You are on page 1of 17

SECTION 1 OVERVIEW OF BASIC SCIENCE

3  Molecular Biology
Thomas N. Darling

INTRODUCTION Placing the sample in formalin (for light microscopy) or glutaraldehyde


(for electron microscopy) before processing and sectioning allows the
Advances in molecular biology are rapidly changing our understanding sample to be analyzed histologically in a variety of ways, including by
of skin biology and disease. Increased knowledge is being translated into routine staining, immunohistochemistry, or in situ hybridization. The
new molecular diagnostic tests that are transforming the clinical prac- advantage of this approach is that the cells are preserved in a very stable
tice of dermatology. Molecular analyses are currently being employed to fashion, with well-preserved architecture, for later analysis. Disadvan-
diagnose genodermatoses1, cutaneous infections2, melanomas3,4, lym- tages are that the cells are no longer living and the fixation procedure
phomas5, and inherited6 or autoimmune blistering disorders7. In order can place limits on the methods that can be used to analyze DNA,
to use these tests in a prudent manner, dermatologists nowadays have RNA, and protein from cells of interest. Nonetheless, PCR analysis of
an even greater need to understand basic concepts in molecular biology. DNA is increasingly being performed on formalin-fixed, paraffin-
This knowledge is also leading to the development of new targeted embedded tissue, especially for infectious diseases and detection of gene
therapies, many of which require determination of the molecular basis rearrangements in lymphoproliferative disorders.
for disease in order to appropriately implement treatment8. In addi- In order to preserve molecules in a more native state, the specimen
tion, molecular analysis is being used to screen for drug efficacy and may be snap frozen. Cryosections prepared from such samples have
susceptibility to adverse reactions to certain medications9. lower quality tissue architecture than do permanent sections from
The goal of this chapter is to outline basic concepts and methodolo- formalin-fixed, paraffin-embedded tissues, but this form of tissue pres-
gies of molecular biology and to give practicing dermatologists a fun- ervation may allow for better analysis of DNA, RNA, and protein. One
damental appreciation of what their colleagues are doing in the may also directly process fresh or frozen tissue with buffers and reagents
laboratory and how their own clinical practice is changing as molecular in order to extract DNA, RNA, and protein from the whole tissue. The
approaches are implemented into patient care. advantage of performing extractions from whole tissue is that the DNA,
RNA, and protein will be fresh and of high quality. The disadvantage
EXPERIMENTAL TECHNIQUES is that the extracts will not be from a pure population of cells.
Another approach is to culture cells obtained from fresh tissue. The
Tissue Processing desired lineage of cells (e.g. keratinocytes, fibroblasts, immunocytes,
Dermatologists are accustomed to placing a biopsy specimen in forma- endothelial cells) can be propagated in vitro by employing selective
lin followed by paraffin embedding and staining tissue sections with culture media and isolation techniques. Culturing cells allows one to
hematoxylin and eosin. However, for molecular analysis, this is just obtain more cells than in the original sample, and then the cells can
one of several possible starting points for tissue processing (Fig. 3.1). be exposed to various conditions. However, the culturing process may
change fundamental characteristics of the cells, so that they do not
accurately represent the cells in vivo.
TISSUE PROCESSING One method for isolating a pure population of cells from a tissue
section is to use laser microdissection (Fig. 3.2). A laser is used to

LASER CAPTURE MICRODISSECTION

Excised tissue Transfer cap


Tissue section
Transfer film
Glass slide
Fixed Frozen Fresh
Laser beam

Electron Light Tissue Flow


microscopy microscopy culture cytometry
and FACS

Laser
microdissection Transfer film

Glass slide
Extraction

Fig. 3.2 Laser capture microdissection. Laser capture microdissection is one


DNA RNA Protein method of microdissection used to selectively procure individual cells or
clusters of cells from tissue sections. A cap coated with a thermoplastic transfer
membrane is placed directly over the tissue section. The operator visually
Fig. 3.1 Tissue processing. A tissue sample can be processed in various ways
  identifies the cells of interest and triggers a low-energy infrared laser to melt
66 for the analysis of DNA, RNA, or protein. FACS, fluorescence-activated cell the transfer film onto cells of interest. The cap is then lifted from the section to
sorting. separate the selected cells from the remainder of the tissue section.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
non-print metadata
ABSTRACT KEYWORDS:
Advances in the methods used to analyze DNA, RNA, and proteins are flow cytometry,
transforming the practice of medicine and research. Traditional methods polymerase chain reaction,
CHAPTER
to assess individual genes or proteins are complemented by advanced PCR,
techniques allowing genomic or proteomic analysis of populations of
cells or individual cells. The functions of genes and encoded proteins
DNA sequencing,
reverse transcription PCR,
3
are determined in vivo using animal models that allow control of when RT-PCR,

Molecular Biology
and where genes are expressed. Molecular techniques enable gene-based fluorescence in situ hybridization,
therapies in which genes are corrected, supplemented, or suppressed FISH,
for therapeutic benefit. The future promise of precision medicine nucleic acid arrays,
depends on our ability to use and develop these techniques for molecu- proteome,
lar analysis. transgenic mice,
gene-based therapy

66.e1

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
retrieve or cut individual cells from a section on a slide while the
POLYMERASE CHAIN REACTION
specimen is viewed with a microscope10. An advantage of microdissec-
tion is that a very pure population of cells is obtained that has been Purpose
precisely identified under the microscope. One limitation is that the
total number of cells isolated is relatively low, as each cell has to be
• Amplify a specific piece of DNA from a complex mixture CHAPTER

individually captured by the laser. Consequently, the procedures to


extract DNA, RNA and protein from these pure (but few) cells have to
Requirements 3
• Need to know sequence of the DNA of interest (at least of its ends)
be robust.

Molecular Biology
A way to isolate and analyze cells in suspension is to use flow cytom- Underlying concepts
etry (Fig. 3.3)11. Cells are passed individually in a stream through a • Double-stranded DNA can be melted or unwound to single strands with
set of lasers and electronic detectors capable of measuring multiple increased temperature (Fig. 3.4A); when cooled, the single strands come
parameters for each cell. As an initial step, cells are typically incu- back together to form double strands (hybridize) if the nucleotide sequences
bated with fluorescent-labeled antibodies that recognize cell-surface are complementary*
markers so that a heterogeneous population of cells can be charac- • During the hybridization process, when two complementary strands bind to

terized on a cell-by-cell basis for levels of expression of each marker. each other, the A nucleotides on one strand bind to T nucleotides on the
Fluorescence-activated cell sorting (FACS), which utilizes an electro- complementary strand, whereas C nucleotides of one strand bind to G
static deflection system, can also be used to obtain pure populations nucleotides on the complementary strand, and vice versa
• In PCR, short DNA strands called oligonucleotide primers are designed for
of cells with specific desired characteristics. Flow cytometry permits
the measurement of multiple characteristics of individual cells at a hybridization to specific sequences in the template DNA
rapid rate. However, with the exception of peripheral blood or bone Outline of method (Fig. 3.4)
marrow cells, a significant limitation is that it requires the cells to be • Oligonucleotide primers are designed to hybridize to specific sequences at
in suspension. Recently, the ability to analyze 40 or more parameters
each end of the DNA of interest. These primers are added to a reaction
for each cell has been accomplished by combining the principles of vessel mixture containing the template DNA along with a thermostable DNA
flow cytometry and elemental mass spectrometry in what is call mass polymerase, the nucleotides dATP (A), dTTP (T), dGTP (G) and dCTP (C), and
cytometry11a. buffer
• The reaction vessel is placed in a thermal cycler, which controls the

temperature of the reaction through many cycles


The Foundations of Molecular Techniques for • Each cycle contains the following steps: (1) denaturation; (2) primer

Analyzing DNA, RNA, and Protein annealing or primer hybridization; (3) primer extension; and (4) repeat of the
complete cycle of PCR 30–40 times
The concepts behind molecular biology are simple and unifying. In
general, they consist of extracting the molecules of interest, amplifying Benefits
them to measurable amounts, and detecting them. Polymerase chain • PCR is simple and rapid
reaction (PCR) is a standard technique for amplifying DNA (Table 3.1; • Because the PCR product is exponentially increased, it is extremely sensitive
in amplifying low amounts of DNA. Each cycle increases the number of PCR
products twofold. The total number of PCR products after n cycles will be 2n
Limitations/errors
• Because of its high degree of sensitivity, laboratory contamination of a DNA
FLOW CYTOMETRY
sample by trace amounts of the PCR product can cause misleading results
• Primers used for PCR can anneal to sequences that are similar, but not

identical, to the sequence of interest. This can be countered with “hot start”
Positive (red) and negative
(clear) cells in suspension techniques (DNA polymerase prevented from acting until after first
that have been pre- denaturation step) or nested PCR (after the PCR amplification, repeat the
incubated with fluorescent- PCR amplification using a second set of primers that hybridize to sequences
labeled antibodies inside first set of primers). In nested PCR, the second set of primers will only
hybridize to correct PCR products resulting from the first PCR amplification
Direction of flow • DNA polymerase occasionally incorporates incorrect nucleotides. For

sequencing by PCR, polymerases that possess proofreading enzymatic


Lasers Detectors activity can be used. This also allows the generation of even longer PCR
products, up to ~50 kb long
Experimental applications
• DNA can be amplified either for detection of a specific sequence or for
  Electrostatic cloning that sequence
  deflection • PCR can be used to label DNA with fluorescent or radioactive nucleotides
  system (FACS) • PCR can be used for rapid haplotype analysis

Modifications/alternatives
• Quantitative PCR – the amount (number of copies) of a specific piece of
DNA can be quantified by a variety of methods utilizing PCR. A relatively simple
and high-throughput method, called real-time quantitative PCR, uses a
specially designed thermal cycler that measures the amount of PCR product
Sorted cells formed after each cycle (see Fig. 3.6B). The higher the level of DNA, the
earlier the product can be detected. This can be used to measure genetic
changes in cancer such as gene amplification, to quantify the amount of
residual cancer following treatment, or to quantify the amount of a pathogen
in a sample. Modifications of this procedure can allow discrimination of gene
Fig. 3.3 Flow cytometry. Cells in suspension, pre-incubated with fluorescent-

polymorphisms
labeled antibodies, flow single file in a liquid stream through lasers. Detectors • Southern blot, in situ hybridization, comparative genomic hybridization
measure the fluorescence intensities of each cell. In fluorescence-activated cell
sorting (FACS), the single cells are divided into charged droplets that are *Hybridization actually forms the basis of several techniques in molecular biology, as the two
separated into different tubes based on the marker(s) of interest as they pass strands can be DNA:DNA (PCR, Southern blotting), DNA:RNA (Northern blotting, in situ
through an electrostatic deflection system. Flow cytometry is commonly hybridization), or RNA:RNA.
employed in the evaluation of patients with cutaneous B- and T-cell Table 3.1 Polymerase chain reaction.
  67
lymphomas.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
POLYMERASE CHAIN REACTION DNA SEQUENCING

Purpose
SECTION $
• Determine the sequence or order of nucleotides (A, G, C, T) in a stretch
1 Double-strand DNA
of DNA
Requirements
Overview of Basic Science

• The piece of DNA to be sequenced can be either a PCR product or a


Step 1 denaturation
cloned piece of DNA present in a plasmid, but it should be pure
Underlying concepts
• Chain termination (Sanger sequencing), or a variation thereof, is a
standard method for DNA sequencing
• In chain termination, the extension of a new strand of DNA is stopped
Step 2 annealing by the addition of an analogue of dATP, dCTP, dGTP or dTTP (ddATP,
ddCTP, ddGTP or ddTTP, respectively) to the sequencing mixture.
When the DNA polymerase incorporates the analogue nucleotide
instead of the correct normal nucleotide, DNA synthesis is terminated
because the polymerase is no longer able to link to the next nucleotide
• Gel electrophoresis is used to separate the different sizes of DNA

Step 3 extension fragments that result from chain termination synthesis. The DNA
fragments are forced to travel through a gel using an electric current;
the smaller molecules are less impeded by the gel and travel faster than
larger molecules*
% Outline of method (Fig. 3.5)
Heat
• An oligonucleotide primer hybridizes to the DNA to be sequenced and
Heat
DNA polymerase synthesizes a second complementary strand
• The synthesis of the second strand is interrupted randomly by the

incorporation of the fluorescent nucleotide analogues (ddATP, ddGTP,


Heat
35 cycles ddCTP, ddTTP), and the DNA fragments containing this final nucleotide
235=34 analogue can be identified because each of the four ddNTPs is labeled
billion with a different color fluorochrome
Template copies • The different DNA fragments are electrophoresed through a
DNA
F+R polyacrylamide gel or capillary tubes
primers • The different-length DNA strands terminating with different

dNTP 1st cycle fluorochrome-labeled nucleotide analogues pass a fluorescence


polymerase 2 =2 copies
1
2nd cycle detector and indicate the order of the DNA sequence
22=4 copies
Benefits
Fig. 3.4 Polymerase chain reaction. A Each cycle contains the following steps:

• The fluorescent chain termination method is able to rapidly sequence
(1) denaturation – separate the two strands of DNA by heating to >90°C; (2) large amounts of DNA with automated analysis of results
primer annealing or primer hybridization – allow the oligonucleotide primers
to bind to the template DNA by cooling to 50–65°C; (3) primer extension Limitations/errors
– DNA polymerase catalyzes the addition of nucleotides (A, G, C, T) that are • Routinely sequence only ~500 bases per run
complementary to the DNA template, beginning with the primer and • Difficulty with G+C-rich regions
extending 3′ at the optimal temperature of 72°C; and (4) repeat the complete • DNA must be high-quality
cycle 30–40 times. B Each cycle increases the number of PCR products twofold.
The total number of PCR products after n cycles will be 2n times the original Experimental applications
amount. F+R, forward and reverse. • Determine previously unknown sequence
• Confirm sequence following the cloning of a DNA fragment of interest
and other manipulations
Modifications/alternatives
• Pyrosequencing
Fig. 3.4)12. The PCR-amplified DNA, typically 50 to 2000 base pairs • Next-generation sequencing (massively parallel sequencing; see Table
in size depending on the primers designed for a particular sequence, 54.6)
can be detected in a gel using an intercalating dye that fluoresces with *Gel electrophoresis is used in many molecular biologic techniques to separate DNA, RNA or
ultraviolet light. The nucleotide sequence can then be determined via protein molecules of differing sizes.
automated fluorescence sequencing techniques (Table 3.2; Fig. 3.5). Table 3.2 DNA sequencing.
This simple and relatively inexpensive approach is still widely used.

However, it is being supplanted by massively parallel sequencing, also


known as next-generation sequencing, in which millions of fragments
of DNA are sequenced in a single run (see Table 54.6)13.
RNA is also easy to purify, but it is much more readily degraded than translation and post-translational modifications that affect protein sta-
DNA. Therefore, a typical first step in the analysis of RNA is to convert bility. One method used to measure levels of protein is referred to as a
it into DNA using reverse transcription (RT; Table 3.3; Fig. 3.6A). Fol- Western blot (Table 3.4; Fig. 3.7); it is also known as an immunoblot
lowing RT, the complementary DNA (cDNA) can be amplified by PCR, because an antibody is employed to detect the protein of interest. In
as described above. The technique of RT-PCR has also been modified addition to measuring protein levels, Western blot analysis can deter-
to allow accurate quantitation of very low levels of mRNA14. Because mine the size of proteins and can reveal whether there are different
the amount of PCR product is monitored throughout each cycle of forms of the protein15. Another method commonly used to measure
amplification, this technique is referred to as “real-time” quantitative protein levels is an enzyme-linked immunosorbent assay (ELISA; see
PCR (Fig. 3.6B). Table 3.4)16. An ELISA can provide very exact quantitation of protein
68 The amount of protein is a complex balance of synthesis and degrada- levels and may be less expensive and easier to perform than a Western
tion controlled at multiple steps, including efficiency of protein blot.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
DNA SEQUENCING REVERSE TRANSCRIPTION PCR

Purpose
Fixed end CHAPTER
CACCGAATACATCTG • To amplify mRNA by PCR, the mRNA is first converted to DNA (called

A T C G
complementary DNA or cDNA), followed by PCR amplification of a
specific region of the cDNA to detectable levels
3

Molecular Biology
A reaction T reaction C reaction G reaction Requirements
dATP dATP dATP dATP
dTTP dTTP dTTP dTTP • Starting material can be total cellular RNA (including ribosomal, transfer
dCTP dCTP dCTP dCTP and messenger RNA [mRNA]) or purified mRNA
dGTP dGTP dGTP dGTP
ddATP ddTTP ddCTP ddGTP
Underlying concept

CA CACCGAAT C CACCG
• In order to facilitate studies of RNA, many techniques that study RNA
CACCGA CACCGAATACAT CAC CACCGAATACATCTG first convert the RNA to cDNA with an enzyme called reverse
CACCGAA CACCGAATACATCT CACC transcriptase, an RNA-dependent DNA polymerase
CACCGAATA CACCGAATAC
Outline of method (Fig. 3.6a)
CACCGAATACA CACCGAATACATC
• Reverse transcriptase can convert mRNA to cDNA by three different
methods, depending on the primer used for the initial RT step*.
3' G CACCGAATACATCTG (1) Random hexamer primers contain six nucleotides (6-mer) that have
T CACCGAATACATCT
CACCGAATACATCTG all possible sequence combinations of the dA, dG, dC and dT
C CACCGAATACATC
T CACCGAATACAT 80 390 nucleotides (46 possible combinations). These random hexamers will
A CACCGAATACA hybridize to the corresponding complementary sequences in the sample
C CACCGAATAC RNA. (2) Oligo dT primers contain only dT nucleotides and hybridize to
A CACCGAATA the complementary string of dA nucleotides that are present at the end
T CACCGAAT of mRNA molecules (poly A tail). (3) The third choice is a primer that will
A CACCGAA
A CACCGA only hybridize to a specific mRNA sequence
• After the mRNA has been converted to cDNA, primers that can
G CACCG
C CACC hybridize to specific sequences are added and PCR amplification is
C CAC performed as described in Table 3.1
A CA
5' C C Automated fluorescent
Benefits
Sequencing gel sequencing scan • As for PCR, RT-PCR is simple and rapid
• RT-PCR is extremely sensitive in detecting low levels of mRNA
Fig. 3.5 DNA sequencing. An oligonucleotide primer hybridizes to the DNA to
  transcripts
be sequenced and DNA polymerase synthesizes a second complementary
strand. The synthesis of the second strand is interrupted randomly by the Limitations/errors
incorporation of fluorescent nucleotide analogues (ddATP, ddGTP, ddCTP, • If the RNA sample is contaminated with DNA (that contains the gene of
ddTTP). The DNA fragments containing this final nucleotide analogue can be interest), a PCR product may be amplified from the DNA even though
identified because each of the four ddNTPs is labeled with a different color the corresponding mRNA for the gene is not present. To control for this,
fluorochrome. Gel electrophoresis is used to separate the different sizes of DNA RT-PCR of RNA samples can be done with and without reverse
fragments. The different-length DNA strands terminating with different transcriptase
fluorochrome-labeled nucleotide analogues pass a fluorescence detector and • RNA is fragile and the absence of a specific mRNA transcript may result
indicate the order of the DNA sequence (see Table 3.2 for more details). from RNA degradation during and following extraction. RNA quality can
be tested by gel electrophoresis and/or by RT-PCR of housekeeping
genes (genes expressed by all cells)
Experimental applications
Measuring Spatial Distribution Within Tissues • The mRNA gene transcripts can be amplified for subsequent cloning or
In populations of cells, PCR, RT-PCR, and Western blots are useful for sequencing
measuring DNA, RNA, and protein, respectively. However, in order to • The mRNA gene transcripts (instead of the gene) could be analyzed for

identify which cells are responsible for observed characteristics, particu- the presence of mutations
larly within complex tissues such as skin, tissue sections are analyzed Modifications/alternatives
by fluorescence in situ hybridization (FISH) for DNA or RNA and
immunohistochemistry for protein. FISH is based on hybridization (as • Quantitative RT-PCR – by adding a reverse transcription step to
in PCR), but it relies on the use of fluorescently labeled nucleic acid quantitative PCR (see Table 3.1), the levels of mRNA transcripts from
probes that are complementary to the desired DNA or RNA sequences genes of interest can be quantified.
• Correspondingly, samples that contain less mRNA transcripts will
to be detected in tissue sections (Table 3.5, Fig. 3.8). The probes are
require more PCR cycles before the exponential phase. Therefore,
hybridized to the target sequences in the tissue and then the slide is
different RNA samples can be precisely compared by measuring the
examined using fluorescence microscopy17.
number of PCR cycles (x axis) needed to produce a defined amount of
FISH can be used to detect DNA deletions or amplifications in cancer
PCR product (y axis) (Fig. 3.6B)
cells. Normally, two fluorescent signals appear in each nucleus for an • By carefully designing the primers, quantitative RT-PCR can be used to
autosomal gene, but in cancer cells there may be only one signal, indi- measure the amounts of alternatively spliced forms of a gene
cating a deletion, or three or more signals if there is amplification. An • Alternative methods to measure RNA levels include digital PCR, in
alternative to FISH is microarray-based comparative genomic hybrid- which the sample is partitioned into individual nucleic acids, and
ization (array CGH) (see Fig. 113.28). Whereas FISH typically only Northern blots
probes up to 4 genetic regions in one tissue section, array CGH tests
for copy number aberrations across the genome. A disadvantage of array *In addition to the RNA, the reaction mixture contains the reverse transcriptase enzyme, an
oligonucleotide primer, dNTPs, and buffer.
CGH, relative to FISH, is that array CGH assesses a population of cells
rather than individual nuclei. In other words, FISH may detect genetic Table 3.3 Reverse transcription PCR (RT-PCR).

abnormalities in a minority of cells that could be masked by normal


surrounding cells in array CGH. Array CGH and FISH may be used to
supplement routine histopathology in the analysis of ambiguous mela-
nocytic lesions, soft tissue tumors (see Fig. 117.19), and myelogenous 69
leukemia involving the skin3,4.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
REVERSE TRANSCRIPTION PCR WESTERN BLOT

Purpose
SECTION $ Reverse transcription
• Western analysis can measure the size and the amount of protein present in
1 a sample
Requirements
Overview of Basic Science

Gene-specific priming • Western analysis requires an antibody that is specific for the protein of
PCR interest (i.e. does not cross-react with other proteins)
AAAAA(A)n AAAAA(A)n
amplification Underlying concepts
RNA cDNA
Oligo d(T) priming
• Polyclonal antibodies that react to several epitopes on a protein antigen are
AAAAA(A)n
obtained by injecting a protein into an animal and later isolating the
antibodies from the serum immunoglobulin fraction
N6 N6 • Monoclonal antibodies that react to only one epitope of a protein antigen are

Random hexamer priming obtained by immunizing mice (or rats, rabbits, or chickens) with the antigen,
then fusing the animal’s reactive lymphocytes with an immortal myeloma cell
% Quantitative real-time PCR line to create cells that are able to provide antibodies indefinitely
• A secondary antibody is used to detect the protein or the primary antibodies

bound to the protein. These detection antibodies can be visualized by


Amplification attaching a fluorescent probe or by attaching an enzyme that can produce
1
either light or a color by enzymatic action on a substrate
Outline of method (Fig. 3.7)
Fluorescent intensity

0 • A solubilized protein mix is separated on a polyacrylamide gel and


transferred electrophoretically to a membrane. The membrane is then
soaked in a buffer containing the antibody. Bound antibody is detected by a
–1 chromogenic or chemiluminescent assay
Benefits
• Western analysis is a simple and sensitive method to detect and quantify
–2
proteins present in a complex mixture
• Western analysis can determine the molecular weight of a specific protein

0 2 4 6 8 10 12 14 16 18 20 22 24 26 28 30 32 34 36 38 40 relative to standard controls


Cycle Limitations/errors
• Proteins are subject to degradation during extraction. The use of protease
Fig. 3.6 Reverse transcription PCR (RT-PCR). A Reverse transcriptase can

inhibitors in the extraction buffer helps to prevent degradation
convert mRNA to cDNA in three different ways, depending on the primer used • To perform a Western analysis, one must possess a highly specific antibody
for the initial RT step: (1) random hexamer primers; (2) oligo dT primers; and (3) that can recognize denatured proteins
gene-specific primers (see Table 3.3 for more details). After the mRNA has been • Western blots may contain a high background of nonspecific staining. To
converted to cDNA, primers that can hybridize to specific sequences are added correct this, blocking agents such as bovine serum albumin or milk protein
and PCR amplification is performed as described in Table 3.1. B Real-time PCR
are used
is able to precisely measure the amount of PCR product continuously (y axis) • Large proteins transfer poorly from the gel to the membrane, and small
after each cycle (x axis). Each plotted line represents the amount of PCR
proteins may transfer through the membrane without binding. Adequate
product present in a different sample. In samples that initially contain more
mRNA gene transcripts, real-time PCR will demonstrate an exponential increase transfer to the membrane can be accomplished by controlling the duration
in PCR product earlier, after fewer PCR cycles. of the transfer procedure
Experimental applications
• Detection, quantification, and characterization of a specific protein
• Identification of antibody activity to a known antigen

Immunohistochemistry is used to assess the expression of proteins Modifications/alternatives


in tissue sections. An antibody is added to a tissue section where it • Dot blot – a drop of the protein mixture is placed on a paper membrane and
binds specifically to the protein of interest. The bound antibody is the protein of interest detected with antibodies, as in the Western blot. The
usually detected based on an enzymatic reaction that generates a disadvantage of this technique is that, owing to the elimination of size
colored product at the site of bound antibody (see Chapter 0). Immuno­ separation by gel electrophoresis, specific binding to the protein of correct
histochemistry can be used to determine the locations and levels of size cannot be distinguished from nonspecific background binding to proteins
expression of a protein within a tissue, including proteins that mark • Immunoprecipitation (IP) – the specific antibody is added to the protein

specific cell lineages or cell states such as differentiation, activation, mixture and the resulting antibody–protein complexes are then isolated.
proliferation, or apoptosis. Because proteins are not first denatured in immunoprecipitation, they can be
detected in a more native configuration
Measuring the Transcriptome and Proteome • IP–Western – protein–protein interactions can be studied by first

immunoprecipitating the protein with one antibody, bringing down a protein


There are several approaches for quantitatively analyzing the levels of complex. The protein complex is then separated on a polyacrylamide gel,
expression of nearly all cellular genes. For example, RNA-seq utilizes followed by Western blotting to detect members of the protein complex
massively parallel sequencing (see Ch. 54) to evaluate RNA samples; • Enzyme-linked immunosorbent assay (ELISA) – this is a sensitive and
it can analyze levels of gene expression, gene splicing, non-coding RNA, specific method for quantifying the amount of a protein. The protein of
microRNA, gene fusions, and mutations17a. Additional methods are interest is captured on a plate coated with a monoclonal antibody and other
based on the hybridization of labeled sample mRNA to known oligo- proteins are washed away. The protein is then detected using a second
nucleotide or cDNA sequences (representing thousands of genes) placed antibody that has been modified for detection using a colorimetric or
on chips, beads, or glass slides (Table 3.6; Fig. 3.9)18. The level of luminescent assay
hybridization to the nucleotide sequence of a given gene indicates how • Immunohistochemistry – this is used to visualize the cellular localization of a

much mRNA of that gene is present. In addition to using this method protein. An antibody to the protein of interest is applied to a tissue section.
to profile mRNAs (the transcriptome), modified approaches allow the The antibody is detected using a secondary antibody coupled to an enzyme
70
measurement of differences in microRNAs or changes in genomic DNA that reacts with a substrate to produce a colored precipitate (see Ch. 0)
Table 3.4 Western blot.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
WESTERN BLOT TECHNIQUE FLUORESCENCE IN SITU HYBRIDIZATION (FISH)

Purpose
Protein samples
• To visualize locations and levels of specific sequences of DNA or RNA CHAPTER

in tissue sections
Requirements
3

Molecular Biology
• Tissue section of formalin-fixed paraffin-embedded tissue or fresh-frozen
Gel electrophoresis Polyacrylamide
tissue
gel
• The target sequence must be known

Underlying concepts
• A fluorescently labeled nucleic-acid probe hybridizes to complementary
Direction of sequences in tissue sections

protein transfer • The signal is detected using fluorescence microscopy

Polyacrylamide Outline of method (Fig. 3.8)


gel
• A probe is designed that is complementary to the target sequence and
Transfer to Membrane is directly or indirectly labeled with a fluorochrome
membrane
• Nucleic acids in the tissue section are denatured and then the FISH
+ probe is applied to the tissue section to hybridize to the target
Primary antibody sequence
Secondary antibody * • Unbound probe is washed away

• Samples are viewed by fluorescence microscope

Benefits
• Able to identify which cells exhibit a specific abnormality in DNA, such
as gene deletion, amplification, or translocation
Detection Membrane • Able to determine which cells are expressing a particular gene

Limitations/errors
• Subject to interlaboratory and interobserver variability
• Technical issues may complicate results, such as incomplete
hybridization or nonspecific binding
Fig. 3.7 Western blot technique. A solubilized protein mix is separated on a

Experimental applications
polyacrylamide gel and transferred electrophoretically to a membrane. The
membrane is soaked in a buffer containing antibody. The bound antibody is • Identification of DNA copy number aberrations in cancer cells
detected by a chromogenic or chemiluminescent assay utilizing a labeled • Determining the spatial pattern of gene expression in a tissue
secondary antibody (green asterisk).
Modifications/alternatives
• Single molecule RNA FISH
• FISH of metaphase preparations to diagnose chromosomal
abnormalities
• Spectral karyotyping or multifluor FISH to detect chromosomal

translocations and large deletions or duplications


• PCR or RT-PCR following laser microdissection of pure cell
copy number. Changes in copy number can be determined by array population
CGH or single nucleotide polymorphism (SNP) arrays (see Figs 54.6 • Array-based comparative genomic hybridization (CGH) (see Ch. 113)

and 113.28)18a.
The proteome of a cell represents all the cellular proteins that are Table 3.5 Fluorescence in situ hybridization (FISH).

being expressed under a particular set of conditions. Technologies that


allow measurement of the proteome continue to be refined19. One
technique relies on mass spectrometry, which can provide extremely
accurate measurements of protein mass by ionizing the protein and
measuring the “time-of-flight” through a tube to a detector on the
opposite end (Table 3.7; Fig. 3.10). In most instances, the proteins
are separated prior to mass spectrometry, using two-dimensional gel and develops into a mouse (Table 3.8; Fig. 3.11). Although the gene
electrophoresis or capillary electrophoresis. This can be technically dif- is present in every cell, the expression of the gene can be limited or
ficult and time-consuming, so mass spectrometry analysis of proteins targeted to specific tissues or cell types by using a promoter/enhancer
continues to be optimized for high-throughput and quantitative analy- that is only expressed in a given cell. Promoters/enhancers are the
sis of proteins. Quantitation is accomplished by labeling proteins using portion of a gene – usually on the 5′ end of the gene or upstream
stable isotopes to provide internal standards, or by label-free shotgun of its coding region – that initiates and regulates the level of mRNA
proteomic approaches that are being enhanced for greater speed and expression. For example, the promoters/enhancers that would ensure
lower costs19. Another technique is the use of reverse-phase protein that the gene is only expressed in melanocytes would include the pro-
arrays, in which whole protein lysates are spotted onto slides and moters of genes involved in melanin biosynthesis, such as tyrosinase.
probed using antibodies to specific proteins20. The field of proteomics Although every mouse cell will contain a randomly integrated copy
promises to provide a tremendous amount of information regarding cell of this new gene, it will be expressed only in melanocytes because of
biology and disease states. the melanocyte-specific promoter/enhancer. To express a gene only in
keratinocytes, a gene construct would contain a promoter/enhancer
from a gene that is only expressed in keratinocytes, such as a keratin
Genetically Engineered Mouse Models promoter21.
Transgenic mice are commonly employed as experimental models for The functional loss of a gene can also be studied by creating a “knock-
elucidating in vivo functions of genes21,22. The gene to be studied is out” transgenic mouse (Table 3.9; Fig. 3.12)23. It is also possible to
microinjected into a fertilized mouse egg where it randomly integrates selectively knock out or delete a gene (e.g. a tumor suppressor gene) in 71
into the mouse genome, and is present in every cell as the egg divides a specific cell type or tissue as opposed to deleting the gene from all

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Fig. 3.8 Fluorescence in situ

FLUORESCENCE IN SITU HYBRIDIZATION (FISH) hybridization (FISH). A tissue


section on a microscope slide is
fixed and permeabilized. The
SECTION nucleotides on the section (yellow)

1 and the fluorescently labeled


(asterisks) probes (dark pink) are
denatured; then the probes are
Permeabilize
Overview of Basic Science

and Hybridize added to the section to hybridize


denature to probe * * * * with complementary sequences.
Unbound probe is washed away
* * * * and the tissue is examined by
Gene of fluorescence microscopy. Normally,
interest two fluorescent signals appear in
each nucleus for an autosomal
gene, but in cancer cells there may
be only one signal, indicating a
deletion, or three or more signals if
Fluorescence there is amplification. Here, a
microscopy normal nucleus contains two green
signals and two dark pink signals
for two separate autosomal genes
whereas a tissue section from an
Tissue section on microscope slide angiosarcoma that was formalin-
fixed and paraffin-embedded
shows multiple separate signals
using a MYC break-apart probe (the
probe consists of two flanking
sequences in which variable
breakpoints have been observed)
instead of only two copies of the
Angiosarcoma − Normal cell − two signals attached to one
tissue section (FFPE) interphase nucleus
another. FFPE, formalin-fixed,
paraffin-embedded.

NUCLEIC ACID ARRAYS

Purpose
• To profile the mRNA expression of thousands of genes in one experiment
Requirements
• Total RNA or mRNA from samples (larger amounts than required for RT-PCR)
Underlying concepts
• Hybridization to DNA – the same principle of hybridization applies as in PCR (described in Table 3.1), except that many different genes are being
evaluated simultaneously. DNA is attached to beads, chemically synthesized on a surface at thousands of specific locations, or spotted onto glass slides
(Fig. 3.9). If cells are expressing mRNA of the corresponding gene, labeled cRNA prepared from mRNA in those cells will hybridize and generate a signal
intensity related to its level of expression
Outline of method (Fig. 3.9)
• RNA is reverse transcribed to generate cDNA. In vitro transcription of the cDNA in the presence of biotinylated nucleotides yields biotin-labeled cRNA.
Labeled cRNA molecules are then hybridized to small DNA fragments attached to beads or on a chip. The samples are stained with streptavidin
phycoerythrin, the streptavidin binding the biotin on the cRNA and the phycoerythrin generating a fluorescent signal. Usually, the patterns of gene
expression of two samples are compared, such as normal versus tumor or treated versus untreated
• Alternatively, oligonucleotide arrays can be used. Using this approach, the RNA is directly labeled with either fluorescent or radioactive nucleotides, and

hybridized to oligonucleotides on a slide or membrane. When using radioactive probes, the samples are hybridized to separate microarrays. If the mRNA
samples to be compared are labeled with differently colored fluorescent probes, the two samples can be hybridized to the same array simultaneously
• A scanner measures the intensity of the signals at each spot and computer software determines those genes over- and underexpressed in one sample

compared to the other


Benefits
• Thousands of genes can be rapidly and quantitatively profiled in one experiment
Limitations/errors
• Genes expressed at low levels may not be detected
• For some genes, the cRNA may not hybridize under the conditions used
• The RNA must be of very high quality

Experimental applications
• Microarray analysis can be used to profile changes in gene expression
• The patterns of gene expression can be used to group samples
Modifications/alternatives
• RNA-seq
• MicroRNA arrays
• Comparative genomic hybridization (CGH) to detect differences in DNA copy number

• Single nucleotide polymorphism (SNP) arrays to assess markers of genetic variation (see Fig. 54.6); useful for linkage analysis, including genome-wide
72
association studies (GWAS; see Fig. 54.7)
Table 3.6 Nucleic acid arrays.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
NUCLEIC ACID ARRAYS

$ Expression bead array % Gene chip microarray & Glass slide microarray: comparative hybridization CHAPTER

Molecular Biology
Cells Cells Control cells (e.g. melanocytes) Test cells (e.g. melanoma cells)
mRNA extraction

mRNA mRNA mRNA Labeling with mRNA


Cy5 fluorescent dye Cy3
Reverse transcription

cDNA cDNA Cy5-labeled cDNA Cy3-labeled cDNA


In vitro transcription Biotin-labeled Biotin-labeled
nucleotides nucleotides
B B B B

B B B B
Biotin-labeled cRNA Biotin-labeled cRNA
Fluorescent streptavidin Fluorescent streptavidin
Hybridization
B B
B B

Address Probe

Beads loaded with 50 bp Gene chip with up to Slide with ~40 000
gene-specific probes 1.4x106 DNA probe sets unique DNA probes

Signal detection and computer analysis

Fig. 3.9 Nucleic acid arrays. A In the expression bead array, RNA is extracted from cells and reverse transcribed to generate cDNA (see Fig. 3.6, Table 3.3). In vitro

transcription of the cDNA in the presence of biotinylated nucleotides yields biotin-labeled cRNA. Labeled cRNAs bind to their respective 50-base probes that are
linked to separate beads using additional nucleotides as an address to decode the gene. Fluorescent streptavidin is added which strongly binds to biotin. The
fluorescent intensities on different beads are related to the amount of cRNA and the latter is a reflection of the level of expression of mRNA by specific genes.
Usually, the patterns of gene expression of two samples are compared, such as normal versus tumor or treated versus untreated. B In the gene chip microarray,
labeled cRNA molecules are hybridized to small DNA fragments chemically synthesized on a surface, with the surface containing thousands of specific sequences
at different locations. C In a third approach using glass slides, two samples can be labeled with differently colored fluorescent probes (e.g. Cy5, Cy3) that are
hybridized to an oligonucleotide array simultaneously (see Table 3.6 for more details). Arrays can also be used to analyze DNA rather than RNA; examples include
single nucleotide polymorphism (SNP) arrays and array comparative genomic hybridization (CGH) (see Chs 54 & 113).

cells in the mouse (Fig. 3.13)23. A further refinement is to trigger dele- therapy may be directly administered to the patient’s skin in vivo, but
tion of the gene from a specific tissue at a particular point in time, there are concerns regarding safety and the potential for genetic altera-
including during development24. One advantage of these inducible tion of the germline. Therefore, ex vivo techniques are being developed
systems, as compared to gene deletion in all mouse cells, is that it may in which cells are grown from a skin sample, treated in vitro, and
more faithfully mimic how gene deletion occurs in a specific tissue of grafted back onto the patient (see Fig. 2.7).
a patient and thereby contributes to a phenotype such as tumors in Because genetic manipulation of stem cells may result in long-term
those tissues. Another advantage is that gene deletion in all mouse cells engraftment and stable benefits26, they are often preferred. However,
may prevent normal mouse development, whereas selective gene dele- the number of stem cells that can be obtained from adult tissues is
tion after development is complete will avoid this problem. Finally, limited, so there is considerable interest in gene therapy utilizing
selective gene deletion limited to specific target tissues provides a less induced pluripotent stem (iPS) cells (see Ch. 2). Fibroblasts grown from
complicated and cleaner animal model with fewer secondary effects human skin can be reprogrammed into iPS cells via exposure to tran-
resulting from gene deletion in tissues not being targeted. In addition scription factors; these iPS cells are pluripotential and can be propa-
to the use of tamoxifen for temporal control of Cre expression (see Fig. gated in vitro indefinitely27. iPS cells from a patient may be genetically
3.13), the “Tet-On/Off” system can be used to address the problem of corrected and then differentiated into the desired cell type, such as
embryonic death when a particular gene is deleted. keratinocytes, for grafting back to the patient (see Fig. 2.7).
When pursuing gene therapy, there are several ways that the DNA
can be manipulated. To compensate for loss-of-function of a gene, a
Gene-Based Therapy for Skin Diseases new copy of the gene may be introduced into cells utilizing either non-
Gene-based therapy is a therapeutic approach that holds future promise viral or viral vectors. Sleeping Beauty is a non-viral, cut-and-paste DNA
as a treatment for skin diseases25. In gene-based therapy, DNA or RNA transposon that improves long-term gene expression, as compared to
is manipulated in order to add or correct a gene in the cells of a target that obtained when only DNA from the gene of interest is used. 73
tissue with the goal of achieving a therapeutic effect (Table 3.10). This However, the DNA transposon poses risks for insertional mutagenesis

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
PROTEOMICS WITH MASS SPECTROMETRY PROTEOMICS WITH MASS SPECTROMETRY

Purpose
SECTION • High-throughput analysis that allows investigators to rapidly and
1 quantitatively assess the complex mixtures of proteins present in a cell
at a given point in time
Overview of Basic Science

Requirements Laser
HPLC
• A mixture of cellular proteins, or peptides derived from these proteins, is
purified from a defined population of cells and then separated into +
+
proteins and/or peptides prior to mass spectrometry +
+
+
Underlying concepts Sample
Analyzer
• Mass spectrometry is able to measure very precisely the size or mass of Detector
proteins, peptides, or peptide fragments by giving these peptides a
positive charge (ionization) and then measuring the time required for the 2D gel Laser Mass
Digestion Ions Detection
positively charged peptide ions to move through a tube to a detector separation excitation analysis
(time-of-flight)
Outline of method (Fig. 3.10)
• The first step is to reduce the complexity of the mixture of cellular
L S P Q D L C D K
proteins or peptides to be analyzed, e.g. by utilizing two-dimensional Intensity
gel electrophoresis and/or liquid chromatography columns Amino acid sequence
• Mass spectrometry analysis is performed on the separated proteins/

peptides by ionizing them into positively charged ions using lasers or


the processes of electrospray ionization and nanospray ionization.
m/z
Based upon the time-of-flight of the charged ion, the peptide mass/ Mass spectrum
charge ratio can be measured and may allow the identification of the
peptide sequence and the protein that contained this peptide
Fig. 3.10 Proteomics with mass spectrometry. The first step is to reduce the

• Tandem mass spectrometry [MS/MS] can “ion trap” a given peptide ion
complexity of the mixture of cellular proteins or peptides to be analyzed. The
and then sequence the peptide by fragmenting it into its component two main methods to separate proteins/peptides from each other are
amino acids. Bioinformatics software can use this sequence information two-dimensional (2D) gel electrophoresis and/or high-performance liquid
to search protein databases for rapid protein identification chromatography (HPLC) columns. Mass spectrometry analysis is then
Benefits performed by first ionizing the separated proteins/peptides into positively
charged ions using lasers. Based on the time-of-flight of the charged ion, mass
• Mass spectrometry is extremely sensitive and able to detect very small spectrometry can measure, record and print out the mass/charge ratio of every
amounts of proteins/peptides peptide along with signal intensity.
Limitations/errors
• Better technology to reduce the complexity of protein/peptide mixtures
and differentially separate the proteins/peptides in a high-throughput
manner is being developed
• Although mass spectrometry can measure the mass of proteins/

peptides very precisely, it is more difficult to measure the amount of


protein/peptide quantitatively, and more robust techniques for
quantification by mass spectrometry are being developed
Experimental applications
• Mass spectrometry can be used to determine the complete set of It may be possible to avoid the complication of insertional mutagen-
proteins present in a defined population of cells esis by correcting the endogenous gene rather than inserting a gene.
• Because of the sensitivity of mass spectrometry analysis, it holds Methods for targeted gene correction include the use of zinc finger
promise as a sensitive diagnostic tool nucleases (ZFNs), transcription activator-like effector nucleases
Modifications/alternatives
(TALENs), and CRISPR-Cas9 (Fig. 3.14)30. These nucleases have
sequence-specific DNA-binding modules that are linked to a DNA
• Isotope labeling approaches for quantitative mass spectrometry, cleavage domain. Thus they can induce a double-strand DNA break at
including isotope-coded affinity tag (ICAT), stable isotope labeling by a specified site. The double-strand break increases the frequency of
amino acids in cell culture (SILAC), tandem mass tags (TT), and isobaric homology-directed repair, so that a co-delivered plasmid with extended
tags for relative and absolute quantification (iTRAQ) homology arms can correct the existing gene. Correction of endogenous
• Label-free quantitative proteomics
genes holds promise for treating genetically based diseases.
• Antibody protein arrays and reverse-phase protein arrays
Another therapeutic approach is to target specific mRNAs for degra-
Table 3.7 Proteomics with mass spectrometry.
  dation. Double-stranded RNAs, 21–23 nucleotides in length, are
designed complementary to the mRNA of interest. These synthesized
antisense oligodeoxynucleotides (AS-ODNs) are delivered to the cell,
where they mediate sequence-specific mRNA degradation using an
endogenous complex of proteins called the RNA-induced silencing
complex (RISC). Ribozymes also target specific sequences based on
short complementary oligonucleotides, but the catalyst for target deg-
radation is provided by the RNA enzyme without the need for protein
because it may randomly insert near a proto-oncogene that then cofactors. However, AS-ODN and ribozyme techniques are being
becomes activated28. Viral vectors can also be used to introduce DNA, replaced by synthetic vectors that deliver short interfering RNA (siRNA)
including self-inactivating lentiviral and gamma retroviral vectors. and microRNA (miRNA) (Fig. 3.15). These synthetic delivery vectors
These vectors integrate into the genome and have less risk of inser- include lipid-based or polymer-based nanoparticles, as well as covalent
tional mutagenesis compared to earlier retroviral vectors25. However, delivery systems such as dynamic polyconjugates and GalNAc conju-
74 viral vectors do have disadvantages such as immunogenicity, broad gates29. These approaches have been used successfully in the laboratory
tropism, and limited DNA packaging capacity29. and are being tested in clinical trials.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
TRANSGENIC MICE
Purpose
• Transgenic mouse models allow investigators to study the effects of a transgene (the gene of interest) on a cellular, tissue, and whole animal level. The
CHAPTER
transgene can be selectively expressed in a particular cell type or tissue by using a promoter/enhancer regulatory region that is specific for that cell type
Requirements 3
• A transgene or gene of interest, whose biologic function is to be characterized in an animal model

Molecular Biology
• A regulatory region (promoter/enhancer) that will selectively express the transgene in a specific tissue
• The facilities and technologies to create transgenic mice

Underlying concepts
• After injection of a transgene into a fertilized mouse egg or single-cell embryo, the transgene randomly integrates into the genome (Fig. 3.11). As the
embryo undergoes many cell divisions and develops into a mouse, the integrated transgene will be present in every cell type, tissue, and organ in the
mouse. Transgene expression only occurs in cells and tissues where the promoter/enhancer regulatory region is active. Any phenotypes that develop in
mice expressing the transgene in the desired tissue can help us understand the biologic effects of the transgene
Outline of method (Fig. 3.11)
• A transgene construct, defined as the transgene and a regulatory region (promoter/enhancer), is prepared for injection
• The transgene is microinjected into fertilized eggs (single-cell stage) and the transgene integrates into the genome, usually at a single site
• These injected eggs are then implanted into a recipient mother, who then gives birth to a heterozygous “founder” mouse that can be either male or female.

The founder mouse is referred to as a transgenic mouse because it only contains the transgene on one of the two paired chromosomes (e.g. only one
copy of chromosome 7 contains the transgene, whereas the other chromosome 7 does not)
• The founder mice are bred with normal non-transgenic mice of the same strain. The progeny mice will be both heterozygous transgenic and non-

transgenic, according to Mendelian genetics


• In order to derive homozygous transgenic mice that contain the transgene on both of the paired chromosomes (e.g. both copies of chromosome 7 contain

the transgene), two heterozygous transgenic mice are mated. Homozygous transgenic mice will have a double dose of the transgene, which may lead to
different phenotypes and biologic effects from those present in the heterozygous transgenic mice
Benefits
• Transgenic mouse models can help us understand the in vivo biologic effects of known and unknown genes when expressed in a specific tissue
Limitations/errors
• The precise regulation of the level of transgene expression as well as the timing of gene expression during development (turning the transgene on or off
during development) have been relatively difficult. Advancements in transgenic design, including inducible promoters, have improved the regulation of
transgene expression
• When expressed in mouse tissues, a human transgene may have different biologic effects compared to its effects in human tissues

• Classic transgenic mouse models do not allow investigators to study the biologic effects of gene mutations or deletions that result in loss of gene

expression, because the technique of creating transgenic mice does not alter endogenous genes, it only adds a new transgene to each cell
Experimental applications
• Transgenic mouse models of disease can be used to test the effectiveness of new therapeutic agents
Modifications/alternatives
• More precise control of transgene expression can be achieved with promoters/enhancers that are inducible and can be regulated (i.e. turned off and on at
different time points)
• Programmable nuclease systems for RNA-guided genome editing such as transcription activator-like effector nuclease (TALEN) and clustered regularly

interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) (CRISPR-Cas9; Fig. 3.14)
• “Knockout” transgenic mouse models (Table 3.9) allow investigators to understand the biologic effects of gene mutations and deletions that cause a loss

of specific proteins
Table 3.8 Transgenic mice.

CONCLUSIONS Advances in our scientific understanding translate into new develop-


ments in disease diagnosis and management. For dermatology, this
In this chapter, we have presented a variety of techniques used in provides hope that diseases can be diagnosed more rapidly and more
dermatology research and clinical practice. Some of these are new, reliably, and that individualized prognostic information can be pro-
whereas others are long established and still widely used. New tech- vided. Therapies have been and will continue to be developed that
nologies continue to appear and change the way research is done. specifically target the genetic abnormality causing the disease. In the
These technologic advances allow experiments to be done faster and future, therapies may be tailored to the individual, taking into consid-
with less and less starting material. They allow the measurement eration the genetic make-up of the individual and his or her disease
of thousands of different genes or proteins in single experiments. process. These future possibilities are exciting.
They also automate experiments that used to require time-consuming
step-by-step human involvement. To handle all the information cur- Copyright notice
rently produced by biologic research, advances in computers and Chapter 3 is US Government work in the public domain and not
bioinformatics are progressing along with the technologic advances. subject to copyright.
The outcome is that the pace of scientific inquiry proceeds at an 75
increasing rate. For additional online figures visit www.expertconsult.com

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Online only content
BIOINFORMATICS SKIN GENE THERAPY

$ Direct injection CHAPTER

PubMed
Full-text
journals
online
3
abstracts

Molecular Biology
3-D
Phylogeny
structure
(Taxonomy)
(MMDB)

Genomes

% Gene gun: Micro-projectiles


Nucleotide Protein
sequences sequences

eFig. 3.1 Bioinformatics. A goal of bioinformatics is to give users access to


huge amounts of biologic information in a way that is easy to use and analyze.
One example is Entrez produced by the National Center for Biotechnology
Information (NCBI), available at http://www.ncbi.nlm.nih.gov. This retrieval
system allows the user to search the biomedical literature (PubMed),
nucleotide sequence database (GenBank), protein sequence database,
three-dimensional macromolecular structures, and complete genome
assemblies and organisms in GenBank (taxonomy). As indicated by arrows,
users may access information from a single database and also integrated
information from several NCBI databases. MMDB, molecular modeling
database.

eFig. 3.2 Skin gene therapy. In the direct in vivo approach, the desired gene is

introduced directly into the skin as shown, either directly injected intradermally
(A) or biolistically discharged into the epidermis and dermis (B). Both viral and
non-viral vectors can be delivered to the skin using these in vivo approaches.

SKIN GENE THERAPY

Grafting genetically modified keratinocytes

Gene introduction

Keratinocyte culture

eFig. 3.3 Skin gene therapy. In the ex vivo approach, keratinocytes are

removed from the donor and, during ex vivo culture, the desired gene is
efficiently introduced, usually with viral vectors. Skin equivalents or raft
cultures containing these genetically modified keratinocytes (along with a
dermal portion containing fibroblasts) are constructed and then grafted back
onto the donor.

75.e1

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Fig. 3.11 Transgenic mice. A transgene construct, defined as

the transgene and regulatory region (promoter/enhancer), is


TRANSGENIC MICE
prepared for injection. The transgene is microinjected into
fertilized eggs (single cell stage) and the transgene integrates
SECTION into the genome, usually at a single site. These injected eggs

1 Transgene (+)
are then implanted into a recipient mother, who then gives
birth to a heterozygous “founder” mouse. The founder mice are
bred with normal non-transgenic mice of the same strain, and
Overview of Basic Science

then two heterozygous transgenic mice are mated.

1 cell
Injection pipet
embryo

Founder
(+/–)

Founder Normal
(+/–) (–/–)

(–/–) (+/–) (+/–) (–/–)

(+/+) (+/–) (+/–) (–/–)

KNOCKOUT TRANSGENIC MICE

Purpose
• Knockout transgenic mice have both alleles of a normal endogenous gene modified, mutated, or deleted in all cells (whole animal or targeted tissue) so
that protein can no longer be produced by that gene. Therefore, knockout transgenic mice allow investigators to understand what happens to a cell or
tissue when a particular gene can no longer be expressed
Requirements
• Murine embryonic stem cells (ES cells) that are totipotent and capable of forming and reconstituting all tissues and organs of a mouse
• A targeting construct defined as a defective gene that can hybridize to the normal endogenous gene in ES cells and modify or delete the targeted gene so
that it is no longer able to express a functional protein
Underlying concepts
• In knockout transgenic mice, all cells (whole animal or targeted tissue) lack the gene that has been “knocked out” and do not express any functional
protein from that gene
• Technology is available to selectively target and delete an endogenous gene in its normal chromosomal location in ES cells

Outline of method (Fig. 3.12)


• A targeting vector that contains some sequences of the gene to be targeted is created and introduced into ES cells. This targeting vector is able to
selectively hybridize to one of the endogenous alleles of a gene, and through a process known as homologous recombination, the targeting vector
modifies or deletes the endogenous gene so that no normal protein is produced. ES cells that contain this defective endogenous gene can be selected for
and isolated
• The ES cells containing the “knocked out” gene are introduced into early mouse embryos, known as blastocysts, which are then implanted into recipient

mothers. The progeny will be chimeric mice; all tissues and organs, including testes and ovaries, will contain a mixture of normal cells and cells that lack
the “knockout” gene allele
• The chimeric mice are next mated to normal mice. Depending on the degree of chimerism, a certain percentage of the offspring will be heterozygous,

containing one normal allele and one allele that has been modified or deleted (knocked out). As described for the mating procedure in transgenic mice
(see Table 3.8), the genotypes of all progeny will follow the rules of Mendelian genetics
• To obtain completely “knocked out” mice, two heterozygous mice are mated. Approximately 25% of the offspring will lack a normal endogenous gene in

every cell and tissue


76
Table 3.9 Knockout transgenic mice.

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
KNOCKOUT TRANSGENIC MICE

Benefits
• This approach yields mice that completely lack the gene product in all cells of the whole animal or targeted tissues CHAPTER
Limitations/errors
• Investigators have to ascertain that no normal gene product is being produced. It is possible that the homologous recombination process used to knock
3
out a gene may result in a gene that encodes a mutant protein that still has biologic effects. This would make it difficult to understand the biologic effects of

Molecular Biology
the gene
Experimental applications
• Unlike regular transgenic mice, knockout transgenic mice allow investigators to study the biologic effects of losing gene expression. This is particularly
useful in the study of genes that may normally suppress cancer development (tumor suppressors); the tumor suppressor gene can be knocked out in all
mouse cells in vivo, and the mechanisms of cancer development in different tissues can be studied
Modifications/alternatives
• In order to more accurately mimic in vivo situations, the gene of interest can be selectively deleted in only one organ or tissue. These conditional knockout
mice allow investigators to study the effects of gene loss in a particular tissue without the complications of losing gene expression in all the other tissues
(Fig. 3.13)
• A similar technology is also available to delete a gene of interest at a particular time during development. Initially, the endogenous gene will be expressed

normally in the knockout mice and then deleted at a later time, either during intrauterine development or after birth. This is advantageous if investigators
want to study the effects of a gene that, if knocked out, would prevent embryo development
• Targeted genome editing using CRISPR-Cas9 (Fig. 3.14)

Table 3.9 Knockout transgenic mice. (cont’d)


KNOCKOUT TRANSGENIC MICE CONDITIONAL KNOCKOUT TRANSGENIC MICE

X
Mutated gene
P Cre X IoxP Gene X IoxP
Early
ES mouse
embryo

Cre gene linked to a Target gene X flanked


tissue-specific promoter by IoxP sites

Chimeric
(–/–)(+/+)

P Cre
X
IoxP Gene X IoxP
Chimeric Normal
(–/–)(+/+) (+/+)

In tissue expressing Cre

(+/+) (+/–) (+/–) (+/+)


IoxP
Gene X

Excision of DNA Cleavage, exchange


between loxP sites and ligation by Cre
IoxP

(+/+) (+/–) (+/–) (–/–)


IoxP
Fig. 3.12 Knockout transgenic mice. A targeting vector that contains some

sequences of the gene to be targeted is created and introduced into cells. This
Tissue-specific deletion of Gene X
targeting vector is able to hybridize selectively to one of the endogenous
alleles of a gene, and modifies or deletes the endogenous gene so that no
normal protein is produced. The embryonic stem (ES) cells containing the Fig. 3.13 Conditional knockout transgenic mice. In the conditional knockout
“knocked out” gene are introduced into early mouse embryos, which are then

model using the Cre-lox system, a target gene can be selectively deleted in a
implanted into recipient mothers. The progeny will be chimeric mice. These particular organ or tissue. Cre is a site-specific DNA recombinase that catalyzes
chimeric mice are then mated to normal mice. To obtain completely knocked recombination between two loxP sites, resulting in cleavage, exchange, and
out mice, two heterozygous mice are mated. ligation; this leads to excision (and subsequent degradation) of the DNA
between the loxP sites. A transgenic mouse with the Cre gene linked to a
tissue-specific promoter is mated to a transgenic mouse with two loxP sites
flanking the target gene. Progeny inherit both of these constructs, and the
target gene is knocked out only in cells that activate the tissue-specific
promoter. By adding a ligand-binding domain to the Cre recombinase, such as
one that binds tamoxifen, investigators can achieve temporal control of Cre 77
expression in these cells. The target gene can be temporally deleted, inverted
or translocated, depending on the orientation of the loxP sites.
Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
SKIN GENE-BASED THERAPY

Purpose
SECTION • In skin gene-based therapy, genes are introduced and expressed, corrected, or knocked down in the skin for therapeutic purposes. Diseases that could
1 be treated by this approach include both skin diseases and systemic diseases where skin is used as a vehicle for the systemic delivery of a needed factor
or substance
Overview of Basic Science

Requirements
• A gene that is able to achieve the desired therapeutic effect when expressed in keratinocytes or other skin cells such as fibroblasts and melanocytes
• A delivery vector that can efficiently introduce the desired gene into cells. Both viral vectors and non-viral vectors can be used to deliver a desired gene to
a target cell type. With viral vectors, the desired gene is incorporated into the genetic material of a modified virus that is able to efficiently infect target
cells but is no longer able to cause disease and spread to other cells
Underlying concepts
• Skin gene therapy is a promising field that has the potential to provide new treatments for both skin and systemic diseases
• There are two general approaches for introducing genes into the skin: a direct in vivo approach and an ex vivo approach
• The viral vectors that have most often been used to deliver genes to skin cells are retroviral, lentiviral, and adenoviral vectors

• Many issues need to be considered when choosing a vector and determining the best approach to deliver the desired gene to the skin. These include: (1)

how many cells (what percentage) need to contain the gene in order to have a therapeutic effect; (2) how long the gene needs to be expressed in order to
be therapeutic. In some situations, short-term or transient expression is sufficient; in other cases, long-term expression is preferable
Outline of method
• In the direct in vivo approach, the desired gene is introduced directly into the skin by injection or physical methods such as gene gun or electroporation
• In the ex vivo approach, keratinocytes are removed from the donor and, during ex vivo culture, the desired gene is efficiently introduced, usually with viral
vectors. Skin equivalents or raft cultures containing these genetically modified keratinocytes (along with a dermal portion containing fibroblasts) are
constructed and then grafted back onto the donor in the correct anatomic location
• The principal advantage of the in vivo approaches is their simple straightforward nature. A disadvantage is that expression is usually transient and not

long-term because selective targeting of keratinocyte stem cells is usually not feasible with in vivo approaches. Another disadvantage is that the
percentage of cells containing the desired transgene may be lower than desired
• In contrast, the ex vivo approaches are superior at achieving long-term expression of a desired gene in a high percentage of skin cells. During the ex vivo

culture period, the desired gene could be targeted to keratinocyte stem cells
• The disadvantage of the ex vivo approach is that it is much more complicated and technically demanding than the in vivo approaches

Benefits
• Genes are the therapeutic agent in this approach to treatment of cutaneous and systemic disease. As our understanding of the molecular basis of disease
increases, genetic therapies will become more important
• The effectiveness of skin gene therapy can be assessed in animal models

Limitations/errors
• Successful skin gene therapy with long-term expression of a desired gene in a high percentage of cells has been difficult to achieve in animal models
• Skin gene therapy for recessive genetic skin diseases is problematic because of the development of an unwanted immune response against the normal
protein
• Risk of oncogenesis, especially with retroviral vectors

Experimental applications
• Treatment of genetic skin diseases
• Treatment of non-genetic skin diseases if a sufficient understanding of the molecular basis of the disease exists
• Systemic delivery of cytokines, hormones, and growth factors

• Genetic immunization

• Suicide gene therapy

Modifications/alternatives
• Use of short (small) interfering RNA (siRNA) that specifically targets and silences expression of dominant-negative mutant genes (Fig. 3.15)
• Gene-based therapy utilizing induced pluripotent stem (iPS) cells (see Fig. 2.7)
• Protein replacement therapies

• Cellular therapies

Table 3.10 Skin gene-based therapy.


78

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Fig. 3.14 Genomic engineering

GENOMIC ENGINEERING MEDIATED BY CRISPR-Cas9 TECHNOLOGY mediated by CRISPR-Cas9


technology. A single-guide RNA
(sgRNA) is engineered to target
genomic DNA adjacent to a CHAPTER

5
Engineered sgRNA
20 nucleotide
protospacer adjacent motif (PAM)
utilizing a 20 nucleotide guide
sequence. The hairpin structure
3
guide sequence
Cas9 

Molecular Biology
nuclease recruits Cas9 to form a complex
– 3 that binds to the target sequence.
Cas9 catalyzes cleavage of both
strands of DNA three nucleotides
upstream of the PAM. A double-
strand break may be repaired by
Target DNA sequence nonhomologous end joining
(NHEJ) that leads to the formation
of short insertions or deletions that
Cas9-sgRNA
complex disrupts gene function. In the
presence of a DNA-repair template,
homology-directed repair (HDR)
Protospacer- may occur in order to introduce
adjacent exogenous DNA at the site of the
motif (PAM)
break.

Cas9-induced double-
stranded DNA breaks

3 – – 5

5 – – 3

PAM
Target DNA

Nonhomologous
end joining (NHEJ) Homology-directed
repair (HDR)

Repair of double-stranded DNA

Donor DNA

indel New DNA

indel, short insertion or deletion that disrupts gene function

79

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Fig. 3.15 Mechanisms of

MECHANISMS OF SEQUENCE-SPECIFIC GENE SILENCING VIA mRNA KNOCKDOWN sequence-specific gene silencing
via mRNA knockdown. A Short
(small) interfering RNA (siRNA) is
SECTION
A B C unwound and the “guide” antisense

1 Chemically synthesized
short interfering RNA
Short hairpin RNA
(shRNA) precursor
MicroRNA (miRNA)
precursor expressed
strand incorporated into an
RNA-induced silencing complex
(RISC) that degrades a specific
expressed from plasmid
Overview of Basic Science

(siRNA) molecule from genomic DNA target mRNA sequence. Processing


or viral vector DNA
of short hairpin RNA (shRNA;  
B) and pre-microRNA (miRNA; C)
Processing and by the Dicer enzyme can generate
export from nucleus siRNA and miRNA, respectively.  
C Endogenously produced miRNA
5 regulates up to a third of human
5 genes and tends to have less
3
shRNA 3 complementarity with target
pre-miRNA mRNA; it recruits RISC proteins and
Dicer Dicer typically inhibits mRNA translation
(rather than decreasing mRNA
5 3 levels).
3 5
siRNA 5 3
Duplex unwinding 3 5
and strand selection miRNA

5 AAA3
3 5
siRISC
5 AAA3
mRNA cleavage 3 5
Ribosome miRISC

AAA

mRNA degradation Translational repression

REFERENCES
1. Schaffer JV. Molecular diagnostics in genodermatoses. 11. Jahan-Tigh RR, Ryan C, Obermoser G, Schwarzenberger 20. Solier C, Langen H. Antibody-based proteomics and
Semin Cutan Med Surg 2012;31:211–20. K. Flow cytometry. J Invest Dermatol 2012;132:e1. biomarker research – current status and limitations.
2. Swick BL. Polymerase chain reaction-based molecular 11a.  Spitzer MH, Nolan GP. Mass cytometry: single cells, Proteomics 2014;14:774–83.
diagnosis of cutaneous infections in dermatopathology. many features. Cell 2016;165:780–91. 21. Schneider MR. Genetic mouse models for skin research:
Semin Cutan Med Surg 2012;31:241–6. 12. Garibyan L, Avashia N. Polymerase chain reaction. J strategies and resources. Genesis 2012;50:652–64.
3. Carless MA, Griffiths LR. Cytogenetics of melanoma and Invest Dermatol 2013;133:e6. 22. Avci P, Sadasivam M, Gupta A, et al. Animal models of
nonmelanoma skin cancer. Adv Exp Med Biol 13. Grada A, Weinbrecht K. Next-generation sequencing: skin disease for drug discovery. Expert Opin Drug
2014;810:160–81. methodology and application. J Invest Dermatol Discov 2013;8:331–55.
4. Griewank KG, Scolyer RA, Thompson JF, et al. Genetic 2013;133:e11. 23. Scharfenberger L, Hennerici T, Király G, et al. Transgenic
alterations and personalized medicine in melanoma: 14. Bustin SA, Nolan T. Analysis of mRNA expression by mouse technology in skin biology: generation of
progress and future prospects. J Natl Cancer Inst real-time PCR. In: Saunders NA, Lee MA, editors. complete or tissue-specific knockout mice. J Invest
2014;106:djt435. Real-time PCR: advanced technologies and applications. Dermatol 2014;134:e16.
5. Deonizio JM, Guitart J. The role of molecular analysis in Norfolk, UK: Caister Academic Press; 2013. p. 51–88. 24. Günschmann C, Chiticariu E, Garg B, et al. Transgenic
cutaneous lymphomas. Semin Cutan Med Surg 15. Nicholas MW, Nelson K. North, south, or east? Blotting mouse technology in skin biology: inducible gene
2012;31:234–40. techniques. J Invest Dermatol 2013;133:e10. knockout in mice. J Invest Dermatol 2014;134: 
6. Fine JD, Bruckner-Tuderman L, Eady RA, et al. Inherited 16. Gan SD, Patel KR. Enzyme immunoassay and e22.
epidermolysis bullosa: updated recommendations on enzyme-linked immunosorbent assay. J Invest 25. Abdul-Wahab A, Qasim W, McGrath JA. Gene therapies
diagnosis and classification. J Am Acad Dermatol Dermatol 2013;133:e12. for inherited skin disorders. Semin Cutan Med Surg
2014;70:1103–26. 17. Chen AY, Chen A. Fluorescence in situ hybridization. J 2014;33:83–90.
7. Otten JV, Hashimoto T, Hertl M, et al. Molecular Invest Dermatol 2013;133:e8. 26. Therrien JP, Pfützner W, Vogel JC. An approach to
diagnosis in autoimmune skin blistering conditions. 17a.  Whitley SK, Horne WT, Kolls JK. Research techniques achieve long-term expression in skin gene therapy.
Curr Mol Med 2014;14:69–95. made simple: methodology and clinical applications of Toxicol Pathol 2008;36:104–11.
8. Miller DM, Flaherty KT, Tsao H. Current status and future RNA sequencing. J Invest Dermatol 2016;136:e77–82. 27. Dinella J, Koster MI, Koch PJ. Use of induced pluripotent
directions of molecularly targeted therapies and 18. Villaseñor-Park J, Ortega-Loayza AG. Microarray stem cells in dermatological research. J Invest Dermatol
immunotherapies for melanoma. Semin Cutan Med technique, analysis, and applications in dermatology. J 2014;134:e23.
Surg 2014;33:60–7. Invest Dermatol 2013;133:e7. 28. Skipper KA, Andersen PR, Sharma N, Mikkelsen JG. DNA
9. Rizzo AE, Maibach HI. Personalizing dermatology: the 18a.  Szuhai K, Vermeer M. Microarray techniques to analyze transposon-based gene vehicles – scenes from an
future of genomic expression profiling to individualize copy-number alterations in genomic DNA: array evolutionary drive. J Biomed Sci 2013;20:92.
dermatologic therapy. J Dermatolog Treat comparative genomic hybridization and single- 29. Yin H, Kanasty RL, Eltoukhy AA, et al. Non-viral vectors
2012;23:161–7. nucleotide polymorphism array. J Invest Dermatol for gene-based therapy. Nat Rev Genet 2014;15: 
10. Legres LG, Janin A, Masselon C, Bertheau P. Beyond 2015;135:e37. 541–55.
laser microdissection technology: follow the yellow 19. Woods AG, Sokolowska I, Wetie AG, et al. Mass 30. Gaj T, Gersbach CA, Barbas CF 3rd. ZFN, TALEN, and
brick road for cancer research. Am J Cancer Res. spectrometry for proteomics-based investigation. Adv CRISPR/Cas-based methods for genome engineering.
2014;4:1–28. Exp Med Biol 2014;806:1–32. Trends Biotechnol 2013;31:397–405.

80

Downloaded for Nicole Aguilar (cnaguilars@gmail.com) at University of Miami from ClinicalKey.com by Elsevier on July 19, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.

You might also like