SlideShare une entreprise Scribd logo
1  sur  36
IN-VIVO PROTEIN SYNTHESIS : FROG OOCYTE SYSTEM
INTRODUCTION
 In 1971 Gurdon demonstrated that oocytes extracted from the South-African
clawed frog Xenopus laevis were able to synthesize hemoglobin consequent to
injection of the corresponding mRNA (Gurdon et al., 1971).
 Oocytes are particularly well suited for expression of proteins following mRNA
injection, since they contain accumulated stores of enzymes, organelles and
proteins that are normally used after fertilization that can be recruited for
heterologous proteins.
 The most common approach for expression of exogenous proteins in Xenopus
oocytes is to inject in vitro transcribed complementary RNA into the oocyte
cytoplasm. Expression can be also achieved by injection of the cDNA into the
oocyte nucleus, but this method requires visual localization of the nucleus and
involves the risk of damaging the nuclear membrane during injection; which
renders this approach more technically challenging and less efficient.
 Oocytes of X. laevis are used because of the following reasons;
1) X. laevis is easily maintained in captivity, making it a low-cost laboratory animal.
2) Oocytes can be readily harvested and, because of their large diameter (1-1.2
mm), are straightforward to injected.
3) frog oocytes are built to survive for long periods of time outside the body
4) Oocytes express a low number of endogenous membrane transporters and
channels because they are virtually independent from exogenous nutrients.
5) Multiple species of mRNA can be simultaneously injected to study protein
complexes formed by multiple subunits, an advantage over transfected
mammalian cells for studying large multi-protein complexes.
6) Standard electrophysiological techniques are easily applicable.
XENOPUS DEVELOPMENT AND CHARACTERISTICS
 A Xenopus oocyte is an immature egg that under the appropriate hormonal
stimulation can become competent for fertilization. The egg production is
controlled by subcutaneous injections of 50-100 units of pregnant mare serum
(PMS) in their dorsal lymph sacs several days before the second injection with
600-800 units of human chorionic gonadotropin (hCG) 12-16 hours before the
desirable egg laying. Right after the second injection, they are placed in
individual tanks and the next day the eggs are collected by by surgical
procedures.
 Oocytes go through six maturation stages (I-VI); stage V and VI oocytes are
generally used for electrophysiological studies and are the largest. These large
cells (1-1.2 mm diameter) have a black pigmented region called the animal pole
and a white (non-pigmented) vegetal pole.
 They contain a huge nucleus (or germinal vesicle), 100,000 times larger than a
somatic cell nucleus, which occupies approximately the one-third of the oocyte’s
volume and they develop rapidly. The vesicle is surrounded by a nuclear envelope
with large pores that facilitate transportation from and to the cytoplasm.
 The oocyte plasma membrane is surrounded by a vitelline membrane, that helps to
maintain the oocyte shape and renders the oocyte more resistant to manipulations.
The vitelline membrane does not affect electrophysiological recordings since it is
devoid of channels and transporters and has a large-enough mesh to allow
permeation of ions and small molecules.
Fig.
1
 However, for single channel recordings the vitelline membrane is removed because it
hinders the formation of high resistance seal between the patch pipette and the oocyte
membrane. Around the vitelline membrane is a layer of follicular cells that separates
the oocyte from the external environment (Figure 1).
 By contrast to the vitelline membrane, the follicular cells express ion channels and
transporters they are electrically coupled to each other and to the oocyte by gap
junctions and therefore can create serious interference during electrophysiological
recordings. For these reasons this layer of cells is eliminated prior to injection by a
combination of collagenase treatment and manual stripping.
 The material that Xenopus can offer in biology is great in terms of quality and quantity.
For example, one frog ovary corresponds to 1000 mouse ovaries and western blots can
be performed with protein samples from just one oocyte (or 1µL of extract).
 Oocytes are very rich in all the necessary RNA and proteins for the first stages of
development until early embryogenesis, i.e., the tadpole stage. They can contain up to
4 µg of ribosomal RNA, therefore each oocyte is able of synthesizing up to 400 ng of
proteins per day.
 In addition, they lack transcriptional activity until the mid-late blastula stage. It is
estimated that the protein content is about 500 µg in each oocyte and approximately
10-50 mg/mL in the extract.
CDNA CLONING INTO THE APPROPRIATE VECTORS
 When a cDNA sequence has not been experimentally confirmed, it is important to keep in mind
that less conserved regions (usually 5′ and 3′ end) may be mispredicted. Moreover, splice variants
with different functional properties or cell-specific expression may exist and should be considered
whenever attempting to fully characterize a protein using an expression system such as Xenopus
oocytes.
 When experimentally confirmed, a cDNA may be available as full length or overlapping EST
(Expression Sequence Tag).
 For genes for which cDNA clones are not available, cDNA can be cloned relying on sequence
information contained in highly conserved regions of the gene or using non-biased methods based
on RACE-PCR to identify extreme 3′ and 5′ ends.
FEATURES OF THE PREFERRED VECTORS
 Cloned cDNA must be inserted into a plasmid that ensures stability of the transcribed
sequence and efficient translation into Xenopus oocytes.
 Vectors such as pSP64T (Promega), pGEM (Promega) and pCDNA3 (Invitrogen) all contain
polyadenylation (poly(A) sequences at the 3′ end of their cloning linker that render
transcribed RNA stable and are suitable for oocytes expression.
 In addition plasmid pSP64T contains the 5′ and 3′ UTRs from Xenopus laevis β-globin. The β-
globin sequences greatly enhance translation efficiency of heterologous mRNA transcripts in
oocytes. The vectors contain also RNA polymerase promoter sites (SP6 in pSP64T and T7 in
PGEM and pCDNA3) that are used for the in vitro transcription.
IN- VITRO RNA SYNTHESIS
1. Choosing the right kit
 In vivo most eukaryotic mRNAs have a 7-methyl guanosine cap structure at their 5′ end
required for efficient message binding to ribosomes and efficient translation.
 When in vitro transcribing RNA for oocyte expression studies, it is important to use a
transcription kit that adds a guanosine cap to newly synthesized cRNA molecules.
 Failing to do so will dramatically reduce the expression of the protein of interest.
(mMESSAGE mMACHINE kits from Ambion contain a capped analog that is incorporated as
the first G of the transcript).
Ambion markets three kits containing three different RNA polymerases: SP6, T7 and T3.
Depending on the promoter sequence of the vector employed and/or on the orientation of
the cDNA, the appropriate kit to generate a sense transcript must be used.
2. Plasmid template
• DNA should be free of contaminating proteins and RNA. Most commercially
available plasmid preparation systems yield DNA that is appropriate for in vitro RNA
transcription using a transcription kit. Alternatively, DNA could be purified by
standard phenol/chloroform/ isoamyl alcohol extraction.
3. Linearization of the DNA template
• Circular plasmid templates generate long and heterogeneous RNA transcripts
because RNA polymerases are highly processive. To produce a homogeneous
population of transcripts, plasmid DNA should be linearized by digesting with a
restriction enzyme that cuts at the 3′ end of the inserted cDNA. The restriction
enzyme should be chosen with care. An enzyme that leaves 5′ end overhangs is
optimal. Restriction enzymes leaving 3′ overhanging ends (for example Kpn I, Pst I,
etc) are the least desirable because they yield low level of transcripts.
PROTOCOL
Digest plasmid DNA (5-10 µg) in a large volume reaction (150-200 µl) for at least 3 hours
to ensure complete digestion of the template. Electrophorese 5 µl of the digestion mix to
check for complete digestion.
Clean the digested DNA from restriction enzyme and restriction buffer either by
phenol/chloroform/isoamyl alcohol extraction or using a DNA purification kit. PCR purification kit
from QIAGEN is appropriate and recovers DNA at high yield. Elute or resuspend the DNA with 50 µl
RNase-free water.
Ethanol precipitate DNA overnight at −20°C using 30 µl RNase-free 3 M sodium acetate and 1.2 ml
100% ethanol. Centrifuge 12,000g for 30 min at 4°C, wash once with 70% ethanol, decant
supernatant completely and resuspend in 8 µl RNase-free water.
Run 1 µl to check recovery of DNA and estimate its concentration by comparison with an
appropriate size standard of known quantity or by measuring O.D.
XENOPUS OOCYTES - ISOLATION & ENUCLEATION
 Microinjection of Xenopus oocytes has proven to be a valuable tool in a broad array of
studies that require expression of DNA or RNA into functional protein.
 These studies are diverse and range from expression cloning to receptor–ligand interaction
to nuclear programming.
 Oocytes offer a number of advantages for such studies, including their large size (1.2 mm in
diameter), capacity for translation, and enormous nucleus (0.3–0.4 mm).
 They are cost effective, easily manipulated, and can be injected in large numbers in a short
time period. Oocytes have a large maternal stockpile of all the essential components for
transcription and translation.
 Consequently, the investigator needs only to introduce by microinjection the specific DNA
or RNA of interest for synthesis.
 Oocytes translate virtually any exogenous RNA regardless of source, and the translated
proteins are folded, modified, and transported to the correct cellular locations.
 Xenopus laevis ovaries are large and transparent and may contain several hundreds of, also large,
oocytes which are easily obtainable.
 Oocytes are prepared from Xenopus laevis ovaries that can be extracted from adult females.
 The oocytes can be manually dissected using forceps from the ovaries or they can be extracted by
digestion with collagenase (enzymatic defolliculation) of the four extracellular connective tissue layers
that surround them.
 Then, from the oocytes, the nucleus can be manually removed. A pair of forceps with sharp tips is
used to pierce a small opening on the cell and by squeezing force applied by another pair of forceps
with blunt tips the nucleus is extracted.
 This results in oocytes without any nucleus (enucleated oocyte) and isolated nuclei. The experimental
protocol with which the enucleation takes place is selected according to the downstream application,
which can be immunoblotting.
 Injection of a somatic nuclei and RNA synthesis for analysis of transcription, Fully functional nuclei
can be isolated and be used to study all nuclear processes including gene expression, chromatin
dynamics, nuclear import and export of fluorescently labeled proteins or function of the nuclear pore
complexes
OOCYTE PROCUREMENT PROTOCOL
Anesthetize the adult female.
Place the frog on crushed ice for approximately 20 min prior to, and throughout, the surgical procedure to
maintain anesthesia.
Make a 1 cm incision in the lower abdominal wall near one leg to expose an ovary.
Carefully remove about one third of one ovary and place it in OR2 with CaCl2 or ND96 with CaCl2.
Dissociate oocytes by collagenase treatment. Incubate for 1hr with gentle agitation at room temperature,
Removal of the follicular layer is actually easier several hours after collagenase treatment; in addition oocytes tend
to be more fragile right after enzymatic treatment.
Manually defolliculated stage V and VI oocytes by forcing them through a pipette with an opening slightly smaller than
the diameter of the oocytes (smooth the opening by fire polishing).
The higher concentration of collagenase allows defolliculation of most oocytes that can be set aside for injection.
OOCYTE MICROINJECTION
 The volume of one Xenopus laevis oocyte is approximately 1µL. They can tolerate an
injection of up to 50 nL using automatic nanoinjectors, without any adverse effect in
the function.
 Injections can be used for either gain-of-function (GOF) or loss-of-function (LOF)
experiments or experiments involving both.
 There are two routes for introducing the foreign genetic information into the oocyte
and both require injection of the cell.
 The chief problem for cytoplasmic injections is the instability of the mRNA, for stability
poly (A) tail is added. The mRNA have a 5’7-methyl guanosine residue cap structure
which functions in the protein synthesis initiation process, protects the mRNA from
degradation and is essential for oocyte expression.
 Oocyte expression importance of both capping and the poly(A) tail of the mRNA
expression in oocytes changes during oocyte development, but the two processes
work synergistically stimulating translation as the oocyte matures, This is one reason
for checking that the oocytes chosen for injection are at the correct developmental
stage to optimize expression.
 A different approach is to directly inject the nucleus with double-stranded cDNA.
However, the cDNA must be cloned into a suitable expression vector before nuclear
1) dissecting microscope
2) cold light source power supply,
3) cold light source goose-necks,
4) microinjector mounted on a coarse
micromanipulator,
5) switch pedal.
• Oocyte injection chamber with
microchip sockets for holding
oocytes during injection.
PROTOCOL
Immobilize oocytes by
positioning them into the
openings of a nylon
mesh at the bottom of a
Petri dish containing
culture medium.
Prepare 50–100 oocytes
at a time for
microinjection. gently
withdraw the pipette and
move to the next oocyte
by moving the
microinjection dish.
Inject the sample, then wait a
few seconds before
withdrawing the pipette to
minimize sample leakage.
Periodically release one
injection shot into solution to
ensure the pipette has not
clogged.
When microinjection is
complete, carefully
separate the oocytes into
individual dishes or
microplate wells
containing the same
medium used during
injection
Return the oocytes to the
18˚C incubator.
Twice a day, monitor the
oocytes for viability,
removing damaged or
dying oocytes to prevent
deleterious effects on
healthy oocytes.
Include antibiotics, such
as gentamycin or
streptomycin, in the
culture medium to
prevent bacterial infection
 The voltage clamp is an electrophysiological technique used to measure ion current
through the cell membrane of excitable cells such as oocytes and neurons while
holding the membrane voltage to desired level.
 The TEVC technique is used to study properties of membrane proteins especially ion
channels.
 Researchers use this method most commonly to investigate membrane structures
expressed in xenopus oocyte.
 This technique allows to measure the current across the cell membrane.
 Voltage clamping allows for the potential in the system to be set to a specific voltage
while recording the current.
TWO ELECTRODE VOLTAGE CLAMP
PRINCIPLE OF TEVC
 The TEVC method utilizes two low-
resistance pipettes, one is for sensing
voltage and the other is for injecting
current.
 The micro electrodes are filled with
conducting solution and inserted into the
cell to artificially control membrane
potential.
 The microelectrodes compare the
membrane potential against a command
voltage, giving an accurate reproduction of
the currents flowing across the membrane.
 Current reading can be used to analyze the
electrical response of the cell to different
application.
TEVC IN XENOPUS OOCYTE
 In Xenopus oocytes, the electrodes are filled with a 3M KCl solution and have an electric
resistance between 0.2 and 3Ω. Other solutions with lower KCl concentrations and
containing Ca2+ buffers such as EGTA can be used.
 Ca2+ buffers are used to inhibit endogenous Ca2+-activated Cl-currents.
 The electrodes are mounted on electrode-holders that contain a silver wire coated with a
layer of AgCl2 which allows transmission of the signal from the KCl solution through the
silver wire to the amplifier. The ground electrodes are made of 3M KCl in 2-3% agar.
 After both electrodes have been inserted, the amplifier is set in voltage-clamp mode and
the voltage-clamp feedback gain, used to compensate for membrane capacitance, is
increased to speed up the time response of the voltage clamp.
 Currents are then filtered and recorded using an analog/digital converter and a computer.
WHY TEVC IS USED OVER OTHER VOLTAGE CLAMP TECHNIQUES
TEVC offers many advantages over other techniques such as:-
1. Having a high current-passing capacity.
2. Able to inject current into the system for increased sensitivity.
3. Increased signal-to-noise ratio (SNR).
Increasing sensitivity and SNR is important in a scanning ion conductance
microscope. It increases the ionic conductance of the system, which allows for
higher sensitivity and low noise when scanning a sample and its features.
DETECTING PROTEINS AT THE OOCYTE SURFACE BY FLUORESCENT
METHOD
 Detecting expressed proteins at the plasma membrane of Xenopus oocytes can be used
to examine their trafficking to the membrane, their subcellular distribution or even
interaction with other proteins.
 When drawing conclusions on protein trafficking based on observations made in
Xenopus oocytes though, it should be kept in mind that oocytes are reared at 19°C–
20°C while the organism from which the protein originates may have a different vital
temperature.
 Studies on mammalian membrane proteins may be complicated by this problem while
for C. elegans proteins this factor may have less of an impact (nematodes can be
reared at a wide range of temperatures from 15°C to 25°C).
 Nevertheless, it is important to keep in mind that certain mutant phenotypes are
temperature-sensitive and a protein that is at the cell surface at 20°C may display a
traffic defect at 25°C .
 Membrane proteins can be visualized at the oocyte plasma membrane in two ways:
1) By tagging them with fluorescent proteins such as Green Fluorescent Protein (GFP) or
2) By immunostaining
 There are at least three different protocols for oocyte immunostaining: two in which
the oocytes are fixed and sliced prior to incubation with the antibodies and one in
which they are sliced after incubation with antibodies.
 For visualization of GFP-tagged proteins at the plasma membrane, oocytes must be
fixed and sliced. Fixing and slicing methods described in protocol 1 are well-suited
for visualization of GFP-tagged proteins.
Protocol. 1
Fix oocytes overnight at 4°C with 4% paraformaldehyde in PBS.
Wash three times for 5 minutes with PBS and imbed the oocytes in 3% low-melting point
agarose in PBS. Pour enough warm (not hot) agarose in a 35 mm Petri dish to cover the
bottom, drop 3 to 4 oocytes on the surface and cover with another layer of agarose.
Incubate the imbedded oocytes at 4°C for at least 2 hours to allow hardening of
the agarose and then, using a vibrotome, section into 50 µm slices.
Block non-specific sites by incubation with 0.2% Bovine Serum Albumin (BSA) plus
0.1% Tween 20 in PBS overnight at 4°C or for 3–4 hours at room temperature.
Transfer the slices from one solution to another using a fire-polished Pasteur pipette.
Incubate with the primary antibody in 1% Bovine Serum Albumin (BSA)
plus 0.1% Tween 20 in PBS preferably over night at 4°C.
Wash three times for 5 minutes with PBS and incubate with a
fluorescent secondary antibody in 1% Bovine Serum Albumin (BSA)
plus 0.1% Tween 20 in PBS for 1 hour at room temperature.
Wash three times for 5 minutes in PBS and mount the slices on glass
slides using a medium appropriate for fluorescent microscopy
Photograph slices using an inverted microscope equipped with 4x and 20x
objectives. Microscopes typically used for C. elegans photography are well
suited for oocyte photography.
Examples of oocyte sections stained using this protocol
.
Immunostaining of C. elegans in Xenopus
oocytes using a polyclonal antibody
(a) Fluorescent photograph of an
immune stained non-injected
oocyte, showing no background
fluorescence at the plasma
membrane.
(b) Oocytes expressing MEC-
4(d) stained with anti-MEC-4
antibodies.
(C) Oocytes expressing MEC-
4(A713T/ S726F) stained with
anti-MEC-4 antibodies.
Protocol.
2
(Use this protocol to detect FLAG-tagged proteins)
Fix the oocytes with 3% paraformaldehyde in PBS for 4 h.
Place fixed oocytes on thin cork disks, embedded into cryo-embedding compound and freeze them
in liquid propane cooled by liquid nitrogen. Store the oocytes at −80°C until further use.
Section the oocytes in 6 µm slices using a cryostat and place them on chrom-alum
gelatin-coated glass slides.
Use the tyramide signal amplification (TSA-Direct) kit according to the manufacturer's instructions.
Incubate with anti-FLAG antibody diluted 1:100 in the TSA blocking buffer for 1 hour at room temperature.
Rinse slices with PBS containing 0.05% Tween and incubate with a 1:100 dilution of horseradish peroxidase-
conjugated sheep anti-mouse Ig (1 hour at room temperature).
Wash with PBS-Tween and incubate with FITC-tyramide conjugates diluted 1:50 in the TSA diluent (1 hour at
room temperature).
Wash slices in PBS-Tween and mount them in DAKO-Glycergel containing 2.5% of 1,4-diazabicyclo (2.2.2)-octane as a
fading retardant.
IMMUNOPRECIPITATION
 Immunoprecipitation (IP) is the technique of precipitating a protein antigen out of
solution using an antibody that specifically binds to that particular protein.
 This process can be used to isolate and concentrate a particular protein from a
sample containing many thousands of different proteins.
 Immunoprecipitation requires that the antibody be coupled to a solid substrate at
some point in the procedure.
 Antigens isolated by IP are analyzed by SDS-PAGE or Western blotting.
TYPES OF IMMUNOPRECIPITATION
There are different types of immunoprecipitation:
1. Individual protein immunoprecipitation (IP): Involves using an antibody that is specific for a
known protein to isolate that particular protein out of a solution containing many different
proteins.
2. Protein complex immunoprecipitation (Co-IP): Immunoprecipitation of intact protein
complexes is known as co-immunoprecipitation (Co-IP). Co-IP works by selecting an antibody
that targets a known protein that is believed to be a member of a larger complex of proteins.
3. Chromatin immunoprecipitation (ChIP): Chromatin immunoprecipitation (ChIP) is a method
used to determine the location of DNA binding sites on the genome for a particular protein of
interest. This technique gives a picture of the protein–DNA interactions that occur inside the
nucleus of living cells or tissues.
4. RNP immunoprecipitation targets ribonucleoproteins (RNPs): Live cells are first lysed and
then the target protein and associated RNA are immunoprecipitated using an antibody
targeting the protein of interest
EXTRACTING PROTEINS FROM XENOPUS OOCYTES FOR
IMMUNOPRECIPITATION
 The most important step when extracting proteins from Xenopus oocytes is to
eliminate the large amount of yolk, which interferes with electrophoresis and
immunodetection.
 This allows elimination of the yolk and extraction of membrane proteins from
Xenopus oocytes.
STEPS OF IMMUNOPRECIPITATION
 Lyse cells and prepare sample for immunoprecipitation.
 Pre-clear the sample by passing the sample over beads alone or
bound to an irrelevant antibody to soak up any proteins that non-
specifically bind to the IP components.
 Incubate solution with antibody against the protein of interest.
Antibody can be attached to solid support before this step (direct
method) or after this step (indirect method). Continue the
incubation to allow antibody-antigen complexes to form.
 Precipitate the complex of interest, removing it from bulk solution.
 Wash precipitated complex several times. Spin each time between
washes when using agarose beads or place tube on magnet when
using superparamagnetic beads and then remove the supernatant.
After the final wash, remove as much supernatant as possible.
 Elute proteins from the solid support using low-pH or SDS sample
loading buffer.
 Analyze complexes or antigens. This can be done by Western Blot.
LIMITATIONS OF USING XENOPUS OOCYTES FOR EXPRESSION OF
EXOGENOUS PROTEINS
 Frog oocytes express endogenous channels and transporters at a low level. These
endogenous proteins can interfere with analysis of expressed proteins in at least two
ways:
1) They can be upregulated by expression of exogenous proteins and interfere with
the analysis of the protein of interest, and
2) They can form heteromultimers with the proteins translated from injected RNA.
 Cellular environment, particularly signaling pathways, may differ from that of the cell
where the ion channel of interest is normally expressed, calling for extreme caution
when studying channel modulation by secondary messengers in oocytes. In addition
there are differences between C. elegans and vertebrates in the types of sugars attached
during glycosylation of membrane proteins and glycosylation affects function and
trafficking of ion channels.
 C. elegans membrane proteins contain a predominance of oligomannose sugars
identical to those found in vertebrates. The more complex and hybrid N-glycans that
are highly abundant in vertebrates are either absent in C. elegans or present at
undetectable levels.
CONCLUSION
 Xenopus laevis oocytes constitute a simple, low-cost and well-controlled system for
expression of exogenous proteins in large quantity.
 This expression system is particularly amenable to the study of membrane proteins using
electrophysiological techniques because of the large size and spherical shape of the
oocyte, both of which facilitate insertion of electrodes and good voltage control.
 Other techniques applied to Xenopus oocytes allow detection and isolation of
membrane proteins using fluorescence and immunochemical tags. These techniques can
be used to complement electrophysiological recordings for a thorough analysis of a
channel or transporter, in its wild type or mutant forms.
 Despite the limitations of oocytes (endogenous channels and transporters, and rearing
temperature) they are still a powerful method for studying several aspects of ion channel
biology, including permeation properties, structure-function relationships, regulation by
intracellular and extracellular molecules and stoichiometry.
REFERENCES
 Gurdon, J.B. (1973). The translation of messenger RNA injected in living oocytes of Xenopus laevis. Acta.
Endocrinol. Suppl. (Copenh) 180, 225–243.
 Gurdon, J.B., Lane, C.D., Woodland, H.R., and Marbaix, G. (1971). Use of frog eggs and oocytes for the study of
messenger RNA and its translation in living cells. Nature 233, 177–182.
 Gurdon, J.B., Lingrel, J.B., and Marbaix, G. (1973). Message stability in injected frog oocytes: long life of
mammalian alpha and beta globin messages. J. Mol. Biol. 80, 539–551.
 Gurdon, J.B., Woodland, H.R., and Lingrel, J.B. (1974). The translation of mammalian globin mRNA injected into
fertilized eggs of Xenopus laevis I. Message stability in development. Dev. Biol. 39, 125–133.
 Hausdorff, S.F., Goldstein, S.A., Rushin, E.E., and Miller, C. (1991). Functional characterization of a minimal K+
channel expressed from a synthetic gene. Biochemistry 30, 3341–3346
 Khanna, R., Myers, M.P., Laine, M., and Papazian, D.M. (2001). Glycosylation increases potassium channel stability
and surface expression in mammalian cells. J. Biol. Chem. 276, 34028–34034.
 Kopito, R.R. (1999). Biosynthesis and degradation of CFTR. Physiol. Rev. 79, S167–S173.
 Krieg, P.A., and Melton, D.A. (1984). Functional messenger RNAs are produced by SP6 in vitro transcription of
cloned cDNAs. Nucleic Acids Res. 12, 7057–7070.
 Sambrook J., S.J., and Maniatis T. (1989). Molecular cloning: a laboratory manual.

Contenu connexe

Similaire à Invivo protein synthesis

Transgenic pig (1) (1)
Transgenic pig (1) (1)Transgenic pig (1) (1)
Transgenic pig (1) (1)
jain_sonia
 
Transgenic pig
Transgenic pig Transgenic pig
Transgenic pig
jain_sonia
 
Lab Differential Expression Differential gene expression provides th.pdf
 Lab Differential Expression Differential gene expression provides th.pdf Lab Differential Expression Differential gene expression provides th.pdf
Lab Differential Expression Differential gene expression provides th.pdf
rita892197
 
Lab Differential Expression Differential gene expression provides .pdf
 Lab Differential Expression Differential gene expression provides .pdf Lab Differential Expression Differential gene expression provides .pdf
Lab Differential Expression Differential gene expression provides .pdf
basilpaul63
 
Nuclear Transport And Its Effect On Breast Cancer Tumor Cells
Nuclear Transport And Its Effect On Breast Cancer Tumor CellsNuclear Transport And Its Effect On Breast Cancer Tumor Cells
Nuclear Transport And Its Effect On Breast Cancer Tumor Cells
Stephanie Clark
 
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Mohamed Khalid Ali Xundhur
 

Similaire à Invivo protein synthesis (20)

Transgenic pig (1) (1)
Transgenic pig (1) (1)Transgenic pig (1) (1)
Transgenic pig (1) (1)
 
Ezhil1
Ezhil1Ezhil1
Ezhil1
 
Exprssion vector
Exprssion vectorExprssion vector
Exprssion vector
 
Synthesis of c dna
Synthesis of c dnaSynthesis of c dna
Synthesis of c dna
 
Transgenic pig
Transgenic pig Transgenic pig
Transgenic pig
 
Protein sorting and transport
Protein sorting and transport Protein sorting and transport
Protein sorting and transport
 
Transplastomics
TransplastomicsTransplastomics
Transplastomics
 
Lab Differential Expression Differential gene expression provides th.pdf
 Lab Differential Expression Differential gene expression provides th.pdf Lab Differential Expression Differential gene expression provides th.pdf
Lab Differential Expression Differential gene expression provides th.pdf
 
Lab Differential Expression Differential gene expression provides .pdf
 Lab Differential Expression Differential gene expression provides .pdf Lab Differential Expression Differential gene expression provides .pdf
Lab Differential Expression Differential gene expression provides .pdf
 
Cloning dna f inal
Cloning dna f inalCloning dna f inal
Cloning dna f inal
 
Intracellular trafficking and protein sorting
Intracellular trafficking and protein sortingIntracellular trafficking and protein sorting
Intracellular trafficking and protein sorting
 
MAMMALIAN CELL EXPRESSION SYSTEM, STRONG PROMOTERS.pptx
MAMMALIAN CELL EXPRESSION SYSTEM, STRONG PROMOTERS.pptxMAMMALIAN CELL EXPRESSION SYSTEM, STRONG PROMOTERS.pptx
MAMMALIAN CELL EXPRESSION SYSTEM, STRONG PROMOTERS.pptx
 
rprotein3
rprotein3rprotein3
rprotein3
 
Abstract conference mbsmb 2009
Abstract conference mbsmb 2009Abstract conference mbsmb 2009
Abstract conference mbsmb 2009
 
Application1
Application1Application1
Application1
 
molecular.pdf
molecular.pdfmolecular.pdf
molecular.pdf
 
Nuclear Transport And Its Effect On Breast Cancer Tumor Cells
Nuclear Transport And Its Effect On Breast Cancer Tumor CellsNuclear Transport And Its Effect On Breast Cancer Tumor Cells
Nuclear Transport And Its Effect On Breast Cancer Tumor Cells
 
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
 
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
Gene expression profiling in apoptotic mcf 7 cells infected with newcastle di...
 
Qualify
QualifyQualify
Qualify
 

Dernier

Conjugation, transduction and transformation
Conjugation, transduction and transformationConjugation, transduction and transformation
Conjugation, transduction and transformation
Areesha Ahmad
 
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
ssuser79fe74
 
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptxSCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
RizalinePalanog2
 

Dernier (20)

❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
❤Jammu Kashmir Call Girls 8617697112 Personal Whatsapp Number 💦✅.
 
Site Acceptance Test .
Site Acceptance Test                    .Site Acceptance Test                    .
Site Acceptance Test .
 
COMPUTING ANTI-DERIVATIVES (Integration by SUBSTITUTION)
COMPUTING ANTI-DERIVATIVES(Integration by SUBSTITUTION)COMPUTING ANTI-DERIVATIVES(Integration by SUBSTITUTION)
COMPUTING ANTI-DERIVATIVES (Integration by SUBSTITUTION)
 
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 60009654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
9654467111 Call Girls In Raj Nagar Delhi Short 1500 Night 6000
 
module for grade 9 for distance learning
module for grade 9 for distance learningmodule for grade 9 for distance learning
module for grade 9 for distance learning
 
Conjugation, transduction and transformation
Conjugation, transduction and transformationConjugation, transduction and transformation
Conjugation, transduction and transformation
 
Kochi ❤CALL GIRL 84099*07087 ❤CALL GIRLS IN Kochi ESCORT SERVICE❤CALL GIRL
Kochi ❤CALL GIRL 84099*07087 ❤CALL GIRLS IN Kochi ESCORT SERVICE❤CALL GIRLKochi ❤CALL GIRL 84099*07087 ❤CALL GIRLS IN Kochi ESCORT SERVICE❤CALL GIRL
Kochi ❤CALL GIRL 84099*07087 ❤CALL GIRLS IN Kochi ESCORT SERVICE❤CALL GIRL
 
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptxPSYCHOSOCIAL NEEDS. in nursing II sem pptx
PSYCHOSOCIAL NEEDS. in nursing II sem pptx
 
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
Chemical Tests; flame test, positive and negative ions test Edexcel Internati...
 
CELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdfCELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdf
 
pumpkin fruit fly, water melon fruit fly, cucumber fruit fly
pumpkin fruit fly, water melon fruit fly, cucumber fruit flypumpkin fruit fly, water melon fruit fly, cucumber fruit fly
pumpkin fruit fly, water melon fruit fly, cucumber fruit fly
 
SAMASTIPUR CALL GIRL 7857803690 LOW PRICE ESCORT SERVICE
SAMASTIPUR CALL GIRL 7857803690  LOW PRICE  ESCORT SERVICESAMASTIPUR CALL GIRL 7857803690  LOW PRICE  ESCORT SERVICE
SAMASTIPUR CALL GIRL 7857803690 LOW PRICE ESCORT SERVICE
 
FAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
FAIRSpectra - Enabling the FAIRification of Spectroscopy and SpectrometryFAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
FAIRSpectra - Enabling the FAIRification of Spectroscopy and Spectrometry
 
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptxSCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
SCIENCE-4-QUARTER4-WEEK-4-PPT-1 (1).pptx
 
Connaught Place, Delhi Call girls :8448380779 Model Escorts | 100% verified
Connaught Place, Delhi Call girls :8448380779 Model Escorts | 100% verifiedConnaught Place, Delhi Call girls :8448380779 Model Escorts | 100% verified
Connaught Place, Delhi Call girls :8448380779 Model Escorts | 100% verified
 
High Profile 🔝 8250077686 📞 Call Girls Service in GTB Nagar🍑
High Profile 🔝 8250077686 📞 Call Girls Service in GTB Nagar🍑High Profile 🔝 8250077686 📞 Call Girls Service in GTB Nagar🍑
High Profile 🔝 8250077686 📞 Call Girls Service in GTB Nagar🍑
 
Zoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdfZoology 5th semester notes( Sumit_yadav).pdf
Zoology 5th semester notes( Sumit_yadav).pdf
 
GBSN - Microbiology (Unit 1)
GBSN - Microbiology (Unit 1)GBSN - Microbiology (Unit 1)
GBSN - Microbiology (Unit 1)
 
9999266834 Call Girls In Noida Sector 22 (Delhi) Call Girl Service
9999266834 Call Girls In Noida Sector 22 (Delhi) Call Girl Service9999266834 Call Girls In Noida Sector 22 (Delhi) Call Girl Service
9999266834 Call Girls In Noida Sector 22 (Delhi) Call Girl Service
 
Unit5-Cloud.pptx for lpu course cse121 o
Unit5-Cloud.pptx for lpu course cse121 oUnit5-Cloud.pptx for lpu course cse121 o
Unit5-Cloud.pptx for lpu course cse121 o
 

Invivo protein synthesis

  • 1. IN-VIVO PROTEIN SYNTHESIS : FROG OOCYTE SYSTEM
  • 2. INTRODUCTION  In 1971 Gurdon demonstrated that oocytes extracted from the South-African clawed frog Xenopus laevis were able to synthesize hemoglobin consequent to injection of the corresponding mRNA (Gurdon et al., 1971).  Oocytes are particularly well suited for expression of proteins following mRNA injection, since they contain accumulated stores of enzymes, organelles and proteins that are normally used after fertilization that can be recruited for heterologous proteins.  The most common approach for expression of exogenous proteins in Xenopus oocytes is to inject in vitro transcribed complementary RNA into the oocyte cytoplasm. Expression can be also achieved by injection of the cDNA into the oocyte nucleus, but this method requires visual localization of the nucleus and involves the risk of damaging the nuclear membrane during injection; which renders this approach more technically challenging and less efficient.
  • 3.  Oocytes of X. laevis are used because of the following reasons; 1) X. laevis is easily maintained in captivity, making it a low-cost laboratory animal. 2) Oocytes can be readily harvested and, because of their large diameter (1-1.2 mm), are straightforward to injected. 3) frog oocytes are built to survive for long periods of time outside the body 4) Oocytes express a low number of endogenous membrane transporters and channels because they are virtually independent from exogenous nutrients. 5) Multiple species of mRNA can be simultaneously injected to study protein complexes formed by multiple subunits, an advantage over transfected mammalian cells for studying large multi-protein complexes. 6) Standard electrophysiological techniques are easily applicable.
  • 4. XENOPUS DEVELOPMENT AND CHARACTERISTICS  A Xenopus oocyte is an immature egg that under the appropriate hormonal stimulation can become competent for fertilization. The egg production is controlled by subcutaneous injections of 50-100 units of pregnant mare serum (PMS) in their dorsal lymph sacs several days before the second injection with 600-800 units of human chorionic gonadotropin (hCG) 12-16 hours before the desirable egg laying. Right after the second injection, they are placed in individual tanks and the next day the eggs are collected by by surgical procedures.  Oocytes go through six maturation stages (I-VI); stage V and VI oocytes are generally used for electrophysiological studies and are the largest. These large cells (1-1.2 mm diameter) have a black pigmented region called the animal pole and a white (non-pigmented) vegetal pole.
  • 5.  They contain a huge nucleus (or germinal vesicle), 100,000 times larger than a somatic cell nucleus, which occupies approximately the one-third of the oocyte’s volume and they develop rapidly. The vesicle is surrounded by a nuclear envelope with large pores that facilitate transportation from and to the cytoplasm.  The oocyte plasma membrane is surrounded by a vitelline membrane, that helps to maintain the oocyte shape and renders the oocyte more resistant to manipulations. The vitelline membrane does not affect electrophysiological recordings since it is devoid of channels and transporters and has a large-enough mesh to allow permeation of ions and small molecules. Fig. 1
  • 6.  However, for single channel recordings the vitelline membrane is removed because it hinders the formation of high resistance seal between the patch pipette and the oocyte membrane. Around the vitelline membrane is a layer of follicular cells that separates the oocyte from the external environment (Figure 1).  By contrast to the vitelline membrane, the follicular cells express ion channels and transporters they are electrically coupled to each other and to the oocyte by gap junctions and therefore can create serious interference during electrophysiological recordings. For these reasons this layer of cells is eliminated prior to injection by a combination of collagenase treatment and manual stripping.  The material that Xenopus can offer in biology is great in terms of quality and quantity. For example, one frog ovary corresponds to 1000 mouse ovaries and western blots can be performed with protein samples from just one oocyte (or 1µL of extract).  Oocytes are very rich in all the necessary RNA and proteins for the first stages of development until early embryogenesis, i.e., the tadpole stage. They can contain up to 4 µg of ribosomal RNA, therefore each oocyte is able of synthesizing up to 400 ng of proteins per day.  In addition, they lack transcriptional activity until the mid-late blastula stage. It is estimated that the protein content is about 500 µg in each oocyte and approximately 10-50 mg/mL in the extract.
  • 7. CDNA CLONING INTO THE APPROPRIATE VECTORS  When a cDNA sequence has not been experimentally confirmed, it is important to keep in mind that less conserved regions (usually 5′ and 3′ end) may be mispredicted. Moreover, splice variants with different functional properties or cell-specific expression may exist and should be considered whenever attempting to fully characterize a protein using an expression system such as Xenopus oocytes.  When experimentally confirmed, a cDNA may be available as full length or overlapping EST (Expression Sequence Tag).  For genes for which cDNA clones are not available, cDNA can be cloned relying on sequence information contained in highly conserved regions of the gene or using non-biased methods based on RACE-PCR to identify extreme 3′ and 5′ ends.
  • 8. FEATURES OF THE PREFERRED VECTORS  Cloned cDNA must be inserted into a plasmid that ensures stability of the transcribed sequence and efficient translation into Xenopus oocytes.  Vectors such as pSP64T (Promega), pGEM (Promega) and pCDNA3 (Invitrogen) all contain polyadenylation (poly(A) sequences at the 3′ end of their cloning linker that render transcribed RNA stable and are suitable for oocytes expression.  In addition plasmid pSP64T contains the 5′ and 3′ UTRs from Xenopus laevis β-globin. The β- globin sequences greatly enhance translation efficiency of heterologous mRNA transcripts in oocytes. The vectors contain also RNA polymerase promoter sites (SP6 in pSP64T and T7 in PGEM and pCDNA3) that are used for the in vitro transcription.
  • 9. IN- VITRO RNA SYNTHESIS 1. Choosing the right kit  In vivo most eukaryotic mRNAs have a 7-methyl guanosine cap structure at their 5′ end required for efficient message binding to ribosomes and efficient translation.  When in vitro transcribing RNA for oocyte expression studies, it is important to use a transcription kit that adds a guanosine cap to newly synthesized cRNA molecules.  Failing to do so will dramatically reduce the expression of the protein of interest. (mMESSAGE mMACHINE kits from Ambion contain a capped analog that is incorporated as the first G of the transcript). Ambion markets three kits containing three different RNA polymerases: SP6, T7 and T3. Depending on the promoter sequence of the vector employed and/or on the orientation of the cDNA, the appropriate kit to generate a sense transcript must be used.
  • 10. 2. Plasmid template • DNA should be free of contaminating proteins and RNA. Most commercially available plasmid preparation systems yield DNA that is appropriate for in vitro RNA transcription using a transcription kit. Alternatively, DNA could be purified by standard phenol/chloroform/ isoamyl alcohol extraction. 3. Linearization of the DNA template • Circular plasmid templates generate long and heterogeneous RNA transcripts because RNA polymerases are highly processive. To produce a homogeneous population of transcripts, plasmid DNA should be linearized by digesting with a restriction enzyme that cuts at the 3′ end of the inserted cDNA. The restriction enzyme should be chosen with care. An enzyme that leaves 5′ end overhangs is optimal. Restriction enzymes leaving 3′ overhanging ends (for example Kpn I, Pst I, etc) are the least desirable because they yield low level of transcripts.
  • 11. PROTOCOL Digest plasmid DNA (5-10 µg) in a large volume reaction (150-200 µl) for at least 3 hours to ensure complete digestion of the template. Electrophorese 5 µl of the digestion mix to check for complete digestion. Clean the digested DNA from restriction enzyme and restriction buffer either by phenol/chloroform/isoamyl alcohol extraction or using a DNA purification kit. PCR purification kit from QIAGEN is appropriate and recovers DNA at high yield. Elute or resuspend the DNA with 50 µl RNase-free water. Ethanol precipitate DNA overnight at −20°C using 30 µl RNase-free 3 M sodium acetate and 1.2 ml 100% ethanol. Centrifuge 12,000g for 30 min at 4°C, wash once with 70% ethanol, decant supernatant completely and resuspend in 8 µl RNase-free water. Run 1 µl to check recovery of DNA and estimate its concentration by comparison with an appropriate size standard of known quantity or by measuring O.D.
  • 12. XENOPUS OOCYTES - ISOLATION & ENUCLEATION  Microinjection of Xenopus oocytes has proven to be a valuable tool in a broad array of studies that require expression of DNA or RNA into functional protein.  These studies are diverse and range from expression cloning to receptor–ligand interaction to nuclear programming.  Oocytes offer a number of advantages for such studies, including their large size (1.2 mm in diameter), capacity for translation, and enormous nucleus (0.3–0.4 mm).  They are cost effective, easily manipulated, and can be injected in large numbers in a short time period. Oocytes have a large maternal stockpile of all the essential components for transcription and translation.  Consequently, the investigator needs only to introduce by microinjection the specific DNA or RNA of interest for synthesis.  Oocytes translate virtually any exogenous RNA regardless of source, and the translated proteins are folded, modified, and transported to the correct cellular locations.
  • 13.  Xenopus laevis ovaries are large and transparent and may contain several hundreds of, also large, oocytes which are easily obtainable.  Oocytes are prepared from Xenopus laevis ovaries that can be extracted from adult females.  The oocytes can be manually dissected using forceps from the ovaries or they can be extracted by digestion with collagenase (enzymatic defolliculation) of the four extracellular connective tissue layers that surround them.  Then, from the oocytes, the nucleus can be manually removed. A pair of forceps with sharp tips is used to pierce a small opening on the cell and by squeezing force applied by another pair of forceps with blunt tips the nucleus is extracted.  This results in oocytes without any nucleus (enucleated oocyte) and isolated nuclei. The experimental protocol with which the enucleation takes place is selected according to the downstream application, which can be immunoblotting.  Injection of a somatic nuclei and RNA synthesis for analysis of transcription, Fully functional nuclei can be isolated and be used to study all nuclear processes including gene expression, chromatin dynamics, nuclear import and export of fluorescently labeled proteins or function of the nuclear pore complexes
  • 14. OOCYTE PROCUREMENT PROTOCOL Anesthetize the adult female. Place the frog on crushed ice for approximately 20 min prior to, and throughout, the surgical procedure to maintain anesthesia. Make a 1 cm incision in the lower abdominal wall near one leg to expose an ovary. Carefully remove about one third of one ovary and place it in OR2 with CaCl2 or ND96 with CaCl2. Dissociate oocytes by collagenase treatment. Incubate for 1hr with gentle agitation at room temperature, Removal of the follicular layer is actually easier several hours after collagenase treatment; in addition oocytes tend to be more fragile right after enzymatic treatment. Manually defolliculated stage V and VI oocytes by forcing them through a pipette with an opening slightly smaller than the diameter of the oocytes (smooth the opening by fire polishing). The higher concentration of collagenase allows defolliculation of most oocytes that can be set aside for injection.
  • 15.
  • 16. OOCYTE MICROINJECTION  The volume of one Xenopus laevis oocyte is approximately 1µL. They can tolerate an injection of up to 50 nL using automatic nanoinjectors, without any adverse effect in the function.  Injections can be used for either gain-of-function (GOF) or loss-of-function (LOF) experiments or experiments involving both.  There are two routes for introducing the foreign genetic information into the oocyte and both require injection of the cell.  The chief problem for cytoplasmic injections is the instability of the mRNA, for stability poly (A) tail is added. The mRNA have a 5’7-methyl guanosine residue cap structure which functions in the protein synthesis initiation process, protects the mRNA from degradation and is essential for oocyte expression.  Oocyte expression importance of both capping and the poly(A) tail of the mRNA expression in oocytes changes during oocyte development, but the two processes work synergistically stimulating translation as the oocyte matures, This is one reason for checking that the oocytes chosen for injection are at the correct developmental stage to optimize expression.  A different approach is to directly inject the nucleus with double-stranded cDNA. However, the cDNA must be cloned into a suitable expression vector before nuclear
  • 17.
  • 18. 1) dissecting microscope 2) cold light source power supply, 3) cold light source goose-necks, 4) microinjector mounted on a coarse micromanipulator, 5) switch pedal. • Oocyte injection chamber with microchip sockets for holding oocytes during injection.
  • 19. PROTOCOL Immobilize oocytes by positioning them into the openings of a nylon mesh at the bottom of a Petri dish containing culture medium. Prepare 50–100 oocytes at a time for microinjection. gently withdraw the pipette and move to the next oocyte by moving the microinjection dish. Inject the sample, then wait a few seconds before withdrawing the pipette to minimize sample leakage. Periodically release one injection shot into solution to ensure the pipette has not clogged. When microinjection is complete, carefully separate the oocytes into individual dishes or microplate wells containing the same medium used during injection Return the oocytes to the 18˚C incubator. Twice a day, monitor the oocytes for viability, removing damaged or dying oocytes to prevent deleterious effects on healthy oocytes. Include antibiotics, such as gentamycin or streptomycin, in the culture medium to prevent bacterial infection
  • 20.  The voltage clamp is an electrophysiological technique used to measure ion current through the cell membrane of excitable cells such as oocytes and neurons while holding the membrane voltage to desired level.  The TEVC technique is used to study properties of membrane proteins especially ion channels.  Researchers use this method most commonly to investigate membrane structures expressed in xenopus oocyte.  This technique allows to measure the current across the cell membrane.  Voltage clamping allows for the potential in the system to be set to a specific voltage while recording the current. TWO ELECTRODE VOLTAGE CLAMP
  • 21. PRINCIPLE OF TEVC  The TEVC method utilizes two low- resistance pipettes, one is for sensing voltage and the other is for injecting current.  The micro electrodes are filled with conducting solution and inserted into the cell to artificially control membrane potential.  The microelectrodes compare the membrane potential against a command voltage, giving an accurate reproduction of the currents flowing across the membrane.  Current reading can be used to analyze the electrical response of the cell to different application.
  • 22. TEVC IN XENOPUS OOCYTE  In Xenopus oocytes, the electrodes are filled with a 3M KCl solution and have an electric resistance between 0.2 and 3Ω. Other solutions with lower KCl concentrations and containing Ca2+ buffers such as EGTA can be used.  Ca2+ buffers are used to inhibit endogenous Ca2+-activated Cl-currents.  The electrodes are mounted on electrode-holders that contain a silver wire coated with a layer of AgCl2 which allows transmission of the signal from the KCl solution through the silver wire to the amplifier. The ground electrodes are made of 3M KCl in 2-3% agar.  After both electrodes have been inserted, the amplifier is set in voltage-clamp mode and the voltage-clamp feedback gain, used to compensate for membrane capacitance, is increased to speed up the time response of the voltage clamp.  Currents are then filtered and recorded using an analog/digital converter and a computer.
  • 23. WHY TEVC IS USED OVER OTHER VOLTAGE CLAMP TECHNIQUES TEVC offers many advantages over other techniques such as:- 1. Having a high current-passing capacity. 2. Able to inject current into the system for increased sensitivity. 3. Increased signal-to-noise ratio (SNR). Increasing sensitivity and SNR is important in a scanning ion conductance microscope. It increases the ionic conductance of the system, which allows for higher sensitivity and low noise when scanning a sample and its features.
  • 24. DETECTING PROTEINS AT THE OOCYTE SURFACE BY FLUORESCENT METHOD  Detecting expressed proteins at the plasma membrane of Xenopus oocytes can be used to examine their trafficking to the membrane, their subcellular distribution or even interaction with other proteins.  When drawing conclusions on protein trafficking based on observations made in Xenopus oocytes though, it should be kept in mind that oocytes are reared at 19°C– 20°C while the organism from which the protein originates may have a different vital temperature.  Studies on mammalian membrane proteins may be complicated by this problem while for C. elegans proteins this factor may have less of an impact (nematodes can be reared at a wide range of temperatures from 15°C to 25°C).  Nevertheless, it is important to keep in mind that certain mutant phenotypes are temperature-sensitive and a protein that is at the cell surface at 20°C may display a traffic defect at 25°C .
  • 25.  Membrane proteins can be visualized at the oocyte plasma membrane in two ways: 1) By tagging them with fluorescent proteins such as Green Fluorescent Protein (GFP) or 2) By immunostaining  There are at least three different protocols for oocyte immunostaining: two in which the oocytes are fixed and sliced prior to incubation with the antibodies and one in which they are sliced after incubation with antibodies.  For visualization of GFP-tagged proteins at the plasma membrane, oocytes must be fixed and sliced. Fixing and slicing methods described in protocol 1 are well-suited for visualization of GFP-tagged proteins.
  • 26. Protocol. 1 Fix oocytes overnight at 4°C with 4% paraformaldehyde in PBS. Wash three times for 5 minutes with PBS and imbed the oocytes in 3% low-melting point agarose in PBS. Pour enough warm (not hot) agarose in a 35 mm Petri dish to cover the bottom, drop 3 to 4 oocytes on the surface and cover with another layer of agarose. Incubate the imbedded oocytes at 4°C for at least 2 hours to allow hardening of the agarose and then, using a vibrotome, section into 50 µm slices. Block non-specific sites by incubation with 0.2% Bovine Serum Albumin (BSA) plus 0.1% Tween 20 in PBS overnight at 4°C or for 3–4 hours at room temperature. Transfer the slices from one solution to another using a fire-polished Pasteur pipette.
  • 27. Incubate with the primary antibody in 1% Bovine Serum Albumin (BSA) plus 0.1% Tween 20 in PBS preferably over night at 4°C. Wash three times for 5 minutes with PBS and incubate with a fluorescent secondary antibody in 1% Bovine Serum Albumin (BSA) plus 0.1% Tween 20 in PBS for 1 hour at room temperature. Wash three times for 5 minutes in PBS and mount the slices on glass slides using a medium appropriate for fluorescent microscopy Photograph slices using an inverted microscope equipped with 4x and 20x objectives. Microscopes typically used for C. elegans photography are well suited for oocyte photography.
  • 28. Examples of oocyte sections stained using this protocol . Immunostaining of C. elegans in Xenopus oocytes using a polyclonal antibody (a) Fluorescent photograph of an immune stained non-injected oocyte, showing no background fluorescence at the plasma membrane. (b) Oocytes expressing MEC- 4(d) stained with anti-MEC-4 antibodies. (C) Oocytes expressing MEC- 4(A713T/ S726F) stained with anti-MEC-4 antibodies.
  • 29. Protocol. 2 (Use this protocol to detect FLAG-tagged proteins) Fix the oocytes with 3% paraformaldehyde in PBS for 4 h. Place fixed oocytes on thin cork disks, embedded into cryo-embedding compound and freeze them in liquid propane cooled by liquid nitrogen. Store the oocytes at −80°C until further use. Section the oocytes in 6 µm slices using a cryostat and place them on chrom-alum gelatin-coated glass slides. Use the tyramide signal amplification (TSA-Direct) kit according to the manufacturer's instructions. Incubate with anti-FLAG antibody diluted 1:100 in the TSA blocking buffer for 1 hour at room temperature. Rinse slices with PBS containing 0.05% Tween and incubate with a 1:100 dilution of horseradish peroxidase- conjugated sheep anti-mouse Ig (1 hour at room temperature). Wash with PBS-Tween and incubate with FITC-tyramide conjugates diluted 1:50 in the TSA diluent (1 hour at room temperature). Wash slices in PBS-Tween and mount them in DAKO-Glycergel containing 2.5% of 1,4-diazabicyclo (2.2.2)-octane as a fading retardant.
  • 30. IMMUNOPRECIPITATION  Immunoprecipitation (IP) is the technique of precipitating a protein antigen out of solution using an antibody that specifically binds to that particular protein.  This process can be used to isolate and concentrate a particular protein from a sample containing many thousands of different proteins.  Immunoprecipitation requires that the antibody be coupled to a solid substrate at some point in the procedure.  Antigens isolated by IP are analyzed by SDS-PAGE or Western blotting.
  • 31. TYPES OF IMMUNOPRECIPITATION There are different types of immunoprecipitation: 1. Individual protein immunoprecipitation (IP): Involves using an antibody that is specific for a known protein to isolate that particular protein out of a solution containing many different proteins. 2. Protein complex immunoprecipitation (Co-IP): Immunoprecipitation of intact protein complexes is known as co-immunoprecipitation (Co-IP). Co-IP works by selecting an antibody that targets a known protein that is believed to be a member of a larger complex of proteins. 3. Chromatin immunoprecipitation (ChIP): Chromatin immunoprecipitation (ChIP) is a method used to determine the location of DNA binding sites on the genome for a particular protein of interest. This technique gives a picture of the protein–DNA interactions that occur inside the nucleus of living cells or tissues. 4. RNP immunoprecipitation targets ribonucleoproteins (RNPs): Live cells are first lysed and then the target protein and associated RNA are immunoprecipitated using an antibody targeting the protein of interest
  • 32. EXTRACTING PROTEINS FROM XENOPUS OOCYTES FOR IMMUNOPRECIPITATION  The most important step when extracting proteins from Xenopus oocytes is to eliminate the large amount of yolk, which interferes with electrophoresis and immunodetection.  This allows elimination of the yolk and extraction of membrane proteins from Xenopus oocytes.
  • 33. STEPS OF IMMUNOPRECIPITATION  Lyse cells and prepare sample for immunoprecipitation.  Pre-clear the sample by passing the sample over beads alone or bound to an irrelevant antibody to soak up any proteins that non- specifically bind to the IP components.  Incubate solution with antibody against the protein of interest. Antibody can be attached to solid support before this step (direct method) or after this step (indirect method). Continue the incubation to allow antibody-antigen complexes to form.  Precipitate the complex of interest, removing it from bulk solution.  Wash precipitated complex several times. Spin each time between washes when using agarose beads or place tube on magnet when using superparamagnetic beads and then remove the supernatant. After the final wash, remove as much supernatant as possible.  Elute proteins from the solid support using low-pH or SDS sample loading buffer.  Analyze complexes or antigens. This can be done by Western Blot.
  • 34. LIMITATIONS OF USING XENOPUS OOCYTES FOR EXPRESSION OF EXOGENOUS PROTEINS  Frog oocytes express endogenous channels and transporters at a low level. These endogenous proteins can interfere with analysis of expressed proteins in at least two ways: 1) They can be upregulated by expression of exogenous proteins and interfere with the analysis of the protein of interest, and 2) They can form heteromultimers with the proteins translated from injected RNA.  Cellular environment, particularly signaling pathways, may differ from that of the cell where the ion channel of interest is normally expressed, calling for extreme caution when studying channel modulation by secondary messengers in oocytes. In addition there are differences between C. elegans and vertebrates in the types of sugars attached during glycosylation of membrane proteins and glycosylation affects function and trafficking of ion channels.  C. elegans membrane proteins contain a predominance of oligomannose sugars identical to those found in vertebrates. The more complex and hybrid N-glycans that are highly abundant in vertebrates are either absent in C. elegans or present at undetectable levels.
  • 35. CONCLUSION  Xenopus laevis oocytes constitute a simple, low-cost and well-controlled system for expression of exogenous proteins in large quantity.  This expression system is particularly amenable to the study of membrane proteins using electrophysiological techniques because of the large size and spherical shape of the oocyte, both of which facilitate insertion of electrodes and good voltage control.  Other techniques applied to Xenopus oocytes allow detection and isolation of membrane proteins using fluorescence and immunochemical tags. These techniques can be used to complement electrophysiological recordings for a thorough analysis of a channel or transporter, in its wild type or mutant forms.  Despite the limitations of oocytes (endogenous channels and transporters, and rearing temperature) they are still a powerful method for studying several aspects of ion channel biology, including permeation properties, structure-function relationships, regulation by intracellular and extracellular molecules and stoichiometry.
  • 36. REFERENCES  Gurdon, J.B. (1973). The translation of messenger RNA injected in living oocytes of Xenopus laevis. Acta. Endocrinol. Suppl. (Copenh) 180, 225–243.  Gurdon, J.B., Lane, C.D., Woodland, H.R., and Marbaix, G. (1971). Use of frog eggs and oocytes for the study of messenger RNA and its translation in living cells. Nature 233, 177–182.  Gurdon, J.B., Lingrel, J.B., and Marbaix, G. (1973). Message stability in injected frog oocytes: long life of mammalian alpha and beta globin messages. J. Mol. Biol. 80, 539–551.  Gurdon, J.B., Woodland, H.R., and Lingrel, J.B. (1974). The translation of mammalian globin mRNA injected into fertilized eggs of Xenopus laevis I. Message stability in development. Dev. Biol. 39, 125–133.  Hausdorff, S.F., Goldstein, S.A., Rushin, E.E., and Miller, C. (1991). Functional characterization of a minimal K+ channel expressed from a synthetic gene. Biochemistry 30, 3341–3346  Khanna, R., Myers, M.P., Laine, M., and Papazian, D.M. (2001). Glycosylation increases potassium channel stability and surface expression in mammalian cells. J. Biol. Chem. 276, 34028–34034.  Kopito, R.R. (1999). Biosynthesis and degradation of CFTR. Physiol. Rev. 79, S167–S173.  Krieg, P.A., and Melton, D.A. (1984). Functional messenger RNAs are produced by SP6 in vitro transcription of cloned cDNAs. Nucleic Acids Res. 12, 7057–7070.  Sambrook J., S.J., and Maniatis T. (1989). Molecular cloning: a laboratory manual.