Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Biochem Soc Trans ; 51(4): 1505-1520, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37560910

RESUMEN

Kinesin motor proteins couple mechanical movements in their motor domain to the binding and hydrolysis of ATP in their nucleotide-binding pocket. Forces produced through this 'mechanochemical' coupling are typically used to mobilize kinesin-mediated transport of cargos along microtubules or microtubule cytoskeleton remodeling. This review discusses the recent high-resolution structures (<4 Å) of kinesins bound to microtubules or tubulin complexes that have resolved outstanding questions about the basis of mechanochemical coupling, and how family-specific modifications of the motor domain can enable its use for motility and/or microtubule depolymerization.


Asunto(s)
Cinesinas , Tubulina (Proteína) , Cinesinas/metabolismo , Tubulina (Proteína)/análisis , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Adenosina Trifosfato/metabolismo , Microtúbulos/metabolismo , Miosinas
2.
Hum Mol Genet ; 29(5): 766-784, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-31919497

RESUMEN

By using the Cre-mediated genetic switch technology, we were able to successfully generate a conditional knock-in mouse, bearing the KIF2A p.His321Asp missense point variant, identified in a subject with malformations of cortical development. These mice present with neuroanatomical anomalies and microcephaly associated with behavioral deficiencies and susceptibility to epilepsy, correlating with the described human phenotype. Using the flexibility of this model, we investigated RosaCre-, NestinCre- and NexCre-driven expression of the mutation to dissect the pathophysiological mechanisms underlying neurodevelopmental cortical abnormalities. We show that the expression of the p.His321Asp pathogenic variant increases apoptosis and causes abnormal multipolar to bipolar transition in newborn neurons, providing therefore insights to better understand cortical organization and brain growth defects that characterize KIF2A-related human disorders. We further demonstrate that the observed cellular phenotypes are likely to be linked to deficiency in the microtubule depolymerizing function of KIF2A.


Asunto(s)
Conducta Animal , Cinesinas/fisiología , Malformaciones del Desarrollo Cortical/patología , Mutación , Neuronas/patología , Proteínas Represoras/fisiología , Animales , Masculino , Malformaciones del Desarrollo Cortical/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo
3.
J Am Chem Soc ; 143(18): 6847-6854, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33938740

RESUMEN

Cancer metastasis is a complex process involving highly motile tumor cells that breach tissue barriers, enter the bloodstream and lymphatic system, and disseminate throughout the body as circulating tumor cells. The primary cellular mechanism contributing to these critical events is the reorganization of the actin cytoskeleton. Mycalolide B (MycB) is an actin-targeting marine macrolide that can suppress proliferation, migration, and invasion of breast and ovarian cancer cells at low nanomolar doses. Through structure-activity relationship studies focused on the actin-binding tail region (C24-C35) of MycB, we identified a potent truncated derivative that inhibits polymerization of G-actin and severs F-actin by binding to actin's barbed end cleft. Biological analyses of this miniature MycB derivative demonstrate that it causes a rapid collapse of the actin cytoskeleton in ovarian cancer cells and impairs cancer cell motility and invasion of the extracellular matrix (ECM) by inhibiting invadopodia-mediated ECM degradation. These studies provide essential proof-of-principle for developing actin-targeting therapeutic agents to block cancer metastasis and establish a synthetically tractable barbed end-binding pharmacophore that can be further improved by adding targeting groups for precision drug design.


Asunto(s)
Actinas/antagonistas & inhibidores , Antineoplásicos/farmacología , Matriz Extracelular/efectos de los fármacos , Toxinas Marinas/farmacología , Oxazoles/farmacología , Actinas/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Matriz Extracelular/metabolismo , Femenino , Humanos , Toxinas Marinas/síntesis química , Toxinas Marinas/química , Modelos Moleculares , Estructura Molecular , Oxazoles/síntesis química , Oxazoles/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
4.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388224

RESUMEN

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Asunto(s)
Anomalías Congénitas/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedades Renales/congénito , Riñón/anomalías , Cinesinas/genética , Mutación con Pérdida de Función , Microcefalia/genética , Proteínas Oncogénicas/genética , Animales , Anomalías Congénitas/metabolismo , Citocinesis/genética , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Genes Letales , Estudios de Asociación Genética/métodos , Sitios Genéticos , Humanos , Riñón/metabolismo , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Masculino , Microcefalia/metabolismo , Microcefalia/patología , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Linaje , Fenotipo , Relación Estructura-Actividad , Pez Cebra
5.
BMC Biochem ; 19(1): 8, 2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30115012

RESUMEN

BACKGROUND: Stilbene cleaving oxygenases (SCOs), also known as lignostilbene-α,ß-dioxygenases (LSDs) mediate the oxidative cleavage of the olefinic double bonds of lignin-derived intermediate phenolic stilbenes, yielding small modified benzaldehyde compounds. SCOs represent one branch of the larger carotenoid cleavage oxygenases family. Here, we describe the structural and functional characterization of an SCO-like enzyme from the soil-born, bio-control agent Pseudomonas brassicacearum. METHODS: In vitro and in vivo assays relying on visual inspection, spectrophotometric quantification, as well as liquid-chormatographic and mass spectrometric characterization were applied for functional evaluation of the enzyme. X-ray crystallographic analyses and in silico modeling were applied for structural investigations. RESULTS: In vitro assays demonstrated preferential cleavage of resveratrol, while in vivo analyses detected putative cleavage of the straight chain carotenoid, lycopene. A high-resolution structure containing the seven-bladed ß-propeller fold and conserved 4-His-Fe unit at the catalytic site, was obtained. Comparative structural alignments, as well as in silico modelling and docking, highlight potential molecular factors contributing to both the primary in vitro activity against resveratrol, as well as the putative subsidiary activities against carotenoids in vivo, for future validation. CONCLUSIONS: The findings reported here provide validation of the SCO structure, and highlight enigmatic points with respect to the potential effect of the enzyme's molecular environment on substrate specificities for future investigation.


Asunto(s)
Dioxigenasas/química , Dioxigenasas/metabolismo , Pseudomonas/enzimología , Cristalografía por Rayos X , Dioxigenasas/clasificación , Simulación del Acoplamiento Molecular , Filogenia , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados , Microbiología del Suelo , Especificidad por Sustrato
6.
Biochem Cell Biol ; 94(2): 197-204, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26935656

RESUMEN

Glucosamine-6-phosphate N-acetyltransferase1 (GNA1) catalyses the transfer of an acetyl group from acetyl coenzyme A (AcCoA) to glucosamine-6-phosphate (GlcN6P) to form N-acetylglucosamine-6-phosphate (GlcNAc6P), which is an essential intermediate in UDP-GlcNAc biosynthesis. An analog of GlcNAc, N-butyrylglucosamine (GlcNBu) has shown healing properties for bone and articular cartilage in animal models of arthritis. The goal of this work was to examine whether GNA1 has the ability to transfer a butyryl group from butyryl-CoA to GlcN6P to form GlcNBu6P, which can then be converted to GlcNBu. We developed fluorescent and radioactive assays and examined the donor specificity of human GNA1. Acetyl, propionyl, n-butyryl, and isobutyryl groups were all transferred to GlcN6P, but isovaleryl-CoA and decanoyl-CoA did not serve as donor substrates. Site-specific mutants were produced to examine the role of amino acids potentially affecting the size and properties of the AcCoA binding pocket. All of the wild type and mutant enzymes showed activities of both acetyl and butyryl transfer and can therefore be used for the enzymatic synthesis of GlcNBu for biomedical applications.


Asunto(s)
Acetilcoenzima A/metabolismo , Carbono/metabolismo , Glucosamina 6-Fosfato N-Acetiltransferasa/metabolismo , Acetilcoenzima A/química , Carbono/química , Fluorescencia , Glucosamina/análogos & derivados , Glucosamina/biosíntesis , Glucosamina/química , Glucosamina 6-Fosfato N-Acetiltransferasa/química , Glucosamina 6-Fosfato N-Acetiltransferasa/genética , Glucosa-6-Fosfato/análogos & derivados , Glucosa-6-Fosfato/biosíntesis , Glucosa-6-Fosfato/química , Humanos , Espectrofotometría
7.
Eukaryot Cell ; 14(8): 755-74, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26024903

RESUMEN

Candida albicans is a major fungal pathogen whose virulence is associated with its ability to transition from a budding yeast form to invasive hyphal filaments. The kinesin-14 family member CaKar3 is required for transition between these morphological states, as well as for mitotic progression and karyogamy. While kinesin-14 proteins are ubiquitous, CaKar3 homologs in hemiascomycete fungi are unique because they form heterodimers with noncatalytic kinesin-like proteins. Thus, CaKar3-based motors may represent a novel antifungal drug target. We have identified and examined the roles of a kinesin-like regulator of CaKar3. We show that orf19.306 (dubbed CaCIK1) encodes a protein that forms a heterodimer with CaKar3, localizes CaKar3 to spindle pole bodies, and can bind microtubules and influence CaKar3 mechanochemistry despite lacking an ATPase activity of its own. Similar to CaKar3 depletion, loss of CaCik1 results in cell cycle arrest, filamentation defects, and an inability to undergo karyogamy. Furthermore, an examination of the spindle structure in cells lacking either of these proteins shows that a large proportion have a monopolar spindle or two dissociated half-spindles, a phenotype unique to the C. albicans kinesin-14 homolog. These findings provide new insights into mitotic spindle structure and kinesin motor function in C. albicans and identify a potentially vulnerable target for antifungal drug development.


Asunto(s)
Candida albicans/metabolismo , Proteínas Fúngicas/metabolismo , Cinesinas/metabolismo , Morfogénesis/fisiología , Huso Acromático/metabolismo , Adenosina Trifosfatasas/metabolismo , Antifúngicos/metabolismo , Puntos de Control del Ciclo Celular/fisiología , Quinasas Ciclina-Dependientes/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Mitosis/fisiología
8.
J Biol Chem ; 288(52): 36957-70, 2013 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-24240171

RESUMEN

Force production by kinesins has been linked to structural rearrangements of the N and C termini of their motor domain upon nucleotide binding. In recent crystal structures, the Kar3-associated protein Vik1 shows unexpected homology to these conformational states even though it lacks a nucleotide-binding site. This conservation infers a degree of commonality in the function of the N- and C-terminal regions during the mechanochemical cycle of all kinesins and kinesin-related proteins. We tested this inference by examining the functional effects on Kar3Vik1 of mutating or deleting residues in Vik1 that are involved in stabilizing the C terminus against the core and N terminus of the Vik1 motor homology domain (MHD). Point mutations at two moderately conserved residues near the Vik1 C terminus impaired microtubule gliding and microtubule-stimulated ATP turnover by Kar3Vik1. Deletion of the seven C-terminal residues inhibited Kar3Vik1 motility much more drastically. Interestingly, none of the point mutants seemed to perturb the ability of Kar3Vik1 to bind microtubules, whereas the C-terminal truncation mutant did. Molecular dynamics simulations of these C-terminal mutants showed distinct root mean square fluctuations in the N-terminal region of the Vik1 MHD that connects it to Kar3. Here, the degree of motion in the N-terminal portion of Vik1 highly correlated with that in the C terminus. These observations suggest that the N and C termini of the Vik1 MHD form a discrete folding motif that is part of a communication pathway to the nucleotide-binding site of Kar3.


Asunto(s)
Secuencia de Aminoácidos , Candida glabrata/química , Proteínas Fúngicas/química , Proteínas Asociadas a Microtúbulos/química , Mutación Puntual , Eliminación de Secuencia , Secuencias de Aminoácidos , Candida glabrata/genética , Candida glabrata/metabolismo , Cristalografía por Rayos X , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/química , Microtúbulos/genética , Microtúbulos/metabolismo , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
9.
J Med Chem ; 67(7): 5315-5332, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38401158

RESUMEN

Actin barbed end-binding macrolides have been shown to inhibit cancer cell motility and invasion of extracellular matrix (ECM), evoking their potential utility as therapies for metastatic cancers. Unfortunately, the direct use of these compounds in clinical settings is impeded by their limited natural abundance, challenging total synthesis, and detrimental effects on normal tissues. To develop potent analogues of these compounds that are simpler to synthesize and compatible with cell-specific targeting systems, such as antibodies, we designed over 20 analogues of the acyclic side chain (tail) of the macrolide Mycalolide B. These analogues probed the contributions of four distinct regions of the tail towards the inhibition of actin polymerization and ECM invasion by human lung cancer A549 cells. We observed that two of these regions tolerate considerable substituent variability, and we identified a specific combination of substituents that leads to the optimal inhibition of the ECM invasion activity of A549 cells.


Asunto(s)
Actinas , Neoplasias Pulmonares , Humanos , Macrólidos/farmacología , Movimiento Celular , Invasividad Neoplásica/prevención & control
10.
J Biol Chem ; 287(48): 40292-301, 2012 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-23043140

RESUMEN

BACKGROUND: Kar3Vik1 is a heterodimeric kinesin with one catalytic subunit (Kar3) and one noncatalytic subunit (Vik1). RESULTS: Vik1 experiences conformational changes in regions analogous to the force-producing elements in catalytic kinesins. CONCLUSION: A molecular mechanism by which Kar3 could trigger Vik1's release from microtubules was revealed. SIGNIFICANCE: These findings will serve as the prototype for understanding the motile mechanism of kinesin-14 motors in general. It is widely accepted that movement of kinesin motor proteins is accomplished by coupling ATP binding, hydrolysis, and product release to conformational changes in the microtubule-binding and force-generating elements of their motor domain. Therefore, understanding how the Saccharomyces cerevisiae proteins Cik1 and Vik1 are able to function as direct participants in movement of Kar3Cik1 and Kar3Vik1 kinesin complexes presents an interesting challenge given that their motor homology domain (MHD) cannot bind ATP. Our crystal structures of the Vik1 ortholog from Candida glabrata may provide insight into this mechanism by showing that its neck and neck mimic-like element can adopt several different conformations reminiscent of those observed in catalytic kinesins. We found that when the neck is α-helical and interacting with the MHD core, the C terminus of CgVik1 docks onto the central ß-sheet similarly to the ATP-bound form of Ncd. Alternatively, when neck-core interactions are broken, the C terminus is disordered. Mutations designed to impair neck rotation, or some of the neck-MHD interactions, decreased microtubule gliding velocity and steady state ATPase rate of CgKar3Vik1 complexes significantly. These results strongly suggest that neck rotation and neck mimic docking in Vik1 and Cik1 may be a structural mechanism for communication with Kar3.


Asunto(s)
Proteínas Fúngicas/química , Proteínas Asociadas a Microtúbulos/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Dominio Catalítico , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Cinesinas/metabolismo , Proteínas de Microtúbulos/química , Proteínas de Microtúbulos/genética , Proteínas de Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Conformación Proteica , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
11.
Front Cell Infect Microbiol ; 13: 1287418, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38239502

RESUMEN

Introduction: The fungal G protein-coupled receptors Ste2 and Ste3 are vital in mediating directional hyphal growth of the agricultural pathogen Fusarium graminearum towards wheat plants. This chemotropism is induced by a catalytic product of peroxidases secreted by the wheat. Currently, the identity of this product, and the substrate it is generated from, are not known. Methods and results: We provide evidence that a peroxidase substrate is derived from F. graminearum conidia and report a simple method to extract and purify the FgSte2-activating ligand for analyses by mass spectrometry. The mass spectra arising from t he ligand extract are characteristic of a 400 Da carbohydrate moiety. Consistent with this type of molecule, glycosidase treatment of F. graminearum conidia prior to peroxidase treatment significantly reduced the amount of ligand extracted. Interestingly, availability of the peroxidase substrate appears to depend on the presence of both FgSte2 and FgSte3, as knockout of one or the other reduces the chemotropism-inducing effect of the extracts. Conclusions: While further characterization is necessary, identification of the F. graminearum-derived peroxidase substrate and the FgSte2-activating ligand will unearth deeper insights into the intricate mechanisms that underlie fungal pathogenesis in cereal crops, unveiling novel avenues for inhibitory interventions.


Asunto(s)
Fusarium , Peroxidasa , Ligandos , Peroxidasas/farmacología , Enfermedades de las Plantas/microbiología
12.
J Biol Chem ; 286(35): 30384-30392, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21757693

RESUMEN

We employed budding yeast and skeletal muscle actin to examine the contribution of the actin isoform to myosin motor function. While yeast and muscle actin are highly homologous, they exhibit different charge density at their N termini (a proposed myosin-binding interface). Muscle myosin-II actin-activated ATPase activity is significantly higher with muscle versus yeast actin. Whether this reflects inefficiency in the ability of yeast actin to activate myosin is not known. Here we optimized the isolation of two yeast myosins to assess actin function in a homogenous system. Yeast myosin-II (Myo1p) and myosin-V (Myo2p) accommodate the reduced N-terminal charge density of yeast actin, showing greater activity with yeast over muscle actin. Increasing the number of negative charges at the N terminus of yeast actin from two to four (as in muscle) had little effect on yeast myosin activity, while other substitutions of charged residues at the myosin interface of yeast actin reduced activity. Thus, yeast actin functions most effectively with its native myosins, which in part relies on associations mediated by its outer domain. Compared with yeast myosin-II and myosin-V, muscle myosin-II activity was very sensitive to salt. Collectively, our findings suggest differing degrees of reliance on electrostatic interactions during weak actomyosin binding in yeast versus muscle. Our study also highlights the importance of native actin isoforms when considering the function of myosins.


Asunto(s)
Actinas/química , Miosina Tipo II/química , Miosina Tipo I/química , Saccharomyces cerevisiae/metabolismo , Actomiosina/química , Secuencia de Aminoácidos , Dictyostelium/metabolismo , Regulación de la Expresión Génica , Humanos , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Miosina Tipo I/metabolismo , Miosina Tipo II/metabolismo , Unión Proteica , Conformación Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína , Electricidad Estática
13.
Proteins ; 80(4): 1016-27, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22493778

RESUMEN

Kar3 kinesins are microtubule (MT) minus-end-directed motors with pleiotropic functions in mitotic spindle formation and nuclear movement in budding and fission yeasts. A Kar3-like kinesin is also expressed by the filamentous fungus Ashbya gossypi, which exhibits different nuclear movement challenges from its yeast relatives. Presented here is a 2.35 Å crystal structure and enzymatic analysis of the AgKar3 motor domain (AgKar3MD). Compared to the previously published Saccharomyces cerevisiae Kar3MD structure (ScKar3MD), AgKar3MD displays differences in the conformation of some of its nucleotide-binding motifs and peripheral elements. Unlike ScKar3MD, the salt bridge between Switch I and Switch II in AgKar3MD is broken. Most of the Switch I, and the adjoining region of helix α3, are also disordered instead of bending into the active site cleft as is observed in ScKar3MD. These aspects of AgKar3MD are highly reminiscent of the ScKar3 R598A mutant that disrupts the Switch I-Switch II salt bridge and impairs MT-stimulated ATPase activity of the motor. Subtle differences in the disposition of secondary structure elements in the small lobe (ß1a, ß1b, and ß1c) at the edge of the MD are also apparent even though it contains approximately the same number of residues as ScKar3. These differences may reflect the unique enzymatic properties we measured for this motor, which include a lower MT-stimulated ATPase rate relative to ScKar3, or they could relate to its interactions with different regulatory companion proteins than its budding yeast counterpart.


Asunto(s)
Ascomicetos/química , Proteínas Fúngicas/química , Cinesinas/química , Adenosina Trifosfatasas/química , Secuencia de Aminoácidos , Ascomicetos/clasificación , Ascomicetos/enzimología , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X/métodos , Activación Enzimática , Proteínas Fúngicas/clasificación , Proteínas Fúngicas/aislamiento & purificación , Cinesinas/clasificación , Cinesinas/aislamiento & purificación , Datos de Secuencia Molecular , Filogenia , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia , Relación Estructura-Actividad
14.
Biochem Biophys Res Commun ; 428(4): 427-32, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23137538

RESUMEN

In the human fungal pathogen Candida albicans, the Kinesin-14 motor protein Kar3 (CaKar3) is critical for normal mitotic division, nuclear fusion during mating, and morphogenic transition from the commensal yeast form to the virulent hyphal form. As a first step towards detailed characterization of this motor of potential medical significance, we have crystallized and determined the X-ray structure of the motor domain of CaKar3 as a maltose-binding protein (MBP) fusion. The structure shows strong conservation of overall motor domain topology to other Kar3 kinesins, but with some prominent differences in one of the motifs that compose the nucleotide-binding pocket and the surface charge distribution. The MBP and Kar3 modules are arranged such that MBP interacts with the Kar3 motor domain core at the same site where the neck linker of conventional kinesins docks during the "ATP state" of the mechanochemical cycle. This site differs from the Kar3 neck-core interface in the recent structure of the ScKar3Vik1 heterodimer. The position of MBP is also completely distinct from the Vik1 subunit in this complex. This may suggest that the site of MBP interaction on the CaKar3 motor domain provides an interface for the neck, or perhaps a partner subunit, at an intermediate state of its motile cycle that has not yet been observed for Kinesin-14 motors.


Asunto(s)
Candida albicans/metabolismo , Proteínas Fúngicas/química , Cinesinas/química , Proteínas de Unión a Maltosa/química , Proteínas Recombinantes de Fusión/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Proteínas Fúngicas/genética , Humanos , Cinesinas/genética , Proteínas de Unión a Maltosa/genética , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética
15.
Nat Commun ; 13(1): 4198, 2022 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-35859148

RESUMEN

Kinesin-8s are dual-activity motor proteins that can move processively on microtubules and depolymerize microtubule plus-ends, but their mechanism of combining these distinct activities remains unclear. We addressed this by obtaining cryo-EM structures (2.6-3.9 Å) of Candida albicans Kip3 in different catalytic states on the microtubule lattice and on a curved microtubule end mimic. We also determined a crystal structure of microtubule-unbound CaKip3-ADP (2.0 Å) and analyzed the biochemical activity of CaKip3 and kinesin-1 mutants. These data reveal that the microtubule depolymerization activity of kinesin-8 originates from conformational changes of its motor core that are amplified by dynamic contacts between its extended loop-2 and tubulin. On curved microtubule ends, loop-1 inserts into preceding motor domains, forming head-to-tail arrays of kinesin-8s that complement loop-2 contacts with curved tubulin and assist depolymerization. On straight tubulin protofilaments in the microtubule lattice, loop-2-tubulin contacts inhibit conformational changes in the motor core, but in the ADP-Pi state these contacts are relaxed, allowing neck-linker docking for motility. We propose that these tubulin shape-induced alternations between pro-microtubule-depolymerization and pro-motility kinesin states, regulated by loop-2, are the key to the dual activity of kinesin-8 motors.


Asunto(s)
Cinesinas , Tubulina (Proteína) , Adenosina Difosfato/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo
16.
mSphere ; 7(6): e0045622, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36377914

RESUMEN

Fungal hyphal chemotropism has been shown to be a major contributor to host-pathogen interactions. Previous studies on Fusarium species have highlighted the involvement of the Ste2 G-protein-coupled receptor (GPCR) in mediating polarized hyphal growth toward host-released peroxidase. Here, the role of the opposite mating type GPCR, Ste3, is characterized with respect to Fusarium graminearum chemotropism and pathogenicity. Fgste3Δ deletion strains were found to be compromised in the chemotropic response toward peroxidase, development of lesions on germinating wheat, and infection of Arabidopsis thaliana leaves. In the absence of FgSte3 or FgSte2, F. graminearum cells exposed to peroxidase showed no phosphorylation of the cell-wall integrity, mitogen-activated protein kinase pathway component Mgv1. In addition, transcriptomic gene expression profiling yielded a list of genes involved in cellular reorganization, cell wall remodeling, and infection-mediated responses that were differentially modulated by peroxidase when FgSte3 was present. Deletion of FgSte3 yielded the downregulation of genes associated with mycotoxin biosynthesis and appressorium development, compared to the wild-type strain, both in the presence of peroxidase. Together, these findings contribute to our understanding of the mechanism underlying fungal chemotropism and pathogenesis while raising the novel hypothesis that FgSte2 and FgSte3 are interdependent on each other for the mediation of the redirection of hyphal growth in response to host-derived peroxidase. IMPORTANCE Fusarium head blight of wheat, caused by the filamentous fungus Fusarium graminearum, leads to devastating global food shortages and economic losses. Fungal hyphal chemotropism has been shown to be a major contributor to host-pathogen interactions. Here, the role of the opposite mating type GPCR, Ste3, is characterized with respect to F. graminearum chemotropism and pathogenicity. These findings contribute to our understanding of the mechanisms underlying fungal chemotropism and pathogenesis.


Asunto(s)
Fusarium , Fusarium/genética , Peroxidasas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Peroxidasa , Receptores Acoplados a Proteínas G/metabolismo
17.
J Bacteriol ; 193(2): 449-59, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21057010

RESUMEN

The O antigen is the outer part of the lipopolysaccharide (LPS) in the outer membrane of Gram-negative bacteria and contains many repeats of an oligosaccharide unit. It contributes to antigenic variability and is essential to the full function and virulence of bacteria. Shigella is a Gram-negative human pathogen that causes diarrhea in humans. The O antigen of Shigella boydii type 14 consists of repeating oligosaccharide units with the structure [→6-d-Galpα1→4-d-GlcpAß1→6-d-Galpß1→4-d-Galpß1→4-d-GlcpNAcß1→]n. The wfeD gene in the O-antigen gene cluster of Shigella boydii type 14 was proposed to encode a galactosyltransferase (GalT) involved in O-antigen synthesis. We confirmed here that the wfeD gene product is a ß4-GalT that synthesizes the Galß1-4GlcNAcα-R linkage. WfeD was expressed in Escherichia coli, and the activity was characterized by using UDP-[³H]Gal as the donor substrate as well as the synthetic acceptor substrate GlcNAcα-pyrophosphate-(CH2)11-O-phenyl. The enzyme product was analyzed by liquid chromatography-mass spectrometry (LC-MS), high-performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR), and galactosidase digestion. The enzyme was shown to be specific for the UDP-Gal donor substrate and required pyrophosphate in the acceptor substrate. Divalent metal ions such as Mn²(+), Ni²(+), and, surprisingly, also Pb²(+) enhanced the enzyme activity. Mutational analysis showed that the Glu101 residue within a DxD motif is essential for activity, possibly by forming the catalytic nucleophile. The Lys211 residue was also shown to be required for activity and may be involved in the binding of the negatively charged acceptor substrate. Our study revealed that the ß4-GalT WfeD is a novel enzyme that has virtually no sequence similarity to mammalian ß4-GalT, although it catalyzes a similar reaction.


Asunto(s)
Galactosiltransferasas/metabolismo , Antígenos O/metabolismo , Shigella boydii/enzimología , Cationes Bivalentes/metabolismo , Cromatografía Liquida , Análisis Mutacional de ADN , Activadores de Enzimas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Galactosidasas/metabolismo , Expresión Génica , Espectroscopía de Resonancia Magnética , Metales/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Uridina Difosfato Galactosa/metabolismo
18.
Nat Struct Mol Biol ; 12(4): 378-9, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15750603

RESUMEN

Molecular motors play a central role in cytoskeletal-mediated cellular processes and thus present an excellent target for cellular control by pharmacological agents. Yet very few such compounds have been found. We report here the structure of blebbistatin, which inhibits specific myosin isoforms, bound to the motor domain of Dictyostelium discoideum myosin II. This reveals the structural basis for its specificity and provides insight into the development of new agents.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Miosina Tipo II/antagonistas & inhibidores , Miosina Tipo II/química , Animales , Sitios de Unión , Dictyostelium/química , Dictyostelium/metabolismo , Modelos Moleculares , Miosina Tipo II/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Especificidad por Sustrato
19.
Protein Sci ; 29(8): 1707-1723, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32472639

RESUMEN

Kinesins are a diverse group of adenosine triphosphate (ATP)-dependent motor proteins that transport cargos along microtubules (MTs) and change the organization of MT networks. Shared among all kinesins is a ~40 kDa motor domain that has evolved an impressive assortment of motility and MT remodeling mechanisms as a result of subtle tweaks and edits within its sequence. Several elegant studies of different kinesin isoforms have exposed the purpose of structural changes in the motor domain as it engages and leaves the MT. However, few studies have compared the sequences and MT contacts of these kinesins systematically. Along with clever strategies to trap kinesin-tubulin complexes for X-ray crystallography, new advancements in cryo-electron microscopy have produced a burst of high-resolution structures that show kinesin-MT interfaces more precisely than ever. This review considers the MT interactions of kinesin subfamilies that exhibit significant differences in speed, processivity, and MT remodeling activity. We show how their sequence variations relate to their tubulin footprint and, in turn, how this explains the molecular activities of previously characterized mutants. As more high-resolution structures become available, this type of assessment will quicken the pace toward establishing each kinesin's design-function relationship.


Asunto(s)
Cinesinas/química , Cinesinas/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo , Animales , Microscopía por Crioelectrón , Cristalografía por Rayos X , Humanos
20.
Sci Rep ; 10(1): 10770, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612109

RESUMEN

Fusarium Head Blight of wheat, caused by the filamentous fungus Fusarium graminearum, leads to devastating global food shortages and economic losses. While many studies have addressed the responses of both wheat and F. graminearum during their interaction, the possibility of fungal chemotropic sensing enabling pathogenicity remains unexplored. Based on recent findings linking the pheromone-sensing G-protein-coupled receptor Ste2 to host-directed chemotropism in Fusarium oxysporum, we investigated the role of the Ste2 receptor and its downstream signaling pathways in mediating chemotropism of F. graminearum. Interestingly, a chemotropic response of growing hyphae towards catalytically active Triticum aestivum 'Roblin' cultivar secreted peroxidases was detected, with deletion of STE2 in F. graminearum leading to loss of the observed response. At the same time, deletion of STE2 significantly decreased infection on germinating wheat coleoptiles, highlighting an association between Ste2, chemotropism and infection by F. graminearum. Further characterization revealed that the peroxidase-directed chemotropism is associated with stimulation of the fungal cell wall integrity mitogen-activated protein kinase signaling cascade. Altogether, this study demonstrates conservation of Ste2-mediated chemotropism by Fusarium species, and its important role in mediating pathogenicity.


Asunto(s)
Proteínas Fúngicas/metabolismo , Fusarium/metabolismo , Receptores del Factor de Conjugación/metabolismo , Triticum/microbiología , Agrobacterium tumefaciens , Catálisis , Pared Celular/metabolismo , Quimiotaxis , Eliminación de Gen , Hifa/metabolismo , Ligandos , Sistema de Señalización de MAP Quinasas , Peroxidasas/metabolismo , Feromonas/metabolismo , Enfermedades de las Plantas/microbiología , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae , Transducción de Señal , Esporas Fúngicas/metabolismo , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA