Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

36 

CHA PTER 2
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

Molecular Genetics in Paediatric Dermatology


Anna C. Thomas1 & Veronica A. Kinsler1,2
1
 Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
2
 Paediatric Dermatology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK

Introduction, 36 Which samples to take for genetic Concept of personalized medicine, 39


Genetics and inheritance in brief, 36 testing, 39 Molecular genetics techniques, 40
Consent for genetic testing and incidental How to interpret genetic results, 39
findings, 38 What to do with a genetic result, 39

Abstract volume of genetic data generation, and have made the transition
from the research sphere to the diagnostic laboratories. The iden-
tification of disease‐causing genetic mutations in both inherited
Molecular genetics is the study of the structure of genes at molecu-
and sporadic paediatric dermatological conditions has begun to
lar level, and the impact of gene expression and regulation on the
influence clinical management, allowing preimplantation genetic
biology of the organism. The interplay between these two fields,
diagnosis, stratification for targeted personalized medical therapies,
genetics and biology, is a burgeoning area in all disease. In recent
or gene therapy in some cases. This chapter reviews the essential
years the process of disease gene identification and clinical genetic
terminology, key techniques and important advances needed to
diagnostic reporting has been revolutionized by the technologies
update paediatric dermatologists in this field, forming a basis for
discussed in this chapter, most prominently next‐generation
the detailed disease‐specific chapters that follow.
­sequencing (NGS). These techniques have increased the speed and

­Introduction confirm a diagnosis or to subclassify disease, the


i­dentification of disease‐causing genetic mutations in
Molecular genetics is the study of the structure of genes at both inherited and sporadic paediatric dermatologi-
a molecular level, and the impact of gene expression and cal conditions has begun to be clinically relevant in
regulation on the biology of the organism. The interplay clinical management. Preimplantation genetic diagno-
between these two fields, genetics and biology, is a bur- sis (PGD) as part of the in vitro fertilization (IVF) pro-
geoning area in all disease. For essential terminology in cess can be offered for future pregnancies where there is
molecular genetics, see Table 2.1. a family history and the genetic defect is known, for
A large number of skin conditions have now had their example in epidermolysis bullosa and the autosomal
genetic aetiology identified and this information can be recessive ichthyoses. In other instances the genetic
easily accessed through many of the online curated defect will stratify patients towards targeted medical
databases available, for example Online Mendelian therapies, such as in the vascular overgrowth disorders.
Inheritance in Man (www.omim.org) and the Decipher In addition, paediatric dermatology is now in the era
database (https://decipher.sanger.ac.uk), as well as from of personalized genetic therapy, and this will surely
published literature. be an area of tremendous growth in the decade to come.
In recent years, the process of disease gene identifica- Techniques of gene therapy will be discussed in
tion and clinical genetic diagnostic reporting has been Chapter 170.
revolutionized by the technologies discussed in this chap-
ter, most prominently next‐generation sequencing (NGS).
These techniques have increased the speed and volume of
genetic data generation, and have made the transition
­Genetics and inheritance in brief
from the research sphere to the diagnostic laboratories. The human genome is comprised of deoxyribonucleic acid
Storage and analysis of this data however, is still fraught (DNA) neatly contained within 23 pairs of chromosomes
with difficulties, not least in the interpretation of novel in the human, one of each pair inherited maternally and
(undescribed) variants. In addition, functional analysis of one paternally. The sex chromosomes are inherited XX for
the implications of any novel findings still requires vali- a female and XY for a male, and the other 22 pairs are
dation by cell biology and animal model generation. described as autosomes. It should also be noted that cel-
The implications of genetic diagnosis have changed lular mitochondria house 37 genes within a circular
in recent years. Whereas this used to serve only to genome that is inherited maternally. DNA is transcribed

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 2  Molecular Genetics in Paediatric Dermatology 37

Table 2.1  Essential terminology in molecular genetics

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Allele The alternative form of a gene’s sequence
Allele‐specific A test or treatment directed at only one allele
Array An ordered arrangement of probes on a solid surface
Base/nucleotide A DNA or RNA nucleotide (ACTG or ACUG)
Biallelic mutations Two different mutations at the same site in a gene
Chimaera An organism resulting from the fusion of two or more zygotes
Compound heterozygous mutations Two different mutations at different sites in a gene
Copy number Number of copies of a DNA sequence, usually at chromosomal or gene level but can be applied to
repeats in the DNA sequence
Copy number variation A change in the copy number of a portion of DNA, which is described in the normal population and
therefore considered to be a variant rather than pathogenic
De novo mutation Not inherited from either parent, but arisen for the first time in the patient
Digenic mutation A condition which is caused by mutations in two different genes
Exons The parts of the DNA sequence that code for proteins
Frameshift mutation A mutation that causes a shift in the reading frame of the DNA sequence
Gain‐of‐function/activating mutation A mutation that increases the function of the gene
Germline mutation A mutation that affects all the cells of an organism including the gametes
Hemizygous mutation Having only a single copy of a gene, which results in monoallelic expression
Heterozygous mutation The occurrence of a mutation in one copy of a gene but not in the other
Homozygous mutation The occurrence of the same mutation in both copies of a gene
Hybridization Binding of DNA or RNA to complementary strands or probes
Imprinting Natural silencing of one allele, leading to monoallelic expression
Introns The parts of the DNA sequence of the genome that do not code for proteins
In silico The analysis of the effects of a mutation using bioinformatics rather than laboratory experiments
Knockdown The deliberate reduction in expression of a gene (e.g. in an experiment to see what it does)
Knockout The deliberate obliteration of expression of a gene (e.g. in a mouse model)
Ligation Joining together of fragments of DNA
Loss‐of‐function mutation A mutation that reduces or obliterates the function of a gene
Missense mutation A single base change in the DNA sequence that alters an amino acid in the protein but does not
truncate it
Monoallelic expression Expression of only one allele instead of two. This occurs with X inactivation and with imprinted genes
Mosaicism The presence of two or more genotypes in an organism arising from a single zygote
Mutant or minor allele frequency The proportion of a sample or a population affected by a mutation
Mutation A change in the DNA sequence that is thought or known to be pathogenic. Non‐synonymous
mutations are those that alter the amino acid code in an exon whereas synonymous mutations do
not
Nonsense mutation A mutation that truncates the protein product
Novel A mutation that has not previously been described in the literature or in public databases
Oligonucleotide A short synthetic sequence of DNA or RNA bases
Overexpression Increased expression of a gene, usually deliberately to analyse the effects
Panel/targeted panel A selected set of genes for DNA sequencing
Polymorphism A change in the sequence of DNA that is commoner than 1% frequency in a population, and
therefore considered to be a variant rather than pathogenic
Post‐zygotic A mutation that occurs in utero
Segregation The co‐occurrence of a phenotype in members of a family who are affected by a particular genotype,
and the absence of that phenotype in unaffected family members
Sequencing Elucidation of the sequence of the bases in DNA or RNA
Single nucleotide polymorphism (SNP) or variant A single base pair change in DNA that is seen in more than 1% of a normal population
(SNV)
Somatic mutation A mutation within a tissue
Splice site A sequence of DNA that indicates the junction of an exon and intron
Sporadic A disorder that does not appear to be inherited or passed on
Transcription The process of making RNA from a DNA template
Translation The process of making protein from an RNA template
Uniparental disomy (UPD) The inheritance of two copies of a gene or genes from one parent
Validation The use of secondary or functional tests to confirm a DNA mutation

into messenger ribonucleic acid (mRNA) and mRNA is The genetic code of DNA occurs in the form of four
translated into protein. Proteins are the molecules in each chemicals or ‘bases’, namely cytosine (C), thymine (T),
cell that perform the vital functional tasks required in the guanine (G) and adenine (A). Complementary pairing
human body. It is the chemical properties of the amino occurs between C bases and G via three hydrogen bonds,
acids and the exact order in which they are aligned that and between T with A via two hydrogen bonds, at the
causes the protein to fold into a particular shape, and centre of the famous DNA double helix [1,2]. Long runs of
therefore determines its functional properties. these four bases are grouped into regions known as exons
38 Section 1  Development, Structure and Physiology of the Skin

and introns. Exons are known to be the regions that ‘code’ or disrupted, and on a larger scale there can be chromo-
STRUCTURE AND PHYSIOLOGY

for RNA and therefore for protein products, and they somal structural aberrations such as translocations and
SECTION 1: DEVELOPMENT,

make up only around 1% of the whole DNA sequence. inversions.


Introns make up the rest of the sequence and their func- In many cases a disease manifests itself because the
tion is still not completely understood. Early on in the flow of molecular information from DNA to RNA to pro-
study of molecular genetics they were considered to be tein contains a mutation, usually originating from the
‘junk DNA’, however it is now well established that DNA code. Many diseases can be described as ‘dominant’
intronic sequences are crucial for the functioning and con- or ‘recessive’ in inheritance pattern, where either one or
trol of the exonic areas and their protein products. The both copies of the gene are required to be faulty respec-
four bases of the exons and their flanking sequences are tively in order to cause the resultant disease. As males
‘transcribed’ into RNA by the process of transcription. In have one copy of the X chromosome, all genes in males
this process an RNA molecule is made from the DNA that do not lie on the pseudoautosomal regions of the X/Y
sequence transcript, with the intronic sequences removed, chromosomes cannot be described as heterozygous or
and the thymine base of DNA replaced by the uracil base homozygous and in this situation are referred to as
of RNA. The four RNA molecule bases (UCGA) can be ‘hemizygous’. X‐linked disorders in general are therefore
arranged into groups of three in 64 different ways, called more likely to be severe in males as there is only one copy
codons. Sixty one of these codons code for one of 20 amino of the gene.
acids, and three code for a so‐called stop codon. This is A disease is described as monogenic if there is only one
known as the genetic code, and the excess of codons for gene involved in the phenotype; however this is a rapidly‐
amino acids is known as the redundancy or degeneracy of disappearing concept, as the background genotype of the
the genetic code. Amino acids are the building blocks patient will almost inevitably have some phenotype‐
of proteins, and stop codons tell the protein when the modifying effect in any disease situation. Polygenic disor-
exonic instructions are finished. The process of forming a ders are those in which more than one gene is known to
string of amino acids from the RNA code is known as be involved in the phenotype. Mosaic disorders are dealt
translation. with in Section 23.
DNA mutations can occur anywhere in the human
genome, and in some cases lead directly to or contribute ­References
to disease. Broadly speaking the most damaging muta- 1 Watson JD, Crick FH. Molecular structure of nucleic acids; a structure
for deoxyribose nucleic acid. Nature 1953;171:737–8.
tions are found within exons, however they are increas-
2 Watson JD, Crick FH. Genetical implications of the structure of deox-
ingly being discovered outside coding regions, for yribonucleic acid. Nature 1953;171:964–7.
example in important regulatory regions outside the
gene itself such as the promoter. Mutations can be clas-
­ onsent for genetic testing and
C
sified into (i) point mutations, similar to a spelling mis-
incidental findings
take where a single base is replaced by another, and (ii)
insertions or deletions, where part of the DNA message Individual hospitals usually have their own standards
is missing or a part is added. Point mutations can be and protocols for obtaining consent for genetic testing.
subdivided into synonymous or non‐synonymous and, This will vary depending on the method to be used, and
where non‐synonymous, into missense or nonsense. whether the test is a diagnostic laboratory test or being
Synonymous mutations are base changes that do not done on a research basis. Oral consent for genetic testing
change the amino acid which is coded for and therefore may be sufficient for established diagnostic tests, but any
the protein product remains the same. These are usually research testing should have fully informed written con-
but not always benign, and therefore usually without sent recorded in the hospital and research notes.
apparent functional consequence. Non‐synonymous In most diagnostic tests the issue of incidental find-
mutations are those that alter the amino acid code in an ings does not arise as only a single gene is being tested.
exon. Within this group missense mutations are those However, with the increase in gene panel testing and of
that change the amino acid but not to a stop codon, clinical exome testing there may be genes being tested
producing a protein, although this protein is abnormal which are known to affect clinical outcome in other
in sequence. Nonsense mutations on the other hand ­diseases, such as tumour suppressor genes or onco-
lead to an absent or truncated protein product by chang- genes. In these cases, there is no international consen-
ing the codon to a stop codon. For mutations that are sus as yet for whether specific consent needs to be
deletions or insertions of sequence this can be ′in‐frame’ taken in advance, however, a gradual consensus is
or leading to a ′frameshift’. In‐frame insertions or dele- emerging, and in general it is good practice to inform
tions lead to one codon being changed, either to a dif- families that there is a possibility that other mutations
ferent codon or a stop codon, and frameshift alter the may be uncovered which could affect their health in
reading frame of the gene – in other words the three‐ other areas. Ideally there is a specific option after
letter codons are no longer read in the correct groups of informed consent for the patient/family to choose
three, which leads to a nonsense protein product and whether they would like to be told of incidental find-
sometimes to a premature stop codon. Large deletions, ings. Recent US and European publications in this area
duplications and insertions can also occur at gene level, may help with consideration of issues around consent
causing whole exons or multiple exons to be removed for incidental findings [1–6].
Chapter 2  Molecular Genetics in Paediatric Dermatology 39

­References result as reported should be inserted into the patient’s


1 Evans BJ. Minimizing liability risks under the ACMG recommenda-

STRUCTURE AND PHYSIOLOGY


medical notes, as the exact mutation may be extremely
tions for reporting incidental findings in clinical exome and genome

SECTION 1: DEVELOPMENT,
sequencing. Genet Med 2013;15:915–20. important. Reading the results of a DNA sequence muta-
2 Green RC, Berg JS, Grody WW et al. ACMG recommendations for tion is explained in Table 2.2.
reporting of incidental findings in clinical exome and genome
sequencing. Genet Med 2013;15:565–74.
3 Hehir‐Kwa JY, Claustres M, Hastings RJ et al. Towards a European ­What to do with a genetic result
consensus for reporting incidental findings during clinical NGS
testing. Eur J Hum Genet 2015;23:1601–6. In general terms it is best to refer the family to Clinical
4 May T. On the justifiability of ACMG recommendations for report- Genetics for the result to be given. Even if the paediatric
ing of incidental findings in clinical exome and genome sequencing.
J Law Med Ethics 2015;43:134–42. dermatologist feels confident of their knowledge in the
5 Anderson JA, Hayeems RZ, Shuman C et al. Predictive genetic testing area, they are not trained in genetic counselling and may
for adult‐onset disorders in minors: a critical analysis of the arguments not be aware of all the implications for all family mem-
for and against the 2013 ACMG guidelines. Clin Genet 2015;87:301–10.
6 Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of
bers. In referral of the family, mention that the whole fam-
secondary findings in clinical exome and genome sequencing, 2016 ily may need to attend, and certainly both parents where
update (ACMG SF v2.0): a policy statement of the American College of possible, and warn that further testing may be required.
Medical Genetics and Genomics. Genet Med 2017;19:249–55. Clinical geneticists can also deal with the issues surround-
ing incidental findings where relevant.
­ hich samples to take for genetic
W
testing ­Concept of personalized medicine
Blood DNA A rapidly evolving area of science is that of personalized
Methods of DNA extraction from whole blood can vary medicine, or precision medicine [1,2]. This is a broad term
dependent on the local laboratory, and it is always worth to describe the use of genetic data on an individual to tai-
checking before obtaining a sample. Very commonly lor the treatment of a disorder to that individual. There
1–5 mL is sufficient from children, collected into an EDTA‐ are two principal ways in which this research is being
containing vial. The sample is safe overnight at room tem- driven. First, genetic information is being analysed retro-
perature if need be. spectively from cohorts of patients who have had success-
ful or unsuccessful responses to therapy to create a model
Cheek swab or saliva DNA of genotypic association with outcomes including side‐
Increasingly it is possible to get good‐quality DNA from effects. Second, where disorders are found to be caused
a cheek swab, however this will also depend on the local by or driven by a specific mutation, researchers and clini-
laboratory. These samples require a specialized swab (not cians are prescribing therapies targeted to these mutations
a normal skin swab), and instructions for collection or to the known effects of these mutations. In paediatric
should be followed. Generally lower quantities of DNA dermatology this is currently being used in trials such as
are obtainable than from blood. The method is, however, of AKT1 inhibitors in Proteus syndrome, rapamycin in the
particularly suitable for young babies if it is difficult to PIK3CA‐related overgrowth spectrum, and collagen VII
obtain a blood sample, or from family members who are
not able to attend clinic to have blood taken, when a cheek Table 2.2  Terminology used in genetic testing results
swab or saliva sample can be sent remotely by following
the manufacturer’s instructions. _ (Underscore) is a range (e.g. c.76_78delACT)
> Indicates a substitution at DNA level (e.g. c.76A>T)
Skin DNA c. Position of base pair in cDNA
Skin biopsy is required for DNA extraction where it is c.(83G=/83G>C) Describes the two genotypes of a mosaic case
del Indicates a deletion (e.g. c.76delA)
possible that the mutation is not present in the germline
dup Indicates a duplication (e.g. c.76dupA)
and therefore not detectable in blood but only in affected fs Frameshift
tissue. A skin biopsy for DNA extraction must not be put fs*# *# Indicates at which codon position the new
into formalin. It can be transported fresh to the laboratory reading frame ends in a stop codon (*). The
on a saline‐soaked gauze, or snap‐frozen in liquid nitro- position of the stop in the new reading frame is
gen, or placed in a medium that stabilizes nucleic acids. calculated starting at the first amino acid that is
Most diagnostic laboratories when presented with a skin changed by the frameshift, and ending at the first
stop codon (*#)
sample for DNA extraction will culture fibroblasts from it
g. Position of base pair in genomic DNA
by default, and then extract DNA from the fibroblasts. This ins Indicates an insertion (e.g. c.76_77insG)
may not be appropriate in a mosaic disorder as the muta- inv Indicates an inversion (e.g. c.76_83inv)
tion may not be present in that cell type, and therefore DNA m. Position in mitochondrial DNA
extraction from whole skin should be specifically requested. n. Position in noncoding RNA
p. Position of amino acid in protein
p.(Arg97Profs*23) Frameshifting change with arginine‐97 as the first
­How to interpret genetic results affected amino acid, changing into a proline, and the
new reading frame ending in a stop at position 23
Once a clinical or research test has been ordered it is use- r. Position in RNA (e.g. r.76a>u)
ful to be able to understand the result. The full genetic
40 Section 1  Development, Structure and Physiology of the Skin

replacement therapy in recessive dystrophic epidermoly- of complementary sequence in the gene of interest.
STRUCTURE AND PHYSIOLOGY

sis bullosa. This personalized medicine approach will Polymerase chain reaction (PCR) is then performed to
SECTION 1: DEVELOPMENT,

change the face of medicine and is likely to improve amplify DNA fragments followed by dideoxynucleotide
patient outcome in response to drug therapies, reducing chain termination sequencing to obtain a sequencing
the requirement for multiple therapies and avoidable readout that can be compared to the known ‘wild‐type’
side‐effects. reference version. An example is shown in Fig. 2.1.

­References Next‐generation sequencing (NGS) of DNA [3]


1 Ashley EA. Towards precision medicine. Nat Rev Genet 2016;
NGS is the best way to sequence DNA on a large scale.
17:507–22.
2 Collins DC, Sundar R, Lim JS, Yap TA. Towards precision medicine
Sequencing that would previously have taken years
in the clinic: from biomarker discovery to novel therapeutics. Trends can now be done in around a week by hand or 3 days
Pharmacol Sci 2017;38:25–40. when using a robotic automated system. Over the years
there have been different technologies for NGS, and
there are still a wide variety of methods available.
­Molecular genetics techniques
However, the principles behind NGS are similar in all
Key reviews are referenced for further reading. cases, and there are also certain major categories of
experiment. These categories vary in how the DNA of
Karyotype interest is captured:
A karyotype test is used to determine chromosomal num- 1 Whole exome: all predicted coding regions of the genome
ber, structure and integrity. The test has to be performed are sequenced.
on actively dividing live cells, and therefore the speci- 2 Whole genome: all DNA regardless of its known function
mens need to be freshly delivered to the laboratory. is sequenced.
Karyotyping can be performed on blood or skin. Single 3 Targeted capture: only preselected DNA is enriched for
nucleotide polymorphism (SNP) arrays are increasingly and sequenced. This is often in the form of a set of
replacing karyotyping for the assessment of chromosomal known and/or candidate genes added to a screening
number and integrity (detection of duplications and dele- panel or it could be a region on a particular chromo-
tions); however if structural rearrangements are to be some. This is the most commonly used type within a
detected (for example inversions or translocations) then diagnostic context, where only known genes are being
karyotyping is still required. investigated. NGS panels are now validated in many
paediatric dermatology diseases [4–7].
Mutation identification by DNA sequencing There are a number of different protocols available
All methods of DNA sequencing rely on the existence of depending on the concentration of DNA collected, for
what is known as the reference sequence, which is the example a total concentration of 3 μg or as low as 200 ng.
accepted and curated version of the human genome. There This is extremely helpful if there are only small quantities
are various version of the reference sequence, and new of material to begin with. Recently adapted protocols
‘builds’ of these become available at intervals of a few have been optimized to obtain good‐quality sequencing
years, when the current version is updated with the latest data from DNA extracted from formalin‐fixed paraffin‐
data. The reference sequence can be found at various web- embedded (FFPE) blocks. This is advantageous as it
sites such as the University of California Santa Cruz means archived pathology samples can be analysed from
(UCSC) genome browser, https://genome.ucsc.edu. The skin samples (as well as other tissues). This has previ-
production of the reference sequence was made possible as ously been difficult as DNA extracted from FFPE blocks is
a result of the revolutionary Human Genome Project [1]. often of lower quality and often more fragmented than a
fresh sample.
Sanger sequencing How NGS works:
Sanger sequencing is a technique of sequencing DNA that 1 A ‘library’ is prepared by randomly fragmenting a
was discovered by Frederick Sanger and coworkers [2]. genomic DNA sample.
The method is to design numerous pairs of oligonucleo- 2 Ligation adapters are then added to both ends of each
tide DNA ‘primers’ that cover the coding region of the DNA fragment.
gene of interest. Primers are essentially small pieces of 3 Adapter‐ligated fragments are PCR amplified and
DNA of around 20 base pairs in length that bind to DNA subjected to purification.

#70 #80
G C T G C G G G T C C G C G T G C C C A C C A C
C G A C G C C C A G G C G C A C G G G T G G T G

#130 #130
Fig. 2.1  A typical Sanger sequencing trace after importing the data into an appropriate analysis software tool. Bases are represented in different colours.
Chapter 2  Molecular Genetics in Paediatric Dermatology 41

4 At this stage the library is loaded onto a flow cell so that 7 Data analysis is then carried out by sequence‐read

STRUCTURE AND PHYSIOLOGY


the DNA fragments can hybridize to the surface of the alignment to a selected reference genome with the use

SECTION 1: DEVELOPMENT,
flow cell via surface‐bound oligonucleotides that are of specialized software. After alignment, any differ-
complementary in sequence to the attached adapters. ences between patient samples and the reference
5 Each bound fragment goes through an amplification genome are identified (Fig. 2.2).
process to produce a cluster that becomes clonally If hunting for new genes on a research basis, the ideal
identical. scenario is to find gene variants in all/most of the affected
6 Sequencing reagents, including a differently labelled individuals that are not present in the reference genome
fluorescent nucleotide for each base, are used to iden- and other control populations and, if looking for an inher-
tify each base. ited disease, are present in the parental DNA where the

Library preparation Cluster amplification

Genomic DNA

Fragmentation
Row cell

Adapters
Bridge amplification
cycles
Ligation

Sequencing
library
2 4
1 3

Clusters
NGS library is prepared by fragmenting a gDNA sample and Library is loaded into a flow cell and the fragments hybridize
ligating specialized adapters to both fragment ends. to the flow cell surface. Each bound fragment is amplified into
a clonal cluster through bridge amplification.

(a) (b)

Sequencing Alignment & data anaylsis

A
T
C
2 4 G
1 3
ATGGCATTGCAATTTGACAT
TGGCATTGCAATTTG
AGATGGTATTG
Reads GATGGCATTGCAA
Sequencing cycles
GCATTGCAATTTGAC
ATGGCATTGCAATT
AGATGGCATTGCAATTTG
T A
G C
Reference
Digital image AGATGG TATTGCAATTTGACAT
genoma
Data is exported to an output file

Cluster 1 > Read 1: GAGT...


Cluster 2 > Read 2: TTGA...
Cluster 3 > Read 3: CTAG...
Cluster 4 > Read 4: ATAC... Text file

Sequencing reagents, including fluorescently labelled nucleo- Reads are aligned to a reference sequence with bioinformatics
tides, are added and the first base is incorporated. The flow software. After alignment, differences between the reference
cell is imaged and the emission from each cluster is recorded. genoma and the newly sequenced reads can be identified.
The emission wavelength and intensity are used to identify
the base. This cycle is repeated ”n” times to create a read
length of “n” bases.

(c) (d)
Fig. 2.2  Overview of the workflow of an Illumina next‐generation sequencing (NGS) method. Source: Courtesy of Illumina, Inc.
42 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

p15.31 p15.1 p14.1 p13.2 p12 q11.2 q12.2 q13.2 q14.1 q14.3 q15 q21.2 q22.2 q23.2 q31.1 q31.3 q33.1 q34 q35.2

41 bp
140,207,950 bp 140,207,960 bp 140,207,970 bp 140,207,980 bp

T
T
C

C
C

Typical representation of the output of NGS, with 'reads' represented as grey horizontal bands aligned with the reference
genome build using Integrative Genomics Viewer (Broad Institute). Each of these is the equivalent to one of the Sanger
sequencing traces shown in Figure 2.1. A heterozygous mutation is demonstrated by the blue cytosine change.

(e)
Fig. 2.2  (Continued)

inheritance can be explained. However, we also know Copy number changes


that diseases can be sporadic and mutations can appear Copy number mutations or copy number variations
de novo so this has to be considered during the analysis (CNVs) can be challenging to identify when using DNA
phase. sequencing methods, especially if they occur in heterozygous
At this point there is still a requirement for DNA varia- form. CNVs occur if a copy of all or part of a gene is either
tions within candidate genes to be validated by Sanger deleted or duplicated by one or more copies. There are a
sequencing. It is always important to ensure that any number of different methods by which to identify CNVs.
result is not due to an NGS artefact that has happened by
chance. Array techniques [8–14]
One preferred method of detection is array‐comparative
Other types of NGS genomic hybridization (aCGH). In simple terms aCGH
In the last few years NGS protocols have been adapted to has RNA oligonucleotides or ‘oligos’ spaced at specific
perform sequencing of RNA in a process known as RNA‐ distances along the human genome that bind DNA
seq. This has the huge advantage of being able to analyse complementary in sequence. In each case a patient DNA
the sequence of alternatively spliced mRNA transcripts. sample under investigation is labelled with a red fluo-
Splicing is the editing of an early mRNA into a mature rescent dye and a reference DNA sample is labelled
form, where introns (noncoding DNA) are removed and with a different dye which fluoresces green. Differentially
exons (protein‐coding parts of DNA) are joined together. labelled samples are mixed 50 : 50 and then hybridized
In this manner a single gene can give rise to different pro- to the array surface. After a time of hybridization, sam-
teins depending on the composition of the exons ligated ples can be analysed for CNVs. In areas of normal copy
together during the splicing stage. Different tissues of the number, equal binding of red and green labelled DNA
body can contain differentially spliced forms of the same is assumed. However, in cases of a patient deletion the
mRNA. RNA‐seq can capture this information and addi- software would detect more or all green fluorescence
tionally gather quantitative information on how highly a compared with red in a genomic location depending on
particular transcript is expressed at a given time point. the presence of a homozygous or heterozygous dele-
There are different types of RNA that have specialized tion. Conversely, in the case of a patient duplication
biological functions and, depending on the precise meth- there would be a higher detection of red fluorescence
ods used, small RNAs, microRNA (miRNA) and transfer than green, depending on the number of duplicated
RNA (tRNA) as well as total RNA can be analysed using copies (Fig. 2.3).
RNA‐seq. SNP DNA arrays can also be used to read copy number.
Other types of NGS include chromatin immunopre- These work by fragmentation of the patient DNA and
cipitation sequencing (CHIP‐seq), which extracts data labelling with fluorescent dye, and then hybridization
on protein interactions with DNA (DNA‐binding sites), onto a solid surface peppered with allele‐specific oligo-
and methylated DNA immunoprecipitation (MeDIP‐seq), nucleotide probes. These probes are allele‐specific at sites
which extracts data on DNA methylation. of known common variation in the population, and are
Chapter 2  Molecular Genetics in Paediatric Dermatology 43

Ratio plot 143.39 Mbp. 9q34.3, no genes, no CNVs


4.00

STRUCTURE AND PHYSIOLOGY


3.75

SECTION 1: DEVELOPMENT,
3.50
3.25
3.00
2.75
2.50
2.25
2.00
1.75
1.50
1.25
1.00
0.75
0.50
0.25
0.00
–0.25
–0.50
–0.75
–1.00
–1.25
–1.50
–1.75
–2.00
–2.25
–2.50
–2.75
–3.00
–3.25
–3.50
–3.75
–4.00
24.3
24.2
24.1

23
22.3
22.1
21.3
21.2
21.1
13.3
13.2
13.1
12
11.2
11.1

12

13
21.11
21.12
21.13
21.2
21.31
21.32
21.33
22.1
22.31
22.32
22.33
31.1
31.2
31.3
32
33.1
33.2
33.3
34.11
34.13
34.3
Fig. 2.3  Array comparative genomic hybridization ratio plot depicting a deletion of a large part of chromosome 9p (shown in red), and two small areas of
closely related deletion/duplication (red and green). The chromosomal bands are laid out at the bottom of the figure.

therefore able to give information not only on copy (cDNA). cDNA contains all the relevant information on
number but also on genotype at these loci, and on large expression level and is preferable to work with as it is
areas of homozygosity or hemizygosity. more stable than mRNA and therefore less likely to
degrade during the handling process.
Multiplex ligation‐dependent probe It is now possible to extract mRNA directly from blood
amplification [15] using specialized kits, however this is only useful if the
Often the results of aCGH require validation by another gene of interest is expressed in white blood cells and this
method; the gold standard here is multiplex ligation‐ is not always the case. Luckily for dermatologists, skin
dependent probe amplification (MLPA). There are com- can be easily biopsied. Different cell types such as fibro-
panies that provide validated and optimized panels with blasts and keratinocytes can be extracted from a fresh skin
probes annealing at many points along a certain gene. biopsy and then cultured to obtain a large number of cells
MLPA works on a multiplex PCR basis after specific to work with. Additionally, the skin biopsy as a whole can
probes have annealed to the designated DNA regions. be used to extract mRNA, resulting in mRNA from all the
Using specialized analysis software the designed probes cell populations in the skin in one sample.
can identify CNVs in the gene under investigation. A Two methods are used, differing in how the experiment
schematic showing the interpretation of MLPA results is technically works: an intercalated dye method such as
explained in Fig.  2.4. MLPA is the preferred method for SYBR® green, or a probe method such as TaqMan® where
diagnostic reporting where pathological CNVs are known a fluorescent reporter dye is analysed. Both obtain data
to be causative of a particular disease. on the basis that the amount of cDNA (equating to the
original relative amounts of mRNA) can be quantified as
Quantitative real‐time PCR [16] each round of PCR cycle causes the amount of cDNA to
Expression analysis of a particular gene can be extremely double during the exponential amplification phase. By
useful for validating involvement in a particular disease, comparing to the expression of a gene that is expressed to
and quantitative real‐time PCR (qRT‐PCR) is often used. the same level in all cells, known as a ‘housekeeping
Put simply, this works by obtaining mRNA and using the gene’, such as glyceraldehyde‐3‐phosphate dehydroge-
retroviral enzyme reverse transcriptase to convert RNA nase (GAPDH) or beta‐actin (ACTB), the expression level
back to DNA to a form known as complementary DNA of the gene of interest can be calculated. For example, if
44 Section 1  Development, Structure and Physiology of the Skin

2.5
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

1.5
Peak ratio

0.5

100 150 200 250


(a) Size (bps)

2.5

1.5 Fig. 2.4  Schematic representation of multiplex


Peak ratio

ligation‐dependent probe amplification (MLPA) data.


Blue squares on the right are reference probes showing
1 normal copy number over different chromosomes.
Squares on the left indicate probes for a particular
gene. (a) In this case the gene under investigation
0.5 shows probes in red and above the cut‐off for normal
copy number (depicted by the green lines). This
indicates a duplication of the whole gene. (b) Shows
0 partial deletion of the same gene as five probes are in
green and below the normal cut‐off, but the rest of the
100 150 200 250 gene is of normal copy number as shown by three blue
(b) Size (bps) squares within the lines.

the gene of interest has a significantly lower level of


expression or lacks detectable expression in patient sam-
ples compared to control samples, this can provide fur-
ther evidence that the mutation is causative of the disease
under investigation.

Fluorescence in situ hybridization [17]


Fluorescence in situ hybridization (FISH) is another
method that can be used for copy number variations of
genes or indeed whole chromosomes. This works via the
attachment of fluorescently labelled probes to a specified
chromosomal location in an individual cell where the
chromosomes can be visualized microscopically. Fig. 2.5
shows the presence of three copies of chromosome 21,
which is the karyotype of an individual with Down syn-
drome, in a cell during metaphase of cell division.

Genome‐wide association studies [18]


Sometimes it is extremely challenging to find specific
gene candidates associated with a disease. This is espe-
cially the case for disorders that are likely to be polygenic. Fig. 2.5  Fluorescence in situ hybridization showing three copies of
In these cases a genome‐wide association study (GWAS) chromosome 21 during metaphase as depicted by the red fluorescent probe
may be helpful. This works by analysing the genotype of in a single cell of an individual with Down syndrome. The two control green
a set of patients with a particular condition, for example probes are hybridized to chromosome 13. Chromosomes are stained in blue.
Chapter 2  Molecular Genetics in Paediatric Dermatology 45

psoriasis, compared to a control cohort without clinical creating double‐stranded RNA. Owing to a n ­atural

STRUCTURE AND PHYSIOLOGY


signs or a family history of psoriasis. By genotype in this mechanism within cells this double‐stranded RNA is then

SECTION 1: DEVELOPMENT,
example we are interested in single base variations at degraded by the cell. This therefore prevents translation
locations throughout the genome known as SNPs. If there of the RNA into protein, and effectively stops or reduces
is an association with a particular set of SNPs in the dis- expression of the protein. This molecular genetic tech-
ease cohort compared to the control cohort, this may give nique is therefore useful for studying the effects of
valuable information regarding areas of the genome that removal of the protein product from the cell, and by infer-
require further investigation. ence can help with working out what that gene usually
does. Alternatively, siRNA has been suggested to be used
Other commonly‐encountered techniques as therapy for diseases in paediatric dermatology where a
Polymerase chain reaction (PCR) mutation is leading to overexpression of a protein. This is
PCR is a technique whose discovery revolutionized therefore a type of gene therapy for skin disease and is
molecular biology [19,20], earning its inventor Kary being researched in paediatric dermatology conditions.
Mullis a Nobel Prize in 1993. It forms the basis of many
other techniques in molecular genetics. It exploits a class ­References
of enzymes known as DNA polymerases, which will copy 1 Lander ES, Linton LM, Birren B et  al. Initial sequencing and
double‐stranded DNA from single‐stranded DNA. The analysis of the human genome. Nature 2001;409:860–921.
function of PCR is to selectively amplify one area of the 2 Sanger F, Coulson AR. A rapid method for determining sequences
in DNA by primed synthesis with DNA polymerase. J Mol Biol
genome, such as the area carrying a mutation of interest, 1975;94:441–8.
so that large quantities of DNA just from that area are pro- 3 Goodwin S, McPherson JD, McCombie WR. Coming of age: ten
duced. Taq polymerase is combined with the DNA tem- years of next‐generation sequencing technologies. Nat Rev Genet
2016;17:333–51.
plate (from a patient), two specific oligonucleotide
4 Lai‐Cheong JE, McGrath JA. Next‐generation diagnostics for
primers which are designed by the researcher to flank inherited skin disorders. J Invest Dermatol 2011;131:1971–3.
either side of the area of interest, and other reagents 5 Takeichi T, Liu L, Fong K, et al. Whole‐exome sequencing improves
including base pairs for construction of new DNA. The mutation detection in a diagnostic epidermolysis bullosa laboratory.
Br J Dermatol 2015;172:94–100.
reaction involves multiple rounds of increasing the tem- 6 Bhoj EJ, Yu Z, Guan Q et al. Phenotypic predictors and final diagnoses
perature to around 96° C to cause double‐stranded tem- in patients referred for RASopathy testing by targeted next‐generation
plate DNA denaturation (separation of the two strands), sequencing. Genet Med 2017;19:715–8.
lowering of the temperature (variable) to allow the prim- 7 Scott CA, Plagnol V, Nitoiu D et al. Targeted sequence capture and
high‐throughput sequencing in the molecular diagnosis of ichthyosis
ers to stick to their relevant complementary strands, and and other skin diseases. J Invest Dermatol 2013;133:573–6.
increasing the temperature again to 72° C which is the 8 Rodriguez‐Revenga L, Mila M, Rosenberg C et  al. Structural
optimum temperature for Taq polymerase function. This variation in the human genome: the impact of copy number variants
on clinical diagnosis. Genet Med 2007;9:600–6.
process is usually repeated 35 times and the production of 9 Lee C, Iafrate AJ, Brothman AR. Copy number variations and
DNA proceeds exponentially. The DNA produced is clinical cytogenetic diagnosis of constitutional disorders. Nat Genet
known as the PCR product, and is used then in Sanger 2007;39:S48–S54.
sequencing, genotyping or cloning. 10 Carter NP. Methods and strategies for analyzing copy number

variation using DNA microarrays. Nat Genet 2007;39:S16–S21.
11 Shen Y, Wu BL. Microarray‐based genomic DNA profiling technol-
Methods for genotyping specific base ogies in clinical molecular diagnostics. Clin Chem 2009;55:659–69.
pair changes 12 Barnes MR. Genetic variation analysis for biomedical researchers: a
There are various methods used for genotyping specific primer. Methods Mol Biol 2010;628:1–20.
13 Dumanski JP, Piotrowski A. Structural genetic variation in the

base pair changes that avoid the need to sequence the ­context of somatic mosaicism. Methods Mol Biol 2012;838:249–72.
DNA. These rely on the difference in the sequence at the 14 Hall IM, Quinlan AR. Detection and interpretation of genomic
particular base pair in question, either by using a restric- structural variation in mammals. Methods Mol Biol 2012; 838:225–48.
tion enzyme which recognizes one of the alleles (wild‐ 15 Schouten JP, McElgunn CJ, Waaijer R et al. Relative quantification
of 40 nucleic acid sequences by multiplex ligation‐dependent probe
type or mutant), or by recognition of the differing amplification. Nucleic Acids Res 2002;30:e57.
properties of the two alleles during changes of tempera- 16 Mocellin S, Rossi CR, Pilati P et al. Quantitative real‐time PCR: a
ture. Examples of such genotyping methods are high‐ powerful ally in cancer research. Trends Mol Med 2003;9:189–95.
17 Trask BJ. Gene mapping by in situ hybridization. Curr Opin Genet
resolution melt PCR (HRM), restriction fragment length Dev 1991;1:82–7.
polymorphisms (RFLPs) and amplification‐refractory 18 Visscher PM, Brown MA, McCarthy MI, Yang J. Five years of GWAS
mutation system (ARMS). discovery. Am J Hum Genet 2012;90:7–24.
19 Mullis K, Faloona F, Scharf S et al. Specific enzymatic amplification
of DNA in vitro: the polymerase chain reaction. Cold Spring Harb
Small interfering RNA [21] Symp Quant Biol 1986;51:263–73.
Small interfering RNAd (siRNA) are synthetic RNA 20 Saiki RK, Gelfand DH, Stoffel S et  al. Primer‐directed enzymatic
­oligonucleotides which are designed to be complemen- amplification of DNA with a thermostable DNA polymerase.
Science 1988;239:487–91.
tary to the RNA transcript of a DNA sequence of interest. 21 Sarkies P, Miska EA. Small RNAs break out: the molecular cell
When the DNA is transcribed to RNA in the presence of the biology of mobile small RNAs. Nat Rev Mol Cell Biol 2014;15:
siRNA, the siRNA binds to the specific RNA transcript, 525–35.

You might also like