Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
FASEB J ; 38(8): e23623, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38656660

The nuclear transport of proteins plays an important role in mediating the transition from egg to embryo and distinct karyopherins have been implicated in this process. Here, we studied the impact of KPNA2 deficiency on preimplantation embryo development in mice. Loss of KPNA2 results in complete arrest at the 2cell stage and embryos exhibit the inability to activate their embryonic genome as well as a severely disturbed nuclear translocation of Nucleoplasmin 2. Our findings define KPNA2 as a new maternal effect gene.


Embryonic Development , alpha Karyopherins , Animals , Female , Mice , alpha Karyopherins/metabolism , alpha Karyopherins/genetics , Embryonic Development/genetics , Fertility/genetics , Mice, Knockout , Maternal Inheritance , Gene Expression Regulation, Developmental , Male , Pregnancy , Nucleoplasmins/metabolism , Nucleoplasmins/genetics , Blastocyst/metabolism
3.
Development ; 148(19)2021 10 01.
Article En | MEDLINE | ID: mdl-34473250

Spermatogenesis is driven by an ordered series of events, which rely on trafficking of specific proteins between nucleus and cytoplasm. The karyopherin α family of proteins mediates movement of specific cargo proteins when bound to karyopherin ß. Karyopherin α genes have distinct expression patterns in mouse testis, implying they may have unique roles during mammalian spermatogenesis. Here, we use a loss-of-function approach to determine specifically the role of Kpna6 in spermatogenesis and male fertility. We show that ablation of Kpna6 in male mice leads to infertility and has multiple cumulative effects on both germ cells and Sertoli cells. Kpna6-deficient mice exhibit impaired Sertoli cell function, including loss of Sertoli cells and a compromised nuclear localization of the androgen receptor. Furthermore, our data demonstrate devastating defects on spermiogenesis, including incomplete sperm maturation and a massive reduction in sperm number, accompanied by disturbed histone-protamine exchange, differential localization of the transcriptional regulator BRWD1 and altered expression of RFX2 target genes. Our work uncovers an essential role of Kpna6 in spermatogenesis and, hence, in male fertility.


Infertility, Male/metabolism , Spermatogenesis , alpha Karyopherins/genetics , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Infertility, Male/genetics , Loss of Function Mutation , Male , Mice , Mice, Inbred C57BL , Receptors, Androgen/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism , alpha Karyopherins/deficiency , alpha Karyopherins/metabolism
4.
Science ; 369(6505): 842-846, 2020 08 14.
Article En | MEDLINE | ID: mdl-32792398

How is neuropathic pain regulated in peripheral sensory neurons? Importins are key regulators of nucleocytoplasmic transport. In this study, we found that importin α3 (also known as karyopherin subunit alpha 4) can control pain responsiveness in peripheral sensory neurons in mice. Importin α3 knockout or sensory neuron-specific knockdown in mice reduced responsiveness to diverse noxious stimuli and increased tolerance to neuropathic pain. Importin α3-bound c-Fos and importin α3-deficient neurons were impaired in c-Fos nuclear import. Knockdown or dominant-negative inhibition of c-Fos or c-Jun in sensory neurons reduced neuropathic pain. In silico screens identified drugs that mimic importin α3 deficiency. These drugs attenuated neuropathic pain and reduced c-Fos nuclear localization. Thus, perturbing c-Fos nuclear import by importin α3 in peripheral neurons can promote analgesia.


Chronic Pain/physiopathology , Neuralgia/physiopathology , Sensory Receptor Cells/physiology , alpha Karyopherins/physiology , Active Transport, Cell Nucleus , Animals , Benzophenones/pharmacology , Chronic Pain/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Isoxazoles/pharmacology , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Proto-Oncogene Proteins c-fos/antagonists & inhibitors , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/metabolism , alpha Karyopherins/genetics
5.
Cell Rep ; 31(3): 107549, 2020 04 21.
Article En | MEDLINE | ID: mdl-32320654

Importin-α adaptor proteins orchestrate dynamic nuclear transport processes involved in cellular homeostasis. Here, we show that importin-α3, one of the main NF-κB transporters, is the most abundantly expressed classical nuclear transport factor in the mammalian respiratory tract. Importin-α3 promoter activity is regulated by TNF-α-induced NF-κB in a concentration-dependent manner. High-level TNF-α-inducing highly pathogenic avian influenza A viruses (HPAIVs) isolated from fatal human cases harboring human-type polymerase signatures (PB2 627K, 701N) significantly downregulate importin-α3 mRNA expression in primary lung cells. Importin-α3 depletion is restored upon back-mutating the HPAIV polymerase into an avian-type signature (PB2 627E, 701D) that can no longer induce high TNF-α levels. Importin-α3-deficient mice show reduced NF-κB-activated antiviral gene expression and increased influenza lethality. Thus, importin-α3 plays a key role in antiviral immunity against influenza. Lifting the bottleneck in importin-α3 availability in the lung might provide a new strategy to combat respiratory virus infections.


Influenza A virus/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , alpha Karyopherins/biosynthesis , A549 Cells , Animals , Cell Line, Tumor , Chlorocebus aethiops , Down-Regulation , Female , HEK293 Cells , Humans , Influenza, Human/genetics , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vero Cells , alpha Karyopherins/genetics , alpha Karyopherins/immunology
6.
Front Med (Lausanne) ; 5: 338, 2018.
Article En | MEDLINE | ID: mdl-30560131

Acute intra-renal infusion of bradykinin increases diuresis and natriuresis via inhibition of vasopressin activity. However, the consequences of chronically increased bradykinin in the kidneys have not yet been studied. A new transgenic animal model producing an excess of bradykinin by proximal tubular cells (KapBK rats) was generated and submitted to different salt containing diets to analyze changes in blood pressure and other cardiovascular parameters, urine excretion, and composition, as well as levels and expression of renin-angiotensin system components. Despite that KapBK rats excrete more urine and sodium, they have similar blood pressure as controls with the exception of a small increase in systolic blood pressure (SBP). However, they present decreased renal artery blood flow, increased intrarenal expression of angiotensinogen, and decreased mRNA expression of vasopressin V1A receptor (AVPR1A), suggesting a mechanism for the previously described reduction of renal vasopressin sensitivity by bradykinin. Additionally, reduced heart rate variability (HRV), increased cardiac output and frequency, and the development of cardiac hypertrophy are the main chronic effects observed in the cardiovascular system. In conclusion: (1) the transgenic KapBK rat is a useful model for studying chronic effects of bradykinin in kidney; (2) increased renal bradykinin causes changes in renin angiotensin system regulation; (3) decreased renal vasopressin sensitivity in KapBK rats is related to decreased V1A receptor expression; (4) although increased renal levels of bradykinin causes no changes in mean arterial pressure (MAP), it causes reduction in HRV, augmentation in cardiac frequency and output and consequently cardiac hypertrophy in rats after 6 months of age.

7.
Cell Rep ; 25(11): 3169-3179.e7, 2018 12 11.
Article En | MEDLINE | ID: mdl-30540948

Importins mediate transport from synapse to soma and from cytoplasm to nucleus, suggesting that perturbation of importin-dependent pathways should have significant neuronal consequences. A behavioral screen on five importin α knockout lines revealed that reduced expression of importin α5 (KPNA1) in hippocampal neurons specifically decreases anxiety in mice. Re-expression of importin α5 in ventral hippocampus of knockout animals increased anxiety behaviors to wild-type levels. Hippocampal neurons lacking importin α5 reveal changes in presynaptic plasticity and modified expression of MeCP2-regulated genes, including sphingosine kinase 1 (Sphk1). Knockout of importin α5, but not importin α3 or α4, reduces MeCP2 nuclear localization in hippocampal neurons. A Sphk1 blocker reverses anxiolysis in the importin α5 knockout mouse, while pharmacological activation of sphingosine signaling has robust anxiolytic effects in wild-type animals. Thus, importin α5 influences sphingosine-sensitive anxiety pathways by regulating MeCP2 nuclear import in hippocampal neurons.


Anxiety/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , alpha Karyopherins/metabolism , Animals , Anti-Anxiety Agents/pharmacology , Behavior, Animal , Carbolines/pharmacology , Hippocampus/pathology , Mice, Knockout , Neurons/metabolism , Phenotype , Synapses/metabolism , Transcription, Genetic , alpha Karyopherins/deficiency
8.
Proc Natl Acad Sci U S A ; 115(11): E2624-E2633, 2018 03 13.
Article En | MEDLINE | ID: mdl-29476013

Spinocerebellar ataxia type 3 (SCA3) is a neurodegenerative disorder caused by a CAG expansion in the ATXN3 gene leading to a polyglutamine expansion in the ataxin-3 protein. The nuclear presence and aggregation of expanded ataxin-3 are critical steps in disease pathogenesis. To identify novel therapeutic targets, we investigated the nucleocytoplasmic transport system by screening a collection of importins and exportins that potentially modulate this nuclear localization. Using cell, Drosophila, and mouse models, we focused on three transport proteins, namely, CRM1, IPO13, KPNA3, and their respective Drosophila orthologs Emb, Cdm, and Kap-α3. While overexpression of CRM1/Emb demonstrated positive effects in Drosophila, KPNA3/Kap-α3 emerged as the most promising target, as knockdown via multiple RNAi lines demonstrated its ability to shuttle both truncated and full-length expanded ataxin-3, rescue neurodegeneration, restore photoreceptor formation, and reduce aggregation. Furthermore, KPNA3 knockout in SCA3 mice resulted in an amelioration of molecular and behavioral disturbances such as total activity, anxiety, and gait. Since KPNA3 is known to function as an import protein and recognize nuclear localization signals (NLSs), this work unites ataxin-3 structure to the nuclear pore machinery and provides a link between karyopherins, NLS signals, and polyglutamine disease, as well as demonstrates that KPNA3 is a key player in the pathogenesis of SCA3.


Active Transport, Cell Nucleus/genetics , Ataxin-3/genetics , Machado-Joseph Disease/genetics , alpha Karyopherins/genetics , Animals , Ataxin-3/metabolism , DNA Repeat Expansion , Disease Models, Animal , Drosophila , Female , HEK293 Cells , Humans , Machado-Joseph Disease/metabolism , Male , Mice , Mice, Knockout , Peptides , alpha Karyopherins/metabolism
9.
PLoS Pathog ; 14(1): e1006823, 2018 01.
Article En | MEDLINE | ID: mdl-29304174

Herpesviruses are large DNA viruses which depend on many nuclear functions, and therefore on host transport factors to ensure specific nuclear import of viral and host components. While some import cargoes bind directly to certain transport factors, most recruit importin ß1 via importin α. We identified importin α1 in a small targeted siRNA screen to be important for herpes simplex virus (HSV-1) gene expression. Production of infectious virions was delayed in the absence of importin α1, but not in cells lacking importin α3 or importin α4. While nuclear targeting of the incoming capsids, of the HSV-1 transcription activator VP16, and of the viral genomes were not affected, the nuclear import of the HSV-1 proteins ICP4 and ICP0, required for efficient viral transcription, and of ICP8 and pUL42, necessary for DNA replication, were reduced. Furthermore, quantitative electron microscopy showed that fibroblasts lacking importin α1 contained overall fewer nuclear capsids, but an increased proportion of mature nuclear capsids indicating that capsid formation and capsid egress into the cytoplasm were impaired. In neurons, importin α1 was also not required for nuclear targeting of incoming capsids, but for nuclear import of ICP4 and for the formation of nuclear capsid assembly compartments. Our data suggest that importin α1 is specifically required for the nuclear localization of several important HSV1 proteins, capsid assembly, and capsid egress into the cytoplasm, and may become rate limiting in situ upon infection at low multiplicity or in terminally differentiated cells such as neurons.


Capsid Proteins/metabolism , Cell Nucleus/metabolism , Fibroblasts/virology , Herpesvirus 1, Human/physiology , Neurons/virology , Virus Assembly/genetics , alpha Karyopherins/physiology , Active Transport, Cell Nucleus/genetics , Animals , Capsid/metabolism , Cell Line , Cell Nucleus/virology , Cricetinae , Fibroblasts/metabolism , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , alpha Karyopherins/genetics
10.
Stem Cells ; 34(11): 2784-2797, 2016 11.
Article En | MEDLINE | ID: mdl-27434733

Satellite cells are stem cells with an essential role in skeletal muscle repair. Precise regulation of gene expression is critical for proper satellite cell quiescence, proliferation, differentiation and self-renewal. Nuclear proteins required for gene expression are dependent on the nucleocytoplasmic transport machinery to access to nucleus, however little is known about regulation of nuclear transport in satellite cells. The best characterized nuclear import pathway is classical nuclear import which depends on a classical nuclear localization signal (cNLS) in a cargo protein and the heterodimeric import receptors, karyopherin alpha (KPNA) and beta (KPNB). Multiple KPNA1 paralogs exist and can differ in importing specific cNLS proteins required for cell differentiation and function. We show that transcripts for six Kpna paralogs underwent distinct changes in mouse satellite cells during muscle regeneration accompanied by changes in cNLS proteins in nuclei. Depletion of KPNA1, the most dramatically altered KPNA, caused satellite cells in uninjured muscle to prematurely activate, proliferate and undergo apoptosis leading to satellite cell exhaustion with age. Increased proliferation of satellite cells led to enhanced muscle regeneration at early stages of regeneration. In addition, we observed impaired nuclear localization of two key KPNA1 cargo proteins: p27, a cyclin-dependent kinase inhibitor associated with cell cycle control and lymphoid enhancer factor 1, a critical cotranscription factor for ß-catenin. These results indicate that regulated nuclear import of proteins by KPNA1 is critical for satellite cell proliferation and survival and establish classical nuclear import as a novel regulatory mechanism for controlling satellite cell fate. Stem Cells 2016;34:2784-2797.


Cell Nucleus/metabolism , Muscle, Skeletal/metabolism , Nuclear Localization Signals/genetics , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , alpha Karyopherins/genetics , Active Transport, Cell Nucleus/genetics , Animals , Barium Compounds/toxicity , Cell Proliferation , Cell Survival , Chlorides/toxicity , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytoplasm/metabolism , Female , Gene Expression Regulation , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/injuries , Nuclear Localization Signals/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction , alpha Karyopherins/deficiency , beta Catenin/genetics , beta Catenin/metabolism
11.
Mol Cell Proteomics ; 13(5): 1286-98, 2014 May.
Article En | MEDLINE | ID: mdl-24623588

The importin α:ß complex is responsible for the nuclear import of proteins bearing classical nuclear localization signals. In mammals, several importin α subtypes are known to exist that are suggested to have individual functions. Importin α 7 was shown to play a crucial role in early embryonic development in mice. Embryos from importin α 7-depleted females stop at the two-cell stage and show disturbed zygotic genome activation. As there is evidence that individual importin α subtypes possess cargo specificities, we hypothesized that importin α 7 binds a unique set of intracellular proteins. With the use of a collection of in vitro and in vivo binding assays, importin α 7 interaction partners were identified that differed from proteins found to bind to importin α 2 and 3. One of the proteins preferentially binding importin α 7 was the maternal effect protein Brg1. However, Brg1 was localized in oocyte nuclei in importin α 7-deficient embryos, albeit in reduced amounts, suggesting additional modes of nuclear translocation of this factor. An additional SILAC-based screening approach identified Ash2l, Chd3, Mcm3, and Smarcc1, whose nuclear import seems to be disturbed in importin α 7-deficient fibroblasts.


Oocytes/metabolism , Proteomics/methods , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , Animals , Cell Nucleus/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Female , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Minichromosome Maintenance Complex Component 3/metabolism , NIH 3T3 Cells , Nuclear Proteins/metabolism , Substrate Specificity , Transcription Factors/metabolism
12.
EMBO J ; 31(6): 1350-63, 2012 Mar 21.
Article En | MEDLINE | ID: mdl-22246183

Retrograde axonal injury signalling stimulates cell body responses in lesioned peripheral neurons. The involvement of importins in retrograde transport suggests that transcription factors (TFs) might be directly involved in axonal injury signalling. Here, we show that multiple TFs are found in axons and associate with dynein in axoplasm from injured nerve. Biochemical and functional validation for one TF family establishes that axonal STAT3 is locally translated and activated upon injury, and is transported retrogradely with dynein and importin α5 to modulate survival of peripheral sensory neurons after injury. Hence, retrograde transport of TFs from axonal lesion sites provides a direct link between axon and nucleus.


Axons/metabolism , Ganglia, Spinal/metabolism , Peripheral Nerve Injuries/metabolism , Sensory Receptor Cells/metabolism , Transcription Factors/metabolism , Animals , Axonal Transport/physiology , Cell Nucleus/metabolism , Dyneins/metabolism , Karyopherins/metabolism , Male , Mice , Protein Transport/physiology , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology
13.
PLoS One ; 6(3): e18310, 2011 Mar 29.
Article En | MEDLINE | ID: mdl-21479251

Importin α is involved in the nuclear import of proteins. It also contributes to spindle assembly and nuclear membrane formation, however, the underlying mechanisms are poorly understood. Here, we studied the function of importin α7 by gene targeting in mice and show that it is essential for early embryonic development. Embryos lacking importin α7 display a reduced ability for the first cleavage and arrest completely at the two-cell stage. We show that the zygotic genome activation is severely disturbed in these embryos. Our findings indicate that importin α7 is a new member of the small group of maternal effect genes.


Embryonic Development/genetics , Genome/genetics , Zygote/metabolism , alpha Karyopherins/metabolism , Animals , DNA Replication , Embryo, Mammalian/embryology , Female , Gene Expression Regulation, Developmental , Gene Targeting , Genes, Essential/genetics , Male , Mice , Mice, Inbred C57BL , Nuclear Envelope/metabolism , Oocytes/cytology , Oocytes/metabolism , Ovary/cytology , Ovary/metabolism , Parthenogenesis/genetics , alpha Karyopherins/deficiency , alpha Karyopherins/genetics
14.
Nat Commun ; 2: 156, 2011 Jan 18.
Article En | MEDLINE | ID: mdl-21245837

Influenza A viruses are a threat to humans due to their ability to cross species barriers, as illustrated by the 2009 H1N1v pandemic and sporadic H5N1 transmissions. Interspecies transmission requires adaptation of the viral polymerase to importin-α, a cellular protein that mediates transport into the nucleus where transcription and replication of the viral genome takes place. In this study, we analysed replication, host specificity and pathogenicity of avian and mammalian influenza viruses, in importin-α-silenced cells and importin-α-knockout mice, to understand the role of individual importin-α isoforms in adaptation. For efficient virus replication, the polymerase subunit PB2 and the nucleoprotein (NP) of avian viruses required importin-α3, whereas PB2 and NP of mammalian viruses showed importin-α7 specificity. H1N1v replication depended on both, importin-α3 and -α7, suggesting ongoing adaptation of this virus. Thus, differences in importin-α specificity are determinants of host range underlining the importance of the nuclear envelope in interspecies transmission.

...