Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Fungal Genet Biol ; 163: 103739, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36089227

RESUMO

Fungal biofilm founder cells experience self-generated hypoxia leading to dramatic changes in their cell biology. For example, during Aspergillus nidulans biofilm formation microtubule (MT) disassembly is triggered causing dispersal of EB1 from MT tips. This process is dependent on SrbA, a sterol regulatory element-binding transcription factor required for adaptation to hypoxia. We show that SrbA, an ER resident protein prior to activation, is proteolytically activated during early stages of biofilm formation and that, like SrbA itself, its activating proteases are also required for normal biofilm MT disassembly. In addition to SrbA, the AtrR transcription factor is also found to be required to modulate cellular responses to gaseous signaling during biofilm development. Using co-cultures, we further show that cells lacking srbA or atrR are capable of responding to biofilm generated gaseous microenvironments but are actually more sensitive to this signal than wild type cells. SrbA is a regulator of ergosterol biosynthetic genes and we find that the levels of seven GFP-tagged Erg proteins differentially accumulate during biofilm formation with various dependencies on SrbA for their accumulation. This uncovers a complex pattern of regulation with biofilm accumulation of only some Erg proteins being dependent on SrbA with others accumulating to higher levels in its absence. Because different membrane sterols are known to influence cell permeability to gaseous molecules, including oxygen, we propose that differential regulation of ergosterol biosynthetic proteins by SrbA potentially calibrates the cell's responsiveness to gaseous signaling which in turn modifies the cell biology of developing biofilm cells.


Assuntos
Aspergillus nidulans , Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Aspergillus fumigatus/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Esteróis/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Gases/metabolismo , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Hipóxia , Biofilmes , Ergosterol/metabolismo
2.
Methods Mol Biol ; 2502: 183-201, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35412239

RESUMO

Targeting a protein of interest to a subcellular location by linking it to another protein is a commonly used approach to help determine function in many model systems. Such targeting strategies rely on the creation of functional protein-protein fusions followed by microscopic examination if one or both proteins have fluorescent tags. In this paper, using the model filamentous fungus Aspergillus nidulans, we describe methods to link GFP-tagged proteins to other proteins in the cell by fusing the latter with a GFP-Binding Protein (GBP) that has a high affinity for GFP. This method enables rapid generation of strains with linked proteins in filamentous fungi by sexual crossing or transformations. Additionally, if these two linked proteins stably associate with subcellular structures, it is possible to link the structures using this approach. For example, we used this method to link Nuclear Pore Complexes (NPCs) with mitotic chromatin in A. nidulans. This was done to show that the NPC protein Nup2, that uniquely transitions from NPC onto mitotic chromatin, couples NPC segregation with chromatin segregation by bridging these two structures. In the absence of Nup2, we used the described approach to show that an artificial NPC-chromatin bridge was sufficient for faithful NPC segregation.


Assuntos
Aspergillus nidulans , Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Cromatina/metabolismo , Mitose , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo
3.
Mol Biol Cell ; 32(12): 1181-1192, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33826367

RESUMO

After growing on surfaces, including those of medical and industrial importance, fungal biofilms self-generate internal microenvironments. We previously reported that gaseous microenvironments around founder Aspergillus nidulans cells change during biofilm formation causing microtubules to disassemble under control of the hypoxic transcription factor SrbA. Here we investigate if biofilm formation might also promote changes to structures involved in exocytosis and endocytosis. During biofilm formation, the endoplasmic reticulum (ER) remained intact but ER exit sites and the Golgi apparatus were modified as were endocytic actin patches. The biofilm-driven changes required the SrbA hypoxic transcription factor and could be triggered by nitric oxide, further implicating gaseous regulation of biofilm cellular architecture. By tracking green fluorescent protein (GFP)-Atg8 dynamics, biofilm founder cells were also observed to undergo autophagy. Most notably, biofilm cells that had undergone autophagy were triggered into further autophagy by spinning disk confocal light. Our findings indicate that fungal biofilm formation modifies the secretory and endocytic apparatus and show that biofilm cells can also undergo autophagy that is reactivated by light. The findings provide new insights into the changes occurring in fungal biofilm cell biology that potentially impact their unique characteristics, including antifungal drug resistance.


Assuntos
Aspergillus nidulans/ultraestrutura , Autofagia , Biofilmes , Retículo Endoplasmático/fisiologia , Luz , Aspergillus nidulans/fisiologia , Endocitose , Retículo Endoplasmático/metabolismo , Exocitose , Proteínas Fúngicas/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/fisiologia , Microtúbulos/metabolismo , Fatores de Transcrição/metabolismo
4.
Fungal Genet Biol ; 130: 72-81, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31026588

RESUMO

Nuclear pore complex (NPC) proteins (Nups) play multiple roles during mitosis. In this study we expand these roles and reveal that in Aspergillus nidulans, compromising the core Nup84-120 subcomplex of the NPC modifies the mitotic behavior of the nuclear envelope (NE). In wildtype cells, the NE undergoes simultaneous double pinching events to separate daughter nuclei during mitotic exit, whereas in Nup84-120 complex mutants, only one restriction of the NE is observed. Investigating the basis for this modified behavior of the NE in Nup deleted cells uncovered previously unrealized roles for core Nups in mitotic exit. During wildtype anaphase, the NE surrounds the two separating daughter DNA masses which typically flank the central nucleolus, to form three distinct nuclear compartments. In contrast, deletion of core Nups frequently results in early nucleolar eviction from the mitotic nucleus, in turn causing an uncharacteristic dumbbell-shaped NE morphology of anaphase nuclei with a nuclear membrane bridge connecting the two forming G1 nuclei. Importantly, the absence of the nucleolus between the separating daughter nuclei during anaphase delays chromosome segregation and progression into G1 as nuclei remain connected by chromatin bridges. Proteins localizing to late segregating chromosome arms are observed between forming daughter nuclei, and the mitotic spindle fails to resolve in a timely manner. These chromatin bridges are occupied by the Aurora kinase until nuclei have fully separated, suggesting involvement of Aurora in monitoring mitotic spindle and nuclear membrane resolution during mitotic exit. Our findings thus reveal a novel requirement for core Nups in mediating nucleolar positioning during mitosis, which dictates the pattern of NE fissions during karyokinesis and facilitates normal chromosome segregation. The findings additionally demonstrate that the mode of mitosis can be dramatically modified by deletion of a single NPC gene and reveals surprising fluidity in mitotic mechanisms.


Assuntos
Aspergillus nidulans/genética , Mitose/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Deleção de Sequência , Anáfase , Aspergillus nidulans/citologia , Nucléolo Celular , Núcleo Celular/metabolismo , Divisão do Núcleo Celular/fisiologia , Cromatina , Segregação de Cromossomos , DNA Ribossômico , Proteínas Fúngicas/genética , Mutação , Membrana Nuclear , Fuso Acromático
5.
Fungal Genet Biol ; 127: 50-59, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30849444

RESUMO

SUMOylation, covalent attachment of the small ubiquitin-like modifier protein SUMO to proteins, regulates protein interactions and activity and plays a crucial role in the regulation of many key cellular processes. Understanding the roles of SUMO in these processes ultimately requires identification of the proteins that are SUMOylated in the organism under study. The filamentous fungus Aspergillus nidulans serves as an excellent model for many aspects of fungal biology, and it would be of great value to determine the proteins that are SUMOylated in this organism (i.e. its SUMOylome). We have developed a new and effective approach for identifying SUMOylated proteins in this organism in which we lock proteins in their SUMOylated state, affinity purify SUMOylated proteins using the high affinity S-tag, and identify them using sensitive Orbitrap mass spectroscopy. This approach allows us to distinguish proteins that are SUMOylated from proteins that are binding partners of SUMOylated proteins or are bound non-covalently to SUMO. This approach has allowed us to identify 149 proteins that are SUMOylated in A. nidulans. Of these, 67 are predicted to be involved in transcription and particularly in the regulation of transcription, 21 are predicted to be involved in RNA processing and 16 are predicted to function in DNA replication or repair.


Assuntos
Aspergillus nidulans/química , Aspergillus nidulans/genética , Proteínas Fúngicas/química , Sumoilação , Proteínas Fúngicas/genética , Espectrometria de Massas , Processamento de Proteína Pós-Traducional , Proteômica , Transcrição Gênica
6.
Curr Opin Cell Biol ; 58: 42-49, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30798206

RESUMO

Eukaryotic cells rely on flux of macromolecules between the nucleus and the cytoplasm for growth and survival. Bidirectional transport is achieved through Nuclear Pore Complexes (NPCs) embedded in the Nuclear Envelope (NE). NPC proteins perform other cellular functions during mitosis, chromatin organization, DNA repair and gene regulation. Dysregulation of NPC number, or defects in their structure and function, are linked to numerous diseases but how NPCs are faithfully inherited during mitosis is poorly understood. In this review, we discuss recent insights to mechanisms of mammalian mitotic NPC segregation and NPC assembly as well as mitotic NPC inheritance via the mitotic chromatin located NPC protein Nup2 in Aspergillus nidulans. We suggest mitotic Nup2 chromatin-based mechanisms could also operate in vertebrate cells.


Assuntos
Células Eucarióticas/citologia , Mitose , Poro Nuclear/metabolismo , Animais , Aspergillus nidulans/citologia , Aspergillus nidulans/metabolismo , Núcleo Celular/metabolismo , Cromossomos/metabolismo , Células Eucarióticas/metabolismo , Humanos , Membrana Nuclear/metabolismo
7.
Mol Biol Cell ; 29(26): 3144-3154, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30355026

RESUMO

The nuclear pore complex (NPC) protein Nup2 plays interphase nuclear transport roles and in Aspergillus nidulans also functions to bridge NPCs at mitotic chromatin for their faithful coinheritance to daughter G1 nuclei. In this study, we further investigate the interphase functions of Nup2 in A. nidulans. Although Nup2 is not required for nuclear import of all nuclear proteins after mitosis, it is required for normal G1 nuclear accumulation of the NPC nuclear basket-associated components Mad2 and Mlp1 as well as the THO complex protein Tho2. Targeting of Mlp1 to nuclei partially rescues the interphase delay seen in nup2 mutants indicating that some of the interphase defects in Nup2-deleted cells are due to Mlp1 mislocalization. Among the inner nuclear membrane proteins, Nup2 affects the localization of Ima1, orthologues of which are involved in nuclear movement. Interestingly, nup2 mutant G1 nuclei also exhibit an abnormally long period of extensive to-and-fro movement immediately after mitosis in a manner dependent on the microtubule cytoskeleton. This indicates that Nup2 is required to limit the transient postmitotic nuclear migration typical of many filamentous fungi. The findings reveal that Nup2 is a multifunctional protein that performs diverse functions during both interphase and mitosis in A. nidulans.


Assuntos
Aspergillus nidulans/metabolismo , Interfase/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Transporte Ativo do Núcleo Celular , Aspergillus nidulans/genética , Núcleo Celular/metabolismo , Proteínas Fúngicas/metabolismo , Interfase/genética , Mitose , Membrana Nuclear/metabolismo , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia
8.
PLoS One ; 12(12): e0189077, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29194456

RESUMO

Endogenously tagging proteins with green fluorescent protein (GFP) enables the visualization of the tagged protein using live cell microscopy. GFP-tagging is widely utilized to study biological processes in model experimental organisms including filamentous fungi such as Aspergillus nidulans. Many strains of A. nidulans have therefore been generated with different proteins endogenously tagged with GFP. To further enhance experimental approaches based upon GFP-tagging, we have adapted the GFP Binding Protein (GBP) system for A. nidulans. GBP is a genetically encoded Llama single chain antibody against GFP which binds GFP with high affinity. Using gene replacement approaches, it is therefore possible to link GBP to anchor proteins, which will then retarget GFP-tagged proteins away from their normal location to the location of the anchor-GBP protein. To facilitate this approach in A. nidulans, we made four base plasmid cassettes that can be used to generate gene replacement GBP-tagging constructs by utilizing fusion PCR. Using these base cassettes, fusion PCR, and gene targeting approaches, we generated strains with SPA10-GBP and Tom20-GBP gene replacements. These strains enabled test targeting of GFP-tagged proteins to septa or to the surface of mitochondria respectively. SPA10-GBP is shown to effectively target GFP-tagged proteins to both forming and mature septa. Tom20-GBP has a higher capacity to retarget GFP-tagged proteins being able to relocate all Nup49-GFP from its location within nuclear pore complexes (NPCs) to the cytoplasm in association with mitochondria. Notably, removal of Nup49-GFP from NPCs causes cold sensitivity as does deletion of the nup49 gene. The cassette constructs described facilitate experimental approaches to generate precise protein-protein linkages in fungi. The A. nidulans SPA10-GBP and Tom20-GBP strains can be utilized to modulate other GFP-tagged proteins of interest.


Assuntos
Aspergillus nidulans/metabolismo , Proteínas Fúngicas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Mitocôndrias/metabolismo , Reação em Cadeia da Polimerase , Transporte Proteico
9.
Mol Microbiol ; 106(2): 285-303, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28787538

RESUMO

Microtubule-organizing centers (MTOCs) are large, multi-subunit protein complexes. Schizosaccharomyces pombe harbors MTOCs at spindle pole bodies, transient MTOCs in the division plane (eMTOCs) and nuclear-envelope associated MTOCs in interphase cells (iMTOCs). In the filamentous fungus Aspergillus nidulans SPBs and septum-associated MTOCs were described. Although comparable to S. pombe eMTOCs, A. nidulans sMTOCS are permanent septum-associated structures. The composition of sMTOCs is poorly understood and how they are targeted to septa was unknown. Here, we show that in A. nidulans several SPB outer plaque proteins also locate to sMTOCs while other SPB proteins do not, including SfiA, a protein required for SPB duplication in Saccharomyces cerevisiae and S. pombe and PcpA, the anchor for γ-TuSCs at the SPB inner plaque. The A. nidulans disordered protein Spa18Mto2 and the centrosomin-domain containing protein ApsBMto1 were required for recruiting the γ-TuRC component GcpC to sMTOCs and for seeding MT formation from septa. Testing different septum-associated proteins for a role in sMTOC function, Spa10 was identified. It forms a septal pore disc structure, recruits Spa18 and ApsB to septa and is required for sMTOC activity. This is the first evidence for a septum-specific protein, Spa10, as anchor for a specific class of MTOCs.


Assuntos
Aspergillus nidulans/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Sequência de Aminoácidos/genética , Proteínas Fúngicas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Ligação Proteica/fisiologia , Transporte Proteico/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
10.
J Cell Biol ; 216(9): 2813-2826, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28747316

RESUMO

Transport through nuclear pore complexes (NPCs) during interphase is facilitated by the nucleoporin Nup2 via its importin α- and Ran-binding domains. However, Aspergillus nidulans and vertebrate Nup2 also locate to chromatin during mitosis, suggestive of mitotic functions. In this study, we report that Nup2 is required for mitotic NPC inheritance in A. nidulans Interestingly, the role of Nup2 during mitotic NPC segregation is independent of its importin α- and Ran-binding domains but relies on a central targeting domain that is necessary for localization and viability. To test whether mitotic chromatin-associated Nup2 might function to bridge NPCs with chromatin during segregation, we provided an artificial link between NPCs and chromatin via Nup133 and histone H1. Using this approach, we bypassed the requirement of Nup2 for NPC segregation. This indicates that A. nidulans cells ensure accurate mitotic NPC segregation to daughter nuclei by linking mitotic DNA and NPC segregation via the mitotic specific chromatin association of Nup2.


Assuntos
Aspergillus nidulans/metabolismo , Proteínas Fúngicas/metabolismo , Mitose , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Poro Nuclear/metabolismo , Aspergillus nidulans/genética , Aspergillus nidulans/crescimento & desenvolvimento , Cromatina/genética , Cromatina/metabolismo , DNA Fúngico/genética , DNA Fúngico/metabolismo , Proteínas Fúngicas/genética , Histonas/metabolismo , Microscopia de Fluorescência , Mutação , Poro Nuclear/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Transdução de Sinais , Fatores de Tempo , Imagem com Lapso de Tempo
11.
Fungal Genet Biol ; 103: 1-15, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28315405

RESUMO

Filamentous fungi have devastating negative impacts as pathogens and agents of food spoilage but also have critical ecological importance and are utilized for industrial applications. The characteristic multinucleate nature of filamentous fungi is facilitated by limiting if, when and where septation, the fungal equivalent of cytokinesis, occurs. In the model filamentous fungus Aspergillus nidulans septation does not occur immediately after mitosis and is an incomplete process resulting in the formation of a septal pore whose permeability is cell cycle regulated. How mitotic regulators, such as the Aurora kinase, contribute to the often unique biology of filamentous fungi is not well understood. The Aurora B kinase has not previously been investigated in any detail during hyphal growth. Here we demonstrate for the first time that Aurora displays cell cycle dependent locations to the region of forming septa, the septal pore and mature septa as well as the mitotic apparatus. To functionally analyze Aurora, we generated a temperature sensitive allele revealing essential mitotic and spindle assembly checkpoint functions consistent with its location to the kinetochore region and spindle midzone. Our analysis also reveals that cellular and kinetochore Aurora levels increase during a mitotic spindle assembly checkpoint arrest and we propose that this could be important for checkpoint inactivation when spindle formation is prevented. We demonstrate that Aurora accumulation at mature septa following mitotic entry does not require mitotic progression but is dependent upon a timing mechanism. Surprisingly we also find that Aurora inactivation leads to cellular swelling and lysis indicating an unexpected function for Aurora in fungal cell growth. Thus in addition to its conserved mitotic functions our data suggest that Aurora has the capacity to be an important regulator of septal biology and cell growth in filamentous fungi.


Assuntos
Aspergillus nidulans/genética , Aurora Quinase B/genética , Ciclo Celular/genética , Mitose/genética , Aspergillus nidulans/enzimologia , Aspergillus nidulans/crescimento & desenvolvimento , Citocinese/genética , Cinetocoros/enzimologia , Microtúbulos/enzimologia , Microtúbulos/genética , Fuso Acromático/enzimologia
12.
Mol Biol Cell ; 28(5): 634-644, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057761

RESUMO

How microtubules (MTs) are regulated during fungal biofilm formation is unknown. By tracking MT +end-binding proteins (+TIPS) in Aspergillus nidulans, we find that MTs are regulated to depolymerize within forming fungal biofilms. During this process, EB1, dynein, and ClipA form transient fibrous and then bar-like structures, novel configurations for +TIPS. Cells also respond in an autonomous manner, with cells separated by a septum able to maintain different MT dynamics. Surprisingly, all cells with depolymerized MTs rapidly repolymerize their MTs after air exchange above the static culture medium of biofilms. Although the specific gasotransmitter for this biofilm response is not known, we find that addition of hydrogen sulfide gas to growing cells recapitulates all aspects of reversible MT depolymerization and transient formation of +TIPs bars. However, as biofilms mature, physical removal of part of the biofilm is required to promote MT repolymerization, which occurs at the new biofilm edge. We further show MT depolymerization within biofilms is regulated by the SrbA hypoxic transcription factor and that without SrbA, MTs are maintained as biofilms form. This reveals a new mode of MT regulation in response to changing gaseous biofilm microenvironments, which could contribute to the unique characteristics of fungal biofilms in medical and industrial settings.


Assuntos
Aspergillus nidulans/fisiologia , Biofilmes/crescimento & desenvolvimento , Microtúbulos/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Aspergillus nidulans/metabolismo , Microambiente Celular/fisiologia , Dineínas/metabolismo , Gases , Proteínas Associadas aos Microtúbulos/metabolismo , Polimerização
13.
Mol Biol Cell ; 2016 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-27630260

RESUMO

During Aspergillus nidulans mitosis peripheral nuclear pore complex (NPC) proteins (Nups) disperse from the core NPC structure. Unexpectedly, one predicted peripheral Nup, Gle1, remains at the mitotic NE via an unknown mechanism. Gle1 affinity purification identified MtgA ( M: itotic T: ether for G: le1), which tethers Gle1 to the NE during mitosis, but not during interphase when Gle1 is at NPCs. MtgA is the ortholog of the Schizosaccharomyces pombe telomere-anchoring inner nuclear membrane protein Bqt4. Like Bqt4, MtgA has meiotic roles but is functionally distinct from Bqt4 as MtgA is not required for tethering telomeres to the NE. Domain analyses revealed MtgA targeting to the NE requires its C-terminal transmembrane domain and a nuclear localization signal. Importantly, MtgA functions beyond Gle1 mitotic targeting and meiosis and impacts nuclear and nucleolar architecture when deleted or overexpressed. Deletion of MtgA generates small, round nuclei whereas overexpressing MtgA generates larger nuclei with altered nuclear compartmentalization resulting from NE expansion around the nucleolus. The accumulation of MtgA around the nucleolus promotes a similar accumulation of the endoplasmic reticulum (ER) protein Erg24 lowering its levels in the ER. This study extends the functions of Bqt4-like proteins to include mitotic Gle1 targeting and modulation of nuclear and nucleolar architecture.

14.
PLoS One ; 10(7): e0132489, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147902

RESUMO

How membranes and associated proteins of the nuclear envelope (NE) are assembled specifically and inclusively around segregated genomes during exit from mitosis is incompletely understood. Inner nuclear membrane (INM) proteins play key roles by providing links between DNA and the NE. In this study we have investigated the highly conserved INM protein Src1 in Aspergillus nidulans and have uncovered a novel cell cycle response during post mitotic formation of G1 nuclei. Live cell imaging indicates Src1 could have roles during mitotic exit as it preferentially locates to the NE abscission points during nucleokinesis and to the NE surrounding forming daughter G1 nuclei. Deletion analysis further supported this idea revealing that although Src1 is not required for interphase progression or mitosis it is required for stable post-mitotic G1 nuclear formation. This conclusion is based upon the observation that in the absence of Src1 newly formed G1 nuclei are structurally unstable and immediately undergo architectural modifications typical of mitosis. These changes include NPC modifications that stop nuclear transport as well as disassembly of nucleoli. More intriguingly, the newly generated G1 nuclei then cycle between mitotic- and interphase-like states. The findings indicate that defects in post-mitotic G1 nuclear formation caused by lack of Src1 promote repeated failed attempts to generate stable G1 nuclei. To explain this unexpected phenotype we suggest a type of regulation that promotes repetition of defective cell cycle transitions rather than preventing progression past the defective cell cycle transition. We suggest the term "reboot regulation" to define this mode of cell cycle regulation. The findings are discussed in relationship to recent studies showing the Cdk1 master oscillator can entrain subservient oscillators that when uncoupled cause cell cycle transitions to be repeated.


Assuntos
Aspergillus nidulans/metabolismo , Proteínas Fúngicas/metabolismo , Fase G1/fisiologia , Mitose/fisiologia , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Aspergillus nidulans/genética , Proteínas Fúngicas/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética
15.
Mol Biol Cell ; 26(4): 605-21, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25540430

RESUMO

Chromatin and nuclear pore complexes (NPCs) undergo dramatic changes during mitosis, which in vertebrates and Aspergillus nidulans involves movement of Nup2 from NPCs to the chromatin region to fulfill unknown functions. This transition is shown to require the Cdk1 mitotic kinase and be promoted prematurely by ectopic expression of the NIMA kinase. Nup2 localizes with a copurifying partner termed NupA, a highly divergent yet essential NPC protein. NupA and Nup2 locate throughout the chromatin region during prophase but during anaphase move to surround segregating DNA. NupA function is shown to involve targeting Nup2 to its interphase and mitotic locations. Deletion of either Nup2 or NupA causes identical mitotic defects that initiate a spindle assembly checkpoint (SAC)-dependent mitotic delay and also cause defects in karyokinesis. These mitotic problems are not caused by overall defects in mitotic NPC disassembly-reassembly or general nuclear import. However, without Nup2 or NupA, although the SAC protein Mad1 locates to its mitotic locations, it fails to locate to NPCs normally in G1 after mitosis. Collectively the study provides new insight into the roles of Nup2 and NupA during mitosis and in a surveillance mechanism that regulates nucleokinesis when mitotic defects occur after SAC fulfillment.


Assuntos
Aspergillus nidulans/metabolismo , Proteínas Fúngicas/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/fisiologia , Poro Nuclear/metabolismo , Aspergillus nidulans/citologia , Aspergillus nidulans/genética , Cromatina/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Deleção de Genes , Mitose/genética , Mitose/fisiologia , Poro Nuclear/fisiologia , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo
16.
Genetics ; 197(4): 1225-36, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24835271

RESUMO

Mitosis is promoted and regulated by reversible protein phosphorylation catalyzed by the essential NIMA and CDK1 kinases in the model filamentous fungus Aspergillus nidulans. Protein methylation mediated by the Set1/COMPASS methyltransferase complex has also been shown to regulate mitosis in budding yeast with the Aurora mitotic kinase. We uncover a genetic interaction between An-swd1, which encodes a subunit of the Set1 protein methyltransferase complex, with NIMA as partial inactivation of nimA is poorly tolerated in the absence of swd1. This genetic interaction is additionally seen without the Set1 methyltransferase catalytic subunit. Importantly partial inactivation of NIMT, a mitotic activator of the CDK1 kinase, also causes lethality in the absence of Set1 function, revealing a functional relationship between the Set1 complex and two pivotal mitotic kinases. The main target for Set1-mediated methylation is histone H3K4. Mutational analysis of histone H3 revealed that modifying the H3K4 target residue of Set1 methyltransferase activity phenocopied the lethality seen when either NIMA or CDK1 are partially functional. We probed the mechanistic basis of these genetic interactions and find that the Set1 complex performs functions with CDK1 for initiating mitosis and with NIMA during progression through mitosis. The studies uncover a joint requirement for the Set1 methyltransferase complex with the CDK1 and NIMA kinases for successful mitosis. The findings extend the roles of the Set1 complex to include the initiation of mitosis with CDK1 and mitotic progression with NIMA in addition to its previously identified interactions with Aurora and type 1 phosphatase in budding yeast.


Assuntos
Aspergillus nidulans/citologia , Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Fúngicas/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Aspergillus nidulans/genética , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/genética , Metilação de DNA , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Mitose/genética , Quinase 1 Relacionada a NIMA , Fosforilação , Proteínas Serina-Treonina Quinases/genética
17.
PLoS One ; 9(3): e90911, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24599037

RESUMO

Filamentous fungi occupy critical environmental niches and have numerous beneficial industrial applications but devastating effects as pathogens and agents of food spoilage. As regulators of essentially all biological processes protein kinases have been intensively studied but how they regulate the often unique biology of filamentous fungi is not completely understood. Significant understanding of filamentous fungal biology has come from the study of the model organism Aspergillus nidulans using a combination of molecular genetics, biochemistry, cell biology and genomic approaches. Here we describe dual localization-affinity purification (DLAP) tags enabling endogenous N or C-terminal protein tagging for localization and biochemical studies in A. nidulans. To establish DLAP tag utility we endogenously tagged 17 protein kinases for analysis by live cell imaging and affinity purification. Proteomic analysis of purifications by mass spectrometry confirmed association of the CotA and NimXCdk1 kinases with known binding partners and verified a predicted interaction of the SldABub1/R1 spindle assembly checkpoint kinase with SldBBub3. We demonstrate that the single TOR kinase of A. nidulans locates to vacuoles and vesicles, suggesting that the function of endomembranes as major TOR cellular hubs is conserved in filamentous fungi. Comparative analysis revealed 7 kinases with mitotic specific locations including An-Cdc7 which unexpectedly located to mitotic spindle pole bodies (SPBs), the first such localization described for this family of DNA replication kinases. We show that the SepH septation kinase locates to SPBs specifically in the basal region of apical cells in a biphasic manner during mitosis and again during septation. This results in gradients of SepH between G1 SPBs which shift along hyphae as each septum forms. We propose that SepH regulates the septation initiation network (SIN) specifically at SPBs in the basal region of G1 cells and that localized gradients of SIN activity promote asymmetric septation.


Assuntos
Aspergillus nidulans/enzimologia , Cromatografia de Afinidade/métodos , Proteínas Quinases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Aspergillus nidulans/citologia , Aspergillus nidulans/efeitos dos fármacos , Aspergillus nidulans/crescimento & desenvolvimento , Benomilo/farmacologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/enzimologia , Vesículas Citoplasmáticas/efeitos dos fármacos , Vesículas Citoplasmáticas/enzimologia , Proteínas Fúngicas/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Interfase/efeitos dos fármacos , Cinetocoros/efeitos dos fármacos , Cinetocoros/enzimologia , Microtúbulos/efeitos dos fármacos , Microtúbulos/enzimologia , Mitose/efeitos dos fármacos , Dados de Sequência Molecular , Proteínas Quinases/química , Transporte Proteico/efeitos dos fármacos , Proteômica , Corpos Polares do Fuso/efeitos dos fármacos , Corpos Polares do Fuso/enzimologia , Vacúolos/efeitos dos fármacos , Vacúolos/enzimologia
18.
PLoS Genet ; 10(3): e1004248, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24675878

RESUMO

The Never in Mitosis A (NIMA) kinase (the founding member of the Nek family of kinases) has been considered a mitotic specific kinase with nuclear restricted roles in the model fungus Aspergillus nidulans. By extending to A. nidulans the results of a synthetic lethal screen performed in Saccharomyces cerevisiae using the NIMA ortholog KIN3, we identified a conserved genetic interaction between nimA and genes encoding proteins of the Endosomal Sorting Complex Required for Transport (ESCRT) pathway. Absence of ESCRT pathway functions in combination with partial NIMA function causes enhanced cell growth defects, including an inability to maintain a single polarized dominant cell tip. These genetic insights suggest NIMA potentially has interphase functions in addition to its established mitotic functions at nuclei. We therefore generated endogenously GFP-tagged NIMA (NIMA-GFP) which was fully functional to follow its interphase locations using live cell spinning disc 4D confocal microscopy. During interphase some NIMA-GFP locates to the tips of rapidly growing cells and, when expressed ectopically, also locates to the tips of cytoplasmic microtubules, suggestive of non-nuclear interphase functions. In support of this, perturbation of NIMA function either by ectopic overexpression or through partial inactivation results in marked cell tip growth defects with excess NIMA-GFP promoting multiple growing cell tips. Ectopic NIMA-GFP was found to locate to the plus ends of microtubules in an EB1 dependent manner, while impairing NIMA function altered the dynamic localization of EB1 and the cytoplasmic microtubule network. Together, our genetic and cell biological analyses reveal novel non-nuclear interphase functions for NIMA involving microtubules and the ESCRT pathway for normal polarized fungal cell tip growth. These insights extend the roles of NIMA both spatially and temporally and indicate that this conserved protein kinase could help integrate cell cycle progression with polarized cell growth.


Assuntos
Proteínas de Ciclo Celular/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Interfase/genética , Microtúbulos/genética , Proteínas Serina-Treonina Quinases/genética , Aspergillus nidulans/genética , Ciclo Celular/genética , Núcleo Celular/genética , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Proteínas de Fluorescência Verde , Mitose/genética , Quinase 1 Relacionada a NIMA
19.
Mol Biol Cell ; 25(6): 763-75, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24451264

RESUMO

Intercellular bridges are a conserved feature of multicellular organisms. In multicellular fungi, cells are connected directly via intercellular bridges called septal pores. Using Aspergillus nidulans, we demonstrate for the first time that septal pores are regulated to be opened during interphase but closed during mitosis. Septal pore-associated proteins display dynamic cell cycle-regulated locations at mature septa. Of importance, the mitotic NIMA kinase locates to forming septa and surprisingly then remains at septa throughout interphase. However, during mitosis, when NIMA transiently locates to nuclei to promote mitosis, its levels at septa drop. A model is proposed in which NIMA helps keep septal pores open during interphase and then closed when it is removed from them during mitosis. In support of this hypothesis, NIMA inactivation is shown to promote interphase septal pore closing. Because NIMA triggers nuclear pore complex opening during mitosis, our findings suggest that common cell cycle regulatory mechanisms might control septal pores and nuclear pores such that they are opened and closed out of phase to each other during cell cycle progression. The study provides insights into how and why cytoplasmically connected Aspergillus cells maintain mitotic autonomy.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Fúngicas/genética , Regulação Fúngica da Expressão Gênica , Mitose , Proteínas Serina-Treonina Quinases/genética , Aspergillus nidulans/citologia , Aspergillus nidulans/genética , Aspergillus nidulans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Fúngicas/metabolismo , Interfase/genética , Quinase 1 Relacionada a NIMA , Poro Nuclear/química , Poro Nuclear/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais
20.
Eukaryot Cell ; 13(1): 99-109, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24186954

RESUMO

The G2-M transition in Aspergillus nidulans requires the NIMA kinase, the founding member of the Nek kinase family. Inactivation of NIMA results in a late G2 arrest, while overexpression of NIMA is sufficient to promote mitotic events independently of cell cycle phase. Endogenously tagged NIMA-GFP has dynamic mitotic localizations appearing first at the spindle pole body and then at nuclear pore complexes before transitioning to within nuclei and the mitotic spindle and back at the spindle pole bodies at mitotic exit, suggesting that it functions sequentially at these locations. Since NIMA is indispensable for mitotic entry, it has been difficult to determine the requirement of NIMA for subaspects of mitosis. We show here that when NIMA is partially inactivated, although mitosis can be initiated, a proportion of cells fail to successfully generate two daughter nuclei. We further define the mitotic defects to show that normal NIMA function is required for the formation of a bipolar spindle, nuclear pore complex disassembly, completion of chromatin segregation, and the normal structural rearrangements of the nuclear envelope required to generate two nuclei from one. In the remaining population of cells that enter mitosis with inadequate NIMA, two daughter nuclei are generated in a manner dependent on the spindle assembly checkpoint, indicating highly penetrant defects in mitotic progression without sufficient NIMA activity. This study shows that NIMA is required not only for mitotic entry but also sequentially for successful completion of stage-specific mitotic events.


Assuntos
Aspergillus nidulans/enzimologia , Proteínas de Ciclo Celular/metabolismo , Proteínas Fúngicas/metabolismo , Mitose/genética , Proteínas Serina-Treonina Quinases/metabolismo , Aspergillus nidulans/genética , Aspergillus nidulans/fisiologia , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , Proteínas Fúngicas/genética , Quinase 1 Relacionada a NIMA , Poro Nuclear/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fuso Acromático/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...