and generate ROS intra-
cellularly. ROS such as superoxide anion radical, hydroxyl radical, and hydrogen
peroxide interact in various ways with components of DNA, thereby damaging the
DNA. Oxidation of nucleotide bases promotes mispairing rather than following nor-
mal Watson-Crick parameter as well as deamination, oxidation, and alkylation.
Depending upon the severity of UV exposure, formation of photodimers and other
mutations occur as a result of mutagenesis in the genome [18]. The G→T transver-
sion, for example, is a very common mutation where guanine is oxidized at 8th
position to yield 8-OHdG (8-hydroxy-2′-deoxyguanine) [19]. 8-OHdG pairs with
an A instead of C finally resulting to A/T pair. Many DNA repair pathways get acti-
vated in response to damage. Base excision repair (BER) is the key pathway to
revert DNA mutagenesis by removing and replacing damaged bases. BER is initi-
ated by the activation of DNA damage-specific glycosylases. Glycosylases scruti-
nize and recognize the DNA for alterations in their structure. Thereafter, damaged
nitrogenous base is removed by cleavage of the N-glycosylic bond between deoxy-
ribose sugar and base. This leaves phosphodiester backbone intact which creates an
abasic or apurinic/apyrimidinic site known as AP site. The site is then cleaved by
AP endonucleases which results in single-strand breaks. Single-strand DNA breaks
are subsequently processed and repaired using the complementary strand as a tem-
plate. In this process, either a single nucleotide or 2–3 nucleotides are synthesized
at a time. Other mechanisms involve antioxidant system that detoxifies ROS to
avoid oxidative damages to DNA. Among all antioxidant systems, glutathione
(GSH) is one of the most important and abundant cellular antioxidant molecules
which neutralizes the reactivity of free radicals and maintains some proteins in
reduced form for cellular homeostasis. In the cell, GSH exists in two forms, reduced
form, i.e., GSH, and oxidized form, i.e., GSSG. Reduced GSH helps the liver to
remove toxic materials from body. Other most important antioxidant enzymes
include catalase that catalyzes breakdown of hydrogen peroxide and superoxide
dismutase (SOD) that dismutates superoxide anion radicals to less toxic forms
[20–23].
22 N. Yadav and M. Banerjee

3.4 Photoproduct Formation

Direct interaction of UVB with DNA may produce photoproducts within the cell.
One of the most damaging effects is the formation of 6–4 pyrimidine dimer photo-
products (6–4PP) and cyclobutane pyrimidine dimers (CPD) which drastically
affect the usual 3D structure of DNA. Photoproducts interfere with usual replication
of DNA. Photoproducts are a great threat to normal cellular functioning and are
responsible for most of the UV-mediated carcinogenesis. 6–4 photoproducts are
formed as a result of covalent bond formation between adjacent thymine residues in
single DNA strand. CPDs are the most common and abundant form of DNA lesions
which contribute to nearly 85% of all UVB-generated DNA lesions. Besides TT
dimer formation, photoproducts can also be formed by interaction between adjacent
CT, TC, and CC residues. 6–4 PP are more mutagenic than CPDs. Although, as soon
as the lesions are formed, BER comes into action and reverts mutations to normal
state, it does not repair 6–4 PP to the same extent. 6–4 photoproducts, if not repaired,
may be converted into even more destructive form known as Dewar products.
Dewar products are hard to repair. A subway of NER is transcription-coupled repair
(TCR) process that slowly removes CPDs from template DNA strand of transcrip-
tionally active genes. C to T and CC to TT transitions are referred to as “UVB fin-
gerprint mutations” as they are formed by UVB irradiation. These transitions are
very common and plentiful. The specific mutations induced may be either single-­
base or double-base substitutions. Single-base substitutions induced by UVB in
DNA sequences are most often C for T. Double-base transitions, i.e., CC to TT, also
take place but at a lower frequency. Occasionally, UVB-induced base substitutions
have also been evident. However, T to G transversions and double-base changes
from TT to GG have been studied. UVB also generates ROS and induces photo-
chemical reactions favoring DNA-protein cross-link formation [24].

3.5 UV and DNA Repair Systems

There are three repair pathways of UV-induced DNA damage, i.e., BER, NER
(nucleotide excision repair), and MMR (mismatch repair). Non-bulky lesions are
specifically repaired by BER and MMR, whereas bulky lesions are repaired by
NER. The photoproducts are not problematic if repaired efficiently. If excessive and
chronic exposure of skin to UV continues, the repair pathway in skin cells become
overactivated, and photoproducts are passed on in subsequent rounds of replication.
NER mechanism repairs photoproducts and removes massive DNA lesions [25, 26].
There are two sub-pathways of NER, i.e., GG-NER (global genomic) and TC-NER
(transcription coupled), which converge after recognition of damaged DNA. This
pathway consists of five crucial steps: (1) recognition of photoproduct, (2) cleavage
on both sides of photoproduct, (3) removal of lesion, (4) synthesis of new nucleic
acid string, and (5) ligation of the string. NER alters the three-dimensional structure
of DNA due to orchestrated interaction of enzymes to DNA. When damaged DNA
is recognized by multi-protein repair complex, the lesion is cleaved off as a single
3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 23

strand, few bases away on each side [27, 28]. The undamaged DNA single strand
acts as a template for DNA polymerase, and a new complementary strand of few
bases is synthesized. Finally, ligation proceeds by ligase to form double-stranded
DNA. Transcription factors involved in NER polymorphism are possibly influenced
by UV irradiation, and thereby, chances of increased skin cancers may be observed.
Xeroderma pigmentosum (XP) is a rare skin disease observed in the form of
UV-mediated hypersensitivity. XP syndrome is result of homozygous genetic defect
in at least one effector protein of the NER pathway. NER pathway involves nine
major proteins named XPA, ERCC1, ERCC3 (XP-B), XPC, ERCC2 (XP-D), DDB2
(XP-E), ERCC4 (XP-F), ERCC5 (XP-G), and POLH. Protein names are associated
with xeroderma pigmentosum. Besides these, there are many other proteins involved
in NER pathway. XP patient is usually identified by high pigmentary abnormalities,
atrophy, and capillary telangiectasias on UV-exposed parts of the body. These
symptoms ultimately transformed to premalignant lesions. Skin cancers develop at
very early ages in XP patients [29]. These XP-associated skin cancers are character-
ized as UV signature mutations.

3.6 DNA Damage and UV Radiation

UV radiation raised mutations in DNA of keratinocyte cells lead to development of


non-melanoma skin cancers including BCC and SCC. These mutations start with
the interaction of cellular DNA with UV photons. When DNA molecules absorb UV
photons, they get excited resulting in the formation of photoproducts. The photo-
products, if not repaired, can restrict DNA replication process, thereby causing
mutations in DNA to occur. UVB radiation has 1000 times more mutagenicity than
UVA.

3.6.1 Genes Under UV Threat

3.6.1.1 Tumor Suppressor Gene p53


Tumor suppressor gene p53 is known as the genomic guardian. In humans, it is
encoded by TP53 gene and checks DNA mutations, thereby preventing cancer. It
has been associated with cancers in nearly all skin types and contributes 50% of all
human cancers [30]. p53 is actively involved in maintaining genomic integrity,
DNA repair, cell cycle arrest, and apoptosis [33]. UV irradiation can induce muta-
tion in tumor suppressor genes and proto-oncogenes. p53 has been known for its
opposing effects. On the one side, it prevents UV-mediated cancer, whereas, on the
other hand, it plays a critical role in the development of precancerous lesions.
Formation of precancerous lesions is the result of mutation in p53 gene. UV irradia-
tion causes TP53 proteins encoded by p53 gene to accumulate in the nucleus and
delay the cell cycle progression. This delay provides extra time for DNA repair and
apoptosis of damaged cells. In UV-irradiated cells, the expression of p53 gene is
upregulated which results in increased synthesis of TP53 proteins. These proteins
24 N. Yadav and M. Banerjee

interact with other regulatory proteins to arrest cell cycle at check points. If UV
irradiation elicits mutation in p53 gene, cells may enter into S phase besides apop-
tosis disruption and DNA damage. Cells continue to replicate with mutated DNA
and, consequently, lead to cancer transformation. High percentage of p53 mutations
has been documented in SCC skin cancer [31]. Most of UVB-mediated p53 muta-
tions are predominantly the result of C to T and CC to TT base substitutions in both
SCCs and BCCs. However, isolated mutations in one allele and loss of the other p53
allele are not uncommon in SCC (Fig. 3.3).
In SCCs, mutation in proto-oncogene or single p53 mutation causes transition
from precursor lesions, actinic keratosis to invasive carcinoma. This is a multistep
process where precursor cells undergo successive genetic lesions prior to tumor
formation. However, in BCCs p53 mutations occur on both alleles clustered in a
specific region, exons 5–9, and there is no loss of allele.

UV radiation

Stratum Corneum

Epidermis

DNA Damage

Genetic Mutations
Cell Cycle Arrest
DNA Repair
Tumor Suppressor Inactivation
Apoptosis
p53 p53
patched
Protooncogene-oncogene activation
H-ras, K-RAS, N-ras

Normal Cell Proliferation

Abnormal Cell Proliferation

Tumorigenesis

Fig. 3.3 Molecular mechanisms in skin under UV radiation


3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 25

In case of melanomas, p53 mutations are usually much less (˂25%) observed and
probably play different roles than in non-melanoma skin cancer. The exact mecha-
nism of p53 mutations still remains to be solved. However, in non-melanoma skin
cancer, initially expression of p53 protein is nonsignificant, while overexpression
occurs in late stages of melanomas which turns into tumor formation. However,
frequent p53 mutations in metastatic melanomas and less common in primary mela-
noma are obvious. The frequency of p53 mutation is 1–5% in primary melanomas
and 11–25% in metastatic melanomas. It is also observed that there is an inverse
relationship between the actual p53 mutations and overexpression of its protein
product, suggesting involvement of other proteins and genes that alter p53 protein
levels in melanoma progression.

3.6.1.2 PTCH Tumor Suppressor Gene


Protein patched homolog 1 (PTCH) is another group of tumor suppressor genes
associated with BCC, sporadic BCCs, and nevoid basal cell carcinoma (NBCC).
NBCC is an autosomal dominant disorder categorized by multiple BCCs in young
age. In 20–30% sporadic BCCs, somatic mutations are common in PTCH gene.
Somatic mutations are similar to p53 mutations.

3.6.1.3 Ras Proto-Oncogenes


The ras proto-oncogene family is another group of genes that is a crucial target for
UVB radiation. The ras family encodes G proteins that hydrolyze GTP (guanosine
5′-triphosphate) and facilitate cell signaling responses of various growth factor
receptors. Cells with ras mutations are important in early stages of skin cancer
development. Proto-oncogenes are normal genes that become oncogenic after muta-
tion and encode proteins with new functions. The mutant proteins cannot hydrolyze
GTP; thereby, cell growth remains no longer dependent on growth factors and con-
tinues even after there are no growth factors available. UVB-induced mutations in
ras genes are also evident in some human BCCs and SCCs. In individuals with XP,
ras gene expression is increased significantly.

3.6.1.4 Other Genes


The tumor suppressor gene p16 on chromosome 9p21, CMM1 gene on chromo-
some 1p36, CDK4 (cyclin dependent kinase) gene on chromosome 12q14, and
other genes related with p53 pathways are also implicated in the development of
melanoma [27, 32].

3.7 Conclusion

Ultraviolet radiation induces DNA strand breaks either directly or through oxidative
pathways that may eventually lead to tumorigenesis. UVA is less carcinogenic than
UVB. UV-induced melanomagenesis can be mediated via various pathways. UVB-­
induced mutations lead to melanoma development. UV irradiation induces
pigmentation-­related aberration lesions in the skin. UV radiation can enhance
26 N. Yadav and M. Banerjee

vitamin D3 synthesis in skin, thereby upsurging the chances of survival of mela-


noma patients. For protection from UV-induced lesions, repair systems of the cells
work efficiently. Nucleotide excision repair (NER) clears DNA damages such as
CPDs and 6–4 PP. p53 gene mutation is most common mutation that consequently
can lead to cancer transformation associated with chronic sun exposure.

References
1. Ito S, Wakamatsu K, Ozeki H (2000) Chemical analysis of melanins and its application to the
study of the regulation of melanogenesis. Pigment Cell Melanoma Res 13(s8):103–109
2. Jablonski NG, Chaplin G (2010) Human skin pigmentation as an adaptation to UV radiation.
Proc Natl Acad Sci 107(Supplement 2):8962–8968
3. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T (2013) UV radiation and the skin. Int J Mol Sci
14(6):12222–12248
4. Narayanan DL, Saladi RN, Fox JL (2010) Ultraviolet radiation and skin cancer. Int J Dermatol
49(9):978–986
5. Tatalovich Z, Wilson JP, Mack T, Yan Y, Cockburn M (2006) The objective assessment of life-
time cumulative ultraviolet exposure for determining melanoma risk. J Photochem Photobiol
B Biol 85(3):198–204
6. Kohen E, Santus R, Hirschberg JG (1995) Photobiology. Elsevier, p 16
7. Norval M (2006) The mechanisms and consequences of ultraviolet-induced immunosuppres-
sion. Prog Biophys Mol Biol 92(1):108–118
8. Yamaguchi Y, Takahashi K, Zmudzka BZ, Kornhauser A, Miller SA, Tadokoro T, Berens W,
Beer JZ, Hearing VJ (2006) Human skin responses to UV radiation: pigment in the upper epi-
dermis protects against DNA damage in the lower epidermis and facilitates apoptosis. FASEB
J 20(9):1486–1488
9. Skobowiat C, Dowdy JC, Sayre RM, Tuckey RC, Slominski A (2011) Cutaneous hypothalamic-­
pituitary-­adrenal axis homolog: regulation by ultraviolet radiation. Am J Physiol-Endocrinol
Metab 301(3):E484–E493
10. Skobowiat C, Sayre RM, Dowdy JC, Slominski AT (2013) Ultraviolet radiation regulates
cortisol activity in a waveband-dependent manner in human skin ex vivo. Br J Dermatol
168(3):595–601
11. Norman AW (2008) From vitamin D to hormone D: fundamentals of the vitamin D endocrine
system essential for good health. Am J Clin Nutr 88(2):491S–499S
12. Chaplin G, Jablonski NG (2009) Vitamin D and the evolution of human depigmentation. Am
J Phys Anthropol 139(4):451–461
13. Khazai N, Judd SE, Tangpricha V (2008) Calcium and vitamin D: skeletal and extraskeletal
health. Curr Rheumatol Rep 10(2):110–117
14. Kawada A (2000) Risk and preventive factors for skin phototype. J Dermatol Sci 23:S27–S29
15. Miller SA, Coelho SG, Zmudzka BZ, Bushar HF, Yamaguchi Y, Hearing VJ, Beer JZ (2008)
Dynamics of pigmentation induction by repeated ultraviolet exposures: dose, dose interval and
ultraviolet spectrum dependence. Br J Dermatol 159(4):921–930
16. Schlenz K, Smuda C, Batzer J, Stä F, Wenck H, Elsaesser HP, Wolber R (2005) Pigmentation
mechanisms induced by different wavelength of Uv light. Pigment Cell Res 18:33
17. Choi W, Miyamura Y, Wolber R, Smuda C, Reinhold W, Liu H, Kolbe L, Hearing VJ (2010)
Regulation of human skin pigmentation in situ by repetitive UV exposure: molecular charac-
terization of responses to UVA and/or UVB. J Investig Dermatol 130(6):1685–1696
18. Schulz I, Mahler HC, Boiteux S, Epe B (2000) Oxidative DNA base damage induced by sin-
glet oxygen and photosensitization: recognition by repair endonucleases and mutagenicity.
Mutat Res/DNA Repair 461(2):145–156
3 Molecular and Genetic Response of Human Skin Under Ultraviolet Radiation 27

19. Kunisada M, Sakumi K, Tominaga Y, Budiyanto A, Ueda M, Ichihashi M, Nakabeppu Y,


Nishigori C (2005) 8-Oxoguanine formation induced by chronic UVB exposure makes Ogg1
knockout mice susceptible to skin carcinogenesis. Cancer Res 65(14):6006–6010
20. Meredith P, Sarna T (2006) The physical and chemical properties of eumelanin. Pigment Cell
Melanoma Res 19(6):572–594
21. Song X, Mosby N, Yang J, Xu A, Abdel-Malek Z, Kadekaro AL (2009) α-MSH activates
immediate defense responses to UV-induced oxidative stress in human melanocytes. Pigment
Cell Melanoma Res 22(6):809–818
22. Kadekaro AL, Chen J, Yang J, Chen S, Jameson J, Swope VB, Cheng T, Kadakia M, Abdel-­
Malek Z (2012) Alpha-melanocyte–stimulating hormone suppresses oxidative stress through a
p53-mediated signaling pathway in human melanocytes. Mol Cancer Res 10(6):778–786
23. Bickers DR, Athar M (2006) Oxidative stress in the pathogenesis of skin disease. J Investig
Dermatol 126(12):2565–2575
24. Agar NS, Halliday GM, Barnetson RS, Ananthaswamy HN, Wheeler M, Jones AM (2004) The
basal layer in human squamous tumors harbors more UVA than UVB fingerprint mutations: a
role for UVA in human skin carcinogenesis. Proc Natl Acad Sci U S A 101(14):4954–4959
25. Nouspikel TD (2009) DNA repair in mammalian cells. Cell Mol Life Sci 66(6):994–1009
26. Kane AB, Kumar V (1999) Environmental and nutritional pathology. Robbins pathologic basis
of disease, pp 403–457
27. Daya-Grosjean L (2008) Xeroderma pigmentosum and skin cancer. In Molecular mechanisms
of xeroderma pigmentosum, Springer, New York, pp 19–27
28. Leibeling D, Laspe P, Emmert S (2006) Nucleotide excision repair and cancer. J Mol Histol
37(5–7):225–238
29. DiGiovanna JJ, Kraemer KH (2012) Shining a light on xeroderma pigmentosum. J Investig
Dermatol 132(3):785–796
30. Ratner D, Peacocke M, Zhang H, Tsou HC (2001) UV-specific p53 and PTCH mutations in
sporadic basal cell carcinoma of sun-exposed skin. J Am Acad Dermatol 44(2):293–297
31. Benjamin CL, Ananthaswamy HN (2007) p53 and the pathogenesis of skin cancer. Toxicol
Appl Pharmacol 224(3):241–248
32. Hussein MR, Haemel AK, Wood GS (2003) p53-related pathways and the molecular patho-
genesis of melanoma. Eur J Cancer Prev 12(2):93–100
33. Hussain SP, Harris CC (2006) p53 biological network: at the crossroads of the cellular-stress
response pathway and molecular carcinogenesis. J Nippon Med Sch 73(2):54–64
Photocarcinogenesis and Molecular
Mechanism 4
Neeraj Agarwal

Abstract
Ultraviolet radiation (UVR) reaches to the earth surface through sunlight, and every
living organism gets exposed to it multiple times during their life span. Excessive
exposure to UVR has adverse effects and could lead to skin aging, eye damage, and
skin cancer. UVA and UVB components of UVR, both can penetrate the skin and
have the potential of causing skin damage. UVR exposure causes DNA damage lead-
ing to somatic mutations either directly or through the generation of reactive oxygen
species (ROS). UVR plays a role in initiation, promotion, and progression of cancer
via affecting the oncogenes, tumor suppressor genes, inflammation, immunosuppres-
sion, signaling pathways, and mitochondrial DNA damage. Although exposure to
sunlight is almost unavoidable, appropriate precautions should be taken while out-
side either for pleasure or work. This chapter comprehensively summarizes the
aspects about photocarcinogenesis, factors and molecular mechanisms involved in
photo-induced skin cancer, treatments available, and photoprotection.

Keywords
UVA · UVB · Photocarcinogenesis · Skin cancer · ROS · DNA damage

4.1 Introduction

It’s a pleasure to have some sunlight exposure, but too much and recurrent sunlight
exposure can cause serious complications. Overexposure to ultraviolet radiation
(UVR) coming through sunlight to the skin can cause acute effects such as sunburn
and long-term chronic effects leading to skin cancer and cataracts. UV radiation is an

N. Agarwal (*)
Urology Division, Department of Surgery, Anschutz Medical Campus,
University of Colorado Denver, Aurora, CO, USA

© Springer Nature Singapore Pte Ltd. 2018 29


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_4
30 N. Agarwal

invisible part of the whole-light spectrum of sunlight reaching the earth. UV radiation
is not visible to naked eye since it has wavelengths shorter than visible light. UVR
comprised of three wavelength ranges termed as UVA, UVB, or UVC. UVA has the
longest wavelength range at 320–400 nanometers (nm). UVB ranges from 290 to
320 nm. UVC has the shorter wavelength <290 nm. UVR reaching the earth surface
through sunlight contains UVB and UVA but not UVC. UVA and UVB can penetrate
the ozone layer, while UVC gets absorbed by the ozone layer and therefore does not
reach the earth. Long-term human exposure to UVA and UVB can result in adverse
events such as premature skin aging, eye damage (including cataracts), and skin can-
cers. They can also suppress the immune system, thereby suppressing the ability to
fight off diseases. UVB radiation has more energy and shorter wavelength causing
more damage quickly compared to UVA. Since UVB wavelength falls in the nucleic
acid absorbance range, UVB exposure causes the DNA damage leading to sunburn
and skin cancer. UVA radiation is less harmful than UVB, but it can penetrate deeper
into the skin. Repeated exposure to even low doses of UVA can cause long-term skin
injury, even with no signs of sunburn. UVA light is considered safe and therefore used
in tanning booths. However, repeated exposure to UVA in tanning booths causes the
same level of skin and eye damage as natural sunlight.
The main reason for skin cells to become cancer cells is the exposure to UV
radiation [1]. Almost all types of skin cancers including non-melanoma skin cancers
and melanoma are initiated by chronic UVR exposure from the sun or other sources
such as solaria (solariums, sunbeds, and sun lamps). Skin cancer arises from the
sensitization of the epidermal cells – the outermost layer of the skin. The longer
wavelength UVA penetrates deep into the skin (up to the dermis) and causes genetic
damage to cells, photoaging, and immunosuppression. The shorter wavelength
UVB only penetrates the epidermis and causes cellular damage. UVB is mostly
responsible for sunburn, which is a significant risk factor for skin cancer, especially
melanoma. If the body fails to repair the extensive damage to the cells, they can
escape the cell-cycle checkpoint and start dividing. This uncontrolled growth of
cells eventually results in tumor formation. Both UVA and UVB contribute to sun-
burn, skin aging, eye damage, melanoma, and other skin cancers.
Annually, about one million patients are diagnosed with skin cancer, making it the
most commonly occurring cancer in the United States alone [2, 3]. Solar UVB radia-
tion is the main environmental factor for causing skin cancer; primarily, it causes
DNA damages in skin epidermal cells which, if unrepaired, potentially lead to initi-
ated cells (Fig. 4.1). The highest risk factors for melanoma are family history,

Fig. 4.1 Repetitive exposure of skin to UV radiation leads to initiation, promotion, and progres-
sion of skin cancers
4 Photocarcinogenesis and Molecular Mechanism 31

multiple moles, fair skin, immunosuppression, and UVR. Epidemiologic studies


have suggested that intense intermittent UVR exposure and severe sunburns during
childhood confers the highest risk [4]. Exposure to UVR due to indoor artificial tan-
ning devices has also been linked to melanoma risk [5]. UVR causes multiple changes
on the skin, including DNA damage, reactive oxygen species (ROS) generation,
alterations in cutaneous immune function, and production of growth factors [6].
Recent studies with mouse models have shown that UVR exposure causes induction
of inflammatory responses including macrophages and neutrophils that can promote
melanocytic cell survival, immunoevasion, and perivascular invasion [7, 8].

4.2 Types of Skin Cancer

Three major types of skin cancers are basal cell carcinoma (BCC), squamous cell
carcinoma (SCC), and melanoma. The first two, BCC and SCC, are grouped
together as non-melanoma skin cancers. Cancer starts with precancerous lesions
termed as dysplasia; for skin cancer these are changes in the skin that could lead
to cancer over time. Some dysplastic changes that occur in the skin are actinic
keratoses (AK), moles, and dysplastic nevi (abnormal moles). Other less common
types of skin cancer include Merkel cell tumors and dermatofibrosarcoma protu-
berans. Non-­melanoma skin cancers SCC and BCC occur in the vast majority.
Although malignant, these are noninvasive and may not spread to other body
parts. Malignant melanomas are comparatively less common in occurrence.
Malignant melanomas are highly invasive with a tendency to spread to other parts
of the body.

4.3 Mechanisms Involved in UVR-Induced Carcinogenesis

4.3.1 ROS and DNA Damage

The advancement of skin cancer occurs in three sequential steps including initia-
tion, promotion, and progression which are mediated via alterations in cellular, bio-
chemical, and molecular processes (Fig. 4.1). Reactive oxygen species are reported
to be involved in all three steps of skin carcinogenesis [9]. The permanent altera-
tions or mutations in the genes cause the first step in carcinogenesis, initiation [10].
The genetic alterations or mutations in proto-oncogenes and tumor suppressor genes
may cause epidermal cells to avoid going into terminal differentiation and start
dividing [11]. During oxidative stress, free radicals can directly damage DNA, and
Ca2+-dependent endonucleases can be activated to cause DNA strand breaks
(Fig. 4.2). Extensive oxidative stress-induced DNA damage can lead to mutation,
altering cellular phenotype leading to aberrant cellular growth or cell death [12].
Evidence exist suggesting the important role of ROS in skin carcinogenesis through
various mechanisms. Reactive oxygen species cause the activation of pro-­
carcinogens like polycyclic aromatic hydrocarbons. For example,
7,12-dimethylbenz(a)anthracene (DMBA) can initiate mouse skin carcinogenesis
32 N. Agarwal

Fig. 4.2 Mutagenic DNA damage by UV light. Exposure to UV radiation causes covalent bond-
ing between thymine molecules, generating cyclobutane thymine dimer, a DNA lesion causing
mutations leading to UVR-induced skin cancers

by causing point mutation in the proto-oncogene c-Ha-Ras [13]. Activation of


DMBA is done by NADPH and cytochrome P450-dependent oxidases [14].
Metabolism of molecular oxygen [15] leads to the formation of various ROS
including superoxide anion radical (O2−.), singlet oxygen (1O2), hydrogen peroxide
(H2O2), and the highly reactive hydroxyl radical (.OH). Normally in all aerobic
cells, there is a balance between the generation of ROS and their quenching by bio-
chemical antioxidants. Disruption of this critical balance leads to oxidative stress
due to the formation of excessive ROS, depletion of antioxidants, or combination of
both. Excessive accumulation of ROS in the tissues causes damage by sensitizing
and altering lipids in cellular membranes, nucleotides in DNA [16], sulphydryl
groups in proteins [17], and cross-linking/fragmentation of ribonucleoproteins [18].
ROS-mediated DNA damage is the major mechanism of cancer initiation [19].
ROS-induced DNA damage leads to random mutations in genes including genes
essential for normal cell division. Most of the genetic mutations caused by ROS
involves modification of guanine, causing G→T transversions [20–23]. Once these
ROS-induced mutations occur in oncogenes or tumor suppressor genes and alter the
expression/function, it will lead to initiation/progression of cancer [24]. These find-
ings strongly suggest that ROS are directly or indirectly involved in both initiation
and progression of cancer [25].

4.3.2 Molecular Mechanisms

Solar radiation exposure mainly from UVB and UVA mutagenize DNA, leading to
UV-signature mutations, C to T or CC to TT, almost always via cyclobutane dimers
(Fig. 4.2). Whenever these mutations occur in oncogenes, tumor suppressors, or
important housekeeping genes can alter their function, leading to uncontrolled cell
cycle, and transformation of keratinocytes and melanocytes [26]. In addition to
mutating DNA directly, UV can alter signal transduction pathways that may indi-
rectly affect mutation frequency, for example, by accelerating the cell-cycle pro-
gression which will give less time for cells to repair any DNA errors before the
subsequent round of replication, or may reduce the levels of enzymes responsible
for the abatement of UV-induced cellular damage [27].
4 Photocarcinogenesis and Molecular Mechanism 33

4.3.3 Tumor Suppressor Genes (TSGs)

Several TSGs have been reported to play a role in photocarcinogenesis including


Tp53 in SCCs and BCCs, p16 in melanoma, and PTCH in BCCs and possibly SCCs.
UVR alters the genetic pathways regulated by these molecules. Notably, the pertur-
bation of TSGs-regulated whole signaling cascade more critically determines the
fate of the lesions than the alteration of TSGs itself at the protein or gene level [28].
Tp53 is a well-known transcription factor; it maintains the genomic stability by
two main mechanisms, apoptosis and cell-cycle arrest. The functional loss of Tp53
leads to transformation of cells in vitro and formation of neoplasms in vivo.
Mutations in Tp53 gene occur in more than 50% of cancers including melanoma
and non-melanoma skin cancers. Point mutations, deletions, or insertional muta-
tions in Tp53 gene may result in its inactivation [29].
A low-molecular-weight CDK inhibitor, p16, is the product of a tumor suppres-
sor gene p16/INK4a gene. p16 is also a tumor suppressor and is frequently mutated
in human tumors, including skin cancer. P16 inhibits the cell-cycle progression by
preventing phosphorylation of the retinoblastoma gene (Rb). The inactivating muta-
tions in p16 gene can disrupt these functions [30–33].
PTCH protein serves as a receptor for sonic hedgehog, a secreted molecule
implicated in tumorigenesis. The normal biological function of a PTCH protein is to
relay extracellular growth regulatory signals to the nucleus [34]. PTCH gene is con-
sidered as the potential tumor suppressor gene for familial and sporadic BCCs.
Mutations in PTCH gene frequently occur in BCCs mainly the UV-signature muta-
tions C:T transitions, representing earlier events in the development of BCCs than
Tp53 gene alterations [35]. These mutations are specifically more frequent in xero-
derma pigmentosum-linked BCCs compared to sporadic BCCs [36, 37] and may
also be linked to an allelic loss at chromosomal region 9q22.3 harboring PTCH
gene. Occasionally, nonsense, missense, and silent PTCH mutations also occur in
SCCs patients with the previous history of multiple BCCs [38]. The expression
level and the mutational status of PTCH gene in normal skin and melanoma are still
unknown.

4.3.4 Oncogenes

A subset of genes involved in the growth and differentiation of normal embryonic


and adult tissues are termed as proto-oncogenes. Genetic mutations like point muta-
tions or chromosomal translocations caused by UVR exposure to some of the proto-
oncogenes can lead to their higher expression levels or structurally different variants.
Due to the altered expression or activity, the proto-oncogenes become oncogenes
leading to initiation and progression of photocarcinogenesis. Some of the known
oncogenes involved in photocarcinogenesis are bcl-2, ras, and c-fos genes [28].
Role of each of these oncogenes is described below.
Bcl-2, originally discovered as a translocated locus in B-cell leukemia/lym-
phoma, is an apoptosis suppressor gene. Apoptosis is a process of programmed cell
34 N. Agarwal

death, which usually occurs in those cells which are no longer needed or are cor-
rupted. By suppressing apoptosis, bcl-2 prevents the elimination of corrupt cells
which eventually turns into neoplastic cells. Based on these findings, bcl-2 func-
tions as an oncogene and plays an important role in tumorigenesis. Higher bcl-2
protein levels have been detected in various types of skin cancer such as AK, SCCs,
BCCs, and melanomas which strongly suggests its potential functional role in skin
cancer development [39–43].
The ras proto-oncogenes belong to the family of small GTP-binding proteins and
mediate the transduction of intracellular mitogenic signals caused by activation of
growth factor receptors. Ras is a critical component of mitogenic signaling path-
ways. Activation of ras leads to the dysregulated growth of cells. Therefore, activa-
tion of ras is an early and possibly initial event in skin cancer development.
c-Fos proto-oncogene encodes a nuclear protein and is formed together with
c-Jun or other Jun family members, the transcription factor-activated protein (API).
c-fos plays a role in proliferation and differentiation of cells. c-fos is detected in the
nuclei of SCCs while in normal squamous cells is present in the cytoplasm [44, 45].
It shows gradual protein upregulation with the progression toward more malignant
phenotype. c-Fos mRNA is expressed in basal keratinocytes, spinous cells, Bowen’s
disease, and SCCs [44]. Interestingly, c-fos and its family member c-Jun are specifi-
cally expressed at protein/mRNA level in melanoma cell lines but not in normal
melanocytes [46, 47].

4.3.5 Other Molecules

Along with molecules described earlier, several other molecules are also involved in
photocarcinogenesis. Even though UV-signature mutations are not present in some
molecules, they can still be induced by UVR. Such molecules include extracellular
matrix (ECM) proteins, cytokines, and DNA repair pathways proteins. ECM is
composed of a network of proteins that interacts with adjacent tumor cells and influ-
ence their proliferation and migration. The cytokines are groups of polypeptides
secreted by certain immune cells and can modulate the growth and proliferation of
tumor cells. Emerging evidence suggests the influential role of various cytokines in
the uncontrollable growth of tumor cells in vitro and in vivo.
Accumulating evidence has suggested that cyclooxygenase-2 (COX-2), an
enzyme responsible for prostaglandin synthesis, may also play a role in the patho-
genesis of non-melanoma skin cancer. Pharmacological inhibition or deficiency of
a COX-2 enzyme in mice leads to suppression of tumor growth compared to control
mice when both are exposed to UV radiation. Epidemiological studies have also
suggested the involvement of COX-2 in UV-induced skin cancers. UVR itself can
also augment the COX-2 expression in human skin. Furthermore, recent studies
suggested that COX-2 inhibitor drugs may prevent the progression of non-­melanoma
skin cancers [48].
Reactivation of telomerase reverse transcriptase (TERT) is a most common fea-
ture of neoplasms. Recently reported highly frequent TERT promoter mutations in
4 Photocarcinogenesis and Molecular Mechanism 35

various types of cancers were also hypothesized to result from UV radiation. Populo
et al. [49] have examined the occurrence of TERT promoter mutations in BCCs
from X-irradiated and X-unexposed patients and in melanomas. TERT promoter
mutations were present in 27% of X-irradiated and 51% of X-non-irradiated BCC
patients. They were also detected in 22% of melanoma patients. Insightfully, in non-­
irradiated patients, BCCs from sun-exposed skin had more mutations, suggesting
the role of UVR in causing mutations. In melanoma patients, TERT promoter muta-
tions were more frequent in intermittent sun-exposed areas and were associated
with nodular and superficial spreading subtypes, increased thickness, ulceration,
increased mitotic rate, and BRAFV600E mutations.

4.3.6 Signal Transduction Pathways

The photooxidative stress alters several pathways predominantly includes the


mitogen-­activated protein kinase (MAPK), the nuclear factor-kappa beta (NF-κB)/
p65, the JAK/STAT (signal transduction and activation of transcription), and the
nuclear factor erythroid 2-related factor 2 (Nrf2) [50, 51]. Tyrosine kinase receptor
activates MAP kinase pathway which in turn activates transcription factor activator
protein-1 (AP-1). AP-1 increases the expression of matrix metalloproteinases
(MMPs), extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun-N-terminal
kinase (JNK), and p38 proteins [51]. The JNK and p38 pathways also mediate the
UVR-induced AP-1 and COX-2 expression and therefore are therapeutically tar-
geted for the cure of skin cancer [52]. Transcription factor Nrf2 and its downstream
target genes are significantly downregulated, thereby causing oxidative stress in
human prostate cancer [53].
The NF-κB activation is linked to UVR-induced oxidative modification of cel-
lular membrane components [53]. Mechanistically, NF-κB gets activated by release
from its inhibitor (I-κB) and translocate to the nucleus, thereby activating the
inflammatory cytokines and prostaglandins [50]. In general, NF-κB inhibition by
antioxidants, proteasome inhibitors, prevention of I-κB phosphorylation, or expres-
sion of overactivated mutant (I-κB) mitigate UV-induced damage [51].

4.3.7 Mitochondrial DNA Damage

Mitochondrial DNA (mtDNA) mutations have been used as a biomarker of


UV-induced DNA damage, commonly termed as sunburnt DNA. Mitochondria lack
the nucleotide excision repair, therefore having high mutation rate and mtDNA
complementation. Since ROS are involved in mutations of cancer-associated genes
and the mitochondrial respiratory chain is a major source of ROS generation, the
mtDNA is more vulnerable to ROS-mediated damage and can amplify ROS stress
in cancer cells. Skin cancer along with other types of cancer such as colorectal,
liver, breast, pancreatic, lung, prostate, and bladder was shown to harbor somatic
mtDNA mutations [54–57].
36 N. Agarwal

4.3.8 Inflammation Cascade

UV radiation can also induce pro-inflammatory genes. Inflammation plays an


important role in photoaging and photocarcinogenesis [58–60]. The inflammatory
mediators are released from various types of cells including keratinocytes, fibro-
blasts, tumor cells, leukocytes, and the endothelial lining of blood vessels. These
inflammatory mediators are plasma mediators (bradykinin, plasmin, fibrin), lipid
mediators (prostaglandins, leukotrienes, and platelet-activating factor), and the
inflammatory cytokines [interleukin-1 (IL-1), IL-6, and tumor necrosis factor
(TNF)-α]. ROS can also activate the lipid mediators, COX-2 and prostaglandin E2
(PGE2) [60–62]. UV radiation has also been reported to activate the ornithine
decarboxylase enzyme, which is known to inhibit the activity of different poly-
amines regulating the cell proliferation [64]. The inflammatory process also triggers
the generation of ROS and RNS (reactive nitrogen species), which in turn generates
peroxynitrite causing degradation and rearrangement of DNA [65, 66].
Bald et al. [8] have reported that recurrent UV exposure in a genetically engi-
neered mouse model with primary cutaneous melanomas can promote metastatic
progression, independent of its tumor-initiating effects. UV irradiation enhances the
tumor growth along abluminal blood vessel surfaces and thereby increases the met-
astatic spread of tumor cells leading to lung metastases. Metastatic spread is facili-
tated by the recruitment and activation of neutrophils. Neutrophil activation is
initiated by the release of high-mobility group box 1 (HMGB1) from UV-damaged
epidermal keratinocytes and driven by Toll-like receptor 4 (TLR4). This UVR-­
mediated neutrophil inflammatory response stimulates angiogenesis and promotes
the migration of melanoma cells toward endothelial cells. These results suggest that
UV-irradiated epidermal keratinocytes sensitize the innate immune system. This
also suggests that the inflammatory response to UV irradiation facilitates reciprocal
melanoma-endothelial cell interactions leading to perivascular invasion, which is
termed as angiotropism in human melanomas by histopathologists.

4.3.9 Immunosuppression

Individuals with the suppressed immune system are more susceptible to tumors due
to decreased immunosurveillance. Exposure to UVR suppresses the cellular
response but can also affect humoral response [67]. UVR exposure depletes the
antigen presenting epidermal Langerhans cells (LC), which are also crucial media-
tors of the cellular immune response [68]. Notably, in addition to depleting the cells,
UVR also impairs the function of LCs, for example, migration and antigen presenta-
tion in lymph nodes. How UVR impaired the LC migration is not well understood,
but impairment of antigenic presentation property is due to loss of co-stimulatory
molecules like B7. Isomerization of trans-urocanic acid to cis isoform directly sup-
presses the LC migration and activity [69–71]. In addition, UVR also promotes
secretion of the immunosuppressive cytokine IL-10. Due to the formation of cis-­
UCA or CPD, keratinocytes secrete IL-10. IL10 is a therapeutically relevant
4 Photocarcinogenesis and Molecular Mechanism 37

cytokine because it plays a crucial role in skin immunosuppression induced by UV


radiation as well as by other skin pathologies, e.g., melanoma, in which high IL-10
correlates with bad prognosis [72]. Also, UVR-induced LC depletion and the pro-­
inflammatory microenvironment cause the influx of macrophages which in turn
activates regulatory T cells (Tregs) and polarizes the Th1/Th2 response toward Th2
[66, 73]. The role of Th2 response in UVR-induced immunosuppression is likely
related to IL-12 expression by LC. IL-12 depletion alters T cell activation toward
Th2 while promoting Treg activation [74–76].

4.3.10 E
 xtracellular Remodeling: Network of Collagen, Elastin,
and Matrix Metalloproteinases

Collagen and elastin are the structural components of the ECM. Rearrangement of
the collagen and elastin fibers promotes the angiogenesis and metastasis process.
Further, damage to collagen and elastin proteins serve as additional sensitizers of
photooxidative stress [77]. Collagen consists of amino acids wound together to
form triple helices. Factors that determine specific properties of different collagens
are the length of triple helices, interruptions to the triple helix, and amino acid
modifications [58]. The elastin fibers, comprised of an elastin core (90%) sur-
rounded by fibrillin microfibrils, provide stretch-recoil properties to the skin. Skin
exposure to UV radiation causes the loss of proper elastin fibers [78]. UV radiation
exposure disrupts the microfibrillar network in the epidermal-dermal layer and the
dermis, which also causes the aberrant elastic fibers [79].
Epidermal keratinocytes and fibroblasts produce ECM proteolytic enzymes
(MMPs/elastases), which mediate ECM remodeling and skin cancer [80–82].
Although the basal levels of ECM proteolytic enzymes increase with aging, it can
also be accelerated by environmental pollutants and UV radiation, resulting in pre-
mature degradation of collagen and elastin fibers. Different MMPs are grouped on
the basis of presence or absence of AP-1 and TATA nucleotide sequences in their
promoters. Group I MMPs (MMP-1, 3, 7, 9, 10, 12, 13, 19, and 26) contain both
TATA box and AP-1 site, group II MMPs (MMP-8, 11, 21) only TATA box, and
group III (MMP-2, 14, 28) lacks both TATA box and AP-1 site [83, 84].
The MAPK pathway stimulated AP-1 activates the transcription of several
MMPs such as MMP-1, MMP-2/9, and MMP-3 that collectively degrade the ECM
[85]. In addition, AP-1 also inhibits the transcription of type I collagen [58].
Therefore, the disruption of ECM and tissue integrity is caused by both the MMPs
and the reduced expression of the structural ECM proteins. The tissue inhibitors of
MMPs (TIMPs) inhibit both the pro- and active forms of MMPs [83, 84]. The
remodeling of collagen and elastin, for angiogenesis, metastasis, and tissue destruc-
tion, is largely caused by either increased expression or activation of MMPs and
also reduced expression of TIMPs [83, 84].
38 N. Agarwal

4.4 Treatment for Skin Cancer

Different treatments are available for patients with non-melanoma skin cancer and
actinic keratosis. Six types of standard treatments are surgery, radiation therapy,
chemotherapy, photodynamic therapy, biologic therapy, and targeted therapy. Novel
therapies are also being tested in clinical trials.

1. Surgery – Different types of surgical procedures are Mohs micrographic sur-


gery, simple excision, shave excision, electrodesiccation and curettage, cryosur-
gery, laser surgery, and dermabrasion. Non-melanoma skin cancer or actinic
keratosis is mainly treated by single or multiple surgical procedures.
2. Radiation therapy – It involves the use of high-energy X-rays or other types of
radiation to eliminate or impair the growth of cancer cells. External and internal
radiation therapy are the two types of radiation therapy used.
3. Chemotherapy – For this treatment patient are given drug compounds to inhibit
the growth of cancer cells by either causing cell death or inducing senescence.
4. Photodynamic therapy – Photodynamic therapy (PDT) is a cancer treatment
that uses the combination of drug and laser light to kill cancer cells. A drug is
first injected into a vein which is not active until it is exposed to light. The drug
collects more in actively dividing cancer cells than in normal cells, and when
laser light is illuminated onto the skin, the drug becomes active and kills the
cancer cells.
5. Immunotherapy – It takes advantage of the patient’s own immune system to
fight cancer. Substances made by the body or made in a laboratory are used to
boost, direct, or restore the body’s own immune system to fight against cancer.
6. Targeted therapy – This type of treatment uses drugs or other substances to
inhibit the targeted protein or signaling pathway most likely driving the tumor
growth and progression. Aberrant protein expression or dysregulated signaling
pathway often leads to uncontrolled cell growth due to overexpression or muta-
tions caused by various external or internal factors.

4.5 Photoprotection

Living organisms have a group of mechanisms to neutralize the molecular damage


caused by sunlight, termed as photoprotection. Humans have the extremely efficient
system to internally convert DNA, proteins, and melanin for the protection of skin
from photodamage. During internal conversion, a photochemical process dissipates
the energy of UV photon into small, harmless amounts of heat. In cases of excessive
sun exposure, the energy of the UV photon was not fully dissipated into heat leading
to the generation of reactive oxygen species or other harmful reactive chemical spe-
cies. To prevent DNA damage, the ultrafast internal conversion of DNA reduced the
excited state lifetime of DNA to only a few femtoseconds, not providing enough
time for the excited DNA to react with other molecules. Pigment melanin present in
the skin is a photoprotective substance which acts as a natural sunscreen. Melanin
4 Photocarcinogenesis and Molecular Mechanism 39

dissipates more than 99.9% of the absorbed UV radiation as heat. Therefore, per-
sons with low melanin or fair skin are more prone to sun damage.

4.6 Photoprotective Agents

There are naturally occurring photoprotective agents present in the atmosphere and
environment and inside the body. Ozone, the major photoprotective agent formed in
the stratosphere, absorbs large amounts of UVB and UVC. However, it only absorbs
very little to negligible amounts of UVA and visible light. Clouds and fog can scat-
ter some of the UVR and thereby decrease the amount of UVR reaching the earth’s
surface. Densely leafed trees can also protect against UVB exposure. Ironically,
pollutants present in the atmosphere can also prevent the UVR from reaching the
earth surface.
Chromophores present in the body such as pyrimidine and purine bases in DNA,
and proteins can absorb light energy. Urocanic acid present in the epidermis has a
peak absorption spectrum at 277 nm, which is within UVR range. Melanin, a large
opaque pigment molecule present in the epidermis, can absorb throughout the UV
and visible ranges.
The most obvious strategy to prevent the detrimental effects of UV radiation is
to minimize its incidence on the skin. Although we have natural protection, still too
much sun exposure or phototoxic substances can overcome natural barriers leading
to skin damage and cancer. Physical photoprotective agents should also be consid-
ered while outside in the sun. Proper clothing, sunglasses, wide-brimmed hat, and
topical sunscreens are excellent sources of photoprotection. Topical sunscreens are
subdivided into reflective and absorbing substances. Maximum effectiveness of
sunscreens can be achieved by application of the correct amount and frequent
replenishment upon changing environmental conditions, e.g., increased perspira-
tion, water immersion, etc.

4.7 Conclusion

Sun exposure is almost unavoidable during normal-life activities either because of


occupation or pleasure activities. Too much sun exposure or adverse effects of pho-
totoxic compounds can lead to severe skin damage and cancer. Accumulation of
mutations overtime caused by either through direct DNA damage from sunlight or
through the generation of ROS can contribute to initiation and progression of skin
cancer. There are various mechanisms for photocarcinogenesis including activation
of oncogenes, suppression of tumor suppressor genes, suppression of immune sys-
tem, and dysregulation of signaling pathways (Fig. 4.3). Most of the skin cancers
are the non-melanoma type which is mostly local and don’t spread to others of the
body. A small percentage of skin cancers are melanoma which is highly malignant
and can rapidly spread to other parts of the body. Currently, six types of therapy
treatments are available for skin cancer, and there are also ongoing clinical trials for
40 N. Agarwal

SUN

UVB & UVA radiation

Epidermis

Direct damage
Dimers formation
Telomeres alteration

ROS production Alteration of proteins (TSGs, Oncogene, structural and enzymatic)


Lipid peroxidation (membrane degradation)

Dermis Inflammation

Immunosuppression

Extracellular matrix
remodeling & angiogenesis

Fig. 4.3 Various mechanisms of UVR-induced photocarcinogenesis

new drug treatment options. Overall, it is advisable to avoid unnecessary sun expo-
sure and use proper protection while in the sun to avoid the incidence of adverse
effects. Also, there should be awareness of the phototoxic potential of commonly
used therapeutic drugs and chemical compounds.

References
1. Koh HK, Geller AC, Miller DR, Grossbart TA, Lew RA (1996) Prevention and early detection
strategies for melanoma and skin cancer. Current status. Arch Dermatol 132(4):436–443
2. Rogers HW, Weinstock MA, Feldman SR, Coldiron BM (2015) Incidence estimate of nonmel-
anoma skin cancer (keratinocyte carcinomas) in the U.S. population, 2012. JAMA Dermatol
151(10):1081–1086
3. Bleyer AOLM, Barr R, Ries LAG (eds) (2006) Cancer epidemiology in older adolescents
and young adults 15 to 29 years of age, including SEER incidence and survival: 1975–2000.
National Cancer Institute, Bethesda NIH Pub. No. 06
4. Whiteman DC, Whiteman CA, Green AC (2001) Childhood sun exposure as a risk factor for
melanoma: a systematic review of epidemiologic studies. Cancer Causes Control 12(1):69–82
5. Boniol M, Autier P, Boyle P, Gandini S (2012) Cutaneous melanoma attributable to sunbed
use: systematic review and meta-analysis. BMJ 345:e4757
4 Photocarcinogenesis and Molecular Mechanism 41

6. Garibyan L, Fisher DE (2010) How sunlight causes melanoma. Curr Oncol Rep 12(5):319–326
7. Zaidi MR, Davis S, Noonan FP, Graff-Cherry C, Hawley TS, Walker RL et al (2011) Interferon-­
gamma links ultraviolet radiation to melanomagenesis in mice. Nature 469(7331):548–553
8. Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D et al (2014) Ultraviolet-­
radiation-­induced inflammation promotes angiotropism and metastasis in melanoma. Nature
507(7490):109–113
9. Marnett LJ (2000) Oxyradicals and DNA damage. Carcinogenesis 21(3):361–370
10. Sung SJS, Kormanik PP, Black CC (1993) Vascular cambial sucrose metabolism and growth in
loblolly-pine (Pinus-Taeda L) in relation to transplanting stress. Tree Physiol 12(3):243–258
11. Perchellet JP, Percheliet E, Gali HU et al (1995) Oxidant stress and multistage skin carcino-
genesis. In: Mukhtar H (ed) Skin cancer: mechanisms and human relevance. CRC Press, Boca
Raton, pp 145–180
12. Taffe BG, Kensler T (1989) Free radicals and signal transduction in tumor promotion. In:
Colburn NH (ed) Genes and signal transduction in multistage carcinogenesis. Marcel Dekker,
New York, pp 391–397
13. Quintanilla M, Brown K, Ramsden M, Balmain A (1986) Carcinogen-specific mutation and
amplification of Ha-ras during mouse skin carcinogenesis. Nature 322(6074):78–80
14. Ioannides C, Parke DV (1987) The cytochromes P-448 – a unique family of enzymes involved
in chemical toxicity and carcinogenesis. Biochem Pharmacol 36(24):4197–4207
15. Halliwell B (1999) Oxygen and nitrogen are pro-carcinogens. Damage to DNA by reactive
oxygen, chlorine and nitrogen species: measurement, mechanism and the effects of nutrition.
Mutat Res 443(1–2):37–52
16. Ahsan H, Ali A, Ali R (2003) Oxygen free radicals and systemic autoimmunity. Clin Exp
Immunol 131(3):398–404
17. Knight JA (1995) Diseases related to oxygen-derived free radicals. Ann Clin Lab Sci
25(2):111–121
18. Waris G, Alam K (1998) Attenuated antigenicity of ribonucleoproteins modified by reactive
oxygen species. Biochem Mol Biol Int 45(1):33–45
19. Ames BN (1983) Dietary carcinogens and anticarcinogens. Oxygen radicals and degenerative
diseases. Science 221(4617):1256–1264
20. Higinbotham KG, Rice JM, Diwan BA, Kasprzak KS, Reed CD, Perantoni AO (1992) GGT to
GTT transversions in codon 12 of the K-ras oncogene in rat renal sarcomas induced with nickel
subsulfide or nickel subsulfide/iron are consistent with oxidative damage to DNA. Cancer Res
52(17):4747–4751
21. Du MQ, Carmichael PL, Phillips DH (1994) Induction of activating mutations in the human
c-Ha-ras-1 proto-oncogene by oxygen free radicals. Mol Carcinog 11(3):170–175
22. Denissenko MF, Venkatachalam S, Ma YH, Wani AA (1996) Site-specific induction and repair
of benzo[a]pyrene diol epoxide DNA damage in human H-ras protooncogene as revealed by
restriction cleavage inhibition. Mutat Res 363(1):27–42
23. Lunec J, Holloway KA, Cooke MS, Faux S, Griffiths HR, Evans MD (2002) Urinary 8-oxo-2′-
deoxyguanosine: redox regulation of DNA repair in vivo? Free Radic Biol Med 33(7):875–885
24. Ames BN, Shigenaga MK (1993) Gold LS. DNA lesions, inducible DNA repair, and cell divi-
sion: three key factors in mutagenesis and carcinogenesis. Environ Health Perspect 101(Suppl
5):35–44
25. Moller P, Wallin H (1998) Adduct formation, mutagenesis and nucleotide excision repair of
DNA damage produced by reactive oxygen species and lipid peroxidation product. Mutat Res
410(3):271–290
26. Kramer M, Stein B, Mai S, Kunz E, Konig H, Loferer H et al (1990) Radiation-induced activa-
tion of transcription factors in mammalian cells. Radiat Environ Biophys 29(4):303–313
27. Daya-Grosjean L, Dumaz N, Sarasin A (1995) The specificity of p53 mutation spectra in sun-
light induced human cancers. J Photochem Photobiol B 28(2):115–124
28. Hussein MR, Wood GS (2002) Molecular aspects of melanocytic dysplastic nevi. J Mol Diagn
4(2):71–80
42 N. Agarwal

29. Hussein MR, Haemel AK (2003) Wood GS. p53-related pathways and the molecular patho-
genesis of melanoma. Eur J Cancer Prev 12(2):93–100
30. Serrano M, Hannon GJ, Beach D (1993) A new regulatory motif in cell-cycle control causing
specific inhibition of cyclin D/CDK4. Nature 366(6456):704–707
31. Stone S, Jiang P, Dayananth P, Tavtigian SV, Katcher H, Parry D et al (1995) Complex struc-
ture and regulation of the P16 (MTS1) locus. Cancer Res 55(14):2988–2994
32. Piepkorn M (2000) Melanoma genetics: an update with focus on the CDKN2A(p16)/ARF
tumor suppressors. J Am Acad Dermatol 42(5 Pt 1):705–722 quiz 23–6
33. Zhang H, Rosdahl I (2004) Deletion in p16INK4a and loss of p16 expression in human skin
primary and metastatic melanoma cells. Int J Oncol 24(2):331–335
34. Chung JH, Han JH, Hwang EJ, Seo JY, Cho KH, Kim KH et al (2003) Dual mechanisms of
green tea extract (EGCG)-induced cell survival in human epidermal keratinocytes. FASEB
J 17(13):1913–1915
35. D’Errico M, Calcagnile A, Canzona F, Didona B, Posteraro P, Cavalieri R et al (2000) UV
mutation signature in tumor suppressor genes involved in skin carcinogenesis in xeroderma
pigmentosum patients. Oncogene 19(3):463–467
36. Daya-Grosjean L, Sarasin A (2000) UV-specific mutations of the human patched gene in basal
cell carcinomas from normal individuals and xeroderma pigmentosum patients. Mutat Res
450(1–2):193–199
37. de Gruijl FR, van Kranen HJ, Mullenders LH (2001) UV-induced DNA damage, repair, muta-
tions and oncogenic pathways in skin cancer. J Photochem Photobiol B 63(1–3):19–27
38. Zhang H, Ping XL, Lee PK, Wu XL, Yao YJ, Zhang MJ et al (2001) Role of PTCH and p53
genes in early-onset basal cell carcinoma. Am J Pathol 158(2):381–385
39. Andrija Stanimirović HČBKB (2003) Expression of p53, bcl-2 and growth hormone receptor
in actinic keratosis, hypertrophic type. Arch Dermatol Res 295(3):102–108
40. Berhane T, Halliday GM, Cooke B, Barnetson RS (2002) Inflammation is associated with
progression of actinic keratoses to squamous cell carcinomas in humans. Br J Dermatol
146(5):810–815
41. Grace VM, Shalini JV, Lekha TT, Devaraj SN, Devaraj H (2003) Co-overexpression of p53 and
bcl-2 proteins in HPV-induced squamous cell carcinoma of the uterine cervix. Gynecol Oncol
91(1):51–58
42. Nakagawa K, Yamamura K, Maeda S, Ichihashi M (1994) bcl-2 expression in epidermal kera-
tinocytic diseases. Cancer 74(6):1720–1724
43. Feinmesser M, Tsabari C, Fichman S, Hodak E, Sulkes J, Okon E (2003) Differential expres-
sion of proliferation- and apoptosis-related markers in lentigo maligna and solar keratosis
keratinocytes. Am J Dermatopathol 25(4):300–307
44. Takahashi S, Pearse AD, Marks R (1994) Expression of c-fos proto-oncogene mRNA in non-­
melanoma skin cancer. J Dermatol Sci 7(1):54–62
45. Pacheco MM, Kowalski LP, Nishimoto IN, Brentani MM (2002) Differential expression of
c-jun and c-fos mRNAs in squamous cell carcinoma of the head and neck: associations with
uPA, gelatinase B, and matrilysin mRNAs. Head Neck 24(1):24–32
46. Kroumpouzos G, Eberle J, Garbe C, Orfanos CE (1994) P53 mutation and c-fos overex-
pression are associated with detection of the antigen VLA-2 in human melanoma cell lines.
Pigment Cell Res 7(5):348–353
47. Yang S, McNulty S, Meyskens FL Jr (2004) During human melanoma progression AP-1 bind-
ing pairs are altered with loss of c-Jun in vitro. Pigment Cell Res 17(1):74–83
48. Elmets CA, Ledet JJ, Athar M (2014) Cyclooxygenases: mediators of UV-induced skin cancer
and potential targets for prevention. J Invest Dermatol 134(10):2497–2502
49. Populo H, Boaventura P, Vinagre J, Batista R, Mendes A, Caldas R et al (2014) TERT promoter
mutations in skin cancer: the effects of sun exposure and X-irradiation. J Invest Dermatol
134(8):2251–2257
50. Lodish HBA, Kaiser CA, Krieger M, Scott MP, Bretscher A (2008) Molecular cell biology.
W.H. Freeman and Company, New York
4 Photocarcinogenesis and Molecular Mechanism 43

51. Magne N, Toillon RA, Bottero V, Didelot C, Houtte PV, Gerard JP et al (2006) NF-kappaB
modulation and ionizing radiation: mechanisms and future directions for cancer treatment.
Cancer Lett 231(2):158–168
52. Bachelor MA, Bowden GT (2004) UVA-mediated activation of signaling pathways involved in
skin tumor promotion and progression. Semin Cancer Biol 14(2):131–138
53. Birch-Machin MA, Swalwell H (2010) How mitochondria record the effects of UV exposure
and oxidative stress using human skin as a model tissue. Mutagenesis 25(2):101–107
54. Eshaghian A, Vleugels RA, Canter JA, McDonald MA, Stasko T, Sligh JE (2006)
Mitochondrial DNA deletions serve as biomarkers of aging in the skin, but are typically absent
in nonmelanoma skin cancers. J Invest Dermatol 126(2):336–344
55. Jakupciak JP, Wang W, Markowitz ME, Ally D, Coble M, Srivastava S et al (2005)
Mitochondrial DNA as a cancer biomarker. J Mol Diagn 7(2):258–267
56. Pelicano H, Carney D, Huang P (2004) ROS stress in cancer cells and therapeutic implications.
Drug Resist Updat 7(2):97–110
57. Birch-Machin MA (2005) Using mitochondrial DNA as a biosensor of early cancer develop-
ment. Br J Cancer 93(3):271–272
58. Callaghan TM, Wilhelm KP (2008) A review of ageing and an examination of clinical methods
in the assessment of ageing skin. Part 2: clinical perspectives and clinical methods in the evalu-
ation of ageing skin. Int J Cosmet Sci 30(5):323–332
59. Grossman D, Leffell DJ (1997) The molecular basis of nonmelanoma skin cancer: new under-
standing. Arch Dermatol 133(10):1263–1270
60. Hofseth LJ, Saito S, Hussain SP, Espey MG, Miranda KM, Araki Y et al (2003) Nitric oxide-­
induced cellular stress and p53 activation in chronic inflammation. Proc Natl Acad Sci U S A
100(1):143–148
61. Athar M, An KP, Morel KD, Kim AL, Aszterbaum M, Longley J et al (2001) Ultraviolet
B(UVB)-induced cox-2 expression in murine skin: an immunohistochemical study. Biochem
Biophys Res Commun 280(4):1042–1047
62. Buckman SY, Gresham A, Hale P, Hruza G, Anast J, Masferrer J et al (1998) COX-2 expres-
sion is induced by UVB exposure in human skin: implications for the development of skin
cancer. Carcinogenesis 19(5):723–729
63. Wang D, Dubois RN (2006) Prostaglandins and cancer. Gut 55(1):115–122
64. Einspahr JG, Xu MJ, Warneke J, Saboda K, Ranger-Moore J, Bozzo P et al (2006)
Reproducibility and expression of skin biomarkers in sun-damaged skin and actinic keratoses.
Cancer Epidemiol Biomark Prev 15(10):1841–1848
65. Maeda H, Akaike T (1998) Nitric oxide and oxygen radicals in infection, inflammation, and
cancer. Biochemistry (Mosc) 63(7):854–865
66. Coussens LM, Werb Z (2001) Inflammatory cells and cancer: think different. J Exp Med
193(6):F23–F26
67. Ullrich SE (2005) Mechanisms underlying UV-induced immune suppression. Mutat Res
571(1–2):185–205
68. Simon JC, Tigelaar RE, Bergstresser PR, Edelbaum D, Cruz PD Jr (1991) Ultraviolet B radia-
tion converts Langerhans cells from immunogenic to tolerogenic antigen-presenting cells.
Induction of specific clonal anergy in CD4+ T helper 1 cells. J Immunol 146(2):485–491
69. Norval M, Simpson TJ, Ross JA (1989) Urocanic acid and immunosuppression. Photochem
Photobiol 50(2):267–275
70. Noonan FP, De Fabo EC (1992) Immunosuppression by ultraviolet B radiation: initiation by
urocanic acid. Immunol Today 13(7):250–254
71. Kurimoto I, Streilein JW (1992) Deleterious effects of cis-urocanic acid and UVB radiation on
Langerhans cells and on induction of contact hypersensitivity are mediated by tumor necrosis
factor-alpha. J Invest Dermatol 99(5):69S–70S
72. Kruger-Krasagakes S, Krasagakis K, Garbe C, Schmitt E, Huls C, Blankenstein T et al (1994)
Expression of interleukin 10 in human melanoma. Br J Cancer 70(6):1182–1185
73. Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and can-
cer progression. Nat Genet 29(2):117–129
44 N. Agarwal

74. Baadsgaard O, Fox DA, Cooper KD (1988) Human epidermal cells from ultraviolet light-­
exposed skin preferentially activate autoreactive CD4+2H4+ suppressor-inducer lymphocytes
and CD8+ suppressor/cytotoxic lymphocytes. J Immunol 140(6):1738–1744
75. Simon JC, Cruz PD Jr, Bergstresser PR, Tigelaar RE (1990) Low dose ultraviolet B-irradiated
Langerhans cells preferentially activate CD4+ cells of the T helper 2 subset. J Immunol
145(7):2087–2091
76. Schmitt DA, Owen-Schaub L, Ullrich SE (1995) Effect of IL-12 on immune suppression and
suppressor cell induction by ultraviolet radiation. J Immunol 154(10):5114–5120
77. Wondrak GT, Roberts MJ, Cervantes-Laurean D, Jacobson MK, Jacobson EL (2003) Proteins
of the extracellular matrix are sensitizers of photo-oxidative stress in human skin cells. J Invest
Dermatol 121(3):578–586
78. Yaar M, Gilchrest BA (2007) Photoageing: mechanism, prevention and therapy. Br J Dermatol
157(5):874–887
79. Watson RE, Griffiths CE, Craven NM, Shuttleworth CA, Kielty CM (1999) Fibrillin-rich
microfibrils are reduced in photoaged skin. Distribution at the dermal-epidermal junction.
J Invest Dermatol 112(5):782–787
80. Philips N, Burchill D, O'Donoghue D, Keller T, Gonzalez S (2004) Identification of benzene
metabolites in dermal fibroblasts as nonphenolic: regulation of cell viability, apoptosis, lipid
peroxidation and expression of matrix metalloproteinase 1 and elastin by benzene metabolites.
Skin Pharmacol Physiol 17(3):147–152
81. Philips N, Hwang H, Chauhan S, Leonardi D, Gonzalez S (2010) Stimulation of cell prolifera-
tion and expression of matrixmetalloproteinase-1 and interluekin-8 genes in dermal fibroblasts
by copper. Connect Tissue Res 51(3):224–229
82. Philips N, Tuason M, Chang T, Lin Y, Tahir M, Rodriguez SG (2009) Differential effects of
ceramide on cell viability and extracellular matrix remodeling in keratinocytes and fibroblasts.
Skin Pharmacol Physiol 22(3):151–157
83. Herouy Y (2001) Matrix metalloproteinases in skin pathology (review). Int J Mol Med
7(1):3–12
84. Yan C, Boyd DD (2007) Regulation of matrix metalloproteinase gene expression. J Cell
Physiol 211(1):19–26
85. Fisher GJ, Datta SC, Talwar HS, Wang ZQ, Varani J, Kang S et al (1996) Molecular basis of
sun-induced premature skin ageing and retinoid antagonism. Nature 379(6563):335–339
Immunomodulation
and Photocarcinogenesis 5
Neeraj Agarwal

Abstract
Ultraviolet radiation exposure has many biological effects, and immunosuppres-
sion is one of the least understood. UVR-suppressed immune reactions have
been known for decades, and it diverges from drug-induced immunosuppression
in many ways. Repeated exposure to lower doses of UVR induces long-term
immunosuppression; UVR-mediated immunosuppression is antigen-specific and
primarily perturbs the T-cell-driven immune reactions. UVR-induced immuno-
suppression is one of the factors for photocarcinogenesis.

Keywords
UV radiation · Immunosuppression · Skin immune system · Immunotherapy

5.1 The Immune System of Skin

The skin is the largest organ of human’s body, accounting for 12–15% of total body
weight. The exposure of skin to vast majority of things, including oil, moisture,
light, cold, or warm, provides favorable conditions for the growth of numerous spe-
cies of bacteria and fungus (called commensal microbiota). Although the skin
immune system can control these skin microbes, it also gets educated from com-
mensal microbiota. Skin is a highly immune-responsive organ; the human skin har-
bors an estimated 20 billion T-cells, which is a far greater number compared to
blood T-cells, suggesting the importance of immune defense in the skin. Besides
T-cells, other major players in the skin immune landscape are diverse group of cells
with innate or innate-like functions (Fig. 5.1). The epidermis has keratinocytes and

N. Agarwal (*)
Urology Division, Department of Surgery, Anschutz Medical Campus,
University of Colorado Denver, Aurora, CO, USA

© Springer Nature Singapore Pte Ltd. 2018 45


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_5
46 N. Agarwal

UVR

CD8+T cell

EPIDERMIS
Langerhans cell

CD4+T cells

DERMIS Macrophage

ILC

Dermal DC HYPODERMIS

Fig. 5.1 Distinct locations of immune cells in the skin

Langerhans cells, while the dermis has dermal dendritic cells, macrophages, and
innate lymphoid cells. On sensitization with a foreign antigen, a population of den-
dritic cells (DCs) called Langerhans cells (LCs) internalize and process it. The LCs
mature and move to the lymph nodes, now termed DCs, present the processed anti-
gen fragments to naïve T-cells. Upon stimulation with DCs, the CD4+ T-cells get
differentiated into the T helper 1 (Th1) subtype or the Th2 subtype cells. The dif-
ferentiated Th1 cells secrete cytokines such as interleukin-2 (IL-2) and interferon-γ
which bolster the cellular immunity, while cytokines such as IL-4 and IL-10 secreted
by Th2 cells stimulate humoral immunity and are generally suppressive for cellular
responses. Along with CD4+ T-cells, the DCs can also activate CD8+ cytotoxic
T-cells, which are particularly essential for the control of corrupted cells or tumor
cells. The DCs also boost the specialized T-cells with regulatory functions termed
as T regulatory (T-reg) cells. Upon stimulation by DC cells, population of T-cells
expressing the skin homing receptors move from lymph nodes to the skin and
extravagate into the tissues. After homing the skin tissues, the T-cells act as effector
cells, often promoting the infiltration of macrophages and neutrophils to the site of
antigenic challenge.
5 Immunomodulation and Photocarcinogenesis 47

5.2 UVR-Induced Immunomodulation

Exposure to UV radiation leads to many biological changes to the skin that contrib-
ute to photocarcinogenesis. Low-dose exposure of UVR increases vitamin D syn-
thesis, which in turn protects from genetic damage and carcinogenesis [1]. Higher
exposure causes sunburn to the cells leading to apoptosis and cell death, thus pro-
tecting from photocarcinogenesis. UV doses in between low and high suppress
immunity and cause genetic mutations.

5.3 UVR-Induced Immunosuppression

In 1974, Kripke and colleagues reported that murine skin tumors induced by chronic
UVR exposure exhibited an unexpectedly high degree of antigenicity compared with
chemically induced tumors. UV-induced spindle cell tumors completely regressed when
transferred to immunocompetent mice but grew progressively in immunosuppressed
mice [2]. This suggests that chronic UV radiation mediates suppression of antitumor
immune responses. These findings marked the beginning of new subject termed photo-
immunology, which progressed rapidly into an exciting research field. Several research
groups have tried to understand the molecular mechanisms behind UVR-induced immu-
nosuppression. The involved pathways are complex and consist of multiple steps.
Depending on the UV dosage, wavelength, and exposure frequency, the sequence of
molecular events also varies. The sequence of events can also vary because of the pres-
ence of different antigens and the particular immune component studied.
The UVR-induced immunosuppression is thought to be initiated by the excitation of
chromophores at or near the body surface termed photoreceptors. The primary changes
in UVR-induced immunosuppression are DNA damage, trans- to cis-isomerization of
urocanic acid (UCA), and changes in membrane integrity. The leading chromophore
that absorbs the UVR in skin is DNA. The UVR-sensitized DNA leads to the formation
of cyclobutane pyrimidine dimers (CPDs) and (6-4)-photoproducts. These photoprod-
ucts then get deposited into keratinocytes and LCS in the epidermis and in DCs which
goes to lymph nodes for draining irradiated sites [3].
Studies on human subjects by Kraemer et al. 1987 provide substantial evidence
that DNA damage by UVR exposure causes immunomodulation and increases the
risk of developing skin cancer. Xeroderma pigmentosum is a rare genetic disorder
in which patients lack the ability to repair the DNA damage caused by UV radiation.
These patients have the higher risk of developing skin cancer due to accumulation
of UV-induced DNA mutations [4]. Examination of transgenic mice lacking DNA
repair mechanism also supports this study [5]. Application of liposome-based lotion
containing DNA repair enzyme or placebo cream onto skin for 1 year on patients
with xeroderma pigmentosum showed decreased incidence of actinic keratosis (a
precursor lesion to non-melanoma skin cancer) and skin cancer [6].
Formation of cis-UCA could also initiate the UV-induced immunosuppression.
Breakdown on histidine in the epidermis by histidase leads to the formation of trans-
isomer of UCA. Since the skin lacks the catabolic enzyme urocanase, trans-­UCA gets
48 N. Agarwal

accumulated in the epidermis. Trans-UCA acts as a chromophore and is a major


absorber of UVR in the skin, isomerizing to cis-UCA in a dose-dependent manner
until the photostationary state is achieved where both isomers are in equal proportion.
In 1983, De Fabo and Noonan [7] suggested that formation and accumulation of cis-
UCA could be an initiator of UV-induced immunosuppression. Since then, several
studies using different approaches have confirmed this hypothesis [8]. Particularly,
cis-UCA may play an important role in conditions involving antigen processing and
presentation, such as those occurring in microbial infections [9].
Devary et al., 1993, have shown that cell membrane is the target of UVR by
evaluating the series of events occurring in cells depleted of nuclei [10]. The UV
exposure causes generation of reactive oxygen species which disbalances the cel-
lular redox equilibrium leading to oxidative stress and membrane lipid peroxida-
tion. Oxidative damage of plasma membrane can activate various enzymes leading
to the synthesis of mediators, which in turn phosphorylates and activates transcrip-
tion factors, such as NF-κB [11]. These transcription factors will then regulate the
yield of immunomodulating cytokines following UV irradiation of keratinocytes
[12]. A further study revealed the involvement of platelet-activating factor (PAF)
secreted by keratinocytes in response to UVR-induced oxidative stress when mem-
brane phosphatidylcholine is oxidized [13]. PAF binds to its receptors expressed on
a variety of cells including monocytes, mast cells, and keratinocytes and activates
the synthesis of prostaglandins and cytokines (IL-4 and IL-10) [14]. Hence, UVR
exposure initiates the immunosuppressive pathway.

5.4 Mechanism of UVR-Mediated Immunomodulation

The mechanisms responsible for UVR-induced immunosuppression have signifi-


cant overlap between local and systemic effects. Dissimilar to the immunosuppres-
sive drugs which act by suppressing the immune system, UVR acts in an
antigen-specific fashion. UVR-mediated suppression of immune reactions is mostly
owed to the generation of regulatory T-cells. Monitoring the suppression of contact
hypersensitivity (CHS) is the best model to study UVR-induced immunosuppres-
sion. Topical application of potent contact allergens to skin results in antigen-­
specific sensitization causing a specific swelling response which develops after few
days of application of the same allergen on the ear. Pre-exposure of skin to the low
doses of UVR suppress and prevent the contact allergen sensitization [15]. Local
effects of UV exposure include the depletion of Langerhans cells or downregulation
of its antigen-presenting capacity [16, 17]. Keratinocytes and macrophages stimula-
tion in response to UV exposure produces cytokines such as IL-10, which are
important for systemic immunosuppression [17, 18]. The chromophores that play a
major role in mediating UV radiation effects are DNA, membrane lipids, and trans-­
urocanic acid (trans-UCA) which is isomerized to cis-UCA. The combination of
DNA damage, generation of reactive oxygen species, and cytokines collude to
mutagenize epidermal cells and create an immunosuppressive environment favoring
the initiation and progression of tumors [14, 19, 20].
5 Immunomodulation and Photocarcinogenesis 49

UVR-mediated DNA damage is the major molecular trigger for immunosuppression


[21]. Major DNA lesions caused by UVR are cyclobutane pyrimidine dimers and 6-4
photoproducts. Kripke et al. have further shown that repair of DNA damage by exoge-
nous DNA repair enzymes leads to inhibition of UVR-induced immunosuppression.
This finding provides direct evidence for involvement of DNA damage in UV-induced
immunosuppression [21]. UV-induced DNA lesions cause immunosuppression by
modification of antigen presentation [22], Langerhans cell depletion from the epidermal
layer, T-cell induction [23], recruitment of immunosuppressive CD11b+ macrophages
into the skin, and change of the cytokine environment, which is important for the cell-
mediated immune response [24]. Cytokine IL-12 has been demonstrated first to prevent
UVR-induced immunosuppression [25–27]. A hallmark feature of UVR-induced
immunosuppression is the depletion of Langerhans cells from the epidermis [15].
UVR also generates reactive oxygen species (ROS) including superoxide anion,
hydrogen peroxide (H2O2), and singlet oxygen, which further cause oxidative damage to
DNA, proteins, and lipids [28, 29]. ROS damage the DNA as well as proteins by chang-
ing the protein structure and function, which may alter various signaling pathways lead-
ing to inflammation, cell proliferation, angiogenesis, and tumorigenesis [30].

5.5 Skin Cancer Immunotherapy

Skin cancer immunotherapy has been most thoroughly studied in melanoma. Using
systemic IFN-α-2b and high-dose IL-2 immune agonists as adjuvants for the treat-
ment of late-stage melanoma has shown modest increase in disease-free survival of
a small proportion of patients [31, 32]. Melanoma-specific expression of antigens
such as Mart1 and gp100 elicits T-cell responses, thereby urging interest in vaccine-­
based and adoptive T-cell immunotherapies. Most of the systemic immunotherapies
now combine the adoptive T-cell immunotherapy with vaccines against melanoma-­
specific antigens or with chemotherapy. Recently, treatment with a humanized anti-
body called ipilimumab, targeting the inhibitory T-cell receptor CTLA-4, alone or
in combination with gf100 vaccine has shown to extend the overall survival of unre-
sectable stage III/VI melanoma patients [33].
For other skin cancers such as NMSC, topical or local application of immuno-
modulatory agents has been used. Treating plasmacytoid DCs with imiquimod, an
immune modifier which activates Toll-like receptor (TLR)7, caused the NF-κB-­
dependent secretion of pro-inflammatory cytokines such as IFN-γ and the chemo-
kines CXCR3, CXCL10, CXCL11, and CCL8 that collectively regulate lymphocyte
trafficking [34–36]. These events lead to further activation of antigen-presenting
cells and enrichment of Th1 and cytotoxic CD8+ T-cell responses [36–38].
Therapeutically, imiquimod treatment has shown effectiveness against superficial
primary skin tumors and cutaneous metastases [39], including BCC, Bowen’s dis-
ease, erythroplasia de Queyrat, and lentigo maligna [40]. Imiquimod treatment in
SCC decreases the infiltration of T-reg cells inside the tumor, suppresses the produc-
tion of IL-10 and transforming growth factor (TGF)-β, and restores the expression
of vascular E-selectin [41].
50 N. Agarwal

References
1. Wong G, Gupta R, Dixon KM, Deo SS, Choong SM, Halliday GM et al (2004)
1,25-Dihydroxyvitamin D and three low-calcemic analogs decrease UV-induced DNA dam-
age via the rapid response pathway. J Steroid Biochem Mol Biol 89–90(1–5):567–570
2. Kripke ML (1974) Antigenicity of murine skin tumors induced by ultraviolet light. J Natl
Cancer Inst 53(5):1333–1336
3. Sontag Y, Guikers CL, Vink AA, de Gruijl FR, van Loveren H, Garssen J et al (1995) Cells
with UV-specific DNA damage are present in murine lymph nodes after in vivo UV irradiation.
J Invest Dermatol 104(5):734–738
4. Kraemer KH, Lee MM, Scotto J (1987) Xeroderma pigmentosum. Cutaneous, ocular, and
neurologic abnormalities in 830 published cases. Arch Dermatol 123(2):241–250
5. Garssen J, Goettsch W, de Gruijl F, Slob W, van Loveren H (1996) Risk assessment of UVB
effects on resistance to infectious diseases. Photochem Photobiol 64(2):269–274
6. Yarosh D, Klein J, O’Connor A, Hawk J, Rafal E, Wolf P (2001) Effect of topically applied T4
endonuclease V in liposomes on skin cancer in xeroderma pigmentosum: a randomised study.
Xeroderma Pigmentosum Study Group. Lancet 357(9260):926–929
7. De Fabo EC, Noonan FP (1983) Mechanism of immune suppression by ultraviolet irradiation
in vivo. I. Evidence for the existence of a unique photoreceptor in skin and its role in photoim-
munology. J Exp Med 158(1):84–98
8. Norval M, El-Ghorr AA (2002) Studies to determine the immunomodulating effects of cis-­
urocanic acid. Methods 28(1):63–70
9. Kim TH, Moodycliffe AM, Yarosh DB, Norval M, Kripke ML, Ullrich SE (2003) Viability of
the antigen determines whether DNA or urocanic acid act as initiator molecules for UV-induced
suppression of delayed-type hypersensitivity. Photochem Photobiol 78(3):228–234
10. Devary Y, Rosette C, DiDonato JA, Karin M (1993) NF-kappa B activation by ultraviolet light
not dependent on a nuclear signal. Science 261(5127):1442–1445
11. Simon MM, Aragane Y, Schwarz A, Luger TA, Schwarz T (1994) UVB light induces nuclear
factor kappa B (NF kappa B) activity independently from chromosomal DNA damage in cell-­
free cytosolic extracts. J Invest Dermatol 102(4):422–427
12. Rosette C, Karin M (1996) Ultraviolet light and osmotic stress: activation of the JNK cascade
through multiple growth factor and cytokine receptors. Science 274(5290):1194–1197
13. Barber LA, Spandau DF, Rathman SC, Murphy RC, Johnson CA, Kelley SW et al (1998)
Expression of the platelet-activating factor receptor results in enhanced ultraviolet B radiation-­
induced apoptosis in a human epidermal cell line. J Biol Chem 273(30):18891–18897
14. Walterscheid JP, Ullrich SE, Nghiem DX (2002) Platelet-activating factor, a molecular sensor
for cellular damage, activates systemic immune suppression. J Exp Med 195(2):171–179
15. Toews GB, Bergstresser PR, Streilein JW (1980) Epidermal Langerhans cell density deter-
mines whether contact hypersensitivity or unresponsiveness follows skin painting with
DNFB. J Immunol 124(1):445–453
16. Schwarz A, Noordegraaf M, Maeda A, Torii K, Clausen BE, Schwarz T (2010) Langerhans
cells are required for UVR-induced immunosuppression. J Invest Dermatol 130(5):1419–1427
17. Loser K, Beissert S (2009) Regulation of cutaneous immunity by the environment: an impor-
tant role for UV irradiation and vitamin D. Int Immunopharmacol 9(5):587–589
18. Aubin F (2003) Mechanisms involved in ultraviolet light-induced immunosuppression. Eur
J Dermatol 13(6):515–523
19. Stege H, Roza L, Vink AA, Grewe M, Ruzicka T, Grether-Beck S et al (2000) Enzyme plus
light therapy to repair DNA damage in ultraviolet-B-irradiated human skin. Proc Natl Acad Sci
U S A 97(4):1790–1795
20. Sreevidya CS, Fukunaga A, Khaskhely NM, Masaki T, Ono R, Nishigori C et al (2010) Agents
that reverse UV-induced immune suppression and photocarcinogenesis affect DNA repair.
J Invest Dermatol 130(5):1428–1437
5 Immunomodulation and Photocarcinogenesis 51

21. Kripke ML, Cox PA, Alas LG, Yarosh DB (1992) Pyrimidine dimers in DNA initiate systemic
immunosuppression in UV-irradiated mice. Proc Natl Acad Sci U S A 89(16):7516–7520
22. Schwarz T, Schwarz A (2011) Molecular mechanisms of ultraviolet radiation-induced immu-
nosuppression. Eur J Cell Biol 90(6–7):560–564
23. Gruber F, Zamolo G, Kastelan M, Massari LP, Cabrijan L, Peharda V et al (2007)
Photocarcinogenesis – molecular mechanisms. Coll Antropol 31(Suppl 1):101–106
24. Nasti TH, Iqbal O, Tamimi IA, Geise JT, Katiyar SK, Yusuf N (2011) Differential roles of
T-cell subsets in regulation of ultraviolet radiation induced cutaneous photocarcinogenesis.
Photochem Photobiol 87(2):387–398
25. Muller G, Saloga J, Germann T, Schuler G, Knop J, Enk AH (1995) IL-12 as mediator and
adjuvant for the induction of contact sensitivity in vivo. J Immunol 155(10):4661–4668
26. Schmitt DA, Owen-Schaub L, Ullrich SE (1995) Effect of IL-12 on immune suppression and
suppressor cell induction by ultraviolet radiation. J Immunol 154(10):5114–5120
27. Schwarz A, Grabbe S, Aragane Y, Sandkuhl K, Riemann H, Luger TA et al (1996)
Interleukin-12 prevents ultraviolet B-induced local immunosuppression and overcomes
UVB-induced tolerance. J Invest Dermatol 106(6):1187–1191
28. Katta R, Brown DN (2015) Diet and skin cancer: the potential role of dietary antioxidants in
nonmelanoma skin cancer prevention. J Skin Cancer 2015:893149
29. Valko M, Izakovic M, Mazur M, Rhodes CJ, Telser J (2004) Role of oxygen radicals in DNA
damage and cancer incidence. Mol Cell Biochem 266(1–2):37–56
30. Bickers DR, Athar M (2006) Oxidative stress in the pathogenesis of skin disease. J Invest
Dermatol 126(12):2565–2575
31. Mocellin S, Pasquali S, Rossi CR, Nitti D (2010) Interferon alpha adjuvant therapy in
patients with high-risk melanoma: a systematic review and meta-analysis. J Natl Cancer Inst
102(7):493–501
32. Tsai KY (2007) Systemic adjuvant therapy for patients with high-risk melanoma. Arch
Dermatol 143(6):779–782
33. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB et al (2010) Improved
survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363(8):711–723
34. Torres A, Storey L, Anders M, Miller RL, Bulbulian BJ, Jin J et al (2007) Immune-mediated
changes in actinic keratosis following topical treatment with imiquimod 5% cream. J Transl
Med 5:7
35. Wenzel J, Uerlich M, Haller O, Bieber T, Tueting T (2005) Enhanced type I interferon signal-
ing and recruitment of chemokine receptor CXCR3-expressing lymphocytes into the skin fol-
lowing treatment with the TLR7-agonist imiquimod. J Cutan Pathol 32(4):257–262
36. Wolf IH, Kodama K, Cerroni L, Kerl H (2007) Nature of inflammatory infiltrate in super-
ficial cutaneous malignancies during topical imiquimod treatment. Am J Dermatopathol
29(3):237–241
37. Huang SJ, Hijnen D, Murphy GF, Kupper TS, Calarese AW, Mollet IG et al (2009)
Imiquimod enhances IFN-gamma production and effector function of T cells infiltrating
human squamous cell carcinomas of the skin. J Invest Dermatol 129(11):2676–2685
38. De Giorgi V, Salvini C, Chiarugi A, Paglierani M, Maio V, Nicoletti P et al (2009) In vivo
characterization of the inflammatory infiltrate and apoptotic status in imiquimod-treated basal
cell carcinoma. Int J Dermatol 48(3):312–321
39. Wagstaff AJ, Perry CM (2007) Topical imiquimod: a review of its use in the management
of anogenital warts, actinic keratoses, basal cell carcinoma and other skin lesions. Drugs
67(15):2187–2210
40. (2009) Imiquimod for superficial and in situ skin malignancy. Drug Ther Bull 47(10):113–6
41. Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, Muthukuru M et al (2008) Human
squamous cell carcinomas evade the immune response by down-regulation of vascular
E-selectin and recruitment of regulatory T cells. J Exp Med 205(10):2221–2234
Epidemiological Aspects
of Photocarcinogenesis 6
Neera Yadav and Monisha Banerjee

Abstract
Epidemiologically, it is hard to distinct the effects of component radiations
because sunlight is considered as a whole entity. Human skin may experience
either acute (immediate reaction) or chronic (repeated) sunlight exposure. UV
radiation coming through sunlight is absorbed by human skin and causes various
degrees of damage. However, melanin pigment in the skin acts as a natural sun-
screen and absorbs and prevents from detrimental consequences of UV radiation.
Repeated and extended UV exposure causes development of photoaging, i.e.,
premature skin aging and photocarcinogenesis. Photo-induced cancers particu-
larly include malignant melanomas and carcinomas, i.e., basal cell carcinoma
(BCC) and squamous cell carcinoma (SCC). High-UV radiation environment
leads to evolution of permanently dark constitutive pigmentation which is resis-
tant to UV radiation. However, light-skinned people experience premature skin
aging on repeated exposure. Therefore, prevalence of photocarcinogenesis is
highest in lightly pigmented people (Halder and Bridgeman-Shah: Cancer
75(S2):667–673, 1995). Development of photocarcinogenesis is dependent on
various factors including ethnic origin, place of residence, anatomic site exposed,
outdoor and indoor activities, total exposure duration, time of exposure, and
ambient sunlight intensity. Photocarcinogenesis is predominantly a disease of
people of European origin. Its rate is very low in Asia and in the United States. It
is more frequently observed in white-skinned than dark-skinned people.

Keywords
Epidemiology · Radiation · Melanin · Photocarcinogenesis · Carcinoma

N. Yadav (*) · M. Banerjee


Molecular and Human Genetics Laboratory, Department of Zoology, University of Lucknow,
Lucknow, Uttar Pradesh, India
e-mail: banerjee_m@lkouniv.ac.in

© Springer Nature Singapore Pte Ltd. 2018 53


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_6
54 N. Yadav and M. Banerjee

6.1 Introduction

Sunlight that reaches at the surface of the earth consists of visible (wavelengths
from about 400–700 nm) and ultraviolet (UV) radiation (290–400 nm). Solar UV
radiation is a combination of UVB (290–320 nm) plus UVA (320–400 nm) wave-
lengths [1]. UVC radiation (less than 290 nm) does not reach at the earth’s surface
as it is absorbed in the atmosphere. UVB is more energetic than UVA and has long
been believed to be significant factor responsible for the damaging effects of sun-
light producing cancer, erythema (sunburn), and DNA damage [2]. Recently, it has
been proved that UVA exposure is also harmful as it is 20 times more abundant. It
penetrates to deeper layers of the skin, i.e., up to the dermis. Of the total UV rays
reaching the ground level, 95% are UVA.
Epidemiologic studies generally deal with sunlight as a whole rather than with
any of its components, i.e., UVB, UVA, and visible light. According to epidemiologic
studies, it is not easy to discrete the overall effect of component radiations. Exposure
of skin to UV radiation is influenced by various factors (season, latitude, time of the
day, etc.). Exposure of skin to UV radiation may result into acute as well as chronic
responses. According to epidemiologic evidence, photocarcinogenesis is:

(i) Recurrent in people residing in regions of high ambient solar radiation


(ii) Observed more frequently in people with elongated sun exposure
(iii) More common on sun-exposed body parts
(iv) More common in sun-sensitive people
(v) More frequent in people suffering from benign sun-related skin disorders
(vi) Reduced by protecting the skin from sun exposure

However, UVA in natural sunlight is a major environmental challenge to the skin.


UV radiation is transmitted through human skin and causes various degrees of dam-
age. Melanin is the main absorber of UV radiation in human skin and acts as a natu-
ral sunscreen [3, 4]. Other functions including biological clock and sexual
maturation are also dependent on solar radiation.
Long-term exposure of skin to UV radiation disrupts skin’s normal organiza-
tion leading to photoaging (premature aging) and photocarcinogenesis. UV expo-
sure upregulates expression of certain types of enzymes found in human skin such
as matrix metalloproteinases (MMPs). These enzymes play essential role in pho-
tocarcinogenesis by regulating tumor initiation, growth, angiogenesis, and metasta-
sis [5]. Photocarcinogenesis can be grouped into two major skin cancer classes
either non-melanoma or malignant melanoma [6]. Non-melanoma skin cancer is
further subdivided into BCC and SCC.

6.2 Basal Cell Carcinoma

BCC is responsible for nearly 80% of all non-melanoma skin cancer cases. The
incidence of BCC is most common in Caucasian populations and rare in Asians and
Black races of Africa. BCC occurs on the body parts that are regular to sunlight
6 Epidemiological Aspects of Photocarcinogenesis 55

exposure. It mostly occurs in superior regions such as the head and neck. BCC is
most often observed on the facial parts especially the nose and the lip. The progres-
sion of BCC is slow and has low metastatic potential. However, BCC may move
into hypodermal fat, muscles, and bones too [7].

6.3 Squamous Cell Carcinoma

SCC is the second most common type of non-melanoma skin cancer. It contributes
to about 20% of all detected skin cancer cases. Characteristic features of SCC
include malignant proliferation of epidermal keratinocytes and unveil an augmented
risk of metastasis (Fig. 6.1) [8]. Metastasis is a multifaceted process which com-
prises degradation of extracellular matrix (ECM) and basement membrane by the
action of MMPs and changes in cell-to-cell adhesion. Subsequently, tumor cells
detach from their original location, enter into blood vessels, and form new tumors at
distant sites.

6.4 Cutaneous Melanoma

Cutaneous melanoma arises from mutated melanocytes. Melanocytes are also known
as the pigment-producing cells of skin. It contributes nearly 75% of all skin cancer
mortality and 3–5% of all cutaneous cancers [9]. Cutaneous melanoma is characterized
by rapid progression and metastasis. It is also accountable for high morbidity and mor-
tality rate among cancer patients. The progression of melanoma is favored by gelatin-
ase and MMP-2 and MMP-9 proteinases. Tumor development requires oxygen,
nutrients, and a direction for migration and invasion of tumor cells.

6.5 Geographical Variation in UV Radiation

Human activities that fetch significant climatic changes contribute to the depletion
of stratospheric ozone layer. Consequences of ozone depletion resulted in an
increased UV irradiance at the earth’s surface. Besides ozone depletion, other fac-
tors include atmospheric and environmental conditions, season, altitude, time of
day, and latitude. Currently, the concentrations of ozone depleting substances (ODS)

Fig. 6.1 Morphology of


normal and tumor cells of
the skin
56 N. Yadav and M. Banerjee

are reducing because of successful implementation of Montreal protocol. Montreal


protocol also led the rate of increase in UVB radiation slow at regions near the
Antarctic ozone hole. At high latitudes, reduction of glaciers and increase of cloud
cover may cause UV radiation to decrease at the earth’s surface. It is expected that
in the absence of proper implementation of Montreal protocol, UV radiation may
rise up to three times by 2065 at mid-northern latitudes. Increase in UV radiation up
to such level may have serious consequences on human health as well as on the
environment. The concentration of ozone has great impact on variability of UV
radiation in different regions of the world. Besides ozone, clouds and aerosols are
also important factors responsible for variability in UV radiation.
Since oxygen, ozone, and water molecules present in the atmosphere have capa-
bility to absorb maximum UVB radiation, therefore, its total amount received by the
earth’s surface is much less than UVA. The intensity and distribution of UVB greatly
vary than UVA because path length of sunlight reaching different parts of the earth
in different seasons is diverse (Fig. 6.2). There are differences in pattern of UVB
distribution all over the world due to random atmospheric scattering and absorption
at different latitudes. Average UVB reaching the earth may be reduced by humidity
and precipitation during rainy season. There are differences in the levels of UV
radiation received by different regions depending upon the geography. High-altitude
areas and areas near the equator receive the highest UVB intensities. African conti-
nent through which the equator passes receives uniformly high amounts of UV
radiation. On the other hand, northern Eurasia receives almost negligible amounts.
In the Arctic regions, high levels of UV radiations have been observed especially
during spring time due to reduced ozone concentration in these parts. According to
scientists, ozone will not reach normal level for at least two decades as it is damaged
by both human activities and natural process.
UVA levels are slightly lower at the equatorial regions than tropical and subtropi-
cal areas. UVA levels are higher toward the poles due to wider latitudinal bands than
UVB. This difference is much abundant to cause UVA at 380 nm wavelength nearly
15 times ampler than UVB at 305 nm. Tropics within the equatorial region receive
extremely high average UVB and UVA radiation. Though intensity of UVA varies

Fig. 6.2 Path length of sunlight reaching different parts of the earth in different seasons
6 Epidemiological Aspects of Photocarcinogenesis 57

greatly in these regions throughout the year, in dry tropical areas near the equator,
UVA varies greatly, but away from the equator less variation is observed. UVB lev-
els are much lower outside the tropics being extremely low average in North
America and Eurasia. Levels of UVA at these latitudes are lower but uniform
throughout the year. Increased UVB radiation is more often observed in most popu-
lated areas of the world which contribute to substantial ozone depletion. Biologically
relevant dose of UV radiation may vary considerably depending on the surface of
the earth. Horizontal surfaces may receive significantly low levels of winter UV
doses as compared to vertical surfaces [10, 11].

6.6 Geographical Variation of Skin Pigmentation

Geographically, the human skin has been divided into six types, viz., Types I, II, III,
IV, V, and VI (Fig. 6.3). Evolution of human skin pigmentation has been always asso-
ciated with alterations in the seasonal distribution, intensity, and biological activity of
UVA and UVB radiation. Natural selection favored dark-skinned phenotypes near
the equator due to high UVB in these regions and fair-skinned phenotypes near poles,
receiving lesser UVB radiation (Fig. 6.4). Pigmentation of human skin is further cor-
related with latitude rather than with altitude, temperature, and humidity [12].
High-UV radiation environments lead to evolution of dark constitutive skin pigmen-
tation which augmented high eumelanin production. Skin pigmentation also increases
in response to seasonal increase in UVB. UVB is more energetic and do not penetrate
the deeper layers of dermis as it is absorbed and scattered by cellular materials. UVA
is capable of penetrating deeply into the dermis of skin. Constitutive pigmentation

Fig. 6.3 Pigmentary phototypes and photocarcinogenesis risk in different geographical regions of
the world
58 N. Yadav and M. Banerjee

Fig. 6.4 Worldwide distribution of human skin type

may be further modified into tanning because of adaptation to seasonally elevated


doses of UV radiation. Tanning is a process of production of new melanin. It is a
protective response to the injury by UV radiation. Light-skinned people are geneti-
cally incapable of tanning, and thus, their skin is damaged repeatedly which conse-
quently develop into premature skin aging [13].

6.7 Geographical Variation of Photocarcinogenesis

Human migration from one place to another plays an important role in determining
the geographical variations in skin cancer. Modern human migrations cause mis-
matches between pigmentation of skin and geographical location or lifestyle of
population [14]. In human skin, solar UV radiation can cause pigmentation, photo-
aging, skin cancer, and the less frequent but very dangerous malignant melanoma.
The damaging or beneficial effects of UV radiation strongly depend on wavelength.
Usually, larger effects have been observed at shorter wavelengths and vice versa.
In some geographical regions, the rate of melanoma incidence in young people
including children is confined to less lethal forms because of rigorous awareness
campaigns regarding human health. People also have shown interest in sound
research findings related to human health. However, older individuals among lightly
pigmented inhabitants of tropics are at high risk of non-melanoma skin cancer
development although they are rarely fatal [15]. However, in many countries, the
trend of photocarcinogenesis incidences is continuously rising. These conditions
claim substantial expenditures to health-care systems and societal impacts.
Geographical regions of higher temperature are supposed to lead to more incidences
6 Epidemiological Aspects of Photocarcinogenesis 59

of skin cancers due to cumulative effect of solar radiation and temperature.


According to some reports, the prevalence of photocarcinogenesis has been found
highest in people with lightly pigmented skin, although such studies are based on
chronic sunlight exposures [16].

6.7.1 Ethnic Origin

Photocarcinogenesis is predominantly a disease of people of European origin. It is


observed that the rate of melanoma formation is very low in Asians in the United
States which is expected to rise in the next 50 years. Its incidence is some 20-folds
higher in Whites than Blacks [17]. Non-melanoma skin cancers, BCC and SCC, are
rare in non-White populations. In populations of European origin, non-melanoma
skin cancer especially SSC, incidence rates are lower in people with ethnically
darker skins.

6.7.2 Place of Residence

The incidence of melanoma is greatest in the sun-sensitive White population of


Australia residing in an area of high ambient sunlight and increases with proximity
to the equator in many populations. In Europe, however, incidence of melanoma is
higher in north including Norway and Sweden as compared to south including
France, Italy, and Spain. The incidence of BCC is greatest in regions of Australia
and the United States closest to the equator [18]. The incidence rate of SCC in
Australia, the United States, and Norway increases with increasing proximity to the
equator.

6.8 Factors Responsible for Photocarcinogenesis

6.8.1 Occupational Exposure

The incidences of non-melanocytic skin cancer are more common with outdoor
occupation. However, more recent studies have shown much less evidence of such
an association. The incidences of melanoma have high rate in indoor as compared
to outdoor workers. The mortality rates are generally high in such populations.
Relationship between melanoma and sun exposure is somewhat complicated.
Socioeconomic status of people is also linked with the incidence of melanoma.
Higher rates have been observed among individuals belonging to high socioeco-
nomic status than that of low socioeconomic status. Socioeconomic status is pre-
sumed to be due to complex effects of sun exposure. The increase may be due to
real-time rise in incidence as a result of increased sun exposure as well as improved
detection rate and awareness about the disease in population [19].
60 N. Yadav and M. Banerjee

There is no direct evidence of melanoma incidence on occupational exposure. A


study from Southern Europe has shown positive correlation between photocarcino-
genesis and occupational exposure. In office workers, photocarcinogenesis inci-
dence is highest on body sites that usually are covered due to heterogeneity in
melanin and melanocyte transformation at those sites, whereas, in outdoor workers,
the incidence is high on sites that are usually exposed. However, photocarcinogen-
esis incidences observed may be uneven because there are differences in the distri-
butions of indoor and outdoor workers in the population, thereby, resultant
differences in distributions of skin cancer by body site are detected [20, 21].
Exposure to sunlight at work is certainly not associated with high BCC risk.

6.8.2 Anatomic Site

Highest density of melanoma is usually observed on the exposed parts such as head
and neck region and lowest density on rarely exposed sites (abdomen and buttocks
in both genders and scalp in women). Low density of melanoma is observed on the
forearms, upper arms, back of hands, and lower limbs. Intermediate density is
observed on the less-exposed areas such as the shoulders and back in both genders
and the chest in males. The highest density of photocarcinogenesis is observed on
the usually exposed sites and the lowest on the rarely exposed sites. Intermediate to
low concentrations of BCC occur on sometimes exposed sites, including the trunk.

6.8.3 Total Lifetime Exposure

Total lifetime exposure of human body to the sun is measured in hours. There is a
strong dose-dependent association between total lifetime exposure and photocarcino-
genesis. However, according to a study conducted in the United States and Canada, no
increased risk of photocarcinogenesis was observed. There is no direct evidence of the
development of BCC after occupational and non-occupational sun exposure.
Although photocarcinogenesis is dependent on age and gender probably due to thick
skin in male body as compared to female, elderly people have more frequent inci-
dence of photocarcinogenesis.Though, some studies conducted in Western Australia
has shown correlation between site of sun exposure and skin cancer incidences.
However, BCC risk does not correlate well with total site-specific sunlight exposure.
Relative photocarcinogenesis risk in the head, neck, trunk, and limb area increases
with increasing overall lifetime exposure to sunlight (Fig. 6.5) [21].

6.8.4 Ambient Sunlight Exposure

Exposure to ambient sunlight affects human skin aging and photocarcinogenesis in


many ways. Exposure of a person to ambient UV radiation may surpass the thresh-
old level, especially in summers. In summers, daily dose received may go beyond
6 Epidemiological Aspects of Photocarcinogenesis 61

Fig. 6.5 A person’s


lifetime risk of melanoma

70 SED (standard erythemal doses) [22]. The SED is independent of skin type
and is the same as 100 J.m−2 erythemal radiant exposure. The median daily exposure
at Antarctic region has been measured 3.2 SED. In Western Australia, increasing
intensity of ambient solar radiation has directly proportional effect on photocar-
cinogenesis risk at all places of residence. In California, increased rates of BCC
have been observed as compared with those who had lived in northeastern states.
US veterans of World War II, who served in the Pacific, were more likely than their
counterparts who served in Europe to have an SCC.

6.8.5 Safety Guidelines

In the United States, the Food and Drug Administration (FDA) and the Environmental
Protection Agency (EPA) have designed safety guidelines to protect both the gen-
eral public and occupational workers from harmful effects of solar radiation. Entire
range of facilities and equipment producing EMR (electromagnetic radiation)
should follow their exposure guidelines. These guidelines bound UV exposure to
normal levels as compared to those levels likely to cause adverse health effects.

6.9 Conclusion

There is large variability in distribution and amount of melanin of the skin. Severe
sunburn during early age may affect the expansion of melanoma cancer on sun-­
exposed areas. Occupation provides little evidence that sunlight causes skin cancer.
Nevertheless, increased melanoma risk has been found more frequent in indoor
workers. The place of residence, anatomic site of exposure, ethnic origin, and ambi-
ent intensity of sunlight affect the rate of incidence of photocarcinogenesis. Growth
of melanoma may be inevitable by consistent application of sunscreen.
62 N. Yadav and M. Banerjee

References
1. Battie C, Verschoore M (2012) Cutaneous solar ultraviolet exposure and clinical aspects of
photodamage. Indian J Dermatol Venereol Leprol 78(7):9
2. Whiteman DC, Watt P, Purdie DM, Hughes MC, Hayward NK, Green AC (2003) Melanocytic
nevi, solar keratoses, and divergent pathways to cutaneous melanoma. J Natl Cancer Inst
95(11):806–812
3. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T (2013) UV radiation and the skin. Int J Mol Sci
14(6):12222–12248
4. Wang SQ, Setlow R, Berwick M, Polsky D, Marghoob AA, Kopf AW, Bart RS (2001)
Ultraviolet A and melanoma: a review. J Am Acad Dermatol 44(5):837–846
5. Pittayapruek P, Meephansan J, Prapapan O, Komine M, Ohtsuki M (2016) Role of matrix
metalloproteinases in photoaging and photocarcinogenesis. Int J Mol Sci 17(6):868
6. Bosch R, Philips N, Suárez-Pérez JA, Juarranz A, Devmurari A, Chalensouk-Khaosaat J,
González S (2015) Mechanisms of photoaging and cutaneous photocarcinogenesis, and pho-
toprotective strategies with phytochemicals. Antioxidants 4(2):248–268
7. Chu CY, Cha ST, Chang CC, Hsiao CH, Tan CT, Lu YC, Jee SH, Kuo ML (2007) Involvement
of matrix metalloproteinase-13 in stromal-cell-derived factor 1 [alpha]-directed invasion of
human basal cell carcinoma cells. Oncogene 26(17):2491
8. Prasad NB, Fischer AC, Chuang AY, Wright JM, Yang T, Tsai HL, Westra WH, Liegeois NJ,
Hess AD, Tufaro AP (2014) Differential expression of degradome components in cutaneous
squamous cell carcinomas. Mod Pathol: Off J U S Can Acad Pathol Inc 27(7):945
9. Leight JL, Tokuda EY, Jones CE, Lin AJ, Anseth KS (2015) Multifunctional bioscaffolds for
3D culture of melanoma cells reveal increased MMP activity and migration with BRAF kinase
inhibition. Proc Natl Acad Sci 112(17):5366–5371
10. Harris NR, Kyrö E, Staehelin J, Brunner D, Andersen SB, Godin-Beekmann S, Dhomse S,
Hadjinicolaou P, Hansen G, Isaksen I Jrrar A (2008) Ozone trends at northern mid-and high
latitudes–a European perspective. Ann Geophys 26(5):1207–1220. Copernicus Publications
on behalf of the European Geosciences Union, Germany
11. Koepke P, Mech M (2005) UV irradiance on arbitrarily oriented surfaces: variation with atmo-
spheric and ground properties. Theor Appl Climatol 81(1):25–32
12. Roberts DF, Kahlon DPS (1976) Environmental correlations of skin colour. Ann Hum Biol
3(1):11–22
13. Slominski A, Tobin DJ, Shibahara S, Wortsman J (2004) Melanin pigmentation in mammalian
skin and its hormonal regulation. Physiol Rev 84(4):1155–1228
14. Nan H, Kraft P, Hunter DJ, Han J (2009) Genetic variants in pigmentation genes, pigmentary
phenotypes, and risk of skin cancer in Caucasians. Int J Cancer 125(4):909–917
15. Ricotti C, Bouzari N, Agadi A, Cockerell CJ (2009) Malignant skin neoplasms. Med Clin N
Am 93(6):1241–1264
16. MacKie RM, Hauschild A, Eggermont AMM (2009) Epidemiology of invasive cutaneous
melanoma. Ann Oncol 20(suppl_6):vi1–vi7
17. Bellew S, Del Rosso JQ, Kim GK (2009) Skin cancer in asians: part 2: melanoma. J Clin
Aesthet Dermatol 2(10):34
18. Marks R, Staples M, Giles GG (1993) Trends in non-melanocytic skin cancer treated in
Australia: the second national survey. Int J Cancer 53(4):585–590
19. Johnson-Obaseki SE, Labajian V, Corsten MJ, McDonald JT (2015) Incidence of cutaneous
malignant melanoma by socioeconomic status in Canada: 1992–2006. J Otolaryngol-Head
Neck Surg 44(1):53
20. Gallagher RP, Hill GB, Bajdik CD, Fincham S, Coldman AJ, McLean DI, Threlfall WJ (1995)
Sunlight exposure, pigmentary factors, and risk of nonmelanocytic skin cancer: I. Basal cell
carcinoma. Arch Dermatol 131(2):157–163
6 Epidemiological Aspects of Photocarcinogenesis 63

21. Vågero D, Ringbäck G, Kiviranta H (1986) Melanoma and other tumors of the skin among
office, other indoor and outdoor workers in Sweden 1961–1979. Br J Cancer 53(4):507–512
22. Kricker A, Armstrong BK, English DR (1994) Sun exposure and non-melanocytic skin cancer.
Cancer Causes Control 5(4):367–392
23. Allen M, McKenzie R (2005) Enhanced UV exposure on a ski-field compared with exposures
at sea level. Photochem Photobiol Sci 4(5):429–437
24. Halder RM, Bridgeman-Shah S (1995) Skin cancer in african americans. Cancer
75(S2):667–673
Photoaging
7
Jyoti Singh, Deepti Chopra, Ashish Dwivedi,
and Ratan Singh Ray

Abstract
The incident of photoaging mainly depends upon the intensity of UV-R and the
amount of melanin present in the skin. UV-R is known to cause photoaging,
photoallergy, and immune suppression to human skin, noted more than a century
ago. From last 10 years, several laboratory studies show that UV rays impaired
the collagen synthesis, blocked collagen expression, and reduced the elasticity of
skin and solar scar formation which is ultimately visible by clinical pattern such
atrophy and wrinkle formation. Which further leads to UV-R induced premature
aging of the skin. UV radiation alters the ECM by raising the level of matrix
metalloproteinases (MMP), decreasing the structural elastin and collagen,
directly or indirectly damaging the DNA, and enhancing the cell surface recep-
tors which are present at the surface of keratinocytes and fibroblasts of skin.
AP-1 and NF-kB are key signaling molecules which involve in UV-R promoted
skin aging. The photoprotective approaches to prevent or treat photocarcinogen-
esis and photoaging involve natural supplements absorption by orally and topi-
cally. Skin accommodates a complex system of endogenous enzymatic and

J. Singh
Photobiology Laboratory, Systems Toxicology and Health Risk Assessment Group,
CSIR-­Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
D. Chopra
Photobiology Laboratory, Systems Toxicology and Health Risk Assessment Group,
CSIR-­Indian Institute of Toxicology Research (CSIR-IITR), Lucknow, Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
A. Dwivedi
Pineal Research Lab, Department of Zoology, Institute of Science, Banaras Hindu University,
Varanasi, Uttar Pradesh, India
R. S. Ray (*)
Photobiology Division, CSIR- Indian Institute of Toxicology Research (IITR),
Lucknow, Uttar Pradesh, India

© Springer Nature Singapore Pte Ltd. 2018 65


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_7
66 J. Singh et al.

nonenzymatic photoprotective antioxidants. However, their role in the UV-R-­


induced oxidative damage has not been fully elucidated. Recently, the research-
ers have elucidated that skin antioxidative defense system is increased in presence
of vitamins and nutritive agents and combination of various kinds of antioxidants
also produce synergistic results.

Keywords
Photoaging · MMPs · AP-1 · NFKB · Collagen

7.1 Introduction

Skin comprises 16% of total body mass; it is the largest body organ which acts as a
physical barrier and protects body organ from direct environmental stress and ultra-
violet radiation (UV-R). Exposure to UV-R is either acute or chronic. Acute expo-
sure of skin to UV-R shows local changes such as (1) inflammation marked by
erythema, edema, and enhanced cytokine level; (2) programmed cell death of dam-
aged cells; and (3) keratinocyte proliferation which ends up in hyperplasia of epi-
dermis [1]. In addition to acute effects, chronic UV-R exposure to skin was a major
cause of photoaging as well as skin cancer development. UV-R exposure to human
or mouse skin leads to initiation of matrix metalloproteinases (MMPs) secretion,
which is a major reason of photoaging. MMPs have shown a key role in photocar-
cinogenesis by regulating/affecting a range of processes like tumor initiation,
growth, angiogenesis, and metastasis which is related to tumor. Experimentally it
has been proved that MPPs are a key mediator for degradation of collagen fiber.
Collagen fibers are mainly responsible for skin strength and resilience; it is synthe-
sized by procollagen also known as collagen precursor molecule secreted by dermal
fibroblast [2]. Procollagen is secreted in extracellular dermal space where it is enzy-
matically processed to collagen and form cross-linked stabilized collagen fibrils.
Estimated half-life of these collagen fibers is 17 years. The clinical pattern for pho-
toaging is recognized by fine and coarse wrinkles, dyspigmentation, fragility, and
coarse skin texture. UV-R-mediated dermal aging is divided in two primary path-
way (1) collagen breakage and (2) inhibition of procollagen biosynthesis. Reactive
oxygen species (ROS) also play a key role in photoaging by increasing the expres-
sion and synthesis of MMPs in dermal fibroblast exposed to UV-R. This chapter
summarized the role of UV-R in photoaging.

7.2  linical and Histopathological Manifestation


C
of Photoaging

Photoaged skin comprises various patterns such as dryness, rough skin, dyspigmen-
tation, telangiectasia, yellowish color, thick plaque, and fine lines [3]. At the same
place, systemically, clinical changes are also exhibited by the change in histology of
7 Photoaging 67

epidermis and dermis of skin. Appearance of photoaged skin's epidermis layer may
be in a normal form or may show change such as hyperplasia (increased number of
cells); loss of dermal bumps; condense basement membrane, with more irregularly
distributed melanocytes and melanosomes; and thick stratum corneum in keratino-
cytes. In response to all these alterations, irregular pigmentations and skin rough-
ness occur [3]. The deposition of elastotic material in dermis layer (solar elastosis)
is an indicator of photoaged skin. Previously it has been demonstrated by transmis-
sion electron microscopy that fragmentation and homogenization of elastotic mate-
rial occurred in a fully developed photoaged skin. Additional examination of
photoaged skin’s dermis and epidermis layer illustrates the occurrence of disordered
collagen fibers, reduced number of collagen, higher quantity of the ground sub-
stance, and enlarge blood vessels [4]. Likewise, the dermis layer of photodamaged
skin is recognized by increased elastosis and collagen fragmentation on the junction
of the dermal-epidermal layer. It is found that photoaged skin showed the elevated
level of glycosaminoglycans and proteoglycans which further induced MMPs pro-
duction, and it also increased the number of hyperplastic fibroblasts [5]. The thick-
ness of the epidermal layer is irregular because of the morphology of epidermal cell
changes. Meanwhile, skin type has also an essential role in the visible identification
of photoaging. People with skin type I and II showed an atopic sign with less amount
of wrinkle formation and dysplastic premalignant induction like actinic keratosis.
However, in the case of type III and IV, skin displays hypertrophic feature with very
vigorous wrinkle formation and leathery skin with bronzed appearance due to the
accumulation of elastosis [6].

7.3 UV-R-Induced Signal Transduction in Photoaging

7.3.1 Role of MMPs in Photoaging

MMPs are an assembly of enzymes which is accountable for the deterioration of


extracellular matrix (ECM) proteins throughout the time of growth, organogenesis,
skin aging, and tissue turnover. In normal condition, the activity and expression of
MMPs are slightly low, while in the case of inflammatory disease, metastasis, and
tumor, its expression raises significantly. Till date approx. 28 MMPs have been
identified in various pathophysiological conditions including photodamaged skin,
wound healing, inflammatory disease, cancer, photoaging, etc. MMPs, zinc-­
containing endopeptidases, are excreted from the keratinocytes and dermal fibro-
blast after exposure to external stimuli like oxidative stress, UV-R, and cytokines.
These enzymes are classified into three domains: N-terminal propeptide, catalytic
domain, and C-terminal domain.
On the basis of substrate specificity and structure, these MMPs are divided into
five specific subgroups which are as follows: (1) Collagenases (MMP-1, MMP-8,
and MMP-13) degraded fibrillar collagen and also recognize the substrate through a
hemopexin-like domain; (2) gelatinases (MMP-2 and MMP-9) have ability to
decay various types of ECM parts such as collagen types I and IV; (3) stromelysins
68 J. Singh et al.

(MMP-3, MMP-10, and MMP-11) have a domain positioning same like collage-
nases, but they do not have tendency to cut collagen type I; and (4) matrilysins
(MMP-7 and MMP-26) don’t have organization like hemopexin domain and only
breakdown collagen IV. Another category of MMPs are (5) MT-MMPs (MMP-14,
MMP-15, and MMP-16); this is membrane-type MMPs and has extra C-terminal
transmembrane domain with a small cytoplasmic tail. Similarly, MMP-14 and
MMP-16 break collagen type I and are situated on the outer surface of cell. UV-R
has two alternate mechanisms to affect/regulate MMPs, first is direct mechanism in
which UV-R (UV-A and UVB) is directly absorbed by dermal fibroblast or kerati-
nocyte. Second mechanism is indirect where UV-R-induced cytokine production
leads to stimulation of ECM and MMPs [7]. Photoaging is mainly caused by loss of
balance in the middle of accumulation and reduction of ECM components that pro-
vide essential anatomical and physiological support to the skin tissue. It is often
seen that continuous exposure to sunlight degraded the skin protein such as elastin
and collagen and also reduces the rate of renewal of these protein synthesis. Dermis
layer of skin has fibroblast which is mainly responsible for the synthesis of collagen
protein and conveys elasticity and strength to the skin. In ECM components, colla-
gen is fibrous protein and found in insoluble form in connective tissue of skin.
However, connective tissue of skin contains type I collagen in plenty amount but
type III protein in small quantity [8]. MMP-1, an enzyme which is deregulated after
UV-R exposure and degrades elastin and collagen fibers (type I and type III in skin)
at single site within its triplet complex [9]. These degraded collagen fibers are fur-
ther cleaved by increased level of MMP-3 and MMP-9 which is experimentally
proved by in vivo study. MMP-1, MMP-3, and MMP-9 have capability to damage
all ECM proteins. However, the role of other member of MMPs in UV-R-mediated
skin aging is also reported but in less frequency. Level of MMP-1 and MMP-3 has
been increased severalfold after irradiation of UV-R. However, MMP-9 mRNA
expression level increases slowly. Earlier studies have also recommended a con-
fined role of MMP-8 in UV-induced collagen disturbance in the skin. However the
expression of this enzyme in UV-induced cases was upregulated at minimum level.
Novel inhibitor of MMP is promising as targets to fight with photoaging. Recently
researcher showed their interest in plant-based additive for the blockage of photo-
damage. A natural conventional Chinese drug, Galla chinensis, considerably sup-
pressed the UVB causes elevated level of MMP-1 and ROS in dermal fibroblasts
[10]. Similarly, Neonauclea reticulata, which belongs from Rubiaceae family, sig-
nificantly downregulates the expression of MMP-1, MMP-3, and MMP-9 by inhib-
iting phosphorylation of ERK, p38, and JNK [11]. Ixora parviflora and Coffea
arabica also from Rubiaceae group showed anti-photoaging property by downregu-
lating the level of MMP-1, MMP-3, and MMP-9 and MAPK activity [12, 13].

7.3.2 Role of Mitochondria in Photoaging

Mitochondria are cellular organ and act as power plant; it behaves like local power
plant which provides electricity to whole city in the same manner mitochondria
7 Photoaging 69

provide strength or energy through the generation of ATP (adenosine triphosphate)


by the breakdown of carbohydrate and fatty acid. Cellular metabolism capacity of
mitochondria declines with age; it is assumed that decline in respiratory function is
related to skin aging, due to accumulation of somatic mitochondrial DNA (mtDNA)
mutation. It is considered that mtDNA mutations are involved in the photoaging
which confirmed by the close examination of chronically UV-exposed skin has clin-
ical signs of damaged photoaged and has a high number of damaged mtDNA [14].
Biopsy and molecular study of photodamaged skin showed increased mutation in
mitochondrial genome. The most frequent mutation is 4977 base-pair mutation
increased up to tenfold in photodamaged skin compared to sun-protected skin [15].
UV-R reaches up to skin; UVB (280–320 nm) is absorbed in the epidermal kerati-
nocytes and induces nuclear DNA and mtDNA damage. UVA (320–400 nm) has
longer wavelength able to penetrate up to basal layer of the epidermis where ROS
generation may initiate and induce damage to mtDNA indirectly. High levels of
ROS were located in mitochondria. It has been found that photoaging is ROS-­
generated, mtDNA mutation, which in the future leads to defective respiratory
chain. This defective respiratory chain and its vicious cycle induced more ROS
generation and mtDNA mutation without depending on primary causing agent. This
leads to increased level of 4977 base deletions also known as “common deletion” up
to 32-fold; it does not depend on UV-R exposure. Recently, in vitro study performed
by Mark Berneburg and colleague showed a clear relation between ROS generation
and most frequently occurred mtDNA deletion known as common deletion. Mark
Berneburg and colleagues have used in vitro model of skin fibroblast and showed
that the “common deletion” can be exhibited in time- and dose-dependent manner
under repetitive exposure of UV-A for 3 weeks. This study also demonstrates that
UV-A mediated “common deletion” caused by the type II phototoxicity mechanism
means by the generation of singlet oxygen [16]. This in vitro study further extended
to in vivo which demonstrates an association between chronically sun-exposed skin
showed photoaging and mtDNA mutations mutation [17].

7.3.3 Signal Transduction in Photoaging

Nucleated and enucleated cells exhibit different types of response under UV-R; one
is nucleus dependent and other is nucleus independent. Nucleus-dependent response
is consequence of direct absorption of UV-R by DNA, lipids, proteins, and urocanic
acid. As a consequence UV-R-irradiated DNA induces mutation due to error in
DNA repair system [18]. Nucleus-independent response occurs at the surface or
near the surface of cell which moves ahead with the help of signal transduction
pathway associated with cytokines and growth factor signaling. This ligand-­
independent receptor is activated by singlet oxygen which initiates signaling path-
way involving MAP kinases and interrelated autocrine cytokine loops (Il-a, Il-b, and
Il-6) which further enhances the expression of MMPs. Study by Fisher et al. indi-
cated that UV-R-induced transcription may play a key role in increased expression
of MMPs. It has been investigated that after the exposure of UVB, skin enhanced
70 J. Singh et al.

the level of transcription factor AP-1 and NF-kB, and these agents are stimulators of
MMPs gene. The transcription factor AP-1 (activator protein-1) is responsible for
many processes such as transformation, cellular proliferation, and cell death [19].
At the same time, AP-1 impaired dermal collagen synthesis and blocked the colla-
gen gene expression in dermal fibroblasts. NF-kB has potential to amplify AP-1 and
releases more NF-kB, resulting in more photodamage. Low dose of UVB induced
c-Jun, which is transcription factor AP-1 which further activates MAP kinases. The
Michigan group has found that UV irradiation affects the regulation of procollagen
synthesis in human skin in vivo and investigated that this effect is reversed with
tretinoin 0.1%. UV-R activates all cell surface receptor which is correlated with
activation of multiple ligands, receptors, and signaling pathways such as MAP
kinases: ERK, JNK, AP-1, PI3K/Akt, and NF-kB. Acute activation of these three
MPKs leads to upregulation of c-Jun transcription factor; c-Jun heterodimerizes by
continuously producing transcription factor c-Fos to make a highly active AP-1
transcription factor complex in skin [20]. AP-1-elevated level of expression is
mainly responsible for promotion of key player of matrix-degrading metalloprotein-
ase (MMP). AP-1 is the main reason for expressing MMP in keratinocytes and skin
fibroblasts [21]. UV-R-induced activation of NF-kB is well known fact. However, it
was found that the activation mechanism NF-kB by UV irradiation is significantly
different from mechanism by which cytokines trigger NF-kB (Fig. 7.1). Earlier

Cytokine FB/KC
Growth factor

Signal transduction pathway


cytoplasm

C-Jun c-Fos
nucleus
DNA damage AP-1
NBKB
cytokine promoter Procollagen MMP
promoter promoter

Increased MMP increased


proinflammtory expression ECM
cytokine Increased pro collagen
synthesis

Fig. 7.1 Schematic of molecular mechanism of photoaging


7 Photoaging 71

studies suggest that UV irradiation stops synthesis of IkB, known as NF-kB inhibi-
tor. This blocking finally leads to NF-kB activation, while on the other hand, IkB
degraded by ubiquitin-proteasome degradation pathway [22, 23]. Already exited
evidences indicate that ROS, which is produced by UV irradiation, can interfere IkB
degradation and NF-kB translocation [24].

7.4 Safety Measures for Photoaging

To protect our skin from overexposure of solar radiation melanin, skin produces
melanin, which can quench free radical naturally. Several studies carried out by
researcher mainly pTpT (thymine dinucleotides) an oligonucleotides are responsi-
ble for skin tanning; in addition to this, pTpT induces enhanced repair of UV-induced
DNA damage. It can also protect skin from photocarcinogenesis and photoaging
naturally (Fig. 7.2). However, there are several other strategies reported by the
researcher for the protection of skin from the harmful effect of UV-R.

7.4.1 Antioxidants

The skin has a collection of protective antioxidant enzymes which play a key role in
neutralizing the toxic effect. The skin is fitted with a huge network of endogenous

Sun
protection

DNA repair enzymes

UV Radiation

Antioxidants, Polyphones

SKIN
Collagen
degradation
increased

Fig. 7.2 Strategy for the protection from photoprotection


72 J. Singh et al.

enzymatic antioxidant protection system like glutathione (GSH peroxidase (GPx),


superoxide (SOD), and catalase as well as nonenzymatic antioxidants (vitamin E,
vitamin C, uric acid, ascorbate, and ubiquinol)). Various examinations have been
completed to protect the skin from photoaging and reported antioxidants which can
protect our skin from a different type of ROS. Laboratory studies conducted on skin
model and animal model suggested that all retinoic acid derivatives are responsible
for inhibition of photoaging by activating the expression of AP-1 with upregulated
gene expression of collagen type 1 (COL1A1) and collagen type 3 (COL3A1)
involved in procollagen I and procollagen III protein expression [19]. Recently,
scientists have paid their more attention to identification of photostable agents
which can protect our skin from photoaging and photocarcinogenesis. Similarly,
derivatives of tocopherols have also showed their protective effect when they are
applied topically on mice skin, which chronically exposed to UVB radiation.
Besides this, resveratrol-enriched rice (RR) and resveratrol (R) inhibited the photo-
aging in UVB-irradiated human dermal through reducing the MMP-1 expression
and enhancing the procollagen production. Photoprotective effects of beta-carotene
originated due to antioxidant properties though some studies showed prooxidant
effects of beta-carotene. It significantly improved facial wrinkles and elasticity in
the low-dose group [25]. Similarly, another study showed tocopherol sorbate have
high potential against skin photoaging in comparison to α-tocopherol and tocoph-
erol acetate. It also showed reduction in radical flux in Skh-1 hairless mouse skin
[26]. In vivo chronic exposure to UVB irradiation in Skh-1 hairless mouse skin
leads to premature skin aging characteristic of photoaged.

7.4.2 DNA Damaging Repair Enzyme

DNA repair enzyme in photoaging is emerging as a new technology. Bacteriophage-­


derived enzyme T4 endonuclease V specific for the CPDs lesions (most common
DNA under UV-R) starts repair system through enhancing their cleavage [27].
Encapsulating the enzyme in liposomes facilitates its delivery into the skin by inci-
sion of UV-irradiated nuclear and mitochondrial DNA damage, DNA repair, and
cell survival in UV-irradiated cell. T4 endonuclease V also suppressed the level of
IL-10 and IL-4 cytokine level, which further reduce the chances of skin cancer [28].
8-oxoguanine DNA glycosylase 1 (OGG1) is a DNA repair enzyme that excises
7,8-dihydro-8-oxoguanine (8oxoG) from damaged DNA. OGG1 decreased level
indicated toward the high risk of skin cancer under the oxidative stress condition.
Previous study confirmed that plant originate liposome-encapsulated OGG1,
increased the number of 8OG removal from human epidermal keratinocytes which
encountered with oxidative damage. Rupert and his colleagues observed that E. coli
embraced with a light-activated enzyme that repaired the UV-R caused DNA dam-
age known as photoreactivating enzyme, which is later called as photolyase enzyme
[29]. Earlier, only CPDs lesion was repaired by these photolyase enzymes; they
were not specific for 6–4 PP. In 1993 a photolyase that repairs the (6–4 PPs) photo-
product was also discovered [30].
7 Photoaging 73

7.4.3 Phytochemical

It is a speculation that naturally occurring compounds, such as phytochemicals, are


accountable for protective health benefits against oxidative stress. Polyphenols act
like an anti-inflammatory, immunomodulatory, and antioxidant agent, and they are
also recommended as a chemopreventive compound in skin disorders and cancer [31].
Researchers focused their attention toward green tea; a polyphenol showed protection
against UV-induced skin damage. Published literature illustrated the role of green tea
in anti-inflammation and anti-carcinogenic as well as anti-photoaging process [32].
Furthermore low and high dose of beta-carotene supplementation increased the
mRNA expression of type I procollagen and enhanced procollagen synthesis with the
reduction in 8-hydroxy-2′-deoxyguanosine in human skin in in vivo; this study con-
cludes that beta-carotene supplementation prevents and repair photoaging [25].
Flavonoids are an assembly of plant phenolics (plant produce a phenolic compound
for defense against oxidative stress) and act as an antioxidant and chelating agent.
Anthocyanin-rich extract from bog blueberry (ATH-BBe) has shown a tendency to
prevent photoaging in UVB-irradiated human dermal fibroblasts [33]. Soybean and
soybean products contain soy isoflavones and have been found to possess many physi-
ological activities like antioxidant activity, free radical scavenger, and anticancerous
activity and prevent osteoporosis. A study has shown that soy isoflavone extract ISO-1
(containing 12 soy isoflavones) from soybean prevents damage in UVB-irradiated
human keratinocyte by increasing the level of catalase and suppressing the expression
of cox2. Therefore, soy isoflavone extract can act as an anti-photoaging agent for skin
[34]. Punica granatum (pomegranate) is a fruit eaten up raw or in drinkable form. It
consists catechin, quercetin, kaempferol, and equol. UVB-irradiated fibroblast treated
with Punica granatum increased the level of procollagen I and reduced the expression
of MMP-1. Catechin is a key compound of Punica granatum and plays an important
role in prevention of photoaging of skin [35]. Artificial skin models irradiated with
UV-A, made of continually dividing epidermis, direct EGCG (epigallocatechin-3-gal-
late) application suppressed the activity of MMP-1, MMP-3, MMP-2, and -9 in skin
model which is mainly involved in degradation of collagen in skin. A number of fla-
vonoids known as scavengers of reactive oxygen species, UV-R neutralizer, cytopro-
tective, anti-­inflammatory anti-apoptotic, and DNA damage inhibitor and to affect
cellular signaling pathways [31].

7.4.4 Sun Protection

The best way to protect ourselves from photoaging is to protect our skin from dam-
aging UV-R exposure by adopting some safety tips that really do work in case of
premature skin protection. Stay in shade or use umbrella in sunny midday especially
between 10am and 4pm when the sunlight exposure is at its highest point in the sky.
Use sunscreen on exposed area of the body every day with SPF 30 or more than this.
Sunscreen should be reapplied after 3–4 h or when exposed to water and excessive
sweating. Cover up with UV-protected clothes and sunglasses. It is suggested to visit
your dermatologist yearly for professional examination of skin.
74 J. Singh et al.

7.5 Conclusions

UV-R is one of the potential factors for the premature skin aging. UV-R-induced
ECM remodeling resulted in wrinkle formation. However, a number of antioxi-
dants, DNA repair enzymes technology, and photochemical are known to prevent
premature aging in human. Therefore, knowledge of photoaging, its clinical pattern,
and molecular mechanism with some protective strategy will be a small initiation to
protect our society from the harmful effect of UV-R.

References
1. D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T (2013) UV radiation and the skin. Int J Mol Sci
14(6):12222–12248
2. Dong KK, Damaghi N, Picart SD, Markova NG, Obayashi K, Okano Y, Masaki H, Grether-­
Beck S, Krutmann J, Smiles KA, Yarosh DB (2008) UV-induced DNA damage initiates release
of MMP-1 in human skin. Exp Dermatol 17(12):1037–1044
3. Gilchrest BA (1989) Skin aging and photoaging: an overview. J Am Acad Dermatol
21(3):610–613
4. Oikarinen A (1990) The aging of skin: chronoaging versus photoaging. Photodermatol
Photoimmunol Photomed 7:3–4
5. Schwarz T (2002) Photoimmunosuppression. Photodermatol Photoimmunol Photomed
18(3):141–145
6. Yaar M, Gilchrest BA (2007) Photoageing: mechanism, prevention and therapy. Br J Dermatol
157(5):874–887
7. Kossodo S, Wong WR, Simon G, Kochevar IE (2004) Effects of UVR and UVR-induced cyto-
kines on production of extracellular matrix proteins and proteases by dermal fibroblasts cul-
tured in collagen gels. Photochem Photobiol 79(1):86–93
8. Chiang HM, Chen HC, Lin TJ, Shih IC, Wen KC (2012) Michelia alba extract attenuates UVB-­
induced expression of matrix metalloproteinases via MAP kinase pathway in human dermal
fibroblasts. Food Chem Toxicol 50(12):4260–4269
9. Sorsa T, Tjäderhane L, Salo T (2004) Matrix metalloproteinases (MMPs) in oral diseases. Oral
Dis 10(6):311–318
10. Sun ZW, Hwang E, Lee HJ, Lee TY, Song HG, Park SY, Shin HS, Lee DG, Yi TH (2015)
Effects of Galla chinensis extracts on UVB-irradiated MMP-1 production in hairless mice.
J Nat Med 69(1):22–34
11. Chiang HM, Chen HC, Chiu HH, Chen CW, Wang SM, Wen KC (2013) Neonauclea reticu-
lata (Havil.) Merr stimulates skin regeneration after UVB exposure via ROS scavenging and
modulation of the MAPK/MMPs/collagen pathway. Evid Based Complement Alternat Med
2013:324864
12. Chiang HM, Lin TJ, Chiu CY, Chang CW, Hsu KC, Fan PC, Wen KC (2011) Coffea arabica
extract and its constituents prevent photoaging by suppressing MMPs expression and MAP
kinase pathway. Food Chem Toxicol 49(1):309–318
13. Wen KC, Fan PC, Tsai SY, Shih I, Chiang HM (2012) Ixora parviflora protects against UVB-­
induced photoaging by inhibiting the expression of MMPs, MAP kinases, and COX-2 and by
promoting type I procollagen synthesis. Evid Based Complement Alternat Med 2012:417346
14. Krutmann J, Schroeder P (2009) Role of mitochondria in photoaging of human skin: the defec-
tive powerhouse model. J Investig Dermatol Symp Proc 14(1):44–49 Elsevier
15. Larsson NG (2010) Somatic mitochondrial DNA mutations in mammalian aging. Annu Rev
Biochem 79:683–706
7 Photoaging 75

16. Berneburg M, Grether-Beck S, Kürten V, Ruzicka T, Briviba K, Sies H, Krutmann J (1999)


Singlet oxygen mediates the UVA-induced generation of the photoaging-associated mitochon-
drial common deletion. J Biol Chem 274(22):15345–15349
17. Berneburg M, Plettenberg H, Krutmann J (2000) Photoaging of human skin. Photodermatol
Photoimmunol Photomed 16(6):239–244
18. Rastogi RP, Kumar A, Tyagi MB, Sinha RP (2010) Molecular mechanisms of ultraviolet
radiation-­induced DNA damage and repair. J Nucleic Acids 2010
19. Fisher GJ, Datta SC, Talwar HS, Wang ZQ, Varani J, Kang S, Voorhees JJ (1996) Molecular
basis of sun-induced premature skin ageing and retinoid antagonism. Nature 379(6563):335
20. Fisher GJ, Talwar HS, Lin J, Lin P, McPhillips F, Wang Z, Li X, Wan Y, Kang S, Voorhees JJ
(1998) Retinoic acid inhibits induction of c-Jun protein by ultraviolet radiation that occurs
subsequent to activation of mitogen-activated protein kinase pathways in human skin in vivo.
J Clin Invest 101(6):1432–1440
21. Fisher GJ, Voorhees JJ (1998) Molecular mechanisms of photoaging and its prevention by
retinoic acid: ultraviolet irradiation induces MAP kinase signal transduction cascades that
induce Ap-1-regulated matrix metalloproteinases that degrade human skin in vivo. J Investig
Dermatol Symp Proc 3(1):61–68 Elsevier
22. Li N, Karin M (1998) Ionizing radiation and short wavelength UV activate NF-κB through two
distinct mechanisms. Proc Natl Acad Sci 95(22):13012–13017
23. Wu S, Tan M, Hu Y, Wang JL, Scheuner D, Kaufman RJ (2004) Ultraviolet light activates
NFκB through translational inhibition of IκBα synthesis. J Biol Chem 279(33):34898–34902
24. Schreck R, Albermann KA, Baeuerle PA (1992) Nuclear factor kB: an oxidative stress-­
responsive transcription factor of eukaryotic cells (a review). Free Radic Res Commun
17(4):221–237
25. Cho S, Lee DH, Won CH, Kim SM, Lee S, Lee MJ, Chung JH (2010) Differential effects of
low-dose and high-dose beta-carotene supplementation on the signs of photoaging and type I
procollagen gene expression in human skin in vivo. Dermatology 221(2):160–171
26. Jurkiewicz BA, Bissett DL, Buettner GR (1995) Effect of topically applied tocopherol on ultra-
violet radiation-mediated free radical damage in skin. J Investig Dermatol 104(4):484–488
27. Karakoula A (n.d.) Studies on UV-induced DNA damage and repair to human DNA. Doctoral
dissertation, Biochemistry
28. Ceccoli J, Rosales N, Tsimis J, Yarosh DB (1989) Encapsulation of the UV-DNA repair
enzyme T4 endonuclease V in liposomes and delivery to human cells. J Investig Dermatol
93(2):190–194
29. Sancar A (2008) Structure and function of photolyase and in vivo enzymology: 50th anniver-
sary. J Biol Chem 283(47):32153–32157
30. Eller MS, Maeda T, Magnoni C, Atwal D, Gilchrest BA (1997) Enhancement of DNA repair
in human skin cells by thymidine dinucleotides: evidence for a p53-mediated mammalian SOS
response. Proc Natl Acad Sci 94(23):12627–12632
31. Evans JA, Johnson EJ (2010) The role of phytonutrients in skin health. Nutrients 2(8):903–928
32. Roh E, Kim JE, Kwon JY, Park JS, Bode AM, Dong Z, Lee KW (2017) Molecular mechanisms
of green tea polyphenols with protective effects against skin photoaging. Crit Rev Food Sci
Nutr 57(8):1631–1637
33. Bae JY, Lim SS, Kim SJ, Choi JS, Park J, Ju SM, Han SJ, Kang IJ, Kang YH (2009) Bog blue-
berry anthocyanins alleviate photoaging in ultraviolet-B irradiation-induced human dermal
fibroblasts. Mol Nutr Food Res 53(6):726–738
34. Huang CC, Hsu BY, Wu NL, Tsui WH, Lin TJ, Su CC, Hung CF (2010) Anti-photoaging
effects of soy isoflavone extract (aglycone and acetyl glucoside form) from soybean cake. Int
J Mol Sci 11(12):4782–4795
35. Park HM, Moon E, Kim AJ, Kim MH, Lee S, Lee JB, Park YK, Jung HS, Kim YB
Drug-Induced Phototoxic Response
8
Syed Faiz Mujtaba, Ajeet K. Srivastav, Shikha Agnihotry,
and Mohammad Anas

Abstract
Phototoxicity induced by drugs is a non-immunological event that refers to the
development of rashes as a result of the combined effects of photosensitizer and
ultraviolet or visible radiation. The combinatorial effect of both light and chemi-
cal is necessary to elicit phototoxicity. Exposure to either light or chemical alone
is insufficient to induce an adverse effect; however, when photoactivation of the
chemical occurs, the abnormal reaction may arise. Mostly, chemicals having
peak absorption within the UVA spectrum (320–400 nm) cause drug-induced
photosensitivity reactions, although occasionally wavelengths within UV-B or
visible range also cause phototoxic reactions. Various drugs are available in the
market, but the awareness about their adverse effect is very little. Phototoxicity
is one of the adverse effects which people should be made aware of. The photo-
behaviour of few such drugs is being discussed here.

Keywords
Phototoxicity · UVR · Drugs · Photosensitizer · Photosensitivity

Authors Syed Faiz Mujtaba and Ajeet K. Srivastav have been equally contributed to this chapter.
S. F. Mujtaba (*)
Department of Zoology, Faculty of Science, Shia P.G. College, University of Lucknow,
Lucknow, Uttar Pradesh, India
A. K. Srivastav · M. Anas
Photobiology Division, CSIR-Indian Institute of Toxicology Research, Lucknow,
Uttar Pradesh, India
S. Agnihotry
Department of Biomedical-informatics, Sanjay Gandhi Post Graduate Institute,
Lucknow, India

© Springer Nature Singapore Pte Ltd. 2018 77


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_8
78 S. F. Mujtaba et al.

8.1 Introduction

The drugs which are being used for its therapeutical property has some side effects.
Phototoxicity is one such adverse effect which has not been much investigated. Not
all the drugs have this property of phototoxic response. Few drugs that elicit this
property have absorption maxima (λmax) in the range of UV radiation and visible
light (Fig. 8.1).
Phototoxicity is also termed as photoirritation, which can be defined as a skin
irritation requiring light and a photosensitizer without any involvement of the
immune system. It can also be called as a type of photosensitivity [1, 2] (Fig. 8.2).
The skin looks like an exaggerated sunburn after photosensitivity reaction. The
chemical may reach the skin by topical administration or through systemic circula-
tion. The chemical to exhibit phototoxicity must be “photoactive,” which means that
it must absorb visible or UV radiation, and then the absorbed energy produces
molecular changes that leads to adverse effects. Many synthetic compounds used to
cure different diseases such as fluoroquinolones, tetracyclines, etc. are known to
cause these effects. Table 8.1 shows the different effects related to photosensitiza-
tion of drugs. The damaging effects of photoactivated compounds on cellular struc-
tures such as DNA, cell membranes, and proteins lead to phototoxic reaction.
Phototoxicity can be elicited by many different compounds. At least one resonating
double bond or an aromatic ring is present in most compounds that can absorb radi-
ant energy. In most instances activation of a compound by light results in electron
excitation from the stable singlet state to an excited triplet state (Fig. 8.3).
Further the excited-state electrons transfer their energy to oxygen and return to a
more stable configuration forming reactive oxygen intermediates (Fig. 8.4).
Reactive oxygen intermediates like superoxide anion, singlet oxygen, and hydro-
gen peroxide damage various biomolecules like cell membranes and DNA (Fig. 8.5).
Psoralen is an exception in drug-induced phototoxic response. Psoralens form
DNA adducts after intercalating within DNA. Exposure to UVA radiation results in
the formation of crosslinks within DNA. Various drugs which produce phototoxic
response fall under the category of antibiotics, antifungal, nonsteroidal anti-­
inflammatory drugs, diuretics, retinoids, anthelmintics, antimalarial, neuroleptic
drugs, etc.

8.2 Antibiotics

Antibiotics such as tetracyclines, fluoroquinolones, sulfonamides, etc. are capable


of producing phototoxic response. Tetracycline phototoxicity is oxygen dependent
and possibly singlet oxygen is involved. Fluoroquinolones are considered relatively
tolerant when compared to other antibiotics that are routinely used [1]. Adverse
effects associated with fluoroquinolones include phototoxicity and photogenotoxic-
ity [2]. Excess exposure of levofloxacin under sunrays may induce levofloxacin-­
mediated phototoxicity/photomutagenesis in vivo. Levofloxacin is unstable under
sunlight exposure and forms photoproducts, which may impose greater health
8 Drug-Induced Phototoxic Response 79

Fig. 8.1 Showed absorption spectra of drug (a). Ketoprofen (NSAIDS) and (b) ofloxacin (antibi-
otic). (c). Mefloquine (antimalarial drug)
80 S. F. Mujtaba et al.

Fig. 8.2 Mechanism of type I and type II reactions

Table 8.1 shows the adverse effect related to phototoxicity of different drugs.
S.
no Photosensitizing drugs Skin reactions
1 Photofrin, amiodarone, chlorpromazine Burning sensation or prickling during
exposure, erythema, edema, urticaria, or
hyperpigmentation
2 Fluoroquinolone antibiotics, Exaggerated sunburn
chlorpromazine, thiazide, diuretics,
quinine, tetracyclines
3 Psoralens Late erythema, blisters, hyperpigmentation
4 Nalidixic acid, tetracycline, naproxen, Increased skin fragility with blisters and
fluoroquinolone antibiotics trauma

hazards to the drug users [3]. Phototoxicity induced by drugs is associated with
toxic photoproduct formation or the generation of short-lived intermediates and
increased level of ROS within the skin and may induce skin diseases [4]. In vivo
phototoxicity study showed the absorption of UVA radiation by tetracyclines which
leads to at least two main processes: (i) photosensitization of biological molecules
by various drugs to induce phototoxicity and (ii) formation of one or more photo-
products that photosensitize by absorption of UV/visible radiation [5].
8 Drug-Induced Phototoxic Response 81

Fig. 8.3 Electromagnetic spectrum and different class of light based at their wavelength

Fig. 8.4 Energy diagram

8.3 Nonsteroidal Anti-inflammatory Drugs (NSAIDs)

NSAIDs are drug class that groups together under analgesic and antipyretic effects
and in higher-dose anti-inflammatory response. The most prominent members of
this group of drugs are aspirin, ibuprofen, and naproxen. Photosensitivity reactions
due to nonsteroidal anti-inflammatory drugs (NSAIDs) are well-known side effects
to these agents usually attributed to ultraviolet radiation. Ketoprofen is one of the
82 S. F. Mujtaba et al.

Fig. 8.5 Diagram of membrane and DNA damage

propionic acid classes of nonsteroidal anti-inflammatory drug with analgesic and


antipyretic effects. Recent studies have shown that the interaction between ketopro-
fen and UV light causes contact dermatitis, lipid peroxidation, and DNA and mem-
brane damage [6]. Ketoprofen-induced phototoxicity may be associated with the
generation of free radicals and formation of toxic photoproducts that results in
increase of ROS within the skin and induces dermatological problems [6]. The tox-
icity of NSAIDs has been associated with metabolism by cytochrome P450, mito-
chondrial dysfunction, and efflux of transporters [7]. Naproxen (NSAIDs) have
been associated with high incidence of both phototoxic and photoallergic skin
responses. UV-exposed cells in presence of naproxen lead to drastic reduction in
cell viability [8]. It is of interest to evaluate the phototoxic response of nonsteroidal
anti-inflammatory drugs.

8.4 Antimalarial

Antimalarial drugs are designed for prevention or cure of malaria. These drugs are
used for treatment of patients with confirmed infection and also for people visiting
a malaria-prone region as a preventive measure. In malaria treatment, most drugs
8 Drug-Induced Phototoxic Response 83

used produce phototoxic side effects in both eyes and skin. Ocular and cutaneous
effects that may be caused by light include corneal opacity, change in skin pigmen-
tation, cataract formation, and other visual disturbances including retinal damage
that may lead to blindness. Mefloquine phototoxicity may be associated with induc-
tion of skin diseases and cancer by altering various biological processes due to
increased level of ROS or by the formation of photoproducts [9, 10]. Adverse pho-
totoxic effect of photosensitive antimalarial drugs has been recognized as undesir-
able side effect, and further testing of photochemical and photobiological properties
can be effective screening method for the prediction of phototoxic potential of anti-
malarials [9, 10]. Drugs are either absorbed locally in the skin or are distributed to
skin via systemic circulation leading to phototoxic reactions [11]. Treatment with
quinine may lead to sub-optimal treatment outcomes [12]. Its accumulation in skin
leads to photosensitization [13].

8.5 Antifungal

Antifungal medication may be fungicidal or fungistatic which is used for treatment


and prevention of mycoses such as dermatophytosis, candidiasis, etc. Adverse
effects of these drugs include liver damage or effecting estrogen levels, but these
drugs can also cause allergic reactions. Voriconazole (antifungal) is known to be
associated with liver toxicity, phototoxicity, and squamous cell carcinoma of skin.
The most common adverse effect of voriconazole is reversible disturbance of vision
(photopsia) that occurs in about 30% of patients [14]. Adverse effects reported from
voriconazole therapy include skin rash, hepatic transaminase level, visual distur-
bances, and photosensitivity. Moreover phototoxicity in children after voriconazole
therapy is also a cause of immense concern [15]. Itraconazole, an antifungal drug
with very few side effects, has also been reported to cause photosensitivity with
symptoms of edema, vesicles, and erythema on sun-exposed areas [16]. Phototoxicity
from the drug results in prominent sunburn-like erythema that is limited to the
exposed surfaces of the skin particularly the head and neck, dorsal hands, and fore-
arms [17].

8.6 Conclusion

Drug-induced photosensitivity refers to the development of dermatological com-


plexity as a result of combinatorial application of light and drug. After photoactiva-
tion of a chemical, one or more clinical manifestations arise. These include
phototoxic and photoallergic reactions, etc. Thus, prevention of photosensitivity
reactions is based on patient’s education. It should be educated to minimize sunlight
exposure during peak hour of exposure. Use of UVA-protected sunscreens and
physical barriers such as clothing provides additional light protection.
84 S. F. Mujtaba et al.

References
1. Lietman PS (1995) Fluoroquinolones toxicities. Updat Drugs 49:794–850
2. Marrot L, Agapakis-Causse C (2000) Differences in the photogenotoxic potential of two flu-
oroquinolones as shown in diploid yeast strain (Saccharomyces cerevisae) and supercoiled
DNA. Muta Res 468:1–9
3. Dwivedi A, Mujtaba SF, Kushwaha HN, Ali D, Yadav N, Singh SK, Ray RS (2012)
Photosensitizing mechanism and identification of levofloxacin photoproducts at ambient UV
radiation. Photochem Photobiol 88:344–355
4. Bagheri H, Lhiaubet V, Montastruc JL, Chouini Lalanne N (2000) Photosensitivity to ketopro-
fen: mechanism and pharmacoepidemiological data. Drug Saf 22:339–349
5. Hasan T, Kochevar IE, McAuliffe DJ, Cooperman BS, Ahdulah D (1984) Mechanism of tetra-
cycline phototoxicity. J Invest Dermatol 84:286
6. Liu S, Mizu H, Yamauchi H (2007) Molecular response to phototoxic stress of UVB irradia-
tion ketoprofen through arresting cell cycle in G2/M phase and inducing apoptosis. Biochem
Biophys Res Commun 364:650–655
7. Jolanda S, Leeuwen V, Unlu B, Nico PE, Vermeulen J, Vos C (2012) Differential involvement
of mitochondrial dysfunction, cytochrome P450 activity and active transport in the toxicity of
structurally related NSAIDs. Toxicol in Vitro 26:197–205
8. Bracchitta G, Catalfo A, Martineau S, Sage E, De Guidi G, Girard PM (2013) Investigation
of the phototoxicity and cytotoxicity of naproxen, a non-steroidal anti-inflammatory drug, in
human fibroblasts. Photochem Photobiol Sci 12(5):911–922
9. Yadav N, Dwivedi A, Mujtaba SF, Verma A, Chaturvedi R, Ray RS, Singh G (2014)
Photosensitized mefloquine induces ROS mediated DNA damage and apoptosis in keratino-
cytes under ambient UVB and sunlight exposure. Cell Biol Toxicol 30:253–268
10. Yadav N, Dwivedi A, Mujtaba SF, Pal MK, Singh G, Ray RS (2014) Photo-damage to kerati-
nocytes by quinine photosensitization under mild ultraviolet B exposure (2014). G-J Environ
Sci Technol 1(6):123–128
11. Lhiaubet V, Paillous N, Chouini LN (2001) Comparison of DNA damage photoinduced by
ketoprofen, fenofibric acid and benzophenone via electron and energy transfer. Photochem
Photobiol 74:670–678
12. Achan J, Talisuna AO, Erhart A, Yeka A, Tibenderana JK, Baliraine FN, Rosenthal PJ,
Alessendro UD (2011) Quinine an old antimalarial drug in a modern world: role in the treat-
ment of malaria. Malar J 10(144):1–12
13. Kristensen S, Orsteen AL, Sande SA, Toennesen HH (1994) Photoreactivity of biologically
active compounds VII. Interactions of antimalarial drugs with melanin in vitro as part of pho-
totoxicity screening. J Photochem Photobiol B Biol 26:87–95
14. Johnson LB, Kauffman CA (2003) Voriconazole a new triazole antifungal agent. Clin Infect
Dis 36:630–637
15. Sheu J, Haryluk EB (2014) Voriconazole phototoxicity in children. Amer Acad Dermatol
10.023
16. Alvarez-Fernandez (2000) Photosensitivity induced by oral itraconazole. Eur Acad Dermatol
Veneriology 14:501–503
17. Malani AN, Aronoff DM (2008) Voriconazole induced photosensitivity. Clin Med Res
6(2):83–85
PAHs and Phototoxicity
9
Ajeet K. Srivastav, Shikha Agnihotry, Syed Faiz Mujtaba,
Sandeep Negi, Ankit Verma, and Ratan Singh Ray

Abstract
PAHs or polycyclic aromatic hydrocarbons belong to group of environmental
pollutants which come under human carcinogens. PAHs is a class which refers to
an immensely colossal number carbon and hydrogen along with two or more
fused aromatic rings. These PAHs produced from chemicals occur naturally in
coal, crude oil, and gasoline. The physical properties of PAHs include flamma-
ble, solid crystals at room temperature, and tobacco leads to the engenderment of
these deleterious chemicals. Their presence is also detected in cigarette smoke
and motor vehicle emissions up to a certain levels. The exposure of PAHs to
general population is in the form of breathing ambient and indoor air, by the
consumption of contaminated aliment containing cigarette smoke, tobacco, and
polluted air. Occupational exposure of PAHs may be reason for breathing quan-
daries, chest pain, and vexation coughing and also cause cancer.

Keywords
PAHs · Photoproducts · Phototoxicity · Skin and cancer

Authors Ajeet K. Srivastav and Shikha Agnihotry have been equally contributed to this chapters.
A. K. Srivastav · S. Negi · A. Verma · R. S. Ray (*)
Photobiology Laboratory, System Toxicology and Health Risk Assessment Group, CSIR-
Indian Institute of Toxicology Research (IITR), Lucknow, Uttar Pradesh, India
S. Agnihotry
Department of Biomedical-informatics, Sanjay Gandhi Post Graduate Institute,
Lucknow, India
S. F. Mujtaba
Department of Zoology, Faculty of Science, Shia P.G. College, University of Lucknow,
Lucknow, Uttar Pradesh, India

© Springer Nature Singapore Pte Ltd. 2018 85


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_9
86 A. K. Srivastav et al.

9.1 Introduction

Polycyclic aromatic hydrocarbons (PAHs) are composed of carbon and hydrogen


and have two or more fused benzene rings in linear, angular, or cluster arrangements
[1], but this is not necessary to fuse with each other. Their presence in the atmo-
sphere enters rain as a result of in-cloud and below-cloud scavenging [2]. Total
amount of PAHs collected on land and water are likely equipollent to PAHs amount
in rainfall [3]. Among the PAHs are some of the most toxic compounds kenned.
Some of the PAHs are kenned to be carcinogenic, mutagenic, and toxic for repro-
duction (linked to birth defects). UV radiation, mainly UV-B (290–320 nm) radia-
tion, induces types of biological changes like cyclobutane pyrimidine dimmers
(CPDs) formation, inflammation, cellular injury, and skin cancer [4]. UV radiation
can cause oxidative damage to DNA, cellular proteins, and lipids [5]. Due to their
wide distribution, it is paramount to monitor these compounds. Mostly PAHs are
considered as PBT substances [6, 7] experts verbalize these substances in this con-
text (assiduous, bioaccumulative, and toxic substances). If such chemicals are relin-
quished, they can no longer be abstracted from the environment due to their
characteristics. They accumulate and can harm plants and animals including entire
flora and fauna [8]. People who are involve in smoking tobacco products, or who are
exposed to smoke, are among the most highly exposed groups; tobacco smoke con-
tributes to 90% of indoor PAH levels in the homes of smokers [9]. Consequently,
PAHs play a paramount role in the etiology of human cancers. Incomplete combus-
tion which results in the formation of PAHs might withal transpire in industrial
processes, cooking, in fires, and in cigarette smoke. A concave exterior region on
some polycyclic aromatic hydrocarbons that have three phenyl rings in a nonlinear
arrangement is defined as bay region. Mutagenic PAHs, like benzo[a]pyrene, have
four or more aromatic rings as well as a “bay region” [10]. PAHs have mutagenic
metabolites like diol epoxides, quinones, and radical PAH cations. The presence of
benzene ring in DBA facilely absorbs the ultraviolet light and modified in photo-
product, and the presence of bay region in DBA makes difference to many of the
PAHs. DBA in reposing phase in ground state when it’s absorbed the light turns in
its exhilarated singlet state; it can withal undergo a process called intersystem cross-
ing, during which there is a transition from ESS to a spin state called the exhilarated
triplet state. DBA in a triplet state can react with molecular oxygen or other biologi-
cal molecules kenned as type-1 photosensitization. All biological molecules subsist
in singlet ground state [11]. Then its triplet state of DBA reacts with these molecules
with high reactivity, and result is apoptosis. The highest intake is shown to emanate
from cereals, oils, and fats. More minuscule amounts emanate from vegetables and
cooked meat. Phototoxic efficiency of DBA showed photochemical generation of
ROS by type-1 reaction; type-1 reaction ROS include superoxide and hydroxyl radi-
cal. Type-1 reaction is predicated at electron transfer mechanism and orbital arrange-
ment in case of superoxide generation is 2px, 2py orbital showed in one orbital is
filled by parallel and antiparallel electron, and the other one remains single antipar-
allel electron due to orbitalic arrangement of superoxide additionally kenned as
superoxide anion radical [1, 12]. It is product of single electron reduction of
9 PAHs and Phototoxicity 87

molecular oxygen to fill one of the π*2p orbital with an additional electron having
an inverse spin quantum number. And this superoxide can be protonated to give the
hydroxyl radical and is an oxidant and reductant. Hydroxyl radical is one of the
most reactive chemical species kenned to be engendered in living organism. It reacts
with astronomically high rate with proteins, carbohydrate, lipids, nucleic acid, and
organic acids and with aromatic as well as aliphatic structure. Among the major
target of hydroxyl in biological system are nucleic acids. The oxidation’s or addita-
ment which takes place on pyrimidine or purine bases as result of hydroxyl radical
attack. One such product is 8-­hydroxyguanosine, which may result from the addita-
ment of hydroxyl on C8 of the heterocycle [13]. Such product has been identified
not only in cellular DNA but additionally in the urine of animals or even human
being exposed to ionizing radiation or oxidative stress. Human keratinocytes are
major target of UV radiation; these play a paramount role in sundry replications to
photodamage caused by UV irradiation by relinquishing and upregulating sundry
activator and effector caspases such as caspase 3, 9, and 12 and antioxidant repres-
sor like Keap-1, Nrf-2, and heme oxygenase-1. Apoptotic cell death is characterized
by nuclear condensation and fragmentation with intact cytoplasmic organelles
induced by photosensitized PAHs. Major source of exposure of PAHs to human
being is victuals [14]. Contact of human skin with PAHs contaminated soil and the
frequent utilization of dermally applied pharmaceutical products predicated on coal
tar have withal been identified as sources of exposure to the human beings [15].
PAHs are commercially not engendered in the United States [16]. The effect of
PAHs on human population depends on its exposure time (length of time, etc.) and
type of exposure too.

9.2 Sources of Exposure and Evaluation of Human Health

The formation of PAHs is mainly due to pyrolytic processes, especially the incom-
plete combustion of organic materials during industrial and other human activities,
such as processing of coal and crude oil, combustion of natural gas, including for
heating, combustion of reluct, conveyance traffic, cooking, and tobacco smoking, as
well as in natural processes like carbonization. It is prominent that human exposure
to intricate coalescences of PAH occurs primarily by three routes: (i) respiratory
tract through the smoking of tobacco products and inhalation of polluted air, (ii)
gastrointestinal tract through the ingestion of contaminated imbibing dihydrogen
monoxide and aliment, and (iii) skin contact, which customarily occurs from occu-
pational exposure (Table 9.1).
Background levels of some representative PAHs in the air are reported to be 0.02–
1.2 nanograms per cubic meter (ng/m3); a nanogram is one-millionth of a milligram)
in rural areas and 0.15–19.3 ng/m3 in urban areas. Person exposed to PAHs in soil near
areas where coal, wood, gasoline, or other products have been burned. The exposure to
PAHs is additionally in the soil at or near hazardous waste sites, such as former manu-
factured-gas factory sites and wood-preserving facilities. PAHs have been found in
some imbibing dihydrogen monoxide supplies in the cumulated states. Background
Table 9.1 List of PAHs
88

List Common name Structure List Common name Structure


EPA,SCF, EU Benzo[a]pyrene EPA,SCF, EU Dibenz[a,h]anthracene

EPA Acenaphthene SCF+EU Dibenzo[a,e]pyrene

EPA Acenaphthylene SCF+EU Dibenzo[a,h]pyrene

EPA Anthracene SCF+EU Dibenzo[a,i]pyrene

EPA, SCF, EU Benz[a]anthracene SCF+EU Dibenzo[a,I]pyrene

EPA, SCF, EU Benzo[b]fluoranthene EPA Fluoranthene

SCF, EU Benzo[J]fluoranthene EPA Fluorene


A. K. Srivastav et al.
9

EPA, SCF, EU Benzo[k]fluoranthene EPA,SCF, EU Indeno[1,2,3-cd]pyrene

EU Benzo[c]fluorene SCF+EU 5-methylchrysene

EPA,SCF, EU Benzo[ghi]perylene EPA Naphthalene


PAHs and Phototoxicity

EPA,SCF, EU Chrysene EPA Phenanthrene

SCF, EU Cyclopenta[cd]pyrene EPA Pyrene


89
90 A. K. Srivastav et al.

levels of PAHs in imbibing dihydrogen monoxide range from 4 to 24 ng/l (ng/L; a liter
is marginally more than a quart). PAHs can enter your body through your lungs when
you breathe air that contains them (customarily stuck to particles or dust). Cigarette
smoke, wood smoke, coal smoke, and smoke from many industrial sites may contain
PAHs. Population living near hazardous waste sites can additionally be exposed by
breathing air containing PAHs. However, it is not kenned how rapidly or thoroughly
your lungs absorb PAHs. Imbibing dihydrogen monoxide and swallowing aliment,
soil, or dust particles that contain PAHs are other routes for these chemicals to enter
your body, but absorption is generally slow when PAHs are swallowed. In mundane
conditions of environmental exposure, PAHs enter the body when skin comes into
contact with soil that contains high calibers of PAHs (this could occur near a hazardous
waste site) or with used crankcase oil or other products (such as creosote) that contain
PAHs. The rate at which PAHs enter your body by victualing, imbibing, or through the
skin can be influenced by the presence of other compounds that you may be exposed
to at the same time with PAHs. PAHs can enter all the tissues of your body that contain
fat. They incline to be stored mostly in your kidneys, liver, and fat. More minute
amounts are stored in your spleen, adrenal glands, and ovaries. PAHs are transmuted
by all tissues in the body into many different substances. Some of these substances are
more inimical, and some are less deleterious than the pristine PAHs. Results of animal
studies show that PAHs do not incline to be stored in your body for a long time. Most
PAHs enter the body leave within a few days, primarily in the feces and urine.

9.3 Photosensitization Mechanism of PAHs

Most of the PAHs and their photoproducts have absorption maxima (λmax) under
UV-R and visible spectrum. Due to the presence of their π orbital system and aromatic
ring structure also, PAHs and their photoproducts can absorb sunlight in the visible
and UV regions of solar spectrum. Photosensitization reactions are two types, i.e., type
1 and type 2. Type 1 is electron transfer mechanism and type 2 is energy transfer pro-
cess. If virtually every environmental pertinent scenario is ineluctable, PAHs and their
photoproducts are exposed to sunlight. When exposed to radiation, molecules capable
of absorbing UV-R and visible light become exhilarated, resulting in different biologi-
cal effects. Photosensitized PAHs and their photoproducts can emit a photon via fluo-
rescence, which in turn returns the molecule to its singlet ground state. PAHs and their
photoproducts can additionally give off their energy through vibrational state as heat
and return to the singlet ground state. However, while the molecule is in its exhilarated
singlet state (ESS), it can withal undergo a process called intersystem crossing, during
which there is a transition from the ESS to a spin state called the exhilarated triplet
state (ETS) designated 3CPs*. Emitted radiation through intersystem crossing by
exhilarated triplet state of molecular oxygen kenned as phosphorescence. Molecular
oxygen in nature is primarily found in a ground triplet state (3O2) (Fig. 9.1).
All biological molecules exist in a singlet ground state. Photoexcited PAHs and
their photoproducts lead to generation of 1O2, O2−, and •OH radical through type-1
and type-2 photosensitized reactions.
9 PAHs and Phototoxicity 91

1PAHs*
Singlet oxygen state

3PAHs*

Intersystem crossing

Fluorescence
Triplet state

Phosphoresence

Absorption

ROS

1PAHs

Ground state

Fig. 9.1 Electronic energy diagram of the physical events accompanying the absorption of photon
by polycyclic aromatic hydrocarbons (PAHs). 1PAHs, ground state; 1PAHs*, excited singlet state;
3
PAHs*, excited triplet state; 3O2, triplet (ground) state oxygen; 1O2, singlet (excited) state
oxygen

9.4 Story Behind PAHs Photoproduct Formation

PAHs are benzene ring containing aromatics compound. Due to the presence of
these benzene rings, they have potential to absorb light. According to the first law of
photochemistry, when these PAHs are absorbed opportune wavelength of light, then
it is degraded and forms their photoproducts. And these photoproducts are present
in environment; anterior study has reported these photoproducts are highly toxic
compared to their parent compound. It is a challenge for researcher on how to
abstract PAHs from environment (Figs. 9.2 and 9.3).

9.5 How to Remove PAHs from Environment

PAHs are diverse group of compound listed by Environmental Protection Agency


reported as hazard for mankind including bird and aquatic organism followed by
mostly living eukaryotes. Study has been reported that like benzanthracene (BNZ),
if BNZ exposed 4–6 h in natural sunlight or in UV, then losing their stability is
completely degraded. It means that when industries are prone for production of such
kind of PAHs, they can remove the BNZ from environment, but the problem is when
Fig. 9.2 A schematic representation showed series of events perform during absorption to photo-
product identification of benzanthracene: (a) absorption spectra, (b) photodegradation spec-
tra(0–7 h), (c) total GC chromatogram, (d) mass spectra compared through NIST library, (e)
schematic representation of photodegradation and photoproduct identification
9 PAHs and Phototoxicity 93

Fig. 9.2 (continued)


94 A. K. Srivastav et al.

Fig. 9.3 (a–d) Schematic representation, (a–b) absorption to photodegradation, (b, d) photodeg-
radation to photoproduct formation of photosensitized anthraquinone in natural sunlight exposure.
(a) Photoabsorption spectra, (b, c) total ion chromatogram of GC at different exposure time period
of ANT, (d) photoproduct identification and NISt comparison

its exposed with lights, it forms photoproducts, and study reported it’s highly toxic
than their parents compound. But if there is continuous exposure of parent PAHs
and their photoproduct, then their parent followed by photoproducts may be broken
in their simplest form. And lower the molecular weight 128 in case of PAHs its safe.
And another way to remove PAHs from environment then is to use bacteria for
degrading PAHs by mixing in industrial waste which have high density of PAHs
contamination without introducing any carbon source. Bacteria utilized PAHs as
carbon source and remove PAHs contamination by degrading in their simplest
metabolite.

9.6 Model for Testing PAHs Phototoxicity

The HaCaT cell line is transformed aneuploid, immortal, keratinocytes cell line
from adult human skin is widely used in scientific research [17]. The skin is the
uppermost part of human body which is directly exposed to sunlight. India is a
9 PAHs and Phototoxicity 95

tropical country that’s why day activity is very mundane and people face a lot of
environmental exposure in their diligent day life from PAHs. Under standard culture
conditions HaCaT cells have a partially to plenarily differentiated phenotype due to
the high calcium content of both standard media and fetal bovine serum. HaCaT
cells are utilized due to their high capacity to differentiate and proliferate in vitro
[18]. This cell line is considered as best model for phototoxicity study in human
skin by utilizing photosensitizers like PAHs. This cell line sanctioned the character-
ization of several processes, such as their utilization as a model system for vitamin
D3 metabolism in the skin [19]. Guinea pigs have been used ecumenical in scientific
research in the 1980s, concretely for dermatological studies. This is considered as
in vivo model for phototoxic study; guinea pigs are one of the few animals which,
like humans and other primates, cannot synthesize vitamin C but must be obtained
from their diet, and they are ideal for researching scurvy due to these characteristics
having no antioxidant property against ROS species engendered by photosensitized
PAHs [20]. Nowadays hairless mice (SKH-1) are adscititiously utilized as an in vivo
model for dermatological studies and phototoxicity studies.

9.7 Receptor for PAHs

Aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that in


humans is encoded by AHR gene which plays very important role in regulation of
biological replications to planar aromatic (aryl) hydrocarbons. This receptor regu-
lates xenobiotic-metabolizing enzymes such as cytochrome P450. After ligand
binding to chemicals such as PAHs, the chaperones dissociate resulting in AhR
translocating into the nucleus and dimerizing with ARNT (AhR nuclear transloca-
tor), leading to transmutations in gene transcription followed by cell death (Fig. 9.4).

9.8  olecular Mechanism Involved in PAHs


M
Photocytotoxicity

PAHs are very stable molecules, which generally nontoxic in dark, whereas they
show phototoxicity under ultraviolet radiation of sunlight [21].Various research
have proved that exposure to sunlight transmutes the chemical structure of PAHs
and enhance its toxicity. Photomodification occurs via an oxidation reaction that
results in the formation of photoproduct which is in many cases more toxic than
their parent compounds. UVA irradiation decremented the mitochondrial activity of
cells when the extracts contained PAHs [22]. Study has been reported PAHs under
sunlight engenders ROS like 1O2 and O2−. which resulted in DNA damage, cell cycle
apprehends, and conclusively cell death [23]. Main sources of intracellular ROS
include both cytoplasmic and mitochondrial. In case engenderment either by mito-
chondria or cytoplasmic is higher, the natural cellular antioxidant defense system is
overwhelmed leading to oxidative stress. Mechanism of cell death by apoptosis is
adscititiously called programmed cell death. Apoptosis is a tightly regulated form of
96 A. K. Srivastav et al.

Fig. 9.4 Diagram showing PAhs and AhR interaction in cellular system (Annu. Rev. Pharmacol.
Toxicol. 43: 309–34. *Reprinted, with permission, from the Annual Review of Pharmacology and
Toxicology, Volume 43 (c)2003 by Annual Reviews)

cell death, which can be initiated by two variants of signals: (1) intracellular stress
signals (intrinsic pathway) and (2) extracellular ligands (extrinsic pathway) [9].
Photosensitized PAHs lead to apoptotic cell death in all membrane-bound organ-
elles by the engendering ROS (Fig. 9.5).

9.9 Genotoxic Potential of PAHs

Earlier studies reported the exposure of UV-B in presence of PAHs (benzanthra-


cene) results in formation of CPDs in HaCaT cell line, which was significantly more
preponderant than UV-B exposure alone [1]. In additament assessment of genotoxic
potential of PAHs in human skin cell by UV exposure, we observed paramount
formation of cyclobutane pyrimidine dimers and 6–4 photoproducts followed by
Dewars isomer. Photosensitized PAHs showed the consequential single-strand DNA
damage [24]. PAHs phototoxicity reported with appearance of micronuclei, nuclear
buds, and nucleoplasmic bridges. These cells were treated with UV light for 30 min
followed by photosensitized PAHs which have caused some cells to initiate apopto-
sis. The nucleus of cells in sundry states stands out in blue, while the rest of the cell
is marginally visible. Cells that designate densely packed and fragmented chromatin
have undergone apoptosis [5] (Figs. 9.6 and 9.7).
9 PAHs and Phototoxicity 97

Fig. 9.5 Showed metabolism of PAHs by cytochromes 1A1 and 1B1 in the liver and after metabo-
lism their toxic effect to cause carcinogenicity

OH
OH

enzymes
O

DNA

OH
OH

DNA
OH

Fig. 9.6 Oxidation of PAHs by enzymes in the liver


98 A. K. Srivastav et al.

Fig. 9.7 DNA’s structure basically has a bunch of flat molecules stacked on top of each other, and
PAH is also flat

References
1. Sims RC, Overcash R (1983) Fate of polynuclear aromatic compounds (PNAs) in soil-plant
systems. Residue Rev 88:1–68
2. Van Noort PCM, Wondergem E (1985) Scavenging of airborne polycyclic aromatic hydrocar-
bons by rain. Environ Sci Technol 19:1044–1048
3. Polynuclear aromatic hydrocarbons (PAH). In: Air quality guidelines for Europe. Copenhagen,
World Health Organization Regional Office for Europe, 1987, pp. 105–117
4. Tatsushi T, Ohnuki G, Ibuki Y (2008) Solar-simulated light-exposed benzo(a)pyrene induces
phosphorylation of histone H2AX. Mutat Res 650:132–139
5. Gao Y, Gao Y, Guan W, Huang L, Xu X, Zhang C, Chen X, Wu Y, Zeng G, Zhong N (2013)
Antitumor effect of para-toluenesulfonamide against lung cancer xenograft in a mouse model.
J Thorac Dis 5(4):472–483. https://doi.org/10.3978/j.issn.2072-1439.2013.08.28
6. Liu S, Mizu H, Yamauchi H (2007) Molecular response to phototoxic stress of UVB-irradiated
ketoprofen through arresting cell cycle in G2/M phase and inducing apoptosis. Biochem
Biophys Res Commun 364:650–655
7. Liu S, Mizu H, Yamauchi H (2010) Photoinflammatory responses to UV-irradiated ketoprofen
mediated by the induction of ROS generation, enhancement of cyclooxygenase-2 expression,
and regulation of multiple signalling pathways. Free Radic Biol Med 48:772–780
8. Harrison RM, Perry R, Wellings RA (1975) Polynuclear aromatic hydrocarbons in raw, pota-
ble and waste waters. Water Res 9:331–346
9. Choi H, Harrison R, Komulainen H, Delgado Saborit J (2010) “Polycyclic aromatic hydrocar-
bons”. WHO guidelines for indoor air quality: selected pollutants. World Health Organization,
Geneva Retrieved 2014-12-12
10. Xue W, Warshawsky D (2005) Metabolic activation of polycyclic and heterocyclic aromatic
hydrocarbons and DNA damage: a review. Toxicol Appl Pharmacol 206(1):73–93. https://doi.
org/10.1016/j.taap.2004 Retrieved 2014-08-20
11. Foote CS (1968) Mechanisms of photosensitized oxidation. There are several different
types of photosensitized oxidation which may be important in biological systems. Science
162(3857):963–970 No abstract available
12. Jongeneelen FJ, Leijdekkers CM, Bos RP, Theuws JLG, Henderson PT (1985) Excretion of
3hydroxy-benzo[a] pyrene and mutagenicity in rat urine after exposure to benzo[a]pyrene.
J Appl Toxicol 5(5):277–282
13. Cosman M, de los Santos C, Fiala R, Hingerty BE, Singh SB, Ibanez V, Margulis LA, Live D,
Geacintov NE, Broyde S, Patel DJ (1992) Proc Natl Acad Sci U S A 89:1914–1918
14. IPCS (1998) Environmental health criteria No. 202. Selected non-heterocyclic polycyclic
aromatic hydrocarbons. International programme on chemical safety. http://www.inchem.org/
documents/ehc/ehc/ehc202.htm
9 PAHs and Phototoxicity 99

15. Jongeneelen FJ (2001) Benchmark guideline for urinary 1hydroxypyrene as biomarker of


occupational exposure to polycyclic aromatic hydrocarbons. Ann Occup Hyg 45(1):3–13
16. IARC (1983) Polynuclear aromatic compounds, part 1. Chemical, environmental and experi-
mental data. IARC monographs on the evaluation of carcinogenic risk of chemicals to humans,
vol 32. International Agency for Research on Cancer, Lyon, p 477
17. Boukamp P, Petrussevska RT, Breitkreutz D, Hornung J, Markham A, Fusenig NE (1988)
Normal keratinization in a spontaneously immortalized aneuploid human keratinocyte cell
line. J Cell Biol 106(3):761–771. https://doi.org/10.1083/jcb.106.3.761. PMID 2450098
18. Schoop VM, Mirancea N, Fusenig NE (1999) Epidermal organization and differentiation
of HaCaT keratinocytes in organotypic coculture with human dermal fibroblasts. J Investig
Dermatol 112(3):343–353. https://doi.org/10.1046/j.1523-1747.1999.00524.x
19. Lehmann B (1997) HaCaT cell line as a model system for vitamin D3 metabolism in human
skin. J Investig Dermatol 108(1):78–82. https://doi.org/10.1111/1523-1747.ep12285640
20. Gad SC (2007) Animal models in toxicology, 2nd edn. Taylor & Francis, Philadelphia,
pp 334–402 ISBN 0-8247-5407-7
21. Daoud A, Verma A, Mujtaba F, Dwivedi A, Hans RK, Ray RS (2011) UVB- induced apoptosis
and DNA damaging potential of chrysene via reactive oxygen species in human keratinocytes.
Toxicol Lett 204:199–207
22. Mujtaba SF, Dwivedi A, Mudiam MKR, Ali D, Yadav N, Ray RS (2011) Production of ROS
by photosensitized anthracene under sunlight and UVR at ambient environmental intensities.
Photochem Photobiol 87:1067–1076
23. Ryter SW, Kim HP, Hoetzel A et al (2007) Mechanisms of cell death in oxidative stress.
Antioxid Redox Signal 9:49–89
24. Şardaş S, Aygün N, Karakaya A (1997) Genotoxicity studies on professional hair colorists
exposed to oxidation hair dyes. Mutat Res Genet Toxicol Environ Mutagen 394:153–161
Phototoxicity of Hair Dyes: Challenge
for Tropical Countries 10
Shruti Goyal, Ajeet Kumar Srivastav, Saroj K. Amar,
Shikha Agnihotry, and Ratan Singh Ray

Abstract
In current scenario, utilization of hair dyes and personal care products (PCPs)
has been increasing globally day to day. The US Aliment, Dye and Cosmetic Act
require prior approbation of color additives, dyes, and cosmetics afore relin-
quishing product in market, but in India there was no guideline yet. According to
survey more than one-third of women over age 18 and about 10% of men over
age 40 utilize some type of hair dye coloring products for better physical appear-
ance. Mostly, hair dye ingredient forms photoproduct after exposure of UV irra-
diation due to the presence of benzene nucleus or bulky structure with strong
functional group. Photocytotoxicity results of hair dye ingredients in human
keratinocyte cells illustrated the paramount reduction in cell survival.
Photogenotoxic potential of hair dye was also reported in various past studies.
Thus, study focused on hair dye which induced photogenotoxicity, photocyto-
toxicity, and apoptotic cell death through disturbing normal cell physiology.

Authors Shruti Goyal and Ajeet Kumar Srivastav have been equally contributed to this
chapter.
S. Goyal · R. S. Ray (*)
Photobiology Laboratory, System Toxicology and Health Risk Assessment Group,
CSIR-­Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
A. K. Srivastav
Photobiology Laboratory, System Toxicology and Health Risk Assessment Group,
CSIR-­Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, India
S. K. Amar
Department of Forensic Science, School of Bioengineering and Biosciences,
Lovely Professional University, Phagwara, Punjab, India
S. Agnihotry
Department of Biomedical-informatics, Sanjay Gandhi Post Graduate Institute,
Lucknow, India

© Springer Nature Singapore Pte Ltd. 2018 101


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_10
102 S. Goyal et al.

Therefore, joint exposure of sunlight and long-term use of hair dyes enhance
oxidative stress in human skin which may lead to various skin diseases in human
beings including mutation and various types of skin cancer.

Keywords
Phototoxicity · Hair dyes · PCPs · UV-R · Sunlight

10.1 Introduction

Sunlight is a component of electromagnetic spectrum of different wavelengths con-


taining ultraviolet radiation (UV-R) and many other radiations [1]. Hair dyes are the
most reactive chemicals utilized in the cosmetic industry [2]. At Roman imperium
time, leaden combs dipped in vinegar were routinely used to darken graying hair.
Earlier, many different plant extracts (husks, flowers, barks, leaves, etc.) were uti-
lized for hair dye preparations afore the advent of modern dyes [3]. Some of these
plant-derived dyes were commixed with metals such as copper and iron, to engen-
der more lasting or richer shades. The first synthetic organic hair dye developed was
pyrogallol often utilized with henna to dye hair brown. The first safe commercial
hair color was engendered in 1909 by French chemist, Eugene Schuller, utilizing
the chemical paraphenylenediamine (PPD) [4]. Some aromatic amines were apper-
ceived to engender an incremented incidence of bladder cancer [5]. European
Scientific Committee on Cosmetics and Non-Food Products has developed guide-
lines for the assessment of the genotoxic potential of hair [6, 7]. During the past
25 years, an independent expert panel, Cosmetics, Toiletry and Fragrance
Association, set up by the Coalesced States has reported 650 ingredients that withal
included hair dyes [8]. US Environmental Protection Agency (EPA) has reported
that 15 million people are potentially exposed with hair dye ingredients [9]. A sta-
tistically consequential dose-dependent sodality between hair dyeing and jeopar-
dize of ovarian cancer was found [10, 11]. Women with perpetuated utilization of
dark, concretely ebony, hair dyes may have incremented risk of fatal non-Hodgkin’s
lymphoma and multiple myeloma [12]. For health risk assessment, phototoxicity of
hair dyes must be taken into knowledge because it is ineluctable for people utilizing
hair dyes to get exposed to both hair dyes and sunlight concurrently. Photo-induced
toxicity by hair dyes should be taken into consideration for the health risk assess-
ment. Phototoxicity is conventionally due to the formation of reactive species (ROS)
and reactive intermediate products [13]. Hair dyes can perforate into the skin. Thus,
there is an exigent need regarding the evaluation of phototoxic potential of the hair
dyes for the safety of people utilizing or working with hair dyes. Since India is a
tropical country (abundance of sunlight is more) and alfresco activities such as agri-
cultural, commercial, sport, etc. are cyclopean, ergo, the phototoxicity study of hair
dyes would be a milestone to obviate people from the hazardous phototoxic effects
of hair dyes.
10 Phototoxicity of Hair Dyes: Challenge for Tropical Countries 103

10.1.1 Hair Dye Types and Its Mechanism of Action

Hair dye is a current fashion trends among the women in global climatic change
scenario. On the basis of hair dye ingredients, they are divided into three different
classes.

10.1.2 Direct Dye

(a) Temporary dyes: Temporary hair color is a more sizably voluminous molecule
that coats the surface of the hair and doesn’t perforate deep into the cuticle.
They generally last for one to two shampoos or washings.
(b) Semipermanent dyes: Semipermanent hair dyes are applied to color the surface
of hair. These dyes perforate into the hair shaft including cuticle and last after
five to ten shampoos or washings.
(c) Permanent (oxidative) hair dyes: Permanent colors are designed to perforate the
hair shaft and deposit their color directly into the cortex of the hair. These dyes
are commonly used and last after new hair growth.
(d) Natural dyes: It consists of the dried leaves of Lawsonia alba plant, growing in
North Africa, in the Midwest, and in India.
(e) Metal salts: Silver salts, lead, and bismuth are major ingredients of metallic
dye.

Types of hair dyes

10.1.3 UV Radiation

Solar ultraviolet radiation (UV-R) is a ubiquitous environmental genotoxic agent to


which everyone is exposed circadianly. Phototoxic effects of solar UV-R have incre-
mented due to the depletion of ozone layer in tropics than other areas. UV-R is more
104 S. Goyal et al.

excruciating in the tropics during noon in the summer. Public health message
regarding the safe exposure of sun is very paramount for hair dye utilizer because
perpetuated exposure of UV-R induced ROS generation and DNA damage [4].
Ultraviolet light is only 5–10% of the total radiant energy received at the earth sur-
face from the sunlight which is divided in UV-R and the rest is divided invisible
(40%) and infrared (50%). The range from 200 to 400 nm is often arbitrarily divided
into UV-A (320–400 nm), UV-B (290–320 nm), and UV-C (200–280 nm) radiation
predicated on skin reaction in human. However, risk of exposure and UV vigor var-
ies greatly depending on several factors like cloud cover, the time of year and time
of day, ozone levels, altitude, and location. The caliber of exposure and UV is rep-
resented by the UV index, and with hourly UV forecast, we can find the UV index.
UV overexposure can be earnest for health and may lead to diseases such as skin
cancer, premature aging of the skin, cataracts, ocular perceiver cancer, immune sys-
tem suppression, and snow visual impairment. While much attention is given to
melanoma, one should be cognizant of the other perils involved. All of us are addi-
tionally exposed to toxic chemicals through our everyday utilization of cosmetics
and personal care products as well as through the air we breathe, victuals we victual,
dihydrogen monoxide we imbibe, and a variety of household products. As a result,
we all carry toxic industrial chemicals inside our bodies.

10.1.4 Physiology of Hair

Mainly hair consists with structural protein keratin. Each fiber of hair consists of
three different layers:

1. An innermost layer or medulla which is only present in immensely colossal thick


hairs.
2. The middle layer kenned as the cortex. The cortex provides vigor and both the
color and the texture of hair.
3. The cuticle is outermost layer has thin, achromic and accommodates as a guard
of the cortex.

10.1.5 Biological Effect of Hair Dyes Under UV Exposure

Cosmetics product is most consequential material for physical appearance of face


by utilizing this in different ways, applying on human skin intended to be rubbed,
sprayed on, introduced into, or any component thereof for cleansing and beautifying
[14]. Research has focused on some aromatic amines utilized in hair dyes which
were identified to engender an incremented incidence of bladder cancer [4, 5, 7].
Hair dyes play an important role in physical appearance for mankind, but people
are unaware from these toxicological and photo-toxicological effects. It is very
harmful and indirectly anthropogenic death warrant for users. Hair dye ingredients
are mostly aromatic hydrocarbons (e.g., PPD, PTD). These aromatic compounds
have an alternative double-bond configuration that has capacity to absorb energy.
10 Phototoxicity of Hair Dyes: Challenge for Tropical Countries 105

After absorbing energy, these molecules become excited and unstable and release
energy or electron in the form of reactive oxygen species (ROS). ROS is a highly
reactive oxygen molecule that can react with any organelles in cell and disturb their
function that finally affects the biological process.
Here we discussed some biological effect of hair dyes with their physical and
molecular level damage including their sophisticated technique.

10.1.6 Apoptosis Detection

Apoptosis is carefully regulated process of program cell death that occurs as a nor-
mal part of development, and it’s one of the major side effects of hair dyes. Certain
detection techniques are available for apoptosis.

10.1.7 Phosphatidylserine Translocation

Phosphatidylserine (PS) is present in inner leaflet of human cell membrane. During


apoptosis, it moves from inner leaflet to outer leaflet of cell membrane. Annexin V/
PI, Ca+2-dependent dye which has high affinity for PS, is used by researchers to
detect apoptosis either via flow cytometer or microscope.

10.1.8 Patch and Photo-Patch Tests

Patch test is very authentic to detect the type of reaction to a particular cosmetic
type either its lipsticks or sunscreens or hair dyes, whether irritant or allergic [14].
Photo-patch test is very supporting in detection of photo contact dermatitis test.

10.1.9 Photosensitivity Test

Two types of photosensitivity test for photosensitize testing: (1) phototoxicity (2)
photoallergy. 3T3 neutral red uptake phototoxicity test (3T3 NRU PT), an alterna-
tive test for replacement of laboratory animal for all photosensitizers, including
personal care products testing for phototoxicity should be done.

10.1.10 Cyclobutane Pyrimidine Dimers Formation

The most customary DNA lesion induced by UV irradiation is the cyclobutane


pyrimidine dimer (CPDs) which forms at two adjacent pyrimidines at single strands.
Solar UV radiation mostly UVB radiation (290–320 nm) is involved in the skin
cancers and bipyrimidine DNA photoproducts formation. CPDs have higher poten-
tial to form when in combined affected with any photosensitizers like hair dye and
cosmetics ingredients.
106 S. Goyal et al.

10.1.11 Micronuclei Formation

Micronuclei (MN) formation occurs in several ways. It might be due to oxidative


DNA damage that shears off a chunk of the DNA from the chromosomes and with-
out centromere, these chunk are left behind and form an extranuclear nuclei, which
we term as MN. In another mechanism, it has been proposed that spindle motor
protein dysfunction might cause a lagging segregation and prior to karyokinesis; the
lagging strand fails to incorporate the nucleic material within the main nucleus, thus
forming an extranuclear inclusion which we find as MN. Displaced chromosome
fragments or chromosomes are further enclosed in a nuclear membrane and appeared
as micronuclei, and it only differs in their size as nuclei in morphology.

10.1.12 AO/EB Staining

Acridine orange is a membrane permeable vital dye and will stain both live and
dead cells. Ethidium bromide membrane will stain only cells that have lost mem-
brane integrity [15].

10.1.13 JC-1 Staining

Disruption of active mitochondria marks the early stages of programmed cell death.
This mitochondrial disruption comprises of alterations to the oxidation–reduction
potential and changes in the membrane potential of the mitochondria. Membrane
potential changes are thought to be a result of the opening of the mitochondrial
permeability transition pore which allows passage of various ions and small mole-
cules. Equilibration of ions as a result of which leads in to the relinquishment of
cytochrome c into the cytosol and decoupling of the respiratory chain. The mem-
brane–permeant JC-1 dye is widely utilized in apoptosis studies to monitor mito-
chondrial health [16].

10.1.14 Photoproduct Identification by LC-MS/MS

Due to presence of benzene ring and bulky structure, hair dye ingredients have abil-
ity to absorb sunlight and UV-R. After absorption it may undergo in photodegrada-
tion and form photoproduct. These hair dye photoproducts will be identified by
liquid chromatography-mass spectrometry [17].

10.1.15 Cellular Toxicity Analysis

Most of the studies related to cell population’s response to external factors mainly
comprise of cell viability and proliferation assays. Tetrazolium salts are now widely
10 Phototoxicity of Hair Dyes: Challenge for Tropical Countries 107

used as a reliable way and forms the basis to examine cell proliferation. The yellow
tetrazolium, i.e., MTT, is reduced by the action of dehydrogenase enzymes present
in metabolically active cells to generate reduced products such as NADPH and
NADH. There is formation of purple formazan crystals that can be quantified and
solubilized by spectrophotometric mean [15].

Graphical representation of hair dye mediated damage at molecular level

10.2 Conclusion

These important research findings are first time related to phototoxicity mechanism
of hair coloring dye and their ingredient. Phototoxic as well as the photogenotoxic
potential under the exposure of environmentally relevant intensity of UV-R irradia-
tion coming on the earth surface through the sunlight. Therefore, the use of hair dye
and concurrent exposure of sunlight induced oxidative stress in the skin. Identification
of photoproduct and its interaction with cellular biomolecules are equally important
to understand the total safety of hair dye. Thus, it is imperative to avoid the use of
photosensitive dyes by users and should also avoid solar radiation exposure espe-
cially peak hour for their health safety.
108 S. Goyal et al.

References
1. CIR (2003) Cosmetic ingredient review: general information
2. Foley P, Nixon R, Marks R, Frowen K, Thompson S (1993) The frequency of reactions to
sunscreens: results of a longitude population based study on the regular use of sunscreen in
Australia. Br J Dermatol 128:512
3. de Groot AC (1990) Labelling cosmetics with their ingredients. Br Med J 300:1636–1638
4. Fischer AA (1973) Cutaneous reactions to cosmetics, 2nd edn. Lea and Febiger, Philadelphia,
pp 217–241
5. EPA (1982) Phenylenediamines: response to interagency testing committee. Fed Registry
47:979
6. de Groot AC, Frosch PJ (1997) Adverse reactions to fragrances: a clinical review. Contact
Dermatitis 36:57–86
7. IARC (1993) International Agency for the Research of Cancer, World Health Organisation.
Occupational exposure of hairdressers and barbers and personal use of hair colorants. Some
hair dyes, cosmetic colorants, industrial dyestuffs and aromatic amines, IARC Monographs on
the Evaluation of Carcinogenic Risk to Humans, vol 57. World Health Organisation, Geneva
8. Teramura AH (1983) Effects of ultraviolet-B radiation on the growth and yield of crop plants.
Physiol Plant 58:415–427
9. Nohynek GJ, Fautz R, Benech-Kieffer F, Toutain H (2004) Toxicity and human health risk of
hair dyes. Food Chem Toxicol 42:517–543
10. Goyal S, Amar SK, Dubey D, Pal M, Singh J, Verma A, Kushwaha HN, Ray RS (2015)
Involvement of cathepsin B in mitochondrial apoptosis byp-phenylenediamine under ambient
UV radiation. J Hazard Mater 300:415–425
11. (1993) Issue 1 frequency of reactions to sunscreens: results of a longitudinal population based
study on the regular use of sunscreen in Australia. Br J Dermatol 128:512–518
12. Förster T, Schwuger MJ (1990) Correlation between adsorption and the effects of surfactants
and polymers on hair. Prog Colloid Polymer Sci 83:104–109
13. Boveri T (2008) Concerning the origin of malignant tumors by Theodor Boveri (translated and
annotated: Harris H). J Cell Sci 121(Supplement 1):1–84
14. SCCNFP (2003) Recommended strategy for testing hair dyes for their potential genotoxicity/
mutagenicity/carcinogenicity. SCCNCP/0720/03, pp 1–10
15. Kirkland DJ, Honeycombe JR, Lawler SD (1981) Sister chromatid exchanges before and after
hair dyeing. Mutat Res 90:279–286
16. Yu H (2002) Environmental carcinogenic polycyclic aromatic hydrocarbons: photochemistry
and phototoxicity. J Environ Sci Health C Environ Carcinog Ecotoxic Rev 20:149–183
17. Rhen L (1895) Bladder tumours in fuchsin workers. Arch Klin Chirurgie 50:588–600
Role of Personal Care Products
and Phototoxicity 11
Syed Faiz Mujtaba, Ajeet K. Srivastav, Shikha Agnihotry,
Sandeep Negi, Jaya Upadhayay, and Ratan S. Ray

Abstract
Cosmetics are chemical substances or products that are used to change the look
or fragrance of the human beings. Mostly cosmetics are applied usually on the
face and hair area. Sun acts as a natural source of different wavelengths that also
contains ultraviolet (UV-A, UV-B, and UV-C) and many other radiations. Many
ingredients present in cosmetics show absorption maxima (λmax) under visible
light followed by ultraviolet radiation. Photosensitization of cosmetics results in
the formation of reactive oxygen species (ROS), i.e., 1O2 and O2−, by the two dif-
ferent photosensitization mechanisms (type-I and type-II). The photosensitiza-
tion of cosmetic products finally leads to ROS production and photoproduct
formation. Oxidative stress mediated damage to biomolecules including DNA,
formation of cyclobutane pyrimidine dimers (CPDs), etc. Various adverse effects

Authors Syed Faiz Mujtaba and Ajeet K. Srivastav have been equally contributed to this chapter.
S. F. Mujtaba
Photobiology Division, C.S.I.R-Indian Institute of Toxicology Research, Lucknow, India
Department of Zoology, Faculty of Science, Shia P.G. College, University of Lucknow,
Lucknow, Uttar Pradesh, India
A. K. Srivastav · S. Negi
Photobiology Division, C.S.I.R-Indian Institute of Toxicology Research, Lucknow, India
Babu Banarasi Das University, Lucknow, India
S. Agnihotry
Department of Biomedical-Informatics, Sanjay Gandhi Post Graduate Institute,
Lucknow, India
J. Upadhayay
Babu Banarasi Das University, Lucknow, India
R. S. Ray (*)
Department of Zoology, Faculty of Science, Shia P.G. College, Lucknow, India
e-mail: rsray@iitr.res.in

© Springer Nature Singapore Pte Ltd. 2018 109


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_11
110 S. F. Mujtaba et al.

of cosmetics are being reported in the research articles that relate to genotoxicity,
mutagenicity, photosensitization, skin irritation, acute toxicity, percutaneous
absorption, and eye irritation. Endogenous and exogenous antioxidants suppress
cosmetic toxicity as well as cellular defense machinery (Keap1/Nrf2 and MAPK)
pathways. Phototoxicity of cosmetics may lead to skin and ocular damage as
well as immune suppression. India is among the tropical countries, where most
of the activities are done by common man in the bright sunlight like agriculture,
commerce, sports, etc. Thus, information is needed regarding the cosmetic pho-
toproducts and its phototoxicity for total human safety.

Keywords
Cosmetics · Photosensitization · Photoproducts · UV-R

11.1 Introduction

Cosmetics are chemical substances that are applied to the body surface of human
being for beautifying the outer look or skin surface. Cosmetics such as hair dyes,
sunscreens, lipsticks, etc. quite frequently cause adverse reactions such as allergy
and contact dermatitis [1]. Maximum number of cosmetic products consists of suf-
ficient water as well as nutrients that are suitable for the growth of microorganisms.
This results to the skin infections from contaminated hair dyes, body lotions, and
sunscreens. Various reports have been published related to blindness caused by con-
taminated mascara [2]. The hair dyes contain various ingredients that responsible
for cancer in human. Today there is not so much information known about the safety
as well as risk to human health by the exposure to chemical mixtures used in per-
sonal care products. As per the US Food and Drug Administration, the products
commonly referred to as personal care products (PCPs) are cosmetics. Cosmetics
can be classified as (a) skin care products, (b) hair care products, (c) face care prod-
ucts, and (d) fragrance products. Figure 11.1 shows the list of cosmetic ingredients
with their products and related health hazard. PCPs, which include sunscreens,
quite frequently cause adverse reactions [1, 3] observed in the case of contact der-
matitis (3.3%) in the patients using various cosmetic products. The adverse reaction
in the person using PCPs was contact allergic dermatitis which is seen mainly due
to lipsticks, shaving creams, and hair dyes. The ingredients present in PCPs can
cause harmful health effects like cancer, mutations in the genome, allergic reactions
in the body, respiratory diseases, as well as developmental and reproductive prob-
lems. The Center for Disease Control and Prevention (CDC) noticed that nowadays
every individual is exposed to phthalates [4]; this group of chemicals is commonly
used in PCPs. CDC also noticed the presence of heavy metals (like lead, mercury,
etc.) in PCP scan across the placenta that affect the nervous system of growing chil-
dren and adults. Studies on low doses of paraben in male juvenile rats had effect on
sperm production (European Food Safety Authority 2004).
11 Role of Personal Care Products and Phototoxicity 111

S.No. Chemicals Name of cosmetics Effects on human health


Products

1. Coal-tar colors Haircolouring dye Some colouring dyes are carcinogen in


nature could leads to cancer on their
application to the skin.

2. Diethanolamine Widely used in shampoo A suspected carcinogen.

3. Formaldehyde Eye shadow, nail paint, Carcinogen, responsible for reproductive


shampoo etc. toxicity.

4. Glycol ethers Nail polish, deodorant, Reproductive toxicity, irritation to skin,


perfumes eyes nose and throat.

5. Lead Hair dye and in eye Neurotoxic, cause decreased learning


makeup ability and behavioral defects,
reproductive toxic and carcinogen.

6. Mercury Skin-lightening cream and Neurotoxic, toxic to respiratory system,


in eye makeup kidneys, gastrointestinal and
reproductive system.

7. Parabens Deodorant, shampoo, Different types of parabens like methyl,


cream, babycare ethyl, propyl and other parabens are
productsetc. mostly reproductive toxic.

8. Phenylenediamine Hair dye Mutagenic and probable human


(PPD) carcinogen. Linked to skin irritation and
respiratory disorders.

9. Phthalates Fragrance, hair products, Liver and kidney lesions, reproductive


cream, lotion. abnormalities, carcinogenic.

Fig. 11.1 List of cosmetic ingredients with their products and related health hazards

11.1.1 Hazardous Chemicals in Cosmetics

The highly reactive chemical that is used in the cosmetics is hair dye [5]. Different
reports have confirmed the dermal absorption of sunscreens and its delivery to blood
and urine. The chemical α-hydroxy acid which is a widely used cosmetic ingredient
is known for reducing aging signs in the human skin [6, 7]. The attention of people
begins regarding increased sensitivity to ultraviolet radiation for α-hydroxy acids
which is responsible for DNA damage and formation of sunburn cells [8].
Benzophenone, an active ingredient in sunscreen, is detected in human breast milk
[9]. Sunscreen ingredients affect the environment by their entry through the skin
during swimming or bathing [10].
112 S. F. Mujtaba et al.

By the application of eye shadow, around 12% of chemical reactions takes place
on the eyelid. The most commonly used eye cosmetic is mascara that causes adverse
effect mainly Pseudomonas aeruginosa corneal infections, which can permanently
destroy visual activity. Kajal and surma contains carbon compounds in addition to
mercury and lead which may cause serious health problems.
The depletion of ozone has increased UV radiation on the Earth’s surface which
leads to the biological damaging of the cells [11]. UV radiation which is reaching to
Earth is divided mainly into two parts, i.e., UV-A (95%) and a very less amount of
UV-B (5%). In day-to-day life, people are constantly being exposed to UV-A [12].
UV light present in sunrays is responsible for the damage of DNA, mutation in
genes, and photocarcinogenesis [13]. UV light is also known to increase the toxicity
of cosmetics through photoactivation and photomodification. The ingredients of
PCPs generally absorb ultraviolet [14]. Studies have demonstrated the lethal effect
of PCPs with sunlight. The most common sensitizers are the benzophenones [15].
Furthermore, the cosmetic products get photomodified under sunlight/UV-R expo-
sure to a variety of photoproducts [16] having different bioactivities compared with
the parent compound [17]. PCPs ingredients and their photoproducts are not
restricted to the human skin, but they can accumulate to different organs [10]. Thus,
this focuses to evaluate the toxicity and phototoxicity of different cosmetic ingredi-
ents as well as to identify the novel cosmetic photoproducts for the human safety
and their total environmental fate.

11.2 Photosensitization Mechanism of Cosmetics

Most of the cosmetic products have absorption maxima (λmax) under UV-R and
visible spectrum. Because of the presence of π orbital system, cosmetic products
can absorb sunlight in the visible and UV regions of solar spectrum. There are two
different mechanisms that may occur during photosensitization reactions, i.e., type-
­I and type-II. Photosensitized cosmetic ingredients get excited to upper energy
states (singlet or triplet) which then undergo electron or energy transfer to molecu-
lar oxygen or biological molecules in the cell to produce reactive oxygen species. In
almost every environmentally relevant scenario, it is inevitable that a cosmetic
ingredient either a hair dye, lipstick, mascara, sunscreen, or others will be exposed
to sunlight. When exposed to radiation, molecules capable of absorbing UV-R and
visible light become excited, resulting in different biological effects. Figure 11.2
shows absorption spectra of cosmetic ingredients (2-amino-3-hydroxypyridine,
2,3-diaminophenazine, eosin Y, phloxine B, benzophenone, benzophenone-1, etc.)
having absorption in visible and UV-R regions of solar spectrum.
Photosensitized cosmetics can emit a photon via fluorescence, which in turn
returns the molecule to its singlet ground state. The cosmetics can also give off their
energy through vibrational state as heat and return to the singlet ground state.
However, while the molecule is in its excited singlet state (ESS), it can also undergo
a process called intersystem crossing, during which there is a transition from the
ESS to a spin state called the excited triplet state (ETS), designated 3CPs*. Emitted
11 Role of Personal Care Products and Phototoxicity 113

Fig. 11.2 Absorption spectra of cosmetic ingredients: (a) 2-amino-3-hydroxypyridine, (b) 2–3
diaminophenazine, (c) eosin Y, (d) phloxine B, (e) benzophenone, and (f) benzophenone-1

radiation through intersystem crossing by excited triplet state of molecular oxygen


is known as phosphorescence. Molecular oxygen in nature is primarily found in a
ground triplet state (3O2).
All biological molecules exist in a singlet ground state. Photoexcited cosmetic
leads to the production of ROS (1O2, O2−, and •OH radical) through type-I and type-
­II photosensitized reactions (Fig. 11.3). Figure 11.4 shows the UNCI and IUPAC
name of some commonly used cosmetic ingredients with their λmax and
solubility.

11.2.1 Phototoxic Potential of Hair Dyes

According to the European Commission Scientific Committee on Consumer Safety,


46 hair dye substances act as sensitizer [18]. Allergy reactions cases have been seen
increasing in recent years by the use of hair dyes [19]. Chemical compounds present
in hair dyes can cause respiratory tract injuries by inhalation or its contact to the
skin [20]. An intermediate oxidative dye like paraphenylenediamine (PPD) is used
in hair dye formulations. PPD generates O2− and •OH radical through type-I photo-
sensitization reaction which leads to damage of genetic material and apoptosis in
skin keratinocytes [17]. The National Cancer Institute suggests that 2% cases of
114 S. F. Mujtaba et al.

1 Singlet oxygen state


CPs*

3
CPs*
Intersystem crossing

Fluorescence
Triplet state

Phosphoresence

Absorption
3
O2
1
O2 ROS
(OH, H2O2, etc)
1
CPs
Ground state

Fig. 11.3 The energy diagram showing the physical events accompanying the absorption of pho-
ton by cosmetic products (CPs). Abbreviations: 1CPs ground state, 1CPs* excited singlet state,
3
CPs* excited triplet state, 3O2 triplet (ground) state oxygen, 1O2 singlet (excited) state oxygen

non-Hodgkin’s lymphoma among women are due to regular use of hair dye prod-
ucts. PPD is found in nearly all hair coloring products and also was shown to be
carcinogenic to the breast. Daily use of hair dyes by the mothers after pregnancy is
a health risk to children [21]. Hair dyes may cause redness and irritation to the eyes.
There may be sores, itching, and burning sensations on the scalp [22]. Various stud-
ies suggested about the penetration of hair dyes inside the hair follicle [23]. From a
public health perspective, such exposure of hair dyes needs special attention by the
adult population worldwide, especially the people of tropical countries in which
outdoor activities and sunlight exposure are much common, which results in a high
attributable risk for mutagenesis and carcinogenesis.

11.2.2 Sunscreens: A Protector or Destructor

Sunscreen is a safeguard against extensive exposure of sunburn and consists UV


filters which either absorb or reflect sunlight. The excessive exposure to sunlight
increased the cases of skin diseases; this leads to increased sunscreen users.
Sunscreen use is considered as a safe practice, but study has reported sunscreens fail
to protect users and also induced ROS generation in the presence of sunlight
11 Role of Personal Care Products and Phototoxicity 115

exposure along with sunscreen and hormones related to reproduction are affected in
human beings [14, 24]. In India, the Bureau of Indian Standards (BIS) has listed
permitted UV filters which PCPs may contain (IS 2009). Benzophenone-1 (sun-
screen ingredient) has harmful effects in the endocrine hormone [25]. Sunscreen
ingredients absorb sunlight to get photosensitized [26]. Photosensitized benzophe-
none generates ROS (1O2, O2−, and •OH) under sunlight exposure, which resulted in
the formation of cyclobutane pyrimidine dimers (CPDs) in skin keratinocytes [14,
27]. Nowadays people switch to safer alternatives which can protect the skin or the
proper use of sunscreens.

11.2.3 Toxic Effects of Lipsticks and Kajal

Lipstick is a PCP which is a mixture of waxes, oils, and various types of pigments.
The constituents are present to provide color, texture, and protection to the lips.
Research done by US consumer group campaign for safety cosmetics noticed 60%
of lipsticks that were tested contained trace amounts of lead mainly in red lipsticks
[28]. Phototoxicity assessment of lipsticks was studied through the production of
ROS which induced hemolysis of RBCs and caused lipid peroxidation. Previous
study suggests that exposure of sunlight should be avoided after the application of
beauty care products such as lipsticks [29].
Kajal is a part of PCPs that is used in the eye. A great concern over the use and
safety of kajal has been raised; its use in children is prevalent. Kajal also known as
surma is extensively used in India mainly in ophthalmology and as an eye care prod-
uct. Maximum commercially produced surma contains high levels of lead. The
regular application of surma may cause high lead concentration in the body, which
harms brain and bone marrow functioning, and also causes convulsions and anemia
[30]. As per the old beliefs, kajal is regularly used in the eyes of small children. A
significant amount of PAHs was found in all samples of kajal and surma [31]. The
concentration of PAHs ranged from 0.14 to 31.18 μg/g in kajal. Benzo(a)pyrene a
carcinogenic PAH is found in highest amount in kajal, which may lead to DNA
adduct formation and cancer. After application of these products in the eye, it is
absorbed through the cornea into the body [32]. Various research documenting toxic
effects of lead and other ingredients emphasize potential health risks of using kajal
and surma and the need for increased surveillance and regulation during manufac-
turing of these products.

11.2.4 Toxic Effect of Cosmetics on Biological System

Many of the cosmetic products generate reactive oxygen species which causes DNA
and membrane damage [33]. DNA is among the main targets for UV-induced dam-
age during the use or application of cosmetic products in humans [34]. DNA dam-
age can be of different types such as single- and double-strand breakage [35].
116 S. F. Mujtaba et al.

PCPs with their IUPAC nomenclature , Absorbance (λmax), solvent and reference.
S.no UNCI Name IUPACnomenclature , UV-absorbance (λmax) Solvent Ref.
Sunscreens

1. PABA 4-Aminobenzoic UV-B (277nm) Methanol [36]


Acid

2. Benzophenone-3 2-hydroxy- UV-B/UV-A Methanol [36]


4 methoxyphenyl (287,325 nm)
Phenylmethanone

3. Bezophenone-5 Sodium 5- benzoyl-4 UV-B/UV-A --- [37]


hydroxyl-2- methoxy (286/323 nm)
benzene sulfonate

4. Diethylamino
ethy Hexyl 2-4- diethylamino- UV-A Ethanol

hydroxybenzoyl 2-hydroxybenzol (benzoate) (3


(354 nm) [38]
hexyl benzoate

5. Avobenzone 1-(4-Methoxyphenyl)3-(4-tert UV-A DMSO [39]


- butylphenly)propane-1,3 dione

6. Mexoryl SX (3z)-3-[4-(z)-[7,7-Dimethyl-2-oxo UV-A Water [40]


-1-(sulfomethyl)-3-bicyclo[2.2.1]
Heptaxylidene]methyl]phenyl]
Methylindene]-7,7,dimethyl-2oxo-
1-bicyclo(2.2.1)heptanyl]methane
sulfonic acid
Hair Dyes

7. p-phenylenedi-amine 1,4- Diaminobenzene UV-B Water


[17]
(282 nm)
8. 2-Aminophenol 2-aminophenol UV-C Water
[41]
(229 nm)

9. Resorcinol Benzene-1,3-diol UV-C Methanol [42]


(276nm)

10. 2,5 diamine


toluene sulphate 2,5 diaminotoluene UV-B Ethanol
(303 nm) [43]

Fig. 11.4 Cosmetic ingredients with their UNCI and IUPAC name as well as their UV absorbance
(λmax) and solubility
11 Role of Personal Care Products and Phototoxicity 117

Lipsticks and Kajal

11. Phloxin B Disodium 2,4,5,7 tetra Visible Water

Bromo 4,5,6,7 tetrachloro 3- (538 nm) [44]


oxospiro [2-benzofuran 1,9
xanthenes] 3,6 diolate

12. Eosin Y 2-(2,4,5,7-tetrabromo 6-oxido Visible Water

3-oxo-3H-xanthen-9yl)benzoate (531 nm)


[45]

13. Ultramarine -------- Visible Water

blue (589 nm)


[46]

14. Tocopheryl [(2R)-2,5,7,8-Tetramethyl-2-[(4R, UV-B Ethanol -


acetate 8R)-4,8,12-trimethyltridecyl] (292 nm)
chroman-6-yl] acetate

Fig. 11.4 (continued)

Figure 11.5 showed photosensitized cosmetics induced ROS-mediated DNA and


membrane damage.
Activation of cell repair machinery such as excision repair (base excision and
nucleotide excision repair) plays an important role in DNA repair. Previous studies
have reported the role of free radicals in sunscreens and hair dye phototoxicity [14,
17].
Figure 11.6 shows a proposed schematic presentation of cosmetic photosensiti-
zation and cellular damage including PS translocation, change in Bax/Bcl2 ratio,
and mitochondrial and lysosomal damage. Sunscreens enhance UV-mediated gen-
eration of free radicals in skin cells [47]. Hair dye components are potent nephro-
toxic agent, with severe nephrotoxicity in humans [48]. The use of mercury in
cosmetic products like skin-whitening creams and prolonged use of PCPs which
contain mercury can lead to inflammation of the liver, kidneys, and urinary tract
[49]. Thus, the use of heavy metal like mercury in skin creams has become a global
public health issue [50].

11.2.5 Cosmetic Photoproducts: Formation and Toxic Effects

PCPs, especially those that are directly in contact to skin for longer time, with the
exposure of sunlight, lead to preservative inactivation and lead to production of
118 S. F. Mujtaba et al.

Fig. 11.5 Cosmetic photosensitization and photomodification. Photosensitized cosmetics gener-


ate 1O2, O2−, and •OH radical which damages the DNA (single- and double-strand breakage)

hazardous photoproducts [51]. Photostability in the case of sunscreens varies. Most


of the sunscreens available in the market with different brand names fail to protect
users and break within 30 min after sunlight exposure [52].
As many of the cosmetic ingredients have absorption maxima (γ max) in visible
and UV range of solar spectrum, after absorption of a particular wavelength of light,
cosmetic products get photomodified into various novel photoproducts possessing
enhanced toxicity [53] (Fig. 11.7). Previous studies showed that benzophenone an
active ingredient of sunscreens photodegrades within 4 h under UV-R and forms
two novel photoproducts, 2-hydroxy-phenylmethylidyne-oxonium and bis2,4-­
dihydroxy-­cyclohexa-1,4-dienyl-methanone [14]. Paraphenylenediamine (PPD) a
hair dye ingredient photodegraded under UV exposure and forms aniline as its pho-
toproduct [17]. Benzophenone-1, a sunscreen UV blocker, is not photostable under
environmental UV radiation [14]. Quinoline yellow is a cosmetic ingredient which
is applied to the skin, lips, or body surface [54]. The photooxidation product of
quinoline yellow dye is detected as 4,4-diaminodiphenylmethane, 2-methoxy-­5-
methylaniline, and 4–4-oxydianiline [34]. Thus, studies show that cosmetic ingredi-
ents photodegrade and form different photoproducts. Therefore, it is necessary to
study the novel photoproducts and their toxic response to understand the total fate
of cosmetics and its effects on human beings.
11

SUN
HAIR DYES

(hair dyes)

TYPE-I
PHOTOMODIFICATION PHOTOTOXICITY

TYPE -II CELL MEMBRANE


PHOTOPRODUCT

PS TRANSLOCATION
HAIR DYES PPs

LYSOSOME
ROS
Mitochondrial depolaristion

LYSOSOMAL Bax
DESTABLISATION
NUCLEUS Bcl-2
Role of Personal Care Products and Phototoxicity

DNA DAMAGE

APOPTOSIS

Fig. 11.6 Cosmetic photosensitization and cellular damage. PS translocation, change in Bax/Bcl2 ratio, lysosomal and mitochondrial damage, and finally
apoptosis
119
120 S. F. Mujtaba et al.

a)
NH2 NH2 NH2
H
-
[ H+]

• NH ⊕ NH2
• 2 ⊕ NH2
p-phenylenediamine
m/z= 109.3

NH2 ⊕
NH2

NH3

aniline
m/z= 92.05 ⊕ NH3

b) •• H
N NH2 H N
N NH NH

OH O
OH
••
2-amino-3-hydroxypyridine
m/z=111.5

N NH
CN


OH H O

3-cyanopropioloyl cyanide H
m/z=105.01

Fig. 11.7 Schematic representation of cosmetic photodegradation and identification of cosmetic


photoproducts under sunlight/UV exposure. (a) Photomodification of p-phenylenediamine into
aniline (m/z = 92.05), (b) photoconversion of 2-amino-3-hydroxypyridine into 3-cyanopropioloyl
cyanide (m/z = 105.01), (c) photodegraded benzophenone forms two photoproducts (P1 and P2)
with m/z = 251 and m/z = 121, respectively. (Adapted from Shruti and Saroj et al. 2015)
11 Role of Personal Care Products and Phototoxicity 121

c) O •O•
• •

hv
Photomodification

Molecular Ion [M+=183]



Benzophenone O

C

Oxidation/
Reduction [O] / [H+] +

[O]

OH O OH OH O

HO OH
Photoproduct (P2) Photoproduct (P1)
[M+=251) [M+=121)
Bis-(2,4-dihydroxy- 2-Hydroxy-
cyclohexa-1,4-dienyl)- phenylmethylidyne-
methanone oxonium

Fig. 11.7 (continued)

11.2.6 Use of Antioxidants in Cosmetic Toxicity

The harmful effects of free radicals are protected by the antioxidants like
α-tocopherol (vitamin E) or ascorbic acid (vitamin C) which can be taken as a sup-
plement to stop the free radicals before they reach the dermis and thus helpful in the
prevention of genetic and cellular damage of skin cells.
Vitamin E provides protection in the presence of sunscreens which has been
studied by measuring the reduction in lipid peroxidation. The topical application of
vitamin C had shown to reduce the number of sunburn cells as compared to control
[55]. Carlotti et al. studied the effect of the different antioxidants like phenylalanine,
sodium ascorbyl phosphate, and ascorbyl palmitate on the oxidation of linoleic acid
with porcine skin in the absence and presence of TiO2. The mixture was irradiated
for 2 h with a UV-B (2.4 W/m2); their studies showed that the inhibition of photo-
peroxidation by all three antioxidants was dose-dependent. The skin has a network
122 S. F. Mujtaba et al.

of protective antioxidants [19]. It includes endogenous enzymatic antioxidants such


as GSH peroxidase, SOD, and catalase and nonenzymatic low-­molecular-­weight
antioxidants such as vitamin E, vitamin C, GSH, uric acid, and ubiquinone [56].
After sunlight exposure on healthy individual, very less catalase expression and
high protein oxidation were detected [57, 58].

11.2.7 Molecular Mechanism of Cosmetic Phototoxicity

Regular exposure to sunlight along with cosmetics eventually leads to accumulation


of several irreversible damages [59]. Most of the chemicals including cosmetic
products have bulky functional group, cyclic ring, or aromatic/aliphatic ring which
reflects the absorption potential toward sunlight. Due to this nature, cosmetics
absorb light, and thereafter they are ionized and degraded into their photoproducts
(which can be more toxic than parent compound) and induced oxidative stress
which may lead genotoxicity followed by carcinogenicity [14, 60–62].
Photosensitized cosmetics generate ROS in biological system and cause two types
of reactions, type-I or type-II or both [63, 64]. Type-II is singlet-mediated, while
type-I generates superoxide which may be responsible for the damage in cellular
systems including DNA, protein, and lipids [65]. But in certain conditions for pro-
tection, the cell starts its own defense mechanism against ROS [66]. Most efficient
mechanism which is applied by cells against ROS is keap-1, Nrf-2, and heme oxy-
genase [67]. Heme oxygenase expression which is induced during oxidative stress
in animal models seems to be protective [68].
Figure 11.8 shows the cellular protective mechanism against generation of ROS
by photosensitized cosmetics. The Keap1-Nrf2 pathway is the major regulator of
cytoprotective responses to endogenous and exogenous stresses caused by reactive
oxygen species (ROS) [69, 70]. Another pathway which is triggered during oxida-
tive stress is MAPK pathway, in which c-fos and c-jun heterodimerize to form acti-
vator protein-1 (AP-1) transcriptional activator complex that activates cells
scavenging enzyme machinery [71]. Thus, by studying such pathways, one should
be able to assess the adverse effects of cosmetic ingredients. With many studies
reporting potential health hazard with cosmetic ingredients [72], some are being
reported as genotoxic as well as probable human carcinogen [73–75]. Not only this,
many cosmetic ingredients have broad-range absorption spectra under UV-A, UV-B,
and visible light [64]. Being directly exposed to sunlight after application on hair,
face, hands, lips etc. After light absorption, cosmetic products get photosensitized
which leads to phototoxic, photoallergic, irritant contact dermatitis, as well as ana-
phylactic reactions [76]. Photoexcited cosmetics generate 1O2, O2−, and •OH radical
through type-I and type-II photosensitization reactions [77]. Previous studies have
described the role of ROS in cosmetic phototoxicity through reduction in cell viabil-
ity and DNA damage [17]. Single- and double-strand breakage, micronuclei, and
cyclobutane pyrimidine dimer formation are the DNA insults caused by photosensi-
tized cosmetics through generation of ROS which leads to mutations as well as
carcinogenesis [20]. Widespread use of these ingredients in cosmetics and their
11 Role of Personal Care Products and Phototoxicity 123

Fig. 11.8 Photosensitized cosmetics induced generation of ROS-mediated DNA and membrane
damage and cellular protective mechanism through Keap1/Nrf2 pathway and increased production
of antioxidants

direct contact exposure to sunlight paid attention for researcher worldwide due to
their toxic effects on human health [53, 78]. Ultraviolet and visible radiation in
sunlight enhances the photodegradation of many cosmetic ingredients such as para-
phenylenediamine, 2-amino-3-hydroxypyridine, benzophenone, etc. [14, 17].
Tomankova et al. [79] revealed that the four colorants frequently used in cosmetics
(P-WS caramel, chlorophyllin, Unicert Red K 7054-J, and Unicert Red 7008-J)
were tested on NIH-3T3 with and without UV radiation. The result showed an
increase of hydrogen peroxide and reduction in cellular viability, and cellular apop-
tosis was detected [80]. published the cytotoxicity of cosmetic ingredients in sea
urchin eggs by analyzing four cosmetic ingredients which showed the effect on
calcium homeostasis, intracellular pH, sodium and potassium contents, protein and
DNA synthesis as well as protein phosphorylation was also affected. Embryo-toxic
potentials of cosmetics including hydroquinone, eugenol, dibutyl phthalate, anti-
mony oxide, neodymium nitrate hexahydrate, etc. were assessed to show the embry-
onic toxicity on embryonic stem cells [81]. In the case of hair dyes like PPD, their
presence in urine sample after 2 days shows their penetration inside the body (SCCP
2013). Thus, for hair dye, further testing may be required. It is estimated that a
maximum one third of the total women above puberty and 10% of men of middle-­
aged group use some type of hair dye [82]. With the excessive use of hair dye all
over the globe, it is very important to examine the safety issues in detail.
Thus, the regulatory bodies should play a major role to prevent the human from
the use of harmful PCPs. They should highlight all hazardous ingredients used in
cosmetics and restrict all agencies about their use in PCPs.
124 S. F. Mujtaba et al.

11.3 Conclusion

Cosmetic ingredients are a silent enemy to humans, as long-term exposure may


pose serious health hazards. Most of the cosmetics, mainly the ingredients in hair
dyes and sunscreens, are classified as known or probable human carcinogen.
Cosmetic ingredients have penetration enhancers which facilitate their penetration
into the human skin. Many cosmetic ingredients have λmax under UV-R and visible
light which results in photosensitization of these ingredients. Phototoxicity and
photomodification of cosmetic ingredients may lead to local damage, and accumu-
lation of novel photoproducts in the skin could be highly toxic than the parent com-
pound. Thus, the review points further to evaluate the total effect of a cosmetic on
humans by identifying its photoproducts and their toxicity as well as suggesting
some safer alternatives.

References
1. Foley P, Nixon R, Marks R, Frowen K, Thompson S (1993) The frequency of reactions to
sunscreens. Results of a longitudinal population based study on the regular use of sunscreen in
Australia. Br J Dermatol 128:512–518
2. Linda B, Sedlewiez BS (2005) Cosmetic preservatives: friend or foe? Skinmed 4:8–100
3. Dogra A, Minocha YC, Kaur S (2003) Adverse reactions to cosmetics. Indian J Dermatol
Venereol Leprol 69:165–167
4. Center for Disease Control (CDC) (2003) Second national report on human exposure to envi-
ronmental chemicals
5. Nohynek GJ, Antignae E, Re T, Toutain H (2010) Safety assessment of personal care products/
cosmetics and their ingredients. Toxicol Appl Pharmacol 243:239–259
6. Bolt HM, Golka K (2007) The debate on carcinogenicity of permanent hair dyes: new insights.
Crit Rev Toxicol 37(6):521–536
7. de Sanjose S, Benavente y, Nieters A (2006) Association between personal use of hair dyes and
lymphoid neoplasms in Europe. Am J Epidemiol 164(1):47–55
8. Kornhauser A, Wei RR, Yamaguchi Y, Coelho SG, Kaidbey K, Barton C, Takahashi K, Beer
JZ, Miller SA, Hearing VJ (2009) The effects of topically applied glycolic acid and salicylic
acid on ultraviolet radiation- induced erythema, DNA damage and sunburn cell formation in
human skin. J Dermatol Sci 1:10–17
9. Gonzalez H, Wahlberg NT, Stromdahl B, Juzeniene A, Moan J, Larko O, Rosen A, Wennberg
AM (2007) Photostability of commercial sunscreens upon sun exposure and irradiation of
ultraviolet lamps. BMC Dermatol 7:1
10. Schlecht C, Klammer H, Franendorf H, Wuttke W, Jarry H (2008) Pharmacokinetics and
metabolism of benzophenone-2 in the rat. Toxicology 245:11–17
11. De Fabo EC (2005) Artic stratospheric ozone depletion and UV-B radiation: potential impacts
to human health. Int J Circumpolar Health 64:509–522
12. Sage E, Guirard PM, Francesconi S (2012) Unravelling, UV-A induced mutagenesis.
Photochem Photobiol Sci 11:74–80
13. Cadet J, Sage E, Douki T (2005) Ultraviolet radiation- mediated damage to cellular DNA. Mutat
Res/Fundam Mol Mech Mutagen 571(1–2):3–17
14. Amar SK, Goyal S, Mujtaba SF, Dwivedi A, Kushwaha HN, Verma A, Chopra D, Chaturvedi
RK, Ray RS (2015) Role of type I and type II reactions in DNA damage and activation of
caspase 3 via mitochondrial pathway induced by photosensitized benzophenone. Toxicol Lett
235:84–95
11 Role of Personal Care Products and Phototoxicity 125

15. Johansen JD, Rastogi SC, Menne T, Johansen JD (1996) Threshold responses in cinnamic-­
aldehyde- sensitive subjects: results and methodological aspects. Contact Dermatitis
34:165–171
16. Aharez-Rivera G, Llompart M, Garcia-Jares C, Lores M (2015) Identification of unwanted
photoproducts of cosmetic preservatives in personal care products under ultraviolet- light
using solid phase micro extraction and micro- matrix solid phase dispersion. J Chromatogr A
1390:1–12
17. Goyal S, Amar SK, Dubey D, Pal MK, Singh J, Verma A, Kushwaha HN, Ray RS (2015)
Involvement of cathepsin B in mitochondrial apoptosis by p-phenylenediamine under ambient
UV radiation. J Hazard Mater 300:415–425
18. Lewis D, Mama J, Hawkes J (2013) A review of aspects of oxidative hair dye chemistry with
special reference to N- nitrosamine formation. Materials 6:517–534
19. McArdle F, Rhodes LE, Parslew R, Jack CIA, Friedmann PS, Jackson MJ (2002) UV-R
induced oxidative stress in human skin in vivo: effects of oral vitamin c supplementation. Free
Rad Biol Med 33(10):1355–1362
20. Conto AC, Ferreira JD, Rosa ACS, Ponnbo-de-oliveira MS, Koifman S, Brazilian Collaboration
study group of infant acute leukemia (2013) Pregnancy, maternal exposure to hair dyes and
hair straightening cosmetics and early age leukemia. Chem-Biol Interact 205:46–52
21. Saitta P, Christopher EC, Jane LM, Ronald B, Benedict CW, Steven KG, Holland J (2013) Is
there a true concern regarding the use of hair dye and malignancy development? J Clin Aesthet
Dermatol 6(1):39–46
22. Thyssen JP, Sosted H, Uter W, Schnuch A, Gimenez-Arnan AM, Vigan M, Rustemeyer T,
Granum B, McFadded J, White JM, White IR, Goossens A, Menne T, Liden C, Johansen JD
(2012) Self testing for contact sensitization to hair dyes-scientific considerations and clinical
concerns of an industry-led screening programme. Contact Dermatitis 66(6):300–311
23. Steiling W, Kreutz J, Hofer H (2001) Percutaneous penetration/dermal absorption of hair dyes
in vitro. Toxicol In Vitro 15(4–5):565–570
24. Krause M, Klit A, Blomberg Jensen M, Soeborg T, Frederiksen H, Schlumpf M, Lichtensteiger
W, Skakkeback NE, Drzewiecki KT (2012) Sunscreens: are they beneficial for health? An
overview of endocrine disrupting properties of UV-filters. Int J Androl 35(3):424–436
25. Suzuki T, Kitamura S, Khota R, Sugihara K, Fujimoto N, Ohta S (2005) Estrogenic and anti-
androgenic activities of 17 benzophenone derivatives used as UV stabilizers and sunscreens.
Toxicol Appl Pharmacol 203:9–17
26. Weiqmann HJ, Schanzer S, Antonion C, Sterry W, Lademann J (2010) Influence of the absorp-
tion behavior of sunscreens in the short-wavelength UV-range (UV-B) and the long wavelength
UV-range (UV-A) on the relation of the UV-B absorption to sun protection factor. J Biomed
Opt 15(5):055008
27. Kockler J, Oelgemoller M, Robertson S, Glass BD (2012) Photostability of sunscreens.
J Photochem Photobiol C: Photochem Rev 13:91–110
28. Zakaria A, Bin Yo Y (2015) Heavy metals contamination in lipsticks and their associated
health risks to lipstick consumers. Regul Toxicol Pharmacol 73(1):191–195
29. Hans RK, Agarwal N, Verma K, Misra RB, Ray RS, Farooq M (2008) Assessment of the pho-
totoxic potential cosmetic products. Food Chem Toxicol 46:1653–1658
30. Mohta A (2010) A dangerous cosmetic. Oman J Opthalmol 3(2):100–101
31. Jain JP, Raiyani CV, Desai NM, Kashyap SK (1988) Polycyclic aromatic hydrocarbons in kajal
and surma eye cosmetic preparations. Hum Toxicol 79(2):195–197
32. Patel DK, Prasad S, Tripathi R, Behari JR (2009) The level of Polyaromatic hydrocarbons in
kajal and surma of major Indian brands. Int J Cosmet Sci 31:177–182
33. Wang L, Yan J, Wang S, Cohly H, Fu PP, Hwang HM, Yu H (2004) Phototoxicity and DNA
damage induced by the cosmetic ingredient chemical azulene in human Jurkat T-cells. Mutat
Res 562(1–2):143–150
34. Chequer FMD, Venancio de Paula V, de Souza Prado MR, Cunha Junior LRC, Lizier TM,
Zanoni MVB, Burbano RR, Bianchi MLP, Antunes LMG (2015) The cosmetic dye quinoline
yellow causes DNA damage in vitro. Mutat Res Genetic Toxicol Environ Mutagen 777:54–61
126 S. F. Mujtaba et al.

35. Kim S, Jin S, Pfeifer GP (2013) Formation of cyclobutane pyrimidine dimers at dipyrimidines
containing 5- hydroxymethylcytosine. Photochem Photobiol Sci 12(8):1409–1415
36. Serpone N, Salinaro A, Emeline AV, Horikoshi S, Kidaka H, Zhao JC (2002) An in vitro
systematic spectroscopic examination of the photostabilities of a random set of commercial
sunscreen lotions and their chemical UVB/UVA active agents. Photochem Photobiol Sci
1:970–981
37. Shaath NA (2010) Ultraviolet filters. Photochem Photobiol Sci 9:464–469
38. Vielhaber G, Grether-Beck S, Koch O, Johncock W, Krutmann J (2006) Sunscreens with an
absorption maximum of ≥ 360 nm provide optimal protection against UVA1- induced expres-
sion of matrix metalloproteinase-1, interleukin-1 and interleukin-6 in human dermal fibro-
blasts. Photochem Photobiol 5:275–282
39. Paris C, Lhianbet-vallet V, Jimenez O, Trullas C, Miranda M (2009) A blocked diketo form of
Avobenzone, photostability, photosensitizing properties and triplet quenching by a Triazine-­
derived UVB filter. Photochem Photobiol 85(1):178–184
40. Seite S, Moyal D, Richard S, de Rigal J, Leveque JL, Hoursean C, Fourtainer A (1998)
MexorylSx: a broad absorption UVA filter protects human skin from the effects of repeated
suberythemal doses of UVA. J Photochem Photobiol 44(1):69–76
41. Mitchell SC, Waring RH (2002) Aminophenols. In: Ullmann’s encyclopedia of industrial
chemistry. Wiley-VCH. https://doi.org/10.1002/14356007.a02_099
42. Boer J, Jemee GB (2010) Resorcinol peels as a possible self treatment of painful nodules in
hidradenitis suppurativa. Clin Exp Dermatol 35(1):36–40
43. Burnett C, Bergfeld W, Velsito D, Klaassen C, Marks J, Shank JR, Slaga T, Snyder P, Andersen
F (2010) Final amended report of the safety assessment of Toluene-2,5 diamine, Toluene-2,5
diamine sulfate and Toluene-3,4- diamine as used in cosmetics. Int J Toxicol 29:61S–83S
44. Inbaraj JJ, Kukielczak BM, Chignell CF (2005) Phloxine B phototoxicity: a mechanistic study
using HaCaT keratinocytes. Photochem Photobiol 81(1):81–88
45. Zheng H, Pan Y, Xiang X (2007) Oxidation of acidic dye Eosin Y by the solar photo fenton
processes. J Hazard Mater 141(3):457–464
46. Paterson I (2003) A dictionary of colour, pp 35, 169, 228, 396
47. Hanson KM, Enrico G, Christopher JB (2006) Sunscreen enhancement of UV-induced reactive
oxygen species in the skin. Free Radic Biol Med 41:1205–1212
48. Senthilkumaran S, Thirumalaikolundusubramanian P (2015) Acute hair dye poisoning: lurk-
ing dangers. J Mahatma Gandhi Inst Med Sci 20(1):33–37
49. Mckelvey W, Jeffery N, Clark N, Kass D, Parsons (2011) Population based inorganic mercury
biomonitoring and the identification of skin care products as a source of exposure in New York
city. Environ Health Perspect 119(2):203–209
50. Anonymous (2011) FDA bans skin whitening products due to mercury content. The manila
paper, 6 August 2011
51. Gonzalez H, Farbrot A, Larko O, Wennberg AM (2006) Percutaneous absorption of the sun-
screen benzophenone-3 after repeated whole body applications with and without UV-radiation.
Br J Dermatol 154:337–340
52. Tarras-Wahlberg N, Stenhagen G, Larko O, Rosen A, Wennberg AM, Wennerstrom O (1999)
Changes in ultraviolet absorption of sunscreens after ultraviolet irradiation. J Invest Dermatol
113:547–553
53. Alvarez-Rivera G, Llompart M, Garcia-Jares C, Lores M (2014) Identification of halogenated
photoproducts generated after ultraviolet-irradiation of parabens and benzoates in water con-
taining chlorine by solid phase microextraction and gas chromatography mass spectrometry.
J Chromatogr A 1349:105–115
54. Allam KV, Kumar GP (2011) Colorants- the cosmetics for the pharmaceutical dosage forms.
Int J Pharm Pharm Sci 3:13–21
55. Darr D, Combs S, Dunston S, Manning T, Pinnell S (1992) Topical vitamin C protects porcine
skin from ultraviolet radiation induced. Br J Dermatol 127:247–253
56. Blatt T, Wenck H, Wittern KP (2010) Alterations of energy metabolism in cutaneous aging. In:
Farage MA, Miller KW, Maibach HI (eds) Text book of aging skin. Springer, Berlin
11 Role of Personal Care Products and Phototoxicity 127

57. Rinnerthaler M, Bischof J, Streubel MK, Trost A, Richter K (2015) Oxidative stress in aging
human skin. Biomol Ther 5(2):545–589
58. Sander CS, Chang H, Salzmann S, Muller CSL, Mudiyanselage SE, Elsner P, Thiele JJ (2002)
Photoaging is associated with protein oxidation in human skin in vivo. J Invest Dermatol
118(4):618–625
59. Saha D, Tamrakar A (2011) Xenobiotics, oxidative stress, free radical vs antioxidants: dance
of death to heaven’s life. Asian J Res Pharm Sci 1(2):36–38
60. Klaunig JE, Kamendulis LM, Hocevar BA (2010) Oxidative stress and oxidative damage in
carcinogenesis. Toxicol Pathol 38(1):96–109
61. Lassalle Y, Kinani A, Rifai A, Sonissi Y, Clavaquera C, Bourcier S, Jaber F, Bouchonnet S
(2014) UV-visible degradation of boscalid- structural characterization of photoproducts and
potential toxicity using in silico tests. Rapid Commun Mass Spectrom 28(10):1153–1163
62. Sinha RP, Hader DP (2002) UV- induced DNA damage and repair. A review. Photochem
Photobiol Sci 1(4):225–236
63. Rastogi RP, Richa, Kumar A, Tyagi MB, Sinha RP (2010) Molecular mechanisms of ultravio-
let radiation induced DNA damage and repair. J Nucleic Acids 592980:p32
64. Tronnier H (1971) Photoallergy and photosensitization due to cosmetics. Med Klin
65(11):510–514
65. Foote CS (1991) Definition of type I and type II photosensitized oxidation. Photochem
Photobiol 54(5):659
66. Kuamagai Y, Kanada H, Shinkai Y, Toyama T (2013) The role of the Keap1/Nrf2 pathway in
the cellular response to methylmercury. Oxidative Med Cell Longev 848279:p8
67. Dodson M, Redmann M, Rajasekaran NS, Darley-Usmar V, Zhang J (2015) KEAP1-NRF2
signalling and autophagy in protection against oxidative and proteotoxicity. Biochem
J 469(3):347–355
68. Ghattas MH, Chuang LT, Kappas A, Abraham NG (2002) Protective effect of HO-1 against
oxidative stress in human hepatoma cell line (HepG2) is independent of telomerase enzyme
activity. Int J Biochem Cell Biol 34(12):1619–1628
69. Jaramillo MC, Zhang DD (2013) The emerging role of the Nrf2-Keap1 signaling pathway in
cancer. Genes Dev 27:2179–2219
70. Kansanen E, Kuosmanen SM, Leinonen H, Levonen AL (2013) The Keap1/Nrf2 pathway:
mechanism of activation and dysregulation in cancer. Redox Biol 1(1):45–49
71. Son Y, Cheong YK, Kin NH, Chung HT, Kang DG, Pae HO (2011) Mitogen-activated pro-
tein kinases and reactive oxygen species: how can ROS activate MAPK pathways? J Signal
Transduct 2011:792639 p6
72. Groot D, Beverdam EG, Ayong Ch T, Coenraads PJ, Nater JP (1988) The role of contact
allergy in the spectrum of adverse effects caused by cosmetics and toiletries. Arch Dermatol
124:1525–1529
73. Galiotte MP, Kohler P, Mussi G, Gattas GJ (2008) Assessment of occupational genotoxic risk
among Brazilian hairdressers. Ann Occup Hyg 52:645–651
74. Sanjose S, Benavente Y, Nicters A, Foretova L, Maynadie M, Cocco PL et al (2006) Association
between personal use of hair dyes and lymphoid neoplasm in Europe. Am J Epidermiol
164:47–55
75. Zhang Y, Kim C, Zheng T (2012) Hair dye use and risk of human cancer. Front Biosci
4:516–528
76. Asschenfeldt BL, Huegel R, Brasch J (2005) Anaphylactic reaction caused by the UV-A
absorber disodium phenyldibenzimidazole tetra sulfonate. Acta Derm Venereol 85:280–281
77. Fernandez-Garcie E (2014) Skin protection against UV-light by dietary antioxidants. Food
Funct 5(9):1994–2003
78. Minamoto K (2010) Skin sensitizers in cosmetics and skin care products. Nihon EiseiqaknZasshi
65(1):20–29
79. Tomankova K, Kejlova K, Binder S, Daskova A, Zapletalova J, Bendova H, Kolarova H, Jirova
D (2011) In vitro cytotoxicity and phototoxicity study of cosmetics colorants. Toxicol In Vitro
25(6):1242–1250
128 S. F. Mujtaba et al.

80. Amouroux I, Pesando D, Noel H, Girard JP (1999) Mechanism of cytotoxicity by cosmetic


ingredients in sea urchin eggs. Arch Environ Contam Toxicol 36(1):28–37
81. Chen R, Chen J, Cheng S, Qin J, Lui W, Zhang L, Jiao H, Yu X, Zhang X, Lahu BT, Xiang AP
(2010) Assessment of embryotoxicity of compounds in cosmetics by the embryonic stem cell
test. Toxicol Mech Methods:1–7
82. Huncharech M, Kupelnick B (2005) Personal use of hair dyes and the risk of bladder cancer:
result of meta-analysis. Public Health Rep 120:31–38
Protective Role of Phytochemicals
Against UVR 12
Deepti Chopra, Jyoti Singh, Ajeet Kumar Srivastav,
Divya Dubey, Ratan Singh Ray, and Kailash Chand Gupta

Abstract
Solar ultraviolet radiation (UVR) is a known environmental skin aggressor and
main causative agent of most types of skin cancer. Skin is the physiological bar-
rier that defends against both pathogens and chemical/physical damage. Sunlight
exposure to skin may cause cellular oxidative damage and mutilation of antioxi-
dant machinery. UVR is the key element responsible for photoageing.
Photoprotection is therefore essential to avoid such undesired instances of
UVR. In day to day life, the use of sunscreens has come up as the best photopro-
tective prerequisite. Apart from the sunscreens, special emphasis is on the use of
chemicals of natural origin having photoshielding, antioxidant, radical scaveng-
ing or repair properties. Key classes of beneficial phytochemicals include pheno-
lic acids, flavonoids, polyphenols, tea polyphenols, curcumin, resveratrol, etc.
These phytochemicals are able to modulate apoptotic machinery (Bax, Caspase-3,
Caspase-9, APAF-1) towards cell survival pathways (Bcl-2, PI3/AKT, mitogen-­
activated kinases). Several studies of medical importance have revealed that phy-
tochemicals proceed through several molecular and cellular mechanisms to
impediment or check photocarcinogenesis and photoageing. Therefore, it is very

D. Chopra · A. K. Srivastav · D. Dubey


Photobiology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-­
Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
J. Singh · R. S. Ray
Photobiology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-­
Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, India
Academy of Scientific and Innovative Research, New Delhi, India
K. C. Gupta (*)
CSIR-Institute of Genomics and Integrative Biology, New Delhi, India

© Springer Nature Singapore Pte Ltd. 2018 129


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_12
130 D. Chopra et al.

much needed to explore more photoprotective phytochemicals and understand


their molecular mechanism to prevent UVR-induced damage.

Keywords
Phytochemicals · Photoprotection · Photoageing · Ultraviolet radiation ·
Apoptosis

12.1 Introduction

Sunlight irradiating earth’s surface comprised of 56% infrared (700–5000 nm), 39%
visible (400–700 nm) and around 5% UV radiations (290–400 nm). UVR is divisi-
ble in three main classes on the basis of wavelength, UVA (320–400 nm), UVB
(290–320 nm) and UVC (200–290 nm). UVR is a known mutagen and primary
cause of human dermal cancers as accounted through epidemiological and molecu-
lar studies [1].
Different chromophores in the skin absorb UVR, viz. proteins, melanin, lipids,
DNA, trans-urocanic acid, aromatic amino acids (like tryptophan, tyrosine), etc.
UVR absorption via different photochemical reactions generates reactive oxygen
species (ROS) and oxidative stress [2]. Further, UVR-induced ROS production
alters gene and protein structure and functions [3]. ROS, viz. singlet oxygen (1O2),
superoxide anion radical (O2∙−), nitric oxide (NO), hydroxyl radical (∙OH), etc.,
cause cutaneous malignancy and skin photosensitivity [4]. ROS may speed up age-
ing and also accountable for irregular pigmentation.
UVA and UVB reaching the earth’s surface penetrate the skin to cause multiple
adverse effects [5]. The role of UVA is not well defined in human skin carcinogen-
esis in spite of it in a major portion in sunlight [6]. A broadly established action of
UVA is reactive oxygen and nitrogen species production ensuing in damage to pro-
teins, lipids and DNA. In cells, such oxidatively modified molecules negatively
affect numerous molecular pathways [7]. In a recent report, it was shown that
cyclobutane pyrimidine dimers (CPDs) are produced in larger yield than oxidative
lesions (8-hydroxy-2-deoxyguanosine) following UVA exposure. The mechanism
of UVA-induced CPD generation is diverse, probably through triplet energy shift
photosensitization [8]. UVB being the most active part of sunlight is more pho-
togenotoxic and damages cell compared to UVA. However, UVB has a lesser
amount of penetrating power over UVA and mainly acts on the skin’s epidermal
basal layer. UVB radiation provokes sunburn, immunosuppression, DNA damage,
oxidative stress, inflammation, free radical generation, photoageing and skin malig-
nancy [9]. UVB radiation has shown enhanced peroxynitrite and NO generation in
keratinocytes. UVC, corresponding to its massive energy, is highly detrimental to
the skin to induce photogenotoxic stress. UVC is completely absorbed by atmo-
spheric ozone layer.
Skin cells possess an intricate system of antioxidant molecules and enzymes that
retain sense of balance among oxidative stress and antioxidant protection and
defend cells [10]. Antioxidant agents thus play a defensive role in the development
12 Protective Role of Phytochemicals Against UVR 131

of ROS-induced dermal disorders. The main proteins involved during oxidative


stress, in response of antioxidant enzymes, are nuclear factor E2-related factor 2
(Nrf2) and kelch-like ECH-associated protein 1 (Keap1). In physiological environ-
ment, Nrf2 kept bounded with Keap1 in cytoplasm. As and when this Nrf2-Keap1
complex gets disturbed through inducers, Nrf2 rapid translocates from cytoplasm
towards the nucleus. Inside the nucleus Nrf2 binds with antioxidant response ele-
ments (ARE) and upregulates antioxidant and phase II enzyme expression [11].
Nrf2 activation was seen as a balanced approach for chemoprevention, specifically
at initiation stage of carcinogenesis [12]. Therefore, exogenous antioxidant-rich
phytochemical supplementation may well be an efficient strategy to revert the harm-
ful effects of UVA-induced ROS on cells. UVR-induced skin malignancy takes
many years to build up and might be prevented in the cancer promotion or progres-
sion stages (rate-limiting stages). The increased skin cancer incidences have raised
the reliability of photoprotection as a grave and realistic approach to control them.
Since UVR exposure is indispensable for individuals, they can modify their food
habits and living sense with a cautious use of personal care products containing
photoprotectant. Phytochemicals have earned substantial notice for the avoidance of
UV-induced dermal damage. Phytochemicals have antimutagenic, anti-­
inflammatory, anticarcinogenic, immunomodulatory and antioxidant properties,
besides having inhibitory effects on various molecular and cellular events [5, 13].
Phytochemical use should be emphasized due to their photosafety, low-down cost
and bioavailability. The chapter deals with the premedical and other clinical studies
of some phytochemicals (pomegranate, tea polyphenols, delphinidin, genistein, res-
veratrol, silymarin, luteolin, sulforaphane, grape seed proanthocyanidins, capsiate,
etc.) in the photosafety of the skin at cellular as well as molecular levels.

12.2 Role of Phytochemicals on UVR-Induced DNA Damage

UVA is a known carcinogen, but still a range of UVA-induced DNA damages are
not well understood. UVA-induced DNA damage, for instance, oxidative base dam-
age, oxidative backbone damage, CPDs or double-stranded breaks, depends upon
cellular sensitizers [14]. Cycloheterophyllin, a prenylflavone (isolated from the root
bark of Artocarpus heterophyllus), is a known antioxidant with radical scavenging
property that effectively reduces UVA-induced damage in fibroblast cells. In in vivo
experiments, cycloheterophyllin significantly increases UVA-induced skin peeling
on mice backs [15]. Hydroponically grown root extracts when treated to cultured
human lung and skin fibroblasts reduced the UVA-induced DNA damage. Total UV
irradiation showed no significant reduction in DNA damage, but the phytochemical
(extracted) principally shows photoprotection against UV-induced oxidative stress.
Resveratrol, also known as 3,5,4′-trihydroxy-trans-stilbene, is a natural polyphe-
nol with an antioxidant effect found in grapes, raspberries, blueberries, etc., which
have been studied widely for its protective effects. It activates Nrf2 to normalize ROS
and perhydride through antioxidant and phase II enzyme upregulation [16, 17].
132 D. Chopra et al.

The mechanism through which UVA-induced damages to the skin is still not well
understood. In physiological conditions, ROS donate electrons (oxidize) to close by
molecules and release additional energy to return in ground state. When ROS are
not further reduced by antioxidants, the oxidative reactions prolong with damaging
consequences. UVA-induced ROS can lead to degraded redox potential, lipid per-
oxidation, initiation of transcription and nuclear factors such as AP-1 and NF-kB
and finally stimulate cytokines (interleukin-4 and -10) that cause systemic immuno-
suppression [18]. Mechanistic studies involving antioxidants and sunscreens explic-
itly associate ROS in UVR-induced immunity suppression, shown as reduction of
epidermal Langerhans cells (LC) and repression of skin contact hypersensitivity in
few reports [19]. The sunscreen and antioxidant application prevents lessening of
epidermal LC and enhances delayed hypersensitivity. The degree of photoprotec-
tion directly relates to UVA protection level [20, 21].
UVB-induced damage to nucleic acids is vital for the commencement of UVB-­
induced carcinogenesis [22]. DNA bases directly absorb UVB to generate DNA
insults in form of CPDs and 6,4 pyrimidine-pyrimidone photoproducts [23]. Base
excision repair (BER) and nucleotide excision repair (NER) play significant role in
UV-induced DNA repair by specifically removing them through programmed cell
death [24]. UVB also causes guanine residues oxidation to form 8-Oxo-7,8-­
dihydrodeoxyguanosine (8-Oxo-dG or 8-OH-dG), a key photo-biomarker of oxida-
tive DNA damage in carcinogenesis [25].
Pomegranate fruit extract (PFE) in SKH-1 hairless mice significantly decreased
CPDs and 8-oxodG positive cells due to increased DNA repair. PFE enhanced
UVB-induced p53 and p21 proteins, which check cell division and DNA replication
for DNA repair [26]. PFE also downregulated pro-inflammatory transcription fac-
tor, NF-κB, and caspase-3 and upregulated G0/G1 phase linked with DNA, thus
shielding human skin fibroblasts from UVR-induced apoptosis [27].
Delphinidin or delphinidine, an anthocyanidin, a plant pigment and an antioxi-
dant found in pigmented fruits (grapes, cranberries, concord grapes, pomegranates,
bilberries) and vegetables (eggplant), has reduced UVB-induced DNA damage in
HaCaT cells and also in mouse skin [28]. Likewise, anthocyanin pretreatment has
negatively modulated UVB-mediated ROS generation and repressed oxidative cell
death by reduction of pro-apoptotic Bax protein levels and downregulation of cas-
pase-­3 pathway activation both at the in vitro and in vivo level [29].
Scientific reports have established that green tea polyphenols (GTP) along with
epigallocatechin-3-gallate (EGCG) decreased UVB-induced DNA damage and skin
cancer. Studies recommended that DNA repair induction (nucleotide excision
repair) by interleukin-12 (IL-12) has mediated UVB-induced CPD reduction [30].
Silibinin, a flavonolignan from Silybum marianum, major active constituent of
silymarin, extracted from milk thistle seeds has surprisingly reduced UVB-mediated
CPD formation in epidermal layer of SKH-1 hairless mice through p53–p21/Cip1
cascade activation [31, 32]. Moore and his coworkers [33] have reported that genis-
tein pretreatment safeguarded cutaneous proliferation along with repair mechanics
of human skin preceding UVB exposure.
Baicalin application notably reduced epidermal CPD formation on Balb/C mice
skin post-UVB exposure in comparison to unexposed mice. Baicalin-pretreated
12 Protective Role of Phytochemicals Against UVR 133

mouse epidermis showed declined UVB-induced apoptosis, less p53 buildup and
upregulated Bcl-2/Bax ratio [34]. Likewise olive oil topical application efficiently
reduced UVB-induced 8-oxodG formation in mouse skin, which is due to radical
scavenging property of olive oil [35]. HaCaT cell treatment with Prunella vulgaris
extract and rosmarinic acid (phenolic acid component of P.vulgaris) significantly
reduced UVB-mediated single-stranded break formation [55]. Pre- as well as post-­
treatment of keratinocytes with Litoria caerulea and Vaccinium myrtillus efficiently
reduced the amount of DNA breakages [36].

12.3  ole of Phytochemicals on UVR-Induced Reactive


R
Oxygen Species and Oxidative Stress Formation

The skin cells profusely contain ROS-detoxifying enzymes together with low-­
molecular-­mass antioxidant molecules to shield cells from ROS-mediated oxidative
stress and restore redox equilibrium/homeostasis (Fig. 12.1). ROS overproduction
results in oxidative stress that acts as an important inducer of cell structure damage
including lipids, cell membranes, DNA and proteins [4]. Studies suggested that
intracellular ROS may act as a prominent secondary messenger for intracellular
signaling cascades and maintain cancer cells oncogenic phenotype [37].
Caffeic acid and ferulic acid are known antioxidants as well as have UVA absorp-
tion properties that can guard against UVA-mediated melanogenesis via Nrf2-ARE
pathway in primary epidermal human melanocytes and melanoma cells, B16F10.
Nrf2 control the antioxidant response in various tissues including the skin. UVB
irradiation damages skin indirectly through ROS formation. Caffeic acid treatment
significantly showed reduction in conjugated dienes, lipid peroxidation and hydro-
peroxide to preserve antioxidant effect in UVB-exposed lymphocytes [38]. EGCG
treatment of normal human epidermal keratinocytes (NHEK) repressed UVB-­
mediated hydrogen peroxide (H2O2) production along with H2O2-induced mitogen-­
activated protein kinases (MAPK) phosphorylation for downstream signaling
pathways. Study reported that EGCG may be effective in reducing oxidative stress
and MAPK-mediated human skin anomalies [39]. Delphinidin-treated HaCaT cells
reduced UVB-induced lipid peroxidation [28]. UVA irradiation generated ROS in
HaCaT cells, and silibinin pretreatment preceding UVA exposure showed aug-
mented ROS and oxidative stress with programmed cell death [40]. Silibinin
reduced survivin levels in hairless mouse SKH-1 skin and induces caspase-3 for
apoptosis. Delphinidin protected HaCaT cells and mouse skin from UVB-induced
oxidative stress and programmed cell death.
In a report Genistein protected human dermis fibroblasts from UVB-mediated
cell deathlike characteristics through maintaining antioxidant enzyme levels along
with mitochondrial oxidative stress modulation via inhibition of p66Shc-dependent
cell signaling pathway [41]. Resveratrol topical application to hairless mice SKH-1
decreased UVB-induced skin edema, leukocytes infiltration, H2O2 generation and
lipid peroxidation [42]. Topical appliance of broccoli sprout extracts (sulforaphane)
134 D. Chopra et al.

UV
GTPs, silymarin, curcumim,
Phytochemicals resveratrol, apigenin,luteolin,
ferulic acid, caffeic acid
Keratinocyte

Cytoplasm

ROS Lipid
oxidative stress peroxidation
Nrf2
Antioxidant defence &
phase I/II enzymes Keap1
(SOD, HO-1) Keap1

Mitochondrial
destabilization

Cytochrome C DNA fragmentation, Nucleus


DNA damage Cell cycle
release & repair
(CPDs, 8-oxoG)
Nrf2
p53
Caspase
cascade Immunosuppression &
activation inflammation
(caspase 9, (IL-10, IL-6, Cox-2) p21 Cell survival &
APAF1, photoprotection
caspase 3)

Imbalance
Bax, BcI2 ratio Apoptosis

Fig. 12.1 Possible mechanisms of cell survival and other cell fate responses under UV irradiation
in presence of phytochemicals

to mice skin increased the levels of cytoprotective enzyme, NAD(P)H:quinone oxi-


doreductase 1 [43].

12.4  hytochemical Response on UVR-Mediated


P
Inflammation

UVB-mediated inflammatory responses include edema, inflammatory mediator


production, inflammatory cells infiltration and ROS generation [13]. UVB-mediated
inflammation is an important player in dermal cancer development. Cancer pro-
ceeds by increasing epidermis hyperplasia via cytokines (pro-inflammatory), growth
factors and cyclooxygenase-2 (COX-2) enzyme induction that consequentially
upregulates levels of prostaglandin (PG). Studies showed that ROS generated by
12 Protective Role of Phytochemicals Against UVR 135

UVB exposure act as a messenger in the inflammation cell signaling pathways [4].
NF-κB and AP-1 (activator protein-1), namely, c-Jun and c-fos, either work inde-
pendently or mutually to regulate inflammatory target gene expression. The
upstream MAPK (ERK1/2, JNK1/2 and p38) modulated the transcription factors.
UVB-induced ROS production activates the MAPK in NHEK and strongly associ-
ated with inflammation as well as carcinogenesis [44].
Sulforaphane is isothiocyanates, isolated from glucosinolate glucoraphanin
found abundantly in broccoli and its sprouts. Topical application of sulforaphane
prevented UVB-mediated sunburn and inflammation of the skin competently in
Nrf2 wild-type mice skin compared to Nrf2 knockout mice [45]. In HaCaT cells
sulforaphane activated Nrf2 and induced cytoprotective proteins expression, namely,
NADP(H):quinone oxidoreductase 1, heme oxygenase 1 (HO-1) and
L-glutamylcysteine ligase [46].
Ellagic acid reduces UVB-mediated inflammatory effects in SKH-1 hairless
mice skin [47]. The antioxidant effect of ellagic acid was straightly linked with the
augmented expression of HO-1 and superoxide dismutase as studied in HaCaT cells
and human fibroblasts. Further it was interceded through downregulation of Keap-1,
Nrf2 activation and elevated nuclear localization [48].
EGCG when topically applied to human skin extensively reduced UVB-mediated
leukocytes infiltration, myeloperoxidase activity and PG metabolites, predomi-
nantly PGE2 [49].
Black tea extract administration preceding UVB exposure had shown reduced inci-
dence and severity of erythema in murine and also in human skin [50]. Kaempferol, a
natural flavonoid, found in elevated levels in cruciferous vegetables, decreased UVB-
mediated COX-2 protein expression and transcriptional changes of COX-2 and
AP-1 in mouse epidermal cells. Capsiate treatment attenuated UVB-­induced increase
in intracellular ROS generation, ERK1/2 phosphorylation and NF-κB/p65 phosphory-
lation and nuclear translocation in keratinocytes. Capsiate repressed UVB-mediated
COX-2, inflammatory cytokines and main angiogenic factors in vitro as well as
in vivo [51]. Lutein along with zeaxanthin administration notably reduced edematous
cutaneous reaction and showed lessening of UVB-­mediated increase of ear bifold
thickening. Luteolin inhibited UVB-mediated PGE2 and IL-1α release in keratino-
cytes, signifying protection against the UVB-mediated sunburn effect [52]. Curcumin-
treated HaCaT cells show repressed COX-2, p38 and JNK expressions and
DNA-binding activity of AP-1. The reports propose that curcumin attenuates COX-2
expression by inhibiting p38 and JNK activities [53]. Anthocyanin treatment inhibited
UVB-mediated COX-2 and PGE2 production via NF-κB-dependent cell signaling
pathway and modulation of PI3K/Akt signaling pathway in HaCaT cells. Thereafter,
topical appliance of anthocyanins on SKH-1 hairless mice preceding UVB exposure
also reduced COX-2 and PGE2 induction [54].
136 D. Chopra et al.

12.5 Conclusion

UVR is a key environmental hazard to the human skin that effectively contributes to
a diversity of pathological disturbances like DNA damage, inflammatory responses,
ROS generation, immunosuppression and phototumorigenesis. Augmented ROS
generates oxidative stress that mediates cell structure damage. Unregulated cell sig-
naling pathways, distorted programmed cell death system, DNA damage, mutations
in crucial target genes and immunological suppression thus result in phototumori-
genesis. The natural agents and phytochemicals discussed in this chapter revoke the
malfunction of cell signaling pathways, programmed cell death system and diverse
cellular, molecular and biochemical processes intervened by the UVR. Globally
reported studies suggest that phytochemicals may possibly be an efficient advance
for shielding UV-mediated phototoxic damage and other dermal anomalies in
humans by means of either food supplement sources or through skin care personal
products or sunscreen formulations.

Acknowledgements KC Gupta thanks the Indian Council of Medical Research (ICMR),


New Delhi, India, for awarding Dr. A.S. Paintal Distinguished Scientist Chair at CSIR-IGIB,
New Delhi.

References
1. Afaq F (2011) Natural agents: cellular and molecular mechanisms of photoprotection. Arch
Biochem Biophys 508(2):144–151
2. González S, Fernández-Lorente M, Gilaberte-Calzada Y (2008) The latest on skin photopro-
tection. Clin Dermatol 26(6):614–626
3. Masaki H (2010) Role of antioxidants in the skin: anti-aging effects. J Dermatol Sci
58(2):85–90
4. Bickers DR, Athar M (2006) Oxidative stress in the pathogenesis of skin disease. J Investig
Dermatol 126(12):2565–2575
5. Bachelor MA, Bowden GT (2004) UVA-mediated activation of signaling pathways involved in
skin tumor promotion and progression. Semin Cancer Biol 14(2):131–138 Academic
6. Wischermann K, Popp S, Moshir S, Scharfetter-Kochanek K, Wlaschek M, De Gruijl F,
Hartschuh W, Greinert R, Volkmer B, Faust A, Rapp A (2008) UVA radiation causes DNA
strand breaks, chromosomal aberrations and tumorigenic transformation in HaCaT skin kera-
tinocytes. Oncogene 27(31):4269
7. Svobodová A, Vostálová J (2010) Solar radiation induced skin damage: review of protective
and preventive options. Int J Radiat Biol 86(12):999–1030
8. Mouret S, Baudouin C, Charveron M, Favier A, Cadet J, Douki T (2006) Cyclobutane pyrimi-
dine dimers are predominant DNA lesions in whole human skin exposed to UVA radiation.
Proc Natl Acad Sci 103(37):13765–13770
9. Timares L, Katiyar SK, Elmets CA (2008) DNA damage, apoptosis and langerhans cells—
activators of UV-induced immune tolerance. Photochem Photobiol 84(2):422–436
10. Svobodová A, Zdařilová A, Mališková J, Mikulková H, Walterová D, Vostalová J (2007)
Attenuation of UVA-induced damage to human keratinocytes by silymarin. J Dermatol Sci
46(1):21–30
12 Protective Role of Phytochemicals Against UVR 137

11. Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD, Yamamoto M (1999) Keap1
represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the
amino-terminal Neh2 domain. Genes Dev 13(1):76–86
12. Pi J, Qu W, Reece JM, Kumagai Y, Waalkes MP (2003) Transcription factor Nrf2 activation by
inorganic arsenic in cultured keratinocytes: involvement of hydrogen peroxide. Exp Cell Res
290(2):234–245
13. Nichols JA, Katiyar SK (2010) Skin photoprotection by natural polyphenols: anti-­inflammatory,
antioxidant and DNA repair mechanisms. Arch Dermatol Res 302(2):71–83
14. Cadet J, Douki T, Ravanat JL, Di Mascio P (2009) Sensitized formation of oxidatively gener-
ated damage to cellular DNA by UVA radiation. Photochem Photobiol Sci 8(7):903–911
15. Huang CH, Li HJ, Wu NL, Hsiao CY, Lin CN, Chang HH, Hung CF (2016) Photoprotective
effects of Cycloheterophyllin against UVA-induced damage and oxidative stress in human
dermal fibroblasts. PLoS One 11(9):e0161767
16. Omata Y, Saito Y, Fujita K, Ogawa Y, Nishio K, Yoshida Y, Niki E (2008) Induction of adaptive
response and enhancement of PC12 cell tolerance by lipopolysaccharide primarily through
the upregulation of glutathione S-transferase A3 via Nrf2 activation. Free Radic Biol Med
45(10):1437–1445
17. Rubiolo JA, Mithieux G, Vega FV (2008) Resveratrol protects primary rat hepatocytes against
oxidative stress damage: activation of the Nrf2 transcription factor and augmented activities of
antioxidant enzymes. Eur J Pharmacol 591(1):66–72
18. Ullrich SE (2005) Mechanisms underlying UV-induced immune suppression. Mutat Res/
Fundam Mol Mech Mutagen 571(1):185–205
19. Bestak R, Halliday GM (1996) Chronic low-dose UVA irradiation induces local suppression of
contact hypersensitivity, Langerhans cell depletion and suppressor cell activation in C3H/HeJ
mice. Photochem Photobiol 64(6):969–974
20. Stoebner PE, Poosti R, Djoukelfit K, Martinez J, Meunier L (2007) Decreased human epider-
mal antigen-presenting cell activity after ultraviolet A exposure: dose–response effects and
protection by sunscreens. Br J Dermatol 156(6):1315–1320
21. Moyal DD, Fourtanier AM (2008) Broad-spectrum sunscreens provide better protection from
solar ultraviolet–simulated radiation and natural sunlight–induced immunosuppression in
human beings. J Am Acad Dermatol 58(5):S149–S154
22. De Gruijl FR, Rebel H (2008) Early events in UV carcinogenesis—DNA damage, target cells
and mutant p53 foci. Photochem Photobiol 84(2):382–387
23. De Gruijl FR, van Kranen HJ, Mullenders LH (2001) UV-induced DNA damage, repair, muta-
tions and oncogenic pathways in skin cancer. J Photochem Photobiol B Biol 63(1):19–27
24. Sarasin A (1999) The molecular pathways of ultraviolet-induced carcinogenesis. Mutat Res/
Fundam Mol Mech Mutagen 428(1):5–10
25. Cooke MS, Loft S, Olinski R, Evans MD, Bialkowski K, Wagner JR, Dedon PC, Møller
P, Greenberg MM, Cadet J (2010) Recommendations for standardized description of and
nomenclature concerning oxidatively damaged nucleobases in DNA. Chem Res Toxicol
23(4):705–707
26. Afaq F, Khan N, Syed DN, Mukhtar H (2010) Oral feeding of Pomegranate fruit extract inhib-
its early biomarkers of UVB radiation-induced Carcinogenesis in SKH-1 hairless mouse epi-
dermis. Photochem Photobiol 86(6):1318–1326
27. Pacheco-Palencia LA, Noratto G, Hingorani L, Talcott ST, Mertens-Talcott SU (2008)
Protective effects of standardized pomegranate (Punica granatum L.) polyphenolic extract in
ultraviolet-irradiated human skin fibroblasts. J Agric Food Chem 56(18):8434–8441
28. Afaq F, Syed DN, Malik A, Hadi N, Sarfaraz S, Kweon MH, Khan N, Zaid MA, Mukhtar
H (2007) Delphinidin, an anthocyanidin in pigmented fruits and vegetables, protects human
HaCaT keratinocytes and mouse skin against UVB-mediated oxidative stress and apoptosis.
J Investig Dermatol 127(1):222–232
29. Tsoyi K, Park HB, Kim YM, Chung JI, Shin SC, Shim HJ, Lee WS, Seo HG, Lee JH, Chang
KC, Kim HJ (2008) Protective effect of anthocyanins from black soybean seed coats on UVB-­
induced apoptotic cell death in vitro and in vivo. J Agric Food Chem 56(22):10600–10605
138 D. Chopra et al.

30. Meeran SM, Mantena SK, Elmets CA, Katiyar SK (2006) (−)-Epigallocatechin-3-Gallate
Prevents Photocarcinogenesis in Mice through Interleukin-12–Dependent DNA Repair.
Cancer Res 66(10):5512–5520
31. Dhanalakshmi S, Mallikarjuna GU, Singh RP, Agarwal R (2004) Silibinin prevents ultraviolet
radiation-caused skin damages in SKH-1 hairless mice via a decrease in thymine dimer positive
cells and an up-regulation of p53–p21/Cip1 in epidermis. Carcinogenesis 25(8):1459–1465
32. Gu M, Dhanalakshmi S, Singh RP, Agarwal R (2005) Dietary feeding of silibinin prevents
early biomarkers of UVB radiation–induced carcinogenesis in SKH-1 hairless mouse epider-
mis. Cancer Epidemiol Prev Biomark 14(5):1344–1349
33. Moore JO, Wang Y, Stebbins WG, Gao D, Zhou X, Phelps R, Lebwohl M, Wei H (2006)
Photoprotective effect of isoflavone genistein on ultraviolet B-induced pyrimidine dimer for-
mation and PCNA expression in human reconstituted skin and its implications in dermatology
and prevention of cutaneous carcinogenesis. Carcinogenesis 27(8):1627–1635
34. Bing-Rong Z, Song-Liang J, Xiao-E C, Xiang-Fei L, Bao-Xiang C, Jie G, Dan L (2008)
Protective effect of the Baicalin against DNA damage induced by ultraviolet B irradiation to
mouse epidermis. Photodermatol Photoimmunol Photomed 24(4):175–182
35. Budiyanto A, Ahmed NU, Wu A, Bito T, Nikaido O, Osawa T, Ueda M, Ichihashi M (2000)
Protective effect of topically applied olive oil against photocarcinogenesis following UVB
exposure of mice. Carcinogenesis 21(11):2085–2090
36. Svobodová A, Zdařilová A, Vostálová J (2009) Lonicera caerulea and Vaccinium myrtillus fruit
polyphenols protect HaCaT keratinocytes against UVB-induced phototoxic stress and DNA
damage. J Dermatol Sci 56(3):196–204
37. Dröge W (2002) Free radicals in the physiological control of cell function. Physiol Rev
82(1):47–95
38. Prasad NR, Jeyanthimala K, Ramachandran S (2009) Caffeic acid modulates ultraviolet radia-
tion-­B induced oxidative damage in human blood lymphocytes. J Photochem Photobiol B Biol
95(3):196–203
39. Katiyar SK, Afaq F, Azizuddin K, Mukhtar H (2001) Inhibition of UVB-induced oxidative
stress-mediated phosphorylation of mitogen-activated protein kinase signaling pathways in
cultured human epidermal keratinocytes by green tea polyphenol (−)-epigallocatechin-3-­
gallate. Toxicol Appl Pharmacol 176(2):110–117
40. Narayanapillai S, Agarwal C, Tilley C, Agarwal R (2012) Silibinin is a potent sensitizer of
UVA radiation-induced oxidative stress and apoptosis in human keratinocyte HaCaT cells.
Photochem Photobiol 88(5):1135–1140
41. Wang YN, Wu W, Chen HC, Fang H (2010) Genistein protects against UVB-induced
senescence-­ like characteristics in human dermal fibroblast by p66Shc down-regulation.
J Dermatol Sci 58(1):19–27
42. Afaq F, Adhami VM, Ahmad N (2003) Prevention of short-term ultraviolet B radiation-­mediated
damages by resveratrol in SKH-1 hairless mice. Toxicol Appl Pharmacol 186(1):28–37
43. Dinkova-Kostova AT, Jenkins SN, Fahey JW, Ye L, Wehage SL, Liby KT, Stephenson KK,
Wade KL, Talalay P (2006) Protection against UV-light-induced skin carcinogenesis in
SKH-1 high-risk mice by sulforaphane-containing broccoli sprout extracts. Cancer Lett
240(2):243–252
44. Bowden GT (2008) Prevention of non-melanoma skin cancer by targeting ultraviolet-B-light
signalling. Nat Rev 2004 4:23–35
45. Saw CL, Huang MT, Liu Y, Khor TO, Conney AH, Kong AN (2011) Impact of Nrf2 on UVB-­
induced skin inflammation/photoprotection and photoprotective effect of sulforaphane. Mol
Carcinog 50(6):479–486
46. Wagner AE, Ernst I, Iori R, Desel C, Rimbach G (2010) Sulforaphane but not ascorbigen,
indole-3-carbinole and ascorbic acid activates the transcription factor Nrf2 and induces phase-2
and antioxidant enzymes in human keratinocytes in culture. Exp Dermatol 19(2):137–144
47. Bae JY, Choi JS, Kang SW, Lee YJ, Park J, Kang YH (2010) Dietary compound ellagic acid
alleviates skin wrinkle and inflammation induced by UV-B irradiation. Exp Dermatol 19(8)
12 Protective Role of Phytochemicals Against UVR 139

48. Hseu YC, Chou CW, Kumar KS, Fu KT, Wang HM, Hsu LS, Kuo YH, Wu CR, Chen SC, Yang
HL (2012) Ellagic acid protects human keratinocyte (HaCaT) cells against UVA-induced oxi-
dative stress and apoptosis through the upregulation of the HO-1 and Nrf-2 antioxidant genes.
Food Chem Toxicol 50(5):1245–1255
49. Katiyar SK, Challa A, McCormick TS, Cooper KD, Mukhtar H (1999) Prevention of UVB-­
induced immunosuppression in mice by the green tea polyphenol (–)-epigallocatechin-­3-­
gallate may be associated with alterations in IL-10 and IL-12 production. Carcinogenesis
20(11):2117–2124
50. Zhao J, Jin X, Yaping E, Zheng ZS, Zhang YJ, Athar M, DeLeo VA, Mukhtar H, Bickers DR,
Wang ZY (1999) Photoprotective effect of Black tea extracts against UVB-induced phototox-
icity in skin. Photochem Photobiol 70(4):637–644
51. Lee EJ, Jeon MS, Kim BD, Kim JH, Kwon YG, Lee H, Lee YS, Yang JH, Kim TY (2010)
Capsiate inhibits ultraviolet B-induced skin inflammation by inhibiting Src family kinases and
epidermal growth factor receptor signaling. Free Radic Biol Med 48(9):1133–1143
52. Verschooten L, Smaers K, Van Kelst S, Proby C, Maes D, Declercq L, Agostinis P, Garmyn M
(2010) The flavonoid luteolin increases the resistance of normal, but not malignant keratino-
cytes, against UVB-induced apoptosis. J Investig Dermatol 130(9):2277–2285
53. Cho JW, Park K, Kweon GR, Jang BC, Baek WK, Suh MH, Kim CW, Lee KS, Suh SI (2005)
Curcumin inhibits the expression of COX-2 in UVB-irradiated human keratinocytes (HaCaT)
by inhibiting activation of AP-1: p38 MAP kinase and JNK as potential upstream targets. Exp
Mol Med 37(3):186
54. Tsoyi K, Park HB, Kim YM, Chung JI, Shin SC, Lee WS, Seo HG, Lee JH, Chang KC, Kim HJ
(2008) Anthocyanins from black soybean seed coats inhibit UVB-induced inflammatory cylo-
oxygenase-­2 gene expression and PGE2 production through regulation of the nuclear factor-κB
and phosphatidylinositol 3-kinase/Akt pathway. J Agric Food Chem 56(19):8969–8974
55. Vostálová J, Zdařilová A, Svobodová A (2010) Prunella vulgaris extract and rosmarinic
acid prevent UVB-induced DNA damage and oxidative stress in HaCaT keratinocytes. Arch
Dermatol Res 302(3):171–181
Role of Nanotechnology in Skin
Remedies 13
Lipika Ray and K. C. Gupta

Abstract
Cosmeceutical-based industries are the fast-growing field, and nanotechnology
played an important role in cosmeceutical growth. Nanotechnology-based cos-
meceuticals have more advantages over traditional products. Nanotechnology
offers different varieties of products with enhanced bioavailability of active com-
ponent which increase visual look of cosmeceutical products for a longer period
of time. Its application covers a wide range of cosmeceutical products ranging
from photoaging, hyperpigmentation, wrinkles, hair, etc. However, augmented
demands of nanotechnology in cosmetic industry have prominent apprehension
regarding the plausible diffusion of nanoparticles in the dipper skin, thus possi-
ble side effects to the human skin. Herein, a brief overview of the various novel
nanocarriers for cosmeceuticals like liposomes, nanoemulsions, dendrimers,
solid lipid nanoparticles (SLNs), inorganic nanoparticles, nanocrystals, etc.,
nanoparticle-based cosmeceutical products existing in the marketplace, possible
health hazards caused by nanoparticles on exposure of nano-based cosmetics,
and the recent regulatory rules applied to avoid the nanotoxicity are described.

Keywords
Nanotechnology · Cosmeceuticals · Nanocarrier · Skin care

L. Ray (*)
Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute,
Lucknow, Uttar Pradesh, India
K. C. Gupta
Department of Biological Sciences and Bioengineering (BSBE)
and Centre for Environmental Science and Engineering (CESE),
Indian Institute of Technology, Kanpur, India

© Springer Nature Singapore Pte Ltd. 2018 141


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_13
142 L. Ray and K. C. Gupta

13.1 Introduction

Nanotechnology is a fast-growing technology where the particle size of particular


substance is decreased to nanometer scale. The concept of nanotechnology is coined
by renowned physicist Richard Feynman in 1959. It has been observed that matter
showed unique properties in its nanoscale. It considers the particle size 1–100 nm
so that the nanoparticles may easily penetrate the barrier and reach to the target.
Thus, nanotechnology is also very emerging and hottest technology to cosmetic
companies. Cosmeceutical industry utilizes nanotechnology to improve their prod-
ucts in respect of better skin penetration, UV protection, quality and color, long-­
term effects, etc. In the present day, this evolving technology acts a crucial role in
rising over the traditional cosmetics and allied products.
Cosmetics are described as material subjected to be applied to any part of the
human body for cleansing, promoting attractiveness, beautifying, or altering the
appearance by the Food and Drug Administration (FDA)[1] (US Food and Drug
Administration). The word “cosmeceuticals” is defined as the combination of cos-
metics and pharmaceuticals. In the beautification field, the term “cosmeceuticals” is
described as a product which improves the skin appearance by different biological
activities on the skin and also takes care of various skin problems ranging from
photoaging, wrinkles, and hyperpigmentation [2–5]. It is proposed that the nanopar-
ticles quickly get absorbed into the skin and efficiently fix the injury [6, 7].
Nanotechnology-based cosmeceuticals offer several advantages like targeting of the
active therapeutic components to the desired site; greater skin retention with
improved stability of the ingredients of vitamins, antioxidants, and unsaturated fats;
greater aesthetic appearance by applying mineral sunscreen where nano-sized active
mineral does not leave any noticeable white cast; and sustained release of active
agent during long time span [5, 8, 9]. Some of the nanotechnology-based novel car-
riers of cosmetics include nanoemulsions, having transparent physical appearance
and surface properties; nanocapsules, mainly applied in skin care products; lipo-
somes, small vesicles contain traditional cosmetic products that protect from light
or oxygen; nanopigments, which give transparency in appearance and enhance the
effectiveness of sunscreen products; nanocrystals; niosomes; carbon nanotubes;
solid lipid nanoparticles; dendrimers; and fullerenes.
Although industrial use of nanoparticles has achieved many new heights, there
are also several concerns about the safety and environmental impacts of this
emerging technology. Its degree of toxicity varies based on the route of exposure
to the nanoparticles [10]. To resolve the safety issue of the nanoparticles has to be
explored by particlular tests related to toxicity, including pre-clinical and clinical
trials [11, 12].
13 Role of Nanotechnology in Skin Remedies 143

13.2 Nanoparticle-Based Cosmeceuticals in Health Care

13.2.1 Nanoemulsions

Nowadays, nanoemulsions are used in a range of cosmetic products with enhanced


activity of both hydrophilic and lipophilic types with size ranges between 50 and
500 nm [13–15]. This is also described as submicron emulsion (SME) which is opti-
cally transparent or translucent with low viscosity and thus having excellent spread-
ability. L’Oreal (French company) has numerous patents on technologies based on
nanoemulsions [16]. Although single or multiemulsions can be produced depending
on the number of core compounds present in emulsions, it has limitation of instabil-
ity of emulsions during storage [17–28]. Multiple emulsions are more popular than
single emulsions because of their low-cost process over single emulsions and, thus,
used widely in cosmeceutical industry. Numerous plant-derived bioactive com-
pounds, like flavonoids and polyphenols, are used in nanoemulsions which improve
cosmeceutical value with minor toxicity [29–32]. Zorzi et al. reported that nano-
emulsions of ethanolic extract of quercetin and Achyrocline satureioides, with size
up to 300 nm, in porcine skin studies showed enhancement of the active ingredients
on the skin with increased activities of the present antioxidant [32]. In another study,
multiple nanoemulsions of genistein (soy isoflavone) and tocomin [tocotrienol
(T3)-rich fraction of red palm oil], with 208 nm particle size, showed increased
chemoprevention of skin damage by UVB [33]. Furthermore, retinyl palmitate
nanoemulsions, having the particle size less than 275 nm, showed increased skin
penetration [34]. Similarly, curcumin nanoemulsions showed enhanced permeabil-
ity of active ingredient, thus increased transdermal availability (size 85 nm) [35].
These nanoemulsions with bioactive compounds have the capability to be useful as
novel skin cream emulsions. Although having several success stories of bioactive
compounds loaded nanoemulsions in in vitro and in vivo systems, the instability of
nanoemulsions is the major obstacle in the production and commercialization in the
cosmeceutical sectors.

13.2.2 Liposomes

Liposomes are self-enclosed spherical vesicles and are constructed by one or more
phospholipid bilayers with dimensions from 20 nm to a few hundred micrometers [5,
6, 36]. They are used in a range of cosmeceuticals as they are biocompatible, biode-
gradable, nontoxic, flexible vesicles and capable to encapsulate both hydrophobic
and hydrophilic active ingredients easily [36]. Phosphatidylcholine, which is the
144 L. Ray and K. C. Gupta

main ingredient in hair and skin care products such as conditioner, shampoo, creams,
moisturizer, lotions, etc., is the key ingredient of liposome synthesis. Some active
components like vitamins A, E, and K, along with some antioxidants such as lyco-
pene, carotenoids, and CoQ10, were used inside the liposomes which increased both
chemical and physical stability in aqueous medium [37, 38]. Recently, curcumin-­
loaded nanoliposomes (size 80 nm) were prepared and showed skin protection [39].
Dior introduced the first liposomal-based antiaging cream named “Capture” in 1986
[40].

13.2.3 Niosomes

Niosomes are vesicles made up of nonionic surfactants, which are biodegradable in


nature. Niosomes have high entrapment efficiency, enhanced penetration, and
improved chemical stability, and thus it has lower production cost as compared to
liposomes. Generally, the diameter of niosomes is varied from 100 nm to 2 μm,
where an aqueous cavity is present in the center and the outside is made by coating
with nonionic surfactant in lamellar phase [41]. Niosomes are known to deliver
active ingredients topically as circulation time of the active ingredients can be
increased in both stratum corneum and epidermis [6, 42]. Interestingly, targeted
delivery is also possible in case of niosomes by attaching targeting ligand on nio-
some surface [43]. Recently, Tavano et al. [44] prepared phyto-derived niosome
which contains multiple antioxidants such as resveratrol, curcumin, and alpha-­
tocopherol in the size up to 565 nm. This niosome improved antioxidant activity
with greater amount of skin penetration. Thus, this fact ascertained that phytochem-
icals entrapped in niosome without altering bioactivity and skin protection proper-
ties can be useful in cosmeceutical applications. In 1987, the first product “Niosome”
was launched by Lancôme [45]. Manconi et al. investigated that unilamellar nio-
somes containing Brij® 30 conferred best protection of tretinoin against photodeg-
radation [5].

13.2.4 Nanocapsules

Nanocapsules also showed promising results in cosmeceutical industry along with


biomedical applications [46–49]. In the nanocapsules, at nanoscale level (10–
1000 nm), polymeric membrane holds inner liquid core [6, 50]. The possible derma-
tological use of nanocapsules was examined when the L’Oreal (French company) in
1995 introduced their first cosmetic product which is nanocapsule-based in order to
improve their cosmetic products [51]. However, synthetic compounds are exclu-
sively nanotized using this technique [52, 53], but phytocompound-based nanocap-
sules in cosmeceuticals were limited. Recently, resveratrol- and curcumin-based
nanocapsules of size 201 nm were prepared to enhance the retention of polyphenols
on the skin [46]. This result confirms that co-encapsulation of active ingredients
improves the bioavailability and thus enhances skin protection. Chitosan hydrogels
13 Role of Nanotechnology in Skin Remedies 145

hold the active compounds of capsaicinoids in the form of nanocapsules, and active
compound reaches the skin by control release method [54–56].

13.2.5 Solid Lipid Nanoparticles (SLNs)

The SLNs are described as colloidal vehicles having size ranges from 50 to 1000 nm.
Specifically, SLNs are made of lipids such as cetyl, stearic, palmitic acid, etc. and
dispersed either in water or in presence of surfactant [57, 58]. In some cases, plant-­
derived phytocompounds such as phenolic acids, flavonoids, etc. are also used with
physiological lipids to form phyto-based SLNs. SLNs are used extensively in cos-
meceuticals as they are small sized and comprised of low toxic physiological and
biodegradable lipids which stay on the surface of the skin and enhance the diffusion
of active compounds inside the body. SLNs also have occlusive properties which are
the cause of increased skin hydration [59]. Up to 31% skin hydration was obtained
in in vivo study after the application of 4% SLNs of conventional cream for 4 weeks
[60]. Recently, vitamin A-loaded SLNs (200 nm) were prepared which improved
skin occlusion in porcine skin [61, 62]. SLNs are also used as a carrier for perfumes/
fragrances, e.g., Chanel’s Allure perfume incorporated into lipid nanoparticles to
slow down the release for a prolonged effect [63, 64]. Interestingly, SLNs also can
act as UV blockers; thus, it can be combined with organic sunscreens which may
reduce the side effects. Moreover, quercetin SLNs attached with silica (483 nm)
have shown improved skin bioavailability of quercetin and therefore showed higher
photostability [65, 66]. Similarly, caffeine-loaded (182 nm) [67], lutein-loaded [68],
and resveratrol-loaded (180 nm) [69, 70] SLNs were prepared, and all of them dis-
played enhanced bioavailability of respective phytochemicals which protected the
skin from UV light immensely. It is important to note that Dr. K. Richter, Lab
GmbH, Germany, launched Nano Repair Q10 cream and Nano Repair Q10 Serum
as antiaging products in 2005 which is the immense achievement in SLN-based
antiaging field [71].

13.2.6 Nanocrystals

Nanocrystals are assembling of several hundred to thousand atoms to form either a


single or polycrystalline arrangement with the size ranging from 10 to 400 nm for
sparingly soluble active ingredient delivery [72]. Scientific reports described that
the bioactivity was increased by 500 times in case of nanocrystal-based rutin
(scantly soluble phyto-antioxidant) compared to water-soluble rutin glucoside
derivative [73]. Similar observation is also found in case of poorly soluble plant
antioxidant, hesperidin. In 2007, Juvena introduced first nanocrystal-based skin
renewing serum (Juvedical) in the market, having rutin (flavonoid) as the active
ingredient [74]. Another nanocrystal-containing product of hesperidin is La Prairie.
The French company, Lancôme, introduced nanocrystal-based anti-wrinkle cream,
146 L. Ray and K. C. Gupta

Renergie Microlift [75, 76]. Nanocrystals are dermally more available as measured
by antioxidant effect and make the customer look much younger.

13.2.7 Dendrimers

Dendrimers are regularly branched symmetrical molecules with a tree-like struc-


ture. The diameters of such dendrimers are varying from 2 nm to 10 nm (Dendrimers
and Dendrons). L’Oreal, Unilever, and the Dow Chemical Company have numerous
patents for the use of dendrimers in cosmeceuticals for skin, nail, and hair applica-
tion [5, 6, 77]. According to Furukawa et al.’s report, carbosiloxane dendrimer can
offer good amount of sebum and glossiness, tactile sensation, water resistance, and/
or adhesive properties to the skin [78]. Thus, dendrimers cover an important part of
the cosmeceutical fields.

13.2.8 Nanogold and Nanosilver

Gold and silver nanoparticles are considered to be more valuable in cosmeceuticals


as they possess antifungal and antibacterial properties. Both silver and gold nanopar-
ticles are mainly used as an active ingredient of deodorant, face pack, and antiaging
cream.

13.2.9 Cubosomes

Cubosomes are defined as discrete nanoparticles of bicontinuous cubic liquid crys-


talline phase comprising much larger specific surface area compared to the parent
cubic phase [5, 79]. Researchers are trying to search the scope of cubosomes in skin
care, hair care, and antiperspirant arena. The well-known cosmetic companies like
L’Oreal and Nivea are trying to employ oil-in-water emulsion stabilizer based and
pollutant absorbents incorporated cubosome particles in cosmeceuticals [80–83].

13.2.10 Fullerenes

Carbon fullerenes (C60) act as an antioxidant and avert early skin aging and have
been used in the formulation of skin rejuvenation cosmeceutical products [84]. It
has been also used to carry phytocompounds in cosmeceutical sectors. Fullerene
nanocapsule with ascorbic acid and vitamin E was developed and showed
enhanced skin protective activity against premature aging by its antioxidant activ-
ities [85, 86]. Unfortunately, being carbon allotropes, fullerenes are extremely
hydrophobic and insoluble in aqueous solutions, thus having limited use in cos-
meceutical field. However, its derivatization with surfactants opens up new front
13 Role of Nanotechnology in Skin Remedies 147

in cosmeceutical field as their capability to solubilize in aqueous medium aug-


mented [87]. Radical Sponge is world’s first fullerene-based cosmetic launched in
2005.

13.2.11 Insoluble, Mineral-Based Nanoparticles

Generally, the mineral UV filters have been used in sun-blocking formulation to


increase their sun protection factor (SPF); however, these types of formulations
leave a noticeable pigmented layer on the surface of the skin. Nowadays, this prob-
lem is largely overcome by the aid of nano-sized oxides such as titanium dioxide
(TiO2) and zinc oxide. TiO2 with size ranging from 65 to 130 nm can effectively
reflect back UV radiation (ultraviolet A and B) [88]. However, the most effective
size of ZnO is ranging from 20 nm to 30 nm [89]. The beneficial effects of TiO2 and
ZnO are as follows: capacity to raise the SPF, greater range of UV defense, and their
inherent nonirritant character [90]. Thus, TiO2 and ZnO nanoformulations are cap-
tured in sunscreen/cosmeceutical market significantly. The first TiO2-based sun-
screen was introduced in 1989, and few years later, nanoformulation of ZnO was
launched in 1991 [91] (Fig. 13.1).

13.3  anotechnology-Based Cosmeceuticals Available


N
in Market

Several nanotechnology-based cosmeceuticals available in the market are listed in


Table 13.1, and phytocompound-based nanoparticles used in cosmeceuticals are
listed in Table 13.2.

13.4 Human Routes of Exposure to Nanoparticles (NPs)

Human health hazards due to nanoparticles mainly depend on the path and amount
of exposure to such materials. Nanoparticles enter into the human body primarily
via three ways, i.e., inhalation, ingestion, and dermal routes [92].

13.4.1 Inhalation

Inhalation is the most frequent way of contact of nanoparticles in the air said by the
National Institute of Occupational Health and Safety [93]. People may inhale
nanoparticles during preparation if the proper protection strategies are not employed.
On the other hand, consumers may inhale aerosolized cosmeceuticals during its
usage such as deodorant, perfumes, etc. Hence, deposition of NPs in the alveolar
areas of the lung may create pulmonary toxicity [94]. Recent research on intratra-
cheal instilled ferric oxide nanoparticles (20 nm) in rats showed some clinical and
148 L. Ray and K. C. Gupta

(a) (b)
Hydrophobic drugs

Hydrophilic drugs

Hydrophobic tails
Hydrophilic head

(c) (d) Hydrophilic


drug
Active molecule

G0
G1
G2 Hydrophobic
G3 drug

(e) (f)

Solid-lipid phase
Active molecules
Surfactant

Polymeric membrane

(g) Active
Ingredient

Inner core

Fig. 13.1 Various categories of nanoparticles. (a) Liposome containing a phospholipid bilayer
with an aqueous interior, (b) nanocrystal, (c) active molecule-loaded dendrimer, (d) cubosome
with drug-loading modalities and its interior, (e) fullerene, (f) solid lipid nanoparticles, (g) differ-
ent drug-loaded nanocapsule

pathological changes such as follicular hyperplasia, protein effusion, pulmonary


capillary vessel hyperemia, and alveolar lipoproteins in lungs [95]. According to the
National Institutes of Health, a small portion of the inhaled nanoparticles may reach
even up to the brain via nasal nerves and get straightforward entry to the blood fol-
lowed by additional organs and nervous system [96].

13.4.2 Ingestion

Ingestion of nanoparticles may take place for those cosmeceuticals that are applied
in the vicinity of the mouth or lips (e.g., lipstick, lip balm, etc.). Although large part
13 Role of Nanotechnology in Skin Remedies 149

Table 13.1 Nanotechnology-based cosmetics in the market


Type of
nanotechnology
S. no. Trade name Proposed use Manufacturer used
1. Hydra flash Bronzer daily Moisturizer Lancôme Nanocapsule
face moisturizer
2. Hydra Zen cream Moisturizer Lancôme Nanoparticles
3. Renergie Microlift Anti-wrinkle Lancôme Nanocrystals
4. Revitalift Double Lifting Anti-wrinkle L’Oreal Nanosomes
5. Eye Tender Anti-wrinkle Kara Vita Nanospheres
6. Eye Contour Nanolift Anti-wrinkle, Euoko Nanocapsules
antiaging
7. Zelens Fullerene C60 Night Antiaging Zelens Fullerene C60
Cream
8. Royal Jelly Lift Concentrate Anti-wrinkle Jafra Cosmetics Liposomes
9. NanoSunTM Sunscreen Micronisers Pty. Nanoparticles
ltd.
10. Elixir Skin Up Makeup Shiseido Nanoparticles
foundation
11. Radical Sponge Skin treatment Vitamin C60 C60 nanoparticles
BioResearch
12. Nanorama—Nano Gold Face mask Lexon Nanotech Nanoparticles
Mask Pack
13. Cosil Whitening Mask Face mask Natural Korea Nanocolloids
14. Cosil Nano Beauty Soap Cleanser Natural Korea Nanoparticles
15. Fresh As A Daisy Body Body lotion Kara Vita Nanospheres
Lotion
16. Lip Tender Lip Kara Vita Nanoparticles
moisturizer
17. Primordiale Optimum Lip Lip treatment Lancôme Nanocapsule
18. Dior Snow Pure UV Base Sunscreen Dior Nano-UV filters
SPF 50
19. Clearly it! Complexion Mist Antiacne Kara Vita Nanosphere
20. Nanosphere Plus Antiaging DermaSwiss Nanosphere
21. Nano-in Hand and Nail Moisturizer Nano-Infinity Nanocrystals
Moisturizing Serum and Nanotech
Foot Moisturizing Serum
22. TEGO® Sun TS Plus Sunscreen Degussa Nanoparticles
23. NanoSalTM Moisture Key Moisturizer Salvona Nanospheres
24. Capture Antiaging Dior Liposomes
25. Anti-Wrinkle Sun Cream Anti-wrinkle, Lancôme Nanocapsules
SPF 15 sunscreen
26. Nano Gold Firming Antiaging Chantecaille Nanoparticles
Treatment
27. Soleil Instant Cooling Sun Sun protection Lancôme Vitamin-containing
Spritz SPF 15 spray nanocapsule
28. Nano Cyclic Cleanser Silver Cleanser Nano Cyclic Silver nanoparticles
29. LifePak Nano Face gel Pharmanex Coenzyme Q10
nanoparticle
150 L. Ray and K. C. Gupta

Table 13.2 Phytocompound-based nanoparticles used in cosmeceuticals


S. no. Phytocompounds Proposed use Type of nanotechnology used
1. Rice bran oil Moisturizers, Nanoemulsion
antiaging, skin care
2. Rice bran and raspberry Sunscreens Lipid nanocarriers
seed oil
3. Lavender extracts Antiaging Polymeric poly(lactic-co-glycolic)
(PLGA) nanoparticle
4. Aloe vera extract Skin care Nanoliposome
5. Lutein Skin care Nanostructured lipid carriers and
nanoemulsion
6. Quercetin Skin care Nanostructured lipid carriers
7. Ganoderma triterpenoids Skin enhancement Nanostructured lipid carriers—gel
8. Curcumin Skin care Nanoencapsulation
9. Tocopherol Skin care Nanostructured lipid carriers
(NLCs) and nanoemulsion (NE)
10. Resveratrol Skin care Solid lipid nanoparticles versus
nanostructured lipid carriers
11. Resveratrol Skin care Niosomes
12. Resveratrol and curcumin Skin care Lipid care nanocapsules
13. Resveratrol, alpha-­ Skin care Niosomes
tocopherol, and curcumin
14. D-limonene Skin care Nanoemulsions
15. Aqueous propolis and Skin care Nanoemulsions
lycopene

of nanoparticles easily escapes from the body after intake, a small fraction might get
absorbed by the body, which subsequently migrates into many vital organs [97].

13.4.3 Dermal Route

The skin is the largest organ of the human body, and it has three layers such as the
epidermis, dermis, and hypodermis. Among them, the epidermis is the outermost
layer of the skin, while the dermis includes tough connective tissue, hair follicles,
and sweat glands. The hypodermis is built up of fat and connective tissues [6].
However, the epidermis, itself, is also divided into several layers, and its outermost
layer is the stratum corneum. The stratum corneum is sole barrier for the skin due to
its lipophilicity. Moreover, it maintains high cohesion between cells [98]. Mainly
three pathways of infiltration are involved across the skin, and these are recognized
as intercellular, trans-follicular, and trans-cellular [99]. The movement of nanopar-
ticles through the skin depends on physicochemical characteristics of the nanopar-
ticles and carriers, the character of the drug [100]. Although cosmeceuticals are
supposed to be used on normal skin, they are also applied on non-healthy and
13 Role of Nanotechnology in Skin Remedies 151

(a) Intracellular pathway


Intercellular pathway Follicular pathway

Epidermis

Dermis

Hair follicle Blood vessels

Hypodermis

Sweat
gland

(b)
Intracellular Intercellular
pathway pathway

Fig. 13.2 (a) Schematic diagram of human skin and skin penetration pathways (intercellular,
intracellular, and follicular) by means of nanoparticles may cross the stratum corneum, (b) sche-
matic diagrams for intracellular and intercellular pathways

broken skin. Thus, nature of the skin is also an important criterion for cosmeceutical
delivery through the skin. Toll et al. reported that nanoparticles penetrate the skin
through skin follicles and pores present in the skin and negligible amount of
nanoparticles found under the stratum corneum [101] (Fig. 13.2).

13.5 Nanoparticle-Based Toxicity

Nanoparticle-based cosmeceutical products applied on the skin may be toxic; owing


to its small size, they may cross the membrane easily and interact with other pro-
teins and cells. A study on toxicity of TiO2 nanoparticles has shown when TiO2
nanoparticles were administered subcutaneously to pregnant mice, TiO2 nanoparti-
cles were reached up to the offspring and damaged the brain [102]. Nanoparticles
could cause skin and organ toxicity and lung damage if they are inhaled and con-
sumed accidentally or absorbed through the skin. They even can harm unborn
152 L. Ray and K. C. Gupta

children [103]. Silver nanoparticles are well known in cosmeceutical field for their
great antimicrobial activity. However, the concentration of silver is very crucial as
those silver concentrations that are lethal for bacteria may be also lethal for both
keratinocytes and fibroblasts [104]. Therefore, clinical support is needed before
nanotechnology proceeds to commercialization and product development.

13.6 Recent Progress in Nanoproduct Regulation

Recently, the USFDA has announced an Import Alert 66-38 for skin care products
labeled as antiaging creams ([6, 105]). According to USFDA, a claim such as “mol-
ecules absorb and expand, exerting upward pressure to lift wrinkles upward” which
cause an inner structural change, and would usually call be a drug. However, several
skin care products available in the market claim that the products counteract, retard,
or control the aging process which infringes USFDA regulation. According to the
FDA, such claims are unlawful on cosmetic labeling. The European Union (EU),
the new Cosmetic Products Regulation 1223/2009, also showed concern about
nanomaterials. According to this regulation, all ingredients having nanomaterials
must be indicated on the package, with the word “nano” in suffix like TiO2-­nano
from July 11, 2013, and each nano-based product needs to provide identification,
specification, quantity, toxicological profile, safety data, and foreseeable exposure
conditions ([106]). Such specifications and detailed data must be submitted at least
6 months before a nanoparticle-based cosmetic product is placed on the market
[107, 108].

13.7 Conclusion

Growth of cosmeceutical industry is rising gradually, and nanotechnology is the


most emerging technology in this field. Nanotechnology can be effectively used to
enhance the safety, efficacy, stability, and visual appearance of the product which
will ultimately lead to greater consumer compliance. Nanoproducts should be made
up and handled in a manner that improve its values and also accomplish the health
of customers and the environment.

Acknowledgment LR gratefully acknowledges the financial support provided by Young Scientist


Grant (SB/FT/CS-034/2013) (GAP-0206), Department of Science and Technology (DST-SERB),
New Delhi, India, and CSIR-CDRI, Lucknow, for providing facility and support.

Conflict of Interests The authors declare that there is no conflict of interests


regarding the publication of this paper.
13 Role of Nanotechnology in Skin Remedies 153

References
1. U.S. Food and Drug Administration. Is it a cosmetic, a drug, or both? (Or is it soap?).
Available from: http://www.fda.gov/cosmetics/guidancecomplianceregulatoryinformation/
ucm074201.htm. Last accessed on 1 Aug 2015
2. Fulekar MH (2010) Nanotechnology: importance and application. IK International Publishing
House, India
3. Mukta S, Adam F (2010) Cosmeceuticals in day-to-day clinical practice. J Drugs Dermatol
9:62–66
4. Brandt FS, Cazzaniga A, Hann M (2011) Cosmeceuticals: current trends and market analysis.
Semin Cutan Med Surg 30:141–143
5. Duarah S, Pujari K, Devidurai R, Narayanan VH (2016) Nanotechnology-based cosmeceuti-
cals: a review. Int J App Pharm 8:8–12
6. Lohani A, Verma A, Joshi H, Yadav N, Karki N (2014) Nanotechnology-based cosmeceuti-
cals. ISRN Dermatol 2014:1–14
7. Singh R, Tiwari S, Tawaniya J (2013) Review on nanotechnology with several aspects. Int
J Res Comp Eng Electron 2:1–8
8. Mu L, Sprando RL (2010) Application of nanotechnology in cosmetics. Pharm Res
27:1746–1749
9. Padamwar MN, Pokharkar VB (2006) Development of vitamin loaded topical liposomal
formulation using factorial design approach: drug deposition and stability. Int J Pharm
320:37–44
10. BBC Research (2007) Nanostructured materials for the biomedical. Pharmaceutical, and cos-
metic markets
11. Papakostas D, Rancan F, Sterry W, Blume-Peytavi U, Vogt A (2011) Nanoparticles in derma-
tology. Arch Dermatol Res 303:533–550
12. Patidar A, Thakur DS, Kumar P, Verma J (2010) A review on novel lipid-based nanocarriers.
Int J Pharm Pharm Sci 2:30–35
13. Bernardi DS, Pereira TA, Maciel NR et al (2011) Formation and stability of oil-in-water
nanoemulsions containing rice bran oil: in vitro and in vivo assessments. J Nanobiotechnol
9:44
14. Nemitz MC, Moraes RC, Koester LS, Bassani VL, Von Poser GL, Teixeira HF (2015)
Bioactive soy isoflavones: extraction and purification procedures, potential dermal use and
nanotechnology-based delivery systems. Phytochem Rev 14:849–869
15. Schwarz JC, Baisaeng N, Hoppel M, Löw M, Keck CM, Valenta C (2013) Ultra-small NLC
for improved dermal delivery of coenyzme Q10. Int J Pharm 447:213–217
16. Patravale VB, Mandawgade SD (2008) Novel cosmetic delivery systems: an application
update. Int J Cosmet Sci 30:19–33
17. Calligaris S, Comuzzo P, Bot F et al (2015) Nanoemulsions as delivery systems of hydropho-
bic silybin from silymarin extract: effect of oil type on silybin solubility, in vitro bioacces-
sibility and stability. LWT Food Sci Technol 63:77–84
18. Cerqueira-Coutinho C, Santos-Oliveira R, dos Santos E, Mansur CR (2015) Development of
a photoprotective and antioxidant nanoemulsion containing chitosan as an agent for improv-
ing skin retention. Eng Life Sci 15:593–604
19. Goncalves VSS, Rodriguez-Rojo S, De Paz E, Mato C, Martin A, Cocero MJ (2015)
Production of water soluble quercetin formulations by pressurized ethyl acetate-in-water
emulsion technique using natural origin surfactants. Food Hydrocoll 51:295–304
20. Ha TVA, Kim S, Choi Y et al (2015) Antioxidant activity and bioaccessibility of size-­different
nanoemulsions for lycopene-enriched tomato extract. Food Chem 178:115–121
154 L. Ray and K. C. Gupta

21. Hategekimana J, Chamba MVM, Shoemaker CF, Majeed H, Zhong F (2015) Vitamin E nano-
emulsions by emulsion phase inversion: effect of environmental stress and long-term storage
on stability and degradation in different carrier oil types. Colloid Surf A 483:70–80
22. Hategekirnana J, Masamba KG, Ma JG, Zhong F (2015) Encapsulation of vitamin E: effect
of physicochemical properties of wall material on retention and stability. Carbohydr Polym
124:172–179
23. Lu LY, Liu Y, Zhang ZF, Gou XJ, Jiang JH, Zhang JZ, Yao Q (2015) Pomegranate seed oil
exerts synergistic effects with trans-resveratrol in a self-nanoemulsifying drug delivery sys-
tem. Biol Pharm Bull 38:1658–1662
24. Sigward E, Corvis Y, Doan BT, Kindsiko K, Seguin J, Scherman D, Brossard D, Mignet N,
Espeau P, Crauste-Manciet S (2015) Preparation and evaluation of multiple nanoemulsions
containing gadolinium (III) chelate as a potential magnetic resonance imaging (MRI) con-
trast agent. Pharm Res 32:2983–2994
25. Walker RM, Decker EA, McClements DJ (2015) Physical and oxidative stability of fish oil
nanoemulsions produced by spontaneous emulsification: effect of surfactant concentration
and particle size. J Food Eng 164:10–20
26. Zhang J, Bing L, Reineccius GA (2015) Formation, optical property and stability of orange
oil nanoemulsions stabilized by Quallija saponins. LWT Food Sci Technol 64:1063–1070
27. McClements DJ (2012) Advances in fabrication of emulsions with enhanced functionality
using structural design principles. Curr Opin Colloid Interf Sci 17:235–245
28. Schwarz JC, Klang V, Karall S, Mahrhauser D, Resch GP, Valenta C (2012) Optimisation of
multiple W/O/W nanoemulsions for dermal delivery of aciclovir. Int J Pharm 435:69–75
29. Bidone J, Argenta DF, Kratz J, Pettenuzzo LF, Horn AP, Koester LS, Bassani VL, Simões
CM, Teixeira HF (2015) Antiherpes activity and skin/mucosa distribution of flavonoids from
achyrocline satureioides extract incorporated into topical nanoemulsions. Biomed Res Int
2015:238010
30. Fasolo D, Bassani VL, Teixeira HF (2009) Development of topical nanoemulsions containing
quercetin and 3-O-methylquercetin. Pharmazie 64:726–730
31. Fasolo D, Schwingel L, Holzschuh M, Bassani V, Teixeira H (2007) Validation of an isocratic
LC method for determination of quercetin and methylquercetin in topical nanoemulsions.
J Pharm Biomed Anal 44:1174–1177
32. Zorzi GK, Caregnato F, Moreira JC, Teixeira HF, Carvalho EL (2016) Antioxidant effect of
nanoemulsions containing extract of achyrocline satureioides (Lam) D.C.-Asteraceae. AAPS
PharmSciTech 17:844
33. Brownlow B, Nagaraj VJ, Nayel A, Joshi M, Elbayoumi T (2015) Development and in vitro
evaluation of vitamin E-enriched nanoemulsion vehicles loaded with genistein for chemopre-
vention against UVB-induced skin damage. J Pharm Sci 104:3510–3523
34. Clares B, Calpena AC, Parra A, Abrego G, Alvarado H, Fangueiro JF, Souto EB (2014)
Nanoemulsions (NEs), liposomes (LPs) and solid lipid nanoparticles (SLNs) for retinyl pal-
mitate: effect on skin permeation. Int J Pharm 473:591–598
35. Kohl E, Steinbauer J, Landthaler M, Szeimies RM (2011) Skin ageing. J Eur Acad Dermatol
Venereol 25:873–884
36. Kaur IP, Agrawal R (2007) Nanotechnology: a new paradigm in cosmeceuticals. Recent Pat
Drug Deliv Formul 1:171–182
37. Aparajita V (2014) Liposomes as carriers in skin ageing. Int J Curr Pharm Res 6:1–7
38. Lasic DD (1998) Novel applications of liposomes. Trends Biotechnol 16:307–321
39. Chen Y, Wu Q, Zhang Z, Yuan L, Liu X, Zhou L (2012) Preparation of curcumin-loaded
liposomes and evaluation of their skin permeation and pharmacodynamics. Molecules
17:5972–5987
40. Kazi KM (2010) Niosome: a future of targeted drug delivery systems. J Adv Pharm Technol
Res 1:374–380
41. Bei D, Meng J, Youan BBC (2010) Engineering nanomedicines for improved melanoma
therapy: progress and promises. Nanomedicine 5:1385–1399
13 Role of Nanotechnology in Skin Remedies 155

42. Anisha S, Kumar SP, Kumar GV, Garima G (2010) Approaches used for penetration enhance-
ment in transdermal drug delivery system. Int J Pharm Sci 2:708–716
43. Sankhyan A, Pawar P Recent trends in niosome as vesicular drug delivery system. J Appl
Pharm Sci 2:20–32
44. Tavano L, Muzzalupo R, Picci N, de Cindio B (2014) Co-encapsulation of lipophilic antioxi-
dants into niosomal carriers: percutaneous permeation studies for cosmeceutical applications.
Colloids Surf B: Biointerfaces 114:144–149
45. L’Oréal (1989) Cosmetic and pharmaceutical compositions containing niosomes and a water-­
soluble polyamide, and a process for preparing these compositions
46. Friedrich RB, Kann B, Coradini K, Offerhaus HL, Beck RC, Windbergs M (2015a) Skin
penetration behavior of lipid-core nanocapsules for simultaneous delivery of resveratrol and
curcumin. Eur J Pharm Sci 78:204–213
47. Calderilla-Fajardo SB, Cázares-Delgadillo J, Villalobos-Garcia R, Quintanar-Guerrero D,
Ganem-Quintanar A, Robles R (2006) Influence of sucrose esters on the in vivo percutane-
ous penetration of octyl methoxycinnamate formulated in nanocapsules, nanoemulsion, and
emulsion. Drug Dev Ind Pharm 32:107–113
48. Olvera-Martínez BI, Cázares-Delgadillo J, Calderilla-Fajardo SB, Villalobos-Garcia R,
Ganem-Quintanar A, Quintanar-Guerrero D (2005) Preparation of polymeric nanocapsules
containing octyl methoxycinnamate by the emulsification-diffusion technique: penetration
across the stratum corneum. J Pharm Sci 94:1552–1559
49. Weiss-Angeli V, Poletto FS, de Marco SL, Salvador M, da Silveira NP, Guterres SS,
Pohlmann AR (2012) Sustained antioxidant activity of quercetin-loaded lipid-core nanocap-
sules. J Nanosci Nanotechnol 12:2874–2880
50. Kothamasu P, Kanumur H, Ravur N, Maddu C, Parasuramrajam R, Thangavel S (2012)
Nanocapsules: the weapons for novel drug delivery systems. Bioimpacts 2:71–81
51. Poletto FS, Beck RCR, Guterres SS, Pohlmann AR (2011) Polymeric nanocapsule: concepts
and applications. In: Beck R, Guterres S, Pohlmann A (eds) Nanocosmetics and nanomedi-
cines: new approaches for skin care. Springer, Berlin, pp 47–51
52. Guterres SS, Alves MP, Pohlmann AR (2007) Polymeric nanoparticles, nanospheres and
nanocapsules, for cutaneous applications. Drug Target Insights 2:147–157
53. Detoni CB, Souto GD, da Silva AL, Pohlmann AR, Guterres SS (2012) Photostability and
skin penetration of different E-resveratrol-loaded supramolecular structures. Photochem
Photobiol 88:913–921
54. Contri RV, Katzer T, Ourique AF, da Silva AL, Beck RC, Pohlmann AR, Guterres SS (2014a)
Combined effect of polymeric nanocapsules and chitosan hydrogel on the increase of capsa-
icinoids adhesion to the skin surface. J Biomed Nanotechnol 10:820–830
55. Contri RV, Kaiser M, Poletto FS, Pohlmann AR, Guterres SS (2011) Simultaneous control of
capsaicinoids release from polymeric nanocapsules. J Nanosci Nanotechnol 11:2398–2406
56. Contri RV, Soares RM, Pohlmann AR, Guterres SS (2014b) Structural analysis of chitosan
hydrogels containing polymeric nanocapsules. Mater Sci Eng C Mater Biol Appl 42:234–242
57. Puri D, Bhandari A, Sharma P, Choudhary D (2010) Lipid nanoparticles (SLN, NLC): a novel
approach for cosmetic and dermal pharmaceutical. J Global Pharma Technol 2:1–15
58. Ekambaram P, Sathali AAH, Priyanka H (2012) Solid lipid nanoparticles: a review. Sci Rev
Chem Commun 2:80–102
59. Pardeike J, Hommoss A, Müller RH (2009) Lipid nanoparticles (SLN, NLC) in cosmetic and
pharmaceutical dermal products. Int J Pharm 366:170–184
60. Mei Z, Wu Q, Hu S, Li X, Yang X (2005) Triptolide loaded solid lipid nanoparticle hydrogel
for topical application. Drug Dev Ind Pharm 31:161–168
61. Jenning V, Gysler A, Schafer-Korting M, Gohla SH (2000a) Vitamin A loaded solid lipid
nanoparticles for topical use: occlusive properties and drug targeting to the upper skin. Eur
J Pharm Biopharm 49:211–218
62. Jenning V, Schäfer-Korting M, Gohla S (2000b) Vitamin A-loaded solid lipid nanoparticles
for topical use: drug release properties. J Control Release 66:115–126
156 L. Ray and K. C. Gupta

63. Souto EB, Muller RH (2008) Cosmetic features and applications of lipid nanoparticles (SLN,
NLC). Int J Cosmet Sci 30:157–165
64. Wissing SA, Mader K, Müller RH (2000) Solid lipid nanoparticles (SLN) as a novel carrier
system offering prolonged release of the perfume Allure (Chanel). In: Proceedings of the
international symposium on controlled release of bioactive materials, vol 27, pp 311–312
65. Scalia S, Franceschinis E, Bertelli D, Iannuccelli V (2013) Comparative evaluation of the
effect of permeation enhancers, lipid nanoparticles and colloidal silica on in vivo human skin
penetration of quercetin. Skin Pharmacol Physiol 26:57–67
66. Scalia S, Mezzena M (2010) Photostabilization effect of quercetin on the UV filter com-
bination, butyl methoxydibenzoylmethane-octyl methoxycinnamate. Photochem Photobiol
86:273–278
67. Puglia C, Offerta A, Tirendi GG, Tarico MS, Curreri S, Bonina F, Perrotta RE (2016) Design
of solid lipid nanoparticles for caffeine topical administration. Drug Deliv 23:36–40
68. Mitri K, Shegokar R, Gohla S, Anselmi C, Müller RH (2011) Lipid nanocarriers for der-
mal delivery of lutein: preparation, characterization, stability and performance. Int J Pharm
414:267–275
69. Caddeo C, Teskac K, Sinico C, Kristl J (2008) Effect of resveratrol incorporated in liposomes
on proliferation and UV-B protection of cells. Int J Pharm 363:183–191
70. Teskac K, Kristl J (2010) The evidence for solid lipid nanoparticles mediated cell uptake of
resveratrol. Int J Pharm 390:61–69
71. Müller RH, Petersen RD, Hommoss A, Pardeike J (2007) Nanostructured lipid carriers
(NLC) in cosmetic dermal products. Adv Drug Deliv Rev 59:522–530
72. Keck CM, Müller RH (2006) Drug nanocrystals of poorly soluble drugs produced by high
pressure homogenization. Eur J Pharm Biopharm 62:3–16
73. Petersen R (2008) Nanocrystals for use in topical cosmetic formulations and method of pro-
duction thereof. Abbott GmbH & Co., US Patent 60/866233
74. http://www.essentialdayspa.com/forum/viewthread.php?tid=26729. Last accessed Jan 2012
75. Bansal S, Bansal M, Kumria R (2012) Nanocrystals: current strategies and trends. Int J Res
Pharmaceut Biomed Sci 3:406–419
76. Dendrimers & dendrons: facets of pharmaceutical nanotechnology. Drug-Dev Newsletter.
http://www.kellerfoundation.com/ME2/dirmod.asp?sid=4306B1E9C3CC4E07A4D64E23F
BDB232C&nm=Back+Issues&type=Publishing&mod=Publications%3A%3AArticle&mid
=8F3A7027421841978F18BE895F8-7F791&tier=4&id=9B9BA1DAA5BE455A85A81D9
7382FE885
77. Tournihac F, Simon P (2001) Cosmetic or dermatological topical compositions comprising
dendritic polyesters. U.S. Patent 6,287,552
78. Furukawa H, Limura T (2012) Copolymer having carbosiloxane dendrimer structure, and
composition and cosmetic containing the same. U.S. Patent 20120263662A1
79. Hyde S, Andersson A, Larsson K (1997) The language of shape, 1st edn. Elsevier, New York
80. Kimmes SC, Feltin C (2013) Cosmetic composition comprising an oil and a polymer
both bearing a hydrogen-bond-generating joining group, and cosmetic treatment process.
European Patent 2575751A1
81. Ribier A, Biatry B (2002) Cosmetic or dermatologic oil/water dispersion stabilized with
cubic gel particles and method of preparation. European Patent 0711540B1
82. Albrecht H, Schreiber J (2002) Hair care products with disperse liquid crystals exhibiting the
cubic phases. W.O. Patent 2002041850A1
83. Simonnet JT, Sonneville O, Legret S (2001) Nanoemulsion based on phosphoric acid fatty
acid esters and its uses in the cosmetics, dermatological, pharmaceutical, and/or ophthalmo-
logical fields. U.S. Patent 6274150 B1
84. Lens M (2009) Use of fullerenes in cosmetics. Recent Pat Biotechnol 3:118–123
85. Ito S, Itoga K, Yamato M, Akamatsu H, Okano T (2010a) The co-application effects of fuller-
ene and ascorbic acid on UV-B irradiated mouse skin. Toxicology 267:27–38
86. Ito Y, Warner JH, Brown R, Zaka M, Pfeiffer R, Aono T, Izumi N, Okimoto H, Morton
JJ, Ardavan A, Shinohara H, Kuzmany H, Peterlik H, Briggs GA (2010b) Controlling
13 Role of Nanotechnology in Skin Remedies 157

i­ ntermolecular spin interactions of La@C(82) in empty fullerene matrices. Phys Chem Chem
Phys 12:1618–1623
87. Cusan C, Da Ros T, Spalluto G, Foley S, Janot J-M, Seta P, Larroque C, Tomasini MC,
Antonelli T, Ferraro L, Prato M (2002) A new multi-charged C60 derivative: synthesis and
biological properties. Eur J Org Chem 17:2928–2934
88. Popov AP, Lademann J, Priezzhev AV, Myllyla R (2005) Effect of size of TiO2 nanoparticles
embedded into stratum corneum on ultraviolet-A and ultraviolet-B sun-blocking properties
of the skin. J Biomed Opt 10:1–9
89. Cross SE, Innes B, Roberts MS, Tsuzuki T, Robertson TA, McCormick P (2007) Human skin
penetration of sunscreen nanoparticles: in vitro assessment of a novel micronized zinc oxide
formulation. Skin Pharmacol Physiol 20:148–154
90. Dransfield GP (2000) Inorganic sunscreens. Radiat Prot Dosim 91:271–273
91. Murphy GM (1999) Sunblocks: mechanisms of action. Photodermatol Photoimmunol
Photomed 15:34–36
92. Oberdorster G, Oberdorster E, Oberdorster J (2005) Nanotoxicology: an emerging discipline
evolving from studies of ultrafine particles. Environ Health Perspect 113:823–839
93. Yah CS, Simate G, Iyuke SE (2012) Nanoparticles toxicity and their routes of exposures. Pak
J Pharm Sci 25:477–491
94. Kreyling WG, Semmler-Behnke M, Moller W (2006) Ultrafine particle-lung interactions:
does size matter? J Aerosol Med 19:74–83
95. Zhu MT, Feng WY, Wang Y, Wang B, Wang M, Ouyang H, Zhao YL, Chai ZF (2009)
Particokinetics and extrapulmonary translocation of intratracheally instilled ferric oxide
nanoparticles in rats and the potential health risk assessment. Toxicol Sci 107:342–351
96. Wang B, Feng WY, Wang M, Wang TC, Gu YQ, Zhu MT (2008) Acute toxicological impact
of nano-and submicro-scaled zinc oxide powder on healthy adult mice. J Nanopart Res
10:263–276
97. Paul JAB, Roel PFS (2006) Toxicological characterization of engineered nanoparticles.
In: Gupta RB, Kompella UB (eds) Nanoparticle technology for drug delivery. Taylor and
Francis, New York, pp 161–170
98. Benson HAE (2005) Transdermal drug delivery: penetration enhancement techniques. Curr
Drug Deliv 2:23–33
99. Buzea C, Pacheco II, Robble K (2007) Nanomaterial and nanoparticles: sources and toxicity.
Biointerphases 2:MR17–MR71
100. Cevc G, Vierl U (2010) Nanotechnology and the transdermal route. A state of the art review
and critical appraisal. J Control Release 141:277–299
101. Toll R, Jacobi U, Richter H, Lademann J, Schaefer H, Blume-Peytavi U (2004) Penetration
profile of microspheres in follicular targeting of terminal hair follicles. J Invest Dermatol
123:168–176
102. Takeda K, Suzuki K-I, Ishihara A, Kubo-Irie M, Fujimoto R, Tabata M, Oshio S, Nihei Y,
Ihara T, Sugamata M (2009) Nanoparticles transferred from pregnant mice to their offspring
can damage the genital and cranial nerve systems. J Health Sci 55:95–102 2009
103. Jong WHD, De H, Borm PJA (2008) Drug delivery and nanoparticles: applications and haz-
ards. Int J Nanomedicine 3:133–149
104. Poon VKM, Burd A (2004) In vitro cytotoxity of silver: implication for clinical wound care.
Burns 30:140–147
105. U.S. Food and Drug Administration, Import Alert 66–38. http://www.accessdata.fda.gov/
cmsia/importalert 188.html
106. Nanomaterials and the EU Cosmetics Regulation: Implications for Your Company. http://
www.gcimagazine.com/business/management/regulation/143553126.html?pa
107. New EU Cosmetics Regulations: A Quick Guide for Busy Formulators. http://chemistscorner.
com/new-eu-cosmeticsregulations-a-quick-guide-for-busyformulators/
108. Stafford N (2009) New nano rule for EU cosmetics. Royal society of Chemistry. http://www.
rsc.org/chemistryworld/News/2009/November/27110901.asp
Role of Photodynamic Therapy in Cancer
Treatment 14
Shikha Agnihotry, Mohammad Anas, Ajeet K. Srivastav,
Deepti Chopra, Jaya Upadhayay, and Syed Faiz Mujtaba

Abstract
Cancer is one of the most fatal diseases next only to cardiovascular diseases
spread all around the globe, and it is the third most fatal disease in India.
Environmental factors such as chemicals, UV light, tobacco products, X-rays,
viruses, and disturbance in oncogenes are the factors which induce mutations
that are inheritable and result in cancer. PDT comprises of three essential com-
ponents: photosensitizer (PS), light, and oxygen. Oxygen in the form of reactive
oxygen species can be toxic and may lead to cell death via necrosis or apoptosis.
PDT is a two-stage procedure. Administration of a light-sensitive PS is followed
by irradiation of tumor loci with a light of appropriate wavelength. This chapter
describes about oncogenes and role of photodynamic therapy in treatment of
oncogenes.

Authors Shikha Agnihotry and Mohammad Anas have been equally contributed to this chapter.
S. Agnihotry · J. Upadhayay
Department of Biomedical-Informatics, Sanjay Gandhi Post Graduate Institute,
Lucknow, Uttar Pradesh, India
M. Anas
Photobiology Division, CSIR-Indian Institute of Toxicology Research,
Lucknow, Uttar Pradesh, India
A. K. Srivastav · D. Chopra
Photobiology Division, System Toxicology and Health Risk Assessment Group, CSIR-Indian
Institute of Toxicology Research,
Lucknow, Uttar Pradesh, India
Babu Banarasi Das University, Lucknow, Uttar Pradesh, India
S. F. Mujtaba (*)
Department of Zoology, Faculty of Science, Shia P.G. College, University of Lucknow,
Lucknow, Uttar Pradesh, India

© Springer Nature Singapore Pte Ltd. 2018 159


R. S. Ray et al. (eds.), Photocarcinogenesis & Photoprotection,
https://doi.org/10.1007/978-981-10-5493-8_14
160 S. Agnihotry et al.

Keywords
Photodynamic therapy · Oncogenes · Photosensitizer · Cancer and light

14.1 Introduction

Cancer remains one of the major public health concerns. The morbidity of cancer had
a high growth in recent decades, and, in this context, there has been marked increase
in skin carcinoma incidences. There were 12 million new cases of cancer worldwide
in 2007 out of which 20% represented cutaneous localization (IARC – International
Agency for Research on Cancer). Skin cancer accounts for about 6% skin diseases
[1]. The most important feature of skin cancer is that it often develops on precancer-
ous lesions and malignant transformation may be prevented by early diagnosis and
treatment. Among the therapeutic means of skin cancer, photodynamic therapy or
PDT is a noninvasive method developed with satisfactory clinical trials.
Photodynamic therapy is a treatment which has applications in dermatology and
involves three components – a photosensitizer drug, light, and oxygen in the tissue.
It is conducted in two stages. The drug works only if it has been activated by a cer-
tain wavelength of light. PDT is also known as photoradiation therapy, photochemo-
therapy, or phototherapy.

14.2 History of PDT

PDT has now been used as a cure for various diseases which include different types of
cancer. Its use has been approved by many countries. The number of both basic and
clinical articles on photodynamic therapy published is steadily increasing every year.

14.3 The Photodynamic Action

Professor Hermann von Tappeiner, one of the pioneers in the field of photobiology, in
1904 introduced the term “photodynamic action.” According to the original definition,
the term photodynamic action should be only used for oxygen-dependent reactions. It
is known that both type I and type II photosensitization reactions require oxygen.

14.4 Phototherapy

The use of light alone for therapeutic purposes is called phototherapy, but in most
treatments, endogenous photosensitizers are usually involved; thus phototherapy
basically depends on photodynamic action. Humans for 3000 years have used sun-
rays for the treatment of psoriasis, vitiligo, skin cancer, and rickets, and its applica-
tion was familiar to Indians, Egyptians, and Chinese. Herodotus termed it as
“heliotherapy” and advised for health restoration. Sunlight effect on rickets was
14 Role of Photodynamic Therapy in Cancer Treatment 161

known in the eighteenth century. Carvin, in 1815, explained the curing effect of
sunlight on rickets, scrofula, paralysis, scurvy, muscle weakness, and rheumatism.
Sniadecki, a Polish physician in 1822, documented the role of sunlight irradiance
for the prevention of rickets. Dane Niels Finsen was awarded Noble Prize in 1903
for the treatment of lupus vulgaris by using light from the carbon arc, and he was
acknowledged as the founder of modern phototherapy for the same work. In the
1950s, Richard Cremer from Essex, England, introduced phototherapy for the treat-
ment of jaundice in newborn babies which is now the most popular form of
phototherapy.

14.5 Photochemotherapy

The treatment of vitiligo with the seeds of plant Psoralea corylifolia has been
described in Atharva Veda (1400 BC). Psoralens are the photoactive component of
these seeds, which is also found in the plant extracts of Ammi majus, growing on the
riverbanks of Nile, and was used by Egyptians for the treatment of vitiligo. There
was no further progress in the field of chemotherapy until 1974, when the treatment
of psoriasis was reported to be performed by PUVA (treatment with psoralens and
UVA radiation) as an efficient method.

14.6 PDT in Its Early Days

Between 1903 and 1905, von Tappeiner made the very first attempts for the treat-
ment of tumors and diseases of skin such as condylomata of female genitalia and
lupus. Many dyes were tried by them such as fluorescein, eosin, “Grubler’s
Magdalene red,” and sodium dichloroanthracene disulfonate. Results were favor-
able, but due to the advent of cancer therapy by the advent of ionizing radiations,
PDT soon lost its value.
Various tests are being conducted on PDT today for the application in the field of
oncology – for the treatment of cancers of various organs of human body.

14.7 Timeline

• In 1900, Oscar Raab explained the combinatorial cytotoxic effects of light and
acridine on a species of Paramecium.
• Niels Finsen treated cutaneous tuberculosis and smallpox with the use of light in
1901.
• Niels Finsen won the Nobel Prize in 1903 for his work on phototherapy. During
the same year, A. Jesionek and Herman von Tappeiner tried treating tumors on
skin by using topical eosin and white light.
• The term “photodynamic” was introduced by von Tappeiner and A. Jodlbauer in
1907.
162 S. Agnihotry et al.

• W. Hausmann in 1911 used mice skin to explain the photosensitive effects of


hematoporphyrin.
• In 1913, Friedrich Meyer–Betz was the first to perform the use of porphyrins in
photodynamic therapy (PD on humans; hematoporphyrin was applied on his own
hands).
• Samuel Schwartz performed acetylation and reduction of hematoporphyrin to
develop hematoporphyrin derivative (HPD); the phototoxic potential of HPD
was twice as compared to hematoporphyrin.
• Baldes and Richard Lipson explained accumulation of HPD in tumors, and its
use in the tumor photodetection was evolved in 1960.
• I. Diamond in 1972 described hematoporphyrin phototoxicity against gliomas by
in vivo and in vitro methods.
• Thomas Dougherty for the first time successfully cured skin cancer using PDT
during 1975. During the same year, HPD was used by J.F. Kelly for the treatment
of bladder cancer in humans. Tumor regression derivative (HPD) was also
observed by reduction and acetylation in concentration of hematoporphyrin.
• Dougherty in 1978 was the pioneer of first clinical study by PDT on humans.
• The first PDT drug was approved in Canada in 1999.

14.8 Photosensitizers in PDT

Until 1990, interest in PDT for the use in dermatology was little, but with the advent
of newer techniques of topical application of photosensitizers, its popularity rose.
5-Aminolevulinic acid (ALA) and its derivative methyl aminolevulinate (MAL)
were used followed by broad-spectrum red radiation which provided an easy and
efficient method. Along with these two drugs, few other synthetic, second-­generation
sensitizing drugs were being developed (derived from porphycenes, phthalocya-
nines, benzoporphyrins, and chlorines).

14.9 Systemic Photosensitizers

In clinical studies of PDT, hematoporphyrin (Hp) and hematoporphyrin derivative


(HpD) were the first systemically used photosensitizers [2]. Various commercial
products like Photosan, Photocan, Photofrin, etc. are manufactured after manipula-
tion and purification of hematoporphyrin derivatives. These photosensitizers are
basically a combination of monomers, dimers, and oligomers obtained from struc-
tural manipulation of hematoporphyrin. Photofrin is FDA approved in the USA for
esophageal obstructing lesions and Barret’s esophagus as well as for early and late
esobronchial lesions [3]. This drug is also approved worldwide for the treatment of
bladder cancer and other cutaneous lesions including basal cell, squamous cell, and
Kaposi sarcoma [4–6].
14 Role of Photodynamic Therapy in Cancer Treatment 163

14.10 Topical Photosensitizers

Approval of PDT use along with topical ALA was given by the US FDA to treat
actinic keratosis since 1999. Many studies related to successful acne treatment and
photorejuvenation have been reported, although FDA is yet to issue approval for its
use. MAL, an esterified ALA derivative, is also lipophilic, and it has high selectivity
for neoplastic cells when compared to ALA. Till date, many countries of Asia,
Europe, and America have already approved MAL for the treatment of actinic kera-
toses, basal cell carcinoma, and Bowen’s disease. MAL, in 2009, got its approval
for treatment of Bowen’s disease in Brazil. The protocol for the combinatorial use
of MAL with PDT along with red light has already been widely used.

14.11 Light Sources

In topical PDT, many different sources of light may be applied. Porphyrin absorbs
maximum light close to 405 nm, also known as Soret band. Q bands are other lower
peaks of absorption found at 510, 545, 580, and 630 nm. Light wavelengths between
625 and 633 nm are used in most of the clinical studies which allow greater skin
penetration. Broad-spectrum lamps, diode lamps, lasers, and optical fibers are the
major groups of light sources being used currently in PDT.

• Lamps – ARC lamps are used as broad-spectrum light sources which are easy
and cheap to operate. But there is difficulty in coupling them to light delivery
fibers without reduction in their optical power. Calculation of the light dose
which has been effective is also difficult. Power output is limited to 1 W. Moreover,
filters are essential for cutting off infrared emission and ultraviolet rays.
• Lasers – These emit precise wavelength of light in a focused beam. Lasers devel-
oped during early years were bulky, costly, and immobile machines that needed
technical support.
• Diode lamps – Systems made of semiconductor diode technology are compact,
portable, and cheaper, and they also retain high power output. Diode lasers, how-
ever, offer single output wavelength which makes its versatility limited. LEDs
are cheaper when compared to other light sources. They are smaller and give a
maximum power output of 150 mW/cm2 at 350–1100 nm range of wavelength.
• Optical fibers – This technology meets the demand of illumination for different
localizations. Illuminating at superficial areas like at oral mucosa, lens tips are
used with optic fibers for spreading the light over the specific area. Cylindrical
diffusers combined with inflated balloons are used in hollow organs like endo-
bronchial, esophagus, and bladder for uniform light distribution. Balloon at one
side is sometimes coated black for shielding of adjacent normal tissue areas.
164 S. Agnihotry et al.

14.12 Mechanism of Action

Experiments with light and combinations of reagents produced the modern version
of photodynamic therapy. PDT comprises two components that are nontoxic, and
they are used in an oxygen-dependent manner to induce tissue and cellular effects.
Photosensitizer is the first component of PDT which is a photosensitive molecule
that localizes to a target cell and/or tissue. Administration of light of a specific
wavelength involves the second component that activates the sensitizer. Reactive
oxygen species (ROS) is produced by energy transfer from light to molecular oxy-
gen. These reactions occur in a light-absorbing photosensitizer. Therefore, light-­
exposed tissues of particular areas only are responsible for activating the biological
responses to the photosensitizer. DNA photoaddition, which is an oxygen-­
independent photochemical reaction, has also been evolved. These reactions are
termed as photochemotherapy. Treatment of psoriasis and vitiligo and enhancement
of immunotherapy have been done by a photochemotherapeutic “psoralens,” in
combination with ultraviolet A.

The chemical is injected The chemical concentrates


into the body. at the tumor site.

LIGHT

The chemical is The tumor is selectively


activated by the light. destroyed.
14 Role of Photodynamic Therapy in Cancer Treatment 165

Administration of the photosensitizer can take place by topical application on the


skin or by intravenous injection which gives an advantage to PDT. However,
biodistribution is affected by these. With time-dependent change in biodistribution,
the effects of PDT can also be regulated with the period of light exposure. After
light absorption, photosensitizer moves from its ground or singlet state into a triplet
state via an excited singlet state that is short lived. This excited triplet state can
undergo reactions of basically two types. First, it can directly react with a substrate,
such as a molecule or the cell membrane, and transfer a hydrogen atom for radical
formation. These radicals through type I reaction convert into oxygenated products.
Alternatively, there may be formation of singlet oxygen through type II reaction
after the triplet state transfers its energy directly to oxygen. Anoxic areas of tissue
are not affected due to photosensitization as most of the PDT drugs are oxygen-­
dependent. In vivo studies have demonstrated that PDT effects of porphyrins were
abolished by induction of tissue hypoxia.

Type I reaction occurs side by side with type II reactions, and the ratio between
these two reactions depends on the concentrations of oxygen and substrate, the type
of sensitizer used, and the binding affinity of the sensitizer to substrate. Due to the
short half-life of the ROS and its high reactivity, only cells that are in close proxim-
ity of ROS production are affected by photodynamic therapy. The half-life of singlet
oxygen in biological systems is less than 0.04 μs, and thus the radius of its action is
less than 0.02 μm [7]. The extent of cytotoxicity and photodamage depends on the
type of sensitizer, its intracellular and extracellular localization, light fluence rate,
oxygen availability, the total light exposure dose, the total dose administered, and
the time between the administration of the drug and exposure of light.
It is already known that there are mainly three mechanisms by which tumor is
destroyed by PDT. In the first case, there is direct killing of tumor cells by ROS
generated by PDT. In the second case, PDT leads to tumor infarction as a result of
tumor-associated vasculature. Finally, an immune response can be activated by PDT
against tumor cells. The abovementioned mechanisms may be interconnected and
thus can influence each other too. The importance of each component relative to
each other for the total tumor response is not clearly understood till date. However,
it is understood that for long-term control of tumor, the combination of all these
components is more effective.

14.13 Clinical Applications of PDT

Canada was the first country to give approval for PDT usage in 1993, for the pro-
phylactic treatment of bladder cancer using the photosensitizer Photofrin. It is a
mixture of mono-, di-, and oligomers of partially purified HPD that contains the
porphyrin unit and is the most commonly used out of all photosensitizer.
5-Aminolevulinic acid and 5-ALA have been approved for treating skin basal cell
carcinoma and actinic keratosis, by topical application followed by red or blue light
exposures.
166 S. Agnihotry et al.

PDT finds its application in dentistry for the treatment of malignant transforma-
tion of oral lesions, treatment of premalignant and malignant oral lesions, and che-
motherapy (PACT) of bacterial and fungal infections. The popular application of
PDT includes age-related muscular degeneration and treatment of many eye dis-
eases. In dermatology PDT is being used for the treatment of acne, scleroderma, and
psoriasis. PDT is also being tested for the treatment of arthritis. Photoimmunotherapy
involves the use of PDT in combination with immunotherapy to enhance the immu-
nostimulating response for the treatment of metastatic cancer.

14.14 Oncogenes

Malignant neoplasia also known as CANCER is considered as a fatal disease which


is caused by the mutations in genes like microRNA genes, oncogenes, and tumor
suppressor genes. The first oncogenes were discovered through the study of retrovi-
ruses, RNA tumor viruses whose genomes are reverse-transcribed into DNA in
infected animal cells [8]. Mutations involved in cancer could be of somatic mutation
or acquired mutation (caused by environmental toxins and radiations exposure,
being not related to reproductive cell; it is not considered as hereditary mutation) or
sporadic mutation. Studies on Burkitt’s lymphoma provided the first evidence that
somatic genetic alterations give rise to cancer where one of the three different trans-
locations causes an oncogene MYC present on chromosome 8q24 to be juxtaposed
on one of the loci for immunoglobulin genes [9]. Second, transfection experiments
have been shown in mouse fibroblasts. DNA transforming activity was traced to a
human homologue of the retroviral RAS oncogene which bears mutations that
makes active the RAS oncogenic protein’s transforming property [10].

14.15 Signal Transduction of Oncogenes

In the normal case of cell division, the protein code for the proto-oncogenes passes
signal toward nucleus in a series of transduction cascade for cell division to occur
(Fig. 14.1). In this pathway signal molecule membrane receptor, cytoplasm signal car-
rying intermediary proteins and cell division activating transcription factors are
included. In this cascade mechanism, more than one protein can be activated by factors.
For example, a mutation in transcription factor proto-oncogene (a family of normal
genes) MYC and RAS changed into an oncogene. In RAS mutation the signaling path-
way remains “on” and leads to cell growth that is uncontrolled. RAS mutation results in
about 30% of tumors which includes thyroid, lung, pancreatic, and colon carcinomas.

14.16 Inflammation and Oncogenes

Inflammation is host-generated response to trauma. It can also be defined as mecha-


nism of defense against non-biological xenobiotic agents and involves a chain of
protective events that mainly leads to pain, swelling, redness, and temperature
14 Role of Photodynamic Therapy in Cancer Treatment 167

Growth Factor

RAS
Tyrosine (G protein)
kinase receptor

Protein kinases

Nucleus

Transcription
DNA factor
Protein that
stimulates cell cycle
Gene expression

Fig. 14.1 Signal transduction of RAS cell division pathway (the growth factor binds outside cell
to the tyrosine kinase receptor which further activates membrane proteins than transcription factor
activation occurs which moves on to the nucleus and ultimately the process results to the gene
expression)

which increases with time until the tissue repair and healing of wound or defense
mechanism is completed. Inflammation mainly involves neutrophils, macrophages,
monocytes, and mast and dendritic cells to invade at the infection site that eventu-
ally leads to destruction of the agent, affect cells or organism, and eventual restora-
tion of complete repair processes [11].
Two recognized oncogenes – myc and ras – are frequently linked to tumor-­
inflammation axis. It has been reported that the ras oncogene through CXCL8
induction is involved in tumor angiogenesis. Bar Sagi and Sparman validated ras
oncogene-dependent induction of the chemokine CXCL8 and tumor angiogenesis
[12], whereas remodeling of angiogenesis and tumor stroma are being related to
myc oncogene [12].The Myc oncogene regulates cell growth with the help of three
168 S. Agnihotry et al.

RNA polymerases – RNA polymerase I, II, and III [13]. Arf, a tumor suppressor
gene, is involved in p53 pathway for the role of protection of mammalian organ-
isms from aging. Low expression levels of this gene are normally found, but when
oncogenes are introduced into normal cells, Arf transcription is highly induced.
The role of Arf/p53 pathway in aging and cancer has thus been discussed by
Matheu et al. [14].

14.17 Proto-oncogene to Oncogene

Some different mechanisms for the conversion of proto-oncogenes to active


oncogenes:

1. Overexpression of proto-oncogene by gain of a novel transcriptional promoter


2. Amplification of the proto-oncogene or oncogene leading to overexpression
3. Effects on transcription level and the amount of gene product
4. Proximity of the oncogene and domains of immunoglobulin, subsequent chro-
mosomal translocations, that appears to result in deregulation of the gene
5. The oncogene protein structure alteration

14.18 Oncogenes: Functional Properties

The proteins encoded by oncogenes control both apoptosis and proliferation of cell.
The oncogenes associated with the tumor enhance the genomic and chromosomal
instability and unscheduled proliferation [15]. The initiation events of tumor pro-
gression are mutation and translocation, and later on amplification occurs during
progression. The classified groups of oncogenic products are chromatin remodelers,
growth factors, apoptosis regulators, growth factor receptors, signal transducers,
and transcription factors [16].

1. Transcription factors: these are multigene family member having common struc-
tural domains. Some of the transcription factors like the Fos transcription protein
form the AP1 transcription factor on dimerization and result in increased expres-
sion of genes that commands cell division [17].
2. Chromatin remodelers: these helps in transcriptional regulation, expression, and
repair of genomic segregation mechanism [18]. Chromatin remodeler enzymes
alter the chromatin structure. They are of two classes:
(a) Enzymes alter the structure of chromatin by histones covalent modi-
fication [19].
(b) ATP-dependent enzymes [20], by using the ATPase along the DNA move-
ment of nucleosomes, occur that ejects histones of DNA and enhances the
exchange of histones that ultimately changes the structure of nucleosomes.
The complete process is energy-dependent process.
Both enzymes regulate the accessibility of packaged DNA.
14 Role of Photodynamic Therapy in Cancer Treatment 169

Fig. 14.2 Nucleosome repositioning (Bizouarn et al. 2014)

Alterations in the DNA methylation, chromatin remodeling, and histone modifi-


cations result in the oncogenesis.
Some oncogenes that targets the chromatin remodeler like ∆Np63α, a skin onco-
gene that promotes squamous cell carcinoma [21] and also suggested that the Lsh-­
mediated chromatin remodeling are important for the process. It is responsible for
many other tumorous growths in the organs like lungs, neck, and head (Fig. 14.2).

3. Growth factors: potential oncogenes may be the cellular genes encoded by pro-
teins involved in the cell response to stimulation by growth factors. Some of the
major growth factors are platelet-derived growth factor (PDGF), epidermal
growth factor (EGF), and brain-derived growth factor (BDGF). Platelet-derived
growth factors (PDGFs) regulate diverse cellular functions in connective tissue
cells and are important for normal embryonic development [22].

The oncogene of simian sarcoma virus, i.e., sis oncogene, is structurally similar
to the gene coding for PDGF β chain [23] (Fig. 14.3).

4. Growth factor receptors


The mutations in the growth factors receptors lead to the continuous activation
of the pathways that ultimately leads to the uncontrolled cell division. The mem-
170 S. Agnihotry et al.

Fig. 14.3 Autocrine growth stimulation of simian sarcoma virus and v-sis PGDF-type growth
factor (The processes are 1. transcription, 2. translation, 3. dimerisation, 4. secretion, 5. receptor
binding, 6. signal transduction, 7. increased transcription)

bers of receptor tyrosine kinase family are mainly involved in the carcinogenesis.
Regulation of cell division, differentiation, apoptosis, and migration by the EGFR
family helps in growth and maintenance of some tissues. Mutations in the regula-
tion of signaling in this system result in growth and development of cancers.
The main epidermal growth factor receptors are EFGR, HER2, HER3, and HER4.
Out of that four EFGR and HER2 are most familiar oncogenes for cancer related to
gastroenterology [24]. In many of the lung and colorectal cancer, the EFGR level is
upregulated [25]. Tyrosine kinase inhibitors gefitinib or erlotinib and receptor
blockers panitumumab or cetuximab act as preventive measures. These agents are
effective in reducing uncontrolled tumor proliferation and cell cycling only if the
mutation should not be in the downstream signaling phosphatases and kinases.
Some reported known [26] oncogenes such as KRAS G12/13 and BRAF V600E
that are constitutively activated making these measures fail to show effect (Fig. 14.4).

5. Signal transducers and apoptosis regulators


The oncogene is related to the Src family of protein tyrosine kinases; it is
derived from proto-oncogene, c-Srca. It belongs to family of kinases that regu-
lates signal transduction in cellular environment by surface receptors. This Src
family of protein kinases enhances the signaling from growth factor receptors. It
promotes modulation of actin cytoskeleton rearrangements by regulating turn-
over of cell surface receptors thus promoting survival and cell motility. FAM83
proteins belong to oncogene family used for the cancer therapies by inhibiting
the MAPK signaling.
14 Role of Photodynamic Therapy in Cancer Treatment 171

Fig. 14.4 Cytoplasmic Extracellular domain


tyrosine kinase domain. (1. Cysteine-rich domain
EGFR, 2. IGFR, 3. FGFR, Ig-like domain
4. PGFR, 5. VEGR,
respectively. On mutation
these factors may cause
carcinogenesis)
Cell membrane

cytosol

1 2 3 4 5

14.19 Gain of Function (Fig. 14.5)

Fig. 14.5 Two genes Gene 1 Gene 2


together on combination
produces hybrid
hyperactive gene
Fusion gene

Fusion transcript

Fusion protein

Cell proliferation
Differentiation Mobility
Apoptosis

Cancer cell
172 S. Agnihotry et al.

17-5p 18a 19a 20a 19b-1


miR-17-92

c-Myc

E2F1 5’ 3’
ORF 3’UTR

Fig. 14.6 E2F1c-Myc-regulated microRNA polycistron miR-17–92 modulation of cell cycle


transcription factor and its function in cancer pathogenesis [28]. The upward part signifies cluster
of microRNA polycistron miR-17–92. Large and small boxes are miRNA precursors and mature
miRNAs, respectively. Transcription of both E2F1and miR-17-92 is promoted by c-Myc on bind-
ing in sites CACGTG or CARGTG. By directly binding miR-20a and miR-17-5pat the 3′UTR of
E2F1 mRNAs through translational repression, they negatively regulate E2F1 gene expression.
Expression levels through the ARF-p53 pathway result in the suppression of apoptosis which leads
to cancer pathogenesis in various organs. miR-17–92 acts as an oncogene in this regulatory mecha-
nism [29]

14.20 MicroRNA as Oncogenes

Loss of apoptosis function along with abnormal growth exhibited by cells usually
results in formation of cancer. Role of miRNA in the regulation of apoptosis and cell
growth have been recently indicated in several studies [13]. Observed study stated
that overexpression of miRNA let-7 in A549 cell lines is suppressed after cancer cell
growth [27]. Expression of RAS and MYC is regulated negatively via let-7 by tar-
geting their mRNAs for translation repression [20]. Along with p53, both RAS and
MYC have been involved, as key oncogenes in lung cancer where at their 3′ UTR
they have many complementary sites to let-7 [20], and also showed decreased levels
of let-7 and significantly higher levels of RAS protein in lung tumor tissues suggest-
ing role RAS regulation by let-7 in lung oncogenesis. In human testicular germ cell
tumors, miR-372 and miR-373 have been reported to work as oncogenes [15]
(Fig. 14.6).

14.21 Oncogenes Target: Cancer Treatment

In most of the cancerous cases, oncogenes act as a target gene so that antibiotics
may be designed by targeting that specified gene (Table 14.1).
14 Role of Photodynamic Therapy in Cancer Treatment 173

Table 14.1 Oncogenic protein targets


Anticancer drug Diseases Targeta
Monoclonal antibodies
Cetuximab Colorectal cancer EGFR
Bevacizumab Cell lung cancer, colorectal cancer VEGF
Trastuzumab Breast cancer ERBB2
Small molecules
Rafenib Renal cell carcinoma PDGFR, VEGFR,
FLT3
Gefitinib Cell lung cancer EGFR
Imatinib Chordoma, chronic myelogenous leukemia, PDGFR,ABL, KIT
gastrointestinal stromal tumors,
Sunitinib Renal cell carcinoma, gastrointestinal stromal PDGFR,VEGFR,
tumors, FLT3
Erlotinib EGFR Cell lung cancer
The New England Journal of Medicine. Copyright © 2008 Massachusetts Medical Society
a
FLT3 FMS-like tyrosine kinase 3, EGFR epidermal growth factor receptor, VEGF vascular endo-
thelial growth factor, PDGFR platelet-derived growth factor receptor

14.22 Oncogenes in Human Cancer

Normal genes help in regulation and proliferation, mutation in that initiates uncon-
trolled cell division that results to carcinoma. Myc enhances the cancer and p53
suppresses the cancer and both are pleiotropic transcription factors. Myc plays
important role in oncogenes signaling. Any mutational inactivation of Myc function
in tumors is driven by oncogenically activated Myc that enhances their fast regres-
sion by both extracellular and intracellular mechanisms, like apoptosis vascular col-
lapse and differentiation. Myc mutations comparatively to others are uncommon,
and aberrant upstream signals result in its formation of Myc in most tumors. Myc,
acting as a factor of upstream oncogenes, is also acts as a therapeutic target depends
how much it contributes to carcinogenesis. On the other side, p53 inactivated by self
only in some tumors, and in most of the cases, mutations inactivate upstream activa-
tors or downstream effectors of p53. These changes summarize the therapeutic util-
ity of p53 restoration in any tumor type.

14.23 Ras Oncogenes in Human Cancer

Active oncogenes are formed by mutations in codon 12, 13, or 61 of one of the three
ras genes, H-ras, k-ras, and N-ras. It was established that the activation of RAS
proteins is done by binding with the GTP. The evidence for the upstream regulators
of RAS has come into knowledge by the genetic studies on the CDC25 protein in
yeast [30]. In most of the human melanomas and also in other tumors, the
174 S. Agnihotry et al.

Fig. 14.7 Ras oncogene


GDP
mutation (oncogene
mutation inhibits active GT
Ras-GTP from converting So
to inactive Ras-GDP that RAS-GDP
results to permanent Ras-GTP
activation of GTP active
form and also continuous Inactive
activation of MAP kinase Active
cascade that leads to Mutation block
cancer) GTPase activity

identification of BRAF oncogenes clears the occurrence of the RAF effectors path-
way in a significant number of human malignancies [31].
Most specific gene for carcinomas is mutation in K-ras gene. The activation of
oncogenic Ras was done by mutation that inhibits the GTP hydrolization that medi-
ates Ras proteins to remain in GTP-bounded active form that become uncontrollable
cell proliferation (Fig. 14.7).

14.24 The Myc Oncogene

This nuclear protein encoded by proto-oncogene acts as a cell proliferating signal


protein which is involved in many carcinomas by its overexpression or excess
amount of presence that makes this protein unable to halt the cells to proliferate.
This mutant nature of protein is formed by chromosomal translocation mechanism.
In this mechanism the large amount of Myc mRNA produced by the strong pro-
moter binds just to the Myc coding sequence (Fig. 14.8).

14.25 Bcl-2 Proteins

For the cause of the lymphoid cancer, common reported translocation is between
chromosome 14 and 18. The antiapoptotic protein Bcl-2 activation done by the
translocation mechanism increased the cells survival that overexpresses the Bcl-2
that ultimately results to the inhibition of the programmed cell death.

14.26 Cyclin D

The overexpression of cyclin D results to cancerous growth. The continuous expres-


sion of this factor is enhanced by the chromosomal translocation process. The
amplification of cyclin D gene occurs like high numbers of cyclin D genes cause
increased level of Cdk4-cyclin D complex that inhibits the Rb gene and then E2F
14 Role of Photodynamic Therapy in Cancer Treatment 175

a Neoplasia
MYC on
Differentiative state: permissive

Epigenetic programme:
cellular proliferation Cell cycle Differentiation Apoptosis

b Normal
MYC off
Differentiative state: non-permissive

?
Epigenetic programme:
differentiation Cell cycle Differentiation Apoptosis

c Apoptosis
MYC off/on
Differentiative state: non-permissive

Epigenetic programme:
apoptosis Cell cycle Differentiation Apoptosis

Fig. 14.8 Complimentary ways of tumorigenesis. (a) MYC activates cell cycle and blocks dif-
ferentiation and apoptosis, (b) MYC inactivation blocks cell cycle and increased the level of dif-
ferentiation, and apoptosis not in all cases. (c) MYC reactivation inhibits cell cycle and
differentiation and induces the apoptosis (W. Felsher Nature Reviews Cancer (2003))

activated that leads to Cyclin E transcription and later on Cdk2-cyclin E complex


activation and finally the cell proliferation stats that result in carcinomas [14].

14.27 Conclusion

The term cancer defines variety of diseases. The main cause of cancer is the changes
in the cellular genome affecting the expression of genes that manages the cell
growth and differentiation. The carcinogenesis follows three major steps like (i)
initiation, reversible change; (ii) promotion, self-activation forms uncontrolled divi-
sion of cells; and (iii) progression, metastatic growth.
Most oncogenes act by copying normal growth signaling by one of the following
mechanisms.

A. Normal mitogenic growth factors use heterotypic, but several cancer cells adopt
the function of their own growth factor synthesis and forms autocrine stimula-
tion. For example, glioblastomas and sarcomas synthesize platelet-derived
growth factor and tumor growth factor α, respectively.
176 S. Agnihotry et al.

B. Cell surface receptors like EGFR and HER2/neu may be structurally altered or
overexpressed, which may lead to signaling that is ligand-independent.
C. Downstream targets involved in the signaling pathway can also be altered. For
example, mutated Ras is found in approximately 25% of human tumors that
leads to activation of the Ras-Raf-MAPK signaling pathway in a ligand-­
independent manner [14].

Mutations like gain of function and loss of function form uncontrolled cell pro-
liferation and suppression of apoptosis, respectively. Oncogenes which are respon-
sible or involved in the process of carcinogenesis are being considered as a target for
the development of anticancer drugs for the treatment of cancer. Multiple targets
could also be involved in the cancerous case, so they may also be in light during
drug development. The presence of microRNA in the cancerous growth steps like
initiation and progression acts as target for therapy.
It has been reported in many literatures that chances of getting cancer increase
exponentially with age. Immune system of body also relates with the cancer phe-
nomenon which includes the involvement of the cytokines and the chemokines. The
cancer results from many cascades, from the first stage of inflammation to the aging
and later to the hypoxia which leads to the cancer involving many factors that medi-
ate or activate aggressiveness of cancer, angiogenesis and migration, etc.
Along with tumor suppressor genes and oncogenes, many extrinsic factors are
also involved in the cancer mechanism; some of these are genetic predisposition,
environmental pollutants, alcohol, tobacco, etc.

References
1. Stern RS (2010) Prevalence of a history of skin cancer in 2007: results of an incidence-based
model. Arch Dermatol 146(3):279–282
2. Calzavara-Pinton PG, Venturini M, Sala R (2007) Photodynamic therapy: update 2006. Part 1:
photochemistry and photobiology. J Eur Acad Dermatol Venereol 21:293–302
3. Dougherty TJ, Gomer CJ, Henderson BW et al (1998) Photodynamic therapy. J Natl Cancer
Inst 90(12):889–905
4. Allison RR, Mang TS, Wilson BD (1998) Photodynamic therapy for the treatment of nonmela-
nomatous cutaneous malignancies. Semin Cutan Med Surg 17(2):153–163
5. Bernstein ZP, Wilson BD, Oseroff AR et al (1999) Photofrin photodynamic therapy for treat-
ment of AIDS-related cutaneous Kaposi’s sarcoma. AIDS 13(13):1697–1704
6. Allison R, Mang T, Hewson G et al (2001) Photodynamic therapy for chest wall progression
from breast carcinoma is an underutilized treatment modality. Cancer 91(1):1–8
7. Nielsen TO, Friis-Hansen L, Poulsen SS, Federspiel B, Sorensen BS (2014) Expression of the
EGF family in gastric cancer: downregulation of HER4 and its activating ligand NRG4. PLoS
One 9(4):e94606
8. Lindahl P, Johansson BR, Leveen P, Betsholtz C (1997) Science 277:242–245
9. Broek D et al (1987) The S. cerevisiae CDC25 gene product regulates the RAS/adenylate
cyclase pathway. Cell 48:789–799
10. Land H, Parada LF, Weinberg RA (1983) Cellular oncogenes and multi-step carcinogenesis.
Science 222:771–778
14 Role of Photodynamic Therapy in Cancer Treatment 177

11. Parsons SJ, Parsons JT (2004) Src family kinases, key regulators of signal transduction.
Oncogene 23:7906–7909. https://doi.org/10.1038/sj.onc.120816
12. Cheng LC, Tavazoie M, Doetsch F (2005) Stem cells: from epigenetics to microRNAs. Neuron
46:363–367
13. O’Donnell KA, Wentzel EA, Zeller KI, Dang CV, Mendell JT (2005) c-Myc-regulated microR-
NAs modulate E2F1 expression. Nature 435:839–843
14. Borrello MG, Degl’innocenti D, Pierotti MA (2008) Inflammation and cancer: the oncogene-­
driven connection. Cancer Lett 267(2):262–270 [PubMed: 18502035]
15. Deindl S, Hwang WL, Hota SK, Blosser TR, Prasad P, Bartholomew B, Zhuang X (2013)
ISWI remodelers slide nucleosomes with coordinated multi-base-pair entry steps and single-­
base-­pair exit steps. Cell 152:442–452
16. Keyes WM, Pecoraro M, Aranda V, Lindahl EV, Li W, Vogel H, Guo X, Garcia EL, Michurina
VT, Enikolopov G, Muthuswamy KS, Mills AA (2011) Np63a is an oncogene that targets
chromatin remodeler Lsh to drive skin stem cell proliferation and tumorigenesis. Cell Stem
Cell 8:164–176
17. Jenuwein T, Allis CD (2001) Translating the histone code. Science 293:1074–1080
18. Davies H et al (2002) Mutations of the BRAF gene in human cancer. Nature 417:949–954
19. Rushdi A, Nishikura K, Erikson J, Watt R, Rovera G, Croce CM (1983) Differential expres-
sion of the translocated and the untranslocated c-myc oncogene in Burkitt lymphoma. Science
222:390–393
20. Lusser A, Kadonaga JT (2003) Chromatin remodeling by ATP dependent molecular machines.
Bioessays 25:1192–1200
21. Dueñas CV, Camarero IR, Cobaleda C, García IS (2013) Function of oncogenes in cancer
development: a changing paradigm. https://doi.org/10.1038/emboj.2013.97
22. Heldin CH, Westermark B (1999) Mechanism of action and in vivo role of platelet derived
growth factor. Physiol Rev 79:1283–1316
23. Shaulian E, Karin M (2001) AP-1 in cell proliferation and survival. Oncogene 20:2390–2400
24. Croce CM (2008) Oncogenes and cancer. N Engl J Med 358:502–511
25. Bizouarn F (2014) Clinical applications using digital PCR. Methods Mol Biol 1160:189–214.
https://doi.org/10.1007/978-1-4939-0733-5_16
26. Takamizawa J, Konish H, Yanagisawa K, Tomida S, Osada H, Endoh H, Harano T, Yatabe
Y, Nagino M, Nimura Y, Mitsudomi T, Takahashi T (2004) Reduced expression of the let-7
microRNAs in human lung cancers in association with shortened postoperative survival.
Cancer Res 64:3753–3756
27. Sparmann A, Bar-Sagi D (2004) Ras-induced interleukin-8 expression plays a critical role in
tumor growth and angiogenesis. Cancer Cell 6(5):447–458 [PubMed: 15542429]
28. Oskarsson T, Trumpp A (2005) The Myc trilogy: lord of RNA polymerases. Nat Cell Biol
7:215–217
29. Grosjean P, Wagnieres G, Fontolliet C (1998) Clinical photodynamic therapy for superficial
cancer in the oesophagus and the bronchi: 514 nm compared with 630 nm light irradiation after
sensitization with photofrin II. Br J Cancer 77(11):1989–1995
30. Varmus H (1988) Retroviruses. Science 240:1427–1435
31. Fischle W, Wang Y, Allis CD (2003) Histone and chromatin cross-talk. Curr Opin Cell Biol
15:172–183 (36, 63, 142)

You might also like