Vous êtes sur la page 1sur 22

Scand J Med Sci Sports 2016: : – ª 2016 The Authors.

Scandinavian Journal of Medicine &


doi: 10.1111/sms.12702 Science in Sports published by John Wiley & Sons Ltd

Review

An overview of technical considerations for Western blotting


applications to physiological research
J. J. Bass, D. J. Wilkinson, D. Rankin, B. E. Phillips, N. J. Szewczyk, K. Smith, P. J. Atherton
MRC/ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of Medicine, University of Nottingham, Derby, UK
Corresponding author: Dr. Philip J. Atherton, MRC-ARUK Centre of Excellence for Musculoskeletal Ageing Research, School of
Medicine, Derby DE22 3DT, UK. Tel: 01332 724725, Fax: 01332 724727, E-mail: philip.atherton@nottingham.ac.uk
Accepted for publication 25 April 2016

The applications of Western/immunoblotting (WB) interpretation of the data can be variable, perhaps
techniques have reached multiple layers of the scientific resulting in spurious conclusions. This may be due to poor
community and are now considered routine procedures in laboratory technique and/or lack of comprehension of the
the field of physiology. This is none more so than in critical steps involved in WB and what quality control
relation to skeletal muscle physiology (i.e., resolving the procedures should be in place to ensure robust data
mechanisms underpinning adaptations to exercise). Indeed, generation. The present review aims to provide a detailed
the inclusion of WB data is now considered an essential description and critique of WB procedures and
aspect of many such physiological publications to provide technicalities, from sample collection through preparation,
mechanistic insight into regulatory processes. Despite blotting and detection, to analysis of the data collected.
this popularity, and due to the ubiquitous and relatively We aim to provide the reader with improved expertise to
inexpensive availability of WB equipment, the quality of critically conduct, evaluate, and troubleshoot the WB
WB in publications and subsequent analysis and process, to produce reproducible and reliable blots.

The Western blot (WB) has diverse applications for Crucial quality control elements of a WB may be
investigating regulatory molecular events underpin- overlooked, leading to poor quality blots, and the
ning energy metabolism, protein turnover and potential for unintentionally misleading data pro-
chronic physiological adaptations. For example, the duction and interpretation.
WB can be used to investigate protein abundance, Outwardly, the principle of the WB is based
kinase activity, cellular localization, protein–protein around a few broad steps: (a) the extraction of cellu-
interactions, or monitoring of post-translational lar proteins from a complex mixture of intracellular
modifications [i.e., events of cleavage, phosphoryla- and extracellular proteins (from tissue, cells, etc.); (b)
tion (Nairn et al., 1982), ubiquitinylation (Paul quantification of protein concentration and elec-
et al., 2012), glycosylation (Pere-Brissaud et al., trophoretic separation of proteins within a gel
2015), methylation (Voelkel et al., 2013), and matrix; (c) transfer to a membrane with a high affin-
SUMOylation (Park-Sarge & Sarge, 2010); to name ity for proteins; (d) “blocking” the membrane to
the main applications]. While such WB approaches reduce non-specific binding; (e) antigen detection by
are routinely used in many fields of biochemical antibodies specific for the protein(s) of interest; (f)
research, the application of the WB to skeletal mus- incubation with a secondary antibody linked to a
cle and exercise physiology is increasing. This is for label (e.g., chemiluminescent or fluorescent); (g)
reasons relating to the pursuit of an improved under- development and detection of the signal, which is
standing of molecular pathways involved in the regu- theoretically proportional to the degree of antigen/
lation of transcription and translation by exercise antibody binding; and (h) quantification of the
and nutrition in health, aging, and disease. This resulting bands using densitometry software (Fig. 1).
expansion in WB applications has led to an increased Originally, the process of “Western blotting” was the
number of users lacking analytical biochemistry aspect of transferring proteins from a gel to a more
backgrounds to appreciate important caveats. stable membrane, although it commonly now refers
to the whole process. To allow for the greatest accu-
racy and interpretation of data, each aspect of the
This is an open access article under the terms of the Creative
WB process must be understood and carefully con-
Commons Attribution License, which permits use, distribution and
reproduction in any medium, provided the original work is properly sidered. In this review, we will describe the stages of
cited. the WB, focusing on the more routine WB gel

1
Bass et al.

Fig. 1. The sequential stages of the Western blot process.

electrophoresis methodologies using standard SDS- • Are additional buffer components (e.g., deter-
PAGE, wet transfers, and chemiluminescence, cri- gents, enzymatic inhibitors) required for solubi-
tiquing and highlighting important points to lization, fractionation, or maintenance of post-
consider throughout. In the main, we will translational modifications?
concentrate on analysis of skeletal muscle tissues • What is the method of protein quantification, will
derived from skeletal muscle biopsies; nonetheless, buffer components interfere?
the details described in each element are inherently
applicable to other tissues or sample types. While Polyacrylamide gel electrophoresis:
performing a WB, there are multiple key aspects to • What concentration of gel is most appropriate
each step: (e.g., 20% for proteins <20 kDa, 7.5% for pro-
Sample preparation: teins >200 kDa)?
• Is the target protein soluble/cytoplasmic, insoluble • What running buffer is most suitable (e.g., MOPS
or membrane bound? for proteins ~75 kDa, MES for proteins <36 kDa)?

2
Technical aspects of Western blotting

• How long and what voltage to run the tank (typi- Sample handling
cally 60 min at 200 V)?
The method of sample collection depends on the
Electro-transfer: sample type: skeletal muscle tissue by biopsy [i.e., for
• What is the most suitable membrane material human tissue typically by conchotome or Bergstr€ om
(e.g., PVDF or nitrocellulose) and pore size (e.g., needle (Dietrichson et al., 1987)], samples dissected
0.45 lm)? post-mortem (i.e., after the terminal procedure of an
• Is methanol excluded in the buffer (i.e., for trans- in vivo experiment), and cell culture using scrapers
fer of larger proteins)? (Quach et al., 2009). Once tissue samples are har-
• Should membrane staining occur to assess transfer vested, they should be immediately washed in an ice-
efficiency (e.g., Ponceau)? cold neutral pH buffer, before removal of visible fat,
snap frozen in liquid N2, and stored at 80 °C.
Blocking: These steps are designed to limit protein degradation
• Which blocking reagent is most suitable (e.g., BSA and preserve post-translational modifications
or milk)? (PTMs), while concurrently reducing non-skeletal
• What concentration (e.g., 2.5%) and what buffer muscle cell contaminants and blood, all of which
(e.g., TBST) should be used? could interfere with downstream processes (e.g.,
hemoglobin may interfere with colorimetric protein
Primary antibody: assays (Doumas et al., 1981) or contaminate muscle
• What are the general characteristics of the primary cell-specific analysis). In order to obtain robust data,
antibody (e.g., monoclonal Rabbit IgG)? samples should remain frozen until use and undergo
• Is it specific toward native or denatured proteins as little manipulation as possible (e.g., periods at
and is the epitope sequence/region know? higher temperatures and multiple freeze thaws, so to
• Are additional bands known/present (i.e., degra- minimize degradation as indicated by gel streaks)
dation products or protein isoforms)? (Mahmood & Yang, 2012). Sometimes, it may be
• Have appropriate controls been run to determine viable for fresh tissue to be utilized for WB sample
specificity? preparation; however, it is more common and in our
• Is the antibody specific to a post-translational experience preferable to snap freeze tissues. This
modification (e.g., phosphorylation)? allows multiple analyses to be performed on a single
sample (e.g., mass spectrometry, quantitative real-
Secondary antibody: time PCR) while controlling the amount of tissue uti-
• What is the label conjugation (e.g., HRP or fluo- lized. Although typically whole skeletal muscle tissue
rescent)? is used, it is possible to isolate and group fibers [i.e.,
• Is the secondary antibody specific toward the pri- based on fiber type (Jensen & Richter, 2011)] or use
mary isotype? individual fibers for WB analysis (Murphy & Lamb,
• Is there detectable/overexposed signal, if so is an 2013). These techniques, however, are time consum-
antibody dilution curve required? ing and may reduce the protein yield, but may be
more informative depending on the experimental
Detection: design (i.e., influence of fiber type on an interven-
• What is the detection method (e.g., chemilumines- tion).
cent or fluorescent)? The extraction of proteins of interest from tissues
• Is fluorescent multiplexing suitable? requires the lysis and disruption of cell membranes
• Have the antibodies been stored correctly? using homogenization techniques, typically in the
• Can the membrane be successfully stripped and form of mechanical, sonication, and/or chemical
reprobed? approaches. For muscle biopsies or other solid tis-
sues, mechanical homogenization is required to
Analysis and normalization: mince large sections of tissue and disrupt membranes
• Is the band of interest within the linear range of (in order to liberate intracellular proteins). High-
the detection system? powered bench-top “polytron” homogenizers, com-
• What method of quantification is most suitable mercial “bead-beaters”, or simple scissor snipping
(i.e., whole lane or boxed analysis)? may be used to effectively release intracellular pro-
• What is the method of background detection (e.g., teins into solution (Goldberg, 2008). It is noteworthy
rolling ball algorithm)? that certain mechanical methods (e.g., polytron
• What is the method of normalization (e.g., Coo- approaches) may retain remnants of tissues if not
massie stain)? thoroughly cleaned, leading to cross-sample contam-
• Is it more suitable to measure the total vs phos- ination. If protein yields are lower than expected
phorylation expression of a protein? after quantification (e.g., in our experience yields for

3
Bass et al.
Table 1. Common buffer components for use in tissue homogenization

Chemical Purpose Typical concentration

Triton x-100 Increase solubility of non-polar proteins 0.1–1%


Urea Disruption of protein hydrogen bonds increasing solubility 6–8 M
NaCl Membrane disruption and protein solubility 1–100 mM
SDS Membrane disruption 0.1–1%
Tris-HCl Solution buffering 50 mM
MOPS/MES Solution buffering 1–5 mM
HEPES Solution buffering 50 mM
Glycerol Solution stabilization 5–10%
EGTA/EDTA Inhibition of metalloproteases and prevention of changes in protein phosphorylation 1 mM
Na3VO4 Tyrosine and alkaline phosphatase inhibition 0.5 mM
NaF Ser/Thr phosphatase inhibition 50 mM
b-glycerophosphate Ser/Thr phosphatase inhibition 10 mM
b-mercaptoethanol Cleavage of disulfide bonds and protein denaturation 1–10 mM
DTT Cleavage of disulfide bonds and protein denaturation 1–10 mM

cultured cells are typically 0.5–1.5 lg/ll, human where possible to avoid or minimize addition before
skeletal muscle biopsies 3–6 lg/ll and excised rat quantification.
kidney 12–15 lg/ll) (discussed below), sonication of The detection of certain proteins may require fur-
samples might be required to further lyse cellular ther optimization of buffer components (typical inclu-
membranes. Sonication utilizes high frequency sound sions and concentrations in Table 1). For instance,
waves to disrupt cellular membranes, nonetheless, high concentrations of salts (i.e., NaCl) or detergents
this should follow mechanical homogenization to to enhance breakdown of organelle and nuclear mem-
remove large pieces of tissue that would not be dis- branes, such as radio-immunoprecipitation buffer to
rupted by sonication alone (Autuori et al., 1982). ensure nuclear disruption (Holden & Horton, 2009).
Notably, homogenization of muscle cell cultures An example of such necessary changes is for the
does not require the same degree of mechanical lys- extraction of the DNA-bound proteins in muscle tis-
ing; instead vigorously pipetting the collected cells sue using hyperosmolar lysis buffers to effectively
through a small gauge syringe or fine tip gel-loading release DNA-bound proteins, which can increase the
pipette is typically sufficient (Crossland et al., 2013). ability to detect low abundant proteins (Girgis et al.,
Tissues should be homogenized in a buffer 2014). Therefore, the cellular location and DNA bind-
designed to solubilize and optimize preservation of ing of target protein(s) should be carefully considered
the target proteins. Accurately buffering a homoge- before determining optimal extraction buffers, which
nization solution proximate to the isoelectric point could impact on quantification and therefore conclu-
of proteins (the pH at which they have neutral sions being drawn.
charge), is necessary (pH 7–9) to ensure solubility, Disruption of cell membranes during homogeniza-
and prevention of protein precipitation, through tion also releases proteases, kinases, and phos-
maintenance of positive or negatively charged amino phatases, and despite reduced storage temperatures
acid functional (R) groups (Grabski, 2009). Addition (i.e., 4 °C), protein degradation may still occur due
of non-ionic detergents (i.e., Triton X-100) are used to retained enzymatic activity (Scopes, 1994). There-
to increase solubility of non-polar insoluble proteins fore, protease inhibitors that suppress the activity of
(Helenius & Simons, 1975). Proteins retaining ter- a variety of proteases must be added either individu-
tiary and quaternary structures remain soluble in ally or as a commercially dissolvable preparation
water, since non-polar hydrophobic regions are gen- with the aim of preventing a broad a range of pro-
erally oriented toward the center of the protein or tease activities (e.g., aprotinin for serine and E-64 for
within cell membranes (Tanford, 1962). Thus, reduc- cysteine proteases) (Grabski, 2009). Metallopro-
ing agents [e.g., dithiothreitol (DTT)] are used to teinases may be inhibited through the use of metal
breakdown disulfide bonds (S-S) between cysteine chelators such as ethylenediaminetetraacetic acid
residues (Cleland, 1964), while sodium dodecyl sul- (EDTA) and ethylene glycol tetraacetic acid (EGTA)
fate (SDS) detergent is added to coat hydrophobic via chelation (binding of metal ions) of Mg2+ and
regions of proteins with negative charge and over- Ca2+ which are required for protease activity (Auld,
whelm positive charges in proteins; this aspect is cru- 1995). Further to this, EDTA and EGTA addition-
cial for resolving proteins in accordance to their ally inhibit serine/threonine (Ser/Thr) phosphatase
molecular mass (discussed below). Reducing agents interactions; however, some caution is warranted
are required in subsequent sample preparation stages since EDTA disrupts Na+ orthovanadate-mediated
and may interfere with determining the protein con- inhibition of Tyr phosphatases (Huyer et al., 1997).
tent (discussed below), and thus it is recommended Other components are also required to maintain

4
Technical aspects of Western blotting
protein phosphorylation states, changeable by phos- (e.g., 1000 g) before removal of the mitochondrial
phatases released by homogenization. As an example containing supernatant. While crudely isolating
of the importance of this step, the addition of phos- mitochondria, pure separation is difficult as skeletal
phatase inhibitors was shown to result in increased muscle mitochondria are integrated with structures
signal intensity for phosphorylated MAP kinase in the muscle (Rasmussen & Rasmussen, 2000). It
within neuronal cells, whereas total-MAP kinase should also be noted that there are two main areas
intensity remained constant (Sharma & Carew, within the muscle where mitochondria are associ-
2002). Interactions of tyrosine (Tyr) and alkaline ated: subsarcolemma and inter-myofibrillar (IM).
phosphatase are generally inhibited through the Robust separation of each fraction requires addi-
addition of Na+ orthovanadate, acting as a competi- tional steps to mechanically or enzymatically (e.g.,
tive Tyr phosphatase inhibitor (Gordon, 1991). trypsinization) release IM mitochondria from the
Additional inhibitors of Ser/Thr phosphatases such myofibrils, which may in turn cause protein degrada-
as sodium fluoride and b–glycerophosphate may be tion to other structures or proteins within the
used in conjunction with these chelators, increasing myofibrillar fraction (Rasmussen & Rasmussen,
the effectiveness of the buffered solution to maintain 2000). It is important to note the addition of trypsin
phosphorylated protein status. Typical buffer com- will require quenching typically through the addition
ponents and concentrations can be found in Table 1. of albumin, increasing the total protein content in
It is important to note that any equipment or buffers subsequent quantification, and samples should,
used within sample processing should be pre-chilled therefore, be thoroughly washed (Beltran Valls et al.,
and ideally kept cold on ice. 2014). A recent method has been described, which
involves short proteinase treatment and homogeniza-
tion in ionic buffers followed by two-stage centrifu-
Cellular subfractionation and immunoprecipitation
gation. This method showed high mitochondrial
Isolating specific cellular fractions (myofibrils, sar- integrity and purity following isolation (Rasmussen
coplasm, mitochondria, collagen) is commonly per- & Rasmussen, 2000). However, even with specific
formed as part of the homogenization process for isolation techniques, the chance of contamination or
multiple analysis (e.g., WB and mass spectrometry; protein degradation remains high. It is, therefore,
Wilkinson et al., 2008) as it may be of interest to crucial to assess the relative purity for fractions isola-
determine proteins with specific localizations (e.g., tion by performing a WB for proteins known to be
GLUT4 translocation across the plasma membrane; associated with the desired location [e.g., GAPDH
Miura et al., 2001) or transcription factor DNA and COXIV or cytochrome C for cytosolic and mito-
binding (Girgis et al., 2014); however, successful iso- chondrial fractions, respectively (Dimauro et al.,
lation may require additional buffer components 2012; Beltran Valls et al., 2014)]. Publications using
(discussed previously). For skeletal muscle, as a first such techniques should be expected to provide good
step, soluble proteins within the sarcoplasm are iso- evidence of fraction(s) purity.
lated and separated from insoluble (in standard WB Additional sample processing by immunoprecipi-
buffers) myofibrillar fractions. The sarcoplasmic tation (IP) may be desirable before SDS-PAGE as it
fraction may be subject to further organelle separa- allows the investigation of potential protein–protein
tion through differential centrifugation and isolation interactions (Crossland et al., 2013), and concentra-
methods (utilizing fraction-specific buffers) into tion of proteins of low abundance to be extracted for
mitochondrial and cytosolic fractions (Huff-Lone- accurate detection by subsequent blotting (Rasc on
rgan et al., 1995; Dimauro et al., 2012). Commonly et al., 1992). Initially, the primary antibody (1°Ab)
mitochondrial isolation may involve the passing of specific to the desired protein is added to the homog-
the homogenized sample through a Dounce homoge- enized lysate to form an immune complex that binds
nizer as it preserves mitochondrial morphology, min- to protein A or G beads (generally agarose), which
imizing membrane disruption (Dounce et al., 1955). are centrifuged at low speed and the pelleted protein-
Many of these isolation methods are relatively crude antibody complex bound beads removed. Proteins
in nature; therefore, steps need to be taken to assess may then be eluted and utilized for WB (Huang &
the purity and specificity of preparations (e.g., mea- Kim, 2013). Through this, physically interacting pro-
suring the presence or absence of fraction-specific teins will be captured by the target 1°Ab, allowing
proteins). Most protocols require differential cen- their subsequent denaturation and separation by
trifugation techniques (Huff-Lonergan et al., 1995; SDS-PAGE. These interacting additional proteins
Wilkinson et al., 2008), whereby insoluble myofibril- may then be blotted for, providing valuable insight
lar fractions (containing mitochondria) are pelleted into potential protein–protein interactions as their
by higher speed centrifugation (e.g., 11 000 g), with detection would only be possible if bound to the
remaining mitochondrial proteins isolated following originally IP-targeted protein. It is important to note
re-suspension, and a slower speed centrifugation that the 1°Ab may also contaminate the sample as

5
Bass et al.
denatured heavy (50 kDa) and light chains (25 kDa) each batch to validate the approach. Once quanti-
will be present within the blot (Anton, 2008). If the fied, aliquoted samples may be diluted to the desired
resulting blot contains either a strong background or concentration with the addition of appropriate buf-
the band of interest is obscured by the denatured fers, in preparation for sample loading.
chains, the use of label conjugated protein A or G The final step in processing samples requires the
(e.g., Protein-A-HRP) may be used as they bind denaturing (unfolding) of secondary/tertiary struc-
almost exclusively to intact antibodies (i.e., the cho- tures in proteins, allowing separation based on the
sen primary antibody) (Lal et al., 2005) primary amino acid sequence theoretically in accor-
dance to the predicted molecular weight. To do this,
standard concentrations of samples (acquired as
Protein quantification and gel loading
above) are mixed with Laemmli buffer (Laemmli,
Following protein extraction, each sample requires 1970), the composition of which serves a number of
the standardization of total protein loading per well. important functions. Thiol containing cysteine resi-
Quantification of protein content may be determined dues form disulfide bonds, which govern protein
through colorimetric (Bradford, 1976) or UV absor- folding and stabilize the secondary/tertiary structure
bance (280 nm) (Desjardins et al., 2009) methods. of proteins (Creighton, 1988). The addition of a
The Bradford assay utilizes a colorimetric change of reducing agent, typically b-MCE (DTT or TCEP
Coomassie Brilliant Blue G-250 from 465 nm (tris(2-carboxyethyl)phosphine) may also be used)
(brown) unbound to 595 nm (blue) when bound to cleaves disulfide bonds destabilizing the secondary
protein (Bradford, 1976), with protein concentration and tertiary structure, unfolding the protein (Anfin-
being proportional to the absorption at 595 nm with sen, 1973). Due to the volatility of b-MCE, its addi-
reference to a standard curve of known concentra- tion should occur immediately before use.
tions (typically 100–1500 lg/ml). This procedure is Furthermore, high concentrations of b-MCE or
simple to perform and requires only basic spec- other denaturing agents (i.e., DTT) can interfere
trophotometric equipment; furthermore the reaction with protein assays and therefore should be added
is rapid (~2 min) and the bound product is stable for post protein quantification (Krieg et al., 2005).
~1 h at room temperature (Bradford, 1976). A num- Finally, to allow separation through the application
ber of other methods based along similar biochemi- of an electrical current, all protein R-groups (func-
cal colorimetric properties such as the Lowry, tional amino acid groups) are coated with negative
bicinchoninic acid, and ortho-phthaladehyde assays charges through addition of SDS. As SDS binds to
are also regularly reported for protein concentration the primary structure of proteins (1.4 g per 1 g of
measures in WB procedures (Noble et al., 2007). protein), the overall charge of the protein becomes
While popular, colorimetric assays have a number of relative to its molecular weight, and it is this that
potential disadvantages in that they require more forms the basis of the established separation of pro-
sample than other modern methods (i.e., UV absor- teins through a polyacrylamide gel matrix (Smith,
bance) and are susceptible to pipetting errors of 1984).
either the samples or during standard curve con- Typically 10–100 lg of total cellular protein per
struction, along with suffering from interference by a lane is loaded (Taylor & Posch, 2014); however, this
number of buffer/tissue components [e.g., b-mercap- is generally within precast gels and will be deter-
toethanol (b-MCE)] or hemoglobin (Doumas et al., mined by multiple factors. These will include the
1981). Measuring protein content through UV thickness of the gel, and the lane width, requiring a
absorption with the use of micro-spectrophotometric greater volume of sample. Nonetheless, the final con-
(e.g., Nanodrop) equipment may be more suitable centration of protein to detect a given antigen should
due to the comparatively small sample volume (0.5– be determined by the end user in accordance to
2 ll), wide quantification range (0.1–3000 lg at detection efficacy (i.e., via running titrations of pro-
280 nm), and potential for complete sample recovery tein). Although it is highly dependent on the protein
once the measurement is complete (Desjardins et al., of interest, it is common for protein concentrations
2009). This procedure utilizes the absorbance of UV of human muscle biopsies to be adjusted to 1–2 lg/
light at 280 nm by amino acids containing aromatic ll, with a total of 15–30 lg protein per lane being
rings (i.e., phenylalanine, tyrosine, and tryptophan) typically loaded (Franchi et al., 2014). Proteins of
allowing accurate quantification (Layne, 1957). lower abundance within the sample may need
Erroneous measurements may occur if the sample increased quantity to be loaded; for example, the
comprises both insoluble and soluble proteins that vitamin D receptor (VDR) is highly expressed within
will affect absorbance measurements, demonstrating the kidney, whereas within skeletal muscle, it is rela-
the importance of sample homogeneity. Moreover, tively low requiring a greater amount to be loaded
to ensure reproducibility, standard curves of known (i.e., ~60 lg; Girgis et al., 2014). This can be
protein concentrations should be performed with achieved by concentrating the sample (i.e.,

6
Technical aspects of Western blotting
evaporation or protein precipitation) or utilizing gels protein (e.g., mTOR ~289 kDa), a lower concentra-
with larger sample wells. tion gel (e.g., 7.5%) may be required for optimal res-
In order to have good resolution and identification olution. Alternatively gradient gels (e.g., 4–12%)
of the band(s) of interest, proteins are separated by provide uniform resolution across the molecular-
their mass (in accordance with their primary struc- weight spectrum (Rath et al., 2013). Other gel
ture/AA sequence), as such it is essential to include types such as agarose may be used, but are less
protein standards containing a mixture of predefined common as they are predominantly used for very
proteins of known molecular weight markers to con- large molecular-weight proteins (e.g., titin isoforms
firm the band of interest is the correct mass (Weber 700–4200 kDa) giving superior separation when
& Osborn, 1969). Such standards are generally compared to polyacrylamide gels (Warren et al.,
loaded into the first and last lane of a gel. The choice 2003). Agarose gels may be hand-cast but require
of standard will be dependent on the resolution storage at 4°C to prevent drying out. Other hand-
required near a molecular-weight region, along with cast gels may also require use soon after casting and
potential analysis requirements, such as confirmation may vary between runs due to the short shelf life of
of the size of protein targets. Standards may already some chemicals. The choice between commercial and
be pre-stained for ease of visualization of separation hand-cast gels is generally the preference of the user,
and confirmation of an effective transfer onto mem- but commercial gels are generally more consistent.
branes. Standards may also provide confirmation of Ultimately, the concentration of the cross-linking
either 1°Ab or 2°Ab binding as they can contain molecules and the molecular weight of the protein(s)
known binding sites for these targets and therefore of interest are the determining factors for gel choice
should produce detectable bands, e.g., a purified pro- as taken together these will allow for efficient migra-
tein of interest. The inclusion of such internal stan- tion and optimal band resolution. The choice of gel
dards allows the optimization of subsequent processes concentration and composition is mainly determined
through confirmation of antibody binding (Mahmood by the molecular weight of the protein(s) of interest,
& Yang, 2012). Therefore, SDS-PAGE systems may as it will allow efficient migration and optimal band
be tailored to ensure optimal separation and resolu- resolution.
tion of the desired protein targets depending on their Whereas electrophoresis of nucleic acids utilizes a
molecular weight. Further to this, the inclusion of constant pH within the buffer and gel to achieve dis-
specific molecular weight standards may be used in cernable separation (Westermeier, 2005), protein
conjunction with additional methods of confirmation samples require a discontinuous buffer system (Orn-
of sufficient separation (e.g., use of stain-free gels or stein, 1964). Discontinuous systems utilize gels sepa-
membrane staining, discussed below). rated into two regions, comprising a “stacking gel”
above a “resolving or separating gel” with larger and
smaller pores, respectively. Discontinuous systems
Polyacrylamide gel electrophoresis (PAGE)
are designed to focus protein samples and allow clear
Following these sample preparation steps, PAGE is resolution of proteins. Initially, proteins migrate
performed to separate out denatured and negatively quickly through the large pore stacking gel until they
charged proteins based on their molecular weight. reach the resolving gel, whereupon the smaller pore
Separation of protein samples within polyacrylamide size slows migration, causing the proteins to stack
gels occurs due to the frictional resistance of a pro- together into compact bands (Ornstein, 1964). The
tein as it migrates through pores formed between second principle utilizes the electrophoretic migra-
polymer chains within the gel (Ornstein, 1964). Poly- tion of both ions and proteins through a pH-buffered
acrylamide gels comprise polymerized acrylamide solution. Chloride ions present within the gel have a
monomers along with cross-linking N,N0 -Methylene- higher mobility and therefore migrate faster than
bisacrylamide monomers (Raymond & Weintraub, denatured proteins, establishing a leading ion bound-
1959), creating uniformly sized pores, dependent on ary (Ornstein, 1964). Within traditional Tris-HCl
both monomer concentration and cross-linker ratio. stacking gels (pH 6.8), a trailing boundary of glyci-
As an electrical current is passed, proteins move nate ions form behind migrating proteins due to
through pores within the gel structure; as such, alter- reduced mobility at a low pH (Walker, 1994). Ulti-
ing the bis-acrylamide concentration regulates pore mately, migrating proteins are sandwiched between
size and consequently the ability of larger proteins to the two ion boundaries, stacking the sample into
migrate. Gels with a higher acrylamide concentration tightly focused bands, whereupon they migrate into
(e.g., 20%) impede the movement of larger proteins the resolving gel containing smaller pores, slowing
to a greater degree than those of a smaller molecular the progression of proteins dependent on their size as
weight but better resolve those of lower molecular previously mentioned. Tris-HCl resolving gels are
weights (e.g., 4EBP1 ~20 kDa) (Chrambach & Rod- typically formed at pH 8.8; this higher pH allows the
bard, 1971). Similarly, if the desired target is a large ionization of the trailing glycinate, increasing its

7
Bass et al.
mobility (Ornstein, 1964). Consequently, both chlo- proteins (discussed later in stripping and re-probing).
ride and glycinate boundaries will migrate past the The development of more durable hydrophobic
protein samples, no longer constricting them into PVDF membranes allows the possibility to strip and
focused bands, allowing the unimpeded separation re-probe, as they are more chemically inert and
of proteins. Alternative gels and discontinuous buf- robust (Kurien & Scofield, 2006). PVDF membranes
fer systems are available that may be better suited to bind proteins through hydrophobic interactions
the resolution of either larger or smaller molecular- (MacPhee, 2010) and are capable of binding greater
weight proteins depending on sample composition. amounts of protein (~150 lg/cm2) than other sub-
For example, Bis-Tris systems utilize different buf- strates (Matsudaira, 1987). Although the use of
fers containing either 3-(N-morpholino)propanesul- PVDF membranes may be advantageous in captur-
fonic acid (MOPS) or 2-(N-morpholino) ing greater amounts of protein, membrane–antibody
ethanesulfonic acid (MES), which function as trail- interactions are more likely to occur, generating
ing boundary ions instead of glycinate (Hachmann & higher backgrounds when exposing the blots, conse-
Amshey, 2005). These allow greater resolution of quently increasing the importance of performing
mid-sized (~75 kDa) or smaller (<36 kDa) proteins thorough wash steps (Mahmood & Yang, 2012). The
with MOPS or MES, respectively. Bis-Tris gels are hydrophobic nature of PVDF membranes requires
cast at pH 6.8, offering significantly longer shelf life an initial pre-soaking in methanol to allow the infil-
compared with Tris-HCl gels as they do not undergo tration of the buffer and the binding of proteins
acrylamide hydrolysis due to their acidic nature. (Mansfield, 1995). Another variable to consider is
Generally, electrophoresis is undertaken using a con- the choice of pore size (i.e., 0.45–0.025 lm) of the
stant voltage, rather than a constant current, due to membrane, as this will affect the binding of larger or
the linear relation of protein migration and voltage. smaller proteins (Burnette, 1981; Tovey & Baldo,
As current is dependent on the voltage and resis- 1987). Smaller proteins, such as cytochrome C
tance, a constant current will not control protein (12.5 kDa), have been shown to have a reduced
migration as changes in resistance (i.e., warming of binding capacity upon membranes with a pore size
the buffer) will cause the voltage to fluctuate. of 0.45 lm (Burnette, 1981). Thus, to probe for
Depending on the apparatus, electrophoresis is typi- smaller molecular weight proteins (<20 kDa), a
cally performed for 60 min with a constant voltage smaller pore size membrane is advisable as this will
of 200 V to give a suitable separation of protein bind larger quantities of protein. It is important to
lysates; however, less time may be required for smal- note that protein transfer onto PVDF membranes
ler molecular-weight proteins. may be inhibited by high SDS concentrations
(Mozdzanowski et al., 1992), so prior to transfer gels
should be thoroughly washed in distilled water (e.g.,
Electrotransfer
2 min for thin gels), and then equilibrated in transfer
Following the separation, the proteins are elec- buffer (e.g., 5 min) to remove excess SDS.
trophoretically transferred to a membrane (elec- Methods of transfer are numerous. Traditional
troblotting; with high affinity for protein), thereby wet transfers involve successive layers of a cassette
immobilizing the separated proteins, allowing subse- backing, fiber pads, blotting paper, the polyacry-
quent probing with antibodies, and providing addi- lamide gel, the chosen membrane, blotting paper,
tional durability compared to gels (Towbin et al., and the cassette front being “sandwiched” together
1979). Utilizing the same principle as PAGE, the and submerged between an anode and cathode
negatively charged proteins in the gel are transferred within a transfer tank (Towbin et al., 1979; Burnette,
across onto the membrane when a lateral electric 1981). The transfer process as described by Burnette
current is applied while immersed in a buffered solu- (1981) was achieved over 22 h; however, this dura-
tion (termed wet transfer). The membrane is placed tion is dependent on the size of the protein(s) of
directly upon the gel ensuring a mirror image trans- interest. Insufficient transfer time will result in weak
fer of proteins occurs. Proteins are typically trans- signal or no intensity, as significant quantities of pro-
ferred onto nitrocellulose or polyvinylidene teins will remain within the gel. Conversely, excessive
difluoride (PVDF) membranes; originally nitrocellu- transfer time will generate a poor signal as proteins
lose membranes were used for their immediate bind- pass through both gel and membrane; however,
ing and immobilization of proteins (Towbin et al., PVDF membranes generally have smaller pores com-
1979; Burnette, 1981). As nitrocellulose membranes pared with nitrocellulose, reducing the amount of
are not hydrophobic, they are easily hydrated for use protein passing through. As with electrophoresis, the
within wet or semi-dry transfers. However, nitrocel- rate of protein movement is dependent on its molec-
lulose membranes can be fragile, making them ular weight, and thus smaller proteins may require
incompatible with the current commonplace prac- less time for transfer. Gel staining allows the evalua-
tices of stripping and re-probing for alternative tion of transfer efficiency, through visualization of

8
Technical aspects of Western blotting
protein remaining within a gel (e.g., using Coomas- complete transfer of proteins, accurate quantitation
sie/silver/Ponceau stains or stain-free gels), removing and analysis will be compromised.
some aspects of transfer uncertainty (Colella et al.,
2012). Other transfer techniques such as semi-dry
Blocking
transfers are frequently utilized as they can offer fas-
ter transfer times (reduced to just over an hour), Although one of the simplest technical steps to per-
depending on individual requirements or time con- form in the WB process, blocking is important as it
straints (Kurien et al., 2015). Most of the conven- can prevent non-specific binding of antibodies (1°Ab
tional commercial systems are easy to use, reliable and/or 2°Ab) to the membrane (Jensen, 2012). As
for transfer of proteins of a wide range of sizes, and membranes have a high affinity for binding proteins
use relatively inexpensive reagents. Still faster trans- and therefore antibodies, blocking reduces back-
fers may be attainable using recently developed sys- ground in subsequent steps. Different solutions may
tems such as the Trans-Blot TurboTM Transfer be used for this stage of the protocol, each having
System, but these may not work well with some pro- their own benefits and limitations. Non-fat-dried
teins (i.e., <100 kDa) and require more expensive milk diluted in Tris Buffer Saline Tween-20 (TBST)
consumables. is often used and is cheap and widely available, and
The composition of the transfer buffer will also milk proteins are not, however, compatible with all
dictate the efficiency of transfer, especially for pro- antibodies. For example, it has been reported that
teins of a high molecular weight. Generally, wet bovine serum albumin (BSA; normally 5%) should
transfer solutions are buffered to pH 8.3 and con- be preferentially used for biotin and anti-phospho-
tain Tris-base along with glycine (Towbin et al., protein antibodies as milk contains casein, which is a
1979). It is important to note that the buffer pH phospho-protein, and biotin, rendering it likely to
should not be adjusted through the addition of interfere with the WB as the phospho-specific anti-
acidic or basic solutions, as this will result in higher body may cross-react with the casein present in the
conductivity through greater ion content, thus milk (Mahmood & Yang, 2012). Awareness of this
increasing the temperature of the solution and issue is ever more relevant with the continued devel-
potential background interference and/or inefficient opment of numerous specific, non-cross-reactive
transfer. These buffers may be pre-chilled to prevent anti-phosphoamino acid antibodies. Although these
overheating, due to the high current, and the defor- antibodies greatly enhance phosphoamino acid-spe-
mation of the membrane, with transfer tanks com- cific analysis and negate the need for radioactive
monly containing ice blocks. Methanol was used reagents, i.e., 32P, high backgrounds have been
within the original transfer buffers (Towbin et al., observed when these antibodies are used during
1979; Burnette, 1981) as it increases the ability of immunoblotting (Michalewski et al., 1999). Possible
membranes to bind proteins and prevents gel swel- explanations for this technical challenge include the
ling. However, since the inclusion of methanol existence of phosphorylated proteins in various
within these buffers may decrease the transfer of lar- blocking solutions (as alluded to above) or inappro-
ger molecular-weight proteins (>100 kDa) as it priate membrane-blocking conditions. Work by
reduces the pore size of within gels (MacPhee, Michalewski et al. reported that the binding of anti-
2010), it may be advisable to exclude methanol phosphoamino acid antibodies to proteins and mem-
when probing for larger proteins. For the majority branes critically depended on blocking conditions,
of proteins analyzed via WB techniques, low-ionic with a combination of amicase (5%), BSA (5%), and
strength buffers and low electrical currents are opti- membrane-blocking agent (5%) to be most effective
mal (Alegria-Schaffer et al., 2009). If these generic in reducing non-specific binding (Michalewski et al.,
rules are followed, then successful transfer is highly 1999).
likely. Reversible staining of the membrane and/or For the preparation of both milk and BSA, a 2.5–
gel with Ponceau, e.g., will reveal protein loading, 5% weight: volume solution is most commonly used
commercial systems for reversible staining also exist (Crossland et al., 2013; Franchi et al., 2014), in
and have been extensively validated for this purpose TBST or phosphate-buffered saline (PBS). The mix-
(Antharavally et al., 2004; Alegria-Schaffer et al., ing must be thorough and the solution should then
2009). Unequal loading will be displayed as notice- be filtered to prevent grains contaminating the blot
able differences in multiple protein band(s)/lane(s) during development. A further (albeit less common)
intensities, in this instance the variability may be option for blocking is using highly purified non-ani-
due to unequal sample protein quantities caused by mal proteins (e.g., isolated casein), although these
different sample concentrations, or mistakes in the are much less cost-effective. Appropriate guidance
sample volume loaded into individual lanes. Protein on blocking conditions is usually provided by com-
lysate concentrations should be reanalyzed to deter- mercial companies alongside the antibody-specific
mine if it was the former. Without efficient and information; however, because not all blocking

9
Bass et al.
conditions are suitable for all target proteins, valida- 1 h, whereupon the background levels are assessed
tion and testing for each individual protein of inter- and concentrations/blocking agent subsequently
est is always recommended (Spinola & Cannon, altered. No single blocking agent is ideal for every
1985). Whichever approach is chosen an optimal WB assay as each antigen-antibody pairing has
blocking, buffer should improve the sensitivity of the unique characteristics. Consequently, the chosen
WB by reducing background and improving the sig- blocking strategy must be optimized for each individ-
nal-to-noise ratio (measured as the signal obtained in ual application, with the above considerations being
a sample containing the target analyte vs that kept in mind.
obtained with a sample without the target analyte).
Ideally, a blocking agent will bind to all sites of non-
Primary antibodies and determining specificity
specific interaction, eliminating all background with-
out altering access/interaction to the protein(s) of The principle of the WB is the detection of protein(s)
interest for antibody binding. The choice of blocking through the binding and recognition of antibodies
agent should be based on the antigen itself and the (Ab) to one or more targets; this interaction should
type of 2°Ab conjugation (see below). For example, be highly specific between a portion of the antigen
in assays where alkaline phosphatase conjugates are (protein) or epitope and the specific recognition sites
used, TBST should be selected, as PBS will interfere found on the fragment antigen-binding (Fab) region
with these phosphatases due to the presence of of the antibody termed a paratope (Kurien et al.,
sodium phosphate (and in some solutions potassium 2011) (Fig. 1). The 1°Ab should be thoroughly
phosphate also). If PBS is to be used for intermediate assessed and validated to be specific and sensitive
steps, the membrane should be sufficiently washed in enough to detect the intended target protein. It is
TBST to remove excess sodium phosphate before important to check that the antibody is specific
addition of the substrate. toward the native or denatured protein, as the dena-
It is not only the choice of blocking agent that can turing treatment of protein samples prior to SDS-
affect WB outcomes; volume of blocking agent and PAGE may alter the exposure and availability of the
indeed the incubation period for blocking (which epitope, affecting antibody binding affinity. In some
may vary from an hour to overnight; Gershoni & cases, it may be necessary to use “native-specific”
Palade, 1983) are also important factors. Too little monoclonal antibodies (Tino et al., 2000). The tar-
blocking agent (low concentration) or too short an geted peptide sequence may be available from the
incubation period will increase the potential for non- supplier to allow confirmation of specificity and
specific binding of the 1°Ab to the membrane-bound region of binding; however, occasionally this may be
proteins which could result in excessive background unavailable proprietary information. Traditionally
and/or reduced signal-to-noise ratio. Conversely, an 1°Ab are produced through immunization of the
incubation period that is too long and/or uses exces- host using purified target proteins, whereas modern
sive blocking agent may interrupt antigen-antibody approaches utilize synthetic peptides, often produc-
interactions, also causing a reduction in signal-to- ing Ab toward short denatured 8–10 amino acid
noise ratio (Mahmood & Yang, 2012). In a compara- sequences. Isolation and purification is generally
tive study assessing the efficacy of commonly used achieved through affinity chromatography isolating
blocking protocols to minimize non-specific back- antibodies and small proteins and/or anion-exchange
ground and promote immunoreactivity of antibodies filtration depending on the class (i.e., IgG, IgM)
(monoclonal, polyclonal or biotin-conjugated) (Clezardin et al., 1986). Predicted and confirmed
against a phospho-amino acids, the authors recom- species cross-reactivity information is often only
mended the use of a solution composed of 5% BSA, available through the vendor; however, binding will
5% Amicase (Sigma; a mixture of free amino acids entirely depend on the antigen region. When choos-
with virtually no unhydrolyzed peptides and minimal ing a 1°Ab, there may be multiple forms available
inorganic components), and 5% membrane-blocking from different vendors, ideally each would bind to a
agent in Tris-buffered saline containing 0.05% unique antigen upon the protein of interest, allowing
Tween 20 [to aid in the removal of residual SDS accurate assessment of the protein’s abundance.
from earlier steps (Zampieri et al., 2000)] for 45 min However, this is often not the case, and assessment
at room temperature to achieve good-quality low of the previous literature utilizing that antibody is
background WB (Michalewski et al., 1999). As with strongly advised. Some proteins may be orthologous
most common blocking agents (Gershoni & Palade, and contain the same or similar sequence to other
1983), incubation for longer than 1.5 h led to species. Thus, if a 1°Ab is specific to an epitope with
reduced signal strength (signal to noise), with 30 min this sequence, it may be used to probe other species
or less resulting in unacceptably high backgrounds. (i.e., GAPDH 1°Ab may be used on human, rat, and
Therefore, in our experience, initial optimization is mouse tissue). Depending on the protein of interest,
undertaken using a 5% milk solution in TBST for extensive testing of multiple antibodies may have

10
Technical aspects of Western blotting
already been undertaken, allowing the most suitable However, the assessment of a bands molecular
antibody to be selected. For example, the VDR has weight may not always be suitable, as some proteins
low expression within skeletal muscle, and in order may migrate to a non-predictable region. For
to find a 1°Ab capable of detecting it by WB and instance, the mTOR regulator REDD1 has a pre-
immunofluorescence, extensive validation of a panel dicted molecular weight of 25 kDa; however, it is
of multiple 1°Abs was required (Wang et al., 2010). detectable at 35 kDa due to multiple lysine residues
The identification of a highly specific VDR 1°Ab (D- (increased positively charged residues) (Chang et al.,
6; Santa Cruz Biotechnology, Cambridge, UK) was 2009). This highlights the importance of knowing the
confirmed and is believed to be the most representa- migrating properties of the target protein, and if
tive. Assessment of new antibodies for a target anti- unexpected bands occur, literature investigation may
gen requires even more careful testing, including the be required.
use of a variety of appropriate positive and negative In order to validate the specificity of a new 1°Ab,
controls (discussed below). it should be tested against a positive lysate or puri-
Specificity and performance of the 1°Ab antibody fied protein control, giving a detectable band at the
is also dependent on whether it is monoclonal (mAb) correct molecular weight and a negative sample from
or polyclonal (pAb). Both have disadvantage/advan- a tissue known not to express the intended target
tage; pAb are produced from differing B-cell lin- [The Human Atlas provides reliable protein expres-
eages, recognizing multiple epitope regions on an sion data (http://www.proteinatlas.org)], resulting in
antigen. They are generally more cost-effective (Lip- no detectable band. Sometimes, it may be appropri-
man et al., 2005) and provide more antibody mole- ate to include a specific knock-in/out (e.g., via
cules that can target the protein of interest, shRNA or siRNA) sample to allow confirmation of
producing potentially a greater level of sensitivity a target within the same tissue type. For example,
upon analysis (MacPhee, 2010). However, their overexpression of AKT isoforms (an essential signal-
specificity can also be compromised, due to greater ing protein for muscle hypertrophy/atrophy) within
possibility of non-specific binding (MacPhee, 2010). rat skeletal muscle following shRNA produced
In contrast, mAb provide highly consistent and detectable bands at the predicted molecular weight
specific binding to a specific and known epitope on (~40 kDa) compared with control samples (Cleasby
an antigen, as they are produced from a single cell et al., 2007). Conversely, knockdown of AKT, again
lineage, raised against a single specific epitope (Lip- within skeletal muscle, demonstrated a reduction in
man et al., 2005; MacPhee, 2010). Yet binding affini- band intensity at the same molecular weight com-
ties of mAb can suffer if the epitope structure is pared with control samples. Similarly protein inhibi-
affected in any way through denaturing or elec- tors (e.g., LY294002, rapamycin, etc.) known to
trophoresis for example (Lipman et al., 2005). block specific phosphorylation pathways can be
Depending on the primary amino acid sequence of used. For example, the addition of LY294002 to cul-
the target protein, similar epitopes may be present tured cells inhibits PI(3)K, resulting in decreased
within degradation products or alternate isoforms, phosphorylation of down-stream intermediates (i.e.,
potentially presenting additional bands. Degradation AKT/P70 S6K1) (Rommel et al., 2001). Thus, if
products will migrate ahead of the band of interest, probing for phosphorylated P70 S6K1, cultured L6
due to the decreased molecular weight. 1°Ab affini- cells treated with/without insulin and LY294002 will
ties toward alternative isoforms may not interfere provide a robust positive (greater band intensity)
with data interpretation if the bands are sufficiently and negative control (reduced intensity), respec-
separated (i.e., have different molecular weights). tively, compared with untreated samples. In this
For example, certain antibodies toward P70 S6K1 instance, the inclusion of an untreated sample along-
(70 kDa), a critical protein in the mRNA transla- side an inhibited negative control will confirm the
tional initiation pathway and one of the most probed reduction in band intensity is in fact due to decreased
of all in the muscle and exercise field, may bind to expression, rather than a loss of detection. Despite
the isoform P80 S6K (80 kDa). P80 S6K encodes a rigorous testing of antibodies with appropriate posi-
nuclear localization signal and contains an addi- tive/negative controls, additional bands may still be
tional 23 amino acids and would be present above present or insufficiently separated making identifica-
P70 S6K1 when blotted (Thomas, 1993). Nonethe- tion and quantitation of the correct band (if present)
less, sufficient electrophoretic separation between the unreliable. Absolute confirmation of the presence of
two isoforms and appropriate controls may allow a protein in a given band may be achieved by mass
correct identification. For example, insulin-treated spectrometry via determination of the peptide
L6 myotubes increased P70 S6K1 phosphorylation sequence (Trauger et al., 2002). Briefly, the band of
(Somwar et al., 1998) but not P85 S6K allowing, in interest is excised and the mixture of proteins
this instance, identification of the correct band; digested by trypsin into small peptide sequences cap-
nonetheless, this does not guarantee specificity. able of being sequenced by liquid chromatography–

11
Bass et al.
mass spectrometry (LC-MS/MS). Utilizing this control samples, demonstrating increased protein
approach, however, requires access to highly specific ubiquitination, suggestive of increased proteolysis.
and costly equipment and technical expertise, but This assessment of ubiquitination needs to be accom-
can provide validation of antibody specificity. Ulti- panied by separate measures of proteins involved
mately, positive and negative controls will help within the proteolytic process, such as FoxO3 and
establish the degree of non-specific binding and AKT, confirming (or otherwise) increases and
potential false-positive bands, along with the confir- decreases in phosphorylation levels in atrophic mus-
mation of increased/decreased protein expression, cle samples (Zhao et al., 2007). This same principle
giving confidence that the highlighted band is indeed of detecting the incorporation of a label onto multi-
the correct one. ple proteins permits the monitoring of the translation
process, e.g., using puromycin in cell culture or pre-
clinical models (Schmidt et al., 2009). Here, puromy-
Post-translational modifications
cin, a structural analog of aminoacyl tRNAs, is
Crucial for determining the relative importance of incorporated into newly synthesized proteins, which
potential signaling mechanisms is the ability to detect are subsequently probed using an anti-puromycin
PTMs such as the alterations in reversible phosphory- 1°Ab. Measuring lane intensity provides a semi-
lation states of various proteins (phosphoproteins). quantitative assessment of global protein synthesis
The phosphorylation of a protein will alter multiple (Schmidt et al., 2009). As previously mentioned,
aspects of its interactions, including localization, known signaling intermediates (i.e., AKT/P70 S6K1)
conformational shape, hydrophobicity, and activity need to be assessed to ensure that changes to puro-
(Polyansky & Zagrovic, 2012). Phosphospecific-anti- mycin incorporation are matched to increased ana-
bodies capable of distinguishing between short epi- bolic signaling (Crossland et al., 2013). Ultimately, a
topes containing phosphorylated or unphosphorylated 1°Ab may be specific for a single phosphorylated
amino acid residues, typically Thr, Ser, and Tyr have protein or for a specific PTM component such as
been developed (Nairn et al., 1982). Proteins may also ubiquitin that may be present within multiple pro-
be phosphorylated at multiple loci through different teins. Thus, even within the same sample, assess-
signaling mechanisms, and thus it is important to ments of signaling cascades (phosphorylation),
choose the correct phosphorylation site, depending on protein degradation (ubiquitination), or protein syn-
the pathway or response of interest. For example, the thesis (puromycin incorporation) may be made,
activity of eukaryotic translation initiation factor 4E allowing a comprehensive investigation of protein
(eIF4E), a subunit of eIF4F (eukaryotic translation metabolism. Further PTMs may be assessed by WB
initiation factor 4F), crucial for the initiation of pro- such as glycosylation of a protein, which has a sub-
tein synthesis is regulated through the phosphorylation stantial impact upon multiple aspects, including pro-
of 4E-BP1 (eIF4E binding protein 1). 4E-BP1 is phos- tein folding, conformational changes, and stability
phorylated by FRAP/MTOR at Thr37/46, but along with solubility (Rudd & Dwek, 1997). The two
requires additional phosphorylation at Ser65 and major forms of glycosylation are serine/threonine O-
Thr70 to disassociate from eiF4E (Gingras et al., linked or asparagine N-linked carbohydrates. This
1999). Simply measuring changes in Thr37/46 phos- will increase the molecular weight of the glycopro-
phorylation may be inadvertently misleading, as addi- tein, and the degree of glycosylation can be assessed
tional phosphorylation may be required for activation by initially treating samples with a endoglycosidase
(Gingras et al., 1999). In this case, both phosphoryla- (e.g., PNGase) to remove N-linked glycans. As a
tion sites (i.e., 37/46 and 65/70) should be probed for result of removing the carbohydrate moieties, addi-
when investigating changes in potential signaling tional bands will be detected at a lower molecular
mechanisms of protein synthesis. weight (Pere-Brissaud et al., 2015).
A myriad of other PTMs have important roles on
various physiological states including but not limited
Secondary antibodies
to ubiquitination (Paul et al., 2012), glycosylation
(Pere-Brissaud et al., 2015), and methylation Secondary antibodies (2°Ab) are required for the
(Voelkel et al., 2013), and changes may be assessed indirect detection of a target antigen bound by a
via WB. For example, starvation-induced skeletal 1°Ab. Typically 1°Ab will not be conjugated to a
muscle atrophy and protein degradation are associ- label for detection, resulting in the need for 2°Ab
ated with an increase in ubiquitinylation of proteins (conjugated to a reporter function, e.g., horseradish
(Paul et al., 2012). This may be assessed using a peroxidase [HRP], specific fluorophores) capable of
1°Ab targeting ubiquitin that will bind to multiple binding to the fragment crystallizable (Fc) region on
ubiquitinated proteins throughout a blot. Thus, in a 1°Ab, allowing subsequent detection by a camera
samples from starved or atrophying muscle, the band or imaging device (discussed below). The crucial
intensity of the lane will be increased compared with aspect of utilizing a 2°Ab is the ability to amplify the

12
Technical aspects of Western blotting
detectable signal since multiple 2°Ab can bind to a solutions are typically made up within the same buf-
single 1°Ab, thus amplifying the detection of low fer and blocking solutions as outlined previously,
abundant proteins. The choice of 2°Ab will initially but may require optimization if a high background
depend on the 1°Ab isotype and which animal it was occurs. Unlike conditions for 1°Ab, secondary incu-
raised within (Lipman et al., 2005). Variations bations are generally undertaken at room tempera-
within Ab heavy chain formations will determine ture for a shorter amount of time, typically 1 h.
their function and class; antibodies from mammals Occasionally, longer incubations may be required;
will be one of five classes, IgM, IgD, IgE, IgA, or however, this can lead to increased non-specific bind-
IgG, with the latter being subdivided into multiple ing within the membrane, generating a higher back-
subclasses within rats, mice, and humans (i.e., IgG2a, ground. For ease of use, in our experience, altering
IgG2b) (Lipman et al., 2005). Primary Ab raised antibody concentrations for different targets, while
within mice or rats may require isotype-specific maintaining incubation times, is preferable.
2°Ab; however, rabbits produce only a single isotype
of IgG, allowing broad specificity 2°Ab to be utilized
Detection
(Manning et al., 2012). For example, an IgG 1°Ab
raised within a rabbit simply requires an anti-rabbit The general principles of detection for WB are the
IgG, whereas a mouse IgG2a would require a 2°Ab same as for other antibody-based assays, such as the
specific for the IgG2a isotype. As rabbit 1°Abs suitable enzyme-linked immunosorbent assay (ELISA) (Vol-
for WB purposes are common, this means a single ler et al., 1978). In general terms, the 2°Ab is conju-
anti-rabbit IgG 2°Ab may serve for multiple targets. gated with a labeled compound (i.e., radio-isotope or
However, the use of 1°Ab from different species fluorophore) or enzyme that permits subsequent
allows the unique potential for multiplex detection detection. Historically, this was a radioactive isotope
using 2°Ab conjugated to different fluorescent wave- or enzyme exposed against X-ray film (Miura et al.,
lengths for multiple detections upon the same blot 2001) but now is normally an enzyme or a fluo-
(discussed below) (Gingrich et al., 2000). For exam- rophore detected by camera (Crossland et al., 2013).
ple, a rabbit IgG 1°Ab specific toward AKT (60 kDa) The two most commonly employed enzymes are
may be probed for along side a mouse IgG2a 1°Ab for alkaline phosphatase (AP) (Bronstein et al., 1989)
P70 S6K1 (70 kDa) using a red fluorescent anti-rabbit and HRP (Kricka, 1991). Both AP and HRP can be
IgG (594 nm) and green fluorescent anti-mouse IgG2a used for colorimetric or chemiluminescent detection.
(488 nm). An additional key aspect is the ability to When employing HRP, it is important to remember
simultaneously probe for both total and phosphory- that sodium azide inhibits this enzyme so solutions
lated expression levels (e.g., pan-AKT vs AKT like those used for blotting and detection should not
Ser473) providing 1°Ab are raised in differing species contain sodium azide, or membranes should be suffi-
and the epitope regions do not overlap (Georgopoulos ciently washed before exposure. In colorimetric
et al., 2010). The ratio between these measures gives detection (Babson et al., 1966), a substrate (e.g., 3,30 -
an indication of a proteins capacity for signal trans- diaminobenzidine) of HRP is oxidized producing a
duction (total) vs activation (phosphorylation), as brown insoluble product. Quantification can then be
discussed below. Ultimately, the use of different spe- achieved by scanning the blot with either a dedicated
cies-specific fluorescent 2°Abs in a single incubation imager or traditional office scanner (although the lat-
potentially allows the measurement of two or more ter is not recommended, as they are not designed to
targets (i.e., total vs phosphorylated or targets of a have a large linear range) and analyzing band inten-
similar molecular weight) within the same blot. sity using freely available software such as NIH
Although 2°Ab are raised toward specific epitopes Image J (http://imagej.nih.gov/ij/). A key disadvan-
upon the 1°Ab, cross-reactivity may occur with other tage of this form of detection is that the chemical
separated proteins or indeed those used for blocking, reaction must be stopped, and therefore, optimal
due to similar peptide sequences (Ramlau, 1987). reaction conditions need to be determined (reaction
Therefore, negative controls should also be under- time, temperature, etc.) prior to quantitation.
taken by omitting the 1°Ab incubation to ensure the With chemiluminescent detection (Kricka, 1991),
observed bands are not the result of non-specific HRP luminol is oxidized in the presence of hydrogen
binding of 2°Ab. The optimal dilution of the 2°Ab peroxide producing 3-aminophthalate, which emits
will depend on the expression level of the target pro- light at 425 nm. A chemiluminescent blot is opti-
tein and the choice of 1°Ab (e.g., GAPDH is highly mally imaged after 3–5 min of incubation with the
expressed, requiring higher dilutions to prevent sig- substrate and may produce a signal for several hours
nal saturation); however, the manufacturer’s recom- (Alegria-Schaffer et al., 2009). A key advantage of
mendation is often a good starting point. A dilution this form of detection is that the blot can be repeat-
curve may then used to optimize the concentration edly rinsed and exposed to substrate and luciferase
needed for the targets being investigated. The 2°Ab to allow multiple exposures, which is useful for

13
Bass et al.
optimization of detection parameters, thereby ensur- generally similar to chemiluminescent, whereas
ing the blot is not underexposed or overexposed. chemiluminescent detection is more sensitive and
However, a potential disadvantage is that the blot produces signal at lower concentrations than colori-
must either be exposed to film, which must then be metric. Second, just as using a labeled 2°Ab boosts
developed, or be placed in a specialized detection sys- the signal compared with using a labeled 1°Ab, use
tem (e.g., Chemidoc, OdysseyTM) with the latter being of a labeled tertiary antibody can boost signal by
the most common and accurate approach. Both providing additional amplification (Delaive et al.,
options for visualization of the blot, however, 2008).
require more expense and equipment than for colori-
metric detection. Quantification of the blot will be
Stripping and re-probing
dependent on the light detection method. If photo-
sensitive film was exposed and developed, subse- The stripping and re-probing of WB membranes pro-
quent scanning and Image J are commonly vides a time-efficient method for determining multi-
employed. More commonly, a specialized detection ple protein targets within a single gel run (Sennepin
system, typically utilizing proprietary software, et al., 2009). It allows a number of different analyses
employing densitometry analysis will be used. The on a single membrane, thus saving time, sample and
proper storage and age of buffers, substrates, stop consumables, and maximizing efficiency. Further-
solutions, and luciferase solutions must similarly be more, there may also be times where a membrane
considered as degradation and/or contamination of requires probing with a 1°Ab to confirm the data
any one of these can affect each of the steps and obtained from the initial analysis of the protein of
potentially prevent detection and/or result in very interest (i.e., a different 1°Ab specific to another epi-
high background signals. It is also important that tope on the same protein) (Kaufmann & Shaper,
the 2°Ab has been properly stored (80 °C, 20 °C, 1992).
4 °C) and is reasonably fresh, as the enzymes will Initially developed in the early 1980s, these meth-
degrade with time. If stored correctly, antibodies ods involved incubation in either urea, b-MCE/BSA
may still be viable for multiple years (Argentieri buffers (Erickson et al., 1982), or highly acidic gly-
et al., 2013); however, suppliers only recommend cine buffers (pH 2.2; Legocki & Verma, 1981) at ele-
storage for approximately a year as extended storage vated temperatures for long periods (up to an hour
will decrease its effectiveness. in some cases), in order to remove the 1°Ab and
These aforementioned limitations can be overcome detection reagent (e.g., enhanced chemiluminescence
using fluorescent detection where the 2°Ab is conju- [ECL]). Ultimately, multiple protein targets could be
gated to a fluorophore. For this type of detection, no detected within a single sample/run, since the 1°Ab
further chemical reactions are required, and the blot have been disassociated from their target proteins,
can be visualized after exposure to 2°Ab and rinsing. allowing other 1°Ab to be added (Yeung & Stanley,
However, it does require further specialized imaging 2009). Since these early experiments, stripping buf-
systems. Fluorescent-based imaging has a greater fers have progressed rapidly and many commercial
upper linear range of detection (250–500 pg) com- preparations exist which claim to be gentler (gener-
pared to chemiluminescent (125 pg) (Gingrich et al., ally consisting of a mixture of SDS, glycine, and
2000). Fluorescent antibodies also have a similar detergents; TBST) and work rapidly (<15 min) at
lower range when compared to chemiluminescent room temperature. However, it is always recom-
detection and so may be more appropriate when mended to check the efficiency of these commercial
investigating higher abundance proteins. A further buffers, by re-exposing the membrane with ECL
benefit of fluorescent antibodies is the ability to uti- reagents or other detection methods post removal of
lize 1°Ab from different species and subsequent dif- the antibody.
ferent wavelength 2°Ab for each to perform Stripping and re-probing is commonly undertaken
multiplexed exposures upon the same blot (discussed to investigate the levels of phosphorylated proteins
previously) (Gingrich et al., 2000). This crucial dif- before assessing the total expression of the same pro-
ference can allow the probing of multiple targets tein (Figueiredo & Nader, 2012), allowing the signal-
with similar molecular weights (e.g., AKT and P70 ing capacity vs activation to be assessed (discussed
S6K1) or PTMs (e.g., pan-AKT vs AKT Ser473) below). In doing so, the generally weaker expressed
using different conjugated fluorophores, specific to phospho-protein is first detected, allowing accurate
different excitation channels (e.g., 594 nm vs measurement, before the total is probed. Despite the
488 nm) (Georgopoulos et al., 2010). When it is obvious benefits of stripping and reprobing of mem-
desirable to improve the sensitivity for the detection branes, these methods are not always foolproof and
of low abundance target proteins, a couple of can have significant limitations. First, care must be
approaches may be employed. First, switching detec- taken as to which protein the membrane is re-probed
tion systems may help as fluorometric detection is for following stripping, as it is difficult to eliminate

14
Technical aspects of Western blotting
detectable signal from highly abundant proteins. expression of the same protein within the same blot
Additionally, if the stripping buffer has failed to is extremely desirable. Therefore, one must consider,
completely remove all antibody and ECL from the when performing WB, whether it is appropriate to
membrane, and the two proteins of interest are of strip and re-probe a membrane, as despite the bene-
similar molecular weights [e.g., GAPDH (molecular fits (phospho vs total protein), the limitations may
weight: 35.8 kDa, but band commonly detected at outweigh the benefit, and consequently the accurate
37 kDa) and ERK 1/2 (molecular weights: 42/ detection and quantification of WB data may only
44 kDa, respectively)], then the signal from the inef- be possible with repeated blots.
fectively stripped antibody may interfere with the
subsequent detection and quantification of the sec-
Analysis
ond target protein. Further to this, stripping of the
membrane cannot be performed indefinitely. As a Analysis of the bands of interest depends on the type
rule of thumb, only three stripping incubations are of detection and the imaging system available, with
recommended, due to loss of the antigen (Sennepin many imaging systems requiring proprietary soft-
et al., 2009). This limits the number of targets that ware for image acquisition and quantification. Each
can be probed from the same membrane but is still software package utilizes slightly different methods
worth doing considering the benefit of measuring for quantification; however, generally peak height or
two or more targets within the identical sample. area is used (Gassmann et al., 2009). During quan-
There are, however, a number of recent develop- tification, it is essential to ensure that the bands of
ments which have aimed to enhance the capacity for interest are within the linear range of detection, as
multiple Ab detection from the same membrane. pixel saturation on imaging sensors may occur
Sennepin et al. (2009) described a technique whereby within highly abundant targets (Mollica et al., 2009);
instead of removing or stripping the HRP activity this is quite a common error and can easily be
linked to the Ab from the membrane, this is instead avoided. However, automatic detection of saturation
irreversibly inhibited with hydrogen peroxide, to is now a standard feature within most imaging pack-
allow up to five different sequential incubations/de- ages. If oversaturation occurs, it may be possible for
tections to be performed on the same membrane the sensitivity of the camera to be reduced (i.e.,
(Sennepin et al., 2009). There are suggestions, how- reduced pixel binning) increasing the image resolu-
ever, that the exposure to such strong oxidizing con- tion (larger image, more pixels) and therefore requir-
ditions may alter certain epitopes, potentially ing more signal to achieve oversaturation. Despite
affecting Ab recognition (Kaufmann, 2001). A simi- this, oversaturated “staining” of the membrane (i.e.,
lar procedure can also be performed using sodium as signified by visible yellowish bands on the mem-
azide, which has been proposed to avoid this epitope brane prior to exposure) during chemiluminescent
issue; however, the method requires lengthy incuba- detection by highly abundant proteins (e.g.,
tions of upwards of 16 h compared to 15 min with GAPDH) may occur requiring the reduction of
hydrogen peroxide, and as sodium azide is known to either sample loading or greater dilution of the 1°Ab.
inhibit HRP detection, it may interfere with subse- Another important consideration for quantifica-
quent detection (Kaufmann, 2001). As previously tion is the choice of single band or whole lane boxed
discussed, both colorimetric and chemiluminescent analysis, with the former suitable for individual pro-
approaches may utilize HRP-conjugated 2°Ab, and tein analysis, and the latter better for the detection of
both may be utilized separately in the same blot for global protein changes (i.e., ubiquitinated proteins
different targets (Kar et al., 2012). Initially, the or puromycin incorporation). Single-band analysis is
weaker target is blotted for using colorimetric undertaken simply through identifying sample lanes
3,30 ,5,50 -Tetramethylbenzidine detection before heat- before determining the band of interest, while whole
ing in a b-MCE containing buffer before re-probing lane analysis will measure the intensity within a des-
for the additional target with ECL detection. ignated area (i.e., single lane) (Taylor & Posch,
Care should be taken with these methods and con- 2014). Whichever method is chosen, it must be con-
ditions should always be optimized; in addition, it sistent throughout the blot, with an equal area of
should also be demonstrated that stripping and analysis used per lane; otherwise different volumes
re-probing does not adversely influence subsequent (size of the analysis box region) will be analyzed pro-
quantification. Thus, the undertaking of stripping ducing potentially distorted data. Although WB con-
and re-probing must be carefully considered as the ditions will have been optimized to produce the
probable loss of an antigen may in fact lead to erro- clearest bands possible, a visible background may
neous measurements. This becomes more important still occur, requiring background subtraction during
when changes in protein expression are small or analysis (Gassmann et al., 2009). Principally, this is
gradual (i.e., time-course experiments). However, the achieved through subtracting the calculated back-
ability to measure both the phosphorylated and total ground density from peak values; however, the

15
Bass et al.
different algorithms used to determine background sample, being shown to differ between proximal and
intensity may not be truly representative. One com- distal regions of a single mouse sciatic nerve (Eaton
mon method is the rolling disk algorithm, which et al., 2013). Such differences in expression may
determines peak origins based on where a specified become more problematic within skeletal muscle as it
disk could “roll,” typically the smaller the value is a large complex tissue, with different regions
(smaller disk), the greater amount of background potentially responding differently to stimulation
becomes subtracted (Taylor & Posch, 2014). This (Seynnes et al., 2007), therein WB results only pro-
approach is better suited to prominent individual vide an average of expression changes within a single
bands since smaller, less intense bands may become sample. The use of a second HKP may help; how-
lost. Another method is to manually designate a ever, some of the same issues may exist and it will
region of consistent background with no bands, need to be demonstrated that the choice of HKP
assigning that density value for background correc- does not affected the interpretation (Fig. 2). As a
tion. This method may be more suitable for whole result of the potential changes in HKP expression in
blot analysis, with minimal fluctuation in back- response to the experiment and the limited linear
ground levels. Ultimately, the choice of analysis must range of some, the use of HKPs for normalization
be representative of the visualized blot and within may mask or confound potentially relevant changes
the range of linear detection of the imaging method in protein expression.
employed. A viable alternative to blotting for HKPs is to
assess the total amount of protein either within
stained or stain-free gels or on stained membranes
Normalization of target protein abundance
(Welinder & Ekblad, 2011; Eaton et al., 2013).
To account for possible errors in sample preparation Assessing total protein offers distinct advantages
and loading, normalization of samples to remove over HKPs as it is unbiased with respect to changes
inter sample/gel variation is paramount. It is typical in the expression of a single (or multiple) HKP, and
for specific “housekeeping” proteins (HKP) such as if utilizing stained membranes, also allows evalua-
glyceraldehyde 3-phosphate dehydrogenase tion of the blotting process and transfer quality
(GAPDH) or b-actin to be probed for, acting as (Taylor et al., 2013). Coomassie staining a gel and
internal loading controls assuming their expression fluorescence detection has shown the existence of a
remains stable under the experimental conditions wide linear range (1–40 lg), negating previously dis-
used (Welinder & Ekblad, 2011). The HKP chosen cussed HKP linearity issues. Membranes may be
should be one that is known to stay constant between stained to visualize total protein by several methods
control and experimental samples and demonstrated (i.e., Ponceau S, colloidal silver, India ink); however,
to be unaffected by the treatment or intervention Coomassie staining is a common, simple approach
undertaken. Errors such as loading more sample that has been demonstrated to be an unbiased
within one well will increase the target signal, likely method of total protein assessment (when analyzing
skewing data interpretation. As such, target measure- total lane volume) with a high linear range of detec-
ments may be normalized to HKP values, removing tion (2.5–25 lg) (Welinder & Ekblad, 2011). If one
loading bias. However, the accuracy and effective- of these approaches is chosen, the quantification of a
ness of these HKPs are dependent on multiple factors single random band that is consistent across each
such as oversaturation of the protein, high back- lane may be used for normalization. In our experi-
ground, and lack of linearity (McDonough et al., ence, Coomassie staining a membrane is an effective
2014) and can easily suffer from technical errors
within the WB process (Eaton et al., 2013). One
study has demonstrated the linear range of up to
5 lg of loaded protein when probing for GAPDH,
showing no increase in band density at higher con-
centrations (Welinder & Ekblad, 2011). Other com-
mon loading controls (e.g., pan-actin and b-tubulin)
have also displayed poor linear ranges in the same
manner (Li & Shen, 2013). This lack of linearity may
be resolved by loading less protein but may reduce
the sensitivity to detect lower abundant proteins.
Expression of HKPs such as b-actin has also been
shown to be extremely variable between tissue types
Fig. 2. Representative blots for multiple housekeeping pro-
(i.e., muscle, heart, fat) (Eaton et al., 2013). Alarm- teins and the Coomassie-stained membrane of IGF-1-trea-
ingly, however, expression of b-actin has been ted C2C12 cells, demonstrating potential variability in
revealed to not be homogeneous within a single tissue housekeeping proteins in response to a treatment.

16
Technical aspects of Western blotting
method of providing reliable quantification of total allow a greater potential for signaling to occur. Mea-
protein that removes potential problems associated surements of a protein’s phosphorylation status (i.e.,
with individual HKP expression. Staining the mem- signaling activation) may change through modifica-
brane or gel allows appropriate quantification nor- tion of the individual proteins phosphorylation level
malization for whole lane analysis providing more (Tremblay et al., 2007) or by alterations within the
robust normalization approach for both ubiquiti- total amount of protein available (Yung et al.,
nated proteins and puromycin incorporation, as dis- 2011). Consequently, the normalization of a phos-
cussed earlier. pho-protein to its total expression allows the ratio of
The final aspect of analysis is the requirement to phosphorylated proteins to be assessed (i.e., the rela-
calculate the changes in protein expression, resulting tive proportion of phosphorylated vs non) (Wilkin-
from a treatment/intervention. Depending on the son et al., 2008). For example, within skeletal
experimental design, multiple blots may be required muscle, the total expression of various anabolic sig-
to analyze all samples, and thus, subtle changes naling intermediates (e.g., AKT, P70 S6K1)
within the process may influence the final data as remained unchanged, while phosphorylation of these
background or band density may be variable across proteins increased (Brook et al., 2015). As such, the
multiple blots. In this instance, a single quality con- ratio of phosphorylated proteins to total expression
trol sample (typically pooled from multiple controls) increased, demonstrating the increased proportion of
is loaded on each gel, providing a control sample phospho-proteins. Importantly, however, if the treat-
across all gels, allowing gel-to-gel comparisons to be ment undertaken increases both total and phospho-
made. Dividing band values (i.e., band density) by rylation levels, this ratio may remain unchanged,
the quality control sample (as a correction factor) masking any potential mechanisms, and thus it may
normalizes differences in loading, separation, trans- be extremely important for both measures, i.e., total
fer, and detection that may have occurred. This and phosphorylation status, to be made simultane-
should be undertaken for the values of the initial ously.
protein(s) of interest (e.g., Pan-AKT, p-AKT Ser
473, or puromycin incorporation), along with the
Presentation of representative blots
normalization values (i.e., HKP or total protein).
Another approach typically used for human studies One essential aspect and ethical concern in the
is to use an initial basal sample for each individual to reporting of scientific findings is the accurate and
assess changes in subsequent samples, permitting the representative presentation of WB data and example
calculation of a fold change from the initial basal images (for a more in-depth review see Rossner &
levels. Whichever approach is chosen, it must be Yamada, 2004). Generally, WB data are presented as
applied consistently throughout analysis and may both a graphical and representative image to demon-
depend on the total number of samples and compar- strate the effect of the intervention and the quality of
isons needed (i.e., control vs treatment or control vs blot. Herein lie ethical considerations in presenting
treatment 1 vs treatment 2). images that are required to present images that accu-
As previously mentioned, measurement of both rately represent the quantified data (Rossner &
the total expression and phosphorylation status of a Yamada, 2004). For example, the splicing of images
protein (either by multiplexing fluorescent 2°Ab or from multiple blots to form one continual image
stripping and re-probing) is important. Determining may obscure the magnitude of change between sam-
changes to a protein’s total expression in response to ples. If possible replicates from all experimental
a treatment or intervention indicates its capacity to groups should be run within the same blot to provide
signal, since a greater abundance or protein will a representative image; however, if this is not

Fig. 3. Examples of image manipulation techniques upon representative blots. (a) Multiple blots spliced together to give the
impression of a single blot. (b) An oversaturated blot with no clear distinction between bands of interest. (c) Contrast adjust-
ment masking additional bands. (d) Image manipulation to intensify, reduce, or remove bands.

17
Bass et al.
possible, clear distinction between blots should be 96-well plate format, increased sensitivity, relative
made (Fig. 3). Additionally, sample replicates should ease of absolute protein concentration (using
be included within the images, demonstrating equiv- supplied standards), and reduced time consumption
alent changes in response to the intervention. The (2–4 h), proteins of varying molecular weights can-
inclusion of a representative loading control (i.e., not be distinguished from one another, and this is a
Coomassie) is essential to demonstrate that changes crucial consideration that must be made as the 1°Ab
in samples are due to the intervention, rather than used may bind to multiple proteins producing a fal-
protein loads. These are required to be from the sely high signal. Additionally, ELISAs are not cap-
same blots as the other representative images and able of re-probing once completed, instead require
should not be repeated unless the same gel has been multiple plates to be run, thus potentially requiring
stripped and reprobed for multiple targets or differ- more sample than multiple WBs. Another emerging
ent molecular-weight targets were measured. As pre- technology is the use of protein arrays to detect the
viously discussed, one method of validation of target presence of multiple target proteins within a single
specificity is the knowledge of a correct molecular sample (Huang et al., 2001), to explore potential
weight. Despite this, it is common to crop represen- protein interactions with other proteins, and even
tative blots to reduce their overall size. The addition DNA and RNA, and to measure enzyme activity.
of an indicator of the precise molecular weight must Utilizing the same principle as WB and ELISAs,
be included even if image cropping occurs to allow arrays typically contain individual spots of labeled
identification. For example, when blotting for phos- 1°Ab immobilized to the array for multiple targets.
phorylated 4E-BP1, three isoforms typically form Although WB can provide important information on
distinctive bands (i.e., c, b, a) at differing molecular modulation of signaling networks, potential targets
weights (Elia et al., 2008). In this instance, each iso- (or pathways) must first be identified, and develop-
form should be indicated with the appropriate ments within array kits allow the probing of dozens
molecular weight. Accordingly indicators of molecu- of targets simultaneously within a single sample, pro-
lar-weight markers should be included to allow the viding lots of data with relative ease. Interesting
independent determination of a targets size and observations may then be further investigated using
specificity. traditional WB techniques. Although the use of ELI-
Representative images should clearly show indi- SAs is now commonplace, the use of mass spectrom-
vidual bands that are not over loaded/saturated. As etry (i.e., LC-MS/MS) for the analysis and extremely
with any representative image for scientific publica- accurate quantification of proteins is becoming more
tion, inappropriate manipulation may be regarded as widespread (Ryder et al., 2015). Mass spectrometry
misconduct. Thus, manipulation of an image to offers distinct advantages over both ELISAs and tra-
change the intensity of a band(s) or even to remove it ditional WB techniques as the distinction between
altogether is highly unethical (Fig. 3). If any alter- multiple protein isoforms is possible with a high
ation of an image is made, it should affect the degree of sensitivity and over a wide dynamic range
entirety of the image (rather than a specific section) (0.5–500 ng/ll) (Hu et al., 2005). However, such
and be clearly stated in the figure legend. Finally, the analysis requires highly specialized, expensive equip-
inclusion of detailed information of the source of ment and technical expertise not commonplace
both the primary and secondary antibodies is essen- within most labs. Mass spectrometry can be used in
tial for reproduction in other laboratories along with conjunction with WB to undertake highly specialized
possible future discussion of antibody specificity. measures, e.g., to confirm antibody specificity, to
determine novel protein–protein interactions after
IP, and to uncover PT modifications (Mann & Jen-
Alternative and emerging methodologies to WB
sen, 2003).
Although this review focuses on the technical aspects
and choices to be made to undertake robust and
Conclusions
accurate WB measures, it is often important to uti-
lize additional complementary techniques to supple- WB has emerged as an essential tool within physio-
ment and support the data that WB generates. One logical research; nevertheless with poor understand-
common approach is the use of an ELISA to quan- ing and implementation, any subsequent analysis can
tify the abundance of a target protein (total or phos- produce misleading and confusing interpretation
pho) (Timen et al., 1976) although this method can (i.e., Ab specificity and validation). Before a sample
be expensive, requiring access to a plate reader. is loaded into a gel, careful consideration must be
Commonly, ELISAs use sample lysates containing given to often overlooked aspects such as the appro-
non-denatured native proteins (Dhingra et al., priate buffer for homogenization and extraction of
2011); however, this may not always be the case. the intended target protein for denaturation. Gel
ELISAs offer distinct advantages such as being in composition should effectively separate proteins by

18
Technical aspects of Western blotting
size, with changes to concentration giving resolution Perspectives
to the intended target by varying migration speed.
Western blotting techniques are now considered
Subsequent transfer onto an immobilizing mem-
routine inclusions within the field of physiology and
brane will allow the probing for one or more targets
are invaluable in providing mechanistic insight into
with 1°Ab and 2°Ab, with emphasis upon Ab speci-
many regulatory processes. The present review aimed
ficity and the ability to assess PT modifications. Vali-
to give a comprehensive insight into the multiple
dation of Ab should always be undertaken, using
aspects within the WB process, providing the reader
both positive and negative controls to try to ensure
with enhanced expertise to critically evaluate and
specificity. Within each study design and group com-
troubleshoot all features to produce reliable and
parisons, QC samples should be used allowing the
reproducible blots.
comparison of multiple gels. The method of detec-
tion will be ultimately be determined by the equip-
Key words: Westen blot, physiology, SDS PAGE,
ment available. However, fluorescent antibodies
skeletal muscle.
have a greater dynamic range and may be multi-
plexed for additional targets if desired. As sample
quantity may be scarce, the ability to strip and Acknowledgements
re-probe membranes for additional targets is desir-
This work was supported by a grant from the Physiological
able; however, potential issues with regard to quan- Society awarded to P. J. Atherton and K. Smith and a grant
tification and potential signal reduction should be from the U.S. National Institutes of Health National Institute
considered carefully and where possible mitigated. for Arthritis and Musculoskeletal and Skin Diseases (AR-
Finally, the quantification and analysis of band 054342) to N. J. Szewczyk. J. J. Bass is funded by the MRC
intensity should be evaluated consistently through- Doctoral Training Programme (J500495). D. J. Wilkinson is a
Medical Research Council-Arthritis Research United King-
out with both single and multiple blots; as doing so dom (MRC-ARUK) Centre-funded postdoctoral research
can produce reliable and accurate data. fellow.

References
Alegria-Schaffer A, Lodge A, Vattem K. serum alkaline phosphatase. Clin Burnette WN. “Western Blotting”:
Performing and optimizing Western Chem 1966: 12(8): 482–490. electrophoretic transfer of proteins
blots with an emphasis on Beltran Valls MR, Wilkinson DJ, from sodium dodecyl sulfate-
chemiluminescent detection. Methods Narici MV, Smith K, Phillips BE, polyacrylamide gels to unmodified
Enzymol 2009: 463(09): 573–599. Caporossi D, Atherton PJ. Protein nitrocellulose and radiographic
Anfinsen CB. Principles that govern the carbonylation and heat shock detection with antibody and
folding of protein chains. Science proteins in human skeletal muscle: radioiodinated protein A. Anal
1973: 181(4096): 223–230. relationships to age and sarcopenia. Biochem 1981: 112(2): 195–203.
Antharavally BS, Carter B, Bell PA, J Gerontol Ser A Biol Sci Med Sci Chang B, Liu G, Yang G, Mercado-
Krishna Mallia A. A high-affinity 2014: 12: 1–8. Uribe I, Huang M, Liu J. REDD1 is
reversible protein stain for Western Bradford MM. A rapid and sensitive required for RAS-mediated
blots. Anal Biochem 2004: 329(2): method for the quantitation of transformation of human ovarian
276–280. microgram quantities of protein epithelial cells. Cell Cycle 2009: 8(5):
Anton P. 2D PAGE: sample utilizing the principle of protein-dye 780–786.
preparation and fractionation. A. binding. Anal Biochem 1976: 72(1– Chrambach A, Rodbard D.
Posch, ed.Munich: Humana Press, 2): 248–254. Polyacrylamide gel electrophoresis.
2008. Bronstein I, Voyta JC, Thorpe GH, Science 1971: 172(3982): 440–451.
Argentieri MC, Pilla D, Vanzati A, Kricka LJ, Armstrong G. Cleasby ME, Reinten TA, Cooney GJ,
Lonardi S, Facchetti F, Doglioni C, Chemiluminescent assay of alkaline James DE, Kraegen EW. Functional
Parravicini C, Cattoretti G. phosphatase applied in an studies of Akt isoform specificity in
Antibodies are forever: a study using ultrasensitive enzyme immunoassay skeletal muscle in vivo; maintained
12-26-year-old expired antibodies. of thyrotropin. Clin Chem 1989: 35 insulin sensitivity despite reduced
Histopathology 2013: 63(6): 869–876. (7): 1441–1446. insulin receptor substrate-1
Auld DS. Removal and replacement of Brook MS, Wilkinson DJ, Mitchell expression. Mol Endocrinol 2007: 21
metal ions in metallopeptidases. WK, Lund JN, Szewczyk NJ, (1): 215–228.
Methods Enzymol 1995: 248: Greenhaff PL, Smith K, Atherton Cleland WW. Dithiothreitol, a new
228–242. PJ. Skeletal muscle hypertrophy protective reagent for SH groups.
Autuori F, Brunk U, Peterson E, adaptations predominate in the early Biochemistry 1964: 3(4): 480–482.
Dallner G. Fractionation of isolated stages of resistance exercise training, Clezardin P, Bougro G, McGregor JL.
liver cells after disruption with a matching deuterium oxide-derived Tandem purification of IgM
nitrogen bomb and sonication. J Cell measures of muscle protein synthesis monoclonal antibodies from mouse
Sci 1982: 57: 1–13. and mechanistic target of rapamycin ascites fluids by anion-exchange and
Babson AL, Greeley SJ, Coleman CM, complex 1 signaling. FASEB J 2015: gel fast protein liquid
Phillips GE. Phenolphthalein 29: 4485–4496. chromatography. J Chromatogr
monophosphate as a substrate for 1986: 354: 425–433.

19
Bass et al.
Colella AD, Chegenii N, Tea MN, Elia A, Constantinou C, Clemens MJ. Grabski AC. Advances in preparation
Gibbins IL, Williams KA, Chataway Effects of protein phosphorylation of biological extracts for protein
TK. Comparison of Stain-Free gels on ubiquitination and stability of the purification. Methods Enzymol 2009:
with traditional immunoblot loading translational inhibitor protein 4E- 463(09): 285–303.
control methodology. Anal Biochem BP1. Oncogene 2008: 27(6): 811–822. Hachmann JP, Amshey JW. Models of
2012: 430(2): 108–110. Erickson PF, Minier LN, Lasher RS. protein modification in Tris-glycine
Creighton TE. Disulphide bonds and Quantitative electrophoretic transfer and neutral pH Bis-Tris gels during
protein stability. BioEssays 1988: of polypeptides from SDS electrophoresis: effect of gel pH.
8(2): 57–63. polyacrylamide gels to nitrocellulose Anal Biochem 2005: 342(2): 237–245.
Crossland H, Kazi AA, Lang CH, sheets: a method for their re-use in Helenius A, Simons K. Solubilization of
Timmons JA, Pierre P, Wilkinson immunoautoradiographic detection membranes by detergents. Biochim
DJ, Smith K, Szewczyk NJ, of antigens. J Immunol Methods Biophys Acta 1975: 415(1): 29–79.
Atherton PJ. Focal adhesion kinase 1982: 51(2): 241–249. Holden P, Horton WA. Crude
is required for IGF-I-mediated Figueiredo VC, Nader GA. Ursolic subcellular fractionation of cultured
growth of skeletal muscle cells via a acid directly promotes protein mammalian cell lines. BMC Res
TSC2/mTOR/S6K1-associated accretion in myotubes but does not Notes 2009: 2: 243.
pathway. Am J Physiol affect myoblast proliferation. Cell Hu Q, Noll RJ, Li H, Makarov A,
Endocrinol Metab 2013: 305(2): Biochem Funct 2012: 30(5): 432–437. Hardman M, Graham Cooks R. The
E183–E193. Franchi MV, Atherton PJ, Reeves ND, Orbitrap: a new mass spectrometer. J
Delaive E, Arnould T, Raes M, Fl€uck M, Williams J, Mitchell WK, Mass Spectrom 2005: 40(4): 430–443.
Renard P. A sensitive three-step Selby A, Beltran Valls RM, Huang BX, Kim HY. Effective
protocol for fluorescence-based Narici MV. Architectural, functional identification of Akt interacting
Western blot detection. J Immunol and molecular responses to proteins by two-step chemical
Methods 2008: 334(1–2): 51–58. concentric and eccentric loading in crosslinking, co-immunoprecipitation
Desjardins P, Hansen JB, Allen M. human skeletal muscle. Acta Physiol and mass spectrometry. PLoS ONE
Microvolume protein concentration 2014: 210(3): 642–654. 2013: 8(4).
determination using the NanoDrop Gassmann M, Grenacher B, Rohde B, Huang RP, Huang R, Fan Y, Lin Y.
2000c spectrophotometer. J Vis Exp Vogel J. Quantifying Western blots: Simultaneous detection of multiple
2009: 33: e1610. pitfalls of densitometry. cytokines from conditioned media
Dhingra S, Bagchi AK, Ludke AL, Electrophoresis 2009: 30(11): 1845– and patient’s sera by an antibody-
Sharma AK, Singal PK. Akt 1855. based protein array system. Anal
regulates IL-10 mediated suppression Georgopoulos NT, Kirkwood LA, Biochem 2001: 294(1): 55–62.
of TNFa-induced cardiomyocyte Walker DC, Southgate J. Differential Huff-Lonergan E, Parrish FC, Robson
apoptosis by upregulating Stat3 regulation of growth-promoting RM. Effects of postmortem aging
phosphorylation. PLoS ONE 2011: 6 signalling pathways by E-cadherin. time, animal age, and sex on
(9): e25009. PLoS ONE 2010: 5(10): e13621. degradation of titin and nebulin in
Dietrichson P, Coakley J, Smith PE, Gershoni JM, Palade GE. Protein bovine longissimus muscle. J Anim
Griffiths RD, Helliwell TR, Edwards blotting: principles and applications. Sci 1995: 73(4): 1064–1073.
RH. Conchotome and needle Anal Biochem 1983: 131(1): 1–15. Huyer G, Liu S, Kelly J, Moffat J,
percutaneous biopsy of skeletal Gingras AC, Gygi SP, Raught B, Payette P, Kennedy B, Tsaprailis G,
muscle. J Neurol Neurosurg Polakiewicz RD, Abraham RT, Gresser MJ, Ramachandran C.
Psychiatry 1987: 50(11): 1461–1467. Hoekstra MF, Aebersold R, Mechanism of inhibition of protein-
Dimauro I, Pearson T, Caporossi D, Sonenberg N. Regulation of 4E-BP1 tyrosine phosphatases by vanadate
Jackson MJ. A simple protocol for phosphorylation: a novel two-step and pervanadate. J Biol Chem 1997:
the subcellular fractionation of mechanism. Genes Dev 1999: 272(2): 843–851.
skeletal muscle cells and tissue. BMC 13(11): 1422–1437. Jensen EC. The basics of western
Res Notes 2012: 5(1): 513. Gingrich JC, Davis DR, Nguyen Q. blotting. Anat Rec 2012: 295(3):
Doumas BT, Bayse DD, Carter RJ, Multiplex detection and quantitation 369–371.
Peters T, Schaffer R. A candidate of proteins on Western blots using Jensen TE, Richter EA. When less is
reference method for determination fluorescent probes. Biotechniques more: a simple Western blotting
of total protein in serum. I. 2000: 29(3): 636–642. amendment allowing data acquisition
Development and validation. Clin Girgis CM, Mokbel N, Cha KM, on human single fibers. J Appl
Chem 1981: 27(10): 1642–1650. Houweling PJ, Abboud M, Fraser Physiol 2011: 110(1522-1601
Dounce AL, Witter RF, Monty KJ, DR, Mason RS, Clifton-Bligh RJ, (Electronic)): 583–584.
Pate S, Cottone MA. A method for Gunton JE. The vitamin D receptor Kar P, Agnihotri SK, Sharma A,
isolating intact mitochondria and (VDR) is expressed in skeletal Sachan R, Lal Bhatt M, Sachdev M.
nuclei from the same homogenate, muscle of male mice and modulates A protocol for stripping and
and the influence of mitochondrial 25-hydroxyvitamin D (25OHD) reprobing of Western blots originally
destruction on the properties of cell uptake in myofibers. Endocrinology developed with colorimetric substrate
nuclei. J Biophys Biochem Cytol 2014: 155(9): 3227–3237. TMB. Electrophoresis 2012: 33(19–
1955: 1(2): 139–153. Goldberg S. Mechanical/physical 20): 3062–3065.
Eaton SL, Roche SL, Llavero Hurtado methods of cell disruption and tissue Kaufmann SH. Reutilization of
M, Oldknow KJ, Farquharson C, homogenization. Methods Mol Biol immunoblots after chemiluminescent
Gillingwater TH, Wishart TM. Total 2008: 424(1): 3–22. detection. Anal Biochem 2001:
protein analysis as a reliable loading Gordon J. Use of vanadate as protein- 296(2): 283–286.
control for quantitative fluorescent phosphotyrosine phosphatase Kaufmann SH, Shaper JH. Erasable
Western blotting. PLoS ONE 2013: 8 inhibitor. Methods Enzymol 1991: Western blots. Methods Mol Biol
(8): e72457. 201(1982): 477–482. 1992: 10: 235–246.

20
Technical aspects of Western blotting
Kricka LJ. Chemiluminescent and antibodies: why choosing the right Park-Sarge OK. Detection of
bioluminescent techniques. Clin secondary is of primary importance. sumoylated proteins. Methods Mol
Chem 1991: 37(9): 1472–1481. PLoS ONE 2012: 7(6): e38313. Biol 2010: 301: 329–338.
Krieg RC, Dong Y, Schwamborn K, Mansfield MA. Rapid Paul PK, Bhatnagar S, Mishra V,
Knuechel R. Protein quantification immunodetection on polyvinylidene Srivastava S, Darnay BG, Choi Y,
and its tolerance for different fluoride membrane blots without Kumar A. The E3 ubiquitin ligase
interfering reagents using the BCA- blocking. Anal Biochem 1995: TRAF6 intercedes in starvation-
method with regard to 2D SDS 229(1): 140–143. induced skeletal muscle atrophy
PAGE. J Biochem Biophys Methods Matsudaira P. Sequence from picomole through multiple mechanisms. Mol
2005: 65(1): 13–19. quantities of proteins electroblotted Cell Biol 2012: 32(7): 1248–1259.
Kurien BT, Scofield RH. Western onto polyvinylidene difluoride Pere-Brissaud A, Blanchet X,
blotting. Methods 2006: 38(4): 283– membranes. J Biol Chem 1987: Delourme D, Pelissier P, Forestier L,
293. 262(21): 10035–10038. Delavaud A, Duprat N, Picard B,
Kurien BT, Dorri Y, Dillon S, Dsouza McDonough AA, Veiras LC, Minas Maftah A, Bremaud L. Expression
A, Scofield RH. An overview of JN, Ralph DL. Considerations when of SERPINA3s in cattle: focus on
Western blotting for determining quantitating protein abundance by bovSERPINA3-7 reveals specific
antibody specificities for immunoblot. Am J Physiol Cell involvement in skeletal muscle. Open
immunohistochemistry. Methods Physiol 2014: doi:10.1152/ Biol 2015: 5(9): 150071.
Mol Biol 2011: 717: 55–67. ajpcell.00400.2014. Polyansky A, Zagrovic, B. Protein
Kurien BT, Danda D, Bachmann MP, Michalewski MP, Kaczmarski W, electrostatic properties predefining
Scofield RH. SDS-PAGE to Golabek A, Kida E, Kaczmarski A, the level of surface hydrophobicity
immunoblot in one hour. Methods Wisniewski KE. Immunoblotting change upon phosphorylation. J
Mol Biol 2015: 1312: 449–454. with antiphosphoamino acid Phys Chem Lett 2012: 3: 973–976.
Laemmli UK. Cleavage of structural antibodies: importance of the Quach NL, Biressi S, Reichardt LF,
proteins during the assembly of the blocking solution. Anal Biochem Keller C, Rando TA. Focal adhesion
head of bacteriophage T4. Nature 1999: 276(2): 254–257. kinase signaling regulates the
1970: 227(5259): 680–685. Miura T, Suzuki W, Ishihara E, Arai I, expression of caveolin 3 and beta1
Lal A, Haynes SR, Gorospe M. Clean Ishida H, Seino Y, Tanigawa K. integrin, genes essential for normal
western blot signals from Impairment of insulin-stimulated myoblast fusion. Mol Biol Cell 2009:
immunoprecipitated samples. Mol GLUT4 translocation in skeletal 20(14): 3422–3435.
Cell Probes 2005: 19(6): 385–388. muscle and adipose tissue in the Ramlau J. Use of secondary antibodies
Layne E. Spectrophotometric and Tsumura Suzuki obese diabetic for visualization of bound primary
turbidimetric methods for measuring mouse: a new genetic animal model reagents in blotting procedures.
proteins. Methods Enzymol 1957: 3: of type 2 diabetes. Eur J Endocrinol Electrophoresis 1987: 8(9): 398–402.
447–455. 2001: 145: 785–790. Rasc on A, Lindgren S, Stavenow L,
Legocki RP, Verma DP. Multiple Mollica JP, Oakhill JS, Lamb GD, Belfrage P, Andersson K-E,
immunoreplica Technique: screening Murphy RM. Are genuine changes Manganiello VC, Degerman E.
for specific proteins with a series of in protein expression being Purification and properties of the
different antibodies using one overlooked? Reassessing Western cGMP-inhibited cAMP
polyacrylamide gel. Anal Biochem blotting Anal Biochem 2009: 386(2): phosphodiesterase from bovine aortic
1981: 111(2): 385–392. 270–275. smooth muscle. Biochim Biophys
Li R, Shen Y. An old method facing a Mozdzanowski J, Hembach P, Speicher Acta Mol Cell Res 1992: 1134(2):
new challenge: re-visiting DW. High yield electroblotting onto 149–156.
housekeeping proteins as internal polyvinylidene difluoride membranes Rasmussen UF, Rasmussen HN.
reference control for neuroscience from polyacrylamide gels. Human quadriceps muscle
research. Life Sci 2013: 92(13): 747– Electrophoresis 1992: 13(1–2): mitochondria: a functional
751. 59–64. characterization. Mol Cell Biochem
Lipman NS, Jackson LR, Trudel LJ, Murphy RM, Lamb GD. Important 2000: 208(1–2): 37–44.
Weis-Garcia F. Monoclonal versus considerations for protein analyses Rath A, Cunningham F, Deber CM.
polyclonal antibodies: distinguishing using antibody based techniques: Acrylamide concentration determines
characteristics, applications, and down-sizing western blotting the direction and magnitude of
information resources. ILAR J 2005: up-sizes outcomes. J Physiol 2013: helical membrane protein gel shifts.
46(3): 258–268. 591: 5823–5831. Proc Natl Acad Sci USA 2013:
MacPhee DJ. Methodological Nairn AC, Detre JA, Casnellie JE, 110(39): 15668–15673.
considerations for improving Greengard P. Serum antibodies that Raymond S, Weintraub L. Acrylamide
Western blot analysis. J Pharmacol distinguish between the phospho- gel as a supporting medium for zone
Toxicol Methods 2010: 61(2): 171– and dephospho-forms of a electrophoresis. Science 1959:
177. phosphoprotein. Nature 1982: 299 130(3377): 711.
Mahmood T, Yang P-C. Western blot: (5885): 734–736. Rommel C, Bodine SC, Clarke BA,
technique, theory, and trouble Noble JE, Knight AE, Reason AJ, Di Rossman R, Nunez L, Stitt TN,
shooting. N Am J Med Sci 2012: MATOLA A, Bailey MJ. A Yancopoulos GD, Glass DJ.
4(9): 429–434. comparison of protein quantitation Mediation of IGF-1-induced skeletal
Mann M, Jensen ON. Proteomic assays for biopharmaceutical myotube hypertrophy by PI(3)K/
analysis of post-translational applications. Mol Biotechnol 2007: Akt/mTOR and PI(3)K/Akt/GSK3
modifications. Nat Biotechnol 2003: 37(2007): 99–111. pathways. Nat Cell Biol 2001: 3(11):
21(3): 255–261. Ornstein L. Disc electrophoresis. I. 1009–1013.
Manning CF, Bundros AM, Trimmer Background and theory. Ann N Y Rossner M, Yamada KM. What’s in a
JS. Benefits and pitfalls of secondary Acad Sci 1964: 121(2): 321–349. picture? The temptation of image

21
Bass et al.
manipulation. J Cell Biol 2004: Taylor SC, Berkelman T, Yadav G, Wang Y, Becklund BR, DeLuca HF.
166(1): 11–15. Hammond M. A defined Identification of a highly specific and
Rudd PM, Dwek RA. Glycosylation: methodology for reliable versatile vitamin D receptor
heterogeneity and the 3D structure quantification of Western blot data. antibody. Arch Biochem Biophys
of proteins. Crit Rev Biochem Mol Mol Biotechnol 2013: 55(3): 217–226. 2010: 494(2): 166–177.
Biol 1997: 32(1): 1–100. Thomas G. p70s6k/p85s6k: mechanism Warren CM, Krzesinski PR, Greaser
Ryder DJ, Judge SM, Beharry AW, of activation, effects of rapamycin ML. Vertical agarose gel
Farnsworth CL, Silva JC, Judge AR. and role in mitogenesis. Biochem Soc electrophoresis and electroblotting of
Identification of the acetylation and Trans 1993: 21(4): 901–904. high-molecular-weight protiens.
ubiquitin-modified proteome during Timen AE, Prokhorenko OK, Zalesskiĭ Electrophoresis 2003: 24(11): 1695–
the progression of skeletal muscle VN. The enzyme-linked 1702.
atrophy. PLoS ONE 2015: 10(8): immunosorbent assay (ELISA). Bull Weber K, Osborn M. The reliability of
e0136247. World Health Organ 1976: 54(2): molecular weight determinations by
Schmidt EK, Clavarino G, Ceppi M, 129–139. dodecyl sulfate-polyacrylamide gel
Pierre P. SUnSET, a nonradioactive Tino WT, Huber MJ, Lake TP, Greene electrophoresis. J Biol Chem 1969:
method to monitor protein synthesis. TG, Murphy GP, Holmes EH. 244(16): 4406–4412.
Nat Methods 2009: 6(4): 275–277. Isolation and characterization of Welinder C, Ekblad L. Coomassie
Scopes RK. Protein purification. New monoclonal antibodies specific for staining as loading control in
York, NY: Springer New York, protein conformational epitopes Western blot analysis. J Proteome
1994. present in prostate-specific Res 2011: 10: 1416–1419.
Sennepin AD, Charpentier S, membrane antigen (PSMA). Westermeier R. Electrophoresis. In:
Normand T, Sarre C, Legrand A, Hybridoma 2000: 19(3): 249–257. eLS. John Wiley & Sons Ltd.
Mollet LM. Multiple reprobing of Tovey ER, Baldo BA. Comparison of Chichester, UK 2005: pp 1–6.
Western blots after inactivation of semi-dry and conventional tank- Wilkinson SB, Phillips SM, Atherton
peroxidase activity by its substrate, buffer electrotransfer of proteins PJ, Patel R, Yarasheski KE,
hydrogen peroxide. Anal Biochem from polyacrylamide gels to Tarnopolsky MA, Rennie MJ.
2009: 393(1): 129–131. nitrocellulose membranes. Differential effects of resistance and
Seynnes OR, de Boer M, Narici MV. Electrophoresis 1987: 8(9): endurance exercise in the fed state on
Early skeletal muscle hypertrophy 384–387. signalling molecule phosphorylation
and architectural changes in response Towbin H, Staehelin T, Gordon J. and protein synthesis in human
to high-intensity resistance training. J Electrophoretic transfer of proteins muscle. J Physiol 2008: 586(Pt 15):
Appl Physiol 2007: 102(1): 368–373. from polyacrylamide gels to 3701–3717.
Sharma SK, Carew TJ. Inclusion of nitrocellulose sheets: procedure and Yeung Y, Stanley ER. A solution for
phosphatase inhibitors during some applications. Proc Natl Acad stripping antibodies from
Western blotting enhances signal Sci USA 1979: 76(9): 4350–4354. polyvinylidene fluoride immunoblots
detection with phospho-specific Trauger SA, Webb W, Siuzdak G. for multiple reprobing. Anal
antibodies. Anal Biochem 2002: Peptide and protein analysis with Biochem 2009: 389(1): 89–91.
307(1): 187–189. mass spectrometry. Spectroscopy Yung HW, Charnock-Jones DS,
Smith BJ. SDS polyacrylamide gel 2002: 16(1): 15–28. Burton GJ. Regulation of AKT
electrophoresis of proteins. Methods Tremblay F, Br^ ule S, Hee Um S, Li Y, phosphorylation at Ser473 and
Mol Biol 1984: 1: 41–55. Masuda K, Roden M, Sun XJ, Thr308 by endoplasmic reticulum
Somwar R, Sumitani S, Taha C, Krebs M, Polakiewicz RD, Thomas G, stress modulates substrate specificity
Sweeney G, Klip A. Temporal Marette A. Identification of IRS-1 in a severity dependent manner V.
activation of p70 S6 kinase and Akt1 Ser-1101 as a target of S6K1 in Cheriyath, ed. PLoS ONE 2011:
by insulin: PI 3-kinase-dependent nutrient- and obesity-induced insulin 6(3): e17894.
and -independent mechanisms. Am J resistance. Proc Natl Acad Sci USA Zampieri S, Ghirardello A, Doria A,
Physiol 1998: 275(4 Pt 1): E618– 2007: 104(35): 14056–14061. Tonello M, Bendo R, Rossini K,
E625. Voelkel T, Andresen C, Unger A, Just Gambari PF. The use of Tween 20 in
Spinola SM, Cannon JG. Different S, Rottbauer W, Linke WA. Lysine immunoblotting assays for the
blocking agents cause variation in methyltransferase Smyd2 regulates detection of autoantibodies in
the immunologic detection of Hsp90-mediated protection of the connective tissue diseases. J Immunol
proteins transferred to nitrocellulose sarcomeric titin springs and cardiac Methods 2000: 239(1–2): 1–11.
membranes. J Immunol Methods function. Biochim Biophys Acta Mol Zhao J, Brault JJ, Schild A, Cao P,
1985: 81(1): 161–165. Cell Res 2013: 1833(4): 812–822. Sandri M, Schiaffino S, Lecker SH,
Tanford C. Contribution of Voller A, Bartlett A, Bidwell DE. Goldberg AL. FoxO3 coordinately
hydrophobic interactions to the Enzyme immunoassays with special activates protein degradation by the
stability of the globular reference to ELISA techniques. J autophagic/lysosomal and
conformation of proteins. J Am Clin Pathol 1978: 31(6): 507–520. proteasomal pathways in atrophying
Chem Soc 1962: 84(22): 4240–4247. Walker JM. Gradient SDS muscle cells. Cell Metab 2007: 6(6):
Taylor SC, Posch A. The design of a polyacrylamide gel electrophoresis of 472–483.
quantitative western blot experiment. proteins. Methods Mol Biol 1994:
BioMed Res Int 2014: 2014: 361590. 32: 35–38.

22

Vous aimerez peut-être aussi