1. dia

advertisement

Manifestation of Novel Social Challenges of the European Union in the Teaching Material of

Medical Biotechnology Master’s Programmes at the

University of Pécs and at the University of Debrecen

Identification number : TÁMOP-4.1.2-08/1/A-2009-0011

Manifestation of Novel Social Challenges of the European Union in the Teaching Material of

Medical Biotechnology Master’s Programmes at the

University of Pécs and at the University of Debrecen

Identification number : TÁMOP-4.1.2-08/1/A-2009-0011

Beáta Scholtz

Molecular Therapies- Lecture 3

RECOMBINANT PROTEINS

RECOMBINANT PROTEINS

TÁMOP-4.1.2-08/1/A-2009-0011

The aim of this lecture is to describe the in vitro and in vivo systems utilized for expression of recombinant proteins, and discuss the advantages and disadvantages of these systems. We will also discuss the basics of affinity-tag based protein purification.

1.1 OVERVIEW: PROTEIN PHARMACEUTICALS

1.2 CELL-FREE SYSTEMS: IN VITRO TRANSCRIPTION AND TRANSLATION

1.3 EXPRESSION OF RECOMBINANT PROTEINS IN CELL CULTURE

1.4 NON-PROKARYOTIC EXPRESSION SYSTEMS

1.4.1 Pichia pastoris

1.4.2 Protein expression in insect cells

1.4.3 Mammalian expression systems

1.5 PURIFICATION OF RECOMBINANT PROTEINS

Pure protein preparations

TÁMOP-4.1.2-08/1/A-2009-011

Uses: medicine and research

Sources:

• natural protein mixtures - human/animal/fungi/plant

• artificial preparations - synthetic peptides, recombinant proteins

Protein pharmaceutical

Insulin

Factor VIII

Human growth hormone

Calcitonin

Anti-venom

Natural Source

Pigs or cattle (pancreas)

Human blood (donated)

Human brains

Salmon

Horse or goat blood

4

TÁMOP-4.1.2-08/1/A-2009-011

Equipment used for blood fractionation

5

Antivenom - specific antiserum from goat or horse

TÁMOP-4.1.2-08/1/A-2009-011

B. Rogge Box jellyfish,

Australia

R. Morante

Lonomia caterpillar, Brasil

P-A. Olsson

Black scorpion, Arabia 6

Protein pharmaceuticals

TÁMOP-4.1.2-08/1/A-2009-011

Natural sources are often rare and expensive

Difficult to keep up with demand

Hard to isolate product

May lead to immune reactions (diff. species)

Viral & pathogen contamination

Most protein pharmaceuticals today are produced recombinantly

Cheaper, safer, abundant supply

7

TÁMOP-4.1.2-08/1/A-2009-011

Peptide drugs

Many hormones are actually small peptides (2-40 amino acids)

Calcitonin (32 residues)

Thyroid hormone to enhance bone mass

Oxytocin (9 residues)

Pituitary hormone to stimulate labor

Vasopressin (9 residues)

Pituitary hormone for antidiuretic/vasoconstriction

8

TÁMOP-4.1.2-08/1/A-2009-011

Peptide drugs

Small enough to synthesize using solid phase chemistry (SPPS)

Method developed by Bruce Merrifield in 1960’s (won Nobel prize)

Very efficient synthesis (>99%/couple)

Still: 50 residue peptide, 99% coupling

Yield = 0.99

50 = 60.5%

Technique limited to small peptides

9

TÁMOP-4.1.2-08/1/A-2009-011

Recombinant proteins

Developed in 1970’s &1980’s

Paul Berg (1973) restriction enzymes

Herbert Boyer (1978) cloning human insulin into E. coli –

Genentech

Four general approaches

Expression in cell-free systems

Expression in isolated cells

Expression in transgenic plants/animals

Gene therapy in humans

10

TÁMOP-4.1.2-08/1/A-2009-011

Cell-free systems:

In vitro transcription and translation

• Rapid identification of gene products

• Functional analyses

• Analyze protein-protein interactions

• Study protein folding

• Incorporate modified amino acids for functional studies

• Engineer truncated gene products

11

Cell-free systems:

In vitro transcription and translation

TÁMOP-4.1.2-08/1/A-2009-011

Advantages over in vivo gene expression:

When the protein is: toxic to the host cell insoluble or forms inclusion bodies degraded rapidly by intracellular proteases

Speed and directness of all procedures

Absence of constraints from a living cell

Pure product

Disadvantages over in vivo gene expression:

Lack of cellular membranes

Lack of post translational modifications

12

Components for in vitro transcription

TÁMOP-4.1.2-08/1/A-2009-011

In vivo In vitro

• Linearized DNA template

• Phage RNA polymerase

• 4dNTP

• Buffer

1998 by Alberts, Bray, Johnson, Lewis, Raff, Roberts, Walter.

Published by Garland Publishing.

13

TÁMOP-4.1.2-08/1/A-2009-011

Phage RNA polymerases

Phage Polymerase

T7 RNA polymerase

T3 RNA polymerase

SP6 RNA polymerase

Host of Phage

E. coli

E. coli

Salmonella typhimurium

Promoter Sequence

5’TAATACGACTCACTATAGGG 3’

5’AAATTAACCCTCACTAAAGGG3’

5’AATTTAGGTGACACTATAGAA3’

14

Characteristics of RNA polymerases

TÁMOP-4.1.2-08/1/A-2009-011

RNA polymerases proceed at a much slower rate than DNA polymerases.

RNA pol (50-100 bases/sec)

DNA pol (1000 bases/sec)

The fidelity of RNA synthesis is much lower than that of DNA.

RNA polymerases do not contain proofreading mechanisms.

15

TÁMOP-4.1.2-08/1/A-2009-011

DNA template

Plasmids

Many commonly used cloning vectors contain phage polymerase promoters outside of the multiple cloning site.

PCR Products

Primer must contain promoter

Oligonucleotides

16

TÁMOP-4.1.2-08/1/A-2009-011

Linearization of template

Plasmids: no RNA polymerase termination signal; templates are linearized

• PCR template: termination signal in the amplified region OR in the primer

17

Translation in eukaryotic cells

TÁMOP-4.1.2-08/1/A-2009-011

18 1998 by Alberts et al.

Published by Garland Publishing.

Components for in vitro translation

• tRNA & aminoacyl-tRNA synthetases

• Ribosomes

• Amino acids

• ATP, GTP

• Initiation, elongation, and termination factors

• Buffer

• RNA template

TÁMOP-4.1.2-08/1/A-2009-011

Much more complex than transcription

Cannot be mixed from a few isolated components

Always provided as crude extract of cells

19

Common in vitro translation systems

TÁMOP-4.1.2-08/1/A-2009-011

Rabbit reticulocyte lysate

Wheat germ extract

E. coli extract

20

TÁMOP-4.1.2-08/1/A-2009-011

Rabbit reticulocyte lysates

Reticulocytes: immature red blood cells no nuclei (DNA) complete translation machinery, for extensive globin synthesis

Endogenous globin mRNA can be eliminated by incubation with a

Ca 2+ dependent micrococcal nuclease. The nuclease is later inactivated by EGTA.

Low background

Efficient utilization of exogenous RNAs, even at low concentrations

Low nuclease activity

Capable of synthesizing large amounts of full-length products

Capable of translating either capped or uncapped RNAs

21

TÁMOP-4.1.2-08/1/A-2009-011

Wheat germ lysates

Low background incorporation due to low levels of endogenous mRNA

Recommended for translation of RNA containing small fragments of doublestranded RNA or oxidized thiols, which are inhibitory to the rabbit reticulocyte lysate

Generally more cap dependent than reticulocyte systems

Often preferable when synthesizing relatively small proteins (12-15kDa) that comigrate with globin, which is abundant in reticulocyte extracts

22

TÁMOP-4.1.2-08/1/A-2009-011

E. coli lysates

Simple translational apparatus and less complicated initiation control mechanisms

BUT: bacterial extracts contain nucleases that rapidly degrade most exogenous RNAs

Extract must be incubated during preparation so that excess endogenous mRNA is translated and subsequently degraded

The exogenous product is easily identifiable

23

TÁMOP-4.1.2-08/1/A-2009-011

Translation systems

Two approaches to cell free protein synthesis:

Standard translation systems (reticulocyte and wheat extracts) use RNA as a template

Linked or coupled transcription+translation systems start with DNA templates

Important elements for translation:

= Eukaryotic translation signal: 5’-GCC A CCAUG G -3’ “Kozak” sequence, if eukaryotic cell free translation system is used

= Prokaryotic translation signals: 5’-UAAGGAGGUGA-3’ Shine-

Delgarno (SD) , if prokaryotic cell free translation system is used

Linked system: tube 1.=transcription, tube 2.= translation.

= Each can be optimized separately.

Coupled system: both reactions in the same tube

24

TÁMOP-4.1.2-08/1/A-2009-011

Main steps of recombinant protein production

In vivo Cell free

Identification/Isolation of gene of interest

Cloning of gene into plasmid

Plasmid: expression vector

Transformation into host cells

Growth of cells through fermentation

Plasmid: source of DNA template for transcription

In vitro transcription

In vitro translation

Isolation & purification of protein

Formulation of protein product

TÁMOP-4.1.2-08/1/A-2009-011

Recombinant protein expression in cells or organisms

Escherichia coli/ Other bacteria

Pichia pastoris/ Other yeast

Insect cell culture (Baculovirus)

Mammalian cell culture

Plants

Sheep/cows/humans

(transgenics and gene therapy)

26

TÁMOP-4.1.2-08/1/A-2009-011

Expression system selection

Choice depends on size and character of protein

 Large proteins (>100 kD)? Choose eukaryote

Small proteins (<30 kD)? Choose prokaryote

 High yields, low cost? Choose E. coli

Post-translational modifications essential? Choose yeast, baculovirus or other eukaryote

 Glycosylation essential? Choose baculovirus or mammalian cell culture

27

TÁMOP-4.1.2-08/1/A-2009-011

Characteristics of (plasmid) vectors

1. Must be compatible with host cell system (prokaryotic vectors for prokaryotic cells, eukaryotic vectors for eukaryotic cells)

2. Features :

• strong promoter/inducible promoter

• transcription START sequences

• ribosome binding sites

• termination sequences, polyA signal sequence

• affinity tag or solubilization sequences

• multi-enzyme restriction site

• origin of replication (ORI)

• bacterial selectable marker (Amp or Tet)

• eukaryotic selectable marker

• recombination sequences 28 protein expression cloning, plasmid propagation

TÁMOP-4.1.2-08/1/A-2009-011

Promoter selection

• Constitutive

• Inducible

- everywhere, all the time

• Tissue- or developmental stage-specific - selected cell types, specific timing

- specific timing, can avoid toxicity to host

• Synthetic

29

30

TÁMOP-4.1.2-08/1/A-2009-011

TÁMOP-4.1.2-08/1/A-2009-011

Inducible promoters: Tet-off system

31

Inducible promoters: Tet-on system

TÁMOP-4.1.2-08/1/A-2009-011

(faster response)

32

TÁMOP-4.1.2-08/1/A-2009-011

Synthetic promoters, inducible systems

Steroid hormone induction: adenovirus promoter glucocorticoid response element inducer: dexamethasone

Tetracycline operon:

Ecdyson-inducible system:

CMV promoter

Tet operator sequence, Tet repressor protein inducer/repressor: tetracycline requires two vectors

SV40 promoter human RXR receptor and Drosophila ecdyson receptor (VgEcR) = transcription factor heterodimer

Activator of transcription factor: pronasteroneA

Nice dose response

33

TÁMOP-4.1.2-08/1/A-2009-011

Bacterial expression systems

Advantages

Grows quickly (8 hrs to produce protein)

High yields (50-500 mg/L)

Low cost of media (simple media constituents)

Low fermentor costs

Disadvantages

Difficulty expressing large proteins (>50 kD)

No glycosylation or signal peptide removal

Eukaryotic proteins are sometimes toxic

Can’t handle S-S rich proteins

34

TÁMOP-4.1.2-08/1/A-2009-011

Promoter selection for prokaryotes

Promoter type lac promoter trc and tac promoter

T7 RNA polymerase promoter

TetA promoter/operon

Phage promoter pL

PPBAD promoter rhaPBAD promoter

Expression level Regulator/inducer low/middle moderatly high very high

IPTG

IPTG

IPTG middle/high moderatly high low/high low/high tetracycline temperature shift

L-arabinose

L-rhamnose

Main characteristics

Low level intracellular expression

Higher expression

Basal level depends on strain

T7-lac system for tight control

High level induction

Low basal level

Tight regulation

Independent of metabolic state

Very low basal level

Temperature sensitive host needed

Very low basal level

Tight regulation

Fine-tuning, dose dependent

Very low basal level

Tight regulation

35

Cloning & transforming in yeast cells

TÁMOP-4.1.2-08/1/A-2009-011

Pichia pastoris

Saccharomyces cerevisiae

36

TÁMOP-4.1.2-08/1/A-2009-011

Pichia pastoris

Yeast are single celled eukaryotes

Behave like bacteria, but have key advantages of eukaryotes

P. pastoris is a methylotrophic yeast that can use methanol as its sole carbon source (using alcohol oxidase)

Has a very strong promoter for the alcohol oxidase (AOX) gene

(~30% of protein produced when induced)

37

TÁMOP-4.1.2-08/1/A-2009-011

Pichia expression system

Advantages

Grow quickly (8 hrs to produce protein)

Very high yields (50-5000 mg/L)

Low cost of media (simple media constituents)

Low fermentor costs

More advantages

Can express large proteins (>50 kD)

Glycosylation & signal peptide removal

Has chaperonins to help fold “tough” prtns

Can handle S-S rich proteins

38

TÁMOP-4.1.2-08/1/A-2009-011

Pichia pastoris cloning

Uses a special plasmid that works both in E. coli and yeast

Once gene of interest is inserted into this plasmid, it must be linearized

Transfect yeast cells with linear plasmid

Double cross-over recombination event occurs to cause the gene of interest to insert directly into P. pastoris chromosome where the old AOX gene used to be

Now gene of interest is under control of the powerful AOX promoter

Stable transfectant

39

Cloning a gene into Pichia vector

TÁMOP-4.1.2-08/1/A-2009-011

40

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus/insect cell expression systems

Spodoptera frugiperda

Spodoptera f. larva

Bastiaan (Bart) Drees

Sf9 cells and baculovirus

41

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus life cycle

2.

3a.

4a.

3b.

4b.

1.

42

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus life phases in culture

1. Early phase: cell entry, shutting down host gene expression viral protein synthesis

2. Late phase: viral DNS replication, virus assembly, release of viral particles from cell

(peak:18-36 hrs post-infection)

Also used to prepare viral stock

3. Very late phase: polyhedrin and p10 genes are expressed, viruses embedded in polyhedrin form occlusion bodies. Cell lysis.

(24-96 hrs post-infection)

Used for protein production

43

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus mediated protein expression in insect cells

Autographica californica multiple nuclear polyhedrosis virus (Baculovirus)

Virus commonly infects insects cells of the alfalfa looper (small beetle) or armyworms (and their larvae)

Uses super-strong promoter from the polyhedrin coat protein to enhance expression of proteins while virus resides inside the insect cell - protein is not required for infection or viral life cycle

Secreted proteins better expressed by stably transfected insect cell lines, from the ie-1 promoter

(infection interferes with secretory pathways)

44

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus expression system workflow

1. Cloning gene of interest into baculovirus genome

2. Use recombinant baculoviral DNA to transfect insect cells

3. Collect viral particles from insect cell culture supernatant

4. Test viral stock titer, freeze stocks

5. Infect new insect cell culture

6. Harvest cells (with occlusion bodies)

Note: not a stable cell line!

45

TÁMOP-4.1.2-08/1/A-2009-011

Cloning a gene into baculovirus (AcMNPV) vector

Site-specific transposition

Transfer vector

5’ x

Cloned gene

3’ x

5’

Cloned gene

3’ modified AcMNPV DNA,

“Bacmid” maintained in E. coli

Recombinant

AcMNPV bacmid

46

TÁMOP-4.1.2-08/1/A-2009-011

Tn7R polyhedrin promoter Gent+ Tn7L

Gene of Interest Transfer vector with insert

PpH

Gene of Interest

Tn7 L

Tn7 R

Bacmid with insert

47

TÁMOP-4.1.2-08/1/A-2009-011

Tn7R GOI Tn7L

Transposition into bacmid

M 13 forward

128bp

Mini att Tn7

145bp

Bacmid DNA

M 13 reverse

48

TÁMOP-4.1.2-08/1/A-2009-011

Baculovirus expression system

Disadvantages Advantages

Grow very slowly (10-12 days for setup)

Cell culture is only sustainable for 4-5 days

Can express large proteins (>50 kD)

(Mostly) Correct glycosylation & signal peptide removal

Has chaperonins to help protein folding

Set-up is time consuming, not as simple as yeast

Very high yields, cheap

49

Baculovirus successes

Alpha and beta interferon

Adenosine deaminase

Erythropoietin

Interleukin 2

Poliovirus proteins

Tissue plasminogen activator (TPA)

TÁMOP-4.1.2-08/1/A-2009-011

50

Mammalian expression systems

TÁMOP-4.1.2-08/1/A-2009-011

51

TÁMOP-4.1.2-08/1/A-2009-011

Mammalian expression systems

Disadvantages Advantages

Selection takes time (weeks for set-up)

Cell culture is only sustainable for limited period of time

Set-up is very time consuming, costly, modest yields

Can express large proteins (>50 kD)

Correct glycosylation & signal peptide removal, generates authentic proteins

Has chaperonins to help protein folding

52

TÁMOP-4.1.2-08/1/A-2009-011

Mammalian expression system

Gene initially cloned into plasmid, and propagated in bacterial cells

Cells are typically derived from the Chinese Hamster Ovary (CHO) cell line

Mammalian cells transformed by electroporation (with linear plasmid) and gene integrates (1 or more times) into random locations within different CHO chromosomes

Multiple rounds of growth and selection using methotrexate to select for those cells with highest expression & integration of DHFR and the gene of interest

Stably transfected cell lines are generated - long term culturing

53

TÁMOP-4.1.2-08/1/A-2009-011

Characteristics of mammalian expression vectors

Recombinant gene expression requires multiple elements in the vector:

• promoter (general or tissue-specific)

• enhancer

• polyA signal

• intron - may enhance expression

• selection marker (ampicylin, neomycin, DHFR etc.)

• Frequently used promoters:

(strong promoters) simian virus 40 (SV40) papovavirus

Rous sarcoma virus human cytomegalovirus (CMV)

54

TÁMOP-4.1.2-08/1/A-2009-011

Methotrexate (MTX) selection

Gene of interest DHFR

Transfect

DHFR minus cells

Grow in nucleoside free medium

Culture a colony of cells

Grow in

0.05 uM Mtx

Culture a colony of cells

55

TÁMOP-4.1.2-08/1/A-2009-011

Methotrexate (MTX) selection

Multiple rounds of selection, increasing MTX concentration

Grow in

0.25 uM Mtx

Culture a

Colony of cells

Grow in

0.5 uM Mtx

Culture a

Colony of cells

Foreign gene expressed at high level in

CHO cells

56

Mammalian cell successes

Factor IX

Factor VIII

Gamma interferon

Interleukin 2

Human growth hormone

Tissue plasminogen activator (TPA)

TÁMOP-4.1.2-08/1/A-2009-011

57

TÁMOP-4.1.2-08/1/A-2009-011

Purification of recombinant proteins

Application

Therapeutic use, in vivo studies

Biochemical assays, X-ray crystallography

N-terminal sequencing, antigen for antibody production, NMR

Required Purity

Extremely high > 99%

High 95-99%

Moderately high < 95%

58

TÁMOP-4.1.2-08/1/A-2009-011

Purification of recombinant proteins

Size

Hydrophobicity

BEHAVIOUR

Charge

Activity

All proteins are different 59

Conventional purification strategy

TÁMOP-4.1.2-08/1/A-2009-011

• Use different properties of protein in purification scheme

• Multiple intermediate steps may be required

• Need to detect low amounts

60

TÁMOP-4.1.2-08/1/A-2009-011

Affinity-tag based purification strategy

• Fusion proteins with affinity tag

• Tag: peptide or protein

• Tag binds something very selectively and w. high affinity

• Very effective purification in initial step

• Tag can be used for detection

• Tag can be cleaved off

61

Affinity-tagging of recombinant proteins

TÁMOP-4.1.2-08/1/A-2009-011 gene for protein of interest insert affinity tag sequence introduce into cells

Purification of tagged protein

Immunolocalization of protein

Other interacting proteins

Tagged protein

62

TÁMOP-4.1.2-08/1/A-2009-011

Histidine tag

His-Ni2+ stable complex at near-neutral aqueous conditions

Solid matrix

Poly-histidine on protein

Nickel ion

(Ni2+)

63

Making proteins bind nickel

TÁMOP-4.1.2-08/1/A-2009-011

64

TÁMOP-4.1.2-08/1/A-2009-011

His-tag based purification strategy

65

His-tag:

T7-tag:

S-tag:

Strep-tag:

Epitope-tags:

TAP-tag:

TÁMOP-4.1.2-08/1/A-2009-011

Examples for affinity and epitope tags

N-or C-terminal 6xHistidin, binds to Ni-resin

• purification starting sequence for T7 gene (11 amino acids)

• enhancer for translation ribonuclease A S-peptid (15 amino acids)

• detection, isolation: biotinylated S-protein, S-protein affinity

C-terminal AWRHPQFGG sequence (affinity to streptavidin) purification recognised by good antibodies (usually monoclonal)

• FLAG-tag ( NYKNNNNK )

• c-myc-tag (

QGKLISGGNL )

„tandem-affinity purification”, calmodulin-binding protein and protein A both fused to protein of interest

• very good system to study protein-protein interactions

66

TÁMOP-4.1.2-08/1/A-2009-011

Fusion proteins in prokaryotic expression systems

Proteins expressed in E. coli are often produced as fusion proteins:

• function of the protein in bacteria is not of interest

• mammalian protein is not expressed effectively by itself

• bacterial fusion partner, (e.g. GST) on the other hand, is expressed effectively – fusion protein is likely to be expressed well, too

• one-step purification from bacterial lysate

67

Bacterial fusion protein systems

TÁMOP-4.1.2-08/1/A-2009-011

Glutathion-S-transferase:

Maltose-binding protein:

Thioredoxin

26 kDa protein

Schistosoma japonica gene product pGEX vector-series fast isolation on glutathion-resin

E. coli malE gene product pMEL vector-series solation on maltose affinity column

17 kDa protein, heat-stable, very good solubility

Ribonucleotide-reductase reducing enzyme

E. coli trxA gene product pTrxFus vector

68

IPTG

TÁMOP-4.1.2-08/1/A-2009-011

Glutathione-S-transferase fusion protein expression system

Polylinker or

Multicloning site

Lac promoter

GST

Repressor protein pGEX

Lac inhibitor gene

Ampicyllin resistance gene

Ori

69

TÁMOP-4.1.2-08/1/A-2009-011

Which tag to use?

Specificity of binding interaction

Cost of resin

Native vs . denaturing elution

Presence of metals

Expression level, solubility & toxicity of target protein

Tag removal

70

Tag removal

NH

2

– tag linker protein

DDDDK protease

TÁMOP-4.1.2-08/1/A-2009-011

Linker/cleavage strategy selection:

• effect on structure

• effect on function

• flexibility

• protein 1 ° sequence

• removal of protease

71

TÁMOP-4.1.2-08/1/A-2009-011

Tag removal

Excision Site Cleavage Enzyme

D-D-D-D-K

X enterokinase

I-D/E-G-R

X factor Xa protease

L-V-P-R

G-S thrombin

E-N-L-Y-F-Q

G TEV protease

L-E-V-L-F-Q

G-P PreScission TM protease

Comment active: pH 4.5-9.5, 4-45 ° C

X cannot be P secondary cleavage sites

X cannot be P/R secondary cleavage sites biotynilated form available secondary cleavage sites active: wide range of T

His-tagged form available engineered with GST tag

72

TÁMOP-4.1.2-08/1/A-2009-011

Purification protocol : as few steps as possible

crude

His-resin I tag cleavage

His-resin II gel filtration

• His-resin I usually provides a major step of the purification

• His-resin II removes cleaved-off

His-tag and persistent contaminant proteins in E.coli

host

• Gel-filtration – “polishing”

Pure protein

73

Download