fbpx
Wikipedia

Green fluorescent protein

The green fluorescent protein (GFP) is a protein that exhibits green fluorescence when exposed to light in the blue to ultraviolet range.[2][3] The label GFP traditionally refers to the protein first isolated from the jellyfish Aequorea victoria and is sometimes called avGFP. However, GFPs have been found in other organisms including corals, sea anemones, zoanithids, copepods and lancelets.[4]

Green fluorescent protein
Structure of the Aequorea victoria green fluorescent protein.[1]
Identifiers
SymbolGFP
PfamPF01353
Pfam clanCL0069
InterProIPR011584
CATH1ema
SCOP21ema / SCOPe / SUPFAM
Available protein structures:
Pfam  structures / ECOD  
PDBRCSB PDB; PDBe; PDBj
PDBsumstructure summary
Green fluorescent protein
Identifiers
OrganismAequorea victoria
SymbolGFP
UniProtP42212
Search for
StructuresSwiss-model
DomainsInterPro

The GFP from A. victoria has a major excitation peak at a wavelength of 395 nm and a minor one at 475 nm. Its emission peak is at 509 nm, which is in the lower green portion of the visible spectrum. The fluorescence quantum yield (QY) of GFP is 0.79. The GFP from the sea pansy (Renilla reniformis) has a single major excitation peak at 498 nm. GFP makes for an excellent tool in many forms of biology due to its ability to form an internal chromophore without requiring any accessory cofactors, gene products, or enzymes / substrates other than molecular oxygen.[5]

In cell and molecular biology, the GFP gene is frequently used as a reporter of expression.[6] It has been used in modified forms to make biosensors, and many animals have been created that express GFP, which demonstrates a proof of concept that a gene can be expressed throughout a given organism, in selected organs, or in cells of interest. GFP can be introduced into animals or other species through transgenic techniques, and maintained in their genome and that of their offspring. To date, GFP has been expressed in many species, including bacteria, yeasts, fungi, fish and mammals, including in human cells. Scientists Roger Y. Tsien, Osamu Shimomura, and Martin Chalfie were awarded the 2008 Nobel Prize in Chemistry on 10 October 2008 for their discovery and development of the green fluorescent protein.

Most commercially available genes for GFP and similar fluorescent proteins are around 730 base-pairs long. The natural protein has 238 amino acids. Its molecular mass is 27 kD.[7] Therefore, fusing the GFP gene to the gene of a protein of interest can significantly increase the protein's size and molecular mass, and can impair the protein's natural function or change its location or trajectory of transport within the cell.[8]

Background edit

 
Aequorea victoria
 
3D reconstruction of confocal image of VEGF-overexpressing neural progenitors (red) and GFP-positive control neural progenitor cells (green) in the rat olfactory bulb. RECA-1-positive blood vessels - blue color.

Wild-type GFP (wtGFP) edit

In the 1960s and 1970s, GFP, along with the separate luminescent protein aequorin (an enzyme that catalyzes the breakdown of luciferin, releasing light), was first purified from the jellyfish Aequorea victoria and its properties studied by Osamu Shimomura.[9] In A. victoria, GFP fluorescence occurs when aequorin interacts with Ca2+ ions, inducing a blue glow. Some of this luminescent energy is transferred to the GFP, shifting the overall color towards green.[10] However, its utility as a tool for molecular biologists did not begin to be realized until 1992 when Douglas Prasher reported the cloning and nucleotide sequence of wtGFP in Gene.[11] The funding for this project had run out, so Prasher sent cDNA samples to several labs. The lab of Martin Chalfie expressed the coding sequence of wtGFP, with the first few amino acids deleted, in heterologous cells of E. coli and C. elegans, publishing the results in Science in 1994.[12] Frederick Tsuji's lab independently reported the expression of the recombinant protein one month later.[13] Remarkably, the GFP molecule folded and was fluorescent at room temperature, without the need for exogenous cofactors specific to the jellyfish. Although this near-wtGFP was fluorescent, it had several drawbacks, including dual peaked excitation spectra, pH sensitivity, chloride sensitivity, poor fluorescence quantum yield, poor photostability and poor folding at 37 °C (99 °F).

The first reported crystal structure of a GFP was that of the S65T mutant by the Remington group in Science in 1996.[14] One month later, the Phillips group independently reported the wild-type GFP structure in Nature Biotechnology.[15] These crystal structures provided vital background on chromophore formation and neighboring residue interactions. Researchers have modified these residues by directed and random mutagenesis to produce the wide variety of GFP derivatives in use today. Further research into GFP has shown that it is resistant to detergents, proteases, guanidinium chloride (GdmCl) treatments, and drastic temperature changes.[16]

GFP derivatives edit

 
The diversity of genetic mutations is illustrated by this San Diego beach scene drawn with living bacteria expressing 8 different colors of fluorescent proteins (derived from GFP and dsRed).

Due to the potential for widespread usage and the evolving needs of researchers, many different mutants of GFP have been engineered.[17][18] The first major improvement was a single point mutation (S65T) reported in 1995 in Nature by Roger Tsien.[19] This mutation dramatically improved the spectral characteristics of GFP, resulting in increased fluorescence, photostability, and a shift of the major excitation peak to 488 nm, with the peak emission kept at 509 nm. This matched the spectral characteristics of commonly available FITC filter sets, increasing the practicality of use by the general researcher. A 37 °C folding efficiency (F64L) point mutant to this scaffold, yielding enhanced GFP (EGFP), was discovered in 1995 by the laboratories of Thastrup[20] and Falkow.[21] EGFP allowed the practical use of GFPs in mammalian cells. EGFP has an extinction coefficient (denoted ε) of 55,000 M−1cm−1.[22] The fluorescence quantum yield (QY) of EGFP is 0.60. The relative brightness, expressed as ε•QY, is 33,000 M−1cm−1.

Superfolder GFP (sfGFP), a series of mutations that allow GFP to rapidly fold and mature even when fused to poorly folding peptides, was reported in 2006.[23]

Many other mutations have been made, including color mutants; in particular, blue fluorescent protein (EBFP, EBFP2, Azurite, mKalama1), cyan fluorescent protein (ECFP, Cerulean, CyPet, mTurquoise2), and yellow fluorescent protein derivatives (YFP, Citrine, Venus, YPet). BFP derivatives (except mKalama1) contain the Y66H substitution.They exhibit a broad absorption band in the ultraviolet centered close to 380 nanometers and an emission maximum at 448 nanometers. A green fluorescent protein mutant (BFPms1) that preferentially binds Zn(II) and Cu(II) has been developed. BFPms1 have several important mutations including and the BFP chromophore (Y66H),Y145F for higher quantum yield, H148G for creating a hole into the beta-barrel and several other mutations that increase solubility. Zn(II) binding increases fluorescence intensity, while Cu(II) binding quenches fluorescence and shifts the absorbance maximum from 379 to 444 nm. Therefore, they can be used as Zn biosensor.[24]

 
A palette of variants of GFP and DsRed.

Chromophore binding. The critical mutation in cyan derivatives is the Y66W substitution, which causes the chromophore to form with an indole rather than phenol component. Several additional compensatory mutations in the surrounding barrel are required to restore brightness to this modified chromophore due to the increased bulk of the indole group. In ECFP and Cerulean, the N-terminal half of the seventh strand exhibits two conformations. These conformations both have a complex set of van der Waals interactions with the chromophore. The Y145A and H148D mutations in Cerulean stabilize these interactions and allow the chromophore to be more planar, better packed, and less prone to collisional quenching.[25]

Additional site-directed random mutagenesis in combination with fluorescence lifetime based screening has further stabilized the seventh β-strand resulting in a bright variant, mTurquoise2, with a quantum yield (QY) of 0.93.[26] The red-shifted wavelength of the YFP derivatives is accomplished by the T203Y mutation and is due to π-electron stacking interactions between the substituted tyrosine residue and the chromophore.[3] These two classes of spectral variants are often employed for Förster resonance energy transfer (FRET) experiments. Genetically encoded FRET reporters sensitive to cell signaling molecules, such as calcium or glutamate, protein phosphorylation state, protein complementation, receptor dimerization, and other processes provide highly specific optical readouts of cell activity in real time.

Semirational mutagenesis of a number of residues led to pH-sensitive mutants known as pHluorins, and later super-ecliptic pHluorins. By exploiting the rapid change in pH upon synaptic vesicle fusion, pHluorins tagged to synaptobrevin have been used to visualize synaptic activity in neurons.[27]

Redox sensitive GFP (roGFP) was engineered by introduction of cysteines into the beta barrel structure. The redox state of the cysteines determines the fluorescent properties of roGFP.[28]

Nomenclature edit

The nomenclature of modified GFPs is often confusing due to overlapping mapping of several GFP versions onto a single name. For example, mGFP often refers to a GFP with an N-terminal palmitoylation that causes the GFP to bind to cell membranes. However, the same term is also used to refer to monomeric GFP, which is often achieved by the dimer interface breaking A206K mutation.[29] Wild-type GFP has a weak dimerization tendency at concentrations above 5 mg/mL. mGFP also stands for "modified GFP," which has been optimized through amino acid exchange for stable expression in plant cells.

In nature edit

 
Live lancelet (B. floridae) under a fluorescent microscope.
 
In the marine copepod Pontella mimocerami

The purpose of both the (primary) bioluminescence (from aequorin's action on luciferin) and the (secondary) fluorescence of GFP in jellyfish is unknown. GFP is co-expressed with aequorin in small granules around the rim of the jellyfish bell. The secondary excitation peak (480 nm) of GFP does absorb some of the blue emission of aequorin, giving the bioluminescence a more green hue. The serine 65 residue of the GFP chromophore is responsible for the dual-peaked excitation spectra of wild-type GFP. It is conserved in all three GFP isoforms originally cloned by Prasher. Nearly all mutations of this residue consolidate the excitation spectra to a single peak at either 395 nm or 480 nm. The precise mechanism of this sensitivity is complex, but, it seems, involves donation of a hydrogen from serine 65 to glutamate 222, which influences chromophore ionization.[3] Since a single mutation can dramatically enhance the 480 nm excitation peak, making GFP a much more efficient partner of aequorin, A. victoria appears to evolutionarily prefer the less-efficient, dual-peaked excitation spectrum. Roger Tsien has speculated that varying hydrostatic pressure with depth may affect serine 65's ability to donate a hydrogen to the chromophore and shift the ratio of the two excitation peaks. Thus, the jellyfish may change the color of its bioluminescence with depth. However, a collapse in the population of jellyfish in Friday Harbor, where GFP was originally discovered, has hampered further study of the role of GFP in the jellyfish's natural environment.

Most species of lancelet are known to produce GFP in various regions of their body.[30] Unlike A. victoria, lancelets do not produce their own blue light, and the origin of their endogenous GFP is still unknown. Some speculate that it attracts plankton towards the mouth of the lancelet, serving as a passive hunting mechanism. It may also serve as a photoprotective agent in the larvae, preventing damage caused by high-intensity blue light by converting it into lower-intensity green light. However, these theories have not been tested.

GFP-like proteins have been found in multiple species of marine copepods, particularly from the Pontellidae and Aetideidae families.[31] GFP isolated from Pontella mimocerami has shown high levels of brightness with a quantum yield of 0.92, making them nearly two-fold brighter than the commonly used EGFP isolated from A. victoria.[32]

Other fluorescent proteins edit

 
Different proteins produce different fluorescent colors when exposed to ultraviolet light.

There are many GFP-like proteins that, despite being in the same protein family as GFP, are not directly derived from Aequorea victoria. These include dsRed, eqFP611, Dronpa, TagRFPs, KFP, EosFP/IrisFP, Dendra, and so on. Having been developed from proteins in different organisms, these proteins can sometimes display unanticipated approaches to chromophore formation. Some of these, such as KFP, are developed from naturally non- or weakly-fluorescent proteins to be greatly improved upon by mutagenesis.[33] When GFP-like barrels of different spectra characteristics are used, the excitation spectra of one chromophore can be used to power another chromophore (FRET), allowing for conversion between wavelengths of light.[34]

FMN-binding fluorescent proteins (FbFPs) were developed in 2007 and are a class of small (11–16 kDa), oxygen-independent fluorescent proteins that are derived from blue-light receptors. They are intended especially for the use under anaerobic or hypoxic conditions, since the formation and binding of the Flavin chromophore does not require molecular oxygen, as it is the case with the synthesis of the GFP chromophore.[35]

 
White light image, or image seen by the eye, of fluorescent proteins in image above.

Fluorescent proteins with other chromophores, such as UnaG with bilirubin, can display unique properties like red-shifted emission above 600 nm or photoconversion from a green-emitting state to a red-emitting state. They can have excitation and emission wavelengths far enough apart to achieve conversion between red and green light.

A new class of fluorescent protein was evolved from a cyanobacterial (Trichodesmium erythraeum) phycobiliprotein, α-allophycocyanin, and named small ultra red fluorescent protein (smURFP) in 2016. smURFP autocatalytically self-incorporates the chromophore biliverdin without the need of an external protein, known as a lyase.[36][37] Jellyfish- and coral-derived GFP-like proteins require oxygen and produce a stoichiometric amount of hydrogen peroxide upon chromophore formation.[38] smURFP does not require oxygen or produce hydrogen peroxide and uses the chromophore, biliverdin. smURFP has a large extinction coefficient (180,000 M−1 cm−1) and has a modest quantum yield (0.20), which makes it comparable biophysical brightness to eGFP and ~2-fold brighter than most red or far-red fluorescent proteins derived from coral. smURFP spectral properties are similar to the organic dye Cy5.[36][39]

 
E. coli colonies expressing fluorescent proteins.

Reviews on new classes of fluorescent proteins and applications can be found in the cited reviews.[40][41]

Structure edit

GFP has a beta barrel structure consisting of eleven β-strands with a pleated sheet arrangement, with an alpha helix containing the covalently bonded chromophore 4-(p-hydroxybenzylidene)imidazolidin-5-one (HBI) running through the center.[3][14][15] Five shorter alpha helices form caps on the ends of the structure. The beta barrel structure is a nearly perfect cylinder, 42Å long and 24Å in diameter (some studies have reported a diameter of 30Å[16]),[14] creating what is referred to as a "β-can" formation, which is unique to the GFP-like family.[15] HBI, the spontaneously modified form of the tripeptide Ser65–Tyr66–Gly67, is nonfluorescent in the absence of the properly folded GFP scaffold and exists mainly in the un-ionized phenol form in wtGFP.[42] Inward-facing sidechains of the barrel induce specific cyclization reactions in Ser65–Tyr66–Gly67 that induce ionization of HBI to the phenolate form and chromophore formation. This process of post-translational modification is referred to as maturation.[43] The hydrogen-bonding network and electron-stacking interactions with these sidechains influence the color, intensity and photostability of GFP and its numerous derivatives.[44] The tightly packed nature of the barrel excludes solvent molecules, protecting the chromophore fluorescence from quenching by water. In addition to the auto-cyclization of the Ser65-Tyr66-Gly67, a 1,2-dehydrogenation reaction occurs at the Tyr66 residue.[16] Besides the three residues that form the chromophore, residues such as Gln94, Arg96, His148, Thr203, and Glu222 all act as stabilizers. The residues of Gln94, Arg96, and His148 are able to stabilize by delocalizing the chromophore charge. Arg96 is the most important stabilizing residue due to the fact that it prompts the necessary structural realignments that are necessary from the HBI ring to occur. Any mutation to the Arg96 residue would result in a decrease in the development rate of the chromophore because proper electrostatic and steric interactions would be lost. Tyr66 is the recipient of hydrogen bonds and does not ionize in order to produce favorable electrostatics.[45]

 
GFP Movie showing entire structure and zoom in to fluorescent chromophore.
 
GFP molecules drawn in cartoon style, one fully and one with the side of the beta barrel cut away to reveal the chromophore (highlighted as ball-and-stick). From PDB: 1GFL​.
 
GFP ribbon diagram. From PDB: 1EMA​.

Autocatalytic formation of the chromophore in wtGFP edit

For a synthetic analogue see also 3,5-Difluoro-4-hydroxybenzylidene imidazolinone.

 

Mechanistically, the process involves base-mediated cyclization followed by dehydration and oxidation. In the reaction of 7a to 8 involves the formation of an enamine from the imine, while in the reaction of 7b to 9 a proton is abstracted.[46] The formed HBI fluorophore is highlighted in green.

The reactions are catalyzed by residues Glu222 and Arg96.[46][47] An analogous mechanism is also possible with threonine in place of Ser65.

Applications edit

Reporter assays edit

Green fluorescent protein may be used as a reporter gene.[48][49]

For example, GFP can be used as a reporter for environmental toxicity levels. This protein has been shown to be an effective way to measure the toxicity levels of various chemicals including ethanol, p-formaldehyde, phenol, triclosan, and paraben. GFP is great as a reporter protein because it has no effect on the host when introduced to the host's cellular environment. Due to this ability, no external visualization stain, ATP, or cofactors are needed. With regards to pollutant levels, the fluorescence was measured in order to gauge the effect that the pollutants have on the host cell. The cellular density of the host cell was also measured. Results from the study conducted by Song, Kim, & Seo (2016) showed that there was a decrease in both fluorescence and cellular density as pollutant levels increased. This was indicative of the fact that cellular activity had decreased. More research into this specific application in order to determine the mechanism by which GFP acts as a pollutant marker.[50] Similar results have been observed in zebrafish because zebrafish that were injected with GFP were approximately twenty times more susceptible to recognize cellular stresses than zebrafish that were not injected with GFP.[51]

Advantages edit

The biggest advantage of GFP is that it can be heritable, depending on how it was introduced, allowing for continued study of cells and tissues it is expressed in. Visualizing GFP is noninvasive, requiring only illumination with blue light. GFP alone does not interfere with biological processes, but when fused to proteins of interest, careful design of linkers is required to maintain the function of the protein of interest. Moreover, if used with a monomer it is able to diffuse readily throughout cells.[52]

Fluorescence microscopy edit

 
Superresolution with two fusion proteins (GFP-Snf2H and RFP-H2A), Co-localisation studies (2CLM) in the nucleus of a bone cancer cell. 120.000 localized molecules in a widefield area (470 µm2).

The availability of GFP and its derivatives has thoroughly redefined fluorescence microscopy and the way it is used in cell biology and other biological disciplines.[53] While most small fluorescent molecules such as FITC (fluorescein isothiocyanate) are strongly phototoxic when used in live cells, fluorescent proteins such as GFP are usually much less harmful when illuminated in living cells. This has triggered the development of highly automated live-cell fluorescence microscopy systems, which can be used to observe cells over time expressing one or more proteins tagged with fluorescent proteins.

There are many techniques to utilize GFP in a live cell imaging experiment. The most direct way of utilizing GFP is to directly attach it to a protein of interest. For example, GFP can be included in a plasmid expressing other genes to indicate a successful transfection of a gene of interest. Another method is to use a GFP that contains a mutation where the fluorescence will change from green to yellow over time, which is referred to as a fluorescent timer. With the fluorescent timer, researchers can study the state of protein production such as recently activated, continuously activated, or recently deactivated based on the color reported by the fluorescent protein.[54] In yet another example, scientists have modified GFP to become active only after exposure to irradiation giving researchers a tool to selectively activate certain portions of a cell and observe where proteins tagged with the GFP move from the starting location.[55] These are only two examples in a burgeoning field of fluorescent microcopy and a more complete review of biosensors utilizing GFP and other fluorescent proteins can be found here [56]

For example, GFP had been widely used in labelling the spermatozoa of various organisms for identification purposes as in Drosophila melanogaster, where expression of GFP can be used as a marker for a particular characteristic. GFP can also be expressed in different structures enabling morphological distinction. In such cases, the gene for the production of GFP is incorporated into the genome of the organism in the region of the DNA that codes for the target proteins and that is controlled by the same regulatory sequence; that is, the gene's regulatory sequence now controls the production of GFP, in addition to the tagged protein(s). In cells where the gene is expressed, and the tagged proteins are produced, GFP is produced at the same time. Thus, only those cells in which the tagged gene is expressed, or the target proteins are produced, will fluoresce when observed under fluorescence microscopy. Analysis of such time lapse movies has redefined the understanding of many biological processes including protein folding, protein transport, and RNA dynamics, which in the past had been studied using fixed (i.e., dead) material. Obtained data are also used to calibrate mathematical models of intracellular systems and to estimate rates of gene expression.[57] Similarly, GFP can be used as an indicator of protein expression in heterologous systems. In this scenario, fusion proteins containing GFP are introduced indirectly, using RNA of the construct, or directly, with the tagged protein itself. This method is useful for studying structural and functional characteristics of the tagged protein on a macromolecular or single-molecule scale with fluorescence microscopy.

The Vertico SMI microscope using the SPDM Phymod technology uses the so-called "reversible photobleaching" effect of fluorescent dyes like GFP and its derivatives to localize them as single molecules in an optical resolution of 10 nm. This can also be performed as a co-localization of two GFP derivatives (2CLM).[58]

Another powerful use of GFP is to express the protein in small sets of specific cells. This allows researchers to optically detect specific types of cells in vitro (in a dish), or even in vivo (in the living organism).[59] Genetically combining several spectral variants of GFP is a useful trick for the analysis of brain circuitry (Brainbow).[60] Other interesting uses of fluorescent proteins in the literature include using FPs as sensors of neuron membrane potential,[61] tracking of AMPA receptors on cell membranes,[62] viral entry and the infection of individual influenza viruses and lentiviral viruses,[63][64] etc.

It has also been found that new lines of transgenic GFP rats can be relevant for gene therapy as well as regenerative medicine.[65] By using "high-expresser" GFP, transgenic rats display high expression in most tissues, and many cells that have not been characterized or have been only poorly characterized in previous GFP-transgenic rats.

GFP has been shown to be useful in cryobiology as a viability assay. Correlation of viability as measured by trypan blue assays were 0.97.[66] Another application is the use of GFP co-transfection as internal control for transfection efficiency in mammalian cells.[67]

A novel possible use of GFP includes using it as a sensitive monitor of intracellular processes via an eGFP laser system made out of a human embryonic kidney cell line. The first engineered living laser is made by an eGFP expressing cell inside a reflective optical cavity and hitting it with pulses of blue light. At a certain pulse threshold, the eGFP's optical output becomes brighter and completely uniform in color of pure green with a wavelength of 516 nm. Before being emitted as laser light, the light bounces back and forth within the resonator cavity and passes the cell numerous times. By studying the changes in optical activity, researchers may better understand cellular processes.[68][69]

GFP is used widely in cancer research to label and track cancer cells. GFP-labelled cancer cells have been used to model metastasis, the process by which cancer cells spread to distant organs.[70]

Split GFP edit

GFP can be used to analyse the colocalization of proteins. This is achieved by "splitting" the protein into two fragments which are able to self-assemble, and then fusing each of these to the two proteins of interest. Alone, these incomplete GFP fragments are unable to fluoresce. However, if the two proteins of interest colocalize, then the two GFP fragments assemble together to form a GFP-like structure which is able to fluoresce. Therefore, by measuring the level of fluorescence it is possible to determine whether the two proteins of interest colocalize.[71]

Macro-photography edit

Macro-scale biological processes, such as the spread of virus infections, can be followed using GFP labeling.[72] In the past, mutagenic ultra violet light (UV) has been used to illuminate living organisms (e.g., see[73]) to detect and photograph the GFP expression. Recently, a technique using non-mutagenic LED lights[74] have been developed for macro-photography.[75] The technique uses an epifluorescence camera attachment[76] based on the same principle used in the construction of epifluorescence microscopes.

Transgenic pets edit

 
Mice expressing GFP under UV light (left & right), compared to normal mouse (center)

Alba, a green-fluorescent rabbit, was created by a French laboratory commissioned by Eduardo Kac using GFP for purposes of art and social commentary.[77] The US company Yorktown Technologies markets to aquarium shops green fluorescent zebrafish (GloFish) that were initially developed to detect pollution in waterways. NeonPets, a US-based company has marketed green fluorescent mice to the pet industry as NeonMice.[78] Green fluorescent pigs, known as Noels, were bred by a group of researchers led by Wu Shinn-Chih at the Department of Animal Science and Technology at National Taiwan University.[79] A Japanese-American Team created green-fluorescent cats as proof of concept to use them potentially as model organisms for diseases, particularly HIV.[80] In 2009 a South Korean team from Seoul National University bred the first transgenic beagles with fibroblast cells from sea anemones. The dogs give off a red fluorescent light, and they are meant to allow scientists to study the genes that cause human diseases like narcolepsy and blindness.[81]

Art edit

Julian Voss-Andreae, a German-born artist specializing in "protein sculptures,"[82] created sculptures based on the structure of GFP, including the 1.70 m (5'6") tall "Green Fluorescent Protein" (2004)[83] and the 1.40 m (4'7") tall "Steel Jellyfish" (2006). The latter sculpture is located at the place of GFP's discovery by Shimomura in 1962, the University of Washington's Friday Harbor Laboratories.[84]

 
Julian Voss-Andreae's GFP-based sculpture Steel Jellyfish (2006). The image shows the stainless-steel sculpture at Friday Harbor Laboratories on San Juan Island (Wash., USA), the place of GFP's discovery.

See also edit

References edit

  1. ^ Ormö M, Cubitt AB, Kallio K, Gross LA, Tsien RY, Remington SJ (September 1996). "Crystal structure of the Aequorea victoria green fluorescent protein". Science. 273 (5280): 1392–5. Bibcode:1996Sci...273.1392O. doi:10.1126/science.273.5280.1392. PMID 8703075. S2CID 43030290.
  2. ^ Prendergast FG, Mann KG (Aug 1978). "Chemical and physical properties of aequorin and the green fluorescent protein isolated from Aequorea forskålea". Biochemistry. 17 (17): 3448–53. doi:10.1021/bi00610a004. PMID 28749.
  3. ^ a b c d Tsien RY (1998). "The green fluorescent protein" (PDF). Annual Review of Biochemistry. 67: 509–44. doi:10.1146/annurev.biochem.67.1.509. PMID 9759496.
  4. ^ Salih A (2019). "Fluorescent Proteins". In Cox G (ed.). Fundamentals of Fluorescence Imaging. Boca Raton: Jenny Stanford Publishing. p. 122. doi:10.1201/9781351129404. ISBN 9781351129404. S2CID 213688192.
  5. ^ Stepanenko OV, Verkhusha VV, Kuznetsova IM, Uversky VN, Turoverov KK (Aug 2008). "Fluorescent proteins as biomarkers and biosensors: throwing color lights on molecular and cellular processes". Current Protein & Peptide Science. 9 (4): 338–69. doi:10.2174/138920308785132668. PMC 2904242. PMID 18691124.
  6. ^ Phillips GJ (Oct 2001). "Green fluorescent protein--a bright idea for the study of bacterial protein localization". FEMS Microbiology Letters. 204 (1): 9–18. doi:10.1016/S0378-1097(01)00358-5. PMID 11682170.
  7. ^ Uckert W, Pedersen L, Günzburg W (2000). "Green fluorescent protein retroviral vector: generation of high-titer producer cells and virus supernatant". Gene Therapy of Cancer. Methods in Molecular Medicine. Vol. 35. pp. 275–85. doi:10.1385/1-59259-086-1:275. ISBN 1-59259-086-1. PMID 21390811.
  8. ^ Goodman SR, ed. (2008). "Chapter 1 - Tools of the Cell Biologist: Green Fluorescent Protein". Medical Cell Biology (3rd ed.). Amsterdam: Elsevier/Academic Press. pp. 14–25. doi:10.1016/B978-0-12-370458-0.50006-2. ISBN 978-0-12-370458-0. S2CID 90224559.
  9. ^ Shimomura O, Johnson FH, Saiga Y (Jun 1962). "Extraction, purification and properties of aequorin, a bioluminescent protein from the luminous hydromedusan, Aequorea". Journal of Cellular and Comparative Physiology. 59 (3): 223–39. doi:10.1002/jcp.1030590302. PMID 13911999.
  10. ^ Morise H, Shimomura O, Johnson FH, Winant J (Jun 1974). "Intermolecular energy transfer in the bioluminescent system of Aequorea". Biochemistry. 13 (12): 2656–62. doi:10.1021/bi00709a028. PMID 4151620.
  11. ^ Prasher DC, Eckenrode VK, Ward WW, Prendergast FG, Cormier MJ (Feb 1992). "Primary structure of the Aequorea victoria green-fluorescent protein". Gene. 111 (2): 229–33. doi:10.1016/0378-1119(92)90691-H. PMID 1347277.
  12. ^ Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC (Feb 1994). "Green fluorescent protein as a marker for gene expression". Science. 263 (5148): 802–5. Bibcode:1994Sci...263..802C. doi:10.1126/science.8303295. PMID 8303295. S2CID 9043327.
  13. ^ Inouye S, Tsuji FI (Mar 1994). "Aequorea green fluorescent protein. Expression of the gene and fluorescence characteristics of the recombinant protein". FEBS Letters. 341 (2–3): 277–80. doi:10.1016/0014-5793(94)80472-9. PMID 8137953.
  14. ^ a b c Ormö M, Cubitt AB, Kallio K, Gross LA, Tsien RY, Remington SJ (Sep 1996). "Crystal structure of the Aequorea victoria green fluorescent protein". Science. 273 (5280): 1392–5. Bibcode:1996Sci...273.1392O. doi:10.1126/science.273.5280.1392. PMID 8703075. S2CID 43030290.
  15. ^ a b c Yang F, Moss LG, Phillips GN (Oct 1996). "The molecular structure of green fluorescent protein" (PDF). Nature Biotechnology. 14 (10): 1246–51. doi:10.1038/nbt1096-1246. hdl:1911/19233. PMID 9631087. S2CID 34713931.
  16. ^ a b c Brejc K, Sixma TK, Kitts PA, Kain SR, Tsien RY, Ormö M, Remington SJ (March 1997). "Structural basis for dual excitation and photoisomerization of the Aequorea victoria green fluorescent protein". Proceedings of the National Academy of Sciences of the United States of America. 94 (6): 2306–2311. Bibcode:1997PNAS...94.2306B. doi:10.1073/pnas.94.6.2306. PMC 20083. PMID 9122190.
  17. ^ Shaner NC, Steinbach PA, Tsien RY (Dec 2005). "A guide to choosing fluorescent proteins" (PDF). Nature Methods. 2 (12): 905–9. doi:10.1038/nmeth819. PMID 16299475. S2CID 10024284.
  18. ^ Wilhelmsson M, Tor Y (2016). Fluorescent Analogs of Biomolecular Building Blocks: Design and Applications. New Jersey: Wiley. ISBN 978-1-118-17586-6.
  19. ^ Heim R, Cubitt AB, Tsien RY (Feb 1995). "Improved green fluorescence" (PDF). Nature. 373 (6516): 663–4. Bibcode:1995Natur.373..663H. doi:10.1038/373663b0. PMID 7854443. S2CID 40179694.
  20. ^ US patent 6172188, Thastrup O, Tullin S, Kongsbak Poulsen L, Bjørn S, "Fluorescent Proteins", published 2001-01-09 
  21. ^ Cormack BP, Valdivia RH, Falkow S (1996). "FACS-optimized mutants of the green fluorescent protein (GFP)". Gene. 173 (1 Spec No): 33–38. doi:10.1016/0378-1119(95)00685-0. PMID 8707053.
  22. ^ McRae SR, Brown CL, Bushell GR (May 2005). "Rapid purification of EGFP, EYFP, and ECFP with high yield and purity". Protein Expression and Purification. 41 (1): 121–127. doi:10.1016/j.pep.2004.12.030. PMID 15802229.
  23. ^ Pédelacq JD, Cabantous S, Tran T, Terwilliger TC, Waldo GS (Jan 2006). "Engineering and characterization of a superfolder green fluorescent protein". Nature Biotechnology. 24 (1): 79–88. doi:10.1038/nbt1172. PMID 16369541. S2CID 2966399.
  24. ^ Barondeau DP, Kassmann CJ, Tainer JA, Getzoff ED (Apr 2002). "Structural chemistry of a green fluorescent protein Zn biosensor". Journal of the American Chemical Society. 124 (14): 3522–3524. doi:10.1021/ja0176954. PMID 11929238.
  25. ^ Lelimousin M, Noirclerc-Savoye M, Lazareno-Saez C, Paetzold B, Le Vot S, Chazal R, Macheboeuf P, Field MJ, Bourgeois D, Royant A (Oct 2009). "Intrinsic dynamics in ECFP and Cerulean control fluorescence quantum yield". Biochemistry. 48 (42): 10038–10046. doi:10.1021/bi901093w. PMID 19754158.
  26. ^ Goedhart J, von Stetten D, Noirclerc-Savoye M, Lelimousin M, Joosen L, Hink MA, van Weeren L, Gadella TW, Royant A (2012). "Structure-guided evolution of cyan fluorescent proteins towards a quantum yield of 93%". Nature Communications. 3: 751. Bibcode:2012NatCo...3..751G. doi:10.1038/ncomms1738. PMC 3316892. PMID 22434194.
  27. ^ Miesenböck G, De Angelis DA, Rothman JE (Jul 1998). "Visualizing secretion and synaptic transmission with pH-sensitive green fluorescent proteins". Nature. 394 (6689): 192–5. Bibcode:1998Natur.394..192M. doi:10.1038/28190. PMID 9671304. S2CID 4320849.
  28. ^ Hanson GT, Aggeler R, Oglesbee D, Cannon M, Capaldi RA, Tsien RY, Remington SJ (Mar 2004). "Investigating mitochondrial redox potential with redox-sensitive green fluorescent protein indicators". The Journal of Biological Chemistry. 279 (13): 13044–53. doi:10.1074/jbc.M312846200. PMID 14722062.
  29. ^ Zacharias DA, Violin JD, Newton AC, Tsien RY (May 2002). "Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells". Science. 296 (5569): 913–16. Bibcode:2002Sci...296..913Z. doi:10.1126/science.1068539. PMID 11988576. S2CID 14957077.
  30. ^ Yue JX, Holland ND, Holland LZ, Deheyn DD (June 2016). "The evolution of genes encoding for green fluorescent proteins: insights from cephalochordates (amphioxus)". Scientific Reports. 6 (1): 28350. Bibcode:2016NatSR...628350Y. doi:10.1038/srep28350. PMC 4911609. PMID 27311567.
  31. ^ Hunt ME, Scherrer MP, Ferrari FD, Matz MV (July 2010). "Very bright green fluorescent proteins from the Pontellid copepod Pontella mimocerami". PLOS ONE. 5 (7): e11517. Bibcode:2010PLoSO...511517H. doi:10.1371/journal.pone.0011517. PMC 2904364. PMID 20644720.
  32. ^ "eGFP". FPbase.
  33. ^ Chudakov DM, Belousov VV, Zaraisky AG, Novoselov VV, Staroverov DB, Zorov DB, Lukyanov S, Lukyanov KA (February 2003). "Kindling fluorescent proteins for precise in vivo photolabeling". Nature Biotechnology. 21 (2): 191–4. doi:10.1038/nbt778. PMID 12524551. S2CID 52887792.
  34. ^ Wiens MD, Shen Y, Li X, Salem MA, Smisdom N, Zhang W, Brown A, Campbell RE (December 2016). "A Tandem Green-Red Heterodimeric Fluorescent Protein with High FRET Efficiency". ChemBioChem. 17 (24): 2361–2367. doi:10.1002/cbic.201600492. PMID 27781394. S2CID 4301322.
  35. ^ Drepper T, Eggert T, Circolone F, Heck A, Krauss U, Guterl JK, et al. (April 2007). "Reporter proteins for in vivo fluorescence without oxygen". Nature Biotechnology. 25 (4): 443–445. doi:10.1038/nbt1293. PMID 17351616. S2CID 7335755.
  36. ^ a b Rodriguez EA, Tran GN, Gross LA, Crisp JL, Shu X, Lin JY, Tsien RY (September 2016). "A far-red fluorescent protein evolved from a cyanobacterial phycobiliprotein". Nature Methods. 13 (9): 763–9. doi:10.1038/nmeth.3935. PMC 5007177. PMID 27479328.
  37. ^ Mattson S, Tran GN, Rodriguez EA (2023). "Directed Evolution of Fluorescent Proteins in Bacteria". In Sharma M (ed.). Fluorescent Proteins. Methods in Molecular Biology. Vol. 2564. New York, NY: Springer US. pp. 75–97. doi:10.1007/978-1-0716-2667-2_4. ISBN 978-1-0716-2666-5. PMID 36107338.
  38. ^ Tsien RY (1998-01-01). "The green fluorescent protein". Annual Review of Biochemistry. 67 (1): 509–44. doi:10.1146/annurev.biochem.67.1.509. PMID 9759496. S2CID 8138960.
  39. ^ Maiti, Atanu; Buffalo, Cosmo Z.; Saurabh, Saumya; Montecinos-Franjola, Felipe; Hachey, Justin S.; Conlon, William J.; Tran, Geraldine N.; Hassan, Bakar; Walters, Kylie J.; Drobizhev, Mikhail; Moerner, W. E.; Ghosh, Partho; Matsuo, Hiroshi; Tsien, Roger Y.; Lin, John Y. (2023-07-12). "Structural and photophysical characterization of the small ultra-red fluorescent protein". Nature Communications. 14 (1): 4155. Bibcode:2023NatCo..14.4155M. doi:10.1038/s41467-023-39776-9. ISSN 2041-1723. PMC 10338489. PMID 37438348.
  40. ^ Rodriguez EA, Campbell RE, Lin JY, Lin MZ, Miyawaki A, Palmer AE, Shu X, Zhang J, Tsien RY (February 2017). "The Growing and Glowing Toolbox of Fluorescent and Photoactive Proteins". Trends in Biochemical Sciences. 42 (2): 111–129. doi:10.1016/j.tibs.2016.09.010. PMC 5272834. PMID 27814948.
  41. ^ Montecinos-Franjola F, Lin JY, Rodriguez EA (2020-11-16). "Fluorescent proteins for in vivo imaging, where's the biliverdin?". Biochemical Society Transactions. 48 (6): 2657–2667. doi:10.1042/BST20200444. PMID 33196077. S2CID 226971864.
  42. ^ Bokman SH, Ward WW (1982). "Reversible denaturation of Aequorea green-fluorescent protein: physical separation and characterization of the renatured protein". Biochemistry. 21 (19): 4535–4540. doi:10.1021/bi00262a003. PMID 6128025.
  43. ^ Pouwels LJ, Zhang L, Chan NH, Dorrestein PC, Wachter RM (Sep 2008). "Kinetic isotope effect studies on the de novo rate of chromophore formation in fast- and slow-maturing GFP variants". Biochemistry. 47 (38): 10111–22. doi:10.1021/bi8007164. PMC 2643082. PMID 18759496.
  44. ^ Chudakov DM, Matz MV, Lukyanov S, Lukyanov KA (Jul 2010). "Fluorescent proteins and their applications in imaging living cells and tissues". Physiological Reviews. 90 (3): 1103–63. doi:10.1152/physrev.00038.2009. PMID 20664080. S2CID 10767597.
  45. ^ Stepanenko OV, Verkhusha VV, Shavlovsky MM, Kuznetsova IM, Uversky VN, Turoverov KK (November 2008). "Understanding the role of Arg96 in structure and stability of green fluorescent protein". Proteins. 73 (3): 539–551. doi:10.1002/prot.22089. PMC 2908022. PMID 18470931.
  46. ^ a b Rosenow MA, Huffman HA, Phail ME, Wachter RM (April 2004). "The crystal structure of the Y66L variant of green fluorescent protein supports a cyclization-oxidation-dehydration mechanism for chromophore maturation". Biochemistry. 43 (15): 4464–4472. doi:10.1021/bi0361315. PMID 15078092.
  47. ^ Ma Y, Yu JG, Sun Q, Li Z, Smith SC (2015-07-01). "The mechanism of dehydration in chromophore maturation of wild-type green fluorescent protein: A theoretical study". Chemical Physics Letters. 631–632: 42–46. Bibcode:2015CPL...631...42M. doi:10.1016/j.cplett.2015.04.061. ISSN 0009-2614.
  48. ^ Jugder BE, Welch J, Braidy N, Marquis CP (2016-07-26). "Construction and use of a Cupriavidus necator H16 soluble hydrogenase promoter (PSH) fusion to gfp (green fluorescent protein)". PeerJ. 4: e2269. doi:10.7717/peerj.2269. PMC 4974937. PMID 27547572.
  49. ^ Arun KH, Kaul CL, Ramarao P (2005). "Green fluorescent proteins in receptor research: an emerging tool for drug discovery". Journal of Pharmacological and Toxicological Methods. 51 (1): 1–23. doi:10.1016/j.vascn.2004.07.006. PMID 15596111.
  50. ^ Song YH, Kim CS, Seo JH (April 2016). "Noninvasive monitoring of environmental toxicity through green fluorescent protein expressing Escherichia coli". Korean Journal of Chemical Engineering. 33 (4): 1331–6. doi:10.1007/s11814-015-0253-1. S2CID 62828580.
  51. ^ Pan Y, Leifert A, Graf M, Schiefer F, Thoröe-Boveleth S, Broda J, Halloran MC, Hollert H, Laaf D, Simon U, Jahnen-Dechent W (March 2013). "High-sensitivity real-time analysis of nanoparticle toxicity in green fluorescent protein-expressing zebrafish". Small. Weinheim an Der Bergstrasse, Germany. 9 (6): 863–9. doi:10.1002/smll.201201173. PMID 23143852.
  52. ^ Chalfie M (Jun 2009). "GFP: Lighting up life". Proceedings of the National Academy of Sciences of the United States of America. 106 (25): 10073–10080. Bibcode:2009PNAS..10610073C. doi:10.1073/pnas.0904061106. PMC 2700921. PMID 19553219.
  53. ^ Yuste R (Dec 2005). "Fluorescence microscopy today". Nature Methods. 2 (12): 902–4. doi:10.1038/nmeth1205-902. PMID 16299474. S2CID 205418407.
  54. ^ Terskikh A, Fradkov A, Ermakova G, Zaraisky A, Tan P, Kajava AV, et al. (November 2000). ""Fluorescent timer": protein that changes color with time". Science. 290 (5496): 1585–8. Bibcode:2000Sci...290.1585T. doi:10.1126/science.290.5496.1585. PMID 11090358.
  55. ^ Patterson GH, Lippincott-Schwartz J (September 2002). "A photoactivatable GFP for selective photolabeling of proteins and cells". Science. 297 (5588): 1873–7. Bibcode:2002Sci...297.1873P. doi:10.1126/science.1074952. PMID 12228718. S2CID 45058411.
  56. ^ Lin W, Mehta S, Zhang J (October 2019). "Genetically encoded fluorescent biosensors illuminate kinase signaling in cancer". The Journal of Biological Chemistry. 294 (40): 14814–14822. doi:10.1074/jbc.REV119.006177. PMC 6779441. PMID 31434714.
  57. ^ Komorowski M, Finkenstädt B, Rand D (Jun 2010). "Using a single fluorescent reporter gene to infer half-life of extrinsic noise and other parameters of gene expression". Biophysical Journal. 98 (12): 2759–2769. Bibcode:2010BpJ....98.2759K. doi:10.1016/j.bpj.2010.03.032. PMC 2884236. PMID 20550887.
  58. ^ Gunkel M, Erdel F, Rippe K, Lemmer P, Kaufmann R, Hörmann C, Amberger R, Cremer C (Jun 2009). "Dual color localization microscopy of cellular nanostructures". Biotechnology Journal. 4 (6): 927–38. doi:10.1002/biot.200900005. PMID 19548231. S2CID 18162278.
  59. ^ Chudakov DM, Lukyanov S, Lukyanov KA (Dec 2005). "Fluorescent proteins as a toolkit for in vivo imaging". Trends in Biotechnology. 23 (12): 605–13. doi:10.1016/j.tibtech.2005.10.005. PMID 16269193.
  60. ^ Livet J, Weissman TA, Kang H, Draft RW, Lu J, Bennis RA, Sanes JR, Lichtman JW (Nov 2007). "Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system". Nature. 450 (7166): 56–62. Bibcode:2007Natur.450...56L. doi:10.1038/nature06293. PMID 17972876. S2CID 4402093.
  61. ^ Baker BJ, Mutoh H, Dimitrov D, Akemann W, Perron A, Iwamoto Y, Jin L, Cohen LB, Isacoff EY, Pieribone VA, Hughes T, Knöpfel T (Aug 2008). "Genetically encoded fluorescent sensors of membrane potential". Brain Cell Biology. 36 (1–4): 53–67. doi:10.1007/s11068-008-9026-7. PMC 2775812. PMID 18679801.
  62. ^ Adesnik H, Nicoll RA, England PM (Dec 2005). "Photoinactivation of native AMPA receptors reveals their real-time trafficking". Neuron. 48 (6): 977–85. doi:10.1016/j.neuron.2005.11.030. PMID 16364901.
  63. ^ Lakadamyali M, Rust MJ, Babcock HP, Zhuang X (Aug 2003). "Visualizing infection of individual influenza viruses". Proceedings of the National Academy of Sciences of the United States of America. 100 (16): 9280–5. Bibcode:2003PNAS..100.9280L. doi:10.1073/pnas.0832269100. PMC 170909. PMID 12883000.
  64. ^ Joo KI, Wang P (Oct 2008). "Visualization of targeted transduction by engineered lentiviral vectors". Gene Therapy. 15 (20): 1384–96. doi:10.1038/gt.2008.87. PMC 2575058. PMID 18480844.
  65. ^ Remy S, Tesson L, Usal C, Menoret S, Bonnamain V, Nerriere-Daguin V, Rossignol J, Boyer C, Nguyen TH, Naveilhan P, Lescaudron L, Anegon I (Oct 2010). "New lines of GFP transgenic rats relevant for regenerative medicine and gene therapy". Transgenic Research. 19 (5): 745–63. doi:10.1007/s11248-009-9352-2. PMID 20094912. S2CID 42499768.
  66. ^ Elliott G, McGrath J, Crockett-Torabi E (Jun 2000). "Green fluorescent protein: A novel viability assay for cryobiological applications". Cryobiology. 40 (4): 360–369. doi:10.1006/cryo.2000.2258. PMID 10924267.
  67. ^ Fakhrudin N, Ladurner A, Atanasov AG, Heiss EH, Baumgartner L, Markt P, Schuster D, Ellmerer EP, Wolber G, Rollinger JM, Stuppner H, Dirsch VM (Apr 2010). "Computer-aided discovery, validation, and mechanistic characterization of novel neolignan activators of peroxisome proliferator-activated receptor gamma". Molecular Pharmacology. 77 (4): 559–66. doi:10.1124/mol.109.062141. PMC 3523390. PMID 20064974.
  68. ^ Gather MC, Yun SH (2011). "Single-cell biological lasers". Nature Photonics. 5 (7): 406–410. Bibcode:2011NaPho...5..406G. doi:10.1038/nphoton.2011.99. S2CID 54971962.
  69. ^ Matson J (2011). "Green Fluorescent Protein Makes for Living Lasers". Scientific American. Retrieved 2011-06-13.
  70. ^ Kouros-Mehr H, Bechis SK, Slorach EM, Littlepage LE, Egeblad M, Ewald AJ, Pai SY, Ho IC, Werb Z (Feb 2008). "GATA-3 links tumor differentiation and dissemination in a luminal breast cancer model". Cancer Cell. 13 (2): 141–52. doi:10.1016/j.ccr.2008.01.011. PMC 2262951. PMID 18242514.
  71. ^ Cabantous S, Terwilliger TC, Waldo GS (January 2005). "Protein tagging and detection with engineered self-assembling fragments of green fluorescent protein". Nature Biotechnology. 23 (1): 102–7. doi:10.1038/nbt1044. PMID 15580262. S2CID 25833063.
  72. ^ Rodman MK, Yadav NS, Artus NN (2002-09-01). "Progression of geminivirus-induced transgene silencing is associated with transgene methylation". New Phytologist. 155 (3): 461–468. doi:10.1046/j.1469-8137.2002.00467.x. PMID 33873315.
  73. ^ Zhu YJ, Agbayani R, Moore PH (Apr 2004). "Green fluorescent protein as a visual selection marker for papaya (Carica papaya L.) transformation". Plant Cell Reports. 22 (9): 660–7. doi:10.1007/s00299-004-0755-5. PMID 14749892. S2CID 23198182.
  74. ^ Niwa Y, Hirano T, Yoshimoto K, Shimizu M, Kobayashi H (1999). "Non-invasive quantitative detection and applications of non-toxic, S65T-type green fluorescent protein in living plants". The Plant Journal. 18 (4): 455–63. doi:10.1046/j.1365-313X.1999.00464.x. PMID 10406127. S2CID 292648.
  75. ^ Baker SS, Vidican CB, Cameron DS, Greib HG, Jarocki CC, Setaputri AW, Spicuzza CH, Burr AA, Waqas MA, Tolbert DA (2012-01-01). "An epifluorescent attachment improves whole-plant digital photography of Arabidopsis thaliana expressing red-shifted green fluorescent protein". AoB Plants. 2012: pls003. doi:10.1093/aobpla/pls003. PMC 3296078. PMID 22479674.
  76. ^ "PlantEdDL - Using SRL digital cameras in quantitative investigations of plants expressing green fluorescent protein (GFP)". planted.botany.org. Retrieved 2016-03-23.
  77. ^ Kac E. "GFP Bunny".
  78. ^ . Archived from the original on February 14, 2009. Retrieved August 30, 2016.
  79. ^ Scientists in Taiwan breed fluorescent green pigs
  80. ^ Wongsrikeao P, Saenz D, Rinkoski T, Otoi T, Poeschla E (2011). "Antiviral restriction factor transgenesis in the domestic cat". Nature Methods. 8 (10): 853–9. doi:10.1038/nmeth.1703. PMC 4006694. PMID 21909101.
  81. ^ "Fluorescent puppy is world's first transgenic dog".
  82. ^ Voss-Andreae J (2005). "Protein Sculptures: Life's Building Blocks Inspire Art". Leonardo. 38: 41–45. doi:10.1162/leon.2005.38.1.41. S2CID 57558522.
  83. ^ Pawlak A (2005). "Inspirierende Proteine". Physik Journal. 4: 12.
  84. ^ "Julian Voss-Andreae Sculpture". Retrieved 2007-06-14.

Further reading edit

  • Pieribone V, Gruber D (2006). Aglow in the Dark: The Revolutionary Science of Biofluorescence. Cambridge: Belknap Press. ISBN 978-0-674-01921-8. OCLC 60321612. Popular science book describing history and discovery of GFP
  • Zimmer M (2005). Glowing Genes: A Revolution In Biotechnology. Buffalo, NY: Prometheus Books. ISBN 978-1-59102-253-4. OCLC 56614624.

External links edit

  • A comprehensive article on fluorescent proteins at Scholarpedia
  • Brief summary of landmark GFP papers
  • Interactive Java applet demonstrating the chemistry behind the
  • Video of 2008 Nobel Prize lecture of Roger Tsien on fluorescent proteins
  • Excitation and emission spectra for various fluorescent proteins
  • Green Fluorescent Protein Chem Soc Rev themed issue dedicated to the 2008 Nobel Prize winners in Chemistry, Professors Osamu Shimomura, Martin Chalfie and Roger Y. Tsien
  • Molecule of the Month, June 2003: an illustrated overview of GFP by David Goodsell.
  • Molecule of the Month, June 2014: an illustrated overview of GFP-like variants by David Goodsell.
  • Green Fluorescent Protein on FPbase, a fluorescent protein database
  • Overview of all the structural information available in the PDB for UniProt: P42212 (Green fluorescent protein) at the PDBe-KB.

green, fluorescent, protein, egfp, redirects, here, airport, with, that, icao, airport, code, pembrey, airport, green, fluorescent, protein, protein, that, exhibits, green, fluorescence, when, exposed, light, blue, ultraviolet, range, label, traditionally, ref. EGFP redirects here For the airport with that ICAO airport code see Pembrey Airport The green fluorescent protein GFP is a protein that exhibits green fluorescence when exposed to light in the blue to ultraviolet range 2 3 The label GFP traditionally refers to the protein first isolated from the jellyfish Aequorea victoria and is sometimes called avGFP However GFPs have been found in other organisms including corals sea anemones zoanithids copepods and lancelets 4 Green fluorescent proteinStructure of the Aequorea victoria green fluorescent protein 1 IdentifiersSymbolGFPPfamPF01353Pfam clanCL0069InterProIPR011584CATH1emaSCOP21ema SCOPe SUPFAMAvailable protein structures Pfam structures ECOD PDBRCSB PDB PDBe PDBjPDBsumstructure summaryGreen fluorescent proteinIdentifiersOrganismAequorea victoriaSymbolGFPUniProtP42212Search forStructuresSwiss modelDomainsInterProThe GFP from A victoria has a major excitation peak at a wavelength of 395 nm and a minor one at 475 nm Its emission peak is at 509 nm which is in the lower green portion of the visible spectrum The fluorescence quantum yield QY of GFP is 0 79 The GFP from the sea pansy Renilla reniformis has a single major excitation peak at 498 nm GFP makes for an excellent tool in many forms of biology due to its ability to form an internal chromophore without requiring any accessory cofactors gene products or enzymes substrates other than molecular oxygen 5 In cell and molecular biology the GFP gene is frequently used as a reporter of expression 6 It has been used in modified forms to make biosensors and many animals have been created that express GFP which demonstrates a proof of concept that a gene can be expressed throughout a given organism in selected organs or in cells of interest GFP can be introduced into animals or other species through transgenic techniques and maintained in their genome and that of their offspring To date GFP has been expressed in many species including bacteria yeasts fungi fish and mammals including in human cells Scientists Roger Y Tsien Osamu Shimomura and Martin Chalfie were awarded the 2008 Nobel Prize in Chemistry on 10 October 2008 for their discovery and development of the green fluorescent protein Most commercially available genes for GFP and similar fluorescent proteins are around 730 base pairs long The natural protein has 238 amino acids Its molecular mass is 27 kD 7 Therefore fusing the GFP gene to the gene of a protein of interest can significantly increase the protein s size and molecular mass and can impair the protein s natural function or change its location or trajectory of transport within the cell 8 Contents 1 Background 1 1 Wild type GFP wtGFP 1 2 GFP derivatives 1 3 Nomenclature 2 In nature 3 Other fluorescent proteins 4 Structure 4 1 Autocatalytic formation of the chromophore in wtGFP 5 Applications 5 1 Reporter assays 5 1 1 Advantages 5 2 Fluorescence microscopy 5 3 Split GFP 5 4 Macro photography 5 5 Transgenic pets 5 6 Art 6 See also 7 References 8 Further reading 9 External linksBackground edit nbsp Aequorea victoria nbsp 3D reconstruction of confocal image of VEGF overexpressing neural progenitors red and GFP positive control neural progenitor cells green in the rat olfactory bulb RECA 1 positive blood vessels blue color Wild type GFP wtGFP edit In the 1960s and 1970s GFP along with the separate luminescent protein aequorin an enzyme that catalyzes the breakdown of luciferin releasing light was first purified from the jellyfish Aequorea victoria and its properties studied by Osamu Shimomura 9 In A victoria GFP fluorescence occurs when aequorin interacts with Ca2 ions inducing a blue glow Some of this luminescent energy is transferred to the GFP shifting the overall color towards green 10 However its utility as a tool for molecular biologists did not begin to be realized until 1992 when Douglas Prasher reported the cloning and nucleotide sequence of wtGFP in Gene 11 The funding for this project had run out so Prasher sent cDNA samples to several labs The lab of Martin Chalfie expressed the coding sequence of wtGFP with the first few amino acids deleted in heterologous cells of E coli and C elegans publishing the results in Science in 1994 12 Frederick Tsuji s lab independently reported the expression of the recombinant protein one month later 13 Remarkably the GFP molecule folded and was fluorescent at room temperature without the need for exogenous cofactors specific to the jellyfish Although this near wtGFP was fluorescent it had several drawbacks including dual peaked excitation spectra pH sensitivity chloride sensitivity poor fluorescence quantum yield poor photostability and poor folding at 37 C 99 F The first reported crystal structure of a GFP was that of the S65T mutant by the Remington group in Science in 1996 14 One month later the Phillips group independently reported the wild type GFP structure in Nature Biotechnology 15 These crystal structures provided vital background on chromophore formation and neighboring residue interactions Researchers have modified these residues by directed and random mutagenesis to produce the wide variety of GFP derivatives in use today Further research into GFP has shown that it is resistant to detergents proteases guanidinium chloride GdmCl treatments and drastic temperature changes 16 GFP derivatives edit nbsp The diversity of genetic mutations is illustrated by this San Diego beach scene drawn with living bacteria expressing 8 different colors of fluorescent proteins derived from GFP and dsRed Due to the potential for widespread usage and the evolving needs of researchers many different mutants of GFP have been engineered 17 18 The first major improvement was a single point mutation S65T reported in 1995 in Nature by Roger Tsien 19 This mutation dramatically improved the spectral characteristics of GFP resulting in increased fluorescence photostability and a shift of the major excitation peak to 488 nm with the peak emission kept at 509 nm This matched the spectral characteristics of commonly available FITC filter sets increasing the practicality of use by the general researcher A 37 C folding efficiency F64L point mutant to this scaffold yielding enhanced GFP EGFP was discovered in 1995 by the laboratories of Thastrup 20 and Falkow 21 EGFP allowed the practical use of GFPs in mammalian cells EGFP has an extinction coefficient denoted e of 55 000 M 1cm 1 22 The fluorescence quantum yield QY of EGFP is 0 60 The relative brightness expressed as e QY is 33 000 M 1cm 1 Superfolder GFP sfGFP a series of mutations that allow GFP to rapidly fold and mature even when fused to poorly folding peptides was reported in 2006 23 Many other mutations have been made including color mutants in particular blue fluorescent protein EBFP EBFP2 Azurite mKalama1 cyan fluorescent protein ECFP Cerulean CyPet mTurquoise2 and yellow fluorescent protein derivatives YFP Citrine Venus YPet BFP derivatives except mKalama1 contain the Y66H substitution They exhibit a broad absorption band in the ultraviolet centered close to 380 nanometers and an emission maximum at 448 nanometers A green fluorescent protein mutant BFPms1 that preferentially binds Zn II and Cu II has been developed BFPms1 have several important mutations including and the BFP chromophore Y66H Y145F for higher quantum yield H148G for creating a hole into the beta barrel and several other mutations that increase solubility Zn II binding increases fluorescence intensity while Cu II binding quenches fluorescence and shifts the absorbance maximum from 379 to 444 nm Therefore they can be used as Zn biosensor 24 nbsp A palette of variants of GFP and DsRed Chromophore binding The critical mutation in cyan derivatives is the Y66W substitution which causes the chromophore to form with an indole rather than phenol component Several additional compensatory mutations in the surrounding barrel are required to restore brightness to this modified chromophore due to the increased bulk of the indole group In ECFP and Cerulean the N terminal half of the seventh strand exhibits two conformations These conformations both have a complex set of van der Waals interactions with the chromophore The Y145A and H148D mutations in Cerulean stabilize these interactions and allow the chromophore to be more planar better packed and less prone to collisional quenching 25 Additional site directed random mutagenesis in combination with fluorescence lifetime based screening has further stabilized the seventh b strand resulting in a bright variant mTurquoise2 with a quantum yield QY of 0 93 26 The red shifted wavelength of the YFP derivatives is accomplished by the T203Y mutation and is due to p electron stacking interactions between the substituted tyrosine residue and the chromophore 3 These two classes of spectral variants are often employed for Forster resonance energy transfer FRET experiments Genetically encoded FRET reporters sensitive to cell signaling molecules such as calcium or glutamate protein phosphorylation state protein complementation receptor dimerization and other processes provide highly specific optical readouts of cell activity in real time Semirational mutagenesis of a number of residues led to pH sensitive mutants known as pHluorins and later super ecliptic pHluorins By exploiting the rapid change in pH upon synaptic vesicle fusion pHluorins tagged to synaptobrevin have been used to visualize synaptic activity in neurons 27 Redox sensitive GFP roGFP was engineered by introduction of cysteines into the beta barrel structure The redox state of the cysteines determines the fluorescent properties of roGFP 28 Nomenclature edit The nomenclature of modified GFPs is often confusing due to overlapping mapping of several GFP versions onto a single name For example mGFP often refers to a GFP with an N terminal palmitoylation that causes the GFP to bind to cell membranes However the same term is also used to refer to monomeric GFP which is often achieved by the dimer interface breaking A206K mutation 29 Wild type GFP has a weak dimerization tendency at concentrations above 5 mg mL mGFP also stands for modified GFP which has been optimized through amino acid exchange for stable expression in plant cells In nature edit nbsp Live lancelet B floridae under a fluorescent microscope nbsp In the marine copepod Pontella mimoceramiThe purpose of both the primary bioluminescence from aequorin s action on luciferin and the secondary fluorescence of GFP in jellyfish is unknown GFP is co expressed with aequorin in small granules around the rim of the jellyfish bell The secondary excitation peak 480 nm of GFP does absorb some of the blue emission of aequorin giving the bioluminescence a more green hue The serine 65 residue of the GFP chromophore is responsible for the dual peaked excitation spectra of wild type GFP It is conserved in all three GFP isoforms originally cloned by Prasher Nearly all mutations of this residue consolidate the excitation spectra to a single peak at either 395 nm or 480 nm The precise mechanism of this sensitivity is complex but it seems involves donation of a hydrogen from serine 65 to glutamate 222 which influences chromophore ionization 3 Since a single mutation can dramatically enhance the 480 nm excitation peak making GFP a much more efficient partner of aequorin A victoria appears to evolutionarily prefer the less efficient dual peaked excitation spectrum Roger Tsien has speculated that varying hydrostatic pressure with depth may affect serine 65 s ability to donate a hydrogen to the chromophore and shift the ratio of the two excitation peaks Thus the jellyfish may change the color of its bioluminescence with depth However a collapse in the population of jellyfish in Friday Harbor where GFP was originally discovered has hampered further study of the role of GFP in the jellyfish s natural environment Most species of lancelet are known to produce GFP in various regions of their body 30 Unlike A victoria lancelets do not produce their own blue light and the origin of their endogenous GFP is still unknown Some speculate that it attracts plankton towards the mouth of the lancelet serving as a passive hunting mechanism It may also serve as a photoprotective agent in the larvae preventing damage caused by high intensity blue light by converting it into lower intensity green light However these theories have not been tested GFP like proteins have been found in multiple species of marine copepods particularly from the Pontellidae and Aetideidae families 31 GFP isolated from Pontella mimocerami has shown high levels of brightness with a quantum yield of 0 92 making them nearly two fold brighter than the commonly used EGFP isolated from A victoria 32 Other fluorescent proteins edit nbsp Different proteins produce different fluorescent colors when exposed to ultraviolet light There are many GFP like proteins that despite being in the same protein family as GFP are not directly derived from Aequorea victoria These include dsRed eqFP611 Dronpa TagRFPs KFP EosFP IrisFP Dendra and so on Having been developed from proteins in different organisms these proteins can sometimes display unanticipated approaches to chromophore formation Some of these such as KFP are developed from naturally non or weakly fluorescent proteins to be greatly improved upon by mutagenesis 33 When GFP like barrels of different spectra characteristics are used the excitation spectra of one chromophore can be used to power another chromophore FRET allowing for conversion between wavelengths of light 34 FMN binding fluorescent proteins FbFPs were developed in 2007 and are a class of small 11 16 kDa oxygen independent fluorescent proteins that are derived from blue light receptors They are intended especially for the use under anaerobic or hypoxic conditions since the formation and binding of the Flavin chromophore does not require molecular oxygen as it is the case with the synthesis of the GFP chromophore 35 nbsp White light image or image seen by the eye of fluorescent proteins in image above Fluorescent proteins with other chromophores such as UnaG with bilirubin can display unique properties like red shifted emission above 600 nm or photoconversion from a green emitting state to a red emitting state They can have excitation and emission wavelengths far enough apart to achieve conversion between red and green light A new class of fluorescent protein was evolved from a cyanobacterial Trichodesmium erythraeum phycobiliprotein a allophycocyanin and named small ultra red fluorescent protein smURFP in 2016 smURFP autocatalytically self incorporates the chromophore biliverdin without the need of an external protein known as a lyase 36 37 Jellyfish and coral derived GFP like proteins require oxygen and produce a stoichiometric amount of hydrogen peroxide upon chromophore formation 38 smURFP does not require oxygen or produce hydrogen peroxide and uses the chromophore biliverdin smURFP has a large extinction coefficient 180 000 M 1 cm 1 and has a modest quantum yield 0 20 which makes it comparable biophysical brightness to eGFP and 2 fold brighter than most red or far red fluorescent proteins derived from coral smURFP spectral properties are similar to the organic dye Cy5 36 39 nbsp E coli colonies expressing fluorescent proteins Reviews on new classes of fluorescent proteins and applications can be found in the cited reviews 40 41 Structure editGFP has a beta barrel structure consisting of eleven b strands with a pleated sheet arrangement with an alpha helix containing the covalently bonded chromophore 4 p hydroxybenzylidene imidazolidin 5 one HBI running through the center 3 14 15 Five shorter alpha helices form caps on the ends of the structure The beta barrel structure is a nearly perfect cylinder 42A long and 24A in diameter some studies have reported a diameter of 30A 16 14 creating what is referred to as a b can formation which is unique to the GFP like family 15 HBI the spontaneously modified form of the tripeptide Ser65 Tyr66 Gly67 is nonfluorescent in the absence of the properly folded GFP scaffold and exists mainly in the un ionized phenol form in wtGFP 42 Inward facing sidechains of the barrel induce specific cyclization reactions in Ser65 Tyr66 Gly67 that induce ionization of HBI to the phenolate form and chromophore formation This process of post translational modification is referred to as maturation 43 The hydrogen bonding network and electron stacking interactions with these sidechains influence the color intensity and photostability of GFP and its numerous derivatives 44 The tightly packed nature of the barrel excludes solvent molecules protecting the chromophore fluorescence from quenching by water In addition to the auto cyclization of the Ser65 Tyr66 Gly67 a 1 2 dehydrogenation reaction occurs at the Tyr66 residue 16 Besides the three residues that form the chromophore residues such as Gln94 Arg96 His148 Thr203 and Glu222 all act as stabilizers The residues of Gln94 Arg96 and His148 are able to stabilize by delocalizing the chromophore charge Arg96 is the most important stabilizing residue due to the fact that it prompts the necessary structural realignments that are necessary from the HBI ring to occur Any mutation to the Arg96 residue would result in a decrease in the development rate of the chromophore because proper electrostatic and steric interactions would be lost Tyr66 is the recipient of hydrogen bonds and does not ionize in order to produce favorable electrostatics 45 nbsp GFP Movie showing entire structure and zoom in to fluorescent chromophore nbsp GFP molecules drawn in cartoon style one fully and one with the side of the beta barrel cut away to reveal the chromophore highlighted as ball and stick From PDB 1GFL nbsp GFP ribbon diagram From PDB 1EMA Autocatalytic formation of the chromophore in wtGFP edit For a synthetic analogue see also 3 5 Difluoro 4 hydroxybenzylidene imidazolinone nbsp Mechanistically the process involves base mediated cyclization followed by dehydration and oxidation In the reaction of 7a to 8 involves the formation of an enamine from the imine while in the reaction of 7b to 9 a proton is abstracted 46 The formed HBI fluorophore is highlighted in green The reactions are catalyzed by residues Glu222 and Arg96 46 47 An analogous mechanism is also possible with threonine in place of Ser65 Applications editReporter assays edit Green fluorescent protein may be used as a reporter gene 48 49 For example GFP can be used as a reporter for environmental toxicity levels This protein has been shown to be an effective way to measure the toxicity levels of various chemicals including ethanol p formaldehyde phenol triclosan and paraben GFP is great as a reporter protein because it has no effect on the host when introduced to the host s cellular environment Due to this ability no external visualization stain ATP or cofactors are needed With regards to pollutant levels the fluorescence was measured in order to gauge the effect that the pollutants have on the host cell The cellular density of the host cell was also measured Results from the study conducted by Song Kim amp Seo 2016 showed that there was a decrease in both fluorescence and cellular density as pollutant levels increased This was indicative of the fact that cellular activity had decreased More research into this specific application in order to determine the mechanism by which GFP acts as a pollutant marker 50 Similar results have been observed in zebrafish because zebrafish that were injected with GFP were approximately twenty times more susceptible to recognize cellular stresses than zebrafish that were not injected with GFP 51 Advantages edit The biggest advantage of GFP is that it can be heritable depending on how it was introduced allowing for continued study of cells and tissues it is expressed in Visualizing GFP is noninvasive requiring only illumination with blue light GFP alone does not interfere with biological processes but when fused to proteins of interest careful design of linkers is required to maintain the function of the protein of interest Moreover if used with a monomer it is able to diffuse readily throughout cells 52 Fluorescence microscopy edit Main article Fluorescence microscope nbsp Superresolution with two fusion proteins GFP Snf2H and RFP H2A Co localisation studies 2CLM in the nucleus of a bone cancer cell 120 000 localized molecules in a widefield area 470 µm2 The availability of GFP and its derivatives has thoroughly redefined fluorescence microscopy and the way it is used in cell biology and other biological disciplines 53 While most small fluorescent molecules such as FITC fluorescein isothiocyanate are strongly phototoxic when used in live cells fluorescent proteins such as GFP are usually much less harmful when illuminated in living cells This has triggered the development of highly automated live cell fluorescence microscopy systems which can be used to observe cells over time expressing one or more proteins tagged with fluorescent proteins There are many techniques to utilize GFP in a live cell imaging experiment The most direct way of utilizing GFP is to directly attach it to a protein of interest For example GFP can be included in a plasmid expressing other genes to indicate a successful transfection of a gene of interest Another method is to use a GFP that contains a mutation where the fluorescence will change from green to yellow over time which is referred to as a fluorescent timer With the fluorescent timer researchers can study the state of protein production such as recently activated continuously activated or recently deactivated based on the color reported by the fluorescent protein 54 In yet another example scientists have modified GFP to become active only after exposure to irradiation giving researchers a tool to selectively activate certain portions of a cell and observe where proteins tagged with the GFP move from the starting location 55 These are only two examples in a burgeoning field of fluorescent microcopy and a more complete review of biosensors utilizing GFP and other fluorescent proteins can be found here 56 For example GFP had been widely used in labelling the spermatozoa of various organisms for identification purposes as in Drosophila melanogaster where expression of GFP can be used as a marker for a particular characteristic GFP can also be expressed in different structures enabling morphological distinction In such cases the gene for the production of GFP is incorporated into the genome of the organism in the region of the DNA that codes for the target proteins and that is controlled by the same regulatory sequence that is the gene s regulatory sequence now controls the production of GFP in addition to the tagged protein s In cells where the gene is expressed and the tagged proteins are produced GFP is produced at the same time Thus only those cells in which the tagged gene is expressed or the target proteins are produced will fluoresce when observed under fluorescence microscopy Analysis of such time lapse movies has redefined the understanding of many biological processes including protein folding protein transport and RNA dynamics which in the past had been studied using fixed i e dead material Obtained data are also used to calibrate mathematical models of intracellular systems and to estimate rates of gene expression 57 Similarly GFP can be used as an indicator of protein expression in heterologous systems In this scenario fusion proteins containing GFP are introduced indirectly using RNA of the construct or directly with the tagged protein itself This method is useful for studying structural and functional characteristics of the tagged protein on a macromolecular or single molecule scale with fluorescence microscopy The Vertico SMI microscope using the SPDM Phymod technology uses the so called reversible photobleaching effect of fluorescent dyes like GFP and its derivatives to localize them as single molecules in an optical resolution of 10 nm This can also be performed as a co localization of two GFP derivatives 2CLM 58 Another powerful use of GFP is to express the protein in small sets of specific cells This allows researchers to optically detect specific types of cells in vitro in a dish or even in vivo in the living organism 59 Genetically combining several spectral variants of GFP is a useful trick for the analysis of brain circuitry Brainbow 60 Other interesting uses of fluorescent proteins in the literature include using FPs as sensors of neuron membrane potential 61 tracking of AMPA receptors on cell membranes 62 viral entry and the infection of individual influenza viruses and lentiviral viruses 63 64 etc It has also been found that new lines of transgenic GFP rats can be relevant for gene therapy as well as regenerative medicine 65 By using high expresser GFP transgenic rats display high expression in most tissues and many cells that have not been characterized or have been only poorly characterized in previous GFP transgenic rats GFP has been shown to be useful in cryobiology as a viability assay Correlation of viability as measured by trypan blue assays were 0 97 66 Another application is the use of GFP co transfection as internal control for transfection efficiency in mammalian cells 67 A novel possible use of GFP includes using it as a sensitive monitor of intracellular processes via an eGFP laser system made out of a human embryonic kidney cell line The first engineered living laser is made by an eGFP expressing cell inside a reflective optical cavity and hitting it with pulses of blue light At a certain pulse threshold the eGFP s optical output becomes brighter and completely uniform in color of pure green with a wavelength of 516 nm Before being emitted as laser light the light bounces back and forth within the resonator cavity and passes the cell numerous times By studying the changes in optical activity researchers may better understand cellular processes 68 69 GFP is used widely in cancer research to label and track cancer cells GFP labelled cancer cells have been used to model metastasis the process by which cancer cells spread to distant organs 70 Split GFP edit GFP can be used to analyse the colocalization of proteins This is achieved by splitting the protein into two fragments which are able to self assemble and then fusing each of these to the two proteins of interest Alone these incomplete GFP fragments are unable to fluoresce However if the two proteins of interest colocalize then the two GFP fragments assemble together to form a GFP like structure which is able to fluoresce Therefore by measuring the level of fluorescence it is possible to determine whether the two proteins of interest colocalize 71 Macro photography edit Macro scale biological processes such as the spread of virus infections can be followed using GFP labeling 72 In the past mutagenic ultra violet light UV has been used to illuminate living organisms e g see 73 to detect and photograph the GFP expression Recently a technique using non mutagenic LED lights 74 have been developed for macro photography 75 The technique uses an epifluorescence camera attachment 76 based on the same principle used in the construction of epifluorescence microscopes Transgenic pets edit nbsp Mice expressing GFP under UV light left amp right compared to normal mouse center Alba a green fluorescent rabbit was created by a French laboratory commissioned by Eduardo Kac using GFP for purposes of art and social commentary 77 The US company Yorktown Technologies markets to aquarium shops green fluorescent zebrafish GloFish that were initially developed to detect pollution in waterways NeonPets a US based company has marketed green fluorescent mice to the pet industry as NeonMice 78 Green fluorescent pigs known as Noels were bred by a group of researchers led by Wu Shinn Chih at the Department of Animal Science and Technology at National Taiwan University 79 A Japanese American Team created green fluorescent cats as proof of concept to use them potentially as model organisms for diseases particularly HIV 80 In 2009 a South Korean team from Seoul National University bred the first transgenic beagles with fibroblast cells from sea anemones The dogs give off a red fluorescent light and they are meant to allow scientists to study the genes that cause human diseases like narcolepsy and blindness 81 Art edit Julian Voss Andreae a German born artist specializing in protein sculptures 82 created sculptures based on the structure of GFP including the 1 70 m 5 6 tall Green Fluorescent Protein 2004 83 and the 1 40 m 4 7 tall Steel Jellyfish 2006 The latter sculpture is located at the place of GFP s discovery by Shimomura in 1962 the University of Washington s Friday Harbor Laboratories 84 nbsp Julian Voss Andreae s GFP based sculpture Steel Jellyfish 2006 The image shows the stainless steel sculpture at Friday Harbor Laboratories on San Juan Island Wash USA the place of GFP s discovery See also editProtein tag pGLO Yellow fluorescent protein Genetically encoded voltage indicatorReferences edit Ormo M Cubitt AB Kallio K Gross LA Tsien RY Remington SJ September 1996 Crystal structure of the Aequorea victoria green fluorescent protein Science 273 5280 1392 5 Bibcode 1996Sci 273 1392O doi 10 1126 science 273 5280 1392 PMID 8703075 S2CID 43030290 Prendergast FG Mann KG Aug 1978 Chemical and physical properties of aequorin and the green fluorescent protein isolated from Aequorea forskalea Biochemistry 17 17 3448 53 doi 10 1021 bi00610a004 PMID 28749 a b c d Tsien RY 1998 The green fluorescent protein PDF Annual Review of Biochemistry 67 509 44 doi 10 1146 annurev biochem 67 1 509 PMID 9759496 Salih A 2019 Fluorescent Proteins In Cox G ed Fundamentals of Fluorescence Imaging Boca Raton Jenny Stanford Publishing p 122 doi 10 1201 9781351129404 ISBN 9781351129404 S2CID 213688192 Stepanenko OV Verkhusha VV Kuznetsova IM Uversky VN Turoverov KK Aug 2008 Fluorescent proteins as biomarkers and biosensors throwing color lights on molecular and cellular processes Current Protein amp Peptide Science 9 4 338 69 doi 10 2174 138920308785132668 PMC 2904242 PMID 18691124 Phillips GJ Oct 2001 Green fluorescent protein a bright idea for the study of bacterial protein localization FEMS Microbiology Letters 204 1 9 18 doi 10 1016 S0378 1097 01 00358 5 PMID 11682170 Uckert W Pedersen L Gunzburg W 2000 Green fluorescent protein retroviral vector generation of high titer producer cells and virus supernatant Gene Therapy of Cancer Methods in Molecular Medicine Vol 35 pp 275 85 doi 10 1385 1 59259 086 1 275 ISBN 1 59259 086 1 PMID 21390811 Goodman SR ed 2008 Chapter 1 Tools of the Cell Biologist Green Fluorescent Protein Medical Cell Biology 3rd ed Amsterdam Elsevier Academic Press pp 14 25 doi 10 1016 B978 0 12 370458 0 50006 2 ISBN 978 0 12 370458 0 S2CID 90224559 Shimomura O Johnson FH Saiga Y Jun 1962 Extraction purification and properties of aequorin a bioluminescent protein from the luminous hydromedusan Aequorea Journal of Cellular and Comparative Physiology 59 3 223 39 doi 10 1002 jcp 1030590302 PMID 13911999 Morise H Shimomura O Johnson FH Winant J Jun 1974 Intermolecular energy transfer in the bioluminescent system of Aequorea Biochemistry 13 12 2656 62 doi 10 1021 bi00709a028 PMID 4151620 Prasher DC Eckenrode VK Ward WW Prendergast FG Cormier MJ Feb 1992 Primary structure of the Aequorea victoria green fluorescent protein Gene 111 2 229 33 doi 10 1016 0378 1119 92 90691 H PMID 1347277 Chalfie M Tu Y Euskirchen G Ward WW Prasher DC Feb 1994 Green fluorescent protein as a marker for gene expression Science 263 5148 802 5 Bibcode 1994Sci 263 802C doi 10 1126 science 8303295 PMID 8303295 S2CID 9043327 Inouye S Tsuji FI Mar 1994 Aequorea green fluorescent protein Expression of the gene and fluorescence characteristics of the recombinant protein FEBS Letters 341 2 3 277 80 doi 10 1016 0014 5793 94 80472 9 PMID 8137953 a b c Ormo M Cubitt AB Kallio K Gross LA Tsien RY Remington SJ Sep 1996 Crystal structure of the Aequorea victoria green fluorescent protein Science 273 5280 1392 5 Bibcode 1996Sci 273 1392O doi 10 1126 science 273 5280 1392 PMID 8703075 S2CID 43030290 a b c Yang F Moss LG Phillips GN Oct 1996 The molecular structure of green fluorescent protein PDF Nature Biotechnology 14 10 1246 51 doi 10 1038 nbt1096 1246 hdl 1911 19233 PMID 9631087 S2CID 34713931 a b c Brejc K Sixma TK Kitts PA Kain SR Tsien RY Ormo M Remington SJ March 1997 Structural basis for dual excitation and photoisomerization of the Aequorea victoria green fluorescent protein Proceedings of the National Academy of Sciences of the United States of America 94 6 2306 2311 Bibcode 1997PNAS 94 2306B doi 10 1073 pnas 94 6 2306 PMC 20083 PMID 9122190 Shaner NC Steinbach PA Tsien RY Dec 2005 A guide to choosing fluorescent proteins PDF Nature Methods 2 12 905 9 doi 10 1038 nmeth819 PMID 16299475 S2CID 10024284 Wilhelmsson M Tor Y 2016 Fluorescent Analogs of Biomolecular Building Blocks Design and Applications New Jersey Wiley ISBN 978 1 118 17586 6 Heim R Cubitt AB Tsien RY Feb 1995 Improved green fluorescence PDF Nature 373 6516 663 4 Bibcode 1995Natur 373 663H doi 10 1038 373663b0 PMID 7854443 S2CID 40179694 US patent 6172188 Thastrup O Tullin S Kongsbak Poulsen L Bjorn S Fluorescent Proteins published 2001 01 09 Cormack BP Valdivia RH Falkow S 1996 FACS optimized mutants of the green fluorescent protein GFP Gene 173 1 Spec No 33 38 doi 10 1016 0378 1119 95 00685 0 PMID 8707053 McRae SR Brown CL Bushell GR May 2005 Rapid purification of EGFP EYFP and ECFP with high yield and purity Protein Expression and Purification 41 1 121 127 doi 10 1016 j pep 2004 12 030 PMID 15802229 Pedelacq JD Cabantous S Tran T Terwilliger TC Waldo GS Jan 2006 Engineering and characterization of a superfolder green fluorescent protein Nature Biotechnology 24 1 79 88 doi 10 1038 nbt1172 PMID 16369541 S2CID 2966399 Barondeau DP Kassmann CJ Tainer JA Getzoff ED Apr 2002 Structural chemistry of a green fluorescent protein Zn biosensor Journal of the American Chemical Society 124 14 3522 3524 doi 10 1021 ja0176954 PMID 11929238 Lelimousin M Noirclerc Savoye M Lazareno Saez C Paetzold B Le Vot S Chazal R Macheboeuf P Field MJ Bourgeois D Royant A Oct 2009 Intrinsic dynamics in ECFP and Cerulean control fluorescence quantum yield Biochemistry 48 42 10038 10046 doi 10 1021 bi901093w PMID 19754158 Goedhart J von Stetten D Noirclerc Savoye M Lelimousin M Joosen L Hink MA van Weeren L Gadella TW Royant A 2012 Structure guided evolution of cyan fluorescent proteins towards a quantum yield of 93 Nature Communications 3 751 Bibcode 2012NatCo 3 751G doi 10 1038 ncomms1738 PMC 3316892 PMID 22434194 Miesenbock G De Angelis DA Rothman JE Jul 1998 Visualizing secretion and synaptic transmission with pH sensitive green fluorescent proteins Nature 394 6689 192 5 Bibcode 1998Natur 394 192M doi 10 1038 28190 PMID 9671304 S2CID 4320849 Hanson GT Aggeler R Oglesbee D Cannon M Capaldi RA Tsien RY Remington SJ Mar 2004 Investigating mitochondrial redox potential with redox sensitive green fluorescent protein indicators The Journal of Biological Chemistry 279 13 13044 53 doi 10 1074 jbc M312846200 PMID 14722062 Zacharias DA Violin JD Newton AC Tsien RY May 2002 Partitioning of lipid modified monomeric GFPs into membrane microdomains of live cells Science 296 5569 913 16 Bibcode 2002Sci 296 913Z doi 10 1126 science 1068539 PMID 11988576 S2CID 14957077 Yue JX Holland ND Holland LZ Deheyn DD June 2016 The evolution of genes encoding for green fluorescent proteins insights from cephalochordates amphioxus Scientific Reports 6 1 28350 Bibcode 2016NatSR 628350Y doi 10 1038 srep28350 PMC 4911609 PMID 27311567 Hunt ME Scherrer MP Ferrari FD Matz MV July 2010 Very bright green fluorescent proteins from the Pontellid copepod Pontella mimocerami PLOS ONE 5 7 e11517 Bibcode 2010PLoSO 511517H doi 10 1371 journal pone 0011517 PMC 2904364 PMID 20644720 eGFP FPbase Chudakov DM Belousov VV Zaraisky AG Novoselov VV Staroverov DB Zorov DB Lukyanov S Lukyanov KA February 2003 Kindling fluorescent proteins for precise in vivo photolabeling Nature Biotechnology 21 2 191 4 doi 10 1038 nbt778 PMID 12524551 S2CID 52887792 Wiens MD Shen Y Li X Salem MA Smisdom N Zhang W Brown A Campbell RE December 2016 A Tandem Green Red Heterodimeric Fluorescent Protein with High FRET Efficiency ChemBioChem 17 24 2361 2367 doi 10 1002 cbic 201600492 PMID 27781394 S2CID 4301322 Drepper T Eggert T Circolone F Heck A Krauss U Guterl JK et al April 2007 Reporter proteins for in vivo fluorescence without oxygen Nature Biotechnology 25 4 443 445 doi 10 1038 nbt1293 PMID 17351616 S2CID 7335755 a b Rodriguez EA Tran GN Gross LA Crisp JL Shu X Lin JY Tsien RY September 2016 A far red fluorescent protein evolved from a cyanobacterial phycobiliprotein Nature Methods 13 9 763 9 doi 10 1038 nmeth 3935 PMC 5007177 PMID 27479328 Mattson S Tran GN Rodriguez EA 2023 Directed Evolution of Fluorescent Proteins in Bacteria In Sharma M ed Fluorescent Proteins Methods in Molecular Biology Vol 2564 New York NY Springer US pp 75 97 doi 10 1007 978 1 0716 2667 2 4 ISBN 978 1 0716 2666 5 PMID 36107338 Tsien RY 1998 01 01 The green fluorescent protein Annual Review of Biochemistry 67 1 509 44 doi 10 1146 annurev biochem 67 1 509 PMID 9759496 S2CID 8138960 Maiti Atanu Buffalo Cosmo Z Saurabh Saumya Montecinos Franjola Felipe Hachey Justin S Conlon William J Tran Geraldine N Hassan Bakar Walters Kylie J Drobizhev Mikhail Moerner W E Ghosh Partho Matsuo Hiroshi Tsien Roger Y Lin John Y 2023 07 12 Structural and photophysical characterization of the small ultra red fluorescent protein Nature Communications 14 1 4155 Bibcode 2023NatCo 14 4155M doi 10 1038 s41467 023 39776 9 ISSN 2041 1723 PMC 10338489 PMID 37438348 Rodriguez EA Campbell RE Lin JY Lin MZ Miyawaki A Palmer AE Shu X Zhang J Tsien RY February 2017 The Growing and Glowing Toolbox of Fluorescent and Photoactive Proteins Trends in Biochemical Sciences 42 2 111 129 doi 10 1016 j tibs 2016 09 010 PMC 5272834 PMID 27814948 Montecinos Franjola F Lin JY Rodriguez EA 2020 11 16 Fluorescent proteins for in vivo imaging where s the biliverdin Biochemical Society Transactions 48 6 2657 2667 doi 10 1042 BST20200444 PMID 33196077 S2CID 226971864 Bokman SH Ward WW 1982 Reversible denaturation of Aequorea green fluorescent protein physical separation and characterization of the renatured protein Biochemistry 21 19 4535 4540 doi 10 1021 bi00262a003 PMID 6128025 Pouwels LJ Zhang L Chan NH Dorrestein PC Wachter RM Sep 2008 Kinetic isotope effect studies on the de novo rate of chromophore formation in fast and slow maturing GFP variants Biochemistry 47 38 10111 22 doi 10 1021 bi8007164 PMC 2643082 PMID 18759496 Chudakov DM Matz MV Lukyanov S Lukyanov KA Jul 2010 Fluorescent proteins and their applications in imaging living cells and tissues Physiological Reviews 90 3 1103 63 doi 10 1152 physrev 00038 2009 PMID 20664080 S2CID 10767597 Stepanenko OV Verkhusha VV Shavlovsky MM Kuznetsova IM Uversky VN Turoverov KK November 2008 Understanding the role of Arg96 in structure and stability of green fluorescent protein Proteins 73 3 539 551 doi 10 1002 prot 22089 PMC 2908022 PMID 18470931 a b Rosenow MA Huffman HA Phail ME Wachter RM April 2004 The crystal structure of the Y66L variant of green fluorescent protein supports a cyclization oxidation dehydration mechanism for chromophore maturation Biochemistry 43 15 4464 4472 doi 10 1021 bi0361315 PMID 15078092 Ma Y Yu JG Sun Q Li Z Smith SC 2015 07 01 The mechanism of dehydration in chromophore maturation of wild type green fluorescent protein A theoretical study Chemical Physics Letters 631 632 42 46 Bibcode 2015CPL 631 42M doi 10 1016 j cplett 2015 04 061 ISSN 0009 2614 Jugder BE Welch J Braidy N Marquis CP 2016 07 26 Construction and use of a Cupriavidus necator H16 soluble hydrogenase promoter PSH fusion to gfp green fluorescent protein PeerJ 4 e2269 doi 10 7717 peerj 2269 PMC 4974937 PMID 27547572 Arun KH Kaul CL Ramarao P 2005 Green fluorescent proteins in receptor research an emerging tool for drug discovery Journal of Pharmacological and Toxicological Methods 51 1 1 23 doi 10 1016 j vascn 2004 07 006 PMID 15596111 Song YH Kim CS Seo JH April 2016 Noninvasive monitoring of environmental toxicity through green fluorescent protein expressing Escherichia coli Korean Journal of Chemical Engineering 33 4 1331 6 doi 10 1007 s11814 015 0253 1 S2CID 62828580 Pan Y Leifert A Graf M Schiefer F Thoroe Boveleth S Broda J Halloran MC Hollert H Laaf D Simon U Jahnen Dechent W March 2013 High sensitivity real time analysis of nanoparticle toxicity in green fluorescent protein expressing zebrafish Small Weinheim an Der Bergstrasse Germany 9 6 863 9 doi 10 1002 smll 201201173 PMID 23143852 Chalfie M Jun 2009 GFP Lighting up life Proceedings of the National Academy of Sciences of the United States of America 106 25 10073 10080 Bibcode 2009PNAS 10610073C doi 10 1073 pnas 0904061106 PMC 2700921 PMID 19553219 Yuste R Dec 2005 Fluorescence microscopy today Nature Methods 2 12 902 4 doi 10 1038 nmeth1205 902 PMID 16299474 S2CID 205418407 Terskikh A Fradkov A Ermakova G Zaraisky A Tan P Kajava AV et al November 2000 Fluorescent timer protein that changes color with time Science 290 5496 1585 8 Bibcode 2000Sci 290 1585T doi 10 1126 science 290 5496 1585 PMID 11090358 Patterson GH Lippincott Schwartz J September 2002 A photoactivatable GFP for selective photolabeling of proteins and cells Science 297 5588 1873 7 Bibcode 2002Sci 297 1873P doi 10 1126 science 1074952 PMID 12228718 S2CID 45058411 Lin W Mehta S Zhang J October 2019 Genetically encoded fluorescent biosensors illuminate kinase signaling in cancer The Journal of Biological Chemistry 294 40 14814 14822 doi 10 1074 jbc REV119 006177 PMC 6779441 PMID 31434714 Komorowski M Finkenstadt B Rand D Jun 2010 Using a single fluorescent reporter gene to infer half life of extrinsic noise and other parameters of gene expression Biophysical Journal 98 12 2759 2769 Bibcode 2010BpJ 98 2759K doi 10 1016 j bpj 2010 03 032 PMC 2884236 PMID 20550887 Gunkel M Erdel F Rippe K Lemmer P Kaufmann R Hormann C Amberger R Cremer C Jun 2009 Dual color localization microscopy of cellular nanostructures Biotechnology Journal 4 6 927 38 doi 10 1002 biot 200900005 PMID 19548231 S2CID 18162278 Chudakov DM Lukyanov S Lukyanov KA Dec 2005 Fluorescent proteins as a toolkit for in vivo imaging Trends in Biotechnology 23 12 605 13 doi 10 1016 j tibtech 2005 10 005 PMID 16269193 Livet J Weissman TA Kang H Draft RW Lu J Bennis RA Sanes JR Lichtman JW Nov 2007 Transgenic strategies for combinatorial expression of fluorescent proteins in the nervous system Nature 450 7166 56 62 Bibcode 2007Natur 450 56L doi 10 1038 nature06293 PMID 17972876 S2CID 4402093 Baker BJ Mutoh H Dimitrov D Akemann W Perron A Iwamoto Y Jin L Cohen LB Isacoff EY Pieribone VA Hughes T Knopfel T Aug 2008 Genetically encoded fluorescent sensors of membrane potential Brain Cell Biology 36 1 4 53 67 doi 10 1007 s11068 008 9026 7 PMC 2775812 PMID 18679801 Adesnik H Nicoll RA England PM Dec 2005 Photoinactivation of native AMPA receptors reveals their real time trafficking Neuron 48 6 977 85 doi 10 1016 j neuron 2005 11 030 PMID 16364901 Lakadamyali M Rust MJ Babcock HP Zhuang X Aug 2003 Visualizing infection of individual influenza viruses Proceedings of the National Academy of Sciences of the United States of America 100 16 9280 5 Bibcode 2003PNAS 100 9280L doi 10 1073 pnas 0832269100 PMC 170909 PMID 12883000 Joo KI Wang P Oct 2008 Visualization of targeted transduction by engineered lentiviral vectors Gene Therapy 15 20 1384 96 doi 10 1038 gt 2008 87 PMC 2575058 PMID 18480844 Remy S Tesson L Usal C Menoret S Bonnamain V Nerriere Daguin V Rossignol J Boyer C Nguyen TH Naveilhan P Lescaudron L Anegon I Oct 2010 New lines of GFP transgenic rats relevant for regenerative medicine and gene therapy Transgenic Research 19 5 745 63 doi 10 1007 s11248 009 9352 2 PMID 20094912 S2CID 42499768 Elliott G McGrath J Crockett Torabi E Jun 2000 Green fluorescent protein A novel viability assay for cryobiological applications Cryobiology 40 4 360 369 doi 10 1006 cryo 2000 2258 PMID 10924267 Fakhrudin N Ladurner A Atanasov AG Heiss EH Baumgartner L Markt P Schuster D Ellmerer EP Wolber G Rollinger JM Stuppner H Dirsch VM Apr 2010 Computer aided discovery validation and mechanistic characterization of novel neolignan activators of peroxisome proliferator activated receptor gamma Molecular Pharmacology 77 4 559 66 doi 10 1124 mol 109 062141 PMC 3523390 PMID 20064974 Gather MC Yun SH 2011 Single cell biological lasers Nature Photonics 5 7 406 410 Bibcode 2011NaPho 5 406G doi 10 1038 nphoton 2011 99 S2CID 54971962 Matson J 2011 Green Fluorescent Protein Makes for Living Lasers Scientific American Retrieved 2011 06 13 Kouros Mehr H Bechis SK Slorach EM Littlepage LE Egeblad M Ewald AJ Pai SY Ho IC Werb Z Feb 2008 GATA 3 links tumor differentiation and dissemination in a luminal breast cancer model Cancer Cell 13 2 141 52 doi 10 1016 j ccr 2008 01 011 PMC 2262951 PMID 18242514 Cabantous S Terwilliger TC Waldo GS January 2005 Protein tagging and detection with engineered self assembling fragments of green fluorescent protein Nature Biotechnology 23 1 102 7 doi 10 1038 nbt1044 PMID 15580262 S2CID 25833063 Rodman MK Yadav NS Artus NN 2002 09 01 Progression of geminivirus induced transgene silencing is associated with transgene methylation New Phytologist 155 3 461 468 doi 10 1046 j 1469 8137 2002 00467 x PMID 33873315 Zhu YJ Agbayani R Moore PH Apr 2004 Green fluorescent protein as a visual selection marker for papaya Carica papaya L transformation Plant Cell Reports 22 9 660 7 doi 10 1007 s00299 004 0755 5 PMID 14749892 S2CID 23198182 Niwa Y Hirano T Yoshimoto K Shimizu M Kobayashi H 1999 Non invasive quantitative detection and applications of non toxic S65T type green fluorescent protein in living plants The Plant Journal 18 4 455 63 doi 10 1046 j 1365 313X 1999 00464 x PMID 10406127 S2CID 292648 Baker SS Vidican CB Cameron DS Greib HG Jarocki CC Setaputri AW Spicuzza CH Burr AA Waqas MA Tolbert DA 2012 01 01 An epifluorescent attachment improves whole plant digital photography of Arabidopsis thaliana expressing red shifted green fluorescent protein AoB Plants 2012 pls003 doi 10 1093 aobpla pls003 PMC 3296078 PMID 22479674 PlantEdDL Using SRL digital cameras in quantitative investigations of plants expressing green fluorescent protein GFP planted botany org Retrieved 2016 03 23 Kac E GFP Bunny Glow In The Dark NeonMice Archived from the original on February 14 2009 Retrieved August 30 2016 Scientists in Taiwan breed fluorescent green pigs Wongsrikeao P Saenz D Rinkoski T Otoi T Poeschla E 2011 Antiviral restriction factor transgenesis in the domestic cat Nature Methods 8 10 853 9 doi 10 1038 nmeth 1703 PMC 4006694 PMID 21909101 Fluorescent puppy is world s first transgenic dog Voss Andreae J 2005 Protein Sculptures Life s Building Blocks Inspire Art Leonardo 38 41 45 doi 10 1162 leon 2005 38 1 41 S2CID 57558522 Pawlak A 2005 Inspirierende Proteine Physik Journal 4 12 Julian Voss Andreae Sculpture Retrieved 2007 06 14 Further reading editPieribone V Gruber D 2006 Aglow in the Dark The Revolutionary Science of Biofluorescence Cambridge Belknap Press ISBN 978 0 674 01921 8 OCLC 60321612 Popular science book describing history and discovery of GFP Zimmer M 2005 Glowing Genes A Revolution In Biotechnology Buffalo NY Prometheus Books ISBN 978 1 59102 253 4 OCLC 56614624 External links edit nbsp Wikimedia Commons has media related to Green fluorescent proteins A comprehensive article on fluorescent proteins at Scholarpedia Brief summary of landmark GFP papers Interactive Java applet demonstrating the chemistry behind the formation of the GFP chromophore Video of 2008 Nobel Prize lecture of Roger Tsien on fluorescent proteins Excitation and emission spectra for various fluorescent proteins Green Fluorescent Protein Chem Soc Rev themed issue dedicated to the 2008 Nobel Prize winners in Chemistry Professors Osamu Shimomura Martin Chalfie and Roger Y Tsien Molecule of the Month June 2003 an illustrated overview of GFP by David Goodsell Molecule of the Month June 2014 an illustrated overview of GFP like variants by David Goodsell Green Fluorescent Protein on FPbase a fluorescent protein database Overview of all the structural information available in the PDB for UniProt P42212 Green fluorescent protein at the PDBe KB Retrieved from https en wikipedia org w index php title Green fluorescent protein amp oldid 1190765697, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.