Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Nature ; 590(7846): 492-497, 2021 02.
Article in English | MEDLINE | ID: mdl-33505027

ABSTRACT

Whole-genome doubling (WGD) is common in human cancers, occurring early in tumorigenesis and generating genetically unstable tetraploid cells that fuel tumour development1,2. Cells that undergo WGD (WGD+ cells) must adapt to accommodate their abnormal tetraploid state; however, the nature of these adaptations, and whether they confer vulnerabilities that can be exploited therapeutically, is unclear. Here, using sequencing data from roughly 10,000 primary human cancer samples and essentiality data from approximately 600 cancer cell lines, we show that WGD gives rise to common genetic traits that are accompanied by unique vulnerabilities. We reveal that WGD+ cells are more dependent than WGD- cells on signalling from the spindle-assembly checkpoint, DNA-replication factors and proteasome function. We also identify KIF18A, which encodes a mitotic kinesin protein, as being specifically required for the viability of WGD+ cells. Although KIF18A is largely dispensable for accurate chromosome segregation during mitosis in WGD- cells, its loss induces notable mitotic errors in WGD+ cells, ultimately impairing cell viability. Collectively, our results suggest new strategies for specifically targeting WGD+ cancer cells while sparing the normal, non-transformed WGD- cells that comprise human tissue.


Subject(s)
Genome, Human/genetics , Mitosis/drug effects , Neoplasms/genetics , Neoplasms/pathology , Tetraploidy , Abnormal Karyotype/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, Lethal/genetics , Humans , Kinesins/deficiency , Kinesins/genetics , Kinesins/metabolism , M Phase Cell Cycle Checkpoints/drug effects , Male , Mitosis/genetics , Proteasome Endopeptidase Complex/metabolism , Reproducibility of Results , Spindle Apparatus/drug effects
2.
Nature ; 590(7846): 486-491, 2021 02.
Article in English | MEDLINE | ID: mdl-33505028

ABSTRACT

Selective targeting of aneuploid cells is an attractive strategy for cancer treatment1. However, it is unclear whether aneuploidy generates any clinically relevant vulnerabilities in cancer cells. Here we mapped the aneuploidy landscapes of about 1,000 human cancer cell lines, and analysed genetic and chemical perturbation screens2-9 to identify cellular vulnerabilities associated with aneuploidy. We found that aneuploid cancer cells show increased sensitivity to genetic perturbation of core components of the spindle assembly checkpoint (SAC), which ensures the proper segregation of chromosomes during mitosis10. Unexpectedly, we also found that aneuploid cancer cells were less sensitive than diploid cells to short-term exposure to multiple SAC inhibitors. Indeed, aneuploid cancer cells became increasingly sensitive to inhibition of SAC over time. Aneuploid cells exhibited aberrant spindle geometry and dynamics, and kept dividing when the SAC was inhibited, resulting in the accumulation of mitotic defects, and in unstable and less-fit karyotypes. Therefore, although aneuploid cancer cells could overcome inhibition of SAC more readily than diploid cells, their long-term proliferation was jeopardized. We identified a specific mitotic kinesin, KIF18A, whose activity was perturbed in aneuploid cancer cells. Aneuploid cancer cells were particularly vulnerable to depletion of KIF18A, and KIF18A overexpression restored their response to SAC inhibition. Our results identify a therapeutically relevant, synthetic lethal interaction between aneuploidy and the SAC.


Subject(s)
Aneuploidy , M Phase Cell Cycle Checkpoints/drug effects , Neoplasms/pathology , Abnormal Karyotype/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromosome Segregation/drug effects , Diploidy , Genes, Lethal , Humans , Kinesins/deficiency , Kinesins/genetics , Kinesins/metabolism , Neoplasms/genetics , Spindle Apparatus/drug effects , Synthetic Lethal Mutations/drug effects , Synthetic Lethal Mutations/genetics , Time Factors
3.
Mol Cell ; 75(1): 26-38.e3, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31130364

ABSTRACT

Growth factor signaling is initiated at the plasma membrane and propagated through the cytoplasm for eventual relay to intracellular organelles such as lysosomes. The serine/threonine kinase mTOR participates in growth factor signaling as a component of two multi-subunit complexes, mTORC1 and mTORC2. mTORC1 associates with lysosomes, and its activity depends on the positioning of lysosomes within the cytoplasm, although there is no consensus regarding the exact effect of perinuclear versus peripheral distribution. mTORC2 and its substrate kinase AKT have a widespread distribution, but they are thought to act mainly at the plasma membrane. Using cell lines with knockout of components of the lysosome-positioning machinery, we show that perinuclear clustering of lysosomes delays reactivation of not only mTORC1, but also mTORC2 and AKT upon serum replenishment. These experiments demonstrate the existence of pools of mTORC2 and AKT that are sensitive to lysosome positioning.


Subject(s)
Cell Nucleus/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Proto-Oncogene Proteins c-akt/genetics , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , CRISPR-Cas Systems , Cell Nucleus/ultrastructure , Culture Media, Serum-Free , Endosomes/metabolism , Endosomes/ultrastructure , Gene Editing , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Kinesins/deficiency , Kinesins/genetics , Lysosomes/ultrastructure , MEF2 Transcription Factors/deficiency , MEF2 Transcription Factors/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
4.
PLoS Genet ; 14(11): e1007817, 2018 11.
Article in English | MEDLINE | ID: mdl-30475797

ABSTRACT

Cerebrospinal fluid flow is crucial for neurodevelopment and homeostasis of the ventricular system of the brain, with localized flow being established by the polarized beating of the ependymal cell (EC) cilia. Here, we report a homozygous one base-pair deletion, c.1193delT (p.Leu398Glnfs*2), in the Kinesin Family Member 6 (KIF6) gene in a child displaying neurodevelopmental defects and intellectual disability. To test the pathogenicity of this novel human KIF6 mutation we engineered an analogous C-terminal truncating mutation in mouse. These mutant mice display severe, postnatal-onset hydrocephalus. We generated a Kif6-LacZ transgenic mouse strain and report expression specifically and uniquely within the ependymal cells (ECs) of the brain, without labeling other multiciliated mouse tissues. Analysis of Kif6 mutant mice with scanning electron microscopy (SEM) and immunofluorescence (IF) revealed specific defects in the formation of EC cilia, without obvious effect of cilia of other multiciliated tissues. Dilation of the ventricular system and defects in the formation of EC cilia were also observed in adult kif6 mutant zebrafish. Finally, we report Kif6-GFP localization at the axoneme and basal bodies of multi-ciliated cells (MCCs) of the mucociliary Xenopus epidermis. Overall, this work describes the first clinically-defined KIF6 homozygous null mutation in human and defines KIF6 as a conserved mediator of neurological development with a specific role for EC ciliogenesis in vertebrates.


Subject(s)
Ependyma/abnormalities , Kinesins/genetics , Mutation , Neurodevelopmental Disorders/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Child , Cilia/metabolism , Cilia/pathology , Consanguinity , Ependyma/metabolism , Female , Gene Expression , Homozygote , Humans , Hydrocephalus/genetics , Intellectual Disability/genetics , Kinesins/deficiency , Kinesins/metabolism , Kinesins/physiology , Male , Mice , Mice, Transgenic , Models, Animal , Neurodevelopmental Disorders/metabolism , Neurodevelopmental Disorders/pathology , Pedigree , Sequence Deletion , Tissue Distribution , Xenopus laevis , Zebrafish
5.
Circulation ; 139(20): 2342-2357, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30818997

ABSTRACT

BACKGROUND: The primary cilium is a singular cellular structure that extends from the surface of many cell types and plays crucial roles in vertebrate development, including that of the heart. Whereas ciliated cells have been described in developing heart, a role for primary cilia in adult heart has not been reported. This, coupled with the fact that mutations in genes coding for multiple ciliary proteins underlie polycystic kidney disease, a disorder with numerous cardiovascular manifestations, prompted us to identify cells in adult heart harboring a primary cilium and to determine whether primary cilia play a role in disease-related remodeling. METHODS: Histological analysis of cardiac tissues from C57BL/6 mouse embryos, neonatal mice, and adult mice was performed to evaluate for primary cilia. Three injury models (apical resection, ischemia/reperfusion, and myocardial infarction) were used to identify the location and cell type of ciliated cells with the use of antibodies specific for cilia (acetylated tubulin, γ-tubulin, polycystin [PC] 1, PC2, and KIF3A), fibroblasts (vimentin, α-smooth muscle actin, and fibroblast-specific protein-1), and cardiomyocytes (α-actinin and troponin I). A similar approach was used to assess for primary cilia in infarcted human myocardial tissue. We studied mice silenced exclusively in myofibroblasts for PC1 and evaluated the role of PC1 in fibrogenesis in adult rat fibroblasts and myofibroblasts. RESULTS: We identified primary cilia in mouse, rat, and human heart, specifically and exclusively in cardiac fibroblasts. Ciliated fibroblasts are enriched in areas of myocardial injury. Transforming growth factor ß-1 signaling and SMAD3 activation were impaired in fibroblasts depleted of the primary cilium. Extracellular matrix protein levels and contractile function were also impaired. In vivo, depletion of PC1 in activated fibroblasts after myocardial infarction impaired the remodeling response. CONCLUSIONS: Fibroblasts in the neonatal and adult heart harbor a primary cilium. This organelle and its requisite signaling protein, PC1, are required for critical elements of fibrogenesis, including transforming growth factor ß-1-SMAD3 activation, production of extracellular matrix proteins, and cell contractility. Together, these findings point to a pivotal role of this organelle, and PC1, in disease-related pathological cardiac remodeling and suggest that some of the cardiovascular manifestations of autosomal dominant polycystic kidney disease derive directly from myocardium-autonomous abnormalities.


Subject(s)
Fibroblasts/ultrastructure , Myocardium/pathology , Polycystic Kidney, Autosomal Dominant/pathology , 3T3 Cells/ultrastructure , Animals , Animals, Newborn , Atrial Remodeling , Cilia , Fetal Heart/cytology , Fibrosis , Heart Injuries/pathology , Humans , Kinesins/deficiency , Kinesins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Rats , Signal Transduction , Smad3 Protein/physiology , TRPP Cation Channels/deficiency , TRPP Cation Channels/physiology , Transforming Growth Factor beta1/physiology , Ventricular Remodeling
6.
Arterioscler Thromb Vasc Biol ; 38(5): 1037-1051, 2018 05.
Article in English | MEDLINE | ID: mdl-29519941

ABSTRACT

OBJECTIVE: Platelet secretion is crucial for many physiological platelet responses. Even though several regulators of the fusion machinery for secretory granule exocytosis have been identified in platelets, the underlying mechanisms are not yet fully characterized. APPROACH AND RESULTS: By studying a mouse model (cKO [conditional knockout]Kif5b) lacking Kif5b (kinesin-1 heavy chain) in its megakaryocytes and platelets, we evidenced unstable hemostasis characterized by an increase of blood loss associated to a marked tendency to rebleed in a tail-clip assay and thrombus instability in an in vivo thrombosis model. This instability was confirmed in vitro in a whole-blood perfusion assay under blood flow conditions. Aggregations induced by thrombin and collagen were also impaired in cKOKif5b platelets. Furthermore, P-selectin exposure, PF4 (platelet factor 4) secretion, and ATP release after thrombin stimulation were impaired in cKOKif5b platelets, highlighting the role of kinesin-1 in α-granule and dense granule secretion. Importantly, exogenous ADP rescued normal thrombin induced-aggregation in cKOKif5b platelets, which indicates that impaired aggregation was because of defective release of ADP and dense granules. Last, we demonstrated that kinesin-1 interacts with the molecular machinery comprising the granule-associated Rab27 (Ras-related protein Rab-27) protein and the Slp4 (synaptotagmin-like protein 4/SYTL4) adaptor protein. CONCLUSIONS: Our results indicate that a kinesin-1-dependent process plays a role for platelet function by acting into the mechanism underlying α-granule and dense granule secretion.


Subject(s)
Blood Platelets/enzymology , Hemostasis , Kinesins/metabolism , Megakaryocytes/enzymology , Platelet Activation , Secretory Vesicles/enzymology , Thrombosis/enzymology , Adenosine Triphosphate/blood , Animals , Blood Platelets/ultrastructure , Disease Models, Animal , Humans , Kinesins/blood , Kinesins/deficiency , Kinesins/genetics , Megakaryocytes/ultrastructure , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/blood , Platelet Aggregation , Platelet Factor 4/blood , Secretory Pathway , Secretory Vesicles/genetics , Secretory Vesicles/ultrastructure , Signal Transduction , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology , Vesicular Transport Proteins/blood , rab27 GTP-Binding Proteins/blood
7.
Nature ; 484(7392): 120-4, 2012 Mar 18.
Article in English | MEDLINE | ID: mdl-22425998

ABSTRACT

The basic unit of skeletal muscle in all metazoans is the multinucleate myofibre, within which individual nuclei are regularly positioned. The molecular machinery responsible for myonuclear positioning is not known. Improperly positioned nuclei are a hallmark of numerous diseases of muscle, including centronuclear myopathies, but it is unclear whether correct nuclear positioning is necessary for muscle function. Here we identify the microtubule-associated protein ensconsin (Ens)/microtubule-associated protein 7 (MAP7) and kinesin heavy chain (Khc)/Kif5b as essential, evolutionarily conserved regulators of myonuclear positioning in Drosophila and cultured mammalian myotubes. We find that these proteins interact physically and that expression of the Kif5b motor domain fused to the MAP7 microtubule-binding domain rescues nuclear positioning defects in MAP7-depleted cells. This suggests that MAP7 links Kif5b to the microtubule cytoskeleton to promote nuclear positioning. Finally, we show that myonuclear positioning is physiologically important. Drosophila ens mutant larvae have decreased locomotion and incorrect myonuclear positioning, and these phenotypes are rescued by muscle-specific expression of Ens. We conclude that improper nuclear positioning contributes to muscle dysfunction in a cell-autonomous fashion.


Subject(s)
Cell Nucleus/metabolism , Kinesins/metabolism , Microtubule-Associated Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Animals , Cell Compartmentation/genetics , Cell Line , Cell Polarity/genetics , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Kinesins/chemistry , Kinesins/deficiency , Kinesins/genetics , Larva/cytology , Larva/genetics , Larva/metabolism , Locomotion/genetics , Locomotion/physiology , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Organ Specificity , Phenotype , Protein Binding , Protein Structure, Tertiary
8.
Clin Exp Pharmacol Physiol ; 45(10): 1010-1018, 2018 10.
Article in English | MEDLINE | ID: mdl-29781531

ABSTRACT

Gastric cancer accounts for a sizeable proportion of global cancer mortality with high morbidity and poor prognosis. Kinesin superfamily proteins (KIFs) are microtubule-dependent motor proteins that function as oncogenes in cancer cells, it has been discovered in recent years. Kinesin family member 2a (KIF2A), a member of the KIFs, has received attention for its role in carcinogenesis and its prognostic value in several human cancers such as breast cancer, colorectal cancer, and squamous cell carcinoma. However, the role of KIF2A in human gastric cancer remains unknown. In this study we aimed to explore the expression and biological functions of KIF2A in human gastric cancer cells, as well as to reveal its potential action mechanism. First, we found that KIF2A was markedly increased in gastric cancer cells (MKN-28, MKN-45, NCI-N87 and SGC-7901) compared to normal gastric mucosa epithelial cells (GES-1). Then KIF2A was successfully silenced in MKN-45 and SGC-7901 cells to facilitate further research into its function. We discovered that KIF2A silencing can significantly inhibit the growth and invasion of MKN-45 and SGC-7901 cells in a time-independent manner, accompanying a decreased expression of Membrane type 1-matrix metalloproteinase (MT1-MMP). When MT1-MMP was reintroduced into MKN-45 and SGC-7901 cells in the KIF2A-siRNA group, only invasion inhibition effects on MKN-45 and SGC-7901 cells induced by KIF2A silencing can be reversed. In conclusion, our study reveals that down-regulation of KIF2A can inhibit gastric cancer cell invasion by suppressing MT1-MMP.


Subject(s)
Down-Regulation , Kinesins/genetics , Kinesins/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Stomach Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Humans , Kinesins/deficiency , Neoplasm Invasiveness , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism
9.
Cell Struct Funct ; 40(1): 31-41, 2015.
Article in English | MEDLINE | ID: mdl-25748359

ABSTRACT

The NACK kinesins (NACK1, NACK2 in tobacco and AtNACK1/HINKEL, AtNACK2/STUD/TETRASPORE in Arabidopsis), members of a plant-specific kinesin-7 family, are required for cytokinesis. Previous studies using tobacco and Arabidopsis cells showed that NACK1 and AtNACK1 at the phragmoplast midzone activate the MAP kinase cascade during the late M phase, which is critical for the cell plate formation. However, the loss-of-function phenotype has not been investigated in details in living cells and the molecular activity of this kinesin remains to be determined. Here, we report the mitotic roles and activity of the NACK kinesins in the moss Physcomitrella patens. When we simultaneously knocked down three PpNACKs by RNA-interference (RNAi) in protonemal cells, we observed a cytokinesis failure following a defect in phragmoplast expansion. In addition, misaligned chromosomes were frequently detected in the pre-anaphase spindle and the anaphase onset was significantly delayed, indicating that PpNACK also plays a role in pre-anaphase. Consistent with the appearance of early and late mitotic phenotypes, endogenous PpNACK was localised to the interpolar microtubule (MT) overlap from prometaphase through telophase. In vitro MT gliding assay and single motor motility assay showed that PpNACK-b is a processive, plus-end-directed motor, suggesting that PpNACK is capable of transporting cargoes along the spindle/phragmoplast MT. Our study using Physcomitrella patens demonstrated that PpNACK is an active motor protein and identified unexpected and conserved roles of PpNACK during the mitosis of P. patens.


Subject(s)
Bryopsida/cytology , Bryopsida/metabolism , Chromosomes, Plant/metabolism , Cytokinesis , Kinesins/metabolism , Metaphase , Plant Proteins/metabolism , Bryopsida/genetics , Kinesins/deficiency , Kinesins/genetics , Microtubules/metabolism , Movement , Plant Proteins/genetics , Prometaphase , RNA Interference , Telophase
10.
Biochem Biophys Res Commun ; 463(1-2): 123-9, 2015.
Article in English | MEDLINE | ID: mdl-26002460

ABSTRACT

Polarization of epithelial cells requires vectorial sorting and transport of polarity proteins to apical or basolateral domains. Kif5b is the mouse homologue of the human ubiquitous Kinesin Heavy Chain (uKHC). To investigate the function of Kif5b in epithelial cells, we examined the phenotypes of Kif5b-deficient MDCK cells. Stable knockdown of Kif5b in MDCK cells resulted in reduced cell proliferation rate, profound changes in cell morphology, loss of epithelial cell marker, and gain of mesenchymal marker, as well as increased cell migration, invasion, and tumorigenesis abilities. E-cadherin and NMMIIA could interact with Kif5b in polarized MDCK cells, and their expression levels were decreased in Kif5b-deficient MDCK cells. Overexpression of E-cadherin and NMMIIA in Kif5b depleted MDCK cells could decrease mesenchymal marker expression and cell migration ability. These results indicate that stable knockdown of Kif5b in MDCK cells can lead to epithelial-mesenchymal transition, which is mediated by defective E-cadherin and NMMIIA expression.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Kinesins/deficiency , Animals , Cadherins/metabolism , Cell Cycle , Cell Line , Cell Polarity/genetics , Cell Polarity/physiology , Cell Proliferation , Dogs , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Humans , Kinesins/antagonists & inhibitors , Kinesins/genetics , Madin Darby Canine Kidney Cells , Mice , Mice, Nude , Myosin Heavy Chains/metabolism , Neoplasm Metastasis
11.
Development ; 139(22): 4152-61, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23034632

ABSTRACT

Abnormal activation of Hedgehog (Hh) signaling leads to basal cell carcinoma (BCC) of the skin, the most common human cancer. Gli2, the major transcriptional activator of Hh signaling, is essential for hair follicle development and its overexpression in epidermis induces BCC formation and maintains tumor growth. Despite its importance in skin development and tumorigenesis, little is known about the molecular regulation of Gli2. Sufu and Kif7 are two evolutionarily conserved regulators of Gli transcription factors. Here, we show that Sufu and Kif7 regulate Gli2 through distinct mechanisms in keratinocytes. Sufu restricts the activity of Gli2 through cytoplasmic sequestration. Kif7 possesses Sufu-dependent and -independent regulatory functions in Hh signaling: while it promotes Hh pathway activity through the dissociation of Sufu-Gli2 complex, it also contributes to the repression of Hh target genes in the absence of Sufu. Deletion of both Sufu and Kif7 in embryonic skin leads to complete loss of follicular fate. Importantly, although inactivation of Sufu or Kif7 alone in adult epidermis cannot promote BCC formation, their simultaneous deletion induces BCC. These studies establish Sufu and Kif7 as crucial components in the regulation of Gli2 localization and activity, and illustrate their overlapping functions in skin development and tumor suppression.


Subject(s)
Carcinoma, Basal Cell/metabolism , Keratinocytes/metabolism , Kinesins/metabolism , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Skin Neoplasms/metabolism , Skin/embryology , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Proliferation , Cell Transformation, Neoplastic , Cytoplasm , Hair Follicle/embryology , Hedgehog Proteins , Kinesins/deficiency , Kinesins/genetics , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/biosynthesis , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Repressor Proteins/deficiency , Repressor Proteins/genetics , Signal Transduction , Skin Neoplasms/pathology , Zinc Finger Protein Gli2
12.
Reproduction ; 150(3): 209-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26245936

ABSTRACT

Male fertility relies on the production of functional spermatozoa. Spermatogenesis is a complex differentiation process that is characterized by meiosis and dramatic morphogenesis of haploid cells. Spermatogenesis involves active changes in the microtubular network to support meiotic divisions, cell polarization, the reshaping of the nucleus, and the formation of a flagellum. Previously, we have demonstrated that a microtubule-based anterograde transport motor protein KIF3A is required for the sperm tail formation and nuclear shaping during spermatogenesis. In this study, we show that KIF3A interacts with a KIF1-binding protein (KBP) in the mouse testis. We have characterized the expression and localization pattern of KBP during spermatogenesis and localized both KIF3A and KBP in the cytoplasm of round spermatids and manchette of elongating spermatids. Interestingly, KBP localized also in the late chromatoid body (CB) of elongating spermatids, whose function involves intracellular movement and association with the microtubular network. Altogether our results suggest a role for KBP in spermatid elongation and in the function of the late CB.


Subject(s)
Carrier Proteins/metabolism , Haploidy , Kinesins/metabolism , Spermatids/metabolism , Spermatogenesis , Testis/metabolism , Animals , Kinesins/deficiency , Kinesins/genetics , Male , Mice, Knockout , Protein Binding , Protein Isoforms , Signal Transduction , Testis/cytology
13.
J Neurosci ; 32(40): 14033-49, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23035110

ABSTRACT

Microtubules are nearly uniformly oriented in the axons of vertebrate neurons but are non-uniformly oriented in their dendrites. Studies to date suggest a scenario for establishing these microtubule patterns whereby microtubules are transported into the axon and nascent dendrites with plus-ends-leading, and then additional microtubules of the opposite orientation are transported into the developing dendrites. Here, we used contemporary tools to confirm that depletion of kinesin-6 (also called CHO1/MKLP1 or kif23) from rat sympathetic neurons causes a reduction in the appearance of minus-end-distal microtubules in developing dendrites, which in turn causes them to assume an axon-like morphology. Interestingly, we observed a similar phenomenon when we depleted kinesin-12 (also called kif15 or HKLP2). Both motors are best known for their participation in mitosis in other cell types, and both are enriched in the cell body and dendrites of neurons. Unlike kinesin-12, which is present throughout the neuron, kinesin-6 is barely detectable in the axon. Accordingly, depletion of kinesin-6, unlike depletion of kinesin-12, has no effect on axonal branching or navigation. Interestingly, depletion of either motor results in faster growing axons with greater numbers of mobile microtubules. Based on these observations, we posit a model whereby these two motors generate forces that attenuate the transport of microtubules with plus-ends-leading from the cell body into the axon. Some of these microtubules are not only prevented from moving into the axon but are driven with minus-ends-leading into developing dendrites. In this manner, these so-called "mitotic" motors coregulate the microtubule patterns of axons and dendrites.


Subject(s)
Axons/ultrastructure , Dendrites/ultrastructure , Microtubules/physiology , Molecular Motor Proteins/physiology , Nerve Tissue Proteins/physiology , Spindle Apparatus/chemistry , Adrenergic Fibers/ultrastructure , Animals , Antibodies, Monoclonal/pharmacology , Cell Polarity/physiology , Cerebral Cortex/chemistry , Cerebral Cortex/cytology , Kinesins/antagonists & inhibitors , Kinesins/deficiency , Kinesins/genetics , Kinesins/physiology , Morphogenesis/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Neurogenesis/physiology , Rats , Superior Cervical Ganglion/chemistry , Superior Cervical Ganglion/cytology
14.
J Neurosci ; 32(16): 5486-99, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22514311

ABSTRACT

Regulation of NMDA receptor trafficking is crucial to modulate neuronal communication. Ca(2+)/calmodulin-dependent protein kinase phosphorylates the tail domain of KIF17, a member of the kinesin superfamily, to control NMDA receptor subunit 2B (GluN2B) transport by changing the KIF17-cargo interaction in vitro. However, the mechanisms of regulation of GluN2B transport in vivo and its physiological significance are unknown. We generated transgenic mice carrying wild-type KIF17 (TgS), or KIF17 with S1029A (TgA) or S1029D (TgD) phosphomimic mutations in kif17(-/-) background. TgA/kif17(-/-) and TgD/kif17(-/-) mice exhibited reductions in synaptic NMDA receptors because of their inability to load/unload GluN2B onto/from KIF17, leading to impaired neuronal plasticity, CREB activation, and spatial memory. Expression of GFP-KIF17 in TgS/kif17(-/-) mouse neurons rescued the synaptic and behavioral defects of kif17(-/-) mice. These results suggest that phosphorylation-based regulation of NMDA receptor transport is critical for learning and memory in vivo.


Subject(s)
Kinesins/metabolism , Memory/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Analysis of Variance , Animals , Biophysics , Biotinylation/methods , CREB-Binding Protein/metabolism , Cells, Cultured , Cycloheximide/pharmacology , DNA-Binding Proteins , Disks Large Homolog 4 Protein , Electric Stimulation , Excitatory Postsynaptic Potentials/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Guanylate Kinases/metabolism , Hippocampus/cytology , Hippocampus/physiology , Immunoprecipitation , In Vitro Techniques , Kinesins/deficiency , Long-Term Potentiation/genetics , Male , Maze Learning/physiology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Mutagenesis, Site-Directed/methods , Mutation/genetics , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Nuclear Proteins/metabolism , Phosphorylation/genetics , Protein Binding/genetics , Protein Synthesis Inhibitors/pharmacology , Protein Transport/genetics , RNA-Binding Proteins , Reaction Time/genetics , Serine/genetics , Serine/metabolism , Synapses/genetics , Synaptophysin/metabolism , Time Factors
15.
Neurogenetics ; 13(2): 169-79, 2012 May.
Article in English | MEDLINE | ID: mdl-22466687

ABSTRACT

Hereditary spastic paraplegia (HSP) is a neurodegenerative disorder preferentially affecting the longest corticospinal axons. More than 40 HSP genetic loci have been identified, among them SPG10, an autosomal dominant HSP caused by point mutations in the neuronal kinesin heavy chain protein KIF5A. Constitutive KIF5A knockout (KIF5A( -/- )) mice die early after birth. In these mice, lungs were unexpanded, and cell bodies of lower motor neurons in the spinal cord swollen, but the pathomechanism remained unclear. To gain insights into the pathophysiology, we characterized survival, outgrowth, and function in primary motor and sensory neuron cultures from KIF5A( -/- ) mice. Absence of KIF5A reduced survival in motor neurons, but not in sensory neurons. Outgrowth of axons and dendrites was remarkably diminished in KIF5A( -/- ) motor neurons. The number of axonal branches was reduced, whereas the number of dendrites was not altered. In KIF5A( -/- ) sensory neurons, neurite outgrowth was decreased but the number of neurites remained unchanged. In motor neurons maximum and average velocity of mitochondrial transport was reduced both in anterograde and retrograde direction. Our results point out a role of KIF5A in process outgrowth and axonal transport of mitochondria, affecting motor neurons more severely than sensory neurons. This gives pathophysiological insights into KIF5A associated HSP, and matches the clinical findings of predominant degeneration of the longest axons of the corticospinal tract.


Subject(s)
Axonal Transport/genetics , Kinesins/metabolism , Spastic Paraplegia, Hereditary/genetics , Animals , Axons/metabolism , Cells, Cultured , Disease Models, Animal , Gene Knockout Techniques , Kinesins/deficiency , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Motor Neurons/cytology , Motor Neurons/metabolism , Mutation, Missense , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Spastic Paraplegia, Hereditary/metabolism
16.
Science ; 375(6581): eabj3944, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35143306

ABSTRACT

Human oocytes are prone to assembling meiotic spindles with unstable poles, which can favor aneuploidy in human eggs. The underlying causes of spindle instability are unknown. We found that NUMA (nuclear mitotic apparatus protein)-mediated clustering of microtubule minus ends focused the spindle poles in human, bovine, and porcine oocytes and in mouse oocytes depleted of acentriolar microtubule-organizing centers (aMTOCs). However, unlike human oocytes, bovine, porcine, and aMTOC-free mouse oocytes have stable spindles. We identified the molecular motor KIFC1 (kinesin superfamily protein C1) as a spindle-stabilizing protein that is deficient in human oocytes. Depletion of KIFC1 recapitulated spindle instability in bovine and aMTOC-free mouse oocytes, and the introduction of exogenous KIFC1 rescued spindle instability in human oocytes. Thus, the deficiency of KIFC1 contributes to spindle instability in human oocytes.


Subject(s)
Cell Cycle Proteins/metabolism , Kinesins/deficiency , Oocytes/physiology , Oocytes/ultrastructure , Spindle Apparatus/physiology , Spindle Poles/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Cattle , Dynactin Complex/metabolism , Dyneins/metabolism , Female , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Organizing Center/physiology , Microtubule-Organizing Center/ultrastructure , Microtubules/metabolism , Recombinant Proteins/metabolism , Spindle Apparatus/ultrastructure , Spindle Poles/ultrastructure , Swine
17.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166335, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34973373

ABSTRACT

BACKGROUND & AIMS: Loss of primary cilia in epithelial cells is known to cause cystic diseases of the liver and kidney. We have previously shown that during experimental and human cirrhosis that primary cilia were predominantly expressed on biliary cells in the ductular reaction. However, the role of primary cilia in the pathogenesis of the ductular reaction is not fully understood. METHODS: Primary cilia were specifically removed in biliary epithelial cells (BECs) by the administration of tamoxifen to Kif3af/f;CK19CreERT mice at week 2 of a 20-week course of TAA treatment. Biliary progenitor cells were isolated and grown as organoids from gallbladders. Cells and tissue were analysed using histology, immunohistochemistry and Western blot assays. RESULTS: At the end of 20 weeks TAA administration, primary cilia loss in liver BECs resulted in multiple microscopic cystic lesions within an unaltered ductular reaction. These were not seen in control mice who did not receive TAA. There was no effect of biliary primary cilia loss on the development of cirrhosis. Increased cellular proliferation was seen within the cystic structures associated with a decrease in hepatocyte lobular proliferation. Loss of primary cilia within biliary organoids was initially associated with reduced cell passage survival but this inhibitory effect was diminished in later passages. ERK but not WNT signalling was enhanced in primary cilia loss-induced cystic lesions in vivo and its inhibition reduced the expansion of primary cilia deficient biliary progenitor cells in vitro. CONCLUSIONS: TAA-treated kif3a BEC-specific knockout mice had an unaltered progression to cirrhosis, but developed cystic lesions that showed increased proliferation.


Subject(s)
Cilia/pathology , Cysts/pathology , Kinesins/genetics , Liver Diseases/pathology , Animals , Biliary Tract/cytology , Cell Proliferation , Cilia/metabolism , Cysts/chemically induced , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratin-19/genetics , Keratin-19/metabolism , Kinesins/deficiency , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thioacetamide/toxicity
18.
Nat Cell Biol ; 4(10): 737-42, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12360284

ABSTRACT

Xkid chromokinesin is required for chromosome alignment on the metaphase plate of spindles formed in Xenopus laevis egg extracts. We have investigated the role of Xkid in Xenopus oocyte meiotic maturation, a progesterone-triggered process that reinitiates the meiotic cell cycle in oocytes arrested at the G2/M border of meiosis I. Here we show that Xkid starts to accumulate at the time of germinal vesicle breakdown and reaches its largest quantities at metaphase II in oocytes treated with progesterone. Both germinal vesicle breakdown and spindle assembly at meiosis I can occur normally in the absence of Xkid. But Xkid-depleted oocytes cannot reactivate Cdc2/cyclin B after meiosis I and, instead of proceeding to meiosis II, they enter an interphase-like state and undergo DNA replication. Expression of a Xkid mutant that lacks the DNA-binding domain allows Xkid-depleted oocytes to complete meiotic maturation. Our results show that Xkid has a role in the meiotic cell cycle that is independent from its role in metaphase chromosome alignment.


Subject(s)
Chromosome Segregation/genetics , DNA-Binding Proteins/genetics , Kinesins/genetics , Meiosis/genetics , Nuclear Proteins/genetics , Oocytes/growth & development , Xenopus Proteins , Xenopus laevis/genetics , Animals , Apoptosis/genetics , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chromosome Segregation/drug effects , Cyclin B/genetics , Cyclin B/metabolism , DNA Replication/drug effects , DNA Replication/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/deficiency , Female , Kinesins/biosynthesis , Kinesins/deficiency , Meiosis/drug effects , Nuclear Proteins/biosynthesis , Nuclear Proteins/deficiency , Oligoribonucleotides, Antisense , Oocytes/drug effects , Oocytes/metabolism , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Xenopus laevis/embryology
19.
Sci Rep ; 11(1): 3191, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542431

ABSTRACT

The Kif26a protein-coding gene has been identified as a negative regulator of the GDNF-Ret signaling pathway in enteric neurons. The aim of this study was to investigate the influence of genetic background on the phenotype of Kif26a-deficient (KO, -/-) mice. KO mice with both C57BL/6 and BALB/c genetic backgrounds were established. Survival rates and megacolon development were compared between these two strains of KO mice. Functional bowel assessments and enteric neuron histopathology were performed in the deficient mice. KO mice with the BALB/c genetic background survived more than 400 days without evidence of megacolon, while all C57BL/6 KO mice developed megacolon and died within 30 days. Local enteric neuron hyperplasia in the colon and functional bowel abnormalities were observed in BALB/c KO mice. These results indicated that megacolon and enteric neuron hyperplasia in KO mice are influenced by the genetic background. BALB/c KO mice may represent a viable model for functional gastrointestinal diseases such as chronic constipation, facilitating studies on the underlying mechanisms and providing a foundation for the development of treatments.


Subject(s)
Enteric Nervous System/metabolism , Intestine, Small/metabolism , Kinesins/genetics , Megacolon/genetics , Neurons/metabolism , Animals , Enteric Nervous System/pathology , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Intestine, Small/innervation , Intestine, Small/pathology , Kinesins/deficiency , Megacolon/metabolism , Megacolon/mortality , Megacolon/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Dehydrogenase/genetics , NADPH Dehydrogenase/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction , Species Specificity , Survival Analysis
20.
Autophagy ; 17(4): 903-924, 2021 04.
Article in English | MEDLINE | ID: mdl-32160081

ABSTRACT

Trimethyltin chloride (TMT) is widely used as a constituent of fungicides and plastic stabilizers in the industrial and agricultural fields, and is generally acknowledged to have potent neurotoxicity, especially in the hippocampus; however, the mechanism of induction of neurotoxicity by TMT remains elusive. Herein, we exposed Neuro-2a cells to different concentrations of TMT (2, 4, and 8 µM) for 24 h. Proteomic analysis, coupled with bioinformatics analysis, revealed the important role of macroautophagy/autophagy-lysosome machinery in TMT-induced neurotoxicity. Further analysis indicated significant impairment of autophagic flux by TMT via suppressed lysosomal function, such as by inhibiting lysosomal proteolysis and changing the lysosomal pH, thereby contributing to defects in autophagic clearance and subsequently leading to nerve cell death. Mechanistically, molecular interaction networks of Ingenuity Pathway Analysis identified a downregulated molecule, KIF5A (kinesin family member 5A), as a key target in TMT-impaired autophagic flux. TMT decreased KIF5A protein expression, disrupted the interaction between KIF5A and lysosome, and impaired lysosomal axonal transport. Moreover, Kif5a overexpression restored axonal transport, increased lysosomal dysfunction, and antagonized TMT-induced neurotoxicity in vitro. Importantly, in TMT-administered mice with seizure symptoms and histomorphological injury in the hippocampus, TMT inhibited KIF5A expression in the hippocampus. Gene transfer of Kif5a enhanced autophagic clearance in the hippocampus and alleviated TMT-induced neurotoxicity in vivo. Our results are the first to demonstrate KIF5A-dependent axonal transport deficiency to cause autophagic flux impairment via disturbance of lysosomal function in TMT-induced neurotoxicity; manipulation of KIF5A may be a therapeutic approach for antagonizing TMT-induced neurotoxicity.Abbreviations: 3-MA: 3-methyladenine; AAV: adeno-associated virus; ACTB: actin beta; AGC: automatic gain control; ATG: autophagy-related; ATP6V0D1: ATPase H+ transporting lysosomal V0 subunit D1; ATP6V1E1: ATPase H+ transporting lysosomal V1 subunit E1; CA: cornu ammonis; CQ: chloroquine; CTSB: cathepsin B; CTSD: cathepsin D; DCTN1: dynactin subunit 1; DG: dentate gyrus; DYNLL1: dynein light chain LC8-type 1; FBS: fetal bovine serum; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor associated protein like 1; GABARAPL2: GABA type A receptor associated protein like 2; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; IPA: Ingenuity Pathway Analysis; KEGG: Kyoto Encyclopedia of Genes and Genomes; KIF5A: kinesin family member 5A; LAMP: lysosomal-associated membrane protein; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; NBR1: NBR1 autophagy cargo receptor; OPTN: optineurin; PBS: phosphate-buffered saline; PFA: paraformaldehyde; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PRM: parallel reaction monitoring; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; SYP: synaptophysin; TAX1BP1: Tax1 binding protein 1; TMT: trimethyltin chloride; TUB: tubulin.


Subject(s)
Autophagy/drug effects , Axonal Transport/drug effects , Kinesins/metabolism , Neurotoxins/toxicity , Trimethyltin Compounds/toxicity , Animals , Animals, Newborn , Autophagosomes/drug effects , Autophagosomes/metabolism , Cell Line , Hippocampus/pathology , Kinesins/deficiency , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Models, Biological , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL