WO1999036516A2 - Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote - Google Patents

Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote Download PDF

Info

Publication number
WO1999036516A2
WO1999036516A2 PCT/US1999/001164 US9901164W WO9936516A2 WO 1999036516 A2 WO1999036516 A2 WO 1999036516A2 US 9901164 W US9901164 W US 9901164W WO 9936516 A2 WO9936516 A2 WO 9936516A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
plant
viral
ofthe
sequence
Prior art date
Application number
PCT/US1999/001164
Other languages
English (en)
Other versions
WO1999036516A3 (fr
Inventor
Guy Della-Cioppa
Robert L. Erwin
Wayne P. Fitzmaurice
Kathleen M. Hanley
Monto H. Kumagai
John A. Lindbo
David R. Mcgee
Hal S. Padgett
Gregory P. Pogue
Original Assignee
Biosource Technologies, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biosource Technologies, Inc. filed Critical Biosource Technologies, Inc.
Priority to KR1020007007808A priority Critical patent/KR20010040337A/ko
Priority to JP2000540219A priority patent/JP2002508957A/ja
Priority to EP99903208A priority patent/EP1045899A2/fr
Priority to AU23286/99A priority patent/AU761367B2/en
Priority to CA002318662A priority patent/CA2318662A1/fr
Publication of WO1999036516A2 publication Critical patent/WO1999036516A2/fr
Publication of WO1999036516A3 publication Critical patent/WO1999036516A3/fr
Priority to US10/236,508 priority patent/US20030167512A1/en
Priority to AU2003220702A priority patent/AU2003220702A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/1034Isolating an individual clone by screening libraries
    • C12N15/1079Screening libraries by altering the phenotype or phenotypic trait of the host
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids

Definitions

  • the present invention relates generally to the field of molecular biology and plant genetics. Specifically, the present invention relates to a method for determining the function of nucleotide sequences and genes by transfecting the same into a host.
  • Arabidopsis thaliana may be an attractive target for gene function discovery because a very large set of ESTs have already been produced in this organism, and these sequences tag more than 50% of the expected Arabidopsis genes.
  • sequence information so generated is enormous if gene function can be determined. It may become possible to engineer commercial seeds for agricultural use to convey any number of desirable traits to food and fiber crops and thereby increase agricultural production and the world food supply. Research and development of commercial seeds has so far focused primarily on traditional plant breeding, however there has been increased interest in biotechnology as it relates to plant characteristics. Knowledge ofthe genomes involved and the function of genes contained therein for both monocotyledonous and dicotyledonous plants is essential to realizing positive effects from such technology.
  • gene profiling in cotton can lead to an understanding ofthe types of genes being expressed primarily in fiber cells.
  • the genes or promoters derived from these genes may be important in genetic engineering of cotton fiber for increased strength or for "built-in" fiber color.
  • gene profiling coupled to physiological trait analysis can lead to the identification of predictive markers that will be increasingly important in marker assisted breeding programs. Mining the DNA sequence of a particular crop for genes important for yield, quality, health, appearance, color, taste, etc., are applications of obvious importance for crop improvement.
  • the recombinant plant viral nucleic acids and recombinant viruses such as those demonstrated by Donson et al. which have been demonstrated to infect plant cells and express the foreign genetic material systemically are generally characterized as comprising a native plant viral subgenomic promoter, at least one non-native plant viral subgenomic promoter, a plant viral coat protein coding sequence, and at least one non-native nucleic acid sequence.
  • the value of using such plant viral nucleic acids to effect systemic expression of non-native nucleic acids in a plant host is significant. This tool, if coupled with a rational design for elucidating the function ofthe non-native nucleic acids, would make significant strides in understanding the large amount of sequence information produced by sequencing efforts.
  • the present invention is directed to a method of determining the function of nucleic acid sequences including genes and the proteins they encode in host organisms such as bacteria, yeast, plants, or animals, by transfecting the nucleic acid sequences into the organisms in a manner so as to effect localized or systemic expression ofthe nucleic acid sequences.
  • the present inventors have determined methods for determining the function of nucleic acid sequences and the proteins they encode by tr.ansfecting organisms with nucleic acids of interest thereby providing a more rapid means for elucidating the function of these nucleic acids including genes and subsequently utilizing the rapidly expanding information in the field of genomics.
  • a nucleic acid is introduced into a plant host wherein the plant host may be a monocotyledonous or dicotyledonous plant, plant tissue or plant cell.
  • the nucleic acid may be introduced by way of a plant viral nucleic acid.
  • plant viral nucleic acids are stable for the maintenance and transcription or expression of non-native nucleic acid sequences and are capable of locally or systemically transcribing or expressing such sequences in the plant host.
  • Especially preferred recombinant plant viral nucleic acids useful in the methods ofthe present invention comprise a native plant viral subgenomic promoter, a plant viral coat protein coding sequence, and at least one non-native nucleic acid sequence.
  • Some viral vectors used in accordance with the present invention may be encapsidated by the coat proteins encoded by the recombinant plant virus.
  • the recombinant plant viral nucleic acid or recombinant plant virus is used to infect appropriate hosts such as plants.
  • the recombinant plant viral nucleic acid is capable of replication in the host, localized or systemic spread in the host, and transcription or expression ofthe non-native nucleic acid in the host to produce the desired product.
  • Such products may be for example, useful polypeptides or proteins including enzymes, complex biomolecules, ribozymes, or polypeptides or protein products resulting from positive-sense or anti-sense RNA expression.
  • the nucleic acid of interest may be expressed with the genomic DNA or RNA ofthe viral vectors and hence be under the control of a genomic promoter.
  • Some other viral vectors used in accordance with the present invention comprise recombinant animal viruses or portions thereof.
  • animal viral vectors are useful to infect appropriate hosts such as animals.
  • the recombinant animal viral nucleic acid is capable if replication in the host, systemic or localized spread in the host, and transcription or expression ofthe non-native nucleic acid in the host to produce the desired product.
  • the present method uses a viral expression vector encoding for at least one protein non-native to the vector that is released from at least one polyprotein expressed by said vector by proteolytic processing.
  • recombinant plant viruses are used which encode for the expression of a fusion between a plant viral coat protein and the amino acid product ofthe nucleic acid of interest.
  • a nucleic acid sequence of interest including a gene may be placed within any suitable vector construct such as a virus for infecting the host organism. That is, the present method may be practiced without concern for the position ofthe nucleic acid sequence of interest within the vector used to infect the host organism.
  • the invention is not intended to be limited to any particular viral constructs but specifically contemplates using all operable constructs. Those skilled in the art will understand that these embodiments are representative only of many constructs which may be useful to produce localized or systemic expression of nucleic acids in host organisms such as plants. All such constructs .are contemplated and intended to be within the scope ofthe present invention.
  • the function of a nucleic acid may be determined by complementation analysis. That is, the function ofthe nucleic acid of interest may be determined by observing the endogenous gene or genes whose function is replaced or augmented by introducing the nucleic acid of interest. A discussion of such phenomenon is provided by Napoli et al, The Plant Cell 2:279-289 (1990).
  • the function of a nucleic acid may be determined by analyzing the biochemical alterations in the accumulation of substrates or products from enzymatic reactions according to any one ofthe means known by those skilled in the art.
  • the function of a nucleic acid may be determined by observing phenotypic changes in the host by methods including morphological, macroscopic or microscopic analysis.
  • the function of a nucleic acid may be determined by observing any changes in biochemical pathways which may be modified in the host organism as a result of expression ofthe nucleic acid.
  • the function of a nucleic acid may be determined utilizing techniques known by those skilled in the art to observe inhibition of endogenous gene expression in the cytoplasm of cells as a result of expression ofthe nucleic acid.
  • the function of a nucleic acid may be determined utilizing techniques known by those skilled in the art to observe changes in the RNA or protein profile as a result of expression ofthe nucleic acid.
  • the function of a nucleic acid may be determined by selection of organisms such as plants or human cells and tissues capable of growing or maintaining viability in the presence of noxious or toxic substances, such as, for example herbicides and pharmaceutical ingredients.
  • a second aspect ofthe present invention is a method of silencing endogenous genes in a host by introducing nucleic acids into the host by way of a viral nucleic acid such as a plant or animal viral nucleic acid suitable to produce expression of a nucleic acid in a transfected host.
  • the host is a plant, but those skilled in the art will understand that other hosts such as bacteria, yeast and animals including humans may also be utilized.
  • This method utilizes the principle of post-transcription gene silencing ofthe endogenous host gene homo log. Since the replication mechanism ofthe transfected non-native nucleic acid produces both sense and antisense RNA sequences, the orientation of the non-native nucleic acid insert is not crucial to providing gene silencing. Particularly, this aspect ofthe invention is especially useful for silencing a multigene family as is frequently found in plants. The prior art has not demonstrated an effective means for silencing a multigene family in plants.
  • a third aspect ofthe present invention is a method for selecting desired functions of RNAs .and proteins by the use of virus vectors to express libraries of nucleic acid sequence variants.
  • Libraries of sequence variants may be generated by means of in vitro mutagenenisis and/or recombination. Rapid in vitro evolution can be used to improve virus-specific or protein-specific functions.
  • plant RNA virus expression vectors may be used as tools to bear libraries containing variants of nucleic acid, genes from virus, plant or other sources, and to be applied to plants or plant cells such that the desired altered effects in the RNA or protein products can be determined, selected and improved.
  • nucleic acid shuffling techniques may be employed to construct shuffled gene libraries.
  • Random, semi-random or known sequences of virus origin may also be inserted in virus expression vectors between native virus sequences and foreign gene sequences, to increase the genetic stability of foreign genes in expression vectors as well as the translation ofthe foreign gene and the stability ofthe mRNA encoding the foreign gene in vivo.
  • the desired function of RNA and protein may include the promoter activities, replication properties, translational efficiencies, movement properties (local and systemic), signaling pathway, or virus host range, among others.
  • the desired function alteration can be identified by assaying infected plants and the nature of mutation can be determined by analysis of sequence variants in the virus vector.
  • Methods to increase the representation of gene sequences in virus expression libraries may also be achieved by bypassing the genetic bottleneck of propagation in E. coli.
  • cell-free methods may be used to clone sequence libraries or individual arrayed sequences into virus expression vectors and reconstruct an infectious virus, such that the final ligation product can be transcribed and the resulting RNA can be used for plant or plant cell inoculation/infection with the output being gene function discovery or protein production.
  • RNA viruses or RNA virus vectors as populations or individuals in parallel to identify individuals with novel and augmented virus-encoded functions in replication and virus movement, foreign gene sequence retention in vectors and proper folding, activity and expression of protein products, novel gene expression, effects on host metabolism, and resistance or susceptibility of plants to exogenous agents.
  • Variation in the sequence of a native virus gene(s) or heterologous nucleotide sequence(s) may be introduced into an RNA virus or an RNA virus expression vector by many methods as a means to screen a population of variants in batch or individuals in parallel for novel properties exhibited by the virus itself or conferred on the host plant or cell by the virus vector.
  • Variant populations can be transfected as populations or individual clones into "host”: 1) protoplasts; 2) whole plants; or 3) inoculated leaves of whole plants and screened for various traits including protein expression (increase or decrease), RNA expression (increase or decrease), secondary metabolites or other host property gained or loss as a result ofthe virus infection.
  • a virus vector which simultaneously expressed the green fluorescent protein (GFP) or other selectable marker gene and the variant sequence, is used to screen quantitatively for levels of resistance or sensitivity to the agent in question conferred upon the host by the variant sequence expressed from the viral vector.
  • GFP green fluorescent protein
  • transcription factors regulating the expression of defense proteins such as systemin peptides, or protease inhibitors could be identified by transfecting hosts with virus libraries and the expression of systemin or protease inhibitors or their RNAs be directly assayed.
  • the promoters responsible for expressing these genes could be genetically fused to the green fluorescent protein .and introduced into hosts as transient expression constructs or into stable transformed host cells/tissues. The resulting cells would be transfected with viral vector libraries.
  • Hosts now could be screened rapidly by following relative GFP expression following vector fransfection.
  • coupling the transfecting of hosts with virus libraries with the treatment of plants with methyl jasmonate could identify sequences that reverse or enhance the gene induction events induced by this metabolite.
  • This approach could be applied to other factors involved in promotion of higher biomass in plants such as Leafy or DET2.
  • the expression of these factors could be directly assayed or via promoters genetically fused to GFP. This technique will enable screening of populations or individuals followed by rescue ofthe sequence from virus vectors conferring desired trait by RT-PCR and re-screening of particular variant sequences in secondary screens.
  • a fourth aspect ofthe present invention is a method for inhibiting an endogenous protease of a plant host comprising the step of treating the plant host with a compound which induces the production of an endogenous inhibitor of said protease.
  • jasmonic acid may be used to treat the plant host to induce the production of an endogenous inhibitor of an endogenous protease.
  • the treatment ofthe plant host with a compound results an increased representation of an exogenous nucleic acid or the protein product thereof.
  • transgenic hosts expressing protease inhibitors may be used to decrease the degradation of proteins expressed by virus expression vectors.
  • jasmonic acid may be used to treat plants infected with virus expression vectors to decrease degradation of proteins expressed by virus expression vectors.
  • a fifth aspect ofthe present invention are genes and fragments thereof, nucleotide sequences, and gene products obtained by way ofthe method ofthe present invention.
  • the present invention features expressing selected nucleotide sequences in a host organism.
  • gene products of such nucleotide sequences may be isolated using techniques known to those skilled in the art. Such gene products may exhibit biological activity as pharmaceuticals, herbicides, and other similar functions.
  • FIG. 1 depicts the vector TTOl/PSY +.
  • FIG. 2 represents the vector TTO1A/PDS+.
  • FIG. 3 represents the vector TT01A/Ca CCS+.
  • FIG. 4 represents the vector TTU51 CTP CrtB.
  • FIG. 5 represents the vector TTOSA1CTP CrtI 491.
  • FIG. 6 represents the Erwinia herbicola phytoene desaturase gene (plasmid pAU211).
  • FIG. 7 represents the plasmid KS+/Crt/* 491.
  • FIG. 8 represents the plasmid pBS736.
  • FIG. 9 represents the plasmid pBS 712.
  • FIG. 10 represents the 72 kDa gene product ofthe genomic clone encoding alcohol oxidase ZZA1.
  • FIG. 11 represents the plasmid TTOS 1 APE ZZA1.
  • FIG. 12 represents the plasmid TTO1 A 103L.
  • FIG. 13 represents the plasmid TTU51A QSEO #3.
  • FIG. 14 represents the plasmid KS+ TVCVK #23.
  • FIG. 15 represents the plasmid pBS735.
  • FIG. 16 represents the plasmid pBS740.
  • FIG. 17 represents the plasmid pBS723.
  • FIG. 18 represents the plasmid pBS731.
  • FIG. 19 represents the plasmid pBS740 AT #120.
  • FIG. 20 represents the nucleotide sequence alignment of 740 AT #120 to human ADP-ribosylation factor (ARF3) M33384.
  • FIG. 21 represents the plasmid pBS740 AT #88.
  • FIG. 22 represents the nucleotide sequence alignment of 740 AT #88 to L33574 mRNA for rhodopsin.
  • FIG. 23 represents the nucleotide sequence alignment of 740 AT #88 to X07797 Octopus mRNA for rhodopsin.
  • FIG. 24 represents the protein sequence alignment of 740 AT #88 to an Arabidopsis est ORF ATTS2938.
  • FIG. 25 represents the protein sequence alignment of 740 AT #88 to Octopus rhodopsin P31356.
  • FIG. 26 represents amino acid sequence comparison of 740 AT #2441 to tobacco RAN-B1 GTP binding protein.
  • FIG. 27 represents nucleotide sequence comparison of 740 AT #2441 to human RAN GTP-binding protein.
  • FIG. 28 represents a schematic diagram of cell free cloning.
  • the present invention is directed to a method of determining the function of a nucleic acid sequence including a gene and a protein encoded thereby in an organism such as bacteria, fungi, yeast, animals and plants by transfecting the nucleic acid sequence into the organism.
  • the present inventors have determined methods for determining the function of nucleic acid sequences by transfecting organisms with the nucleic acids thereby providing a more rapid means for determining gene function and utilizing the rapidly expanding sequence information in the field of genomics.
  • a nucleic acid is introduced into a plant host.
  • the nucleic acid may be introduced by way of a viral nucleic acid.
  • Such recombinant viral nucleic acids are stable for the maintenance and transcription or expression of non-native nucleic acid sequences and are capable of systemically transcribing or expressing such non-native sequences in the plant host.
  • Especially preferred recombinant plant viral nucleic acids useful in the present invention comprise a native plant viral subgenomic promoter, a plant viral coat protein coding sequence, and at least one non-native nucleic acid sequence.
  • plant viral nucleic acid sequences used in the method ofthe present invention are characterized by the deletion ofthe native coat protein coding sequence and comprise a non-native plant viral coat protein coding sequence and a non-native promoter, preferably the subgenomic promoter ofthe non-native coat protein coding sequence, capable of expression in the plant host, packaging ofthe recombinant plant viral nucleic acid, and ensuring a systemic infection ofthe host by the recombinant plant viral nucleic acid.
  • the recombinant plant viral nucleic acid may contain one or more additional native or non-native subgenomic promoters.
  • Each non-native subgenomic promoter is capable of transcribing or expressing adjacent genes or nucleic acid sequences in the plant host and incapable of recombination with each other and with native subgenomic promoters.
  • One or more non-native nucleic acids may be inserted adjacent to the native plant viral subgenomic promoter or the native and non- native plant viral subgenomic promoters if more than one nucleic acid sequence is included.
  • two or more heterologous non-native subgenomic promoters may be used.
  • the non-native nucleic acid sequences may be transcribed or expressed in the host plant under the control ofthe subgenomic promoter to produce the products ofthe nucleic acids of interest.
  • plant viral nucleic acids are used in the present invention wherein the native coat protein coding sequence is placed adjacent one ofthe non-native coat protein subgenomic promoters instead of a non-native coat protein coding sequence.
  • plant viral nucleic acids are used in the present invention wherein the native coat protein gene is adjacent its subgenomic promoter and one or more non-native subgenomic promoters have been inserted into the viral nucleic acid.
  • the inserted non-native subgenomic promoters are capable of transcribing or expressing adjacent genes in a plant host and are incapable of recombination with each other and with native subgenomic promoters.
  • Non-native nucleic acid sequences may be inserted adjacent the non- native subgenomic plant viral promoters such that the sequences are transcribed or expressed in the host plant under control ofthe subgenomic promoters to produce the product ofthe non-native nucleic acid.
  • the native coat protein coding sequence may be replaced by a non-native coat protein coding sequence.
  • the viral vectors used in accordance with the present invention may be encapsidated by the coat proteins encoded by the recombinant plant virus.
  • the recombinant plant viral nucleic acid or recombinant plant virus is used to infect appropriate hosts such as plants.
  • the recombinant plant viral nucleic acid is capable of replication in the host, localized or systemic spread in the host, and transcription or expression ofthe non-native nucleic acid in the host to produce the desired product.
  • Such products may be for example, therapeutics and other useful polypeptides or proteins including enzymes, complex biomolecules, ribozymes, or polypeptides or protein products resulting from positive-sense or anti-sense RNA expression.
  • the nucleic acid of interest may be under the control of a genomic promoter and therefore be expressed with the genome of the virus.
  • the present method uses a viral expression vector encoding at least one protein non-native to the vector that is released from at least one polyprotein expressed by said vector by proteolytic processing catalyzed by at least one protease in said polyprotein wherein said vector comprises at least one promoter, DNA having a sequence which codes for at least one polyprotein from a polyprotein-producing virus, at least one restriction site flanking a 3' terminus of said DNA and a cloning vehicle.
  • Additional embodiments use a viral expression vector encoding for at least one protein non-native to the vector that is released from at least one polyprotein expressed by the vector by proteolytic processing catalyzed by at least one protease in the polyprotein wherein the vector comprises at least one promoter, DNA having a sequence which codes for at least one polyprotein from a polyprotein-producing virus, may contain at least one restriction site flanking a 3' terminus of said cDNA and a cloning vehicle.
  • Preferred embodiments include using a potyvirus as the polyprotein-producing virus, and especially preferred embodiments may use TEV (tobacco etch virus).
  • TEV tobacco etch virus
  • recombinant plant viruses which encode for the expression of a fusion between a plant viral coat protein and the amino acid product ofthe nucleic acid of interest.
  • a recombinant plant virus provides for high level expression of a nucleic acid of interest.
  • the location or locations where the viral coat protein is joined to the amino acid product ofthe nucleic acid of interest may be referred to as the fusion joint.
  • a given product of such a construct may have one or more fusion joints.
  • the fusion joint may be located at the carboxyl terminus ofthe viral coat protein or the fusion joint may be located at the amino terminus ofthe coat protein portion ofthe construct.
  • the nucleic acid of interest may be located 5', 3', upstream, downstream or within the coat protein.
  • a "leaky” start or stop codon may occur at a fusion joint which sometimes does not result in translational termination.
  • a more detailed description of some recombinant plant viruses according to this embodiment of the invention may be found in co-pending U.S. Patent Application Serial No. 08/324,003 the disclosure of which is incorporated herein by reference.
  • a nucleic acid sequence of interest or a gene may be placed within any suitable vector construct such as a virus for infecting the host organism.
  • the present method may be practiced without concern for the position ofthe nucleic acid sequence of interest within the vector used to infect the host organism.
  • the invention is not intended to be limited to any particular viral constructs but specifically contemplates using all operable constructs. Specifically, those skilled in the art may choose to transfer DNA or RNA of any size up to and including an entire genome into a host organism in order to determine the function thereof.
  • Adjacent A position in a nucleotide sequence proximate to and 5' or 3' to a defined sequence. Generally, adjacent means within 2 or 3 nucleotides ofthe site of reference.
  • Animal cell A single functional cell found within an animal organism.
  • Animal tissue refers to one or more cells grouped or organized to perform one or more functions.
  • Animal organ refers to one or more tissues morphologically arranged to perform one or more functions within .an organism.
  • Anti-Sense Inhibition A type of gene regulation based on cytoplasmic, nuclear or organelle inhibition of gene expression due to the presence in a cell of an RNA molecule complementary to at least a portion ofthe mRNA being translated. It is specifically contemplated that DNA molecules may be from either an RNA virus or mRNA from the host cells genome or from a DNA virus.
  • Cell Culture A proliferating group of cells which may be in either an undifferentiated or differentiated state, growing contiguously or non- contiguously.
  • Chimeric Sequence or Gene A nucleotide sequence derived from at least two heterologous parts.
  • the sequence may comprise DNA or RNA.
  • Coding Sequence A deoxyribonucleotide or ribonucleotide sequence which, when either transcribed and translated or simply translated, results in the formation of a cellular polypeptide or a ribonucleotide sequence which, when translated, results in the formation of a cellular polypeptide.
  • Compatible The capability of operating with other components of a system.
  • a vector or plant or animal viral nucleic acid which is compatible with a host is one which is capable of replicating in that host.
  • a coat protein which is compatible with a viral nucleotide sequence is one capable of encapsidating that viral sequence.
  • Complementation Analysis As used herein, this term refers to observing the changes produced in an organism when a nucleic acid sequence is introduced into that organism after a selected gene has been deleted or mutated so that it no longer functions fully in its normal role.
  • a complementary gene to the deleted or mutated gene can restore the genetic phenotype ofthe selected gene.
  • Constitutive expression Gene expression which features substantially constant or regularly cyclical gene transcription. Generally, genes which are constitutively expressed are substantially free of induction from an external stimulus.
  • Differentiated cell A cell which has substantially matured to perform one or more biochemical or physiological functions.
  • Dual Heterologous Subgenomic Promoter Expression System a plus stranded RNA vector having a dual heterologous subgenomic promoter expression system to increase, decrease, or change the expression of proteins, peptides or RNAs, preferably those described in U.S. Patent Nos. 5,316,931, 5,811,653, 5,589,367, and 5,866,785, the disclosure of which is incorporated herein by reference.
  • ESTs Relatively short single-pass DNA sequences obtained from one or more ends of cDNA clones and RNA derived therefrom. They may be present in either the 5' or the 3' orientation. ESTs have been shown useful for identifying particular genes.
  • Gene A discrete nucleic acid sequence responsible for producing one or more cellular products and/or performing one or more intercellular or intracellular functions.
  • Gene silencing A reduction in gene expression.
  • a viral vector expressing gene sequences from a host may induce gene silencing of homologous gene sequences.
  • Growth cycle As used herein, the term is meant to include the replication of a nucleus, an organelle, a cell, or an organism.
  • Host A cell, tissue or organism capable of replicating a nucleic acid such as a vector or plant viral nucleic acid and which is capable of being infected by a virus containing the viral vector or viral nucleic acid.
  • This term is intended to include prokaryotic and eukaryotic cells, organs, tissues or organisms, where appropriate. Bacteria, fungi, yeast, animal (cell, tissues, or organisms), and plant (cell, tissues, or organisms) are examples of a host.
  • induction refer generally to a gene and a promoter operably linked thereto which is in some manner dependent upon an external stimulus, such as a molecule, in order to actively transcribe and/or translate the gene.
  • Infection The ability of a virus to transfer its nucleic acid to a host or introduce a viral nucleic acid into a host, wherein the viral nucleic acid is replicated, viral proteins are synthesized, and new viral particles assembled.
  • transmissible and infective are used interchangeably herein.
  • the term is also meant to include the ability of a selected nucleic acid sequence to integrate into a genome, chromosome or gene of a target organism.
  • Multigene family A set of genes descended by duplication and variation from some ancestral gene. Such genes may be clustered together on the same chromosome or dispersed on different chromosomes.
  • multigene families include those which encode the histones, hemoglobins, immunoglobulins, histocompatibility antigens, actins, tubulins, keratins, collagens, heat shock proteins, salivary glue proteins, chorion proteins, cuticle proteins, yolk proteins, and phaseolins.
  • Non-Native Any RNA or DNA sequence that does not normally occur in the cell or organism in which it is placed. Examples include recombinant plant viral nucleic acids and genes or ESTs contained therein. That is, an RNA or DNA sequence may be non-native with respect to a viral nucleic acid. Such an RNA or DNA sequence would not naturally occur in the viral nucleic acid. Also, an RNA or DNA sequence may be non-native with respect to a host organism. That is, such a RNA or DNA sequence would not naturally occur in the host organism. Conversely, the term non-native does not imply that an RNA or DNA sequence must be non-native with respect to both a viral nucleic acid and a host organism concurrently. The present invention specifically contemplates placing an RNA or DNA sequence which is native to a host organism into a viral nucleic acid in which it is non-native. Nucleic acid: As used herein the term is meant to include any DNA or
  • RNA sequence from the size of one or more nucleotides up to and including a complete gene sequence.
  • the term is intended to encompass all nucleic acids whether naturally occurring in a particular cell or organism or non-naturally occurring in a particular cell or organism.
  • Nucleic acid of interest The term is used interchangeably with the term
  • nucleic acid and is intended to refer to the nucleic acid sequence whose function is to be determined. The sequence will normally be non-native to the viral vector but may be native or non-native to the host organism.
  • Organism The term organism and "host organism” as used herein is specifically intended to include animals including humans, plants, viruses, fungi, .and bacteria.
  • Phenotypic Trait An observable, measurable or detectable property resulting from the expression or suppression of a gene or genes.
  • Plant Cell The structural and physiological unit of plants, consisting of a protoplast and the cell wall.
  • Plant Organ A distinct and visibly differentiated part of a plant, such as root, stem, leaf or embryo.
  • Plant Tissue Any tissue of a plant in planta or in culture. This term is intended to include a whole plant, plant cell, plant organ, protoplast, cell culture, or any group of plant cells organized into a structural and functional unit.
  • Positive-sense inhibition A type of gene regulation based on cytoplasmic inhibition of gene expression due to the presence in a cell of an RNA molecule substantially homologous to at least a portion ofthe mRNA being translated.
  • Promoter The 5 '-flanking, non-coding sequence substantially adjacent a coding sequence which is involved in the initiation of transcription ofthe coding sequence.
  • Protoplast An isolated plant or bacterial cell without some or all of its cell wall.
  • Plant viral nucleic acid which has been modified to contain non-native nucleic acid sequences. These non-native nucleic acid sequences may be from any organism or purely synthetic, however, they may also include nucleic acid sequences naturally occurring in the organism into which the recombinant plant viral nucleic acid is to be introduced.
  • Recombinant Plant Virus A plant virus containing the recombinant plant viral nucleic acid.
  • Subgenomic Promoter A promoter of a subgenomic mRNA of a viral nucleic acid.
  • Substantial Sequence Homology Denotes nucleotide sequences that are substantially functionally equivalent to one another. Nucleotide differences between such sequences having substantial sequence homology will be de minimis in affecting function ofthe gene products or an RNA coded for by such sequence.
  • Transposon A nucleotide sequence such as a DNA or RNA sequence which is capable of transferring location or moving within a gene, a chromosome or a genome.
  • Transgenic plant A plant which contains a foreign nucleotide sequence inserted into either its nuclear genome or organellar genome.
  • RNA polymerase Production of an RNA molecule by RNA polymerase as a complementary copy of a DNA sequence or subgenomic mRNA.
  • Vector A self-replicating RNA or DNA molecule which transfers an
  • RNA or DNA segment between cells such as bacteria, yeast, plant, or animal cells.
  • Virus An infectious agent composed of a nucleic acid which may or may not be encapsidated in a protein.
  • a virus may be a mono-, di-, tri-, or multi- partite virus, as described above.
  • the present invention provides for the infection of a plant host by a recombinant plant virus containing a recombinant plant viral nucleic acid or by the recombinant plant viral nucleic acid which contains one or more non-native nucleic acid sequences which are subsequently transcribed or expressed in the infected tissues ofthe plant host.
  • the product ofthe coding sequences may be recovered from the plant, produce a phenotypic trait in the plant, effect biochemical pathways within the plant or effect endogenous gene expression within the plant.
  • the present invention has a number of advantages.
  • the instant invention allows practitioners to determine the function of a nucleic acid sequence which has been heretofore unknown.
  • chimeric genes and vectors and recombinant plant viral nucleic acids used in this invention are constructed using techniques well known in the art. Suitable techniques have been described in Sambrook et al. (2nd ed.), Cold Spring Harbor Laboratory, Cold Spring Harbor (1982, 1989); Methods in
  • An important feature ofthe present invention is the use of recombinant plant viral nucleic acids which are capable of replication, local and/or systemic spread in a compatible plant host, and which contain one or more non-native subgenomic promoters which are capable of transcribing or expressing adjacent nucleic acid sequences in the plant host.
  • the recombinant plant viral nucleic acids may be further modified to delete all or part ofthe native coat protein coding sequence and to contain a non-native coat protein coding sequence under control ofthe native or one ofthe non-native subgenomic promoters, or put the native coat protein coding sequence under the control of a non-native plant viral subgenomic promoter.
  • the recombinant plant viral nucleic acids have substantial sequence homology to plant viral nucleotide sequences.
  • a partial listing of suitable viruses is described, infra.
  • the nucleotide sequence may be or may be derived from an RNA, DNA, cDNA or a chemically synthesized RNA or DNA.
  • the first step in producing recombinant plant viral nucleic acids according to this particular embodiment for use in the present invention is to modify the nucleotide sequences ofthe plant viral nucleotide sequence by known conventional techniques such that one or more non-native subgenomic promoters are inserted into the plant viral nucleic acid without destroying the biological function ofthe plant viral nucleic acid.
  • the subgenomic promoters are capable of transcribing or expressing adjacent nucleic acid sequences in a plant host infected by the recombination plant viral nucleic acid or recombinant plant virus.
  • the native coat protein coding sequence may be deleted in some embodiments, placed under the control of a non-native subgenomic promoter in other embodiments, or retained in a further embodiment. If it is deleted or otherwise inactivated, a non- native coat protein gene is inserted under control of one ofthe non-native subgenomic promoters, or optionally under control ofthe native coat protein gene subgenomic promoter.
  • the non-native coat protein is capable of encapsidating the recombinant plant viral nucleic acid to produce a recombinant plant virus.
  • the recombinant plant viral nucleic acid contains a coat protein coding sequence, which may be native or a normative coat protein coding sequence, under control of one ofthe native or non-native subgenomic promoters.
  • the coat protein is involved in the systemic infection ofthe plant host.
  • viruses which meet this requirement, and therefore have been shown to be suitable for use according to the methods ofthe present invention, include viruses from the tobamovirus group such as Tobacco Mosaic virus (TMV), Ribgrass Mosaic Virus (RGM), Cowpea Mosaic virus (CMV), Alfalfa Mosaic virus (AMV), Cucumber Green Mottle Mosaic virus watermelon strain (CGMMV-W) and Oat Mosaic virus (OMV) and viruses from the brome mosaic virus group such as Brome Mosaic virus (BMV), broad bean mottle virus and cowpea chlorotic mottle virus.
  • TMV Tobacco Mosaic virus
  • RGM Ribgrass Mosaic Virus
  • CMV Alfalfa Mosaic virus
  • AMV Alfalfa Mosaic virus
  • CGMMV-W Cucumber Green Mottle Mosaic virus watermelon strain
  • Oat Mosaic virus Oat Mosaic virus
  • viruses from the brome mosaic virus group such as Brome Mosaic virus (BMV
  • Suitable viruses include Rice Necrosis virus (RNV), and geminiviruses such as Tomato Golden Mosaic virus (TGMV), Cassava Latent virus (CLV) and Maize Streak virus (MS V).
  • RMV Rice Necrosis virus
  • TGMV Tomato Golden Mosaic virus
  • CLV Cassava Latent virus
  • MS V Maize Streak virus
  • TOBAMOVIRUS GROUP Tobacco Mosaic virus is a member ofthe Tobamo viruses.
  • the TMV virion is a tubular filament, and comprises coat protein sub-units arranged in a single right-handed helix with the single-stranded RNA intercalated between the turns ofthe helix.
  • TMV infects tobacco as well as other plants. TMV is transmitted mechanically and may remain infective for a year or more in soil or dried leaf tissue.
  • the TMV virions may be inactivated by subjection to an environment with a pH of less than 3 or greater than 8, or by formaldehyde or iodine. Preparations of TMV may be obtained from plant tissues by (NH 4 ) 2 SO 4 precipitation, followed by differential centrifugation.
  • the TMV single-stranded RNA genome is about 6400 nucleotides long, and is capped at the 5 '-end but not polyadenylated.
  • the genomic RNA can serve as mRNA for protein of a molecular weight of about 130,000 (130K) and another produced by read-through of molecular weight about 180,000 (180K). However, it cannot function as a messenger for the synthesis of coat protein.
  • Other genes are expressed during infection by the formation of monocistronic, 3'-coterminal subgenomic mRNAs, including one (LMC) encoding the 17.5K coat protein and another (I 2 ) encoding a 3 OK protein.
  • the 3 OK protein has been detected in infected protoplasts as described in Miller, J., Virology 132:71 (1984), and it is involved in the cell-to-cell transport ofthe virus in an infected plant as described by Deom et al, Science 237:389 (1987).
  • the functions ofthe two large proteins are unknown, however, they are thought to function in RNA replication and transcription.
  • RNA molecules including double-str.anded RNAs corresponding to the genomic, I 2 and LMC RNAs, have been detected in plant tissues infected with TMV. These RNA molecules are presumably intermediates in genome replication and/or mRNA synthesis processes which appear to occur by different mechanisms.
  • TMV assembly apparently occurs in plant cell cytoplasm, although it has been suggested that some TMV assembly may occur in chloroplasts since transcripts of ctDNA have been detected in purified TMV virions. Initiation of TMV assembly occurs by interaction between ring-shaped aggregates ("discs") of coat protein (each disc consisting of two layers of 17 subunits) and a unique internal nucleation site in the RNA; a hairpin region about 900 nucleotides from the 3 '-end in the common strain of TMV. Any RNA, including subgenomic RNAs containing this site, may be packaged into virions. The discs apparently assume a helical form on interaction with the RNA, and assembly (elongation) then proceeds in both directions (but much more rapidly in the 3'- to 5'- direction from the nucleation site).
  • CGMMV-W Cucumber Green Mottle Mosaic virus watermelon strain
  • Subgroup I which includes the vulgare, OM, and tomato strain, has an origin of assembly about 800-1000 nucleotides from the 3 '-end ofthe RNA genome, and outside the coat protein cistron. Lebeurier et al, Proc. Natl. Acad. Sci. USA 74:149 (1977); and Fukuda et al, Virology 101:493 (1980).
  • Subgroup II which includes CGMMV-W and cornpea strain (Cc) has an origin of assembly about 300-500 nucleotides from the 3 '-end ofthe RNA genome and within the coat-protein cistron.
  • the coat protein cistron of CGMMV-W is located at nucleotides 176-661 from the 3 '-end.
  • the 3' noncoding region is 175 nucleotides long.
  • the origin of assembly is positioned within the coat protein cistron. Meshi et al, Virology 127:54 (1983).
  • BMV BROME MOSAIC VIRUS GROUP Brome Mosaic virus
  • BMV BROME MOSAIC VIRUS GROUP Brome Mosaic virus
  • Each member of the bromo viruses infects a narrow range of plants. Mechanical transmission of bromoviruses occurs readily, and some members are transmitted by beetles.
  • other bromoviruses include broad bean mottle virus and cowpea chlorotic mottle virus.
  • a bromovirus virion is icosahedral, with a diameter of about 26 ⁇ m, containing a single species of coat protein.
  • the bromovirus genome has three molecules of linear, positive-sense, single-stranded RNA, and the coat protein mRNA is also encapsidated.
  • the RNAs each have a capped 5 '-end, and a tRNA-like structure (which accepts tyrosine) at the 3 '-end.
  • Virus assembly occurs in the cytoplasm.
  • the complete nucleotide sequence of BMV has been identified and characterized as described by Ahlquist et al., J. Mol. Biol. 153:23 (1981).
  • RICE NECROSIS VIRUS Rice Necrosis virus is a member ofthe Potato Virus Y Group or Potyviruses.
  • the Rice Necrosis virion is a flexuous filament comprising one type of coat protein (molecular weight about 32,000 to about 36,000) and one molecule of linear positive-sense single-stranded RNA.
  • the Rice Necrosis virus is transmitted by Polymyxa oraminis (a eukaryotic intracellular parasite found in plants, algae and fungi).
  • Geminiviruses are a group of small, single-stranded DNA-containing plant viruses with virions of unique morphology. Each virion consists of a pair of isometric particles (incomplete icosahedral), composed of a single type of protein (with a molecular weight of about 2.7-3.4X10 4 ). Each geminivirus virion contains one molecule of circular, positive-sense, single-stranded DNA. In some geminiviruses (i.e., Cassava latent virus and bean golden mosaic virus) the genome appears to be bipartite, containing two single-str.anded DNA molecules.
  • TEV tobacco etch virus
  • TEV tobacco etch virus
  • TEV is a well characterized potyvirus and contains a positive-strand RNA genome of 9.5 kilobases encoding for a single, large polyprotein that is processed by three virus- specific proteinases.
  • the nuclear inclusion protein "a" proteinase is involved in the maturation of several replication-associated proteins and capsid protein.
  • the helper component-proteinase (HC-Pro) and 35-kDa proteinase both catalyze cleavage only at their respective C-termini.
  • the proteolytic domain in each of these proteins is located near the C-terminus.
  • the 35-kDa proteinase and HC-Pro derive from the N-terminal region ofthe TEV polyprotein.
  • viruses which are associated with animal hosts. Some of these viruses are discussed, infra.
  • the alphaviruses are a genus ofthe viruses ofthe family Togaviridae. Almost all ofthe members of this genus are transmitted by mosquitoes, and may cause diseases in man or animals. Some of the alphaviruses are grouped into three serologicallly defined complexes. The complex-specific antigen is associated with the El protein ofthe virus, and the species-specific antigen is associated with the E2 protein ofthe virus.
  • the Semliki Forest virus complex includes Bebaru virus, Chikungunya Fever virus, Getah virus, Mayaro Fever virus, O'nyongnyong Fever virus, Ross River virus, Sagiyama virus, Semliki Forest virus and Una virus.
  • the Venezuelan Equine Encephalomyelitis virus complex includes Cabassou virus, Everglades virus, Mucambo virus, Pixuna virus and Venezuelan Equine Encephalomyelitis virus.
  • the Western Equine Encephalomyelitis virus complex includes Aura virus, Fort Morgan virus, Highlands J virus, Kyzylagach virus, Sindbis virus, Western Equine Encephalomyelitis virus and Whataroa virus.
  • the alphaviruses contain an icosahedral nucleocapsid consisting of 180 copies of a single species of capsid protein complexed with a plus-stranded mRNA.
  • the alphaviruses mature when preassembled nucleocapsid is surrounded by a lipid envelope containing two virus-encoded integral membrane glycoproteins, called El and E2.
  • the envelope is acquired when the capsid, assembled in the cytoplasm, buds through the plasma membrane.
  • the envelope consists of a lipid bilayer derived from the host cell.
  • the mRNA encodes a glycoprotein which is cotranslationally cleaved into nonstructural proteins and structural proteins.
  • the 3' one-third ofthe RNA genome consists of a 26S mRNA which encodes for the capsid protein and the E3, E2, K6 and El glycoproteins.
  • the capsid is cotranslationally cleaved from the E3 protein. It is hypothesized that the amino acid triad of His, Asp and Ser at the COOH terminus ofthe capsid protein comprises a serine protease responsible for cleavage. Hahn et al, Proc. Natl. Acad. Sci. USA 82:4648 (1985). Cotranslational cleavage also occurs between E2 and K proteins.
  • two proteins PE2 which consists of E3 and E2 prior to cleavage and an El protein comprising K6 and El are formed. These proteins are cotranslationally inserted into the endoplasmic reticulum ofthe host cell, glycosylated and transported via the Golgi apparatus to the plasma membrane where they can be used for budding. At the point of virion maturation the E3 and E2 proteins are separated. The El and E2 proteins are incorporated into the lipid envelope. It has been suggested that the basic amino-terminal half ofthe capsid protein stabilizes the interaction of capsid with genomic RNA or interacts with genomic RNA to initiate a encapsidation, Strauss et al, in the Togaviridae and Flaviviridaei.
  • Temperature sensitivity results from an A-U change at nucleotide 9502.
  • the t s lesion present cleavage of PE2 to E2 and E3 and the final maturation of progeny virions at the nonpermissive temperature.
  • Hahn et al, supra reported three temperature sensitive mutations in the capsid protein which prevents cleavage ofthe precursor polyprotein at the nonpermissive temperature. The failure of cleavage resulted in no capsid formation and very little envelope protein.
  • Dl particles Defective interfering RNAs (Dl particles) of Sindbis virus are helper- dependent deletion mutants which interfere specifically with the replication ofthe homologous standard virus. Perrault, J., Microbiol. Immunol. 93:151 (1981). Dl particles have been found to be functional vectors for introducing at least one foreign gene into cells. Levis, R., Proc. Natl. Acad. Sci. USA 84:4811 (1987). It has been found that it is possible to replace at least 1689 internal nucleotides of a Dl genome with a foreign sequence and obtain RNA that will replicate and be encapsidated. Deletions ofthe Dl genome do not destroy biological activity.
  • a 42S mRNA region which is packaged into nature virions and functions as the message for the nonstructural proteins
  • 26S mRNA which encodes the structural polypeptides.
  • the 26S mRNA is homologous to the 3' third ofthe 42S mRNA. It is translated into a 13 OK polyprotein that is cotranslationally cleaved and processed into the capsid protein and two glycosylated membrane proteins, El and E2.
  • EEEE Eastern Equine Encephalomyelitis
  • the 26S mRNA region encodes the capsid proteins, E3, E2, 6K and El.
  • the .amino terminal end ofthe capsid protein is thought to either stabilize the interaction of capsid with mRNA or to interact with genomic RNA to initiate encapsidation.
  • PE2 Uncleaved E3 and E2 proteins called PE2 is inserted into the host endoplasmic reticulum during protein synthesis.
  • the PE2 is thought to have a region common to at least five alphaviruses which interacts with the viral nucleocapsid during morphogenesis.
  • the 6K protein is thought to function as a signal sequence involved in translocation ofthe El protein through the membrane.
  • the El protein is thought to mediate virus fusion and anchoring ofthe El protein to the virus envelope.
  • the rhino viruses are a genus of viruses ofthe family Picornaviridae.
  • the rhinoviruses are acid-labile, and are therefore rapidly inactivated at pH values of less than about 6.
  • the rhinoviruses commonly infect the upper respiratory tract of mammals.
  • Human rhinoviruses are the major causal agents ofthe common cold, and many serotypes are known.
  • Rhinoviruses may be propagated in various human cell cultures, and have an optimum growth temperature of about 33°C. Most strains of rhinoviruses are stable at or below room temperature and can withstand freezing.
  • Rhinoviruses can be inactivated by citric acid, tincture of iodine or phenol/alcohol mixtures.
  • HRV2 human rhinovirus 2
  • the genome consists of 7102 nucleotides with a long open reading frame of 6450 nucleotides which is initiated 611 nucleotides from the 5'- end and stops 42 nucleotides from the poly(A) tract. Three capsid proteins and their cleavage cites have been identified.
  • Rhinovirus RNA is single-stranded and positive-sense.
  • the RNA is not capped, but is joined at the 5 '-end to a small virus-encoded protein, virion-protein genome-linked (VPg). Translation is presumed to result in a single polyprotein which is broken by proteolytic cleavage to yield individual virus proteins.
  • An icosahedral viral capsid contains 60 copies each of 4 virus proteins VP1, VP2, VP3 and VP4 and surrounds the RNA genome. Medappa, K., Virology 44:259 (1971). Analysis of the 610 nucleotides preceding the long open reading frame shows several short open reading frames.
  • the capsid proteins of HRV2: VP4, VP2, VP3 and VP1 begin at nucleotide 611, 818, 1601 .and 2311, respectively.
  • the cleavage point between VP1 and P2A is thought to be .around nucleotide 3255. Skern et al, Nucleic Acids Research 13:2111 (1985).
  • HRV89 Human rhinovirus type 89
  • Polioviruses are the causal agents of poliomyelitis in man, and are one of three groups of enteroviruses. Entero viruses are a genus ofthe family Picornaviridae (also the family of rhinoviruses). Most enteroviruses replicate primarily in the mammalian gastrointestinal tract, although other tissues may subsequently become infected. Many enteroviruses can be propagated in primarily cultures of human or monkey kidney cells and in some cell lines (e.g.
  • enteroviruses may be accomplished with heat (about 50°C), formaldehyde (3%), hydrochloric acid (0.1N) or chlorine (ca. 0.3-0.5 ppm free residual Cl 2 ).
  • Poliovirus translation produces a single polyprotein which is cleaved by proteolytic processing. Kitamura et al, Nature
  • the structures ofthe serotypes of poliovirus have a high degree of sequence homology. Their coding sequences code for the same proteins in the same order. Therefore, genes for structural proteins are similarly located.
  • PV1, PV2 and PV3 the polyprotein begins translation near the 750 nucleotide.
  • the four structural proteins VP4, VP2, VP3 and VPI begin at about 745, 960, 1790 and 2495, respectively, with VPI ending at about 3410. They are separated in vivo by proteolytic cleavage, rather than by stop/start codons.
  • Simian virus 40 is a virus ofthe genus Polyomavirus, and was originally isolated from the kidney cells of the rhesus monkey. The virus is commonly found, in its latent form, in such cells. Simian virus 40 is usually non- pathogenic in its natural host.
  • Simian virus 40 virions are made by the assembly of three structural proteins, VPI, VP2 and VP3. Girard et al, Biochem. Biophys. Res. Commun. 40:97 (1970); Prives et al, Proc. Natl. Acad. Sci. USA 21 :302 (1974); and Jacobson et al, Proc. Natl. Acad. Sci. USA 73:2747 (1976).
  • the three conesponding viral genes are organized in a partially overlapping manner. They constitute the late genes portion ofthe genome. Tooze, J., Molecular Biology of Tumor Viruses Appendix A The SV40 Nucleotide Sequence, 2nd Ed. Part 2, pp.
  • Capsid proteins VP2 and VP3 are encoded by nucleotides 545 to 1601 and 899 to 1601, respectively, and both are read in the same frame. VP3 is therefore a subset of VP2.
  • Capsid protein VPI is encoded by nucleotides 1488-2574. The end of the VP2-VP3 open reading frame therefore overlaps the VPI by 113 nucleotides but is read in an alternative frame. Tooze, J., supra. Wychowski et al, J. Virology 61:3862 (1987).
  • ADENOVIRUSES Adenovirus type 2 is a member ofthe adenovirus family or adeno virus.
  • This family of viruses are non-enveloped, icosahedral, linear, double-stranded DNA-containing viruses which infect mammals or birds.
  • the adenovirus virion consists of an icosahedral capsid enclosing a core in which the DNA genome is closely associated with a basic (argmine-rich) viral polypeptide VII.
  • the capsid is composed of 252 capsomeres: 240 hexons
  • Each penton consists of a penton base (composed of viral polypeptide III) associated with one (in mammalian adenoviruses) or two (in most avian adenoviruses) glycoprotein fibres (viral polypeptide IV).
  • the fibres can act as haemagglutinins and are the sites of attachment ofthe virion to a host cell-surface receptor.
  • the hexons each consist of three molecules of viral polypeptide II; they make up the bulk ofthe icosahedron. Various other minor viral polypeptides occur in the virion.
  • the adenovirus dsDNA genome is covalently linked at the 5 '-end of each strand to a hydrophobic 'terminal protein', TP (molecular weight about 55,000 Da); the DNA has an inverted terminal repeat of different length in different adenoviruses. In most adenoviruses examined, the 5 '-terminal residue is dCMP.
  • the virion attaches via its fibres to a specific cell-surface receptor, and enters the cell by endocytosis or by direct penetration of the plasma membrane. Most ofthe capsid proteins are removed in the cytoplasm. The virion core enters the nucleus, where the uncoating is completed to release viral DNA almost free of virion polypeptides.
  • Virus gene expression then begins.
  • the viral dsDNA contains genetic information on both strands.
  • Early genes (regions Ela, Elb, E2a, E3, E4) are expressed before the onset of viral DNA replication.
  • Late genes (regions LI, L2, L3, L4 .and L5) are expressed only after the initiation of DNA synthesis.
  • Intermediate genes (regions E2b and Iva 2 ) are expressed in the presence or absence of DNA synthesis.
  • Region Ela encodes proteins involved in the regulation of expression of other early genes, and is also involved in transformation.
  • the RNA transcripts are capped (with m 7 G 5 ppp 5 N) .and polyadenylated in the nucleus before being transferred to the cytoplasm for translation.
  • Viral DNA replication requires the terminal protein, TP, as well as virus- encoded DNA polymerase and other viral and host proteins.
  • TP is synthesized as an 80K precursor, pTP, which binds covalently to nascent replicating DNA strands.
  • pTP is cleaved to the mature 55K TP late in virion assembly; possibly at this stage, pTP reacts with a dCTP molecule and becomes covalently bound to a dCMP residue, the 3' OH of which is believed to act as a primer for the initiation of DNA synthesis.
  • Late gene expression resulting in the synthesis of viral structural proteins, is accompanied by the cessation of cellular protein synthesis, and virus assembly may result in the production of up to 10 5 virions per cell.
  • viral expression system in bacteria and yeast cells may also be employed. See
  • the nucleic acid of any suitable plant virus can be utilized to prepare a recombinant plant viral nucleic acid for use in the present invention, and the foregoing are only exemplary of such suitable plant viruses.
  • the nucleotide sequence ofthe plant virus is modified, using conventional techniques, by the insertion of one or more subgenomic promoters into the plant viral nucleic acid.
  • the subgenomic promoters are capable of functioning in the specific host plant. For example, if the host is tobacco, TMV, TEV, or other viruses containing subgenomic promoter may be utilized.
  • the inserted subgenomic promoters should be compatible with the TMV nucleic acid and capable of directing transcription or expression of adjacent nucleic acid sequences in tobacco.
  • the native coat protein gene could also be retained and a non-native nucleic acid sequence inserted within it to create a fusion protein.
  • the native or non-native coat protein gene is utilized in the recombinant plant viral nucleic acid. Whichever non-native nucleic acid is utilized may be positioned adjacent its natural subgenomic promoter or adjacent one ofthe other available subgenomic promoters.
  • the non-native coat protein as is the case for the native coat protein, is capable of encapsidating the recombinant plant viral nucleic acid and providing for systemic spread ofthe recombinant plant viral nucleic acid in the host plant.
  • the coat protein is selected to provide a systemic infection in the plant host of interest.
  • the TMV-O coat protein provides systemic infection in N benthamiana
  • TMV-U1 coat protein provides systemic infection in N tabacum.
  • the recombinant plant viral nucleic acid is prepared by cloning a viral nucleic acid. If the viral nucleic acid is D ⁇ A, it can be cloned directly into a suitable vector using conventional techniques. One technique is to attach an origin of replication to the viral D ⁇ A which is compatible with the cell to be transfected. If the viral nucleic acid is R ⁇ A, a full-length D ⁇ A copy ofthe viral genome is first prepared by well-known procedures.
  • the viral R ⁇ A is transcribed into D ⁇ A using reverse transcriptase to produce subgenomic D ⁇ A pieces, and a double-stranded D ⁇ A made using D ⁇ A polymerases.
  • the cD ⁇ A is then cloned into appropriate vectors and cloned into a cell to be transfected.
  • the cD ⁇ A's ligated into the vector may be directly transcribed into infectious R ⁇ A in vitro and inoculated onto the plant host.
  • the cD ⁇ A pieces are mapped and combined in proper sequence to produce a full-length D ⁇ A copy of the viral R ⁇ A genome, if necessary.
  • D ⁇ A sequences for the subgenomic promoters, with or without a coat protein gene are then inserted into the nucleic acid at non-essential sites, according to the particular embodiment ofthe invention utilized.
  • Non-essential sites are those that do not affect the biological properties ofthe plant viral nucleic acid.
  • the RNA genome is the infective agent
  • the cDNA is positioned adjacent a suitable promoter so that the RNA is produced in the production cell.
  • the RNA is capped using conventional techniques, if the capped RNA is the infective agent.
  • the capped RNA can be packaged in vitro with added coat protein from TMV to make assembled virions. These assembled virions can then be used to inoculate plants or plant tissues.
  • an uncapped RNA may also be employed in the embodiments ofthe present mvention. Contrary to the practiced art in scientific literature .and in issued patent (Ahlquist et al, U.S. Patent No. 5,466,788), uncapped transcripts for virus expression vectors are infective on both plants and in plant cells. Capping is not a prerequisite for establishing an infection of a virus expression vector in plants, although capping increases the efficiency of infection.
  • nucleotides may be added between the transcription start site ofthe promoter and the start ofthe cDNA of a viral nucleic acid to construct an infectious viral vector. One or more nucleotides may be added.
  • the inserted nucleotide sequence contains a G at the 5 '-end.
  • the inserted nucleotide sequence is GNN, GTN, or their multiples, (GNN) X or (GTN) X .
  • nucleic acid sequences capable of being transcribed in the plant host.
  • These nucleic acid sequences may be native nucleic acid sequences which occur in the host organism or they may be non-native nucleic acid sequences which do not normally occur in the host organism.
  • the nucleic acid sequence is placed adjacent one ofthe non- native viral subgenomic promoters and/or the native coat protein gene promoter depending on the particular embodiment used.
  • the nucleic acid is inserted by conventional techniques, or the nucleic acid sequence can be inserted into or adjacent the native coat protein coding sequence such that a fusion protein is produced.
  • the nucleic acid sequence which is transcribed may be transcribed as an RNA which is capable of regulating the expression of a phenotypic trait by an anti-sense or a positive-sense mechanism.
  • the nucleic acid sequence in the recombinant plant viral nucleic acid may be transcribed and translated in the plant host to produce a phenotypic trait.
  • the nucleic acid sequence(s) may also code for the expression of more than one phenotypic trait.
  • the recombinant plant viral nucleic acid containing the nucleic acid sequence is constructed using conventional techniques such that the nucleic acid sequence(s) are in proper orientation to whichever viral subgenomic promoter is utilized.
  • a double-stranded DNA ofthe recombinant plant viral nucleic acid or a complementary copy ofthe recombinant plant viral nucleic acid is cloned into the cell to be transfected.
  • the viral nucleic acid is a RNA molecule
  • the nucleic acid (cDNA) is first attached to a promoter which is compatible with the production cell.
  • the recombinant plant viral nucleic acid can then be cloned into any suitable vector which is compatible with the production cell. In this manner, only RNA copies ofthe chimeric nucleotide sequence are produced in the production cell.
  • the CaMV promoter can be used when plant cells are to be transfected.
  • the recombinant plant viral nucleic acid is inserted in a vector adjacent a promoter which is compatible with the production cell. If the viral nucleic acid is a DNA molecule, it can be cloned directly into a production cell by attaching it to an origin of replication which is compatible with the cell to be transfected. In this manner, DNA copies ofthe chimeric nucleotide sequence are produced in the transfected cell.
  • a further alternative when creating the recombinant plant viral nucleic acid is to prepare more than one nucleic acid (i.e., to prepare the nucleic acids necessary for a multipartite viral vector construct). In this case, each nucleic acid would require its own origin of assembly. Each nucleic acid could be prepared to contain a subgenomic promoter and a non-native nucleic acid.
  • the insertion of a non-native nucleic acid into the nucleic acid of a monopartite virus may result in the creation of two nucleic acids (i.e., the nucleic acid necessary for the creation of a bipartite viral vector).
  • Each nucleic acid would have to have its own origin of assembly.
  • the host can be infected with the recombinant plant virus by conventional techniques. Suitable techniques include, but are not limited to, leaf abrasion, abrasion in solution, high velocity water spray and other injury of a host as well as imbibing host seeds with water containing the recombinant plant virus. More specifically, suitable techniques include:
  • Hand inoculations of the encapsidated vector are performed using a neutral pH, low molarity phosphate buffer, with the addition of celite or carborundum (usually about 1%).
  • celite or carborundum usually about 1%.
  • One to four drops ofthe preparation is put onto the upper surface of a leaf and gently rubbed.
  • Plant bed inoculations are performed by spraying (gas-propelled) the vector solution into a tractor- driven mower while cutting the leaves. Alternatively, the plant bed is mowed and the vector solution sprayed immediately onto the cut leaves.
  • High Pressure Spray of Single Leaves Single plant inoculations can also be performed by spraying the leaves with a narrow, directed spray (50 psi, 6-12 inches from the leaf) containing approximately 1% carborundum in the buffered vector solution.
  • Vacuum Infiltration may be accomplished by subjecting the host organism to a substantially vacuum pressure environment in order to facilitate infection.
  • An alternative method for introducing a recombinant plant viral nucleic acid into a plant host is a technique known as agroinfection or Agrobacterium- mediated transformation (sometimes called Agro-infection) as described by Grimsley et al, Nature 325:177 (1987).
  • This technique makes use of a common feature of Agrobacterium which colonizes plants by transferring a portion of their DNA (the T-DNA) into a host cell, where it becomes integrated into nuclear DNA.
  • the T-DNA is defined by border sequences which are 25 base pairs long, and any DNA between these border sequences is transfened to the plant cells as well.
  • the insertion of a recombinant plant viral nucleic acid between the T-DNA border sequences results in transfer ofthe recombinant plant viral nucleic acid to the plant cells, where the recombinant plant viral nucleic acid is replicated, and then spreads systemically through the plant.
  • Agro-infection has been accomplished with potato spindle tuber viroid (PSTV) (Gardner et al, Plant Mol. Biol. 6:221 (1986); CaV (Grimsley et al, Proc. Natl. Acad. Sci. USA 83:3282 (1986)); MSV (Grimsley et al, Nature 325:177 (1987)), and Lazarowitz, S., Nucl. Acids Res.
  • PSTV potato spindle tuber viroid
  • agro-infection of a susceptible plant could be accomplished with a virion containing a recombinant plant viral nucleic acid based on the nucleotide sequence of any ofthe above viruses. Particle bombardment or electrosporation or any other methods known in the art may also be used.
  • Infection may also be attained by placing a selected nucleic acid sequence into an organism such as E. coli, or yeast, either integrated into the genome of such organism or not and then applying the organism to the surface ofthe host organism. Such a mechanism may thereby produce secondary transfer ofthe selected nucleic acid sequence into the host organism. This is a particularly practical embodiment when the host organism is a plant.
  • infection may be attained by first packaging a selected nucleic acid sequence in a pseudovirus. Such a method is described in WO 94/10329, the teachings of which are inco ⁇ orated herein by reference. Though the teachings of this reference may be specific for bacteria, those of skill in the art will readily appreciate that the same procedures could easily be adapted to other organisms.
  • the function of a nucleic acid may be determined by complementation analysis. That is, the function ofthe nucleic acid of interest may be determined by observing the endogenous gene or genes whose function is replaced or augmented by introducing the nucleic acid of interest.
  • a discussion of this principle is provided by Napoli et al, The Plant Cell 2:279-289 (1990) which is inco ⁇ orated herein by reference. Further teachings in these regards are provided by WO 97/42210, the disclosure of which is also inco ⁇ orated herein by reference.
  • the function of a nucleic acid may be determined by analyzing the biochemical alterations in the accumulation of substrates or products from enzymatic reactions according to any one ofthe means known by those skilled in the art.
  • the function of a nucleic acid may be determined by observing phenotypic changes in the host by methods including mo ⁇ hological, macroscopic or microscopic analysis.
  • the function of a nucleic acid may be determined by observing the change in biochemical pathways which may be modified in the host as a result ofthe local and/or systemic expression ofthe non- native nucleic acids.
  • the function of a nucleic acid may be determined utilizing techniques known by those skilled in the art to observe inhibition of gene expression in the cytoplasm of cells as a result of expression of the non-native nucleic acid.
  • a particularly useful way to determine gene function is by observing the phenotype in a whole plant when a particular gene function has been silenced.
  • Useful phenotypic traits in plant cells which may be observed microscopically, macroscopically or by other methods include, but are not limited to, improved tolerance to herbicides, improved tolerance to extremes of heat or cold, drought, salinity or osmotic stress; improved resistance to pests (insects, nematodes or arachnids) or diseases (fungal, bacterial or viral) production of enzymes or secondary metabolites; male or female sterility; dwarfhess; early maturity; improved yield, vigor, heterosis, nutritional qualities, flavor or processing properties, and the like.
  • phenotypic traits include the production of important proteins or other products for commercial use, such as lipase, melanin, pigments, alkaloids, antibodies, hormones, pharmaceuticals, antibiotics and the like.
  • Another useful phenotypic trait is the production of degradative or inhibitory enzymes, such as are utilized to prevent or inhibit root development in malting barley or that determine response or non-response to a systemically administered drug in a human.
  • the phenotypic trait may also be a secondary metabolite whose production is desired in a bioreactor.
  • Another particularly useful means to determine function of nucleic acids transfected into a host is to observe the effects of gene silencing.
  • EST/DNA library from an organism for example Arabidopsis thaliana
  • the DNA sequences in the transcription plasmid library may then be introduced into plant cells as part of a functional RNA virus which post-transcriptionally silences the homologous target gene.
  • the EST/DNA sequences may be introduced into a plant viral vector in either the plus or minus sense orientation, and the orientation can be either directed or random based on the cloning strategy.
  • a high-throughput, automated cloning scheme based on robotics may be used to assemble and characterize the library.
  • double stranded RNA may also be an effective stimulator of gene silencing/co- suppression in transgenic plant. Gene silencing/co-suppression of plant genes may be induced by delivering an RNA capable of base pairing with itself to form double stranded regions. This approach could be used with any plant or non- plant gene to assist in the identification ofthe function of a particular gene sequence.
  • a particularly troublesome problem with gene silencing in plant hosts is that many plant genes exist in a multigene family. Therefore, effective silencing of a gene function may be especially problematic.
  • nucleic acids may be inserted into the genome to effectively silence a particular gene function or to silence the function of a multigene family. It is presently believed that about 20% of plant genes exist in multigene families.
  • a single nucleotide sequence of about 20 to 100 or more bases having about 70% or more homology to a gene may silence an entire plant gene family having two or more homologous genes.
  • RNA can reduce the expression of a target gene through inhibitory RNA interactions with target mRNA that occur in the cytoplasm and/or the nucleus of a cell.
  • Full-length cDNAs may be accessed from public and private repositories or extracted from field samples for insertion of unknown open reading frames into viral vectors for expression of nucleic acids in the host organism and thereby utilized as an alternative to antisense gene knockout.
  • This technology may be implemented by PCR amplification and cloning of all cDNAs that do not share homology with gene sequences in public and or private databases.
  • the cDNAs may be expressed in plants transfected with one or more plant viral vectors for subsequent analysis of novel phenotype ofthe whole plant (biochemical and mo ⁇ hological).
  • Selected cDNA sequences from maize, rice, soybean canola and other crop species may be used to assemble the cDNA libraries. This method may thus be used to search for useful dominant gene phenotypes from novel cDNA libraries through the gene expression.
  • An EST/cDNA library from an organism such as Arabidopsis thaliana may be assembled into a plant viral transcription plasmid background.
  • the cDNA sequences in the transcription plasmid library can then be introduced into plant cells as cytoplasmic RNA in order to post-transcriptionally silence the endogenous genes.
  • the EST/cDNA sequences may be introduced into the plant viral transcription plasmid in either the plus or anti-sense orientation (or both), and the orientation can be either directed or random based on the cloning strategy.
  • a high-throughput, automated cloning strategy using robotics can be used to assemble the library.
  • the EST clones can be inserted behind a duplicated subgenomic promoter such that they are represented as subgenomic transcripts during viral replication in plant cells.
  • the EST/cDNA sequences can be inserted into the genomic RNA of a plant viral vector such that they are represented as genomic RNA during the viral replication in plant cells.
  • the library of EST clones is then transcribed into infectious RNA and inoculated onto individual platelets of Arabidopsis thaliana (or other plant species).
  • the viral RNA containing the EST/cDNA sequences contributed from the original library are now present in a sufficiently high concentration in the cytoplasm such that they cause post-transcriptional gene silencing ofthe endogenous plant-gene homologs.
  • the orientation ofthe EST/cDNA insert is normally irrelevant in terms of producing the desired gene-silenced phenotype in the tissue.
  • Partial cDNA sequences cloned into a plant viral vector in the sense orientation have previously been shown to also confer a gene silencing phenotype (Kumagai et al, Proc. Natl. Acad. Sci. USA 92:1679 (1995)), the teachings of which are inco ⁇ orated herein by reference.
  • the actual mechanism of gene silencing has not been fully determined. This phenomenon may be similar to the gene silencing via cosuppression observed in transgenic plants.
  • the plant tissue may then be taken for sophisticated biochemical analysis in order to determine which metabolic pathway has been affected by the EST/DNA gene silencing, and in particular, which steps in a given metabolic pathway have been affected by the EST/DNA gene silencing.
  • Biochemical analysis may be done, for example, in a high-throughput, fully automated fashion using robotics. Suitable biochemical analysis may include MALDI-TOF, LC/MS, GC/MS, two-dimensional IEF/SDS-PAGE, ELISA or other methods of analyses.
  • the clones in the EST/plant viral vector library may then be functionally classified based on metabolic pathway affected or visual/selectable phenotype produced in the plant. This process enables the rapid determination of gene function for unknown EST/DNA sequences of plant origin. Furthermore, this process can be used to rapidly confirm function of full-length DNA's of unknown gene function. Functional identification of unknown ESTDNA sequences in a plant library may then rapidly lead to identification of similar unknown sequences in expression libraries for other crop species based on
  • Links may be made between sequences from various species predicted to carry out similar biochemical or regulatory functions. Links may also be generated between predicted enzymatic activities and visually displayed biochemical and regulatory pathways. Likewise, links may be generated between predicted enzymatic or regulatory activity and known small molecule inhibitors, activators, substrates or substrate analogs. Phenotypic data from expression libraries expressed in transfected hosts maybe automatically linked within such a relational database. Genes with similar predicted roles of interest in other crop plants or crop plant pests may thereby be more rapidly discovered. A complete classification scheme of gene functionality for a fully sequenced eukaryotic organism has been established for yeast. This classification scheme may be modified for plants and divided into the appropriate categories. Such organizational structure may be utilized to rapidly identify herbicide target loci which may confer dominant lethal phenotypes, and thereby is useful in helping to design rational herbicide programs.
  • a second aspect ofthe present invention is a method of silencing endogenous genes in a host by introducing nucleic acids into the host by way of a viral nucleic acid suitable to produce the local and systemic expression ofthe nucleic acid of interest.
  • the host is a plant, but those skilled in the art will imderstand that other hosts may also be utilized.
  • This method utilizes the principle of post-transcription gene silencing ofthe endogenous host gene homolog as described above. Since the replication mechanism produces both sense and anti-sense RNA sequences as disclosed above, the orientation of the non-native nucleic acid insert is not crucial to providing gene silencing. More information describing some aspects ofthe "gene silencing" effect is provided in co-pending U.S. Patent Application Serial No.
  • RNA can reduce the expression of a target gene through inhibitory RNA interactions with target mRNA that occur in the cytoplasm and/or the nucleus of a cell.
  • Silencing of endogenous genes can be achieved with homologous (but not identical) sequences from distant plant species.
  • the Nicotiana benthamiana gene for phytoene desaturase may be silenced by fransfection with a partial tomato cDNA for PDS (cloned in either the positive or antisense orientation).
  • the tomato PDS cDNA is 92% homologous at the nucleotide level yet is still able to confer efficient gene silencing in an unrelated plant species (Kumagai et al, Proc. Natl. Acad. Sci. USA 92:1679 (1995)).
  • Identification of EST/cDNA gene function in Arabidopsis thaliana could then be extrapolated to similar EST/cDNA sequences of unknown function that exist in other libraries (e.g., soybean, maize, rice, oilseed rape, etc.).
  • a third aspect ofthe present invention is a method for selecting desired functions of RNAs and proteins by the use of virus vectors to express libraries of nucleic acid sequence variants.
  • Libraries of sequence variants may be generated by means of in vitro mutagenenisis and/or recombination. Rapid in vitro evolution can be used to improve virus-specific or protein-specific functions.
  • plant RNA virus expression vectors may be used as tools to bear libraries containing variants of nucleic acid, genes from virus, plant or other sources, and to be applied to plants or plant cells such that the desired altered effects in the RNA or protein products can be determined, selected and improved.
  • nucleic acid shuffling techniques may be employed to construct shuffled gene libraries.
  • Random, semi-random or known sequences of virus origin may also be inserted in virus expression vectors between native virus sequences and foreign gene sequences, to increase the genetic stability of foreign genes in expression vectors as well as the translation ofthe foreign gene and the stability ofthe mRNA encoding the foreign gene in vivo.
  • the desired function of RNA and protein may include the promoter activities, replication properties, translational efficiencies, movement properties (local and systemic), signaling pathway, or virus host range, among others.
  • the desired function alteration can be identified by assaying infected plants and the nature of mutation can be determined by analysis of sequence variants in the virus vector.
  • Methods to increase the representation of gene sequences in virus expression libraries may also be achieved by bypassing the genetic bottleneck of propagation in E. coli.
  • cell-free methods may be used to clone sequence libraries or individual arrayed sequences into virus expression vectors and reconstruct an infectious virus, such that the final ligation product can be transcribed and the resulting RNA can be used for plant or plant cell inoculation/infection with the output being gene function discovery or protein production.
  • RNA viruses or RNA virus vectors as populations or individuals in parallel to identify individuals with novel and augmented virus-encoded functions in replication and virus movement, foreign gene sequence retention in vectors and proper folding, activity and expression of protein products, novel gene expression, effects on host metabolism, and resistance or susceptibility of plants to exogenous agents.
  • Variation in the sequence of a native virus gene(s) or heterologous nucleotide sequence(s) may be introduced into an RNA virus or an RNA virus expression vector by many methods as a means to screen a population of variants in batch or individuals in parallel for novel properties exhibited by the virus itself or conferred on the host plant or cell by the virus vector.
  • Variant populations can be transfected as populations or individual clones into "host”: 1) protoplasts; 2) whole plants; or 3) inoculated leaves of whole plants and screened for various traits including protein expression (increase or decrease), RNA expression (increase or decrease), secondary metabolites or other host property gained or loss as a result ofthe virus infection.
  • a virus vector which simultaneously expressed the green fluorescent protein (GFP) or other selectable marker gene and the variant sequence, is used to screen quantitatively for levels of resistance or sensitivity to the agent in question conferred upon the host by the variant sequence expressed from the viral vector.
  • GFP green fluorescent protein
  • a new mode of enhanced or reduced expression could be identified by finding the factors that respond to cellular signals that in turn alter its particular expression.
  • transcription factors regulating the expression of defense proteins such as systemin peptides, or protease inhibitors could be identified by transfecting hosts with virus libraries and the expression of systemin or protease inhibitors or their RNAs be directly assayed.
  • the promoters responsible for expressing these genes could be genetically fused to the green fluorescent protein and introduced into hosts as transient expression constructs or into stable transformed host cells/tissues. The resulting cells would be transfected with viral vector libraries.
  • Hosts now could be screened rapidly by following relative GFP expression following vector fransfection.
  • coupling the transfecting of hosts with virus libraries with the treatment of plants with methyl jasmonate could identify sequences that reverse or enhance the gene induction events induced by this metabolite.
  • This approach could be applied to other factors involved in promotion of higher biomass in plants such as Leafy or DET2.
  • the expression of these factors could be directly assayed or via promoters genetically fused to GFP. This technique will enable screening of populations or individuals followed by rescue ofthe sequence from virus vectors conferring desired trait by RT-PCR and re-screening of particular variant sequences in secondary screens.
  • a fourth aspect ofthe present invention is a method for inhibiting an endogenous protease of a plant host comprising the step of treating the plant host with a compound which induces the production of an endogenous inhibitor of said protease.
  • jasmonic acid may be used to treat the plant host to induce the production of an endogenous inhibitor of an endogenous protease.
  • the treatment ofthe plant host with a compound results an increased representation of an exogenous nucleic acid or the protein product thereof.
  • transgenic hosts expressing protease inhibitors may be used to decrease the degradation of proteins expressed by virus expression vectors.
  • jasmonic acid may be used to treat plants infected with virus expression vectors to decrease the degradation of proteins expressed by virus expression vectors.
  • a fifth aspect ofthe present invention are genes and fragments thereof, nucleotide sequences, and gene products obtained by way ofthe method ofthe present invention.
  • the present invention features expressing selected nucleotide sequences in a host organism such as, for example, a plant.
  • a host organism such as, for example, a plant.
  • Such gene products may exhibit biological activity as pharmaceuticals, herbicides, and other similar functions.
  • a hybrid virus consisting of TMV-U1 and ToMV-F was constructed by swapping an 874- bp BamRl-Kpnl ToMV fragment into pBGC152, creating plasmid TTO1. The inserted fragment was verified by dideoxynucleotide sequencing.
  • a unique Avrll site was inserted downstream ofthe Xhol site in TTO1 by PCR mutagenesis, creating plasmid TTO1 A, using the following oligonucleotides: 5'-
  • Partial cDNAs were isolated from ripening tomato fruit RNA by polymerase chain reaction (PCR) using the following oligonucleotides: PSY, 5'-TATGTATGGTGCAGAAGAACAGAT-3' (upstream) (SEQ ID NO: 5), 5 * -AGTCGACTCTTCCTCTTCTGGCAT C-3' (downstream) (SEQ ID NO: 6); PDS, 5'- TGCTCGAGTGTGTTCTTCAGTTTTCTGTCA-3' (SEQ ID NO: 7)
  • TTO1/PSY + (FIGURE 1) contains the phytoene synthase cDNA in the positive orientation under the control ofthe TMV-U1 coat protein subgenomic promoter; while, the vector TTOl/PSY - contains the phytoene synthase cDNA in the antisense orientation.
  • TTO1A/PDS + (FIGURE 2) contains the phytoene desaturase cDNA in the positive orientation under the control ofthe TMV-U1 coat protein subgenomic promoter; while the vector TTO1A/PDS - contains the phytoene desaturase cDNA in the antisense orientation.
  • TTO1/PSY+ Transfection and analysis of N. benthamiana ITTO1/PSY+.
  • Infectious RNAs from TTO1/PSY+ (FIGURE 1), TTO 1 /PS Y-TTO 1 /PDS +, TTO1/PDS+ were prepared by in vitro transcription using SP6 DNA-dependent RNA polymerase as described previously (Dawson et al, Proc. Natl. Acad. Sci. USA 83:1832 (1986)) and were used to mechanically inoculate N benthamiana.
  • the hybrid viruses spread throughout all the non- inoculated upper leaves as verified by transmission electron microscopy, local lesion infectivity assay, and polymerase chain reaction (PCR) amplification.
  • the viral symptoms resulting from the infection consisted of distortion of systemic leaves and plant stunting with mild chlorosis.
  • the leaves from plants transfected with TTO1/PSY+ turned orange and accumulated high levels of phytoene while those transfected with TTO1 ⁇ /PDS+ and TTO1 ⁇ /PDS- turned white.
  • Agarose gel eletrophoresis of PCR cD ⁇ A isolated from virion R ⁇ A and Northern blot analysis of virion RNA indicate that the vectors are maintained in an extrachromosomal state and have not undergone any detectable intramolecular rearrangements.
  • the carotenoids were isolated from systemically infected tissue and analyzed by HPLC chromatography. Carotenoids were extracted in ethanol and identified by their peak retention time and abso ⁇ tion spectra on a 25-cm Spherisorb® ODS-1 5- m column using acetonitrile/methanol/2-propanol (85:10:5) as a developing solvent at a flow rate of 1 ml/min. They had identical retention time to a synthetic phytoene standard and ⁇ -carotene standards from carrot and tomato. The phytoene peak from N benthamiana transfected with TTO1/PSY + had an optical absorbance maxima at 276, 285, and 298 nm.
  • Plants transfected with viral encoded phytoene synthase showed a ten- fold increase in phytoene compared to the levels in noninfected plants.
  • the expression of sense and antisense R ⁇ A to a partial phytoene desaturase in transfected plants inhibited the synthesis of colored carotenoids and caused the systemically infected leaves to turn white. HPLC analysis of these plants revealed that they also accumulated phytoene. The white leaf phenotype was also observed in plants treated with the herbicide norflurazon which specifically inhibits phytoene desaturase.
  • This change in the levels of phytoene represents one ofthe largest increases of any carotenoid (secondary metabolite) in any genetically engineered plant.
  • Plants transfected with viral-encoded phytoene synthase showed a ten-fold increase in phytoene compared to the levels in noninfected plants.
  • the accumulation of phytoene in plants transfected with positive-sense or antisense phytoene desaturase suggests that viral vectors can be used as a potent tool to manipulate pathways in the production of secondary metabolites through cytoplasmic .antisense inhibition.
  • EXAMPLE 2 Expression of bell pepper cD ⁇ A in transfected plant confirms that it encodes capsanthin-capsorubin svnthase.
  • capsanthin In contrast to the situation prevailing in chromoplasts, capsanthin was not esterified and its increased level was balanced by a concomitant decrease ofthe major leaf xanthophylls, suggesting an autoregulatory control of chloroplast carotenoid composition. Capsanthin was exclusively recruited into the trimeric and monomeric light-harvesting complexes of Photosystem II. This demonstration that higher plant antenna complexes can accommodate non-native carotenoids provides compelling evidence for functional remodeling of photosynthetic membranes by rational design of carotenoids.
  • Ccs bell pepper capsanthin-capsorubin synthase
  • the 1.6-kb Xhol, Avrll cDNA fragment was placed under the control ofthe TMV-U1 coat protein subgenomic promoter by subcloning into TTO1A, creating plasmid TTO1A CCS+ (FIGURE 3) in the sense orientation as represented by FIGURE 3.
  • EXAMPLE 3 Expression of bacterial Crt-9 gene in transfected plants confirms that it encodes phytoene svnthase.
  • TTU51 tobacco mosaic virus strain Ul (TMV-U1) (Goelet et al, Proc. Natl. Acad. Sci. USA 79:5818- 5822 (1982)), and tobacco mild green mosaic virus (TMGMV; U5 strain) (Solis et al, "The complete nucleotide sequence ofthe genomic RNA ofthe tobamovirus tobacco mild green mosaic virus” (1990)).
  • TMV tobacco mosaic virus
  • This construct also contained the gene encoding the chloroplast targeting peptide (CTP) for the small subunit of ribulose- 1,5-bisphosphate carboxylase (RUBISCO) (O'Neal et al, Nucl. Acids Res. L5: 8661-8677 (1987)) and was called TTU51 CTP Crt-5 as represented by FIGURE 4.
  • CTP chloroplast targeting peptide
  • RUBISCO ribulose- 1,5-bisphosphate carboxylase
  • Phytoene synthase activity from plants transfected with TTU51 CTP Crt-5 was assayed using isolated chloroplasts and labeled [ 14 C] geranylgeranyl PP. There was a large increase in phytoene and an unidentified C 40 alcohol in the CrtB plants. Phytoene synthetase assay.
  • the chloroplasts were prepared as described previously (Camara, Methods Enzymol. 214:352-365 (1993)).
  • the phytoene synthase assays were carried out in an incubation mixture (0.5 ml final volume) buffered with Tris-HCL, pH 7.6, containing [ 14 C] geranylgeranyl PP (100,000 cpm) (prepared using pepper GGPP synthase expressed in E. coli), 1 mM ATP, 5 mM MnCl , 1 mM MgCl 2 , Triton X-100 (20 mg per mg of chloroplast protein) and chloroplast suspension equivalent to 2 mg protein.
  • reaction products were extracted with chloroform methanol (Camara, supra) and subjected to TLC onto silicagel plate developed with benzene/ethyl acetate (90/10) followed by autoradiography.
  • Chlorophyll synthetase assay Chlorophyll synthetase assay.
  • the isolated chloroplasts were lysed by osmotic shock before incubation.
  • the reaction mixture (0.2 ml, final volume) consisting of 50 mM Tris-HCL (pH 7.6) containing [ 14 C] geranylgeranyl PP (100,000 cpm), 5 MgCl 2 , 1 mM ATP, and ruptured plasmid suspension equivalent to 1 mg protein was incubated for 1 hr at 30°C.
  • the reaction products were analyzed as described previously.
  • TTU51 Tobamoviral vector, TTU51, was constructed.
  • PDS Erwinia phytoene desaturase
  • the chloroplast targeting, bacterial phytoene desaturase expression vector, TTOSAl CTP CrtI 491 #7 was constructed as follows. First, a unique Sphl site was inserted in the start codon for the Erwinia herbicola phytoene desaturase gene (plasmid pAU211, (FIGURE 6) by polymerase chain reaction (PCR) mutagenesis using the oligonucleotides CrtI HSM1 5'-GA CAG AAG CTT TGC AGC ATG CAA AAA ACC GTT-3' (upstream) (SEQ ID NO: 16) and IQ419A 5'-CGC GGT CAT TGC AGA TCC TCA ATC ATC AGG C-3' (downstream) (SEQ ID NO: 17).
  • the 1504 bp CrtI PCR fragment was subcloned into pBluescript® (Stratagene) by inserting it between the EcoRV and Hindlll sites, creating plasmid KS+/Crt/* 491 (FIGURE 7).
  • a 1481 bp Sphl, Avrll CrtI fragment from plasmid KS+ICrtI* 491 was then subcloned into the tobamo viral vector TTOSAl , creating TTOSAl CTP CrtI 491 #7.
  • Leaf tissue from a TTOSAl CTP CrtI 491 #7 infected plant was examined for ⁇ -carotene levels. Treating an uninoculated control plant with norflurazon resulted in severely depressed ⁇ -carotene levels (7.8% ofthe wild- type level). However, when a plant which had been previously inoculated with the viral construct TTOSAl CTP CrtI 491 #7 was treated with norflurazon, the ⁇ - carotene level were partially restored (28.3% ofthe wild-type level).
  • EXAMPLE 5 Expression of 5-enolpyruvylshikimate-3-phosphate svnthase -.EPSPS ("EPSPS”) genes in plants confers resistance to Roundup® herbicide.
  • EPSPS 5- enolpyruvylshikimate-3-phosphate synthase
  • the purpose of this experiment is to provide a method to systemically express EPSPS genes via a recombinant viral vector in fully-grown pl-ants.
  • Transfected plants that ove ⁇ roduce the enzyme EPSPS in vegetative tissue (root, stem, and leaf) are resistant to Roundup® herbicide.
  • the present invention provides a method for the production of plasmid-targeted EPSPS in plants via an RNA viral vector.
  • a dual subgenomic promoter vector encoding the full-length EPSPS gene from Nicotiana tabacum (Class I EPSPS) is shown in plasmid pBS736.
  • Systemic expression ofthe Nicotiana tabacum Class I EPSPS confers resistance to Roundup® herbicide in whole plants and tissue culture.
  • FIGURE 8 shows plasmid pBS736.
  • EXAMPLE 6 Cvtoplasmic inhibition of 5-enolpyruwlshikimate-3-phosphate svnthase (EPSPS) genes in plants blocks aromatic amino acid biosynthesis.
  • EPSPS 5-enolpyruwlshikimate-3-phosphate svnthase
  • EPSPS 5-enolpyruvylshikimate-3-phosphate synthase
  • a transgenic plant or naturally occurring plant mutant may have a nonfunctional gene such as the one which produces EPSP synthase.
  • a plant deficient or lacking in the EPSP synthase gene could grow only in the presence of added aromatic amino acids.
  • Transfection of plants with a viral vector containing a functional EPSP synthase gene or cDNA sequence encoding the same would cause the plant to produce a functional EPSP synthase gene product.
  • a plant so transfected would then be able to grow normally without added aromatic amino acids to its environment.
  • the EPSP synthase mutation in the plant would be complemented in trans by the viral nucleic acid sequence containing the native or foreign EPSP synthase cDNA sequence.
  • EXAMPLE 8 Expression of methylotrophic veast ZZA1 gene in transfected plants confirms that it encodes alcohol oxidase.
  • Infectious RNA from TTO1 APE ZZA1 (FIGURE 11) was prepared by in vitro transcription using SP6 DNA-dependent RNA polymerase and used to mechanically inoculate N benthamiana.
  • the 72-kDa protein accumulated in systemically infected tissue and was analyzed by immunoblotting, using Pichia pastoris alcohol oxidase as a standard. No detectable cross-reacting protein was observed in the noninfected N benthamiana control plant extracts.
  • Thermus aquaticus DNA polymerase (2.5U; Perkin-Elmer Cetus) consisted of an initial 2 minute incubation at 97°C followed by two cycles at 97°C (Imin.), 45°C (Imin.), 60°C (1 min.), thirty-five cycles at 94°C (1 min.), 45°C (1 min.), 60°C (1 min.), and a final DNA polymerase extension at 60°C for 7 min.
  • the 3273 base pair fragment containing ZZA1 gene was phenol/chloroform treated and precipitated with ammonium acetate/ethanol.
  • the alcohol oxidase genomic clone KS-AO7'8' was characterized by restriction mapping and dideoxynucleotide sequencing.
  • Rapid, high-level expression of rice OS 103 cDNA in transfected plants confirms that it encodes glycosylated rice ⁇ -amylase.
  • the hybrid virus spread throughout the noninoculated upper leaves as verified by transmission electron microscopy, local lesion infectivity assay, and PCR amplification.
  • the viral symptoms consisted of plant stunting with mild chlorosis and distortion of systemic leaves.
  • the 46-kDa ⁇ -amylase accumulated to levels of at least 5% total soluble protein, and was analyzed by immunoblotting, using yeast expressed ⁇ -amylase as a standard. No detectable cross-reacting protein was observed in the noninfected N benthamiana control plant extracts.
  • the expression level ofthe recombinant enzyme produced in transfected plants was at least ten times higher than the amount of thermostable bacterial ⁇ -amylase produced in transgenic tobacco.
  • the ⁇ -amylase was purified using ion exchange chromatography and its structural and biological properties were analyzed.
  • the secreted protein had an approximate relative molecular mass of 46 kDa, cross-reacted with anti- ⁇ -amylase antibody, and hydro lyzed starch and oligomaltose in an in vitro assay.
  • the recombinant enzyme from transfected N benthamiana was glycosylated at an asparagine residue via an N-glycosidic linkage.
  • the heterologously expressed ⁇ -amylases from transfected N benthamiana .and from transformed strains of S. cerevisiae and P. pastoris were treated with endo-H and were compared by Western blot SDS-PAGE analysis. There was an equivalent mobility shift for the enzymes expressed in S. cerevisiae and P. pastoris. The extent ofthe change in mobility suggests that the yeast expressed enzymes are hyperglycosylated while the recombinant protein from transfected plants is similar to that ofthe native rice ⁇ -amylase.
  • the 1.4-kb Xhol, Avrll OS 103 PCR fragment was subcloned into pTTOlA, creating plasmid TTOl A 103L.
  • the ⁇ -amylase was eluted with a linear gradient of 0.0- 1 M ⁇ aCl in 50 mM Tris-bis propane pH 7.0.
  • the ⁇ -amylase eluted in fraction 16, 17 and its enzyme activity was analyzed (Sigma Kit #576-3).
  • Fractions containing cross-reacting material to ⁇ -amylase antibody were concentrated with a Centriprep-30® (Amicon) and the buffer was exchanged by diafiltration (50 mM Tris-bis propane, pH 7.0).
  • the sample was then loaded on a POROS HQ/M column (Perceptive Biosystems), eluted with a linear gradient of 0.0-1 M ⁇ aCl in 50 mM Tris-bis propane pH 7.0, and assayed for ⁇ -amylase activity. Fractions containing cross-reacting material to ⁇ -amylase antibody were concentrated with a Centriprep-30 and the buffer was exchanged by diafiltration (20 mM Sodium Acetate/HEPES/MES, pH 6.0). The sample was finally loaded on a POROS HQ/M column (Perceptive Biosystems), eluted with a linear gradient of 0.0-1 M ⁇ aCl in 50 mM Tris-bis propane pH 7.0, and assayed for ⁇ -amylase activity. Fractions containing cross-reacting material to ⁇ -amylase antibody were concentrated with a Centriprep-30 and the buffer was exchanged by diafiltration (20 mM Sodium Acetate/HEPES/MES,
  • HS/TVI column Perceptive Biosystems
  • Total soluble plant protein concentrations were determined using bovine serum albumin as a standard.
  • the proteins were analyzed on a 0.1 % SDS/ 10% polyacrylamide gel and fransfened by elecfrob lotting for 1 hour to a nitrocellulose membrane. The blotted membrane was incubated for 1 hr with a 2000- fold dilution of anti- ⁇ -amylase antiserum.
  • the antisera was raised in rabbits against S cerevisiae expressed rice ⁇ -amylase.
  • the enhanced chemiluminescence horseradish peroxidase-linked, goat anti-rabbit IgG assay (Cappel Laboratories) was performed according to the manufacturer's (Amersham) specifications.
  • the blotted membrane was subjected to film exposure times of up to 10 sec.
  • the quantity of total recombinant ⁇ -amylase in an extracted leaf sample was determined (using a 1-sec exposure ofthe blotted membrane) by comparing the crude extract chemiluminescent signal to the signal obtained from known quantities of ⁇ -amylase. Shorter and longer chemiluminescent exposure times ofthe blotted membrane gave the same quantitative results.
  • recombinant protein Approximately 5 ⁇ g of recombinant protein was dissolved in 1 M acetic acid and subjected to matrix-assisted laser deso ⁇ tion/ionization time of flight (MALDI-TOF) analysis (Karas et al, Anal Chem. 60:2299-2301 (1988)).
  • MALDI-TOF matrix-assisted laser deso ⁇ tion/ionization time of flight
  • endo H endo--5-N-acetylglucosaminidase H
  • 2 ⁇ g ofthe recombinant ⁇ -amylases were denatured in 0.5% SDS/ 1% ⁇ -mercaptoethanol at 100°C for 10 minutes.
  • EXAMPLE 10 Expression of Chinese cucumber cDNA clone pO21D in transfected plants confirms that it encodes ⁇ -trichosanthin.
  • the ⁇ - trichosanthin open reading frame was verified by dideoxy sequencing, and placed under the control ofthe TMV-U1 coat protein subgenomic promoter by subcloning into TTU51 A, creating plasmid TTU51 A QSEO #3.
  • N benthaminana plants were inoculated with in vitro transcripts of Kpn I- digested TTU51A QSEO #3 as previously described (Dawson et al, supra). Virions were isolated from N benthamiana leaves infected with TTU51A QSEO #3 transcripts.
  • the hemoglobin expression vector, RED1 was constructed in several subcloning steps. A unique Sphl site was inserted in the start codon for the human ⁇ -globin and a Xbal site was placed downstream ofthe stop codon by
  • This construct consists of a 1020 bp fragment from the tobacco mild green mosaic virus (TMGMV; U5 strain) containing the viral subgenomic promoter, coat protein gene, and the 3'-end that was isolated by PCR using TMGMV primers 5' ⁇ GGC TGT GAA ACT CGA AAA GGT TCC GG-3* (upstream) (SEQ ID NO: 28) and 5'-CGG GGT ACC TGG GCC GCT ACC GGC GGT TAG GGG AGG-3' (downstream) (SEQ ID NO: 29).
  • TMGMV tobacco mild green mosaic virus
  • a hybrid sequence encoding rice alpha-amylase signal peptide and ⁇ - chain of human hemoglobin was placed under the control ofthe tobacco mosaic virus (TMV-U1) coat protein subgenomic promoter. Infectious RNA was made in vitro and directly applied to N benthamiana. One to two weeks post- inoculation transfected plants had accumulated recombinant hemoglobin. The 16-KDa ⁇ -globin accumulated in systemically infected leaves and was analyzed by immunoblotting, using hum.an hemoglobin as a standard. The recombinant hemoglobin was detected in transfected plants using a rabbit anti-human hemoglobin antibody. No detectable cross-reacting protein was observed in the noninfected N benthamiana control plants.
  • TMV-U1 tobacco mosaic virus
  • EXAMPLE 12 Construction of a tobamoviral vector for expression of heterologous genes in A. thaliana. Virions that were prepared as a crude aqueous extract of tissue from turnip infected with RMV were used to inoculate N benthamiana, N tabacum, A. thaliana, and oilseed rape (canola). Two to three weeks after fransfection, systemically infected plants were analyzed by immunoblotting, using purified RMV as a standard.
  • Total soluble plant protein concentrations were determined using bovine serum albumin as a standard. The proteins were analyzed on a 0.1% SDS/12.5% polyacrylamide gel and transfened by electroblotting for 1 hr to a nitrocellulose membrane. The blotted membrane was incubated for 1 hr with a 2000-fold dilution of anti-ribgrass mosaic virus coat antiserum. Using standard protocols, the antisera was raised in rabbits against purified RMV coat protein. The enhanced chemiluminescence horseradish peroxidase- linked, goat anti-rabbit IgG assay (Cappel Laboratories) was performed according to the manufacturer's (Amersham) specifications. The blotted membrane was subjected to film exposure times of up to 10 sec.
  • Ribgrass mosaic virus is a member ofthe tobamovirus group that infects crucifers.
  • a partial RMV cD ⁇ A containing the 30K subgenomic promoter, 30K ORF, coat subgenomic promoter, coat ORF, and 3 '-end was isolated by RT-PCR using by using oligonucleotides TVCV183X 5'-TAC TCG AGG TTC ATA AGA CCG CGG TAG GCG G-3' (upstream) (SEQ ID NO: 30) and TVCV Kpnl 5*-CGG GGT ACC TGG GCC CCT ACC CGG GGT TTA GGG AGG-3' (downstream) (SEQ ID NO: 31), and subcloned into the EcoRV site of KS+, creating plasmid KS+ TVCV #23 (FIGURE 14).
  • the RMV cDNA was characterized by restriction mapping and dideoxy nucleotide sequencing.
  • the partial nucleotide sequence is as follows: 5'- ctcgaggttcataagaccgcggtaggcggagcgtttgtttactgtagtataattaaatatttgtcagataaaggttgttt aaagatttgttttttgtttgactgagtcgataATGTCTTACGAGCCTAAAGTTAGTGACTTC CTTGCTCTTACGAAAAAGGAGGAAATTTTACCCAAGGCTTTGACGAGA TTAAAGACTGTCTCTATTAGTACTAAGGATGTTATATCTGTTAAGGAG TCTGAGTCCCTGTGATATTGATTTGTTAGTGAATGCCATTAGATA AGTATAGGTATGGGTGTTTTGGGTGTTGTTTTC ACCGGTGAATGGC TGGTACCGGATTTCGTTAAAGGTGGGGTAACAGTGAGCGTGATT
  • the 1543 base pair from the partial RMV cDNA was compared (PCGENE) to oilseed rape mosaic virus (ORMV).
  • ORMV oilseed rape mosaic virus
  • the nucleotide sequence identity was 97.8%.
  • the RMV 30K and coat ORF were compared to ORMV and the amino acid identity was 98.11% (30K) and 98.73% (coat), respectively.
  • RMV cDNA containing the 5 '-end and part ofthe replicase was isolated by RT-PCR from RMV RNA using by using oligonucleotides RGMV1 5'"GAT GGC GCC TTA ATA CGA CTC ACT ATA GTT TTA TTT TTG TTG CAA CAA CAA C-3' (upstream) (SEQ ID NO: 33) and RGR 132 5'-CTT GTG CCC TTC ATG ACG AGC TAT ATC ACG-3' (downstream) (SEQ ID NO: 34).
  • the RMV cDNA was characterized by dideoxy nucleotide sequencing.
  • the partial nucleotide sequence containing the T7 RNA polymerase promoter and part ofthe RMV cDNA is as follows: 5'- ccttaatacgactcactataGTTTTATTTTTGTTGCAACAACAACAACAAATTACAA TAACAACAAAACAAATACAAACAACAACAACAACATGGCACAATTTCAAC AAACAGTAAACATGCAAACATTGCAGGCTGCCGCAGGGCGCAACAGC CTGGTGAATGATTTAGCCTCACGACGTGTTTATGACAATGCTGTCGAG GAGCTAAATGCACGCTCGAGACGCCCTAAGGTTCATTACTCCAAATCA GTGTCTACGGAACAGACGCTGTTAGCTTCAAACGCTTATCCGGAGTTT GAGATTTCCTTTACTCATACCCAACATGCCGTACACTCCCTTGCGGGT GGCCTAAGGACTCTTGAGTTAGTATCTCATGATGCAAGTTCCGTTC GGTTCTCTGACGTACGACATCGGTGGTAACTTTGCAGCGCACCTTTTC AAAGGA
  • the uppercase letters are nucleotide sequences from RMV cDNA.
  • the lower case letters are nucleotide sequences from T7 RNA polymerase promoter.
  • the nucleotide sequences from the 5' and 3' oligonucleotides are underlined.
  • Full length infectious RMV cDNA clones were obtained by RT-PCR from
  • RMV RNA using by using oligonucleotides RGMV1 5'"GAT GGC GCC TTA ATA CGA CTC ACT ATA GTT TTA TTT TTG TTG CAA CAA CAA C- 3' (upstream) (SEQ ID NO: 36) and RG1 APE 5'-ATC GTT TAA ACT GGG CCC CTA CCC GGG GTT AGG GAG G-3' (downstream) (SEQ ID NO: 37).
  • the RMV cDNA was characterized by dideoxy nucleotide sequencing.
  • the partial nucleotide sequence containing the T7 RNA polymerase promoter and part of the RMV cDNA is as follows: 5'-
  • the uppercase letters are nucleotide sequences from RMV cDNA.
  • the nucleotide sequences from the 5' .and 3' oligonucleotides are underlined.
  • Full length infectious RMV cDNA clones were obtained by RT-PCR from RMV RNA using oligonucleotides
  • ABRC Biological Resource Center
  • the cDNA inserts in the CD4-13 were recovered by digestion with NotI. Digestion with NotI in most cases liberates the entire Arabidopsis thaliana cD ⁇ A insert because the original library was assembled with NotI adapters. NotI is an 8-base cutter that infrequently cleaves plant D ⁇ A.
  • the pBS735 transcription plasmid (FIGURE 15) was digested with PacVXhol and ligated to an adapter D ⁇ A sequence created from the oligonucleotides 5'-TCGAGCGGCCGCAT-3' (SEQ ID NO: 41) and 5'-
  • the resulting plasmid pBS740 (FIGURE 16) contains a unique NotI restriction site for bidirectional insertion of NotI fragments from the CD4-13 library. Recovered colonies were prepared from these for plasmid minipreps with a Qiagen BioRobot 9600®. The plasmid D ⁇ A preps performed on the BioRobot 9600® are done in 96-well format and yield transcription quality D ⁇ A.
  • An Arabidopsis cD ⁇ A library was transformed into the plasmid and analyzed by agarose gel electrophoresis to identify clones with inserts. Clones with inserts may be transcribed in vitro .and inoculated onto N benthamiana .and/or Arabidopsis thaliana. Selected leaf disks from transfected plants may be then taken for biochemical analysis.
  • EXAMPLE 14 Expression and targeting to the chloroplasts of a green fluorescent protein in Arabidopsis thaliana via a recombinant viral nucleic acid vector.
  • the gene encoding green fluorescent protein (GFP) was fused at the ⁇ - terminus to the chloroplast transit peptide (CTP) sequence of RuBPCase to create plasmid pBS723 (FIGURE 17).
  • Plasmid pBS723 was modified by PCR mutagenesis to create a unique Pad site upstream ofthe ATG start codon ofthe CTP-GFP gene fusion.
  • the PCR .amplification product obtained from plasmid pBS723 was digested PacllSall and cloned into plasmid GFP-30B/clone 60 (also digested with PacllSall) to create plasmid pBS731 (FIGURE 18).
  • Plasmid pBS731 was linearized at a unique Kpnl restriction site and transcribed into infectious R ⁇ A with T7 R ⁇ A polymerase according to standard procedures. Infectious RNA transcripts that were inoculated onto Nicotiana benthamiana plants showed systemic expression in the upper leaves of CTP-GFP within six days.
  • RNA transcripts from plasmid pBS731 were harvested by grinding the leaves with a mortar and pestle to obtain recombinant virions derived from pBS731 infectious RNA transcripts.
  • Virions from pBS731 were inoculated onto Arabidopsis thaliana leaves.
  • the inoculated leaves of Arabidopsis thaliana plants showed strong green fluorescence under UV light, thus indicating successful expression ofthe CTP-GFP reporter gene.
  • Inoculation of subject organisms such as plants may be effected by using means of high throughput robotics.
  • Arabidopsis thaliana were grown in microtiter plates such as the standard 96-well and 384- well micro titer plates.
  • a robotic handling arm then moved the plates containing the organism to a colony picker or other robot that may deliver inoculations to each plant in the well.
  • inoculation was performed in a very high speed and high throughput manner. It is preferable in the case of plants that the organism be a germinating seed at least in the development cycle to enable access to the cells to be transfected.
  • Equipment used for automated robotic production line could include, but not be limited to, robots of these types: electronic multichannel pipetmen, Qiagen BioRobot 9600®, Robbins Hydra liquid handler, Flexys Colony Picker, New Brunswick automated plate pourer, GeneMachines HiGro shaker incubator, New Brunswick floor shaker, three Qiagen BioRobots, MJ Research PCR machines (PTC-200, Tetrad), ABI 377 sequencer and Tecan Genesis RSP200 liquid handler.
  • robots of these types electronic multichannel pipetmen, Qiagen BioRobot 9600®, Robbins Hydra liquid handler, Flexys Colony Picker, New Brunswick automated plate pourer, GeneMachines HiGro shaker incubator, New Brunswick floor shaker, three Qiagen BioRobots, MJ Research PCR machines (PTC-200, Tetrad), ABI 377 sequencer and Tecan Genesis RSP200 liquid handler.
  • Genomic DNA library construction in a recombinant viral nucleic acid vector Genomic DNA represented in BAC (bacterial artificial chromosome) or
  • YAC (yeast artificial chromosome) libraries may be obtained from the Arabidopsis Biological Resource Center (ABRC).
  • the BAC/YAC DNA can be mechanically size- fractionated, ligated to adapters with cohesive ends, and shotgun-cloned into recombinant viral nucleic acid vectors.
  • mechanically size-fractionated genomic DNA can be blunt-end ligated into a recombinant viral nucleic acid vector.
  • Recovered colonies can be prepared for plasmid minipreps with a Qiagen BioRobot 9600®.
  • the plasmid DNA preps done on the BioRobot 9600® may be assembled in 96-well format and yield transcription quality DNA.
  • the recombinant viral nucleic acid; 'Arabidopsis genomic DNA library may be analyzed by agarose gel electrophoresis (template quality control step) to identify clones with inserts. Clones with inserts can then be transcribed in vitro and inoculated onto N benthamiana and/or Arabidopsis thaliana. Selected leaf disks from transfected plants can then be taken for biochemical analysis.
  • Genomic D ⁇ A from Arabidopsis typically contains a gene every 2.5 kb (kilobases) on average. Genomic D ⁇ A fragments of 0.5 to 2.5 kb obtained by random shearing of D ⁇ A were shotgun assembled in a recombinant viral nucleic acid expression/knockout vector library. Given a genome size of Arabidopsis of approximately 120,000 kb, a random recombinant viral nucleic acid genomic D ⁇ A library would need to contain minimally 48,000 independent inserts of 2.5 kb in size to achieve IX coverage of ' the Arabidopsis genome.
  • a random recombinant viral nucleic acid genomic D ⁇ A library would need to contain minimally 240,000 independent inserts of 0.5 kb in size to achieve IX coverage ofthe Arabidopsis genome.
  • Assembling recombinant viral nucleic acid expression/knockout vector libraries from genomic D ⁇ A rather than cD ⁇ A has the potential to overcome known difficulties encountered when attempting to clone rare, low-abundance mR ⁇ A's in a cD ⁇ A library.
  • a recombinant viral nucleic acid expression/knockout vector library made with genomic D ⁇ A would be especially useful as a gene silencing knockout library.
  • DHSPES expression/knockout vector library made with genomic D ⁇ A would be especially useful for expression of genes lacking introns.
  • other plant species with moderate to small genomes e.g. rose, approximately 80,000 kb
  • a recombinant viral nucleic acid expression/knockout vector library could be made from existing BAC/YAC genomic D ⁇ A or from newly-prepared genomic D ⁇ A for any plant species.
  • a recombinant viral nucleic acid expression/knockout vector library could be made with genomic DNA obtained from yeast, bacteria, or animals including humans.
  • Genomic DNA or cDNA library construction in a DHSPES vector and fransfection of individual clones from said vector library onto T-DNA tagged or transposon tagged or mutated plants.
  • Genomic DNA or cDNA library construction in a recombinant viral nucleic acid vector, and fransfection of individual clones from the vector library onto T-DNA tagged or transposon tagged or mutated plants may be performed according the procedure set forth in Example 16.
  • Such a protocol may be easily designed to complement mutations introduced by random insertional mutagenesis of T-DNA sequences or transposon sequences.
  • Clone B is one ocytotoxic T lymphocyte (CTL) cell clone shown to recognize an epitope present in both the P. yoelii and P. berghei CS proteins. Clone B recognizes the following amino acid sequence; SYVPSAEQILEFVKQISSQ (SEQ ID NO: 43) and when adoptively fransfened to mice protects against infection from both species of malaria sporozoites. Construction of a genetically modified tobamovirus designed to carry this malarial CTL epitope fused to the surface of virus particles is set forth herein.
  • PBGC215 was formed by cloning the 0.15 kb Accl-Nsil fragment of pBGC214 into pBGC235. The 0.9 kb Ncol-Kp ⁇ l fragment from pBGC215 was cloned in pBGC152 to form pBGC216.
  • a 0.07 kb synthetic fragment was formed by annealing PYCS.2p with PYCS.2m and the resulting double stranded fragment, encoding the P. yoelii CTL malarial epitope, was cloned into the -4vrII site of pBGC215 made blunt ended by treatment with mung bean nuclease and creating a unique -4 ⁇ tII site, to form pBGC262.
  • a 0.03 kb synthetic -4 ⁇ tII fragment was formed by annealing TLS.1EXP with TLS.1EXM and the resulting double stranded fragment, encoding the leaky-stop sequence and a stuffer sequence used to facilitate cloning, was cloned into Aaill digested ⁇ BGC262 to form pBGC263.
  • PBGC262 was digested with-4 ⁇ tll and ligated to itself removing the 0.02 kb stuffer fragment to form pBGC264.
  • the 1.0 kb Ncol-Kpnl fragment of pBGC264 was cloned into pSNC004 to form pBGC289.
  • the virus TMV289 produced by transcription of plasmid pBGC289 in vitro contains a leaky stop signal resulting in the removal of four amino acids from the C terminus ofthe wild type TMV coat protein gene and is therefore predicted to synthesize a truncated coat protein and coat protein with a CTL epitope fused at the C terminus at a ratio of 20: 1.
  • the recombinant TMVCP/CTL epitope fusion present in TMV289 is with the stop codon decoded as the amino acid Y (amino acid residue 156).
  • the amino acid sequence ofthe coat protein of virus TMV216 produced by transcription ofthe plasmid pBGC216 in vitro is truncated by four amino acids.
  • the epitope SYVPSAEQILEFVKQISSQ (SEQ ID NO: 43) is calculated to be present at approximately 0.5% ofthe weight ofthe virion using the same assumptions confirmed by quantitative ELISA analysis.
  • Sample ID No. TMV289.1 lBla Fifteen tobacco plants were grown for 33 days post inoculation accumulating 595 g fresh weight of harvested leaf biomass not including the two lower inoculated leaves. Purified Sample ID No. TMV289.11B2 was recovered (383 mg) at a yield of 0.6 mg virion per gram of fresh weight. Therefore, 3 g of 19-mer peptide was obtained per gram of fresh weight extracted. Tobacco plants infected with TMV289 accumulated greater than 1.4 micromoles of peptide per kilogram of leaf tissue.
  • TMV289 Partial confirmation ofthe sequence ofthe epitope coding region of TMV289 was obtained by restriction digestion analysis of PCR amplified cDNA using viral RNA isolated from Sample ID No. TMV289.11B2. The presence of proteins in TMV289 with the predicted mobility ofthe cp fusion at 20 kD and the truncated cp at 17.1 kD was confirmed by denaturing polyacrylamide gel electrophoresis.
  • EXAMPLE 19 Identification of nucleotide sequences involved in the regulation of plant growth by cvtoplasmic inhibition of gene expression using viral derived RNA.
  • Antisense RNA has been used to down regulate gene expression in transgenic and transfected plants. The effectiveness of antisense on the inhibition of eukaryotic gene expression was first demonstrated by Izant et al. (Cell 36(4): 1007-1015 (1984)). Since then, the down-regulation of numerous genes from transgenic plants has been reported. In addition, there is evidence that "co- suppression" of genes occurs in transgenic plants containing sense RNA by readthrough transcription from distal promoters located on the opposite strand of the DNA (Van der Krol et al, Plant Cell 2(4):291-299 (1990) and Napoli et al, Plant Cell 2:279-289 (1990)).
  • RNA viral vectors which replicate solely in the cytoplasm, can be used to identify genes involved in the regulation of plant growth by inhibiting the expression of specific endogenous genes. This example will enable one to characterize specific genes and biochemical pathways in transfected plants using an RNA viral vector.
  • Tobamoviral vectors have been developed for the heterologous expression of uncharacterized nucleotide sequences in transfected plants.
  • a partial Arabidopsis thaliana cDNA library was placed under the transcriptional control of a tobamovirus subgenomic promoter in a RNA viral vector. Colonies from transformed E. coli were automatically picked using a Flexys robot and transfened to a 96 well flat bottom block containing terrific broth (TB) Amp 50 ug/ml. Approximately 2000 plasmid DNAs were isolated from overnight cultures using a BioRobot and infectious RNAs from 430 independent clones were directly applied to plants.
  • cDNA encoding Nicotiana benthamiana ADP-ribosylation factor Partial cDNAs from Nicotiana benthamiana leaf RNA may be isolated by polymerase chain reaction (PCR) using the following oligonucleotides: ATARFM1X, 5'-GCC TCG AGT GCA GCA TGG GGT TGT CAT TCG GAA AGT TGT TC-3' (upstream) (SEQ ID NO: 45) and ATARFA181 A, 5'-TAC CTA GGC CTT GCT TGC GAT GTT GTT GGA GAG-3' (downstream) (SEQ ID NO: 46).
  • PCR polymerase chain reaction
  • a full-length cDNA encoding ARF may be isolated by screening a cDNA library by colony hybridization using a 32 P labeled Arabidopsis thaliana ARF PCR product. Hybridization can be carried out at 42°C for 48h in 50% formamide, 5X SSC, 0.02M phosphate buffer, 5X Denhart's solution, and 0.1 mg/ml sheared calf thymus DNA. Filters may be washed at 65°C in 0.1X SSC and 0.1% SDS, prior to autoradiography. PCR products and the ARF cDNA clones may be verified by dideoxynucleotide sequencing.
  • EXAMPLE 20 Identification of nucleotide sequences involved in the regulation of plant development by cvtoplasmic inhibition of gene expression using viral derived RNA.
  • a partial Arabidopsis thaliana cD ⁇ A library was placed under the transcriptional control of a tobamovirus subgenomic promoter in a R ⁇ A viral vector. Colonies from transformed E. coli were automatically picked using a Flexys robot and transfened to a 96 well flat bottom block containing terrific broth (TB) Amp 50 ug/ml. Approximately 2000 plasmid D ⁇ As were isolated from overnight cultures using a BioRobot and infectious R ⁇ As from 430 independent clones were directly applied to plants. One to two weeks after inoculation, transfected Nicotiana benthamiana plants were visually monitored for changes in growth rates, mo ⁇ hology, and color.
  • An Arabidopsis thaliana CD4-13 cDNA library was digested with NotI. D ⁇ A fragments between 500 and 1000 bp were isolated by trough elution and subcloned into the NotI site of pBS740. E. coli C600 competent cells were transformed with the pBS740 AT library and colomes containing Arabidopsis cD ⁇ A sequences were selected on LB Amp 50 ug/ml. Recombinant C600 cells were automatically picked using a Flexys robot and then transfened to a 96 well flat bottom block containing terrific broth (TB) Amp 50 ug/ml. Approximately 2000 plasmid D ⁇ As were isolated from overnight cultures using a BioRobot (Qiagen) and infectious R ⁇ As from 430 independent clones were directly applied to plants.
  • D ⁇ A sequencing and computer analysis A 750 bp NotI fragment of 740 AT #88 containing the G-protein coupled receptor cD ⁇ A was characterized. D ⁇ A sequence of NotI fragment of 740 AT #88 (750 bp) is as follows: 5'-
  • nucleotide sequence analysis and amino acid sequence comparisons were performed using DNA Strider, PCGENE and NCBI Blast programs.
  • the nucleotide sequence from 740 AT #88 was compared to Brassica rapa cDNA L33574 (FIGURE 22), the octopus rhodopsin mRNA X07797 (FIGURE 23).
  • the amino acid sequence derived from 740 AT #88 was compared to an Arabidopsis EST ORF ATTS2938 (FIGURE 24) and octopus rhodopsin P31356 (FIGURE 25).
  • EXAMPLE 21 Identification of nucleotide sequences involved in the regulation of plant growth bv cvtoplasmic inhibition of gene expression using viral derived RNA.
  • Antisense RNA has been used to down regulate gene expression in transgenic and transfected plants.
  • the pu ⁇ ose of this example is again to demonstrate that novel positive strand viral vectors, which replicate solely in the cytoplasm, can be used to identify genes involved in the regulation of plant growth by inhibiting the expression of specific endogenous genes. This example will enable one to characterize specific genes and biochemical pathways in transfected plants using an RNA viral vector.
  • Tobamoviral vectors have been developed for the heterologous expression of uncharacterized nucleotide sequences in transfected plants.
  • a partial Arabidopsis thaliana cDNA library was placed under the transcriptional control of a tobamovirus subgenomic promoter in a RNA viral vector.
  • Colonies from transformed E. coli were automatically picked using a Flexys robot and transfered to a 96 well flat bottom block containing terrific broth (TB) Amp 50 ug/ml.
  • TB terrific broth
  • Approximately 2000 plasmid DNAs were isolated from overnight cultures using a BioRobot and infectious RNAs from 430 independent clones were directly applied to plants.
  • An Arabidopsis thaliana CD4-13 cDNA library was digested with NotI. D ⁇ A fragments between 500 and 1000 bp were isolated by trough elution and subcloned into the NotI site of pBS740. E. coli C600 competent cells were transformed with the pBS740 AT library and colonies containing Arabidopsis cD ⁇ A sequences were selected on LB Amp 50 ug/ml. Recombinant C600 cells were automatically picked using a Flexys robot and then transfered to a 96 well flat bottom block containing terrific broth (TB) Amp 50 ug/ml. Approximately 2000 plasmid D ⁇ As were isolated from overnight cultures using a BioRobot (Qiagen) and infectious R ⁇ As from 430 independent clones were directly applied to plants.
  • DNA sequence of NotI fragment of 740 AT #2441 (350 bp) is as follows: 5'- CTTCACTTTCGCCGATGGCTCTACCTAACCAGCAAACCGTGGATTACC CTAGCTTCAAGCTCGTTATCGTTGGCGATGGAGGCACAGGGAAGACC ACATTTGTAAAGAGACATCTTACTGGAGAGTTTGAGAAGAAGTATGA ACCCACTATTGGTGTTGAGGTTCATCCTCTTGATTTCTTCACTAACTGT GGCAAGATCCGTTTCTACTGTTGGGATACTGCTGGCCAAGAGAAATTT GGTGGTCTTAGGGATGGTTACTACATCCATGGACAATGTGCTATCATC ATGTTTGATGTCACAAGCACGACTGACATACAAGAATGTTCC AACATG GCACCGTGATCTTTG-3' (SEQ ID NO. 48)
  • the nucleotide sequencing of 740 AT #2441 was carried out by dideoxy termination using double stranded templates (Sanger et al, Proc. Natl. Acad. Sci. USA 24(12):5463-5467 (1977)). Nucleotide sequence analysis and amino acid sequence comparisons were performed using DNA Strider, PCGENE and NCBI Blast programs.
  • the nucleotide sequence from 740 AT #2441 was compared to tobacco RAN-Bl GTP binding protein (FIGURE 26).
  • the nucleotide sequence from 740 AT #2441 was compared to human RAN GTP-binding protein (FIGURE 27).
  • EXAMPLE 22 Gene silencing/co-supression of genes induced by delivering an RNA capable of base pairing with itself to form double stranded regions.
  • RNA silencing has been used to down regulate gene expression in transgenic plants. Recent experimental evidence suggests that double stranded RNA may be an effective stimulator of gene silencing/co-suppression phenomenon in transgenic plant. For example, Waterhouse et al. (Proc. Natl. Acad. Sci. USA 95:13959-13964 (1998), inco ⁇ orated herein by reference) described that virus resistance and gene silencing in plants could be induced by simultaneous expression of sense and antisense RNA. Gene silencing/co- suppression of plant genes may be induced by delivering an RNA capable of base pairing with itself to form double stranded regions. This example shows: (1) a novel method for generating an RNA virus vector capable of producing an RNA capable of forming double stranded regions, and (2) a process to silence plant genes by using such a viral vector.
  • Step 1 Construction of a DNA sequence which after it is transcribed would generate an RNA molecule capable of base pairing with itself.
  • Two identical, or nearly identical, ds DNA sequences can be ligated together in an inverted orientation to each other (i.e., in either a head to tail or tail to head orientation) with or without a linking nucleotide sequence between the homologous sequences.
  • the resulting DNA sequence can then be cloned into a cDNA copy of a plant viral vector genome.
  • Step 2 Cloning, screening, transcription of clones of interest using known methods in the art.
  • Step 3 Infect plant cells with transcripts from clones.
  • RNA from foreign gene will base pair upon itself, forming double-stranded RNA regions.
  • This approach could be used with any plant or non-plant gene and used to silence plant gene homologous to assist in identification ofthe function of a particular gene sequence.
  • a full- length cDNA copy ofthe Eastern Equine Encephalomyelitis Virus (EEEV) genome is prepared and inserted into the Pstl site of pUC18 as described by Chang et al, J. Gen. Virol. 68:2129 (1987).
  • the sequence for the viral coat protein and its adjacent El and E2 glycoprotein fransmissibility factors are located on the region conesponding to the 26S RNA region.
  • the vector containing the cDNA copy ofthe EEEV genome is digested with the appropriate restriction enzymes and exonucleases to delete the coding sequence ofthe coat protein and the El and E2 proteins (structural protein coding sequence).
  • the structural protein coding sequence is removed by partial digestion with Mbol, followed by religation to remove a vital portion ofthe structural gene.
  • the vector is cut at the 3 '-end ofthe viral structural gene.
  • the viral DNA is sequentially removed by digestion with Bal31 or Micrococcal SI nuclease up through the start codon ofthe structural protein sequence.
  • the DNA sequence containing the sequence ofthe viral 3 '-tail is then ligated to the remaining 5 '-end.
  • the deletion ofthe coding sequence for the structural proteins is confirmed by isolating EEEV RNA and using it to infect an equine cell culture.
  • the isolated EEEV RNA is found to be non-infective under natural conditions.
  • the coat protein coding sequence is removed by partial digestion with Mbol followed by religation to reattach the 3 '-tail ofthe virus. This will remove a vital portion ofthe coat protein gene.
  • a second alternative method for removing only the coat protein sequence is to cut the vector at the 3 '-end ofthe viral coat protein gene.
  • the viral DNA is removed by digestion with Bal31 or Micrococcal S 1 nuclease up through the start codon ofthe coat protein sequence.
  • the synthetic DNA sequence containing the sequence ofthe 3 '-tail is then ligated to the remaining 5 '-end.
  • the deletion ofthe coding sequence for the coat protein is confirmed by isolating EEEV RNA and using it to infect an equine cell culture.
  • the isolated EEEV RNA is found to be non-infective under natural conditions.
  • Sindbis viruses Methods for genetic manipulation of Sindbis viruses are described in Garoff et al, Curr. Opin. Biotechnol. 9(5):464-9 (1998); Agapov et al, Proc. Natl. Acad. Sci. USA 95(22): 12989-94 (1998); Frolov et al, J. Virol. Apr;71(4):2819-29 (1997), all of which are inco ⁇ orated herein by reference.
  • a full-length cDNA copy ofthe Sindbis virus genome is prepared and inserted into the Sm ⁇ l site of a plasmid derived from pBR322 as described by Lindquist et al, Virology 151:10 (1986).
  • the sequence for the viral coat protein and the adjacent El and E2 glycoprotein fransmissibility factors are located on the region conesponding to the 26S RNA region.
  • the vector containing the cDNA copy of the Sindbis virus genome is digested with the appropriate restriction enzymes and exonucleases to delete the coding sequence for the structural proteins.
  • the structural protein coding sequence is removed by partial digestion with Bi ⁇ l, followed by religation to remove a vital portion ofthe structural gene.
  • the vector is cut at the 3'-end ofthe viral nucleic acid.
  • the viral DNA is removed by digestion with Ba 1 or Micrococcal S 1 nuclease up through the start codon ofthe structural protein sequence.
  • the synthetic DNA sequence containing the sequence ofthe viral 3 '-tail is then ligated to the remaining 5 '-end.
  • the deletion ofthe coding sequence for the structural proteins is confirmed by isolating Sindbis RNA and using it to infect an avian cell culture. The isolated Sindbis RNA is found to be non-infective under natural conditions.
  • the coding sequence for the coat protein is deleted and the sequence for the El and E2 glycoproteins remain in the vector containing the cDNA copy ofthe Sindbis genome.
  • the coat protein coding sequence is removed by partial digestion with Aflll followed by religation to reattach the 3 '-tail ofthe virus.
  • a second alternative method for removing only the coat protein sequence is to cut the vector at the 3 '-end ofthe viral nucleic acid.
  • the viral DNA is removed by digestion with BaB 1 or Micrococcal S 1 nuclease up through the start codon ofthe coat protein sequence (the same start codon as for the sequence for all the structural proteins).
  • the synthetic DNA sequence containing the sequence ofthe 3 '-tail is then ligated to the remaining 5 '-end.
  • the deletion ofthe coding sequence for the coat protein is confirmed by isolating Sindbis RNA and using it to infect an avian cell culture.
  • the isolated Sindbis RNA is found to be non-infective under natural conditions.
  • EXAMPLE 25 Preparation of a Non-Transmissible Western Equine Encephalomyelitis Virus Nucleotide Sequence.
  • the vector containing the cDNA copy ofthe WEEV genome is digested with the appropriate restriction enzymes and exonucleases to delete the coding sequence ofthe coat protein and the El and E2 proteins (structural protein coding sequence).
  • structural protein coding sequence is removed by partial digestion with N ⁇ cI, followed by religation to remove a vital portion ofthe structural protein sequence.
  • the vector is cut at the 3 '-end ofthe structural protein D ⁇ A sequence.
  • the viral D ⁇ A is removed by digestion with -5 ⁇ /31 or Micrococcal SI nuclease up through the start codon ofthe structural protein sequence.
  • the D ⁇ A sequence ofthe viral 3 '-tail is then ligated to the remaining 5 '-end.
  • the deletion ofthe coding sequence for the structural proteins is confirmed by isolating WEEV R ⁇ A and using it to infect a Vero cell culture.
  • the isolated WEEV R ⁇ A is found to be non-infective under natural conditions.
  • only the coding sequence for the coat protein is deleted and the sequence for the El and E2 glycoproteins remain in the vector containing the cD ⁇ A copy ofthe WEEV genome.
  • the coat protein coding sequence is removed by partial digestion with HgiAI followed by religation to reattach the 3 '-tail of the vims.
  • a second alternative method for removing only the coat protein sequence is to cut the vector at the 3'-end ofthe viral coat protein sequence.
  • D ⁇ A is removed by digestion with -5 ⁇ /31 or Micrococcal SI nuclease up through the a vital portion ofthe coat protein sequence.
  • the D ⁇ A sequence containing the sequence ofthe 3'-tail is then ligated to the remaining 5'-end.
  • the deletion ofthe coding sequence for the coat protein is confirmed by isolating WEEV RNA and using it to infect a Vero cell culture.
  • the isolated WEEV RNA is found to be non-infective, i.e., biologically contained, under natural conditions.
  • the VPI coat protein coding sequence is removed by partial digestion with -5 ⁇ mHI nuclease, and then treated with EcoRI, filled in with Klenow enzyme and recircularized.
  • the deletion ofthe coding sequence for the coat protein VPI is confirmed by isolating SV40 RNA and using it to infect simian cell cultures.
  • the isolated SV40 RNA is found to be non-infective, i.e., biologically contained, under natural conditions.
  • Novel requirements for production of infectious viral vector in vitro derived RNA transcripts Novel requirements for production of infectious viral vector in vitro derived RNA transcripts.
  • This example demonstrates the production of highly infectious viral vector transcripts containing 5' nucleotides with reference to the vims vector. Construction of a library of subgenomic cDNA clones of TMV and BMV has been described in Dawson et al, Proc. Natl. Acad. Sci. USA 83:1832-1836
  • Nucleotides were added between the transcriptional start site ofthe promoter for in vitro transcription, in this case T7, .and the start ofthe cDNA of TMV in order to maximize transcription product yield and possibly obviate the need to cap vims transcripts to insure infectivity.
  • the relevant sequence is the T7 promoter ...TATAG ⁇ TATTTT.... where the ⁇ indicates the base preceding is the start site for transcription and the bold letter is the first base ofthe TMV cDNA.
  • Three approaches were taken: 1) addition of G, GG or GGG between the start site of transcription and the TMV cDNA ( ... TATAGGTATTT...
  • the GTN would allow the mimicking of two potential sites for initiation, the added and the native sequence, and facilitate more ready mis-initiation of transcription in vivo to restore the native TMV cDNA sequence.
  • Approaches included cloning GFP expressing TMV vector sequences into vectors containing extra G, GG or GGG bases using standard molecular biology techniques.
  • full length PCR of TMV expression clone 1056 was done to add N2, N3 and GTN bases between the T7 promoter and the TMV cDNA. Subsequently, these PCR products were cloned into pUC based vectors.
  • Capped and uncapped transcripts were made in vitro .and inoculated to tobacco protoplasts or Nicotiana benthamiana plants, wild type and 30k expressing transgenics.
  • the results are that an extra G, ... TATAGGTATTTT..., or a GTC, ... TATAGTCGTATTTT..., were found to be well tolerated as additional 5' nucleotides on the 5' of TMV vector RNA transcripts and were quite infectious on both plant types and protoplasts as capped or non-capped franscripts. Other sequences may be screened to find other options.
  • infectious transcripts may be derived with extra 5' nucleotides.
  • the replicated progeny may exhibit the results of reversion events that would yield the wild type vims 5' vims sequence, but may include portions or entire sets of introduced additional base sequences.
  • This strategy can be applied to a range of RNA vimses or RNA viral vectors of various genetic anangements derived from wild type vims genome. This would require the use of sequences particular to that ofthe vims used as a vector.
  • TMV-based vims expression vectors were initially used in these studies pBTI 1056 which contains the T7 promoter followed directly by the vims cDNA sequence f... TAT AGT ATT).
  • pBTI SBS60-29 which contains the T7 promoter (underlined) followed by an extra guanine residue then the vims cDNA sequence (...TATAGGTATT).
  • Both expression vectors express the cycle 3 shuffled green fluorescent protein (GFPc3) in localized infection sites and systemically infected tissue of infected plants. Transcriptions of each plasmid were carried out in the absence of cap analogue (uncapped) or in the presence of 8-fold greater concentration of RNA cap analogue than rGTP (capped).
  • Nicotiana tabacum protoplasts were infected with either capped or uncapped transcriptions (as described above) of pBTI SBS60 which contains the T7 promoter followed directly by the vims cDNA sequence ( " TATAGTATT). This expression vector also expresses the GFPc3 gene in infected cells and tissues. Nicotiana tabacum protoplasts were transfected with 1 mcl of each transcriptions. Approximately 36 hours post infection fransfected protoplasts were viewed under UV illumination and cells showing GFPc3 expression. Approximately 80% cells transfected with the capped PBTI SBS60 transcripts showed GFP expression while 5% of cells transfected with uncapped franscripts showed GFP expression.
  • the expression ofthe 30K movement protein of TMV in transgenic plants also has the unexpected effect of equalizing the relative specific infectivity of uncapped verses capped franscripts.
  • the mechanism behind this effect is not fully understood, but could arise from the RNA binding activity ofthe movement protein stabilizing the uncapped transcript in infected cells from prereplication cytosolic degradation.
  • TMV-based vims expression vector pBTI 1056 GTN#28 which contains the T7 promoter (underlined) followed GTC bases (bold) then the vims cDNA sequence (...TATAGTCGTATT).
  • This expression vector expresses the cycle 3 shuffled green fluorescent protein (GFPc3) in localized infection sites and systemically infected tissue of infected plants.
  • GFPc3 cycle 3 shuffled green fluorescent protein
  • This vector was transcribed in vitro in the presence (capped) and absence (uncapped) of cap analogue. Transcriptions were mixed with abrasive and inoculated on expanded older leaves of a wild type Nicotiana benthamiana (Nb) plant and a Nb plant expressing a TMV Ul 30k movement protein transgene (Nb 30K).
  • Elicitor recognition and the response cascades occurring in plants form an essential link between the environmental stress and plant survival responses.
  • Many products are induced following induction by environmental stimuli or pathogen infection, which include, but are not limited to, proteases, protease inhibitors, alkaloids and other metabolites. Glazebrook, et al, Annu. Rev. Gen. 31:547-569 (1997); Grahm, et al, J. Biol. Chem. 260:6555-6560 (1985); and Ryan, et al, Ann. Rev. Cell Dev. Biol. 14:1-17 (1998), all inco ⁇ orated herein by reference. The components ofthe recognition and response pathways are poorly understood, yet have tremendous practical value for input traits in genetically improved crops.
  • Vims expression vectors capable of either overexpressing gene products or suppressing the expression of particular endogenous host genes provide a unique tool to discover the nature of the genes whose products contribute to the response pathways.
  • This example describes methods for inhibiting endogenous plant proteases which interfere with the expression and purification of recombinant proteins in plants.
  • this example shows methods for inhibiting proteolytic activity in planta which is responsible for the degradation of a viral vector- expressed recombinant protein.
  • These methods are also applicable to the protection of recombinant proteins expressed via a stable transformation system or endogenous plant proteins.
  • Viral vectors have been constructed to include an N-terminal signal peptide sequence. This sequence directs the recombinant protein through the secretory pathway to the cell surface and ultimately accumulating in the plant intercellular fluid (IF) (Kermode, Critical Reviews in Plant Sciences 5(4):285-423 (1996), inco ⁇ orated herein by reference).
  • IF plant intercellular fluid
  • the target protein was cleaved abenantly in vivo.
  • Three examples include a mammalian growth hormone and single chain antibody and an avian interferon.
  • In vivo residence time in the IF led to the accumulation ofthe cleavage product(s) as detected by immunoblotting. Cleavage was either complete in vivo or continued in vitro following IF extraction (Co-pending U.S. Patent Application Serial No.
  • transgenic plants and virally- expressed proteins Another possible approach is to combine transgenic plants and virally- expressed proteins.
  • JA jasmonic acid
  • JA is produced as part of a general plant defense mechanism and is known to induce specific proteinase inhibitors (Lightner et al, JMol Gen Genet. 241:595-601 (1993), inco ⁇ orated herein by reference).
  • Exogenous application of JA as been used to induce a plant defense response in Nicotiana attenuata to against herbivore attack (Baldwin, PNAS, 95(14):8113-8118 (1998), inco ⁇ orated herein by reference).
  • JA jasmonic acid
  • JA is produced as part of a general plant defense mechanism and is known to induce specific proteinase inhibitors (Lightner et al, JMol Gen Genet. 241:595-601 (1993), inco ⁇ orated herein by reference).
  • Exogenous application of JA as been used to induce a plant defense response in Nicotiana attenuata to against herbivore attack (Baldwin, PNAS,
  • the desired phenotype in host plants used for gene discovery program using vims expression vectors is reduced proteolytic activities in the cytosol, secretory pathway or apoplast so to increase the half-life of virally produced proteins. This will allow virally expressed proteins to exert their influence on plant biochemistry, development and growth optimally. Rapid or premature degradation may reduce the amount ofthe expressed protein below the necessary threshold to exert a measurable effect.
  • Transgenic expression of protease inhibitors such as those induced by the systemin pathway (Ryan, et al, Ann. Rev. Cell Dev. Biol. 14:1-17 (1998)), will provide a continuous source of inhibitor to slow particular degradation processes.
  • EXAMPLE 30 Selection of optimized RNA and protein activities by use of vims vectors to express libraries of sequence variants generated by means of in vitro mutagenenisis and/or recombination.
  • DNA shuffling is a process for recursive mutation and in vitro recombination, performed by random fractionation .and re-assembly of a gene of interest to generate a pool of related, yet not identical, gene sequences.
  • Fractionation occurs through the treatment of DNA sequences with limiting amounts of nuclease and re-assembly typically requires two steps, first primerless PCR to re-align fragments based on local homology and then primer driven PCR to recover full length assembled fragments.
  • DNA shuffling has been successfully applied to prokaryotic or cell-based systems to select sequences of desired protein activities.
  • the ability to introduce shuffled sequences throughout an organism in a rapid and high throughput manner necessary to harness the full potential of this technology has not been demonstrated.
  • the properties confened by the selected shuffled sequences were demonstrated to be inherited by progeny vimses.
  • Two aspects that must be continually improved in vims expression vectors are: 1) their ability to move in a facile manner both locally and systemically in plants, and 2) the need for greater levels of foreign gene expression.
  • Both of these functions can potentially be affected by modifications to the 30 kDa ORF.
  • Functions within the 30 kDa coding region include the movement protein (MP), the vims origin of virion assembly and the subgenomic promoter used for coat protein synthesis. This is the promoter used for expression of foreign gene sequences in most tobamovirus vectors. It has been demonstrated that natural variation in viral populations can be the substrates for selection of improved characters in viral vectors can lead to dramatic improvements in their performance. This work further showed that single or multiple amino acid substitutions in the 30 kDa ORF can significantly effect the movement properties of vims vectors.
  • Vimses function genomically, as an integrated whole of RNA and protein sequences, suggesting that either individual elements, such as the 30 kDa ORF, or the entire plant vims genomes could be subjected to shuffling so to improve plant vims vector performance.
  • Obvious following the application of shuffling in this context is the use of plant vims vectors to house shuffled foreign gene populations which, following inoculation onto plants, gene products with optimized activities can be selected.
  • Plant vims vectors are the ultimate tool for shuttling genes into plants for selection of optimized activities. No other tool, transient or stable expression methods, can match the ability of plant vims vectors to develop optimized genes for plant activities.
  • p30B GFP vector is the TMV Ul infectious cDNA (bases 1-5756) containing the 5' NTR, replicase genes (126 and 183 kDa proteins), movement protein gene with associated subgenomic promoter and an RNA leader derived from the Ul coat protein gene. Following the RNA leader is a unique Pad site and the green fluorescent protein (GFP) gene. Following a unique Xhol site, the clone continues with a portion ofthe TMV Ul 3' NTR followed by a subgenomic promoter, coat protein gene and 3' NTR from TMV U5 strain.
  • GFP green fluorescent protein
  • the first stage ofthe project required the construction of a vector into which shuffled DNA fragments could be reintroduced.
  • the polymerase chain reaction (PCR) was used to amplify a DNA fragment from the TMV vector p30B comprising the T7 promoter, 5' non-translated region (NTR), and the reading frames for the 126 and 183 kDa replicase proteins.
  • the 5' primer covered the T7 promoter and initial bases ofthe TMV genome while the second primer modified the context surrounding the start codon for the 30 kDa MP of TMV. This allowed DNA fragments to be ligated into the modified vector, designated 3 OB GFP d30K, as vrll, Pa restriction endonuclease digested fragments.
  • This modification allowed the ready insertion of modified 30 kDa gene fragments into a vims vector and have them expressed in plant cells, tissues or systemically.
  • the wild type GFP ORF is the reporter gene since the visual level of fluorescence as observed under long wave UV light conelates directly with levels of GFP protein present in plant tissues. This has been demonstrated by looking at different vims vectors expressing GFP, each having different strength subgenomic promoters, that were infected in plants and GFP levels determined by UV fluorescence and Western blotting using anti-GFP antibodies.
  • the procedure for shuffling ofthe 30 kDa gene is similar to that described by Crameri et al, Nature Biotech. 15:436 (1997), and contained the following steps.
  • the 30 kDa gene fragment also containing the coat protein RNA leader was amplified from tobamoviras expression vectors using primers: TMVUl 3 OK 5'A (5'-GGCCCTAGGATGGCTCTAGTTGTTAAAGG-3') (SEQ ID NO: 49) and 3-5' Pac primer (5'-GTTCTTCTCCTTTGCTAGCCATTTAATTAATGAC- 3') (SEQ ID NO: 50).
  • the PCR DNA product was gel isolated and then incompletely digested with DNasel.
  • DNA fragments of 500 bp or smaller were isolated by using DEAE blotting paper technique and then eluted. Purified DNA fragments were mixed together with taq DNA polymerase and allowed to "reassemble” for 40 cycles. "Reassembly” reaction was assayed by gel electrophoresis for DNA bands of approximately 800-850 bp. Approximately 1 mcl ofthe "reassembly” reaction was then subjected to PCR using primers TMV Ul 30K 5'A and 3-5' Pac that hybridize to terminal DNA ends of reassembled fragments.
  • the reassembled fragments will be gel isolated and digested with restriction enzymes Avrll and Pad (sites present in the terminal primers) to allow for facile cloning back into the p30B d30k ANP digested with -4vrII and Pad.
  • Ligations of shuffled genes into p30B d30k ANP resulted in pooled libraries of sequences containing 100 to 50,000 members in five separate experiments. Pooled vims vectors with libraries of variant 30 kDa coding regions were transcribed with T7 RNA polymerase and then inoculated by standard PEG fransfection into 0.5 x 10 6 Nicotiana tabacum protoplasts per sample.
  • the protoplast extracts were then inoculated on leaves of wild type and transgenic Nicotiana tabacum c.v. MD609 expressing the TMV Ul 30 kDa movement protein.
  • Three to five days post inoculation localized infection sites were observed expressing GFP.
  • a variety of intensities of GFP fluorescence were observed varying from that observed with the wild type GFP gene to much duller to very bright, as observed from the viral expression ofthe shuffled GFP gene of Crameri et al, Nature Biotech. (1996) (GFPc3).
  • the occurrence of vimses expressing enhanced GFP fluorescence varied between libraries tested from 1/200 to 1/50 infection foci depending on libraries tested.
  • the protoplast extracts isolated from transfections with vims libraries were inoculated on one half of wild type Nicotiana tabacum c.v. MD609 and Nicotiana benthamiana leaves. To the other leaf half, vims derived from p30B GFP was inoculated. Some infection sites resulting from infection of vimses containing shuffled 30 kDa ORFs grew more rapidly than those ofthe average from p30B GFP. These events occurred at a frequency of 1/100 to 1/500 infection foci depending on the vims library analyzed. These more rapidly growing infection foci were excised and inoculated on young Nicotiana tabacum c.v. MD609 plants.
  • p30B GFP was inoculated on similar sized and aged plants.
  • the p30B GFP vector does not move systemically on tobacco plants.
  • some shuffled 30 kDa ORF variant vectors that were identified as rapidly growing local infection sites, were able to move systemically on tobacco plants. The movement was primarily on phloem source tissue and were localized to veins and circular spots in green lamina. This movement ability was reproducible in multiple inoculations of these individual vims variants.
  • Sequence analysis ofthe vimses containing shuffled 30 kDa ORFs capable of systemic movement on Nicotiana tabacum plants demonstrated that localized amino acid substitutions were present and responsible for altered movement phenotype.
  • the phenotypes that can be assayed are protein activity in planta, as with the movement activities ofthe 30 kDa protein, enzyme activities in planta or in plant extracts or other surrogate features such as substrate or product accumulation.
  • These data demonstrate the power of vims expression vectors to be effective tools for shuttling sequence variants into plants and allow the selection of genes encoding the desired altered property. This tool allows one to mine the hidden activities, enhance the isolated activities of enzymes or eliminate allosteric inhibition of enzyme activities. This could be applied to any plant gene or genes from other sources to optimize the activities desired for agronomic, pharmaceutical or developmental effects caused by altered genes.
  • EXAMPLE 31 Composite clomng to facilitate cloning of libraries in vims vectors and/or their introduction into host cells for expression of sequences.
  • Vims vector clones could be integrated into lambda phage or cosmid clones to facilitate library constmction, clone representation, elimination of cell based amplification by direct transcription and archiving of individual clones.
  • cis-acting elements allowing for expression in plant cells or integration into plant DNA could be included into such plasmids to facilitate inoculation of DNA for direct expression, obviating the need for transcription of vector cDNA, or construction of dedicated plant transformation vectors.
  • Vims vectors are tools housing libraries of sequences that can be screened for novel gene discovery. However libraries are often first constructed in plasmid or phage shuttle vectors before excising and introduction into vims vectors. Likewise, sequences can be screened in hosts using vims vectors, but must be subcloned into appropriate eukaryotic expression vectors before the trait identified in the vector transfected host will become a stable trait in the host by gene integration. Additional hurdles to overcome are: (1) constmction of libraries to most efficiently represent the clones in a cDNA library, (2) obtaining maximal fransfection efficiency into bacterial hosts (if used), and (3) archiving DNA samples without the need for fransfection into bacteria and transcription of ligated DNA.
  • a virus vector into a cosmid clone, or lambda phage itself, (both termed phagmids here) could allow a multi-pu ⁇ ose vector to be generated to be both the repository of primary generated library sequences, source for ligation transcriptions, high efficiency bacterial fransfection and direct expression in higher eukaryotic hosts.
  • a non symmetrical restriction such as BstXI: CCANNNNNNTGG
  • the 3' part ofthe vector will be inserted into another arm with a non-symmetrical restriction (such as BstXI: CCANNNNNNTGG) containing a second unique sticky sequence (the N's).
  • the vector would be split at the determined restriction site (e.g. BstXI) within the site for foreign sequence expression in the vims vector.
  • the 5'-end ofthe vims cDNA would be appropriately fused to a promoter for in vitro transcription (e.g. T7) or for in vivo expression (e.g. an appropriate higher eukaryotic RNA polymerase promoter).
  • the 3'-end ofthe vims cDNA would terminate with a ribozyme for in vitro cleavage and/or a 3' terminator from a gene from host organism to lead to in vivo termination of transcription.
  • Left and right T-DNA borders that promote the integration of sequences in between into plant genomic DNA, could flank the promoter and terminator sequences.
  • At the terminus of each arm would be cos sequences to allow complete regeneration ofthe phagmid upon ligation in the presence of foreign library DNA containing the two unique sticky sequences at each respective termini.
  • These library DNA fragments could be generated by PCR amplification using determined restriction sites (e.g., BstXI) to generate unique sticky ends complementary to those in the phagmid-vector arms integrated in the PCR primers.
  • the 5' and 3' primers would each have unique recognition sequences in the BstXI restriction site (the N's) that would match the sticky sites on the respective sides ofthe vims vector.
  • the sites could be switched on a second set of PCR primers to allow the amplification of DNA to be ligated into the phagmid-viral vector arms in the "sense" and "anti-sense” orientation.
  • These constructions would allow for efficient in vitro ligation and use of cmde ligation mix as template for E. coli transformation, plant transformation, in vitro lambda packaging to 10 9 pfu/mcg or in vitro transcription. In this manner, the vector and flexibility for its screening could be maximized.
  • the goal of this example is to improve the host plant by introducing foreign genetic material via interspecific hybridization.
  • Host plant species vary in their ability to support expression of a sequence inserted into a plant viral vector. Some species support expression to a high specific activity, such as
  • Nicotiana benthamiana but have relatively low biomass.
  • Other species such as N tabacum, have high biomass and/or other desirable properties for growth in the field, but have a relatively low specific activity ofthe expressed sequence.
  • the desirable properties of two or more species are combined by making an interspecific hybrid by standard methods. After chromosome doubling to restore fertility, the primary hybrid may have suitable properties, or it may be desirable to backcross toward either parent selecting or screening at each generation for the desired property(ies) ofthe non-recunent parent, for example, infrogress the superior biomass of N tabacum into N benthamiana, or infrogress the superior viral vector performance of N benthamiana into N tabacum, among others.
  • a viral vector expressing the green fluorescent protein (GFP) is one example of a useful tool for screening the level of systemic expression in candidate hybrid plants.
  • EXAMPLE 33 Libraries of heterologous nucleic acid sequences in DHSPES constructs generated in a restriction-endonuclease-free and cell-free manner.
  • the goal of this example is to generate libraries of DHSPES constmcts containing heterologous sequences while avoiding the potential problems associated with the use of restriction enzymes for preparation ofthe inserted nucleic acids and with passage ofthe resultant constructions through E. coli.
  • DNA fragments are generated by restriction endonuclease treatment and ligated into a DHSPES vector with compatible termini.
  • libraries of DHSPES constructs harboring cDNA molecules in a restriction endonuclease-free and E. co/t-free manner are constructed. Such a system will permit the inclusion into DHSPES constmcts of molecules that harbor inconvenient internal restriction sites. This method of "cell-free cloning" will also allow us to obtain DHSPES- derived vimses containing genes that are not well tolerated by E. coli in traditional cloning approaches.
  • cell-free cloning will entail the in vitro assembly of partial viral sequences with a DNA fragment into a configuration that that will yield infectious viral RNA molecules upon in vitro transcription.
  • the viral sequences are divided into two "arms"; the left arm and the right arm.
  • the left arm encodes a T7 RNA polymerase promoter followed by viral sequences encoding replicase followed by the gene encoding movement protein and the subgenomic promoter that controls expression ofthe desired gene.
  • the right arm will contain sequences ofthe viral genome that encode the viral coat protein and the sequences that control its expression, the viral 3' untranslated region, and a ribozyme sequence for generating the desired 3' terminus on the transcribed molecules.
  • FIGURE 28 A schematic diagram for cell free cloning is shown in FIGURE 28.
  • the left arm and right arm will each have separate asymmetric (non- palindromic, thus self-incompatible) overhangs that will permit the two arms to be brought together by an intervening insert that is derived either from PCR product, cDNA reaction, or elsewhere.
  • the insert will have termini that are compatible with both the left and right arms. The termini of these molecules are such that ligation of left and right arms to insert will ensure assembly into the proper configuration to yield infectious viral transcripts.
  • the sequence contained in the insert will then be in the conect orientation and genomic position to permit its expression from the vims in plant cells.
  • the right arm will be synthesized by PCR and will have a biotin group inco ⁇ orated into the reverse (3') primer.
  • the resulting biotinylated PCR product representing the right arm will then be immobilized upon streptavidin paramagnetic beads.
  • Treatment ofthe DNA with T4 DNA polymerase and a single dNTP (in the present case, dGTP) will give a 5' overhang as a result ofthe exonuclease activity ofthe polymerase.
  • the insert DNA being PCR product, restriction fragment, or cDNA will be treated with T4 DNA polymerase with a single dNTP to generate 5' overhangs on its termini; the 3' of which is compatible with the 5' ofthe right arm.
  • the 5' terminus ofthe insert DNA will be compatible with the left .arm 3' terminus that had been generated similarly.
  • ligation reactions in the assembly ofthe vims on the paramagnetic beads will be carried out sequentially, with the insert being ligated to the immobilized right .arm first, followed by washing ofthe bead complex and then ligation ofthe left arm. Following the subsequent wash, in vitro transcription will be carried out to generate infectious RNA transcripts.
  • a DNA fragment comprising the left arm ofthe vims was then ligated to the resulting DNA-bead complex to generate a full-length vims clone that was subsequently used as template for in vitro transcription.
  • DNA-bead complexes were washed by sedimenting them in a magnetic field and resuspending them in the appropriate buffer.
  • aliquots were taken for analysis to confirm that the desired reaction had occurred.
  • the infectious RNA products ofthe franscription reaction were introduced into protoplasts of tobacco cell suspension cultures.
  • EXAMPLE 34 Use of undefined sequences to increase the genetic stability of foreign genes in vims expression vectors.
  • Insertion of foreign gene sequences into vims expression vectors can result in anangements of sequences that interfere with normal vims function and thereby, establish a selection landscape that favors the genetic deletion ofthe foreign sequence. Such events are adverse to the use of such expression vectors to stably express gene sequences systemically in plants.
  • a method that would allow sequences to be identified that may "insulate" functional virus sequences from the potential adverse effects of insertion of foreign gene sequences would greatly augment the expression potential of vims expression vectors.
  • identification of such "insulating" sequences that simultaneously enhanced the translation ofthe foreign gene product or the stability ofthe mRNA encoding the foreign gene would be quite helpful.
  • the example below demonstrates how libraries of random sequences can be introduced into vims vectors flanking foreign gene sequences.
  • the genetic stability ofthe human growth hormone gene (hGH) or an Ubiquitin fusion to hGH (Ubiq hGH) in the tob-amo virus expression vector p30B is rather poor, such that no stable vims preparations could not be made to serially passage infection onto plants and detect the expression of hGH recombinant protein.
  • the site of gene insertion is following a Pad site (underlined) in the vims vector. This sequence is known as a leader sequence and has been derived from the native leader and coding region from the native TMV Ul coat protein gene. In this leader, the normal coat protein ATG has been mutated to a Aga sequence (underlined in
  • a particular subset of this leader sequence (TCTTACAGTATCACTACTCCATCTCAGTTCGTGTTCTTGTCA) has been known to increase genetic stability and gene expression when compared with vims construct lacking the leader sequence.
  • the start site of subgenomic RNA synthesis is found at the GTTTT...
  • An oligonucleotide RL-1 (GTTTTAAATAGATCTTAC N(20)TTAATTAAGGCC ) was used with a primer homologous to the NcollApal region ofthe TMV genome to amplify a portion ofthe TMV movement protein.
  • the population of sequences were cloned into the Apal and Pad sites ofthe p30B hGH vector.
  • Vectors containing the undefined sequences leading the hGH genes were transcribed and inoculated onto Nicotiana benthamiana plants. 14 days post inoculation, systemic leaves were ground .and the plant extracts were inoculated onto a second set of plants.
  • a selectable marker could be used to discover which ofthe undefined sequences yield the desired function.
  • the amount ofthe GFP protein conelates with the level of fluorescence seen under long wave UV light and the amount of herbicide resistance gene product conelates with survival of plant cells or plants upon treatment with the herbicide. Therefore introduction of undefined sequences sunounding the GFP or herbicide resistance genes and then screening for individual vimses that either express the greatest level of fluorescence or cells that survive the highest amount of herbicide.
  • EXAMPLE 35 Method for using reporter genes fused to regulated or constitutive promoters as a surrogate marker for identifying genes impacting gene regulation.
  • transgenic hosts expressing a reporter gene under the control of various promoter types; 2) means to use such hosts to identify genes from libraries expressed in vims expression vectors that alter gene regulation.
  • the initial constmction ofthe reporter gene expression cassette will require identification ofthe appropriate reporter gene, which could include GFP (fluorescent in live plants under long wave UV light), GUS (fluorescent and color-based assay in detected tissue), herbicide resistance genes (live or death phenotype upon treatment with herbicide) or other scoreable gene products known to the art.
  • Promoter sequences can express RNA in constitutive or induced conditions.
  • An example of a regulated promoter would be that of tomato or potato protease inhibitor type I gene (Graham, et al, J. Biol. Chem. 260:6555- 6560 (1985)). These promoters are up regulated in the presence of jasmonic acid or herbivore damage to plant tissues.
  • Constitutive promoters are readily identifiable from anyone skilled in the art inspecting the relevant literature. Such combinations of inducible or constitutive promoters using appropriate reporter genes would be integrated into binary plant transformation vectors, fransformed into Agrobacterium and transformed into Nicotiana benthamiana leaf disks. Upon identification ofthe appropriate gene construct in regenerated tissues, the primary transformants would be selfed to obtain the first stable line of plants for assay. Libraries of cDNAs, full-length for gene overexpression or gene fragments for sense or anti-sense based gene suppression, would be ligated into vims expression vectors by normal molecular biology techniques. These libraries would be prepared for inoculation by the methods described in this patent application.
  • hosts with inducible promoters fused to reporter genes Once inoculated, hosts with inducible promoters fused to reporter genes, maintained in uninduced state, would be monitored for abenant expression ofthe reporter gene in tissue that contains replicating vims. If hosts containing constitutive promoter fusions to reporter genes .are used, monitoring for hyper- or hypo-expression conditions ofthe reporter gene would be the focus. In this manner, genes that augment pathways that induce or upregulate the activity of certain promoters could be identified by following the surrogate marker of reporter gene expression. Conversely, gene that down-regulate or halt reporter gene expression could be identified as products that negatively effect the activities ofthe promoter or signaling pathway to which it is responsive. Vims vectors containing sequences that effected reporter gene expression by overexpression or suppression positive or negative regulatory factors can be isolated, and foreign gene contained may be sequenced and analyzed by bioinformatic methods.
  • EXAMPLE 36 Method to induce the expression of alternative splicing variants to discover biological effects in host organisms and to use said host organism as a source for novel cDNA libraries enriched for alternatively spliced variants of genes.
  • Transcription of nuclear genes in higher eukaryotic organisms results in a primary RNA transcript that contains both coding (exon) and non-coding (intron) information.
  • a cmcial step in RNA maturation before exporting to the cytosol for translation is the splicing of introns from the primary transcript and the rendering of contiguous exons for coding ofthe desired product.
  • Gene families can include the SF2/ASF-like group of splicing factors (Lopato et al, PNAS 92:7672- 7676 (1995)), the RS-rich family of splicing factors (Lapato et al, The Plant Cell :2255-2264 (1996)) and other splicing families that have been identified in the literature in lower or upper eukaryotic systems.
  • the gene libraries will be subcloned into vims expression vectors and vims libraries will be inoculated as individuals or pools onto plants or plant cells.
  • splicing factors Once individual or groups of splicing factors are overexpressed or have their expression suppressed in plant cells, novel forms of splicing will occur due to the role of these proteins in alternative splicing of many transcription factors, splicing factors or other gene products.
  • the high level of expression achieved by vims expression vectors and their ability to infect most cell types in plants should raise the overall level of abenantly expressed mRNAs in the plant.
  • the transfected plants will be used as the starting point for the isolation of poly A(+) RNA for the constmction of cDNAs enriched for alternatively spliced genes.
  • the alterations in the alternative splicing could be the splicing of a greater or lesser number of introns from the primary mRNA than normally occurs in non-transfected plants.
  • These enriched cDNA libraries can now be cloned into vims expression vectors and the functions of these novel spliced forms of genes can be assayed on plants transfected with these vector libraries.
  • Similar methods could be to derive novel cDNA libraries by using vims vectors to express factors responsible for transcriptional regulation of genes in plants.
  • targeted cloning of transcription factor families would be ligated into vims expression vectors. Families could include homeodomain, Zn finger, leucine zipper and other transcription factor families appearing in pro or eukaryotic genomes. Schwechheimer, et al., Ann. Rev. Plant Phys. and Plant Mol. Biol. 49 (1998), in press.
  • the gene libraries will be sub-cloned into vims expression vectors and vims libraries will be inoculated as individuals or pools onto plants or plant cells.
  • cDNAs would be used to construct expression or gene suppression libraries that will be enriched for these rare or abenantly expressed cDNAs.
  • These enriched cDNA libraries can now be cloned into vims expression vectors and the functions of these novel spliced forms of genes can be assayed on plants transfected with these vector libraries.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Plant Pathology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Breeding Of Plants And Reproduction By Means Of Culturing (AREA)

Abstract

La présente invention concerne des procédés de détermination rapide de la fonction de séquences d'acides nucléiques qui consiste en la transfection desdites séquences dans un organisme hôte afin d'y effectuer une expression; ensuite à analyser les changements phénotypiques et biochimiques ainsi produits afin de déterminer la fonction des acides nucléiques transfectés dans l'organisme hôte. L'invention concerne aussi des procédés de blocage des gènes endogènes par la transfection des hôtes ayant des séquences d'acides nucléiques pour en effectuer l'expression. L'invention concerne en outre des procédés de sélection des fonctions désirées des ARN et des protéines au moyen de vecteurs viraux en vue d'exprimer des banques des allèles des séquences d'acides nucléiques. Finalement l'invention concerne des procédés d'inhibition d'une protéase endogène d'une plante hôte.
PCT/US1999/001164 1998-01-16 1999-01-15 Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote WO1999036516A2 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
KR1020007007808A KR20010040337A (ko) 1998-01-16 1999-01-15 누클레오티드 서열을 숙주로 형질감염시켜 이누클레오티드 서열의 기능 및 이 누클레오티드 서열이코딩하는 단백질을 결정하는 방법
JP2000540219A JP2002508957A (ja) 1998-01-16 1999-01-15 宿主へのトランスフェクションによりヌクレオチド配列及びそれがコードするタンパク質の機能を決定する方法
EP99903208A EP1045899A2 (fr) 1998-01-16 1999-01-15 Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote
AU23286/99A AU761367B2 (en) 1998-01-16 1999-01-15 Method of determining the function of nucleotide sequences and the proteins they encode by transfecting the same into a host
CA002318662A CA2318662A1 (fr) 1998-01-16 1999-01-15 Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote
US10/236,508 US20030167512A1 (en) 1998-01-16 2002-09-06 Method of determining the presence of a trait in a plant by transfecting a nucleic acid sequence of a donor plant into a different host plant in a positive orientation
AU2003220702A AU2003220702A1 (en) 1998-01-16 2003-07-18 Method of determining the function of nucleotide sequences and the proteins they encode by transfecting the same into a host

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US818698A 1998-01-16 1998-01-16
US09/008,186 1998-01-16

Publications (2)

Publication Number Publication Date
WO1999036516A2 true WO1999036516A2 (fr) 1999-07-22
WO1999036516A3 WO1999036516A3 (fr) 2000-03-16

Family

ID=21730222

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/001164 WO1999036516A2 (fr) 1998-01-16 1999-01-15 Procede de determination de la fonction de sequences nucleotidiques et les proteines pour lesquelles elles codent par transfection de telles sequences dans un hote

Country Status (6)

Country Link
EP (1) EP1045899A2 (fr)
JP (1) JP2002508957A (fr)
KR (1) KR20010040337A (fr)
AU (1) AU761367B2 (fr)
CA (1) CA2318662A1 (fr)
WO (1) WO1999036516A2 (fr)

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000063397A2 (fr) * 1999-04-20 2000-10-26 Aventis Cropscience N.V. Methodes et moyens d'administration d'arn inhibiteur a des vegetaux et applications associees
WO2001007613A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede pour renforcer la production d'arn ou de proteines a l'aide de sequences 5' non traduites non natives dans des acides nucleiques viraux de recombinaison
WO2001007600A1 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede de correlation de fonction de sequence par transfection en orientation antisens ou en sens positif dans un hote vegetal d'une sequence d'acide nucleique d'un organisme donneur
WO2001007641A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede permettant d'exprimer une banque de variants de sequences d'acide nucleique et de selectionner des traits souhaites
WO2001007601A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede permettant de conferer a des plantes hotes, une resistance aux herbicides, aux parasites et aux maladies
WO2001038513A2 (fr) * 1999-11-23 2001-05-31 Maxygen, Inc. Techniques de brassage de genes de virus et d'agrobacteries, de plasmides, et de genomes permettant d'ameliorer une transformation de plante
US6300134B1 (en) 1998-01-16 2001-10-09 Large Scale Biology Corporation RNA transformation vectors derived from a single-component RNA virus and contain an intervening sequence between the cap and the 5′ end
US6300133B1 (en) 1998-01-16 2001-10-09 Large Scale Biology Corporation RNA transformation vectors derived from an uncapped single-component RNA virus
WO2001079514A2 (fr) * 2000-04-18 2001-10-25 Syngenta Participations Ag Genes d' « acide amine » glyoxylate aminotransferase d'origine vegetale et utilisations de ceux-ci
WO2002029068A2 (fr) * 2000-10-06 2002-04-11 Icon Genetics Ag Systeme vectoriel pour plantes
WO2002059335A2 (fr) * 2001-01-25 2002-08-01 Large Scale Biology Corporation Inhibition ou expression d'un gene dans le cytoplasme effectuee a l'aide d'un vecteur de tobravirus dans des plantes transfectees
WO2002059378A2 (fr) * 2001-01-25 2002-08-01 Virxsys Procedes et compositions servant a identifier la fonction d'un gene
JP2002238574A (ja) * 2001-02-16 2002-08-27 Japan International Research Center For Agricultural Services パパイア奇形葉モザイクウイルスの全長ゲノムrna
WO2003060134A1 (fr) * 2002-01-16 2003-07-24 Cropdesign N.V. Determination du mode d'action d'un herbicide
WO2004011656A1 (fr) * 2002-07-29 2004-02-05 UNIVERSITé LAVAL Procede d'amelioration du rendement de production de proteine recombinante a partir de vegetaux
WO2003077642A3 (fr) * 2002-03-19 2004-03-11 Metanomics Gmbh & Co Kgaa Population de plantes transgeniques, element biologique derive de ces plantes, collection de plasmides correspondante, population d'organismes hotes transformes, utilisation et procede de production de ces populations
EP1431392A1 (fr) * 2001-08-31 2004-06-23 Riken Systeme vegetal permettant une analyse approfondie de fonctions geniques a l'aide d'adnc pleine longueur
WO2004079000A1 (fr) * 2003-03-07 2004-09-16 National Research Council Of Canada Determination de la fonction de proteines mammaliennes par analyse de profils globaux d'expression genique
US6800748B2 (en) 2001-01-25 2004-10-05 Large Scale Biology Corporation Cytoplasmic inhibition of gene expression and expression of a foreign protein in a monocot plant by a plant viral vector
US6830885B1 (en) 2000-08-18 2004-12-14 Phenogene Therapeutiques Inc. Nucleic acid molecule, method and kit for selecting a nucleic acid having a desired feature
US6861229B2 (en) 2000-07-06 2005-03-01 Genvec, Inc. Method of identifying a gene product
US6998263B2 (en) 2000-02-09 2006-02-14 Genvec, Inc. Methods of preparing and using a viral vector library
US7193131B2 (en) 2001-01-19 2007-03-20 Icon Genetics Ag Processes and vectors for plastid transformation of higher plants
US7371923B2 (en) 2001-07-06 2008-05-13 Icon Genetics Ag Process of generating transplastomic plants or plant cells devoid of a selection marker
US7485774B2 (en) 2001-12-18 2009-02-03 Bayer Bioscience, N.V. Methods and means for delivering inhibitory RNA to plants and applications thereof
US7635798B2 (en) 2001-08-31 2009-12-22 Dow Agrosciences, Llc Nucleic acid compositions conferring altered metabolic characteristics
US7652194B2 (en) 2000-12-08 2010-01-26 Icon Genetics Gmbh Processes and vectors for producing transgenic plants
US7667091B2 (en) 2001-03-29 2010-02-23 Icon Genetics Gmbh Method of encoding information in nucleic acids of a genetically engineered organism
US7667092B2 (en) 2001-04-30 2010-02-23 Icon Genetics Gmbh Processes and vectors for amplification or expression of nucleic acid sequences of interest in plants
US8058506B2 (en) 2001-03-23 2011-11-15 Icon Genetics Gmbh Site-targeted transformation using amplification vectors
US8192984B2 (en) 2001-09-04 2012-06-05 Icon Genetics, Inc. Creation of artificial internal ribosome entry site (IRES) elements
CN102498998A (zh) * 2011-11-24 2012-06-20 湖南杂交水稻研究中心 一种水稻光温敏不育系种子的提纯保真繁殖方法
US8257945B2 (en) 2001-09-04 2012-09-04 Icon Genetics, Inc. Identification of eukaryotic internal ribosome entry site (IRES) elements
WO2014195214A1 (fr) * 2013-06-06 2014-12-11 Henkel Ag & Co. Kgaa Alcool oxydase ayant une activité pour le glycérol
WO2019027861A1 (fr) 2017-07-31 2019-02-07 R. J. Reynolds Tobacco Company Procédés et compositions pour l'édition de gènes à base de virus dans des plantes
WO2019049111A1 (fr) 2017-09-11 2019-03-14 R. J. Reynolds Tobacco Company Méthodes et compositions permettant d'augmenter l'expression de gènes d'intérêt dans une plante par co-expression avec p21
US20200165463A1 (en) * 2017-06-30 2020-05-28 Dow Global Technologies Llc Coating for aldehyde remediation and method of making

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991001375A1 (fr) * 1989-07-14 1991-02-07 Imperial Chemical Industries Plc Structures d'adn, cellules et plantes derivees de celles-ci
WO1995034668A2 (fr) * 1994-06-16 1995-12-21 Biosource Technologies, Inc. Inhibition cytoplasmique de l'expression genique
WO1998036083A1 (fr) * 1997-02-14 1998-08-20 Plant Bioscience Limited Procedes et moyens de blocage de gene dans des plantes transgeniques

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991001375A1 (fr) * 1989-07-14 1991-02-07 Imperial Chemical Industries Plc Structures d'adn, cellules et plantes derivees de celles-ci
WO1995034668A2 (fr) * 1994-06-16 1995-12-21 Biosource Technologies, Inc. Inhibition cytoplasmique de l'expression genique
WO1998036083A1 (fr) * 1997-02-14 1998-08-20 Plant Bioscience Limited Procedes et moyens de blocage de gene dans des plantes transgeniques

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BAULCOMBE , D.C.: "fast foward genetics based on virus-induced gene silencing" CURRENT OPINION IN PLANT BIOLOGY, vol. 2, 1999, pages 109-113, XP002118432 *
SABLOWSKI, R.W.M., ET AL.: "expression of a flower-specific Myb protein in leaf cells using a viral vector causes ectopic activation of a target promoter" PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE USA, vol. 92, July 1995, pages 6901-6905, XP002118431 *

Cited By (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6468745B1 (en) 1998-01-16 2002-10-22 Large Scale Biology Corporation Method for expressing a library of nucleic acid sequence variants and selecting desired traits
US6303848B1 (en) 1998-01-16 2001-10-16 Large Scale Biology Corporation Method for conferring herbicide, pest, or disease resistance in plant hosts
US6300133B1 (en) 1998-01-16 2001-10-09 Large Scale Biology Corporation RNA transformation vectors derived from an uncapped single-component RNA virus
US6300134B1 (en) 1998-01-16 2001-10-09 Large Scale Biology Corporation RNA transformation vectors derived from a single-component RNA virus and contain an intervening sequence between the cap and the 5′ end
WO2000063397A2 (fr) * 1999-04-20 2000-10-26 Aventis Cropscience N.V. Methodes et moyens d'administration d'arn inhibiteur a des vegetaux et applications associees
WO2000063397A3 (fr) * 1999-04-20 2001-02-01 Aventis Cropscience Nv Methodes et moyens d'administration d'arn inhibiteur a des vegetaux et applications associees
WO2001007601A3 (fr) * 1999-07-21 2001-08-30 Large Scale Biology Corp Procede permettant de conferer a des plantes hotes, une resistance aux herbicides, aux parasites et aux maladies
WO2001007641A3 (fr) * 1999-07-21 2001-08-09 Large Scale Biology Corp Procede permettant d'exprimer une banque de variants de sequences d'acide nucleique et de selectionner des traits souhaites
WO2001007601A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede permettant de conferer a des plantes hotes, une resistance aux herbicides, aux parasites et aux maladies
WO2001007641A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede permettant d'exprimer une banque de variants de sequences d'acide nucleique et de selectionner des traits souhaites
WO2001007600A1 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede de correlation de fonction de sequence par transfection en orientation antisens ou en sens positif dans un hote vegetal d'une sequence d'acide nucleique d'un organisme donneur
WO2001007613A3 (fr) * 1999-07-21 2002-01-17 Large Scale Biology Corp Procede pour renforcer la production d'arn ou de proteines a l'aide de sequences 5' non traduites non natives dans des acides nucleiques viraux de recombinaison
WO2001007613A2 (fr) * 1999-07-21 2001-02-01 Large Scale Biology Corporation Procede pour renforcer la production d'arn ou de proteines a l'aide de sequences 5' non traduites non natives dans des acides nucleiques viraux de recombinaison
WO2001038513A2 (fr) * 1999-11-23 2001-05-31 Maxygen, Inc. Techniques de brassage de genes de virus et d'agrobacteries, de plasmides, et de genomes permettant d'ameliorer une transformation de plante
WO2001038513A3 (fr) * 1999-11-23 2002-01-03 Maxygen Inc Techniques de brassage de genes de virus et d'agrobacteries, de plasmides, et de genomes permettant d'ameliorer une transformation de plante
US6998263B2 (en) 2000-02-09 2006-02-14 Genvec, Inc. Methods of preparing and using a viral vector library
WO2001079514A3 (fr) * 2000-04-18 2002-04-18 Syngenta Participations Ag Genes d' « acide amine » glyoxylate aminotransferase d'origine vegetale et utilisations de ceux-ci
WO2001079514A2 (fr) * 2000-04-18 2001-10-25 Syngenta Participations Ag Genes d' « acide amine » glyoxylate aminotransferase d'origine vegetale et utilisations de ceux-ci
US6861229B2 (en) 2000-07-06 2005-03-01 Genvec, Inc. Method of identifying a gene product
US6830885B1 (en) 2000-08-18 2004-12-14 Phenogene Therapeutiques Inc. Nucleic acid molecule, method and kit for selecting a nucleic acid having a desired feature
US7763458B2 (en) 2000-10-06 2010-07-27 Icon Genetics Gmbh Vector system for plants
WO2002029068A2 (fr) * 2000-10-06 2002-04-11 Icon Genetics Ag Systeme vectoriel pour plantes
WO2002029068A3 (fr) * 2000-10-06 2002-10-03 Icon Genetics Ag Systeme vectoriel pour plantes
AU2002214003B2 (en) * 2000-10-06 2007-07-26 Icon Genetics Gmbh Virus-based amplification vector for plants
US7652194B2 (en) 2000-12-08 2010-01-26 Icon Genetics Gmbh Processes and vectors for producing transgenic plants
US7193131B2 (en) 2001-01-19 2007-03-20 Icon Genetics Ag Processes and vectors for plastid transformation of higher plants
WO2002059335A2 (fr) * 2001-01-25 2002-08-01 Large Scale Biology Corporation Inhibition ou expression d'un gene dans le cytoplasme effectuee a l'aide d'un vecteur de tobravirus dans des plantes transfectees
WO2002059378A3 (fr) * 2001-01-25 2003-12-18 Virxsys Procedes et compositions servant a identifier la fonction d'un gene
WO2002059335A3 (fr) * 2001-01-25 2003-11-20 Large Scale Biology Corp Inhibition ou expression d'un gene dans le cytoplasme effectuee a l'aide d'un vecteur de tobravirus dans des plantes transfectees
US7427474B2 (en) 2001-01-25 2008-09-23 Virxsys Corporation High-throughput methods for identifying gene function using lentiviral vectors
US6800748B2 (en) 2001-01-25 2004-10-05 Large Scale Biology Corporation Cytoplasmic inhibition of gene expression and expression of a foreign protein in a monocot plant by a plant viral vector
AU2002241980B2 (en) * 2001-01-25 2008-01-31 Virxsys Corporation Methods and compositions for identifying gene function
WO2002059378A2 (fr) * 2001-01-25 2002-08-01 Virxsys Procedes et compositions servant a identifier la fonction d'un gene
JP2002238574A (ja) * 2001-02-16 2002-08-27 Japan International Research Center For Agricultural Services パパイア奇形葉モザイクウイルスの全長ゲノムrna
US8058506B2 (en) 2001-03-23 2011-11-15 Icon Genetics Gmbh Site-targeted transformation using amplification vectors
US7667091B2 (en) 2001-03-29 2010-02-23 Icon Genetics Gmbh Method of encoding information in nucleic acids of a genetically engineered organism
US7667092B2 (en) 2001-04-30 2010-02-23 Icon Genetics Gmbh Processes and vectors for amplification or expression of nucleic acid sequences of interest in plants
US7371923B2 (en) 2001-07-06 2008-05-13 Icon Genetics Ag Process of generating transplastomic plants or plant cells devoid of a selection marker
EP1431392A4 (fr) * 2001-08-31 2005-04-06 Riken Systeme vegetal permettant une analyse approfondie de fonctions geniques a l'aide d'adnc pleine longueur
US7608758B2 (en) 2001-08-31 2009-10-27 Riken Plant system for comprehensive gene function analysis with the use of full-length cDNA
US7635798B2 (en) 2001-08-31 2009-12-22 Dow Agrosciences, Llc Nucleic acid compositions conferring altered metabolic characteristics
EP1431392A1 (fr) * 2001-08-31 2004-06-23 Riken Systeme vegetal permettant une analyse approfondie de fonctions geniques a l'aide d'adnc pleine longueur
US8257945B2 (en) 2001-09-04 2012-09-04 Icon Genetics, Inc. Identification of eukaryotic internal ribosome entry site (IRES) elements
US8192984B2 (en) 2001-09-04 2012-06-05 Icon Genetics, Inc. Creation of artificial internal ribosome entry site (IRES) elements
US7485774B2 (en) 2001-12-18 2009-02-03 Bayer Bioscience, N.V. Methods and means for delivering inhibitory RNA to plants and applications thereof
WO2003060134A1 (fr) * 2002-01-16 2003-07-24 Cropdesign N.V. Determination du mode d'action d'un herbicide
WO2003077642A3 (fr) * 2002-03-19 2004-03-11 Metanomics Gmbh & Co Kgaa Population de plantes transgeniques, element biologique derive de ces plantes, collection de plasmides correspondante, population d'organismes hotes transformes, utilisation et procede de production de ces populations
US8247652B2 (en) 2002-03-19 2012-08-21 Metanomics Gmbh & Co. Kgaa Population of transgenic plants individually comprising distinct codogenic gene segments, the population having at least 50% of the codogenic gene segments from a donor organism
WO2004011656A1 (fr) * 2002-07-29 2004-02-05 UNIVERSITé LAVAL Procede d'amelioration du rendement de production de proteine recombinante a partir de vegetaux
WO2004079000A1 (fr) * 2003-03-07 2004-09-16 National Research Council Of Canada Determination de la fonction de proteines mammaliennes par analyse de profils globaux d'expression genique
CN102498998A (zh) * 2011-11-24 2012-06-20 湖南杂交水稻研究中心 一种水稻光温敏不育系种子的提纯保真繁殖方法
WO2014195214A1 (fr) * 2013-06-06 2014-12-11 Henkel Ag & Co. Kgaa Alcool oxydase ayant une activité pour le glycérol
US20200165463A1 (en) * 2017-06-30 2020-05-28 Dow Global Technologies Llc Coating for aldehyde remediation and method of making
US11597845B2 (en) * 2017-06-30 2023-03-07 Dow Global Technologies Llc Coating for aldehyde remediation and method of making
WO2019027861A1 (fr) 2017-07-31 2019-02-07 R. J. Reynolds Tobacco Company Procédés et compositions pour l'édition de gènes à base de virus dans des plantes
WO2019049111A1 (fr) 2017-09-11 2019-03-14 R. J. Reynolds Tobacco Company Méthodes et compositions permettant d'augmenter l'expression de gènes d'intérêt dans une plante par co-expression avec p21

Also Published As

Publication number Publication date
EP1045899A2 (fr) 2000-10-25
KR20010040337A (ko) 2001-05-15
CA2318662A1 (fr) 1999-07-22
JP2002508957A (ja) 2002-03-26
AU761367B2 (en) 2003-06-05
AU2328699A (en) 1999-08-02
WO1999036516A3 (fr) 2000-03-16

Similar Documents

Publication Publication Date Title
AU761367B2 (en) Method of determining the function of nucleotide sequences and the proteins they encode by transfecting the same into a host
US20050005319A1 (en) Method of determining the function of nucleotide sequences and the proteins they encode by transfecting the same into a host
US6800748B2 (en) Cytoplasmic inhibition of gene expression and expression of a foreign protein in a monocot plant by a plant viral vector
AU2002361997A1 (en) Improved methods and means for delivering inhibitory rna to plants and applications thereof
EP0999281A1 (fr) Transformation chromosomique non-nucléaire
US7498480B2 (en) Cytoplasmic gene inhibition or gene expression in transfected plants by a tobraviral vector
Vidal et al. Use of gene transfer technology for functional studies in grapevine
JPH11507217A (ja) 組換えウイルスの核酸
US6426185B1 (en) Method of compiling a functional gene profile in a plant by transfecting a nucleic acid sequence of a donor plant into a different host plant in an anti-sense orientation
US6468745B1 (en) Method for expressing a library of nucleic acid sequence variants and selecting desired traits
US6300133B1 (en) RNA transformation vectors derived from an uncapped single-component RNA virus
AU2003220702A1 (en) Method of determining the function of nucleotide sequences and the proteins they encode by transfecting the same into a host
US20020164585A1 (en) Method for enhancing RNA or protein production using non-native 5' untranslated sequences in recombinant viral nucleic acids
US20030167512A1 (en) Method of determining the presence of a trait in a plant by transfecting a nucleic acid sequence of a donor plant into a different host plant in a positive orientation
AU6608300A (en) Method of correlating sequence function by transfecting a nucleic acid sequence of a donor organism into a plant host in an anti-sense or positive sense orientation
US6300134B1 (en) RNA transformation vectors derived from a single-component RNA virus and contain an intervening sequence between the cap and the 5′ end
US20030166169A1 (en) Method for constructing viral nucleic acids in a cell-free manner
US20030041355A1 (en) Method of humanizing plant cDNA
US20030064392A1 (en) Method of humanizing plant cDNAs by transfecting a nucleic acid sequence of a non-plant donor into a host plant in an anti-sense orientation
WO2001007613A2 (fr) Procede pour renforcer la production d'arn ou de proteines a l'aide de sequences 5' non traduites non natives dans des acides nucleiques viraux de recombinaison
US20030097683A1 (en) Single-component RNA vectors derived from a virus and containing an intervening sequence between the cap and the 5' end and able to replicate in a host plant cell within a host plant
AU2002242001A1 (en) Cytoplasmic gene inhibition or gene expression in transfected plants by a tobraviral vector
AU3400100A (en) Recombinant viral nucleic acids

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW SD SZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

ENP Entry into the national phase

Ref document number: 2318662

Country of ref document: CA

Ref country code: CA

Ref document number: 2318662

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1020007007808

Country of ref document: KR

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 540219

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 23286/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 505879

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 1999903208

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999903208

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1020007007808

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 10236508

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 23286/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1020007007808

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1999903208

Country of ref document: EP