Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
PLoS Biol ; 20(10): e3001811, 2022 10.
Article in English | MEDLINE | ID: mdl-36215313

ABSTRACT

Nuclear envelope membrane proteins (NEMPs) are a conserved family of nuclear envelope (NE) proteins that reside within the inner nuclear membrane (INM). Even though Nemp1 knockout (KO) mice are overtly normal, they display a pronounced splenomegaly. This phenotype and recent reports describing a requirement for NE openings during erythroblasts terminal maturation led us to examine a potential role for Nemp1 in erythropoiesis. Here, we report that Nemp1 KO mice show peripheral blood defects, anemia in neonates, ineffective erythropoiesis, splenomegaly, and stress erythropoiesis. The erythroid lineage of Nemp1 KO mice is overrepresented until the pronounced apoptosis of polychromatophilic erythroblasts. We show that NEMP1 localizes to the NE of erythroblasts and their progenitors. Mechanistically, we discovered that NEMP1 accumulates into aggregates that localize near or at the edge of NE openings and Nemp1 deficiency leads to a marked decrease of both NE openings and ensuing enucleation. Together, our results for the first time demonstrate that NEMP1 is essential for NE openings and erythropoietic maturation in vivo and provide the first mouse model of defective erythropoiesis directly linked to the loss of an INM protein.


Subject(s)
Nuclear Envelope , Splenomegaly , Mice , Animals , Erythroblasts/metabolism , Cell Nucleus/metabolism , Erythropoiesis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout
2.
FASEB J ; 34(8): 10242-10249, 2020 08.
Article in English | MEDLINE | ID: mdl-32539195

ABSTRACT

The nuclei of cone photoreceptors are located on the apical side of the outer nuclear layer (ONL) in vertebrate retinas. However, the functional role of this evolutionarily conserved localization of cone nuclei is unknown. We previously showed that Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) are essential for the apical migration of cone nuclei during development. Here, we developed an efficient genetic strategy to disrupt cone LINC complexes in mice. Experiments with animals from both sexes revealed that disrupting cone LINC complexes resulted in mislocalization of cone nuclei to the basal side of ONL in mouse retina. This, in turn, disrupted cone pedicle morphology, and appeared to reduce the efficiency of synaptic transmission from cones to bipolar cells. Although we did not observe other developmental or phototransduction defects in cones with mislocalized nuclei, their dark adaptation was impaired, consistent with a deficiency in chromophore recycling. These findings demonstrate that the apical localization of cone nuclei in the ONL is required for the timely dark adaptation and efficient synaptic transmission in cone photoreceptors.


Subject(s)
Cell Nucleus/physiology , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Animals , Cytoskeleton/physiology , Dark Adaptation/physiology , Female , Male , Mice
3.
Genesis ; 54(3): 129-35, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26789558

ABSTRACT

The developing mouse retina is a tractable model for studying neurogenesis and differentiation. Although transgenic Cre mouse lines exist to mediate conditional genetic manipulations in developing mouse retinas, none of them act specifically in early developing rods. For conditional genetic manipulations of developing retinas, a Nrl-Cre mouse line in which the Nrl promoter drives expression of Cre in rod precursors was created. The results showed that Nrl-Cre expression was specific to the retina where it drives rod-specific recombination with a temporal pattern similar to endogenous Nrl expression during retinal development. This Nrl-Cre transgene does not negatively impact retinal structure and function. Taken together, the data suggested that the Nrl-Cre mouse line was a valuable tool to drive Cre-mediated recombination specifically in developing rods.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Eye Proteins/genetics , Integrases/metabolism , Neurogenesis , Recombination, Genetic , Retinal Rod Photoreceptor Cells/cytology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Eye Proteins/metabolism , Integrases/genetics , Mice , Mice, Transgenic , Promoter Regions, Genetic , Retina
4.
Neurobiol Dis ; 78: 57-67, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25843669

ABSTRACT

Nonsense mutations across the whole coding sequence of Syne1/Nesprin1 have been linked to autosomal recessive cerebellar ataxia Type I (ARCA1). However, nothing is known about the molecular etiology of this late-onset debilitating pathology. In this work, we report that Nesprin1 giant is specifically expressed in CNS tissues. We also identified a CNS-specific splicing event that leads to the abundant expression of a KASH-LESS variant of Nesprin1 giant (KLNes1g) in the cerebellum. KLNes1g displayed a noncanonical localization at glomeruli of cerebellar mossy fibers whereas Nesprin2 exclusively decorated the nuclear envelope of all cerebellar neurons. In immunogold electron microscopy, KLNes1g colocalized both with synaptic vesicles within mossy fibers and with dendritic membranes of cerebellar granule neurons. We further identified vesicle- and membrane-associated proteins in KLNes1g immunoprecipitates. Together, our results suggest that the loss of function of KLNes1g resulting from Nesprin1 nonsense mutations underlies the molecular etiology of ARCA1.


Subject(s)
Cerebellum/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/metabolism , Animals , Brain/metabolism , Cerebellum/ultrastructure , Cytoskeletal Proteins , Mice , Nerve Fibers/metabolism , Nerve Fibers/ultrastructure , Neurons/metabolism , Neurons/ultrastructure
5.
Genesis ; 52(4): 359-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24550182

ABSTRACT

Migration and anchorage of nuclei within developing and adult tissues rely on Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes). These macromolecular assemblies span the nuclear envelope and physically couple chromatin and nuclear lamina to cytoplasmic cytoskeletal networks. LINC complexes assemble within the perinuclear space through direct interactions between the respective evolutionary-conserved SUN and KASH domains of Sun proteins, which reside within the inner nuclear membrane, and Nesprins, which reside within the outer nuclear membrane. Here, we describe and validate a dominant-negative transgenic strategy allowing for the disruption of endogenous SUN/KASH interactions through the inducible expression of a recombinant KASH domain. Our approach, which is based on the Cre/Lox system, allows for the targeted disruption of LINC complexes in a wide array of mouse tissues or specific cell types thereof and bypasses the perinatal lethality and potential cell nonautonomous effects of current mouse models based on germline inactivation of genes encoding Sun proteins and Nesprins. For these reasons, this mouse model provides a useful tool to evaluate the physiological relevance of LINC complexes integrity during development and homeostasis in a wide array of mammalian tissues.


Subject(s)
Multiprotein Complexes/metabolism , Animals , Cell Line , Female , Gene Expression , Gene Expression Regulation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Integrases/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Organ Specificity
6.
Adv Exp Med Biol ; 773: 471-90, 2014.
Article in English | MEDLINE | ID: mdl-24563361

ABSTRACT

Hauling and anchoring the nucleus within immobile or motile cells, tissues, and/or syncytia represents a major challenge. In the past 15 years, Linkers of the Nucleoskeleton to the Cytoskeleton (LINC complexes) have emerged as evolutionary-conserved molecular devices that span the nuclear envelope and provide interacting interfaces for cytoskeletal networks and molecular motors to the nuclear envelope. Here, we review the molecular composition of LINC complexes and focus on how their genetic alteration in vivo has provided a wealth of information related to the relevance of nuclear positioning during tissue development and homeostasis with a special emphasis on the central nervous system. As it may be relevant for metastasis in a range of cancers, the involvement of LINC complexes in migration of nonneuronal cells via its interaction with the perinuclear actin cap will also be developed.


Subject(s)
Cell Movement/physiology , Nuclear Envelope/physiology , Cell Adhesion , Humans , Single-Cell Analysis
7.
Nat Commun ; 14(1): 1111, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36849521

ABSTRACT

Transcription factors (TFs) are transported from the cytoplasm to the nucleus and disappear from the nucleus after they regulate gene expression. Here, we discover an unconventional nuclear export of the TF, orthodenticle homeobox 2 (OTX2), in nuclear budding vesicles, which transport OTX2 to the lysosome. We further find that torsin1a (Tor1a) is responsible for scission of the inner nuclear vesicle, which captures OTX2 using the LINC complex. Consistent with this, in cells expressing an ATPase-inactive Tor1aΔE mutant and the LINC (linker of nucleoskeleton and cytoskeleton) breaker KASH2, OTX2 accumulated and formed aggregates in the nucleus. Consequently, in the mice expressing Tor1aΔE and KASH2, OTX2 could not be secreted from the choroid plexus for transfer to the visual cortex, leading to failed development of parvalbumin neurons and reduced visual acuity. Together, our results suggest that unconventional nuclear egress and secretion of OTX2 are necessary not only to induce functional changes in recipient cells but also to prevent aggregation in donor cells.


Subject(s)
Cell Nucleus , Genes, Homeobox , Animals , Mice , Lysosomes , Cell Division , Nuclear Matrix , Blister
8.
J Cell Biol ; 222(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37071483

ABSTRACT

The Hippo pathway is a conserved and critical regulator of tissue growth. The FERM protein Expanded is a key signaling hub that promotes activation of the Hippo pathway, thereby inhibiting the transcriptional co-activator Yorkie. Previous work identified the polarity determinant Crumbs as a primary regulator of Expanded. Here, we show that the giant cadherin Fat also regulates Expanded directly and independently of Crumbs. We show that direct binding between Expanded and a highly conserved region of the Fat cytoplasmic domain recruits Expanded to the apicolateral junctional zone and stabilizes Expanded. In vivo deletion of Expanded binding regions in Fat causes loss of apical Expanded and promotes tissue overgrowth. Unexpectedly, we find Fat can bind its ligand Dachsous via interactions of their cytoplasmic domains, in addition to the known extracellular interactions. Importantly, Expanded is stabilized by Fat independently of Dachsous binding. These data provide new mechanistic insights into how Fat regulates Expanded, and how Hippo signaling is regulated during organ growth.


Subject(s)
Cell Adhesion Molecules , Drosophila Proteins , Drosophila melanogaster , Hippo Signaling Pathway , Membrane Proteins , Animals , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism
9.
Cardiovasc Res ; 119(11): 2074-2088, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37067297

ABSTRACT

AIMS: Nuclear envelope integrity is essential for the compartmentalization of the nucleus and cytoplasm. Importantly, mutations in genes encoding nuclear envelope (NE) and associated proteins are the second highest cause of familial dilated cardiomyopathy. One such NE protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in the heart remains poorly understood. METHODS AND RESULTS: We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or in adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue, and cellular analyses. High-resolution episcopic microscopy was used for three-dimensional reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided a physiological assessment of Lem2 iCKO adult mice. We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels. Conversely, reducing Lem2 levels to ∼45% in adult cardiomyocytes did not lead to overt cardiac dysfunction up to 18 months of age. CONCLUSIONS: Our data suggest that Lem2 is critical for integrity at the nascent NE in foetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by partial Lem2 depletion, perhaps owing to a more established NE and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.


Subject(s)
Nuclear Envelope , Nuclear Proteins , Animals , Humans , Mice , DNA Damage , Heart , Mutation , Myocytes, Cardiac/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics
10.
J Cell Biol ; 178(5): 785-98, 2007 Aug 27.
Article in English | MEDLINE | ID: mdl-17724119

ABSTRACT

Sun1 and 2 are A-type lamin-binding proteins that, in association with nesprins, form a link between the inner nuclear membranes (INMs) and outer nuclear membranes of mammalian nuclear envelopes. Both immunofluorescence and immunoelectron microscopy reveal that Sun1 but not Sun2 is intimately associated with nuclear pore complexes (NPCs). Topological analyses indicate that Sun1 is a type II integral protein of the INM. Localization of Sun1 to the INM is defined by at least two discrete regions within its nucleoplasmic domain. However, association with NPCs is dependent on the synergy of both nucleoplasmic and lumenal domains. Cells that are either depleted of Sun1 by RNA interference or that overexpress dominant-negative Sun1 fragments exhibit clustering of NPCs. The implication is that Sun1 represents an important determinant of NPC distribution across the nuclear surface.


Subject(s)
Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Nuclear Proteins/metabolism , Amino Acid Motifs , Animals , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Structure, Quaternary , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
11.
Proc Natl Acad Sci U S A ; 106(45): 19017-22, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19850871

ABSTRACT

Defects in nuclear morphology often correlate with the onset of disease, including cancer, progeria, cardiomyopathy, and muscular dystrophy. However, the mechanism by which a cell controls its nuclear shape is unknown. Here, we use adhesive micropatterned surfaces to control the overall shape of fibroblasts and find that the shape of the nucleus is tightly regulated by the underlying cell adhesion geometry. We found that this regulation occurs through a dome-like actin cap that covers the top of the nucleus. This cap is composed of contractile actin filament bundles containing phosphorylated myosin, which form a highly organized, dynamic, and oriented structure in a wide variety of cells. The perinuclear actin cap is specifically disorganized or eliminated by inhibition of actomyosin contractility and rupture of the LINC complexes, which connect the nucleus to the actin cap. The organization of this actin cap and its nuclear shape-determining function are disrupted in cells from mouse models of accelerated aging (progeria) and muscular dystrophy with distorted nuclei caused by alterations of A-type lamins. These results highlight the interplay between cell shape, nuclear shape, and cell adhesion mediated by the perinuclear actin cap.


Subject(s)
Actins/metabolism , Cell Adhesion/physiology , Cell Nucleus Shape/physiology , Cell Shape/physiology , Myosins/metabolism , Animals , Mice , Microscopy, Fluorescence , Muscular Dystrophies/pathology , Progeria/pathology
12.
Hum Mol Genet ; 18(4): 607-20, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19008300

ABSTRACT

Mutations in the gene encoding the inner nuclear membrane proteins lamins A and C produce cardiac and skeletal muscle dysfunction referred to as Emery Dreifuss muscular dystrophy. Lamins A and C participate in the LINC complex that, along with the nesprin and SUN proteins, LInk the Nucleoskeleton with the Cytoskeleton. Nesprins 1 and 2 are giant spectrin-repeat containing proteins that have large and small forms. The nesprins contain a transmembrane anchor that tethers to the nuclear membrane followed by a short domain that resides within the lumen between the inner and outer nuclear membrane. Nesprin's luminal domain binds directly to SUN proteins. We generated mice where the C-terminus of nesprin-1 was deleted. This strategy produced a protein lacking the transmembrane and luminal domains that together are referred to as the KASH domain. Mice homozygous for this mutation exhibit lethality with approximately half dying at or near birth from respiratory failure. Surviving mice display hindlimb weakness and an abnormal gait. With increasing age, kyphoscoliosis, muscle pathology and cardiac conduction defects develop. The protein components of the LINC complex, including mutant nesprin-1alpha, lamin A/C and SUN2, are localized at the nuclear membrane in this model. However, the LINC components do not normally associate since coimmunoprecipitation experiments with SUN2 and nesprin reveal that mutant nesprin-1 protein no longer interacts with SUN2. These findings demonstrate the role of the LINC complex, and nesprin-1, in neuromuscular and cardiac disease.


Subject(s)
Gene Silencing , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Cytoskeletal Proteins , Disease Models, Animal , Female , Humans , Lamins/genetics , Lamins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Emery-Dreifuss/embryology , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/pathology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Phenotype , Protein Binding , Protein Structure, Tertiary
13.
Biochem Soc Trans ; 39(6): 1790-4, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22103527

ABSTRACT

Appropriate tissue morphogenesis strictly requires the developmental regulation of different types of nuclear movements. LINC (linker of nucleoskeleton and cytoskeleton) complexes are macromolecular scaffolds that span the nuclear envelope and physically connect the nuclear interior to different cytoskeletal elements and molecular motors, thereby playing essential roles in nucleokinesis. Recent studies dedicated to the in vivo disruption of LINC complexes not only confirmed their widespread role in nuclear dynamics, but also led to a vigorous regain of interest in the physiological relevance of nuclear positioning within cells and syncitia. In the present paper, we review the results of LINC complex disruption in vivo across different organisms and the potential implications of observed phenotypes in human diseases.


Subject(s)
Multiprotein Complexes/metabolism , Nuclear Envelope/metabolism , Animals , Central Nervous System/metabolism , Humans , Muscle, Skeletal/metabolism
14.
J Cell Biol ; 172(1): 41-53, 2006 Jan 02.
Article in English | MEDLINE | ID: mdl-16380439

ABSTRACT

The nuclear envelope defines the barrier between the nucleus and cytoplasm and features inner and outer membranes separated by a perinuclear space (PNS). The inner nuclear membrane contains specific integral proteins that include Sun1 and Sun2. Although the outer nuclear membrane (ONM) is continuous with the endoplasmic reticulum, it is nevertheless enriched in several integral membrane proteins, including nesprin 2 Giant (nesp2G), an 800-kD protein featuring an NH(2)-terminal actin-binding domain. A recent study (Padmakumar, V.C., T. Libotte, W. Lu, H. Zaim, S. Abraham, A.A. Noegel, J. Gotzmann, R. Foisner, and I. Karakesisoglou. 2005. J. Cell Sci. 118:3419-3430) has shown that localization of nesp2G to the ONM is dependent upon an interaction with Sun1. In this study, we confirm and extend these results by demonstrating that both Sun1 and Sun2 contribute to nesp2G localization. Codepletion of both of these proteins in HeLa cells leads to the loss of ONM-associated nesp2G, as does overexpression of the Sun1 lumenal domain. Both treatments result in the expansion of the PNS. These data, together with those of Padmakumar et al. (2005), support a model in which Sun proteins tether nesprins in the ONM via interactions spanning the PNS. In this way, Sun proteins and nesprins form a complex that links the nucleoskeleton and cytoskeleton (the LINC complex).


Subject(s)
Cell Nucleus/physiology , Cytoplasm/physiology , Multiprotein Complexes/physiology , Nuclear Envelope/chemistry , Amino Acid Sequence , Animals , Cells, Cultured , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/analysis , Macromolecular Substances/metabolism , Membrane Proteins/analysis , Mice , Microfilament Proteins/analysis , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Nuclear Envelope/physiology , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Protein Structure, Tertiary/physiology , Sequence Alignment
15.
Biol Open ; 10(6)2021 06 15.
Article in English | MEDLINE | ID: mdl-34100899

ABSTRACT

Genetic studies have linked FAT1 (FAT atypical cadherin 1) with autism spectrum disorder (ASD); however, the role that FAT1 plays in ASD remains unknown. In mice, the function of Fat1 has been primarily implicated in embryonic nervous system development with less known about its role in postnatal development. We show for the first time that FAT1 protein is expressed in mouse postnatal brains and is enriched in the cerebellum, where it localizes to granule neurons and Golgi cells in the granule layer, as well as inhibitory neurons in the molecular layer. Furthermore, subcellular characterization revealed FAT1 localization in neurites and soma of granule neurons, as well as being present in the synaptic plasma membrane and postsynaptic densities. Interestingly, FAT1 expression was decreased in induced pluripotent stem cell (iPSC)-derived neural precursor cells (NPCs) from individuals with ASD. These findings suggest a novel role for FAT1 in postnatal development and may be particularly important for cerebellum function. As the cerebellum is one of the vulnerable brain regions in ASD, our study warrants further investigation of FAT1 in the disease etiology.


Subject(s)
Autistic Disorder/etiology , Cadherins/genetics , Disease Susceptibility , Animals , Autistic Disorder/metabolism , Biomarkers , Cadherins/metabolism , Cerebellum/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Hippocampus/metabolism , Induced Pluripotent Stem Cells/metabolism , Interneurons/metabolism , Mice , Neurites/metabolism , Protein Transport , Transcriptome
16.
Cell Death Discov ; 6(1): 90, 2020.
Article in English | MEDLINE | ID: mdl-33024575

ABSTRACT

The canonical function of Bcl-2 family proteins is to regulate mitochondrial membrane integrity. In response to apoptotic signals the multi-domain pro-apoptotic proteins Bax and Bak are activated and perforate the mitochondrial outer membrane by a mechanism which is inhibited by their interaction with pro-survival members of the family. However, other studies have shown that Bax and Bak may have additional, non-canonical functions, which include stress-induced nuclear envelope rupture and discharge of nuclear proteins into the cytosol. We show here that the apoptotic stimuli cisplatin and staurosporine induce a Bax/Bak-dependent degradation and subcellular redistribution of nesprin-1 and nesprin-2 but not nesprin-3, of the linker of nucleoskeleton and cytoskeleton (LINC) complex. The degradation and redistribution were caspase-independent and did not occur in Bax/Bak double knockout (DKO) mouse embryo fibroblasts (MEFs). Re-expression of Bax in Bax/Bak DKO MEFs restored stress-induced redistribution of nesprin-2 by a mechanism which requires Bax membrane localization and integrity of the α helices 5/6, and the Bcl-2 homology 3 (BH3) domain. We found that nesprin-2 interacts with Bax in close proximity to perinuclear mitochondria in mouse and human cells. This interaction requires the mitochondrial targeting and N-terminal region but not the BH3 domain of Bax. Our results identify nesprin-2 as a Bax binding partner and also a new function of Bax in impairing the integrity of the LINC complex.

17.
Sci Adv ; 6(35): eabb4591, 2020 08.
Article in English | MEDLINE | ID: mdl-32923640

ABSTRACT

Human genome-wide association studies have linked single-nucleotide polymorphisms (SNPs) in NEMP1 (nuclear envelope membrane protein 1) with early menopause; however, it is unclear whether NEMP1 has any role in fertility. We show that whole-animal loss of NEMP1 homologs in Drosophila, Caenorhabditis elegans, zebrafish, and mice leads to sterility or early loss of fertility. Loss of Nemp leads to nuclear shaping defects, most prominently in the germ line. Biochemical, biophysical, and genetic studies reveal that NEMP proteins support the mechanical stiffness of the germline nuclear envelope via formation of a NEMP-EMERIN complex. These data indicate that the germline nuclear envelope has specialized mechanical properties and that NEMP proteins play essential and conserved roles in fertility.

18.
Biophys J ; 95(11): 5462-75, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18790843

ABSTRACT

Laminopathies encompass a wide array of human diseases associated to scattered mutations along LMNA, a single gene encoding A-type lamins. How such genetic alterations translate to cellular defects and generate such diverse disease phenotypes remains enigmatic. Recent work has identified nuclear envelope proteins--emerin and the linker of the nucleoskeleton and cytoskeleton (LINC) complex--which connect the nuclear lamina to the cytoskeleton. Here we quantitatively examine the composition of the nuclear envelope, as well as the architecture and functions of the cytoskeleton in cells derived from two laminopathic mouse models, including Hutchinson-Gilford progeria syndrome (Lmna(L530P/L530P)) and Emery-Dreifuss muscular dystrophy (Lmna(-/-)). Cells derived from the overtly aphenotypical model of X-linked Emery-Dreifuss muscular dystrophy (Emd(-/y)) were also included. We find that the centrosome is detached from the nucleus, preventing centrosome polarization in cells under flow--defects that are mediated by the loss of emerin from the nuclear envelope. Moreover, while basal actin and focal adhesion structure are mildly affected, RhoA activation, cell-substratum adhesion, and cytoplasmic elasticity are greatly lowered, exclusively in laminopathic models in which the LINC complex is disrupted. These results indicate a new function for emerin in cell polarization and suggest that laminopathies are not directly associated with cells' inability to polarize, but rather with cytoplasmic softening and weakened adhesion mediated by the disruption of the LINC complex across the nuclear envelope.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Microtubules/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Progeria/pathology , Animals , Biomechanical Phenomena , Cell Adhesion , Cell Line , Cell Movement , Cytoplasm/metabolism , Cytoskeleton/metabolism , Disease Models, Animal , Humans , Membrane Proteins/metabolism , Mice , Muscular Dystrophy, Emery-Dreifuss/genetics , Muscular Dystrophy, Emery-Dreifuss/metabolism , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Phenotype , Progeria/genetics , Progeria/metabolism , Rats , rhoA GTP-Binding Protein/metabolism
19.
Methods Mol Biol ; 1840: 25-33, 2018.
Article in English | MEDLINE | ID: mdl-30141035

ABSTRACT

The biochemical characterization of proteins most often require their identification by immunoblotting. Whereas SDS-PAGE provides satisfactory results for most proteins, the identification of larger proteins requires alternative methods to ensure their separation and complete transfer onto nitrocellulose membranes. Here, we describe the application of vertical agarose gel electrophoresis to identify large isoforms of nesprin-1 and nesprin-2.


Subject(s)
Electrophoresis, Agar Gel , Molecular Weight , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Animals , Cytoskeletal Proteins , Immunoprecipitation , Mice , Protein Isoforms
20.
Sci Transl Med ; 10(460)2018 09 26.
Article in English | MEDLINE | ID: mdl-30257952

ABSTRACT

Hutchinson-Gilford progeria syndrome is a disorder of premature aging in children caused by de novo mutations in LMNA that lead to the synthesis of an internally truncated form of prelamin A (commonly called progerin). The production of progerin causes multiple disease phenotypes, including an unusual vascular phenotype characterized by the loss of smooth muscle cells in the arterial media and fibrosis of the adventitia. We show that progerin expression, combined with mechanical stress, promotes smooth muscle cell death. Disrupting the linker of the nucleoskeleton and cytoskeleton (LINC) complex in smooth muscle cells ameliorates the toxic effects of progerin on smooth muscle cells and limits the accompanying adventitial fibrosis.


Subject(s)
Aortic Diseases/complications , Multiprotein Complexes/metabolism , Myocytes, Smooth Muscle/metabolism , Progeria/complications , Progeria/metabolism , Adventitia/metabolism , Adventitia/pathology , Animals , Aorta/metabolism , Aorta/pathology , Cell Death , Cells, Cultured , Collagen Type VIII/biosynthesis , Disease Models, Animal , Lamin Type A/metabolism , Lamin Type B/metabolism , Mice , Myocytes, Smooth Muscle/ultrastructure , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL