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










Publication year range
1.
bioRxiv ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38979312

ABSTRACT

Sweet's syndrome is a poorly understood inflammatory skin disease characterized by neutrophil infiltration to the dermis. Single-nucleus and bulk transcriptomics of archival clinical samples of Sweet's syndrome revealed a prominent interferon signature in Sweet's syndrome skin that was reduced in tissue from other neutrophilic dermatoses. This signature was observed in different subsets of cells, including fibroblasts that expressed interferon-induced genes. Functionally, this response was supported by analysis of cultured primary human dermal fibroblasts that were observed to highly express neutrophil chemokines in response to activation by type I interferon. Furthermore, single-molecule resolution spatial transcriptomics of skin in Sweet's syndrome identified positionally distinct immune acting fibroblasts that included a CXCL1+ subset proximal to neutrophils and a CXCL12+ subset distal to the neutrophilic infiltrate. This study defines the cellular landscape of neutrophilic dermatoses and suggests dermal immune acting fibroblasts play a role in the pathogenesis of Sweet's syndrome through recognition of type I interferons.

2.
Bioorg Med Chem Lett ; 46: 128171, 2021 08 15.
Article in English | MEDLINE | ID: mdl-34098081

ABSTRACT

We have previously reported the development of indole-based CNS-active antivirals for the treatment of neurotropic alphavirus infection, but further optimization is impeded by a lack of knowledge of the molecular target and binding site. Herein we describe the design, synthesis and evaluation of a series of conformationally restricted analogues with the dual objectives of improving potency/selectivity and identifying the most bioactive conformation. Although this campaign was only modestly successful at improving potency, the sharply defined SAR of the rigid analogs enabled the definition of a three-dimensional pharmacophore, which we believe will be of value in further analog design and virtual screening for alternative antiviral leads.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Indoles/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Drug Design , Indoles/chemical synthesis , Indoles/chemistry , Microbial Sensitivity Tests , Molecular Conformation , Structure-Activity Relationship , Virus Replication/drug effects
3.
Front Cell Dev Biol ; 9: 630188, 2021.
Article in English | MEDLINE | ID: mdl-33644069

ABSTRACT

Flavin-Containing Monooxygenases are conserved xenobiotic-detoxifying enzymes. Recent studies have revealed endogenous functions of FMOs in regulating longevity in Caenorhabditis elegans and in regulating aspects of metabolism in mice. To explore the cellular mechanisms of FMO's endogenous function, here we demonstrate that all five functional mammalian FMOs may play similar endogenous roles to improve resistance to a wide range of toxic stresses in both kidney and liver cells. We further find that stress-activated c-Jun N-terminal kinase activity is enhanced in FMO-overexpressing cells, which may lead to increased survival under stress. Furthermore, FMO expression modulates cellular metabolic activity as measured by mitochondrial respiration, glycolysis, and metabolomics analyses. FMO expression augments mitochondrial respiration and significantly changes central carbon metabolism, including amino acid and energy metabolism pathways. Together, our findings demonstrate an important endogenous role for the FMO family in regulation of cellular stress resistance and major cellular metabolic activities including central carbon metabolism.

4.
PLoS Genet ; 11(10): e1005564, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26447709

ABSTRACT

Yeast pseudohyphal filamentation is a stress-responsive growth transition relevant to processes required for virulence in pathogenic fungi. Pseudohyphal growth is controlled through a regulatory network encompassing conserved MAPK (Ste20p, Ste11p, Ste7p, Kss1p, and Fus3p), protein kinase A (Tpk2p), Elm1p, and Snf1p kinase pathways; however, the scope of these pathways is not fully understood. Here, we implemented quantitative phosphoproteomics to identify each of these signaling networks, generating a kinase-dead mutant in filamentous S. cerevisiae and surveying for differential phosphorylation. By this approach, we identified 439 phosphoproteins dependent upon pseudohyphal growth kinases. We report novel phosphorylation sites in 543 peptides, including phosphorylated residues in Ras2p and Flo8p required for wild-type filamentous growth. Phosphoproteins in these kinase signaling networks were enriched for ribonucleoprotein (RNP) granule components, and we observe co-localization of Kss1p, Fus3p, Ste20p, and Tpk2p with the RNP component Igo1p. These kinases localize in puncta with GFP-visualized mRNA, and KSS1 is required for wild-type levels of mRNA localization in RNPs. Kss1p pathway activity is reduced in lsm1Δ/Δ and pat1Δ/Δ strains, and these genes encoding P-body proteins are epistatic to STE7. The P-body protein Dhh1p is also required for hyphal development in Candida albicans. Collectively, this study presents a wealth of data identifying the yeast phosphoproteome in pseudohyphal growth and regulatory interrelationships between pseudohyphal growth kinases and RNPs.


Subject(s)
Hyphae/genetics , Phosphotransferases/biosynthesis , Ribonucleoproteins/biosynthesis , Saccharomyces cerevisiae/genetics , Candida albicans/genetics , Gene Expression Regulation, Fungal , Hyphae/growth & development , Phenotype , Phosphorylation , Phosphotransferases/genetics , Ribonucleoproteins/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
5.
Bioorg Med Chem ; 23(7): 1569-87, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740634

ABSTRACT

Neurotropic alphaviruses are debilitating pathogens that infect the central nervous system (CNS) and are transmitted to humans via mosquitoes. There exist no effective human vaccines against these viruses, underlining the need for effective antivirals, but no antiviral drugs are available for treating infection once the viruses have invaded the CNS. Previously, we reported the development of novel indole-2-carboxamide-based inhibitors of alphavirus replication that demonstrate significant reduction of viral titer and achieve measurable brain permeation in a pharmacokinetic mouse model. Herein we report our continued efforts to improve physicochemical properties predictive of in vivo blood-brain barrier (BBB) permeability through reduction of overall molecular weight, replacing the indole core with a variety of aromatic and non-aromatic monocyclics. These studies culminated in the identification of simple anthranilamides that retain excellent potency with improved metabolic stability and significantly greater aqueous solubility. Furthermore, in a live virus study, we showed that two new compounds were capable of reducing viral titer by two orders of magnitude and that these compounds likely exert their effects through a mechanism similar to that of our indole-2-carboxamide inhibitors.


Subject(s)
Alphavirus/drug effects , Antiviral Agents/pharmacology , Drug Discovery/methods , Virus Replication/drug effects , ortho-Aminobenzoates/pharmacology , Alphavirus/physiology , Animals , Antiviral Agents/chemistry , Mice , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Microsomes, Liver/virology , Virus Replication/physiology , ortho-Aminobenzoates/chemistry
6.
J Virol ; 88(19): 11199-214, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031353

ABSTRACT

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, Western Equine/drug effects , Encephalomyelitis, Equine/drug therapy , Indoles/pharmacology , Pyridines/pharmacology , Virus Replication/drug effects , Animals , Antiviral Agents/chemical synthesis , Bunyaviridae/drug effects , Bunyaviridae/growth & development , Cell Line , Encephalitis Virus, Western Equine/growth & development , Encephalitis Virus, Western Equine/pathogenicity , Encephalomyelitis, Equine/mortality , Encephalomyelitis, Equine/virology , Female , Indoles/chemical synthesis , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/virology , Paramyxoviridae/drug effects , Paramyxoviridae/growth & development , Picornaviridae/drug effects , Picornaviridae/growth & development , Protein Biosynthesis/drug effects , Pyridines/chemical synthesis , Replicon/drug effects , Structure-Activity Relationship , Survival Analysis
7.
PLoS One ; 8(12): e82318, 2013.
Article in English | MEDLINE | ID: mdl-24349254

ABSTRACT

Natural products provide a vast array of chemical structures to explore in the discovery of new medicines. Although secondary metabolites produced by microbes have been developed to treat a variety of diseases, including bacterial and fungal infections, to date there has been limited investigation of natural products with antiviral activity. In this report, we used a phenotypic cell-based replicon assay coupled with an iterative biochemical fractionation process to identify, purify, and characterize antiviral compounds produced by marine microbes. We isolated a compound from Streptomyces kaviengensis, a novel actinomycetes isolated from marine sediments obtained off the coast of New Ireland, Papua New Guinea, which we identified as antimycin A1a. This compound displays potent activity against western equine encephalitis virus in cultured cells with half-maximal inhibitory concentrations of less than 4 nM and a selectivity index of greater than 550. Our efforts also revealed that several antimycin A analogues display antiviral activity, and mechanism of action studies confirmed that these Streptomyces-derived secondary metabolites function by inhibiting the cellular mitochondrial electron transport chain, thereby suppressing de novo pyrimidine synthesis. Furthermore, we found that antimycin A functions as a broad spectrum agent with activity against a wide range of RNA viruses in cultured cells, including members of the Togaviridae, Flaviviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Finally, we demonstrate that antimycin A reduces central nervous system viral titers, improves clinical disease severity, and enhances survival in mice given a lethal challenge with western equine encephalitis virus. Our results provide conclusive validation for using natural product resources derived from marine microbes as source material for antiviral drug discovery, and they indicate that host mitochondrial electron transport is a viable target for the continued development of broadly active antiviral compounds.


Subject(s)
Actinobacteria/chemistry , Antiviral Agents/pharmacology , Geologic Sediments/microbiology , Animals , Antimycin A/chemistry , Antimycin A/pharmacology , Antimycin A/therapeutic use , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Biological Products/chemistry , Biological Products/isolation & purification , Biological Products/pharmacology , Cell Line , Central Nervous System/drug effects , Central Nervous System/pathology , Central Nervous System/virology , Chemical Fractionation , Electron Transport/drug effects , Encephalitis Viruses/drug effects , Encephalitis, Arbovirus/drug therapy , Encephalitis, Arbovirus/pathology , Encephalitis, Arbovirus/virology , High-Throughput Screening Assays , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Viral/metabolism , Reference Standards , Reproducibility of Results , Streptomyces/chemistry , Survival Analysis , Transcription, Genetic/drug effects
8.
J Med Chem ; 56(22): 9222-41, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24151954

ABSTRACT

Neurotropic alphaviruses, which include western equine encephalitis virus (WEEV) and Fort Morgan virus, are mosquito-borne pathogens that infect the central nervous system causing acute and potentially fatal encephalitis. We previously reported a novel series of indole-2-carboxamides as alphavirus replication inhibitors, one of which conferred protection against neuroadapted Sindbis virus infection in mice. We describe here further development of this series, resulting in 10-fold improvement in potency in a WEEV replicon assay and up to 40-fold increases in half-lives in mouse liver microsomes. Using a rhodamine123 uptake assay in MDR1-MDCKII cells, we were able to identify structural modifications that markedly reduce recognition by P-glycoprotein, the key efflux transporter at the blood-brain barrier. In a preliminary mouse PK study, we were able to demonstrate that two new analogues could achieve higher and/or longer plasma drug exposures than our previous lead and that one compound achieved measurable drug levels in the brain.


Subject(s)
Drug Design , Encephalitis Virus, Western Equine/drug effects , Encephalitis Virus, Western Equine/physiology , Indoles/chemistry , Indoles/pharmacology , Virus Replication/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Indoles/metabolism , Indoles/pharmacokinetics , Mice , Molecular Weight , Solubility , Structure-Activity Relationship
9.
Genetics ; 193(4): 1297-310, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23410832

ABSTRACT

The budding yeast Saccharomyces cerevisiae can respond to nutritional and environmental stress by implementing a morphogenetic program wherein cells elongate and interconnect, forming pseudohyphal filaments. This growth transition has been studied extensively as a model signaling system with similarity to processes of hyphal development that are linked with virulence in related fungal pathogens. Classic studies have identified core pseudohyphal growth signaling modules in yeast; however, the scope of regulatory networks that control yeast filamentation is broad and incompletely defined. Here, we address the genetic basis of yeast pseudohyphal growth by implementing a systematic analysis of 4909 genes for overexpression phenotypes in a filamentous strain of S. cerevisiae. Our results identify 551 genes conferring exaggerated invasive growth upon overexpression under normal vegetative growth conditions. This cohort includes 79 genes lacking previous phenotypic characterization. Pathway enrichment analysis of the gene set identifies networks mediating mitogen-activated protein kinase (MAPK) signaling and cell cycle progression. In particular, overexpression screening suggests that nuclear export of the osmoresponsive MAPK Hog1p may enhance pseudohyphal growth. The function of nuclear Hog1p is unclear from previous studies, but our analysis using a nuclear-depleted form of Hog1p is consistent with a role for nuclear Hog1p in repressing pseudohyphal growth. Through epistasis and deletion studies, we also identified genetic relationships with the G2 cyclin Clb2p and phenotypes in filamentation induced by S-phase arrest. In sum, this work presents a unique and informative resource toward understanding the breadth of genes and pathways that collectively constitute the molecular basis of filamentation.


Subject(s)
Gene Regulatory Networks , Genome, Fungal , Hyphae/growth & development , Saccharomyces cerevisiae/genetics , Cyclin B/genetics , Cyclin B/metabolism , Epistasis, Genetic , Gene Deletion , Hyphae/genetics , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phenotype , S Phase/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic
10.
PLoS Genet ; 7(4): e1002058, 2011 Apr.
Article in English | MEDLINE | ID: mdl-22103005

ABSTRACT

The morphogenetic transition between yeast and filamentous forms of the human fungal pathogen Candida albicans is regulated by a variety of signaling pathways. How these pathways interact to orchestrate morphogenesis, however, has not been as well characterized. To address this question and to identify genes that interact with the Regulation of Ace2 and Morphogenesis (RAM) pathway during filamentation, we report the first large-scale genetic interaction screen in C. albicans.Our strategy for this screen was based on the concept of complex haploinsufficiency (CHI). A heterozygous mutant of CBK1(cbk1Δ/CBK1), a key RAM pathway protein kinase, was subjected to transposon-mediated, insertional mutagenesis. The resulting double heterozygous mutants (6,528 independent strains) were screened for decreased filamentation on SpiderMedium (SM). From the 441 mutants showing altered filamentation, 139 transposon insertion sites were sequenced,yielding 41 unique CBK1-interacting genes. This gene set was enriched in transcriptional targets of Ace2 and, strikingly, the cAMP-dependent protein kinase A (PKA) pathway, suggesting an interaction between these two pathways. Further analysis indicates that the RAM and PKA pathways co-regulate a common set of genes during morphogenesis and that hyperactivation of the PKA pathway may compensate for loss of RAM pathway function. Our data also indicate that the PKA­regulated transcription factor Efg1 primarily localizes to yeast phase cells while the RAM­pathway regulated transcription factor Ace2 localizes to daughter nuclei of filamentous cells, suggesting that Efg1 and Ace2 regulate a common set of genes at separate stages of morphogenesis. Taken together, our observations indicate that CHI­based screening is a useful approach to genetic interaction analysis in C. albicans and support a model in which these two pathways regulate a common set of genes at different stages of filamentation.


Subject(s)
Candida albicans/genetics , Gene Expression Regulation, Fungal , Haploinsufficiency , Morphogenesis , Candida albicans/growth & development , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genes, Fungal , Genomic Library , Heterozygote , Hyphae/growth & development , Hyphae/metabolism , Mutagenesis, Insertional , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
11.
J Biol Chem ; 285(20): 15476-15488, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20228058

ABSTRACT

Yeast filamentous growth is a stress response to conditions of nitrogen deprivation, wherein yeast colonies form pseudohyphal filaments of elongated and connected cells. As proteins mediating adhesion and transport are required for this growth transition, we expect that the protein complement at the yeast cell periphery plays a critical and tightly regulated role in pseudohyphal filamentation. To identify proteins differentially abundant at the yeast cell periphery during pseudohyphal growth, we generated quantitative proteomic profiles of plasma membrane protein preparations under conditions of vegetative growth and filamentation. By isobaric tags for relative and absolute quantification chemistry and two-dimensional liquid chromatography-tandem mass spectrometry, we profiled 2463 peptides and 356 proteins, identifying 11 differentially abundant proteins that localize to the yeast cell periphery. This protein set includes Ylr414cp, herein renamed Pun1p, a previously uncharacterized protein localized to the plasma membrane compartment of Can1. Pun1p abundance is doubled under conditions of nitrogen stress, and deletion of PUN1 abolishes filamentous growth in haploids and diploids; pun1Delta mutants are noninvasive, lack surface-spread filamentation, grow slowly, and exhibit impaired cell adhesion. Conversely, overexpression of PUN1 results in exaggerated cell elongation under conditions of nitrogen stress. PUN1 contributes to yeast nitrogen signaling, as pun1Delta mutants misregulate amino acid biosynthetic genes during nitrogen stress. By chromatin immunoprecipitation and reverse transcription-PCR, we find that the filamentous growth factor Mss11p directly binds the PUN1 promoter and regulates its transcription. In total, this study provides the first profile of differential protein abundance during pseudohyphal growth, identifying a previously uncharacterized membrane compartment of Can1 protein required for wild-type nitrogen signaling and filamentous growth.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Cell Adhesion , Chromatin Immunoprecipitation , Chromatography, High Pressure Liquid , Mass Spectrometry , Membrane Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/growth & development , Spectrophotometry, Ultraviolet
12.
Cell Biochem Biophys ; 53(3): 127-34, 2009.
Article in English | MEDLINE | ID: mdl-19159085

ABSTRACT

In eukaryotes, reversible shuttling between the nucleus and cytoplasm is an important regulatory mechanism, particularly for many kinases and transcription factors. Inspired by the natural system, we recently developed a technology to control protein position in budding yeast using a chemical inducer of dimerization (CID). In this method, a nuclear export or localization signal is reversibly appended to a protein of interest by the CID, which effectively places its subcellular location under direct control of the chemical stimulus. Here, we explicitly tested the ability of this system to direct the nucleocytoplasmic transport of a panel of 16 representative kinases and transcription factors. From this set, we found that 12 targets (75%) are susceptible to re-positioning, suggesting that this method might be applicable to a range of targets. Interestingly, the four proteins that resisted mislocalization (Fun20p, Hcm1p, Pho4p, and Ste12p) are known to engage in a large number of protein-protein contacts. We suspect that, for these highly connected targets, the strength of the chemical signal may be insufficient to drive mislocalization and that proteins with relatively few partners might be most amenable to this approach. Collectively, these studies provide a necessary framework for the design of large-scale applications.


Subject(s)
Cell Nucleus/metabolism , Protein Multimerization/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Sirolimus/pharmacology , Active Transport, Cell Nucleus/drug effects , Biomimetics , Drug Resistance, Fungal , Genes, Fungal/genetics , Phenotype , Phosphotransferases/chemistry , Phosphotransferases/metabolism , Protein Structure, Quaternary/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Transcription Factors/chemistry , Transcription Factors/metabolism
13.
Yeast ; 25(8): 577-94, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18668531

ABSTRACT

Protein localization is tightly linked with function, such that the subcellular distribution of a protein serves as an important control point regulating activity. Exploiting this regulatory mechanism, we present here a general approach by which protein location, and hence function, may be controlled on demand in the budding yeast. In this system a small molecule, rapamycin, is used to temporarily recruit a strong cellular address signal to the target protein, placing subcellular localization under control of the selective chemical stimulus. The kinetics of this system are rapid: rapamycin-directed nucleo-cytoplasmic transport is evident 10-12 min post-treatment and the process is reversible upon removal of rapamycin. Accordingly, we envision this platform as a promising approach for the systematic construction of conditional loss-of-function mutants. As proof of principle, we used this system to direct nuclear export of the essential heat shock transcription factor Hsf1p, thereby mimicking the cell-cycle arrest phenotype of an hsf1 temperature-sensitive mutant. Our drug-induced localization platform also provides a method by which protein localization can be uncoupled from endogenous cell signalling events, addressing the necessity or sufficiency of a given localization shift for a particular cell process. To illustrate, we directed the nuclear import of the calcineurin-dependent transcription factor Crz1p in the absence of native stimuli; this analysis directly substantiates that nuclear translocation of this protein is insufficient for its transcriptional activity. In total, this technology represents a powerful method for the generation of conditional alleles and directed mislocalization studies in yeast, with potential applicability on a genome-wide scale.


Subject(s)
Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Active Transport, Cell Nucleus/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal , Heat-Shock Proteins/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Phenotype , Protein Transport/drug effects , Saccharomyces cerevisiae/genetics , Signal Transduction/genetics , Sirolimus/pharmacokinetics , Sirolimus/pharmacology , Transcription Factors/metabolism
14.
Autophagy ; 4(6): 792-800, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18497569

ABSTRACT

Plasmid-based collections of fluorescent protein fusions are valuable and versatile resources, facilitating systematic studies of protein localization in multiple genetic backgrounds. At present, however, few such collections exist for the analysis of protein localization in any organism. To address this deficiency, we present here a plasmid-based set of resources for the analysis of protein localization in the budding yeast. Specifically, we constructed a suite of low-copy destination vectors for recombination-based cloning of yeast genes as fluorescent protein fusions. We cloned a set of 384 yeast genes encoding kinases, transcription factors and signaling proteins as "recombination-ready" cassettes; by Gateway cloning, these genes with native promoters can be easily introduced into the destination vectors described above, generating carboxy-terminal fusions to fluorescent proteins. Using these reagents, we constructed a subcollection of 276 genes encoding carboxy-terminal fusions to yellow fluorescent protein (vYFP). This collection encompasses 14 autophagy-related (ATG) genes, and we localized these Atgp-vYFP chimeras during rapamycin-induced autophagy. To illustrate further the utility of this collection as a tool in exploring the functions and interactions of proteins in a pathway, we localized a subset of these Atg-vYFP chimeras in a strain deleted for the scaffolding protein Atg11p. In addition, we validated previous results identifying the integral membrane protein Atg9p at the pre-autophagosomal structure upon overexpression of ATG11 and upon deletion of ATG1. Collectively, this plasmid-based resource of yeast gene-vYFP fusions provides an initial toolkit for a variety of systematic and large-scale localization studies exploring pathway biology in the budding yeast.


Subject(s)
Autophagy/physiology , Luminescent Proteins/metabolism , Plasmids/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Antifungal Agents/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Luminescent Proteins/genetics , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sirolimus/metabolism
15.
Mol Biol Cell ; 19(7): 2708-17, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417610

ABSTRACT

The subcellular distribution of kinases and other signaling proteins is regulated in response to cellular cues; however, the extent of this regulation has not been investigated for any gene set in any organism. Here, we present a systematic analysis of protein kinases in the budding yeast, screening for differential localization during filamentous growth. Filamentous growth is an important stress response involving mitogen-activated protein kinase and cAMP-dependent protein kinase signaling modules, wherein yeast cells form interconnected and elongated chains. Because standard strains of yeast are nonfilamentous, we constructed a unique set of 125 kinase-yellow fluorescent protein chimeras in the filamentous Sigma1278b strain for this study. In total, we identified six cytoplasmic kinases (Bcy1p, Fus3p, Ksp1p, Kss1p, Sks1p, and Tpk2p) that localize predominantly to the nucleus during filamentous growth. These kinases form part of an interdependent, localization-based regulatory network: deletion of each individual kinase, or loss of kinase activity, disrupts the nuclear translocation of at least two other kinases. In particular, this study highlights a previously unknown function for the kinase Ksp1p, indicating the essentiality of its nuclear translocation during yeast filamentous growth. Thus, the localization of Ksp1p and the other kinases identified here is tightly controlled during filamentous growth, representing an overlooked regulatory component of this stress response.


Subject(s)
Gene Expression Regulation, Fungal , Protein Serine-Threonine Kinases/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Active Transport, Cell Nucleus , Alleles , Bacterial Proteins/chemistry , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Cytoplasm/enzymology , Fungal Proteins/chemistry , Luminescent Proteins/chemistry , MAP Kinase Signaling System , Models, Genetic , Phenotype , Protein Interaction Mapping , Protein Serine-Threonine Kinases/physiology , Recombinant Fusion Proteins/chemistry , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/physiology , Signal Transduction
16.
Eukaryot Cell ; 7(8): 1289-98, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18310357

ABSTRACT

The genomic architecture of the budding yeast Saccharomyces cerevisiae is typical of other eukaryotes in that genes are spatially organized into discrete and nonoverlapping units. Inherent in this organizational model is the assumption that protein-coding sequences do not overlap completely. Here, we present evidence to the contrary, defining a previously overlooked yeast gene, NAG1 (for nested antisense gene) nested entirely within the coding sequence of the YGR031W open reading frame in an antisense orientation on the opposite strand. NAG1 encodes a 19-kDa protein, detected by Western blotting of hemagglutinin (HA)-tagged Nag1p with anti-HA antibodies and by beta-galactosidase analysis of a NAG1-lacZ fusion. NAG1 is evolutionarily conserved as a unit with YGR031W in bacteria and fungi. Unlike the YGR031WP protein product, however, which localizes to the mitochondria, Nag1p localizes to the cell periphery, exhibiting properties consistent with those of a plasma membrane protein. Phenotypic analysis of a site-directed mutant (nag1-1) disruptive for NAG1 but silent with respect to YGR031W, defines a role for NAG1 in yeast cell wall biogenesis; microarray profiling of nag1-1 indicates decreased expression of genes contributing to cell wall organization, and the nag1-1 mutant is hypersensitive to the cell wall-perturbing agent calcofluor white. Furthermore, production of Nag1p is dependent upon the presence of the cell wall integrity pathway mitogen-activated protein kinase Slt2p and its downstream transcription factor Rlm1p. Thus, NAG1 is important for two reasons. First, it contributes to yeast cell wall biogenesis. Second, its genomic context is novel, raising the possibility that other nested protein-coding genes may exist in eukaryotic genomes.


Subject(s)
Antisense Elements (Genetics)/genetics , Gene Expression Regulation, Fungal/genetics , Membrane Proteins/genetics , Nested Genes/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Cell Wall/genetics , Cell Wall/metabolism , Genome, Fungal/genetics , MADS Domain Proteins , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/isolation & purification , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism
17.
Mol Biol Cell ; 19(1): 284-96, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17989363

ABSTRACT

Under certain conditions of nutrient stress, the budding yeast Saccharomyces cerevisiae initiates a striking developmental transition to a filamentous form of growth, resembling developmental transitions required for virulence in closely related pathogenic fungi. In yeast, filamentous growth involves known mitogen-activated protein kinase and protein kinase A signaling modules, but the full scope of this extensive filamentous response has not been delineated. Accordingly, we have undertaken the first systematic gene disruption and overexpression analysis of yeast filamentous growth. Standard laboratory strains of yeast are nonfilamentous; thus, we constructed a unique set of reagents in the filamentous Sigma1278b strain, encompassing 3627 integrated transposon insertion alleles and 2043 overexpression constructs. Collectively, we analyzed 4528 yeast genes with these reagents and identified 487 genes conferring mutant filamentous phenotypes upon transposon insertion and/or gene overexpression. Using a fluorescent protein reporter integrated at the MUC1 locus, we further assayed each filamentous growth mutant for aberrant protein levels of the key flocculence factor Muc1p. Our results indicate a variety of genes and pathways affecting filamentous growth. In total, this filamentous growth gene set represents a wealth of yeast biology, highlighting 84 genes of uncharacterized function and an underappreciated role for the mitochondrial retrograde signaling pathway as an inhibitor of filamentous growth.


Subject(s)
Genes, Fungal , Mutation/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/genetics , Alleles , DNA Transposable Elements/genetics , Gene Expression Regulation, Fungal , Membrane Glycoproteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/metabolism , Mutagenesis, Insertional , Phenotype , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
18.
Genetics ; 177(1): 205-14, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17890363

ABSTRACT

Over the last 15 years, yeast pseudohyphal growth (PHG) has been the focus of intense research interest as a model of fungal pathogenicity. Specifically, PHG is a stress response wherein yeast cells deprived of nitrogen form filaments of elongated cells. Nitrogen limitation also induces autophagy, a ubiquitous eukaryotic stress response in which proteins are trafficked to the vacuole/lysosome for degradation and recycling. Although autophagy and filamentous growth are both responsive to nitrogen stress, a link between these processes has not been investigated to date. Here, we present several studies describing an interrelationship between autophagy and filamentous growth. By microarray-based expression profiling, we detect extensive upregulation of the pathway governing autophagy during early PHG and find both processes active under conditions of nitrogen stress in a filamentous strain of budding yeast. Inhibition of autophagy results in increased PHG, and autophagy-deficient yeast induce PHG at higher concentrations of available nitrogen. Our results suggest a model in which autophagy mitigates nutrient stress, delaying the onset of PHG; conversely, inhibition of autophagy exacerbates nitrogen stress, resulting in precocious and overactive PHG. This physiological connection highlights the central role of autophagy in regulating the cell's nutritional state and the responsiveness of PHG to that state.


Subject(s)
Autophagy , Fungal Proteins/metabolism , Hyphae/growth & development , Protein Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales/cytology , Saccharomycetales/physiology , Autophagy-Related Protein 8 Family , Autophagy-Related Proteins , Blotting, Western , Cell Survival , Fungal Proteins/genetics , Gene Expression Profiling , Hyphae/cytology , Hyphae/drug effects , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nitrogen , Oligonucleotide Array Sequence Analysis , Phagosomes , Protein Kinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomycetales/drug effects
19.
Autophagy ; 3(6): 604-9, 2007.
Article in English | MEDLINE | ID: mdl-17700056

ABSTRACT

Under conditions of nitrogen stress, the budding yeast S. cerevisiae initiates a cellular response involving the activation of autophagy, an intracellular catabolic process for the degradation and recycling of proteins and organelles. In certain strains of yeast, nitrogen stress also drives a striking developmental transition to a filamentous form of growth, in which cells remain physically connected after cytokinesis. We recently identified an interrelationship between these processes, with the inhibition of autophagy resulting in exaggerated filamentous growth. Our results suggest a model wherein autophagy mitigates nutrient stress, and filamentous growth is responsive to the degree of this stress. Here, we extended these studies to encompass a phenotypic analysis of filamentous growth upon overexpression of autophagy-related (ATG) genes. Specifically, overexpression of ATG1, ATG3, ATG7, ATG17, ATG19, ATG23, ATG24 and ATG29 inhibited filamentous growth. From our understanding of autophagy in yeast, overexpression of these genes does not markedly affect the activity of the pathway; thus, we do not expect that this filamentous growth phenotype is due strictly to diminished nitrogen stress in ATG overexpression mutants. Rather, these results highlight an additional undefined regulatory mechanism linking autophagy and filamentous growth, possibly independent of the upstream nitrogen-sensing machinery feeding into both processes.


Subject(s)
Autophagy/genetics , Autophagy/physiology , Genes, Fungal , Hyphae/growth & development , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Hyphae/cytology , Models, Biological , Mutation , Nitrogen , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae Proteins/genetics
20.
Genome Res ; 14(10A): 1975-86, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15466296

ABSTRACT

We present here an unbiased and extremely versatile insertional library of yeast genomic DNA generated by in vitro mutagenesis with a multipurpose element derived from the bacterial transposon Tn7. This mini-Tn7 element has been engineered such that a single insertion can be used to generate a lacZ fusion, gene disruption, and epitope-tagged gene product. Using this transposon, we generated a plasmid-based library of approximately 300,000 mutant alleles; by high-throughput screening in yeast, we identified and sequenced 9032 insertions affecting 2613 genes (45% of the genome). From analysis of 7176 insertions, we found little bias in Tn7 target-site selection in vitro. In contrast, we also sequenced 10,174 Tn3 insertions and found a markedly stronger preference for an AT-rich 5-base pair target sequence. We further screened 1327 insertion alleles in yeast for hypersensitivity to the chemotherapeutic cisplatin. Fifty-one genes were identified, including four functionally uncharacterized genes and 25 genes involved in DNA repair, replication, transcription, and chromatin structure. In total, the collection reported here constitutes the largest plasmid-based set of sequenced yeast mutant alleles to date and, as such, should be singularly useful for gene and genome-wide functional analysis.


Subject(s)
DNA Transposable Elements , Genome, Fungal , Mutagenesis, Insertional , Amino Acid Sequence , Base Sequence , Cisplatin/pharmacology , DNA Primers , Molecular Sequence Data , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...