Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Plant Mol Biol ; 82(4-5): 339-51, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23625359

RESUMO

Besides the KU-dependent classical non-homologous end-joining (C-NHEJ) pathway, an alternative NHEJ pathway first identified in mammalian systems, which is often called the back-up NHEJ (B-NHEJ) pathway, was also found in plants. In mammalian systems PARP was found to be one of the essential components in B-NHEJ. Here we investigated whether PARP1 and PARP2 were also involved in B-NHEJ in Arabidopsis. To this end Arabidopsis parp1, parp2 and parp1parp2 (p1p2) mutants were isolated and functionally characterized. The p1p2 double mutant was crossed with the C-NHEJ ku80 mutant resulting in the parp1parp2ku80 (p1p2k80) triple mutant. As expected, because of their role in single strand break repair (SSBR) and base excision repair (BER), the p1p2 and p1p2k80 mutants were shown to be sensitive to treatment with the DNA damaging agent MMS. End-joining assays in cell-free leaf protein extracts of the different mutants using linear DNA substrates with different ends reflecting a variety of double strand breaks were performed. The results showed that compatible 5'-overhangs were accurately joined in all mutants, that KU80 protected the ends preventing the formation of large deletions and that PARP proteins were involved in microhomology mediated end joining (MMEJ), one of the characteristics of B-NHEJ.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Reparo do DNA por Junção de Extremidades/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Poli(ADP-Ribose) Polimerases/genética
2.
Proc Natl Acad Sci U S A ; 106(24): 9643-8, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19482939

RESUMO

Agrobacterium tumefaciens VirC2 stimulates processing of single-stranded T-DNA that is translocated into plants to induce tumor formation, but how VirC2 functions is unclear. Here, we report the 1.7-A X-ray crystal structure of its trypsin-resistant C-terminal domain, VirC2(82-202), which reveals a form of the ribbon-helix-helix (RHH) DNA-binding fold contained within a single polypeptide chain. DNA-binding assays and mutagenesis indicate that VirC2 uses this RHH fold to bind double-stranded DNA but not single-stranded DNA. Mutations that severely affect VirC2 DNA binding are highly deleterious for both T-DNA transfer into yeast and the virulence of A. tumefaciens in different plants including Nicotiana glauca and Kalanchoe daigremontiana. These data suggest that VirC2 enhances T-DNA transfer and virulence through DNA binding with its RHH fold. The RHH fold of VirC2 is the first crystal structure representing a group of predicted RHH proteins that facilitate endonucleolytic processing of DNA for horizontal gene transfer.


Assuntos
Agrobacterium tumefaciens/metabolismo , Proteínas de Bactérias/fisiologia , DNA Bacteriano/genética , Virulência/fisiologia , Agrobacterium tumefaciens/genética , Agrobacterium tumefaciens/patogenicidade , Sequência de Aminoácidos , Proteínas de Bactérias/química , Sítios de Ligação , DNA Bacteriano/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Modelos Moleculares , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
3.
Methods Mol Biol ; 344: 465-73, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17033086

RESUMO

The yeast Saccharomyces cerevisiae is one of the best characterized eukaryotic organisms. This species has enabled a detailed study of the (genetic) requirements for Agrobacterium-mediated DNA transformation. For instance research with this yeast has led to the recognition that the transforming DNA molecules integrate into the eukaryotic chromosomes either by homologous recombination, which is the preferred pathway in S. cerevisiae, or by nonhomologous end-joining. Based on the protocol for Agrobacterium-mediated transformation of S. cerevisiae methodology has been developed for the transformation of many other yeast and fungal species.


Assuntos
Agrobacterium tumefaciens/genética , Saccharomyces cerevisiae/genética , Transformação Genética , Agrobacterium tumefaciens/citologia , Técnicas de Cultura de Células , Técnicas de Cocultura , Meios de Cultura , DNA Bacteriano , Marcação de Genes , Vetores Genéticos , Plasmídeos , Saccharomyces cerevisiae/isolamento & purificação , Saccharomyces cerevisiae/fisiologia
4.
Nucleic Acids Res ; 31(3): 860-8, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12560481

RESUMO

Agrobacterium tumefaciens causes crown gall disease on a variety of plants. During the infection process Agrobacterium transfers a nucleoprotein complex, the VirD2 T-complex, and at least two Vir proteins, VirE2 and VirF, into the plant cell via the VirB/VirD4 type IV secretion system. Recently, we found that T-DNA could also be transferred from Agrobacterium to Saccharomyces cerevisiae. Here, we describe a novel method to also detect trans-kingdom Vir protein transfer from Agrobacterium to yeast, using the Cre/lox system. Protein fusions between Cre and VirE2 or VirF were expressed in AGROBACTERIUM: Transfer of the Cre-Vir fusion proteins from Agrobacterium to yeast was monitored by a selectable excision event resulting from site-specific recombination mediated by Cre on a lox-flanked transgene in yeast. The VirE2 and VirF proteins were transported to yeast via the virB-encoded transfer system in the presence of coupling factor VirD4, analogous to translocation into plant cells. The yeast system therefore provides a suitable and fast model system to study basic aspects of trans-kingdom protein transport from Agrobacterium into host cells. Using this method we showed that VirE2 and VirF protein transfer was inhibited by the presence of the Osa protein. Besides, we found evidence for a novel third effector protein, VirE3, which has a similar C-terminal signature to VirE2 and VirF.


Assuntos
Agrobacterium tumefaciens/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Canais Iônicos/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Virulência , Agrobacterium tumefaciens/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Proteínas de Ligação a DNA/genética , Integrases/genética , Canais Iônicos/genética , Modelos Biológicos , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais/genética
5.
Methods Mol Biol ; 1197: 103-21, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25172277

RESUMO

Many pathogenic bacteria introduce virulence proteins, also called effector proteins, into host cells to accomplish infection. Such effector proteins are often translocated into host cells by bacterial type III (T3SS) or type IV secretion systems (T4SS). To better understand the molecular mechanisms underlying virulence, it is essential to identify the effector proteins and determine their functions. Several reporter assays have been established to identify translocated effector proteins and verify T3SS- or T4SS-dependent transport into host cells. Here we describe a protocol to monitor the translocation of candidate effector proteins using Cre recombinase as a reporter, and more specifically how this Cre Reporter Assay for Translocation (CRAfT) can be used to detect translocation of Vir proteins from Agrobacterium tumefaciens into yeast and plant cells. The assay can be adapted for the study of the T3SS or T4SS of human pathogens.


Assuntos
Transporte Proteico/fisiologia , Agrobacterium tumefaciens/metabolismo , Agrobacterium tumefaciens/fisiologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/fisiologia , Sistemas de Secreção Bacterianos/fisiologia , Humanos , Integrases/metabolismo , Transporte Proteico/genética
6.
Genome Announc ; 2(2)2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24675863

RESUMO

We have sequenced the complete genome of the plant pathogen Agrobacterium tumefaciens strain LBA4213, a derivative of the wild-type strain A. tumefaciens Ach5 and the ancestor of A. tumefaciens strain LBA4404 used in genetic engineering. The genome consists of a circular chromosome and a linear chromosome, as well as a megaplasmid and a tumor-inducing plasmid.

7.
Artigo em Inglês | MEDLINE | ID: mdl-22919588

RESUMO

Ehrlichia chaffeensis has type 1 and 4 secretion systems (T1SS and T4SS), but the substrates have not been identified. Potential substrates include secreted tandem repeat protein (TRP) 47, TRP120, and TRP32, and the ankyrin repeat protein, Ank200, that are involved in molecular host-pathogen interactions including DNA binding and a network of protein-protein interactions with host targets associated with signaling, transcriptional regulation, vesicle trafficking, and apoptosis. In this study we report that E. chaffeensis TRP47, TRP32, TRP120, and Ank200 were not secreted in the Agrobacterium tumefaciens Cre recombinase reporter assay routinely used to identify T4SS substrates. In contrast, all TRPs and the Ank200 proteins were secreted by the Escherichia coli complemented with the hemolysin secretion system (T1SS), and secretion was reduced in a T1SS mutant (ΔTolC), demonstrating that these proteins are T1SS substrates. Moreover, T1SS secretion signals were identified in the C-terminal domains of the TRPs and Ank200, and a detailed bioinformatic analysis of E. chaffeensis TRPs and Ank200 revealed features consistent with those described in the repeats-in-toxins (RTX) family of exoproteins, including glycine- and aspartate-rich tandem repeats, homology with ATP-transporters, a non-cleavable C-terminal T1SS signal, acidic pIs, and functions consistent with other T1SS substrates. Using a heterologous E. coli T1SS, this investigation has identified the first Ehrlichia T1SS substrates supporting the conclusion that the T1SS and corresponding substrates are involved in molecular host-pathogen interactions that contribute to Ehrlichia pathobiology. Further investigation of the relationship between Ehrlichia TRPs, Ank200, and the RTX exoprotein family may lead to a greater understanding of the importance of T1SS substrates and specific functions of T1SS in the pathobiology of obligately intracellular bacteria.


Assuntos
Proteínas de Bactérias/fisiologia , Ehrlichia chaffeensis/fisiologia , Ehrlichia chaffeensis/patogenicidade , Agrobacterium tumefaciens/genética , Sequência de Aminoácidos , Repetição de Anquirina , Arabidopsis/genética , Arabidopsis/microbiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Ehrlichia chaffeensis/genética , Escherichia coli/genética , Escherichia coli/fisiologia , Interações Hospedeiro-Patógeno , Dados de Sequência Molecular , Doenças das Plantas/microbiologia , Plantas Geneticamente Modificadas , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
8.
Cell Microbiol ; 9(11): 2644-57, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17587335

RESUMO

Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis, is an obligate intracellular bacterium of granulocytes. A. phagocytophilum specifically induces tyrosine phosphorylation of a 160 kDa protein (P160) in host cells. However, identity of P160, kinases involved, and effects of tyrosine phosphorylation on bacterial infection remain largely unknown. Here, we demonstrated through proteomic analysis that P160, an abundant and rapidly tyrosine-phosphorylated protein throughout infection, was AnkA of bacterial origin. Differential centrifugation and confocal microscopy revealed that AnkA was rarely retained within A. phagocytophilum or its inclusion, but localized mainly in the cytoplasm of infected cells. Using Cre recombinase reporter assay of Agrobacterium tumefaciens, we proved that AnkA could be secreted by VirB/D4-dependent type IV secretion (T4S) system. Yeast two-hybrid and coimmunoprecipitation analyses demonstrated that AnkA could bind to Abl-interactor 1 (Abi-1), an adaptor protein that interacts with Abl-1 tyrosine kinase, thus mediating AnkA phosphorylation. AnkA and Abl-1 were critical for bacterial infection, as infection was inhibited upon host cytoplasmic delivery of anti-AnkA antibody, Abl-1 knockdown with targeted siRNA, or treatment with a specific pharmacological inhibitor of Abl-1. These data establish AnkA as the first proven T4S substrate in members of obligate intracellular alpha-proteobacteria; furthermore, it demonstrated that AnkA plays an important role in facilitating intracellular infection by activating Abl-1 signalling pathway, and suggest a novel approach to treatment of human granulocytic anaplasmosis through inhibition of host cell signalling pathways.


Assuntos
Anaplasma phagocytophilum/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Tirosina/metabolismo , Anaplasma phagocytophilum/crescimento & desenvolvimento , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Ehrlichiose/microbiologia , Células HL-60 , Humanos , Imunoprecipitação , Microscopia Confocal , Peso Molecular , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-abl/genética , Técnicas do Sistema de Duplo-Híbrido
9.
Infect Immun ; 74(1): 108-17, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368963

RESUMO

Pathogenic Brucella species replicate within mammalian cells, and their type IV secretion system is essential for intracellular survival and replication. The options for biochemical studies on the Brucella secretion system are limited due to the rigidity of the cells and biosafety concerns, which preclude large-scale cell culture and fractionation. To overcome these problems, we heterologously expressed the Brucella suis virB operon in the closely related alpha(2)-proteobacterium Agrobacterium tumefaciens and showed that the VirB proteins assembled into a complex. Eight of the twelve VirB proteins were detected in the membranes of the heterologous host with specific antisera. Cross-linking indicated protein-protein interactions similar to those in other type IV secretion systems, and the results of immunofluorescence analysis supported the formation of VirB protein complexes in the cell envelope. Production of a subset of the B. suis VirB proteins (VirB3-VirB12) in A. tumefaciens strongly increased its ability to receive IncQ plasmid pLS1 in conjugation experiments, and production of VirB1 further enhanced the conjugation efficiency. Plasmid recipient competence correlated with periplasmic leakage and the detergent sensitivity of A. tumefaciens, suggesting a weakening of the cell envelope. Heterologous expression thus permits biochemical characterization of B. suis type IV secretion system assembly.


Assuntos
Agrobacterium tumefaciens/citologia , Agrobacterium tumefaciens/imunologia , Brucella suis/imunologia , Brucelose/imunologia , Brucelose/microbiologia , Plasmídeos/genética , Brucella suis/genética , Brucelose/metabolismo , Membrana Celular/imunologia , Imunofluorescência , Óperon
10.
J Bacteriol ; 188(23): 8222-30, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17012398

RESUMO

Agrobacterium tumefaciens and Agrobacterium rhizogenes are closely related plant pathogens that cause different diseases, crown gall and hairy root. Both diseases result from transfer, integration, and expression of plasmid-encoded bacterial genes located on the transferred DNA (T-DNA) in the plant genome. Bacterial virulence (Vir) proteins necessary for infection are also translocated into plant cells. Transfer of single-stranded DNA (ssDNA) and Vir proteins requires a type IV secretion system, a protein complex spanning the bacterial envelope. A. tumefaciens translocates the ssDNA-binding protein VirE2 into plant cells, where it binds single-stranded T-DNA and helps target it to the nucleus. Although some strains of A. rhizogenes lack VirE2, they are pathogenic and transfer T-DNA efficiently. Instead, these bacteria express the GALLS protein, which is essential for their virulence. The GALLS protein can complement an A. tumefaciens virE2 mutant for tumor formation, indicating that GALLS can substitute for VirE2. Unlike VirE2, GALLS contains ATP-binding and helicase motifs similar to those in TraA, a strand transferase involved in conjugation. Both GALLS and VirE2 contain nuclear localization sequences and a C-terminal type IV secretion signal. Here we show that mutations in any of these domains abolished the ability of GALLS to substitute for VirE2.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Núcleo Celular/metabolismo , Rhizobium/química , Transdução de Sinais , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/metabolismo , Teste de Complementação Genética , Canais Iônicos/metabolismo , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Doenças das Plantas/microbiologia , Raízes de Plantas/microbiologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Alinhamento de Sequência
11.
Proc Natl Acad Sci U S A ; 102(3): 832-7, 2005 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-15644442

RESUMO

Several human pathogens and the plant pathogen Agrobacterium tumefaciens use a type IV secretion system for translocation of effector proteins into host cells. How effector proteins are selected for transport is unknown, but a C-terminal transport signal is present in the proteins translocated by the A. tumefaciens VirB/D4 type IV secretion system. We characterized this signal in the virulence protein VirF by alanine scanning and further site-directed mutagenesis. The Cre recombinase was used as a reporter to measure the translocation efficiency of Cre-Vir fusions from A. tumefaciens to Arabidopsis. The data unambiguously showed that positive charge is an essential characteristic of the C-terminal transport signal. We increased the sensitivity of this translocation assay by modifying the Cre-induced readout in host cells from kanamycin resistance to GFP expression. This improvement allowed us to detect translocation of the VirD2 relaxase protein in the absence of transferred DNA, showing that attachment to the transferred DNA is not essential for transport by the VirB/D4 system. We also found another translocated effector protein, namely the VirD5 protein encoded by the tumor-inducing plasmid. According to secondary structure predictions, the C termini of all VirB/D4-translocated proteins identified so far are unstructured; however, they contain a characteristic hydropathic profile. Based on sequence alignments and mutational analysis of VirF, we conclude that the C-terminal transport signal for recruitment and translocation of effector proteins by the A. tumefaciens VirB/D4 system is hydrophilic and has a net positive charge with a consensus motif of R-X(7)-R-X-R-X-R-X-X(n)>.


Assuntos
Proteínas de Membrana Transportadoras/química , Sinais Direcionadores de Proteínas , Rhizobium/patogenicidade , Fatores de Virulência/química , Arabidopsis/química , Arabidopsis/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Transporte Biológico , Sequência Consenso , Genes Reporter , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Mutagênese Sítio-Dirigida , Sinais Direcionadores de Proteínas/genética , Rhizobium/química , Eletricidade Estática , Fatores de Virulência/metabolismo
12.
Yeast ; 19(6): 529-36, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11921101

RESUMO

Insertional mutagenesis is a powerful tool for the isolation of novel mutations. The gene delivery system of the bacterium Agrobacterium tumefaciens, which mediates transfer not only to plants but also to yeasts and fungi, could be exploited to generate collections of yeasts containing insertional mutations if there were no bias towards particular integration sites, as is the case in plants. To test this, we have analysed a small collection of Saccharomyces cerevisiae strains with T-DNA copies integrated in the S. cerevisiae genome. The position of 54 of these T-DNAs was determined. The T-DNA showed no clear preference for certain DNA sequences or genomic regions. We have isolated insertions in the coding regions of the genes YGR125w, YDR250c, YGR141w, YGR045c, YPL017c, YGR040w, YDL052c, YJL148w, YCL033c, YFL061w, YJR033c, YDR175c and YLR309c confirming that these genes are non-essential for S. cerevisiae haploid growth on minimal medium. Given the advantages of T-DNA, we propose its use as an ideal mobile DNA element for insertional mutagenesis in yeasts.


Assuntos
Agrobacterium tumefaciens/genética , DNA Bacteriano , DNA de Cadeia Simples , Genes Fúngicos , Haplótipos , Mutagênese Insercional , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento
13.
Mol Microbiol ; 47(4): 891-901, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12581347

RESUMO

Agrobacterium uses a mechanism similar to conjugation for trans-kingdom transfer of its oncogenic T-DNA. A defined VirB/VirD4 Type IV secretion system is responsible for such a genetic transfer. In addition, certain virulence proteins as VirE2 can be mobilized into host cells by the same apparatus. VirE2 is essential to achieve plant but not yeast transformation. We found that the limited host range plasmid CloDF13 can be recruited by the virulence apparatus of Agrobacterium for transfer to eukaryotic hosts. As expected the VirB transport complex was required for such trans-kingdom DNA transfer. However, unexpectedly, the coupling factor VirD4 turned out to be necessary for transfer to plants but not for transport into yeast. The CloDF13 encoded coupling factor (Mob) was essential for transfer to both plants and yeast though. This is interpreted by the different specificities of Mob and VirD4. Hence, Mob being required for the transport of the CloDF13 transferred DNA (to both plants and yeast) and VirD4 being required for transport of virulence proteins such as VirE2. Nevertheless, the presence of the VirE2 protein in the host plant was not sufficient to restore the deficiency for VirD4 in the transforming bacteria. We propose that Mob functions encoded by the plasmid CloDF13 are sufficient for DNA mobilization to eukaryotic cells but that the VirD4-mediated pathway is essential to achieve DNA nuclear establishment specifically in plants. This suggests that other Agrobacterium virulence proteins besides VirE2 are translocated and essential for plant transformation.


Assuntos
Agrobacterium tumefaciens/genética , Proteínas de Bactérias/genética , Bacteriocinas , Proteínas de Escherichia coli , Plantas/genética , Plantas/microbiologia , Transformação Genética , Fatores de Virulência , Agrobacterium tumefaciens/patogenicidade , Quimera/genética , DNA Bacteriano/genética , Proteínas de Ligação a DNA/genética , Genes Bacterianos , Teste de Complementação Genética , Canais Iônicos/genética , Mutação , Doenças das Plantas/genética , Doenças das Plantas/microbiologia , Plantas Geneticamente Modificadas , Plasmídeos/genética , Saccharomyces cerevisiae/genética , Nicotiana/genética , Nicotiana/microbiologia , Transativadores/genética , Virulência/genética
14.
Plant Physiol ; 133(3): 978-88, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14551327

RESUMO

Agrobacterium tumefaciens uses a type IV secretion system to deliver a nucleoprotein complex and effector proteins directly into plant cells. The single-stranded DNA-binding protein VirE2, the F-box protein VirF and VirE3 are delivered into host cells via this VirB/D4 encoded translocation system. VirE1 functions as a chaperone of VirE2 by regulating its efficient translation and preventing VirE2-VirE2 aggregation in the bacterial cell. We analyzed whether the VirE1 chaperone is also essential for transport recognition of VirE2 by the VirB/D4 encoded type IV secretion system. In addition, we assayed whether translocation of VirF and VirE3, which also forms part of the virE operon, is affected by the absence of VirE1. We employed the earlier developed CRAFT (Cre recombinase Reporter Assay For Translocation) assay to detect transfer of Cre::Vir fusion proteins from A. tumefaciens into plants, monitored by stable reconstitution of a kanamycin resistance marker, and into yeast, screened by loss of the URA3 gene. We show that the C-terminal 50 amino acids of VirE2 and VirE3 are sufficient to mediate Cre translocation into host cells, confirming earlier indications of a C-terminal transport signal. This transfer was independent of the presence or absence of VirE1. Besides, the translocation efficiency of VirF is not altered in a virE1 mutant. The results unambiguously show that the VirE1 chaperone is not essential for the recognition of the VirE2 transport signal by the transport system and the subsequent translocation across the bacterial envelope into host cells.


Assuntos
Agrobacterium tumefaciens/genética , Proteínas de Bactérias/metabolismo , Proteínas de Ligação a DNA/metabolismo , Canais Iônicos/metabolismo , Chaperonas Moleculares/metabolismo , Sinais Direcionadores de Proteínas/genética , Agrobacterium tumefaciens/patogenicidade , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA/genética , Deleção de Genes , Integrases/genética , Canais Iônicos/genética , Chaperonas Moleculares/genética , Tumores de Planta/genética , Tumores de Planta/microbiologia , Plasmídeos/genética , Transporte Proteico/genética , Deleção de Sequência , Transformação Genética/genética , Proteínas Virais/genética , Virulência/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA