Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28.727
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 170(4): 693-700.e7, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28802041

RESUMEN

The TOM complex is the main entry gate for protein precursors from the cytosol into mitochondria. We have determined the structure of the TOM core complex by cryoelectron microscopy (cryo-EM). The complex is a 148 kDa symmetrical dimer of ten membrane protein subunits that create a shallow funnel on the cytoplasmic membrane surface. In the core of the dimer, the ß-barrels of the Tom40 pore form two identical preprotein conduits. Each Tom40 pore is surrounded by the transmembrane segments of the α-helical subunits Tom5, Tom6, and Tom7. Tom22, the central preprotein receptor, connects the two Tom40 pores at the dimer interface. Our structure offers detailed insights into the molecular architecture of the mitochondrial preprotein import machinery.


Asunto(s)
Proteínas Portadoras/química , Proteínas Fúngicas/química , Neurospora crassa/enzimología , Sistemas de Translocación de Proteínas/química , Secuencia de Aminoácidos , Proteínas Portadoras/genética , Proteínas Portadoras/ultraestructura , Microscopía por Crioelectrón , Proteínas Fúngicas/genética , Proteínas Fúngicas/ultraestructura , Espectrometría de Masas , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/ultraestructura , Membranas Mitocondriales/enzimología , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales , Modelos Moleculares , Conformación Proteica en Lámina beta , Sistemas de Translocación de Proteínas/genética , Sistemas de Translocación de Proteínas/ultraestructura , Proteínas de Saccharomyces cerevisiae/química
2.
Mol Cell ; 79(1): 127-139.e4, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32437639

RESUMEN

C.neoformans Dnmt5 is an unusually specific maintenance-type CpG methyltransferase (DNMT) that mediates long-term epigenome evolution. It harbors a DNMT domain and SNF2 ATPase domain. We find that the SNF2 domain couples substrate specificity to an ATPase step essential for DNA methylation. Coupling occurs independent of nucleosomes. Hemimethylated DNA preferentially stimulates ATPase activity, and mutating Dnmt5's ATP-binding pocket disproportionately reduces ATPase stimulation by hemimethylated versus unmethylated substrates. Engineered DNA substrates that stabilize a reaction intermediate by mimicking a "flipped-out" conformation of the target cytosine bypass the SNF2 domain's requirement for hemimethylation. This result implies that ATP hydrolysis by the SNF2 domain is coupled to the DNMT domain conformational changes induced by preferred substrates. These findings establish a new role for a SNF2 ATPase: controlling an adjoined enzymatic domain's substrate recognition and catalysis. We speculate that this coupling contributes to the exquisite specificity of Dnmt5 via mechanisms related to kinetic proofreading.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN de Hongos/metabolismo , Proteínas Fúngicas/metabolismo , Nucleosomas/metabolismo , Adenosina Trifosfatasas/genética , Cryptococcus neoformans/genética , Cryptococcus neoformans/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN de Hongos/química , ADN de Hongos/genética , Proteínas Fúngicas/genética , Hidrólisis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Annu Rev Genet ; 53: 149-170, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31451036

RESUMEN

Fungi see light of different colors by using photoreceptors such as the White Collar proteins and cryptochromes for blue light, opsins for green light, and phytochromes for red light. Light regulates fungal development, promotes the accumulation of protective pigments and proteins, and regulates tropic growth. The White Collar complex (WCC) is a photoreceptor and a transcription factor that is responsible for regulating transcription after exposure to blue light. In Neurospora crassa, light promotes the interaction of WCCs and their binding to the promoters to activate transcription. In Aspergillus nidulans, the WCC and the phytochrome interact to coordinate gene transcription and other responses, but the contribution of these photoreceptors to fungal photobiology varies across fungal species. Ultimately, the effect of light on fungal biology is the result of the coordinated transcriptional regulation and activation of signal transduction pathways.


Asunto(s)
Proteínas Fúngicas/genética , Hongos/fisiología , Regulación Fúngica de la Expresión Génica , Fotorreceptores Microbianos/genética , Aspergillus nidulans/fisiología , Luz , Neurospora crassa/genética , Neurospora crassa/fisiología , Fotorreceptores Microbianos/metabolismo , Transducción de Señal , Transcripción Genética
4.
Annu Rev Genet ; 53: 417-444, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31537103

RESUMEN

Cryptococcus species utilize a variety of sexual reproduction mechanisms, which generate genetic diversity, purge deleterious mutations, and contribute to their ability to occupy myriad environmental niches and exhibit a range of pathogenic potential. The bisexual and unisexual cycles of pathogenic Cryptococcus species are stimulated by properties associated with their environmental niches and proceed through well-characterized signaling pathways and corresponding morphological changes. Genes governing mating are encoded by the mating-type (MAT) loci and influence pathogenesis, population dynamics, and lineage divergence in Cryptococcus. MAT has undergone significant evolutionary changes within the Cryptococcus genus, including transition from the ancestral tetrapolar state in nonpathogenic species to a bipolar mating system in pathogenic species, as well as several internal reconfigurations. Owing to the variety of established sexual reproduction mechanisms and the robust characterization of the evolution of mating and MAT in this genus, Cryptococcus species provide key insights into the evolution of sexual reproduction.


Asunto(s)
Cryptococcus/fisiología , Cryptococcus/patogenicidad , Genes del Tipo Sexual de los Hongos , Reproducción/fisiología , Evolución Biológica , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genética de Población , Interacciones Huésped-Patógeno , Humanos , Esporas Fúngicas/patogenicidad , Esporas Fúngicas/fisiología
5.
Plant Cell ; 36(9): 3260-3276, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38923940

RESUMEN

Secreted immune proteases "Required for Cladosporium resistance-3" (Rcr3) and "Phytophthora-inhibited protease-1" (Pip1) of tomato (Solanum lycopersicum) are both inhibited by Avirulence-2 (Avr2) from the fungal plant pathogen Cladosporium fulvum. However, only Rcr3 acts as a decoy co-receptor that detects Avr2 in the presence of the Cf-2 immune receptor. Here, we identified crucial residues in tomato Rcr3 that are required for Cf-2-mediated signaling and bioengineered various proteases to trigger Avr2/Cf-2-dependent immunity. Despite substantial divergence in Rcr3 orthologs from eggplant (Solanum melongena) and tobacco (Nicotiana spp.), minimal alterations were sufficient to trigger Avr2/Cf-2-mediated immune signaling. By contrast, tomato Pip1 was bioengineered with 16 Rcr3-specific residues to initiate Avr2/Cf-2-triggered immune signaling. These residues cluster on one side of the protein next to the substrate-binding groove, indicating a potential Cf-2 interaction site. Our findings also revealed that Rcr3 and Pip1 have distinct substrate preferences determined by two variant residues and that both are suboptimal for binding Avr2. This study advances our understanding of Avr2 perception and opens avenues to bioengineer proteases to broaden pathogen recognition in other crops.


Asunto(s)
Cladosporium , Péptido Hidrolasas , Proteínas de Plantas , Solanum lycopersicum , Solanum lycopersicum/genética , Solanum lycopersicum/microbiología , Solanum lycopersicum/inmunología , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Cladosporium/patogenicidad , Péptido Hidrolasas/metabolismo , Péptido Hidrolasas/genética , Inmunidad de la Planta/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Bioingeniería , Enfermedades de las Plantas/microbiología , Enfermedades de las Plantas/inmunología , Enfermedades de las Plantas/genética , Transducción de Señal , Nicotiana/genética , Nicotiana/microbiología , Nicotiana/metabolismo , Nicotiana/inmunología
6.
Plant Cell ; 36(5): 2000-2020, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38299379

RESUMEN

The flower-infecting fungus Ustilaginoidea virens causes rice false smut, which is a severe emerging disease threatening rice (Oryza sativa) production worldwide. False smut not only reduces yield, but more importantly produces toxins on grains, posing a great threat to food safety. U. virens invades spikelets via the gap between the 2 bracts (lemma and palea) enclosing the floret and specifically infects the stamen and pistil. Molecular mechanisms for the U. virens-rice interaction are largely unknown. Here, we demonstrate that rice flowers predominantly employ chitin-triggered immunity against U. virens in the lemma and palea, rather than in the stamen and pistil. We identify a crucial U. virens virulence factor, named UvGH18.1, which carries glycoside hydrolase activity. Mechanistically, UvGH18.1 functions by binding to and hydrolyzing immune elicitor chitin and interacting with the chitin receptor CHITIN ELICITOR BINDING PROTEIN (OsCEBiP) and co-receptor CHITIN ELICITOR RECEPTOR KINASE1 (OsCERK1) to impair their chitin-induced dimerization, suppressing host immunity exerted at the lemma and palea for gaining access to the stamen and pistil. Conversely, pretreatment on spikelets with chitin induces a defense response in the lemma and palea, promoting resistance against U. virens. Collectively, our data uncover a mechanism for a U. virens virulence factor and the critical location of the host-pathogen interaction in flowers and provide a potential strategy to control rice false smut disease.


Asunto(s)
Quitina , Flores , Hypocreales , Oryza , Enfermedades de las Plantas , Oryza/microbiología , Oryza/metabolismo , Oryza/genética , Enfermedades de las Plantas/microbiología , Quitina/metabolismo , Flores/microbiología , Hypocreales/patogenicidad , Hypocreales/genética , Hypocreales/metabolismo , Transducción de Señal , Interacciones Huésped-Patógeno , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Virulencia , Factores de Virulencia/metabolismo , Factores de Virulencia/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética
7.
PLoS Biol ; 22(7): e3002705, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38950075

RESUMEN

We show here that in the fungus Sordaria macrospora, the meiosis-specific HORMA-domain protein Hop1 is not essential for the basic early events of chromosome axis development, recombination initiation, or recombination-mediated homolog coalignment/pairing. In striking contrast, Hop1 plays a critical role at the leptotene/zygotene transition which is defined by transition from pairing to synaptonemal complex (SC) formation. During this transition, Hop1 is required for maintenance of normal axis structure, formation of SC from telomere to telomere, and development of recombination foci. These hop1Δ mutant defects are DSB dependent and require Sme4/Zip1-mediated progression of the interhomolog interaction program, potentially via a pre-SC role. The same phenotype occurs not only in hop1Δ but also in absence of the cohesin Rec8 and in spo76-1, a non-null mutant of cohesin-associated Spo76/Pds5. Thus, Hop1 and cohesins collaborate at this crucial step of meiotic prophase. In addition, analysis of 4 non-null mutants that lack this transition defect reveals that Hop1 also plays important roles in modulation of axis length, homolog-axis juxtaposition, interlock resolution, and spreading of the crossover interference signal. Finally, unexpected variations in crossover density point to the existence of effects that both enhance and limit crossover formation. Links to previously described roles of the protein in other organisms are discussed.


Asunto(s)
Proteínas Fúngicas , Sordariales , Complejo Sinaptonémico , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Sordariales/genética , Sordariales/metabolismo , Complejo Sinaptonémico/metabolismo , Meiosis , Profase Meiótica I , Profase , Mutación
8.
PLoS Biol ; 22(7): e3002724, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39052688

RESUMEN

Alternative transcription start site (TSS) usage regulation has been identified as a major means of gene expression regulation in metazoans. However, in fungi, its impact remains elusive as its study has thus far been restricted to model yeasts. Here, we first re-analyzed TSS-seq data to define genuine TSS clusters in 2 species of pathogenic Cryptococcus. We identified 2 types of TSS clusters associated with specific DNA sequence motifs. Our analysis also revealed that alternative TSS usage regulation in response to environmental cues is widespread in Cryptococcus, altering gene expression and protein targeting. Importantly, we performed a forward genetic screen to identify a unique transcription factor (TF) named Tur1, which regulates alternative TSS (altTSS) usage genome-wide when cells switch from exponential phase to stationary phase. ChiP-Seq and DamID-Seq analyses suggest that at some loci, the role of Tur1 might be direct. Tur1 has been previously shown to be essential for virulence in C. neoformans. We demonstrated here that a tur1Δ mutant strain is more sensitive to superoxide stress and phagocytosed more efficiently by macrophages than the wild-type (WT) strain.


Asunto(s)
Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Genoma Fúngico , Factores de Transcripción , Sitio de Iniciación de la Transcripción , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Cryptococcus/genética , Cryptococcus/patogenicidad , Cryptococcus/metabolismo , Cryptococcus neoformans/genética , Cryptococcus neoformans/patogenicidad , Cryptococcus neoformans/metabolismo , Macrófagos/microbiología , Macrófagos/metabolismo , Animales , Ratones , Virulencia/genética , Fagocitosis/genética
9.
PLoS Biol ; 22(7): e3002726, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39078817

RESUMEN

The importance of fungi in ecological systems and pathogenicity hinges on their ability to search for nutrients, substrates, and hosts. Despite this, the question of whether fungal hyphae exhibit chemotropism toward them remains largely unresolved and requires close examination at the cellular level. Here, we designed a microfluidic device to assess hyphal chemotropism of Aspergillus nidulans in response to carbon and nitrogen sources, as well as pH. Within this device, hyphae could determine their growth direction in a two-layer flow with distinct compositions that were adjacent but non-mixing. Under conditions with and without a carbon source, hyphae changed growth direction to remain in the presence of a carbon source, but it was still difficult to distinguish between differences in growth and chemotropism. Although nitrogen sources such as ammonia and nitrate are important for growth, the hyphae indicated negative chemotropism to avoid them depending on the specific transporters. This fungus grows equally well at the colony level in the pH range of 4 to 9, but the hyphae exhibited chemotropism to acidic pH. The proton pump PmaA is vital for the chemotropism to acid pH, while the master regulatory for pH adaptation PacC is not involved, suggesting that chemotropism and adaptive growth via gene expression regulation are distinct regulatory mechanisms. Despite various plasma membrane transporters are distributed across membranes except at the hyphal tip, the control of growth direction occurs at the tip. Finally, we explored the mechanisms linking these two phenomena, tip growth and chemotropism.


Asunto(s)
Aspergillus nidulans , Hifa , Aspergillus nidulans/metabolismo , Aspergillus nidulans/fisiología , Hifa/metabolismo , Hifa/crecimiento & desarrollo , Concentración de Iones de Hidrógeno , Nitrógeno/metabolismo , Nutrientes/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Carbono/metabolismo , Regulación Fúngica de la Expresión Génica , Amoníaco/metabolismo
10.
PLoS Biol ; 22(6): e3002693, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38905306

RESUMEN

Candida albicans is a commensal of the human microbiota that can form biofilms on implanted medical devices. These biofilms are tolerant to antifungals and to the host immune system. To identify novel genes modulating C. albicans biofilm formation, we performed a large-scale screen with 2,454 C. albicans doxycycline-dependent overexpression strains and identified 16 genes whose overexpression significantly hampered biofilm formation. Among those, overexpression of the ZCF15 and ZCF26 paralogs that encode transcription factors and have orthologs only in biofilm-forming species of the Candida clade, caused impaired biofilm formation both in vitro and in vivo. Interestingly, overexpression of ZCF15 impeded biofilm formation without any defect in hyphal growth. Transcript profiling, transcription factor binding, and phenotypic microarray analyses conducted upon overexpression of ZCF15 and ZCF26 demonstrated their role in reprogramming cellular metabolism by regulating central metabolism including glyoxylate and tricarboxylic acid cycle genes. Taken together, this study has identified a new set of biofilm regulators, including ZCF15 and ZCF26, that appear to control biofilm development through their specific role in metabolic remodeling.


Asunto(s)
Biopelículas , Candida albicans , Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Factores de Transcripción , Biopelículas/crecimiento & desarrollo , Candida albicans/genética , Candida albicans/metabolismo , Candida albicans/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Animales , Plancton/metabolismo , Glioxilatos/metabolismo , Perfilación de la Expresión Génica/métodos , Ratones , Ciclo del Ácido Cítrico , Hifa/metabolismo , Hifa/crecimiento & desarrollo , Hifa/genética , Candidiasis/microbiología , Reprogramación Metabólica
11.
Cell ; 151(1): 80-95, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23021217

RESUMEN

We examine how different transcriptional network structures can evolve from an ancestral network. By characterizing how the ancestral mode of gene regulation for genes specific to a-type cells in yeast species evolved from an activating paradigm to a repressing one, we show that regulatory protein modularity, conversion of one cis-regulatory sequence to another, distribution of binding energy among protein-protein and protein-DNA interactions, and exploitation of ancestral network features all contribute to the evolution of a novel regulatory mode. The formation of this derived mode of regulation did not disrupt the ancestral mode and thereby created a hybrid regulatory state where both means of transcription regulation (ancestral and derived) contribute to the conserved expression pattern of the network. Finally, we show how this hybrid regulatory state has resolved in different ways in different lineages to generate the diversity of regulatory network structures observed in modern species.


Asunto(s)
Evolución Molecular , Proteínas Fúngicas/genética , Redes Reguladoras de Genes , Proteínas de la Membrana/genética , Saccharomycetales/genética , Factores de Transcripción/genética , Filogenia , Saccharomycetales/metabolismo
12.
Nature ; 592(7853): 309-314, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33692541

RESUMEN

The genome-wide architecture of chromatin-associated proteins that maintains chromosome integrity and gene regulation is not well defined. Here we use chromatin immunoprecipitation, exonuclease digestion and DNA sequencing (ChIP-exo/seq)1,2 to define this architecture in Saccharomyces cerevisiae. We identify 21 meta-assemblages consisting of roughly 400 different proteins that are related to DNA replication, centromeres, subtelomeres, transposons and transcription by RNA polymerase (Pol) I, II and III. Replication proteins engulf a nucleosome, centromeres lack a nucleosome, and repressive proteins encompass three nucleosomes at subtelomeric X-elements. We find that most promoters associated with Pol II evolved to lack a regulatory region, having only a core promoter. These constitutive promoters comprise a short nucleosome-free region (NFR) adjacent to a +1 nucleosome, which together bind the transcription-initiation factor TFIID to form a preinitiation complex. Positioned insulators protect core promoters from upstream events. A small fraction of promoters evolved an architecture for inducibility, whereby sequence-specific transcription factors (ssTFs) create a nucleosome-depleted region (NDR) that is distinct from an NFR. We describe structural interactions among ssTFs, their cognate cofactors and the genome. These interactions include the nucleosomal and transcriptional regulators RPD3-L, SAGA, NuA4, Tup1, Mediator and SWI-SNF. Surprisingly, we do not detect interactions between ssTFs and TFIID, suggesting that such interactions do not stably occur. Our model for gene induction involves ssTFs, cofactors and general factors such as TBP and TFIIB, but not TFIID. By contrast, constitutive transcription involves TFIID but not ssTFs engaged with their cofactors. From this, we define a highly integrated network of gene regulation by ssTFs.


Asunto(s)
Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Genoma Fúngico/genética , Complejos Multiproteicos/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Factores de Transcripción/genética , Coenzimas/metabolismo , Complejos Multiproteicos/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa I/metabolismo , ARN Polimerasa II/metabolismo , ARN Polimerasa III/metabolismo , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factor de Transcripción TFIIB/genética , Factor de Transcripción TFIIB/metabolismo , Factor de Transcripción TFIID , Factores de Transcripción/metabolismo
13.
Mol Cell ; 75(6): 1256-1269.e7, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31378463

RESUMEN

Eukaryotic ribosome biogenesis involves RNA folding and processing that depend on assembly factors and small nucleolar RNAs (snoRNAs). The 90S (SSU-processome) is the earliest pre-ribosome structurally analyzed, which was suggested to assemble stepwise along the growing pre-rRNA from 5' > 3', but this directionality may not be accurate. Here, by analyzing the structure of a series of 90S assembly intermediates from Chaetomium thermophilum, we discover a reverse order of 18S rRNA subdomain incorporation. Large parts of the 18S rRNA 3' and central domains assemble first into the 90S before the 5' domain is integrated. This final incorporation depends on a contact between a heterotrimer Enp2-Bfr2-Lcp5 recruited to the flexible 5' domain and Kre33, which reconstitutes the Kre33-Enp-Brf2-Lcp5 module on the compacted 90S. Keeping the 5' domain temporarily segregated from the 90S scaffold could provide extra time to complete the multifaceted 5' domain folding, which depends on a distinct set of snoRNAs and processing factors.


Asunto(s)
Chaetomium/metabolismo , Proteínas Fúngicas/metabolismo , Conformación de Ácido Nucleico , ARN de Hongos/metabolismo , ARN Ribosómico 18S/metabolismo , Ribosomas/metabolismo , Chaetomium/genética , Proteínas Fúngicas/genética , ARN de Hongos/genética , ARN Ribosómico 18S/genética , Ribosomas/genética
14.
Mol Cell ; 74(4): 771-784.e3, 2019 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-30954403

RESUMEN

In the negative feedback loop driving fungal and animal circadian oscillators, negative elements (FREQUENCY [FRQ], PERIODS [PERs], and CRYPTOCHROMES [CRYs]) are understood to inhibit their own expression, in part by promoting the phosphorylation of their heterodimeric transcriptional activators (e.g., White Collar-1 [WC-1]-WC-2 [White Collar complex; WCC] and BMAL1/Circadian Locomotor Output Cycles Kaput [CLOCK]). However, correlations between heterodimer activity and phosphorylation are weak, contradictions exist, and mechanistic details are almost wholly lacking. We report mapping of 80 phosphosites on WC-1 and 15 on WC-2 and elucidation of the time-of-day-specific code, requiring both a group of phosphoevents on WC-1 and two distinct clusters on WC-2, that governs circadian repression, leading to feedback loop closure. Combinatorial control via phosphorylation also governs rhythmic WCC binding to the promoters of clock-controlled genes mediating the essential first step in circadian output, a group encoding both transcription factors and signaling proteins. These data provide a basic mechanistic understanding for fundamental events underlying circadian negative feedback and output, key aspects of circadian biology.


Asunto(s)
Ritmo Circadiano/genética , Proteínas de Unión al ADN/genética , Proteínas Fúngicas/genética , Neurospora crassa/genética , Factores de Transcripción/genética , Factores de Transcripción ARNTL/genética , Retroalimentación Fisiológica , Regulación Fúngica de la Expresión Génica , Neurospora crassa/fisiología , Fosforilación , Regiones Promotoras Genéticas/genética , Transducción de Señal/genética
15.
Mol Cell ; 76(2): 268-285, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31585693

RESUMEN

The clearance of surplus, broken, or dangerous components is key for maintaining cellular homeostasis. The failure to remove protein aggregates, damaged organelles, or intracellular pathogens leads to diseases, including neurodegeneration, cancer, and infectious diseases. Autophagy is the evolutionarily conserved pathway that sequesters cytoplasmic components in specialized vesicles, autophagosomes, which transport the cargo to the degradative compartments (vacuoles or lysosomes). Research during the past few decades has elucidated how autophagosomes engulf their substrates selectively. This type of autophagy involves a growing number of selective autophagy receptors (SARs) (e.g., Atg19 in yeasts, p62/SQSTM1 in mammals), which bind to the cargo and simultaneously engage components of the core autophagic machinery via direct interaction with the ubiquitin-like proteins (UBLs) of the Atg8/LC3/GABARAP family and adaptors, Atg11 (in yeasts) or FIP200 (in mammals). In this Review, we critically discuss the biology of the SARs with special emphasis on their interactions with UBLs.


Asunto(s)
Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Proteínas Fúngicas/metabolismo , Transducción de Señal , Levaduras/metabolismo , Animales , Autofagosomas/patología , Proteínas Relacionadas con la Autofagia/genética , Sitios de Unión , Proteínas Fúngicas/genética , Humanos , Ligandos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Ubiquitinación , Ubiquitinas/metabolismo , Levaduras/genética
16.
Proc Natl Acad Sci U S A ; 121(13): e2319998121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38513096

RESUMEN

Lytic polysaccharide monooxygenases (LPMOs) are monocopper enzymes that oxidatively degrade various polysaccharides, such as cellulose. Despite extensive research on this class of enzymes, the role played by their C-terminal regions predicted to be intrinsically disordered (dCTR) has been overlooked. Here, we investigated the function of the dCTR of an LPMO, called CoAA9A, up-regulated during plant infection by Colletotrichum orbiculare, the causative agent of anthracnose. After recombinant production of the full-length protein, we found that the dCTR mediates CoAA9A dimerization in vitro, via a disulfide bridge, a hitherto-never-reported property that positively affects both binding and activity on cellulose. Using SAXS experiments, we show that the homodimer is in an extended conformation. In vivo, we demonstrate that gene deletion impairs formation of the infection-specialized cell called appressorium and delays penetration of the plant. Using immunochemistry, we show that the protein is a dimer not only in vitro but also in vivo when secreted by the appressorium. As these peculiar LPMOs are also found in other plant pathogens, our findings open up broad avenues for crop protection.


Asunto(s)
Proteínas Fúngicas , Polisacáridos , Multimerización de Proteína , Dispersión del Ángulo Pequeño , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Difracción de Rayos X , Polisacáridos/metabolismo , Celulosa/metabolismo
17.
PLoS Genet ; 20(2): e1011114, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38346076

RESUMEN

Filamentous fungi display allorecognition genes that trigger regulated cell death (RCD) when strains of unlike genotype fuse. Podospora anserina is one of several model species for the study of this allorecognition process termed heterokaryon or vegetative incompatibility. Incompatibility restricts transmission of mycoviruses between isolates. In P. anserina, genetic analyses have identified nine incompatibility loci, termed het loci. Here we set out to clone the genes controlling het-B incompatibility. het-B displays two incompatible alleles, het-B1 and het-B2. We find that the het-B locus encompasses two adjacent genes, Bh and Bp that exist as highly divergent allelic variants (Bh1/Bh2 and Bp1/Bp2) in the incompatible haplotypes. Bh encodes a protein with an N-terminal HET domain, a cell death inducing domain bearing homology to Toll/interleukin-1 receptor (TIR) domains and a C-terminal domain with a predicted lectin fold. The Bp product is homologous to PII-like proteins, a family of small trimeric proteins acting as sensors of adenine nucleotides in bacteria. We show that although the het-B system appears genetically allelic, incompatibility is in fact determined by the non-allelic Bh1/Bp2 interaction while the reciprocal Bh2/Bp1 interaction plays no role in incompatibility. The highly divergent C-terminal lectin fold domain of BH determines recognition specificity. Population studies and genome analyses indicate that het-B is under balancing selection with trans-species polymorphism, highlighting the evolutionary significance of the two incompatible haplotypes. In addition to emphasizing anew the central role of TIR-like HET domains in fungal RCD, this study identifies novel players in fungal allorecognition and completes the characterization of the entire het gene set in that species.


Asunto(s)
Podospora , Podospora/genética , Alelos , Lectinas/genética , Lectinas/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Polimorfismo Genético
18.
PLoS Genet ; 20(2): e1011158, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38359090

RESUMEN

Elucidating gene function is a major goal in biology, especially among non-model organisms. However, doing so is complicated by the fact that molecular conservation does not always mirror functional conservation, and that complex relationships among genes are responsible for encoding pathways and higher-order biological processes. Co-expression, a promising approach for predicting gene function, relies on the general principal that genes with similar expression patterns across multiple conditions will likely be involved in the same biological process. For Cryptococcus neoformans, a prevalent human fungal pathogen greatly diverged from model yeasts, approximately 60% of the predicted genes in the genome lack functional annotations. Here, we leveraged a large amount of publicly available transcriptomic data to generate a C. neoformans Co-Expression Network (CryptoCEN), successfully recapitulating known protein networks, predicting gene function, and enabling insights into the principles influencing co-expression. With 100% predictive accuracy, we used CryptoCEN to identify 13 new DNA damage response genes, underscoring the utility of guilt-by-association for determining gene function. Overall, co-expression is a powerful tool for uncovering gene function, and decreases the experimental tests needed to identify functions for currently under-annotated genes.


Asunto(s)
Criptococosis , Cryptococcus neoformans , Humanos , Cryptococcus neoformans/genética , Criptococosis/genética , Criptococosis/microbiología , Reparación del ADN/genética , Fenotipo , Daño del ADN/genética , Proteínas Fúngicas/genética
19.
PLoS Genet ; 20(8): e1011156, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39137212

RESUMEN

Phosphorus is essential in all cells' structural, metabolic and regulatory functions. For fungal cells that import inorganic phosphate (Pi) up a steep concentration gradient, surface Pi transporters are critical capacitators of growth. Fungi must deploy Pi transporters that enable optimal Pi uptake in pH and Pi concentration ranges prevalent in their environments. Single, triple and quadruple mutants were used to characterize the four Pi transporters we identified for the human fungal pathogen Candida albicans, which must adapt to alkaline conditions during invasion of the host bloodstream and deep organs. A high-affinity Pi transporter, Pho84, was most efficient across the widest pH range while another, Pho89, showed high-affinity characteristics only within one pH unit of neutral. Two low-affinity Pi transporters, Pho87 and Fgr2, were active only in acidic conditions. Only Pho84 among the Pi transporters was clearly required in previously identified Pi-related functions including Target of Rapamycin Complex 1 signaling, oxidative stress resistance and hyphal growth. We used in vitro evolution and whole genome sequencing as an unbiased forward genetic approach to probe adaptation to prolonged Pi scarcity of two quadruple mutant lineages lacking all 4 Pi transporters. Lineage-specific genomic changes corresponded to divergent success of the two lineages in fitness recovery during Pi limitation. Initial, large-scale genomic alterations like aneuploidies and loss of heterozygosity eventually resolved, as populations gained small-scale mutations. Severity of some phenotypes linked to Pi starvation, like cell wall stress hypersensitivity, decreased in parallel to evolving populations' fitness recovery in Pi scarcity, while severity of others like membrane stress responses diverged from Pi scarcity fitness. Among preliminary candidate genes for contributors to fitness recovery, those with links to TORC1 were overrepresented. Since Pi homeostasis differs substantially between fungi and humans, adaptive processes to Pi deprivation may harbor small-molecule targets that impact fungal growth, stress resistance and virulence.


Asunto(s)
Adaptación Fisiológica , Candida albicans , Proteínas Fúngicas , Fosfatos , Fosfatos/metabolismo , Candida albicans/genética , Candida albicans/metabolismo , Adaptación Fisiológica/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Concentración de Iones de Hidrógeno , Proteínas de Transporte de Fosfato/genética , Proteínas de Transporte de Fosfato/metabolismo , Mutación , Regulación Fúngica de la Expresión Génica , Humanos , Transporte Biológico/genética
20.
Proc Natl Acad Sci U S A ; 121(32): e2404770121, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39074265

RESUMEN

Repression of facultative heterochromatin is essential for developmental processes in numerous organisms. Methylation of histone H3 lysine 27 (H3K27) by Polycomb repressive complex 2 is a prominent feature of facultative heterochromatin in both fungi and higher eukaryotes. Although this methylation is frequently associated with silencing, the detailed mechanism of repression remains incompletely understood. We utilized a forward genetics approach to identify genes required to maintain silencing at facultative heterochromatin genes in Neurospora crassa and identified three previously uncharacterized genes that are important for silencing: sds3 (NCU01599), rlp1 (RPD3L protein 1; NCU09007), and rlp2 (RPD3L protein 2; NCU02898). We found that SDS3, RLP1, and RLP2 associate with N. crassa homologs of the Saccharomyces cerevisiae Rpd3L complex and are required for repression of a subset of H3K27-methylated genes. Deletion of these genes does not lead to loss of H3K27 methylation but increases acetylation of histone H3 lysine 14 at up-regulated genes, suggesting that RPD3L-driven deacetylation is a factor required for silencing of facultative heterochromatin in N. crassa, and perhaps in other organisms.


Asunto(s)
Proteínas Fúngicas , Regulación Fúngica de la Expresión Génica , Heterocromatina , Histonas , Neurospora crassa , Neurospora crassa/genética , Neurospora crassa/metabolismo , Heterocromatina/metabolismo , Heterocromatina/genética , Histonas/metabolismo , Histonas/genética , Proteínas Fúngicas/metabolismo , Proteínas Fúngicas/genética , Acetilación , Silenciador del Gen , Metilación , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA