Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
MedComm (2020) ; 5(4): e469, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38525108

RESUMEN

Motor proteins, encoded by Kinesin superfamily (KIF) genes, are critical for brain development and plasticity. Increasing studies reported KIF's roles in neurodevelopmental disorders. Here, a 6 years and 3 months-old Chinese boy with markedly symptomatic epilepsy, intellectual disability, brain atrophy, and psychomotor retardation was investigated. His parents and younger sister were phenotypically normal and had no disease-related family history. Whole exome sequencing identified a novel heterozygous in-frame deletion (c.265_267delTCA) in exon 3 of the KIF5C in the proband, resulting in the removal of evolutionarily highly conserved p.Ser90, located in its ATP-binding domain. Sanger sequencing excluded the proband's parents and family members from harboring this variant. The activity of ATP hydrolysis in vitro was significantly reduced as predicted. Immunofluorescence studies showed wild-type KIF5C was widely distributed throughout the cytoplasm, while mutant KIF5C was colocalized with microtubules. The live-cell imaging of the cargo-trafficking assay revealed that mutant KIF5C lost the peroxisome-transporting ability. Drosophila models also confirmed p.Ser90del's essential role in nervous system development. This study emphasized the importance of the KIF5C gene in intracellular cargo-transport as well as germline variants that lead to neurodevelopmental disorders and might enable clinicians for timely and accurate diagnosis and disease management in the future.

2.
EMBO Rep ; 24(11): e56864, 2023 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-37575008

RESUMEN

Kinesin-driven intracellular transport is essential for various cell biological events and thus plays a crucial role in many pathological processes. However, little is known about the molecular basis of the specific and dynamic cargo-binding mechanism of kinesins. Here, an integrated structural analysis of the KIF3/KAP3 and KIF3/KAP3-APC complexes unveils the mechanism by which KIF3/KAP3 can dynamically grasp APC in a two-step manner, which suggests kinesin-cargo recognition dynamics composed of cargo loading, locking, and release. Our finding is the first demonstration of the two-step cargo recognition and stabilization mechanism of kinesins, which provides novel insights into the intracellular trafficking machinery.


Asunto(s)
Comunicación Celular , Cinesinas , Cinesinas/metabolismo , Transporte Biológico , Microtúbulos/metabolismo
3.
Dev Cell ; 58(16): 1447-1461.e6, 2023 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-37413993

RESUMEN

Left-dominant [Ca2+]i elevation on the left margin of the ventral node furnishes the initial laterality of mouse embryos. It depends on extracellular leftward fluid flow (nodal flow), fibroblast growth factor receptor (FGFR)/sonic hedgehog (Shh) signaling, and the PKD1L1 polycystin subunit, of which interrelationship is still elusive. Here, we show that leftward nodal flow directs PKD1L1-containing fibrous strands and facilitates Nodal-mediated [Ca2+]i elevation on the left margin. We generate KikGR-PKD1L1 knockin mice in order to monitor protein dynamics with a photoconvertible fluorescence protein tag. By imaging those embryos, we have identified fragile meshwork being gradually transferred leftward involving pleiomorphic extracellular events. A portion of the meshwork finally bridges over the left nodal crown cells in an FGFR/Shh-dependent manner. As PKD1L1 N-term is predominantly associated with Nodal on the left margin and that PKD1L1/PKD2 overexpression significantly augments cellular Nodal sensitivity, we propose that leftward transfer of polycystin-containing fibrous strands determines left-right asymmetry in developing embryos.


Asunto(s)
Proteínas Hedgehog , Canales Catiónicos TRPP , Ratones , Animales , Proteínas Hedgehog/metabolismo , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo , Cilios/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Proteína Nodal/metabolismo
4.
J Cell Biol ; 222(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36482480

RESUMEN

Epilepsy is a common neurological disease worldwide, and one of its causes is genetic abnormalities. Here, we identified a point mutation in KIF4A, a member of kinesin superfamily molecular motors, in patients with neurological disorders such as epilepsy, developmental delay, and intellectual disability. KIF4 is involved in the poly (ADP-ribose) polymerase (PARP) signaling pathway, and the mutation (R728Q) strengthened its affinity with PARP1 through elongation of the KIF4 coiled-coil domain. Behavioral tests showed that KIF4-mutant mice exhibited mild developmental delay with lower seizure threshold. Further experiments revealed that the KIF4 mutation caused aberrant morphology in dendrites and spines of hippocampal pyramidal neurons through PARP1-TrkB-KCC2 pathway. Furthermore, supplementing NAD, which activates PARP1, could modulate the TrkB-KCC2 pathway and rescue the seizure susceptibility phenotype of the mutant mice. Therefore, these findings indicate that KIF4 is engaged in a fundamental mechanism regulating seizure susceptibility and could be a potential target for epilepsy treatment.


Asunto(s)
Epilepsia , Convulsiones , Ratones , Animales , Convulsiones/genética , Transducción de Señal , Cinesinas/genética
5.
Dev Cell ; 57(19): 2273-2289.e11, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36220081

RESUMEN

Digit determination in limb buds is driven by a posteriorizing Sonic hedgehog (Shh) protein gradient; however, the mechanism regulating this is unclear. Here, we propose a diffusion-and-trapping hypothesis for Shh gradient formation based on data from the preaxial polydactyly phenotype of KIF3B motor hypomorphic mice. In the limb buds of these mice, a distal-to-proximal gradient of fibroblast growth factor (FGF) and phosphatidylinositol 3-kinase (PI3K) signaling and a posterior-to-anterior gradient of Shh were disorganized. This phenotype was reproduced by transplanting FGF8b-soaked beads. At the subcellular level, KIF3B transported the phosphatase and tensin homolog (PTEN)-like phosphatase Talpid3 to terminate PI3K signaling. High and low PI3K signaling strengths differentially sorted endocytosed Shh toward exosome-like particles and cytonemal punctata, respectively. These results indicate that the Shh-containing particles undergo either the diffusional movement in the periphery or cytonemal trapping in the center and form a spatial gradient along the periphery of developing limb buds.


Asunto(s)
Proteínas Hedgehog , Polidactilia , Animales , Extremidades , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Cinesinas , Esbozos de los Miembros/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/genética , Monoéster Fosfórico Hidrolasas/genética , Polidactilia/genética , Polidactilia/metabolismo , Tensinas/genética , Tensinas/metabolismo
6.
EMBO J ; 41(5): e108899, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35132656

RESUMEN

The mechanochemical coupling of ATPase hydrolysis and conformational dynamics in kinesin motors facilitates intramolecular interaction cycles between the kinesin motor and neck domains, which are essential for microtubule-based motility. Here, we characterized a charge-inverting KIF1A-E239K mutant that we identified in a family with axonal-type Charcot-Marie-Tooth disease and also in 24 cases in human neuropathies including spastic paraplegia and hereditary sensory and autonomic neuropathy. We show that Glu239 in the ß7 strand is a key residue of the motor domain that regulates the motor-neck interaction. Expression of the KIF1A-E239K mutation has decreased ability to complement Kif1a+/- neurons, and significantly decreases ATPase activity and microtubule gliding velocity. X-ray crystallography shows that this mutation causes an excess positive charge on ß7, which may electrostatically interact with a negative charge on the neck. Quantitative mass spectrometric analysis supports that the mutation hyper-stabilizes the motor-neck interaction at the late ATP hydrolysis stage. Thus, the negative charge of Glu239 dynamically regulates the kinesin motor-neck interaction, promoting release of the neck from the motor domain upon ATP hydrolysis.


Asunto(s)
Adenosina Trifosfatasas/genética , Cinesinas/genética , Mutación/genética , Neuronas/fisiología , Anciano , Secuencia de Aminoácidos , Axones/fisiología , Enfermedad de Charcot-Marie-Tooth , Humanos , Masculino , Microtúbulos/genética , Persona de Mediana Edad , Alineación de Secuencia
7.
Cell Rep ; 35(2): 108971, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852848

RESUMEN

In schizophrenia (SCZ), neurons in the brain tend to undergo gross morphological changes, but the related molecular mechanism remains largely elusive. Using Kif3b+/- mice as a model with SCZ-like behaviors, we found that a high-betaine diet can significantly alleviate schizophrenic traits related to neuronal morphogenesis and behaviors. According to a deficiency in the transport of collapsin response mediator protein 2 (CRMP2) by the KIF3 motor, we identified a significant reduction in lamellipodial dynamics in developing Kif3b+/- neurons as a cause of neurite hyperbranching. Betaine administration significantly decreases CRMP2 carbonylation, which enhances the F-actin bundling needed for proper lamellipodial dynamics and microtubule exclusion and may thus functionally compensate for KIF3 deficiency. Because the KIF3 expression levels tend to be downregulated in the human prefrontal cortex of the postmortem brains of SCZ patients, this mechanism may partly participate in human SCZ pathogenesis, which we hypothesize could be alleviated by betaine administration.


Asunto(s)
Betaína/farmacología , Péptidos y Proteínas de Señalización Intercelular/genética , Cinesinas/genética , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Seudópodos/efectos de los fármacos , Esquizofrenia/dietoterapia , Actinas/genética , Actinas/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Transporte Biológico , Dieta/métodos , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Cinesinas/deficiencia , Masculino , Ratones , Ratones Noqueados , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Proteínas del Tejido Nervioso/deficiencia , Neuronas/metabolismo , Neuronas/ultraestructura , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Unión Proteica , Carbonilación Proteica , Seudópodos/metabolismo , Seudópodos/ultraestructura , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patología
8.
Sci Adv ; 6(51)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33328231

RESUMEN

Synaptic weight changes among postsynaptic densities within a single dendrite are regulated by the balance between localized protein degradation and synthesis. However, the molecular mechanism via these opposing regulatory processes is still elusive. Here, we showed that the molecular motor KIF17 was locally degraded and synthesized in an N-methyl-d-aspartate receptor (NMDAR)-mediated activity-dependent manner. Accompanied by the degradation of KIF17, its transport was temporarily dampened in dendrites. We also observed that activity-dependent local KIF17 synthesis driven by its 3' untranslated region (3'UTR) occurred at dendritic shafts, and the newly synthesized KIF17 moved along the dendrites. Furthermore, hippocampus-specific deletion of Kif17 3'UTR disrupted KIF17 synthesis induced by fear memory retrieval, leading to impairment in extinction of fear memory. These results indicate that the regulation of the KIF17 transport is driven by the single dendrite-restricted cycle of degradation and synthesis that underlies cognitive flexibility.

9.
EMBO J ; 39(1): e101090, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31746486

RESUMEN

The transport of N-methyl-d-aspartate receptors (NMDARs) is crucial for neuronal plasticity and synapse formation. Here, we show that KIF3B, a member of the kinesin superfamily proteins (KIFs), supports the transport of vesicles simultaneously containing NMDAR subunit 2A (NR2A) and the adenomatous polyposis coli (APC) complex. Kif3b+/- neurons exhibited a reduction in dendritic levels of both NR2A and NR2B due to the impaired transport of NR2A and increased degradation of NR2B. In Kif3b+/- hippocampal slices, electrophysiological NMDAR response was found decreased and synaptic plasticity was disrupted, which corresponded to a common feature of schizophrenia (SCZ). The histological features of Kif3b+/- mouse brain also mimicked SCZ features, and Kif3b+/- mice exhibited behavioral defects in prepulse inhibition (PPI), social interest, and cognitive flexibility. Indeed, a mutation of KIF3B was specifically identified in human SCZ patients, which was revealed to be functionally defective in a rescue experiment. Therefore, we propose that KIF3B transports NR2A/APC complex and that its dysfunction is responsible for SCZ pathogenesis.


Asunto(s)
Cinesinas/genética , Cinesinas/fisiología , Mutación , Neuronas/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Esquizofrenia/etiología , Sinapsis/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Conducta Animal , Movimiento Celular , Humanos , Relaciones Interpersonales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Fenotipo , Subunidades de Proteína , Transporte de Proteínas , Esquizofrenia/metabolismo , Esquizofrenia/patología , Sinapsis/metabolismo
10.
Cell Rep ; 29(5): 1261-1273.e6, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31665638

RESUMEN

Acute kidney injury (AKI) is characterized by mitochondrial dysfunction and activation of the innate immune system. The cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway detects cytosolic DNA and induces innate immunity. Here, we investigate the role of mitochondrial damage and subsequent activation of the cGAS-STING pathway using a genetically engineered animal model of cisplatin-induced AKI and cultured tubular cells. Cisplatin induced mtDNA leakage into the cytosol-probably through BCL-2-like protein 4 (BAX) pores in the mitochondrial outer membrane-in tubules, with subsequent activation of the cGAS-STING pathway, thereby triggering inflammation and AKI progression, which is improved in STING-deficient mice. STING knockdown in cultured tubular cells ameliorates inflammatory responses induced by cisplatin. mtDNA depletion and repletion studies support tubular inflammatory responses via the cGAS-STING signal activation by cytosolic mtDNA. Therefore, we conclude that mitochondrial dysfunction and subsequent activation of the mtDNA-cGAS-STING pathway is a critical regulator of kidney injury.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Inflamación/patología , Proteínas de la Membrana/metabolismo , Mitocondrias/patología , Nucleotidiltransferasas/metabolismo , Lesión Renal Aguda/inducido químicamente , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Cisplatino/efectos adversos , Citosol/metabolismo , ADN Mitocondrial/metabolismo , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína X Asociada a bcl-2/metabolismo
11.
Life Sci Alliance ; 2(5)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31591136

RESUMEN

Enhanced carbonyl stress underlies a subset of schizophrenia, but its causal effects remain elusive. Here, we elucidated the molecular mechanism underlying the effects of carbonyl stress in iPS cells in which the gene encoding zinc metalloenzyme glyoxalase I (GLO1), a crucial enzyme for the clearance of carbonyl stress, was disrupted. The iPS cells exhibited significant cellular and developmental deficits, and hyper-carbonylation of collapsing response mediator protein 2 (CRMP2). Structural and biochemical analyses revealed an array of multiple carbonylation sites in the functional motifs of CRMP2, particularly D-hook (for dimerization) and T-site (for tetramerization), which are critical for the activity of the CRMP2 tetramer. Interestingly, carbonylated CRMP2 was stacked in the multimer conformation by irreversible cross-linking, resulting in loss of its unique function to bundle microtubules. Thus, the present study revealed that the enhanced carbonyl stress stemmed from the genetic aberrations results in neurodevelopmental deficits through the formation of irreversible dysfunctional multimer of carbonylated CRMP2.


Asunto(s)
Mutación del Sistema de Lectura , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lactoilglutatión Liasa/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Esquizofrenia/genética , Diferenciación Celular , Células Cultivadas , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Espectrometría de Masas , Modelos Moleculares , Carbonilación Proteica , Conformación Proteica , Multimerización de Proteína , Esquizofrenia/metabolismo
12.
EMBO Mol Med ; 11(12): e10695, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31657521

RESUMEN

Mice with the C3H background show greater behavioral propensity for schizophrenia, including lower prepulse inhibition (PPI), than C57BL/6 (B6) mice. To characterize as-yet-unknown pathophysiologies of schizophrenia, we undertook proteomics analysis of the brain in these strains, and detected elevated levels of Mpst, a hydrogen sulfide (H2 S)/polysulfide-producing enzyme, and greater sulfide deposition in C3H than B6 mice. Mpst-deficient mice exhibited improved PPI with reduced storage sulfide levels, while Mpst-transgenic (Tg) mice showed deteriorated PPI, suggesting that "sulfide stress" may be linked to PPI impairment. Analysis of human samples demonstrated that the H2 S/polysulfides production system is upregulated in schizophrenia. Mechanistically, the Mpst-Tg brain revealed dampened energy metabolism, while maternal immune activation model mice showed upregulation of genes for H2 S/polysulfides production along with typical antioxidative genes, partly via epigenetic modifications. These results suggest that inflammatory/oxidative insults in early brain development result in upregulated H2 S/polysulfides production as an antioxidative response, which in turn cause deficits in bioenergetic processes. Collectively, this study presents a novel aspect of the neurodevelopmental theory for schizophrenia, unraveling a role of excess H2 S/polysulfides production.


Asunto(s)
Sulfuro de Hidrógeno/metabolismo , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Sulfuros/metabolismo , Animales , Electroforesis en Gel Bidimensional , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Epigenómica , Masculino , Ratones , Proteómica , Esquizofrenia/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Cell Rep ; 28(9): 2413-2426.e7, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31461655

RESUMEN

The axon initial segment (AIS) is a compartment that serves as a molecular barrier to achieve axon-dendrite differentiation. Distribution of specific proteins during early neuronal development has been proposed to be critical for AIS construction. However, it remains unknown how these proteins are specifically targeted to the proximal axon within this limited time period. Here, we reveal spatiotemporal regulation driven by the microtubule (MT)-based motor KIF3A/B/KAP3 that transports TRIM46, influenced by a specific MARK2 phosphorylation cascade. In the proximal part of the future axon under low MARK2 activity, the KIF3/KAP3 motor recognizes TRIM46 as cargo and transports it to the future AIS. In contrast, in the somatodendritic area under high MARK2 activity, KAP3 phosphorylated at serine 60 by MARK2 cannot bind with TRIM46 and be transported. This spatiotemporal regulation between KIF3/KAP3 and TRIM46 under specific MARK2 activity underlies the specific transport needed for axonal differentiation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transporte Axonal , Axones/metabolismo , Proteínas del Citoesqueleto/metabolismo , Cinesinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Animales , Células COS , Chlorocebus aethiops , Femenino , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Proteína Quinasa 1 Activada por Mitógenos/metabolismo
14.
J Cell Biol ; 217(12): 4164-4183, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30297389

RESUMEN

Kinesin-1, the founding member of the kinesin superfamily of proteins, is known to use only a subset of microtubules for transport in living cells. This biased use of microtubules is proposed as the guidance cue for polarized transport in neurons, but the underlying mechanisms are still poorly understood. Here, we report that kinesin-1 binding changes the microtubule lattice and promotes further kinesin-1 binding. This high-affinity state requires the binding of kinesin-1 in the nucleotide-free state. Microtubules return to the initial low-affinity state by washing out the binding kinesin-1 or by the binding of non-hydrolyzable ATP analogue AMPPNP to kinesin-1. X-ray fiber diffraction, fluorescence speckle microscopy, and second-harmonic generation microscopy, as well as cryo-EM, collectively demonstrated that the binding of nucleotide-free kinesin-1 to GDP microtubules changes the conformation of the GDP microtubule to a conformation resembling the GTP microtubule.


Asunto(s)
Cinesinas , Microtúbulos , Adenilil Imidodifosfato/química , Adenilil Imidodifosfato/farmacología , Animales , Transporte Biológico Activo , Chlorocebus aethiops , Perros , Guanosina Difosfato/química , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Células HeLa , Humanos , Cinesinas/química , Cinesinas/metabolismo , Células de Riñón Canino Madin Darby , Microtúbulos/química , Microtúbulos/metabolismo , Células Vero
15.
Cell Rep ; 24(11): 2894-2907, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30208315

RESUMEN

Kinesin superfamily proteins (KIFs) are molecular motors that typically alter the subcellular localization of their cargos. However, the atypical kinesin KIF26A does not serve as a motor but can bind microtubules and affect cellular signaling cascades. Here, we show that KIF26A maintains intracellular calcium homeostasis and negatively regulates nociceptive sensation. Kif26a-/- mice exhibit intense and prolonged nociceptive responses. In their primary sensory neurons, excessive inhibitory phosphorylation of plasma membrane Ca2+ ATPase (PMCA) mediated by focal adhesion kinase (FAK) rendered the Ca transients resistant to termination, and the peripheral axonal outgrowth was significantly enhanced. Upstream, KIF26A is directly associated with a FERM domain of FAK and antagonizes FAK function in integrin-Src family kinase (SFK)-FAK signaling, possibly through steric hindrance and localization to cytoplasmic microtubules.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Cinesinas/metabolismo , Animales , Axones/metabolismo , Calcio/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Hiperalgesia/metabolismo , Cinesinas/genética , Masculino , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Nociceptores/metabolismo , Nervios Periféricos/citología , Fosforilación , Transducción de Señal/genética , Transducción de Señal/fisiología
16.
J Cell Biol ; 217(10): 3480-3496, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30126838

RESUMEN

KIF1Bß is a kinesin-3 family anterograde motor protein essential for neuronal development, viability, and function. KIF1Bß mutations have previously been reported in a limited number of pedigrees of Charcot-Marie-Tooth disease type 2A (CMT2A) neuropathy. However, the gene responsible for CMT2A is still controversial, and the mechanism of pathogenesis remains elusive. In this study, we show that the receptor tyrosine kinase IGF1R is a new direct binding partner of KIF1Bß, and its binding and transport is specifically impaired by the Y1087C mutation of KIF1Bß, which we detected in hereditary neuropathic patients. The axonal outgrowth and IGF-I signaling of Kif1b-/- neurons were significantly impaired, consistent with decreased surface IGF1R expression. The complementary capacity of KIF1Bß-Y1087C of these phenotypes was significantly impaired, but the binding capacity to synaptic vesicle precursors was not affected. These data have supported the relevance of KIF1Bß in IGF1R transport, which may give new clue to the neuropathic pathogenesis.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth/embriología , Regulación del Desarrollo de la Expresión Génica , Cinesinas/metabolismo , Mutación Missense , Proyección Neuronal , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Animales , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/patología , Cinesinas/genética , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Transporte de Proteínas/genética , Receptor IGF Tipo 1/genética
17.
Cell Rep ; 23(13): 3864-3877, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29949770

RESUMEN

Fear extinction is a component of cognitive flexibility that is relevant for important psychiatric diseases, but its molecular mechanism is still largely elusive. We established mice lacking the kinesin-4 motor KIF21B as a model for fear extinction defects. Postsynaptic NMDAR-dependent long-term depression (LTD) is specifically impaired in knockouts. NMDAR-mediated LTD-causing stimuli induce dynamic association of KIF21B with the Rac1GEF subunit engulfment and cell motility protein 1 (ELMO1), leading to ELMO1 translocation out of dendritic spines and its sequestration in endosomes. This process may essentially terminate transient activation of Rac1, shrink spines, facilitate AMPAR endocytosis, and reduce postsynaptic strength, thereby forming a mechanistic link to LTD expression. Antagonizing ELMO1/Dock Rac1GEF activity by the administration of 4-[3'-(2″-chlorophenyl)-2'-propen-1'-ylidene]-1-phenyl-3,5-pyrazolidinedione (CPYPP) significantly reverses the knockout phenotype. Therefore, we propose that KIF21B-mediated Rac1 inactivation is a key molecular event in NMDAR-dependent LTD expression underlying cognitive flexibility in fear extinction.


Asunto(s)
Miedo/fisiología , Cinesinas/metabolismo , Plasticidad Neuronal , Neuropéptidos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Espinas Dendríticas/metabolismo , Endocitosis , Endosomas/metabolismo , Guanosina Trifosfato/metabolismo , Cinesinas/deficiencia , Cinesinas/genética , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal/efectos de los fármacos , Neuropéptidos/antagonistas & inhibidores , Unión Proteica , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Pirazoles/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores
18.
Elife ; 72018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29313800

RESUMEN

Kinesin super family protein 2A (KIF2A), an ATP-dependent microtubule (MT) destabilizer, regulates cell migration, axon elongation, and pruning in the developing nervous system. KIF2A mutations have recently been identified in patients with malformed cortical development. However, postnatal KIF2A is continuously expressed in the hippocampus, in which new neurons are generated throughout an individual's life in established neuronal circuits. In this study, we investigated KIF2A function in the postnatal hippocampus by using tamoxifen-inducible Kif2a conditional knockout (Kif2a-cKO) mice. Despite exhibiting no significant defects in neuronal proliferation or migration, Kif2a-cKO mice showed signs of an epileptic hippocampus. In addition to mossy fiber sprouting, the Kif2a-cKO dentate granule cells (DGCs) showed dendro-axonal conversion, leading to the growth of many aberrant overextended dendrites that eventually developed axonal properties. These results suggested that postnatal KIF2A is a key length regulator of DGC developing neurites and is involved in the establishment of precise postnatal hippocampal wiring.


Asunto(s)
Hipocampo/citología , Cinesinas/metabolismo , Neuronas/citología , Proteínas Represoras/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Hipocampo/patología , Cinesinas/genética , Ratones , Ratones Noqueados , Proteínas Represoras/genética
19.
Biophys Rev ; 10(2): 299-306, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29204883

RESUMEN

The need for accurate description of protein behavior in solution has gained importance in various fields, including biophysics, biochemistry, structural biology, drug discovery, and antibody drugs. To achieve the desired accuracy, multiple precise analyses should be performed on the target molecule, compared, and effectively combined. This review focuses on the combination of multiple analyses in solution: size-exclusion chromatography (SEC), multi-angle light scattering (MALS), small-angle X-ray scattering (SAXS), analytical ultracentrifugation (AUC), and their complementary methods, such as atomic force microscopy (AFM) and mass spectrometry (MS). We also discuss the comparison between the determined molar mass value of not only the standard proteins, but of a target molecule tubulin and its depolymerizing protein, KIF2, as an example. The comparison of the estimated molar mass value from the different methods provides additional information about the target molecule, because the value reflects the dynamically changing states of the target molecule in solution. The combination and integration of multiple methods will permit a deeper understanding of protein dynamics in solution.

20.
Sci Rep ; 7(1): 10681, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28878401

RESUMEN

Microtubule associated protein Collapsin response mediator protein 2 (CRMP2) regulates neuronal polarity in developing neurons through interactions with tubulins or microtubules. However, how CRMP2 promotes axonal formation by affecting microtubule behavior remains unknown. This study aimed to obtain the structural basis for CRMP2-tubulin/microtubule interaction in the course of axonogenesis. The X-ray structural studies indicated that the main interface to the soluble tubulin-dimer is the last helix H19 of CRMP2 that is distinct from the known C-terminal tail-mediated interaction with assembled microtubules. In vitro structural and functional studies also suggested that the H19-mediated interaction promoted the rapid formation of GTP-state microtubules directly, which is an important feature of the axon. Consistently, the H19 mutants disturbed axon elongation in chick neurons, and failed to authorize the structural features for axonal microtubules in Caenorhabditis elegans. Thus, CRMP2 induces effective axonal microtubule formation through H19-mediated interactions with a soluble tubulin-dimer allowing axonogenesis to proceed.


Asunto(s)
Axones/metabolismo , Péptidos y Proteínas de Señalización Intercelular/química , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Microtúbulos/química , Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Eliminación de Gen , Guanosina Trifosfato/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Modelos Moleculares , Mutación , Proteínas del Tejido Nervioso/genética , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Soluciones , Relación Estructura-Actividad , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...