Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Bacteriol ; 206(2): e0033723, 2024 02 22.
Article in English | MEDLINE | ID: mdl-38299858

ABSTRACT

Genome sequencing has demonstrated that Staphylococcus aureus encodes arginine biosynthetic genes argDCJBFGH synthesizing proteins that mediate arginine biosynthesis using glutamate as a substrate. Paradoxically, however, S. aureus does not grow in a defined, glutamate-replete medium lacking arginine and glucose (CDM-R). Studies from our laboratory have found that specific mutations are selected by S. aureus that facilitate growth in CDM-R. However, these selected mutants synthesize arginine utilizing proline as a substrate rather than glutamate. In this study, we demonstrate that the ectopic expression of the argDCJB operon supports the growth of S. aureus in CDM-R, thus documenting the functionality of this pathway. Furthermore, suppressor mutants of S. aureus JE2 putA::Tn, which is defective in synthesizing arginine from proline, were selected on CDM-R agar. Genome sequencing revealed that these mutants had compensatory mutations within both spoVG, encoding an ortholog of the Bacillus subtilis stage V sporulation protein, and sarA, encoding the staphylococcal accessory regulator. Transcriptional studies document that argD expression is significantly increased when JE2 spoVG sarA was grown in CDM-R. Lastly, we found that a mutation in ahrC was required to induce argD expression in JE2 spoVG sarA when grown in an arginine-replete medium (CDM), suggesting that AhrC also functions to repress argDCJB in an arginine-dependent manner. In conclusion, these data indicate that the argDCJB operon is functional when transcribed in vitro and that SNPs within potential putative regulatory proteins are required to alleviate the repression.IMPORTANCEAlthough Staphylococcus aureus has the capability to synthesize all 20 amino acids, it is phenotypically auxotrophic for several amino acids including arginine. This work identifies putative regulatory proteins, including SpoVG, SarA, and AhrC, that function to inhibit the arginine biosynthetic pathways using glutamate as a substrate. Understanding the ultimate mechanisms of why S. aureus is selected to repress arginine biosynthetic pathways even in the absence of arginine will add to the growing body of work assessing the interactions between metabolism and S. aureus pathogenesis.


Subject(s)
Glutamic Acid , Staphylococcus aureus , Staphylococcus aureus/metabolism , Glutamic Acid/metabolism , Arginine/metabolism , Bacterial Proteins/metabolism , Transcription Factors/metabolism , Amino Acids/metabolism , Proline/genetics , Proline/metabolism , Gene Expression Regulation, Bacterial
2.
PLoS Pathog ; 19(7): e1011536, 2023 07.
Article in English | MEDLINE | ID: mdl-37486930

ABSTRACT

Central metabolic pathways control virulence and antibiotic resistance, and constitute potential targets for antibacterial drugs. In Staphylococcus aureus the role of the pentose phosphate pathway (PPP) remains largely unexplored. Mutation of the 6-phosphogluconolactonase gene pgl, which encodes the only non-essential enzyme in the oxidative phase of the PPP, significantly increased MRSA resistance to ß-lactam antibiotics, particularly in chemically defined media with physiologically-relevant concentrations of glucose, and reduced oxacillin (OX)-induced lysis. Expression of the methicillin-resistance penicillin binding protein 2a and peptidoglycan architecture were unaffected. Carbon tracing and metabolomics revealed extensive metabolic reprogramming in the pgl mutant including increased flux to glycolysis, the TCA cycle, and several cell envelope precursors, which was consistent with increased ß-lactam resistance. Morphologically, pgl mutant cells were smaller than wild-type with a thicker cell wall and ruffled surface when grown in OX. The pgl mutation reduced resistance to Congo Red, sulfamethoxazole and oxidative stress, and increased resistance to targocil, fosfomycin and vancomycin. Levels of lipoteichoic acids (LTAs) were significantly reduced in pgl, which may limit cell lysis, while the surface charge of pgl cells was significantly more positive. A vraG mutation in pgl reversed the increased OX resistance phenotype, and partially restored wild-type surface charge, but not LTA levels. Mutations in vraF or graRS from the VraFG/GraRS complex that regulates DltABCD-mediated d-alanylation of teichoic acids (which in turn controls ß-lactam resistance and surface charge), also restored wild-type OX susceptibility. Collectively these data show that reduced levels of LTAs and OX-induced lysis combined with a VraFG/GraRS-dependent increase in cell surface positive charge are accompanied by significantly increased OX resistance in an MRSA pgl mutant.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Pentose Phosphate Pathway/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Oxacillin/pharmacology , Cell Wall/metabolism , Monobactams/metabolism , beta-Lactam Resistance/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Microbial Sensitivity Tests
3.
bioRxiv ; 2023 Mar 07.
Article in English | MEDLINE | ID: mdl-36945400

ABSTRACT

Central metabolic pathways controls virulence and antibiotic resistance, and constitute potential targets for antibacterial drugs. In Staphylococcus aureus the role of the pentose phosphate pathway (PPP) remains largely unexplored. Mutation of the 6-phosphogluconolactonase gene pgl, which encodes the only non-essential enzyme in the oxidative phase of the PPP, significantly increased MRSA resistance to ß-lactam antibiotics, particularly in chemically defined media with glucose, and reduced oxacillin (OX)-induced lysis. Expression of the methicillin-resistance penicillin binding protein 2a and peptidoglycan architecture were unaffected. Carbon tracing and metabolomics revealed extensive metabolic reprogramming in the pgl mutant including increased flux to glycolysis, the TCA cycle, and several cell envelope precursors, which was consistent with increased ß-lactam resistance. Morphologically, pgl mutant cells were smaller than wild-type with a thicker cell wall and ruffled surface when grown in OX. Further evidence of the pleiotropic effect of the pgl mutation was reduced resistance to Congo Red, sulfamethoxazole and oxidative stress, and increased resistance to targocil, fosfomycin and vancomycin. Reduced binding of wheat germ agglutinin (WGA) to pgl was indicative of lower wall teichoic acid/lipoteichoic acid levels or altered teichoic acid structures. Mutations in the vraFG or graRS loci reversed the increased OX resistance phenotype and restored WGA binding to wild-type levels. VraFG/GraRS was previously implicated in susceptibility to cationic antimicrobial peptides and vancomycin, and these data reveal a broader role for this multienzyme membrane complex in the export of cell envelope precursors or modifying subunits required for resistance to diverse antimicrobial agents. Altogether our study highlights important roles for the PPP and VraFG/GraRS in ß-lactam resistance, which will support efforts to identify new drug targets and reintroduce ß-lactams in combination with adjuvants or other antibiotics for infections caused by MRSA and other ß-lactam resistant pathogens. Author summary: High-level resistance to penicillin-type (ß-lactam) antibiotics significantly limits the therapeutic options for patients with MRSA infections necessitating the use of newer agents, for which reduced susceptibility has already been described. Here we report for the first time that the central metabolism pentose phosphate pathway controls MRSA resistance to penicillin-type antibiotics. We comprehensively demonstrated that mutation of the PPP gene pgl perturbed metabolism in MRSA leading to increased flux to cell envelope precursors to drive increased antibiotic resistance. Moreover, increased resistance was dependent on the VraRG/GraRS multienzyme membrane complex previously implicated in resistance to antimicrobial peptides and vancomycin. Our data thus provide new insights on MRSA mechanisms of ß-lactam resistance, which will support efforts to expand the treatment options for infections caused by this and other antimicrobial resistant pathogens.

4.
PLoS Pathog ; 19(1): e1011098, 2023 01.
Article in English | MEDLINE | ID: mdl-36652494

ABSTRACT

Proline acquired via specific transporters can serve as a proteinogenic substrate, carbon and nitrogen source, or osmolyte. Previous reports have documented that Staphylococcus aureus, a major community and nosocomial pathogen, encodes at least four proline transporters, PutP, OpuC, OpuD, and ProP. A combination of genetic approaches and 3H-proline transport assays reveal that a previously unrecognized transporter, ProT, in addition to PutP, are the major proline transporters in S. aureus. Complementation experiments using constitutively expressed non-cognate promoters found that proline transport via OpuD, OpuC, and ProP is minimal. Both proline biosynthesis from arginine and proline transport via ProT are critical for growth when S. aureus is grown under conditions of high salinity. Further, proline transport mediated by ProT or PutP are required for growth in media with and without glucose, indicating both transporters function to acquire proline for proteinogenic purposes in addition to acquisition of proline as a carbon/nitrogen source. Lastly, inactivation of proT and putP resulted in a significant reduction (5 log10) of bacterial burden in murine skin-and-soft tissue infection and bacteremia models, suggesting that proline transport is required to establish a S. aureus infection.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Mice , Staphylococcus aureus/genetics , Staphylococcal Infections/microbiology , Proline
5.
mBio ; 13(3): e0039522, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35475645

ABSTRACT

Previous studies have found that arginine biosynthesis in Staphylococcus aureus is repressed via carbon catabolite repression (CcpA), and proline is used as a precursor. Unexpectedly, however, robust growth of S. aureus is not observed in complete defined medium lacking both glucose and arginine (CDM-R). Mutants able to grow on agar-containing defined medium lacking arginine (CDM-R) were selected and found to contain mutations within ahrC, encoding the canonical arginine biosynthesis pathway repressor (AhrC), or single nucleotide polymorphisms (SNPs) upstream of the native arginine deiminase (ADI) operon arcA1B1D1C1. Reverse transcription-PCR (RT-PCR) studies found that mutations within ccpA or ahrC or SNPs identified upstream of arcA1B1D1C1 increased the transcription of both arcB1 and argGH, encoding ornithine carbamoyltransferase and argininosuccinate synthase/lyase, respectively, facilitating arginine biosynthesis. Furthermore, mutations within the AhrC homologue argR2 facilitated robust growth within CDM-R. Complementation with arcB1 or arcA1B1D1C1, but not argGH, rescued growth in CDM-R. Finally, supplementation of CDM-R with ornithine stimulated growth, as did mutations in genes (proC and rocA) that presumably increased the pyrroline-5-carboxylate and ornithine pools. Collectively, these data suggest that the transcriptional regulation of ornithine carbamoyltransferase and, in addition, the availability of intracellular ornithine pools regulate arginine biosynthesis in S. aureus in the absence of glucose. Surprisingly, ~50% of clinical S. aureus isolates were able to grow in CDM-R. These data suggest that S. aureus is selected to repress arginine biosynthesis in environments with or without glucose; however, mutants may be readily selected that facilitate arginine biosynthesis and growth in specific environments lacking arginine. IMPORTANCE Staphylococcus aureus can cause infection in virtually any niche of the human host, suggesting that it has significant metabolic versatility. Indeed, bioinformatic analysis suggests that it has the biosynthetic capability to synthesize all 20 amino acids. Paradoxically, however, it is conditionally auxotrophic for several amino acids, including arginine. Studies in our laboratory are designed to assess the biological function of amino acid auxotrophy in this significant pathogen. This study reveals that the metabolic block repressing arginine biosynthesis in media lacking glucose is the transcriptional repression of ornithine carbamoyltransferase encoded by arcB1 within the native arginine deiminase operon in addition to limited intracellular pools of ornithine. Surprisingly, approximately 50% of S. aureus clinical isolates can grow in media lacking arginine, suggesting that mutations are selected in S. aureus that allow growth in particular niches of the human host.


Subject(s)
Ornithine Carbamoyltransferase , Staphylococcus aureus , Amino Acids/metabolism , Arginine/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Glucose/metabolism , Ornithine/metabolism , Ornithine Carbamoyltransferase/genetics , Ornithine Carbamoyltransferase/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics
6.
mBio ; 12(3): e0053021, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34182779

ABSTRACT

Penicillin binding protein 2a (PBP2a)-dependent resistance to ß-lactam antibiotics in methicillin-resistant Staphylococcus aureus (MRSA) is regulated by the activity of the tricarboxylic acid (TCA) cycle via a poorly understood mechanism. We report that mutations in sucC and sucD, but not other TCA cycle enzymes, negatively impact ß-lactam resistance without changing PBP2a expression. Increased intracellular levels of succinyl coenzyme A (succinyl-CoA) in the sucC mutant significantly perturbed lysine succinylation in the MRSA proteome. Suppressor mutations in sucA or sucB, responsible for succinyl-CoA biosynthesis, reversed sucC mutant phenotypes. The major autolysin (Atl) was the most succinylated protein in the proteome, and increased Atl succinylation in the sucC mutant was associated with loss of autolytic activity. Although PBP2a and PBP2 were also among the most succinylated proteins in the MRSA proteome, peptidoglycan architecture and cross-linking were unchanged in the sucC mutant. These data reveal that perturbation of the MRSA succinylome impacts two interconnected cell wall phenotypes, leading to repression of autolytic activity and increased susceptibility to ß-lactam antibiotics. IMPORTANCEmecA-dependent methicillin resistance in MRSA is subject to regulation by numerous accessory factors involved in cell wall biosynthesis, nucleotide signaling, and central metabolism. Here, we report that mutations in the TCA cycle gene, sucC, increased susceptibility to ß-lactam antibiotics and was accompanied by significant accumulation of succinyl-CoA, which in turn perturbed lysine succinylation in the proteome. Although cell wall structure and cross-linking were unchanged, significantly increased succinylation of the major autolysin Atl, which was the most succinylated protein in the proteome, was accompanied by near complete repression of autolytic activity. These findings link central metabolism and levels of succinyl-CoA to the regulation of ß-lactam antibiotic resistance in MRSA through succinylome-mediated control of two interlinked cell wall phenotypes. Drug-mediated interference of the SucCD-controlled succinylome may help overcome ß-lactam resistance.


Subject(s)
Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , beta-Lactams/pharmacology , Acyl Coenzyme A/analysis , Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Mutation , Proteome , beta-Lactam Resistance
7.
J Infect Dis ; 221(6): 1000-1016, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31628459

ABSTRACT

Prolonging the clinical effectiveness of ß-lactams, which remain first-line antibiotics for many infections, is an important part of efforts to address antimicrobial resistance. We report here that inactivation of the predicted d-cycloserine (DCS) transporter gene cycA resensitized methicillin-resistant Staphylococcus aureus (MRSA) to ß-lactam antibiotics. The cycA mutation also resulted in hypersusceptibility to DCS, an alanine analogue antibiotic that inhibits alanine racemase and d-alanine ligase required for d-alanine incorporation into cell wall peptidoglycan. Alanine transport was impaired in the cycA mutant, and this correlated with increased susceptibility to oxacillin and DCS. The cycA mutation or exposure to DCS were both associated with the accumulation of muropeptides with tripeptide stems lacking the terminal d-ala-d-ala and reduced peptidoglycan cross-linking, prompting us to investigate synergism between ß-lactams and DCS. DCS resensitized MRSA to ß-lactams in vitro and significantly enhanced MRSA eradication by oxacillin in a mouse bacteremia model. These findings reveal alanine transport as a new therapeutic target to enhance the susceptibility of MRSA to ß-lactam antibiotics.


Subject(s)
Alanine/metabolism , Anti-Bacterial Agents/pharmacology , Cycloserine/pharmacology , Methicillin Resistance/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , beta-Lactams/pharmacology , Animals , Antimetabolites/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacteriological Techniques , Biological Transport , Female , Gene Expression Regulation, Bacterial/drug effects , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mutation , Polysaccharides/chemistry , Polysaccharides/metabolism , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
8.
Proc Natl Acad Sci U S A ; 117(1): 761-770, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871206

ABSTRACT

Small RNAs (sRNAs) associate with Argonaute (AGO) proteins in effector complexes, termed RNA-induced silencing complexes (RISCs), which regulate complementary transcripts by translation inhibition and/or RNA degradation. In the unicellular alga Chlamydomonas, several metazoans, and land plants, emerging evidence indicates that polyribosome-associated transcripts can be translationally repressed by RISCs without substantial messenger RNA (mRNA) destabilization. However, the mechanism of translation inhibition in a polyribosomal context is not understood. Here we show that Chlamydomonas VIG1, an ortholog of the Drosophila melanogaster Vasa intronic gene (VIG), is required for this process. VIG1 localizes predominantly in the cytosol and comigrates with monoribosomes and polyribosomes by sucrose density gradient sedimentation. A VIG1-deleted mutant shows hypersensitivity to the translation elongation inhibitor cycloheximide, suggesting that VIG1 may have a nonessential role in ribosome function/structure. Additionally, FLAG-tagged VIG1 copurifies with AGO3 and Dicer-like 3 (DCL3), consistent with it also being a component of the RISC. Indeed, VIG1 is necessary for the repression of sRNA-targeted transcripts at the translational level but is dispensable for cleavage-mediated RNA interference and for the association of the AGO3 effector with polyribosomes or target transcripts. Our results suggest that VIG1 is an ancillary ribosomal component and plays a role in sRNA-mediated translation repression of polyribosomal transcripts.


Subject(s)
Chlamydomonas reinhardtii/physiology , Plant Proteins/metabolism , Protein Biosynthesis/physiology , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/metabolism , Argonaute Proteins/metabolism , Cycloheximide/pharmacology , Cytosol/metabolism , Gene Expression Regulation, Plant , Introns/genetics , Mutation , Plant Proteins/genetics , Plants, Genetically Modified , Polyribosomes/genetics , Polyribosomes/metabolism , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Ribosomes/metabolism
9.
Nucleic Acids Res ; 46(17): 8803-8816, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29986060

ABSTRACT

RsaE is a regulatory RNA highly conserved amongst Firmicutes that lowers the amount of mRNAs associated with the TCA cycle and folate metabolism. A search for new RsaE targets in Staphylococcus aureus revealed that in addition to previously described substrates, RsaE down-regulates several genes associated with arginine catabolism. In particular, RsaE targets the arginase rocF mRNA via direct interactions involving G-rich motifs. Two duplicated C-rich motifs of RsaE can independently downregulate rocF expression. The faster growth rate of ΔrsaE compared to its parental strain in media containing amino acids as sole carbon source points to an underlying role for RsaE in amino acid catabolism. Collectively, the data support a model in which RsaE acts as a global regulator of functions associated with metabolic adaptation.


Subject(s)
Arginine/metabolism , RNA, Bacterial/physiology , Regulatory Sequences, Ribonucleic Acid , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Amino Acids/metabolism , Amino Acids/pharmacology , Conserved Sequence , Culture Media/chemistry , Culture Media/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Organisms, Genetically Modified , Regulatory Sequences, Ribonucleic Acid/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
10.
mBio ; 8(4)2017 08 01.
Article in English | MEDLINE | ID: mdl-28765220

ABSTRACT

Macrophage-derived nitric oxide (NO·) is a crucial effector against invading pathogens. Yet, paradoxically, several bacterial species, including some pathogens, are known to endogenously produce NO· via nitric oxide synthase (NOS) activity, despite its apparent cytotoxicity. Here, we reveal a conserved role for bacterial NOS in activating aerobic respiration. We demonstrate that nitrite generated from endogenous NO· decomposition stimulates quinol oxidase activity in Staphylococcus aureus and increases the rate of cellular respiration. This not only supports optimal growth of this organism but also prevents a dysbalance in central metabolism. Further, we also show that activity of the SrrAB two-component system alleviates the physiological defects of the nos mutant. Our findings suggest that NOS and SrrAB constitute two distinct but functionally redundant routes for controlling staphylococcal respiration during aerobic growth.IMPORTANCE Despite its potential autotoxic effects, several bacterial species, including pathogenic staphylococcal species, produce NO· endogenously through nitric oxide synthase (NOS) activity. Therefore, how endogenous NO· influences bacterial fitness remains unclear. Here we show that the oxidation of NO· to nitrite increases aerobic respiration and consequently optimizes central metabolism to favor growth. Importantly, we also demonstrate that cells have a "fail-safe" mechanism that can maintain respiratory activity through the SrrAB two-component signaling regulon should NOS-derived nitrite levels decrease. These findings identify NOS and SrrAB as critical determinants of staphylococcal respiratory control and highlight their potential as therapeutic targets.


Subject(s)
Nitric Oxide Synthase/metabolism , Nitrites/metabolism , Staphylococcus aureus/enzymology , Aerobiosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Electron Transport Complex IV/metabolism , Mutation , Nitric Oxide/metabolism , Nitric Oxide Synthase/genetics , Oxidation-Reduction , Oxidative Stress , Oxidoreductases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism
11.
Plant Biotechnol J ; 15(2): 227-236, 2017 02.
Article in English | MEDLINE | ID: mdl-27496594

ABSTRACT

Soya bean (Glycine max (L.) Merr.) is sought after for both its oil and protein components. Genetic approaches to add value to either component are ongoing efforts in soya bean breeding and molecular biology programmes. The former is the primary vegetable oil consumed in the world. Hence, its primary usage is in direct human consumption. As a means to increase its utility in feed applications, thereby expanding the market of soya bean coproducts, we investigated the simultaneous displacement of marine ingredients in aquafeeds with soya bean-based protein and a high Omega-3 fatty acid soya bean oil, enriched with alpha-linolenic and stearidonic acids, in both steelhead trout (Oncorhynchus mykiss) and Kampachi (Seriola rivoliana). Communicated herein are aquafeed formulations with major reduction in marine ingredients that translates to more total Omega-3 fatty acids in harvested flesh. Building off of these findings, subsequent efforts were directed towards a genetic strategy that would translate to a prototype design of an optimal identity-preserved soya bean-based feedstock for aquaculture, whereby a multigene stack approach for the targeted synthesis of two value-added output traits, eicosapentaenoic acid and the ketocarotenoid, astaxanthin, were introduced into the crop. To this end, the systematic introduction of seven transgenic cassettes into soya bean, and the molecular and phenotypic evaluation of the derived novel events are described.


Subject(s)
Animal Feed , Aquaculture/methods , Fishes/metabolism , Glycine max/growth & development , Animal Nutritional Physiological Phenomena , Animals , Eicosapentaenoic Acid/metabolism , Fatty Acids, Omega-3/administration & dosage , Oncorhynchus mykiss/metabolism , Plant Oils , Plants, Genetically Modified , Soybean Oil/administration & dosage , Glycine max/genetics , Xanthophylls/metabolism , alpha-Linolenic Acid
12.
Plant Physiol ; 159(2): 710-20, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496509

ABSTRACT

Multicellular eukaryotes demonstrate nongenetic, heritable phenotypic versatility in their adaptation to environmental changes. This inclusive inheritance is composed of interacting epigenetic, maternal, and environmental factors. Yet-unidentified maternal effects can have a pronounced influence on plant phenotypic adaptation to changing environmental conditions. To explore the control of phenotypy in higher plants, we examined the effect of a single plant nuclear gene on the expression and transmission of phenotypic variability in Arabidopsis (Arabidopsis thaliana). MutS HOMOLOG1 (MSH1) is a plant-specific nuclear gene product that functions in both mitochondria and plastids to maintain genome stability. RNA interference suppression of the gene elicits strikingly similar programmed changes in plant growth pattern in six different plant species, changes subsequently heritable independent of the RNA interference transgene. The altered phenotypes reflect multiple pathways that are known to participate in adaptation, including altered phytohormone effects for dwarfed growth and reduced internode elongation, enhanced branching, reduced stomatal density, altered leaf morphology, delayed flowering, and extended juvenility, with conversion to perennial growth pattern in short days. Some of these effects are partially reversed with the application of gibberellic acid. Genetic hemicomplementation experiments show that this phenotypic plasticity derives from changes in chloroplast state. Our results suggest that suppression of MSH1, which occurs under several forms of abiotic stress, triggers a plastidial response process that involves nongenetic inheritance.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/growth & development , Chloroplasts/metabolism , MutS DNA Mismatch-Binding Protein/metabolism , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Base Sequence , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chloroplasts/genetics , DNA Methylation , Flowers/growth & development , Flowers/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant , Genes, Plant , Genetic Complementation Test/methods , Gibberellins/pharmacology , Inheritance Patterns , Mitochondria/genetics , Mitochondria/metabolism , Molecular Sequence Data , MutS DNA Mismatch-Binding Protein/genetics , Phenotype , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , RNA Interference , Sorghum/drug effects , Sorghum/genetics , Sorghum/growth & development , Sorghum/metabolism , Stress, Physiological , Transcription, Genetic , Transgenes
13.
Plant J ; 38(1): 107-18, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15053764

ABSTRACT

Viroid infection is associated with the production of short interfering RNAs (siRNAs), a hallmark of post-transcriptional gene silencing (PTGS). However, viroid RNAs autonomously replicating in the nucleus have not been shown to trigger the degradation of homologous RNA in the cytoplasm. To investigate the potential of viroids for the induction of gene silencing, non-infectious fragments of potato spindle tuber viroid (PSTVd) cDNA were transcriptionally fused to the 3' end of the green fluorescent protein (GFP)-coding region. Introduction of such constructs into tobacco plants resulted in stable transgene expression. Upon PSTVd infection, transgene expression was suppressed and partial de novo methylation of the transgene was observed. PSTVd-specific siRNA was detected but none was found corresponding to the gfp gene. Methylation was restricted almost entirely to the PSTVd-specific part of the transgene. Neither a gfp transgene construct lacking viroid-specific elements was silenced nor was de novo methylation detected, when it was introduced into the genetic background of the PSTVd-infected plant lines containing silenced GFP:PSTVd transgenes. The absence of gfp-specific siRNAs and of significant methylation within the gfp-coding region demonstrated that neither silencing nor DNA methylation spread from the initiator region into adjacent 5' regions.


Subject(s)
Nicotiana/genetics , Base Sequence , DNA Methylation , DNA, Complementary/genetics , DNA, Plant/chemistry , DNA, Plant/genetics , DNA, Viral/genetics , Gene Expression , Green Fluorescent Proteins , Luminescent Proteins/genetics , Phenotype , Plants, Genetically Modified , RNA Interference , RNA, Small Interfering/genetics , RNA, Viral/genetics , Recombinant Fusion Proteins/genetics , Nicotiana/metabolism , Nicotiana/virology , Viroids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...