Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 444
1.
Commun Biol ; 7(1): 640, 2024 May 25.
Article En | MEDLINE | ID: mdl-38796645

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common autosomal dominant muscle disorders, yet no cure or amelioration exists. The clinical presentation is diverse, making it difficult to identify the actual driving pathomechanism among many downstream events. To unravel this complexity, we performed a meta-analysis of 13 original omics datasets (in total 171 FSHD and 129 control samples). Our approach confirmed previous findings about the disease pathology and specified them further. We confirmed increased expression of former proposed DUX4 biomarkers, and furthermore impairment of the respiratory chain. Notably, the meta-analysis provides insights about so far not reported pathways, including misregulation of neuromuscular junction protein encoding genes, downregulation of the spliceosome, and extensive alterations of nuclear envelope protein expression. Finally, we developed a publicly available shiny app to provide a platform for researchers who want to search our analysis for genes of interest in the future.


Muscular Dystrophy, Facioscapulohumeral , Neuromuscular Junction , Nuclear Envelope , Spliceosomes , Humans , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Nuclear Envelope/metabolism , Nuclear Envelope/genetics , Spliceosomes/metabolism , Spliceosomes/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Gene Expression Regulation
2.
J Clin Invest ; 134(7)2024 Feb 01.
Article En | MEDLINE | ID: mdl-38300705

Stromal interaction molecule 1 (STIM1) is a Ca2+ sensor located in the sarcoplasmic reticulum (SR) of skeletal muscle, where it is best known for its role in store-operated Ca2+ entry (SOCE). Genetic syndromes resulting from STIM1 mutations are recognized as a cause of muscle weakness and atrophy. Here, we focused on a gain-of-function mutation that occurs in humans and mice (STIM1+/D84G mice), in which muscles exhibited constitutive SOCE. Unexpectedly, this constitutive SOCE did not affect global Ca2+ transients, SR Ca2+ content, or excitation-contraction coupling (ECC) and was therefore unlikely to underlie the reduced muscle mass and weakness observed in these mice. Instead, we demonstrate that the presence of D84G STIM1 in the nuclear envelope of STIM1+/D84G muscle disrupted nuclear-cytosolic coupling, causing severe derangement in nuclear architecture, DNA damage, and altered lamina A-associated gene expression. Functionally, we found that D84G STIM1 reduced the transfer of Ca2+ from the cytosol to the nucleus in myoblasts, resulting in a reduction of [Ca2+]N. Taken together, we propose a novel role for STIM1 in the nuclear envelope that links Ca2+ signaling to nuclear stability in skeletal muscle.


Muscle Weakness , Nuclear Envelope , Stromal Interaction Molecule 1 , Animals , Humans , Mice , Calcium/metabolism , Calcium Signaling , Muscle Weakness/genetics , Muscle Weakness/metabolism , Muscle, Skeletal/metabolism , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
4.
C R Biol ; 346: 89-93, 2023 Sep 21.
Article En | MEDLINE | ID: mdl-37779383

The nucleus has been viewed as a passenger during cell migration that functions merely to protect the genome. However, increasing evidence shows that the nucleus is an active organelle, constantly sensing the surrounding environment and translating extracellular mechanical inputs into intracellular signaling. The nuclear envelope has a large membrane reservoir which serves as a buffer for mechanical inputs as it unfolds without increasing its tension. In contrast, when cells cope with mechanical strain, such as migration through solid tumors or dense interstitial spaces, the nuclear envelope folds stretch, increasing nuclear envelope tension and sometimes causing rupture. Different degrees of nuclear envelope tension regulate cellular behaviors and functions, especially in cells that move and grow within dense matrices. The crosstalk between extracellular mechanical inputs and the cell nucleus is a critical component in the modulation of cell function of cells that navigate within packed microenvironments. Moreover, there is a link between regimes of nuclear envelope unfolding and different cellular behaviors, from orchestrated signaling cascades to cellular perturbations and damage.


Le noyau a longtemps été considéré comme un passager lors de la migration cellulaire, servant simplement à protéger le génome. Cependant, de plus en plus de preuves montrent que le noyau est un organite actif, qui sonde le milieu environnant et traduit les entrées mécaniques extracellulaires en signalisation intracellulaire. L'enveloppe nucléaire possède un grand réservoir membranaire qui sert de tampon face aux entrées mécaniques en se dépliant sans augmenter sa tension. En revanche, lorsque les cellules font face à des contraintes mécaniques, telles que la migration au travers de tumeurs solides ou despaces interstitiels denses, les plis de l'enveloppe nucléaire s'étirent, augmentant sa tension et provoquant parfois sa rupture. Différents degrés de tension de l'enveloppe nucléaire régulent les comportements et les fonctions cellulaires, en particulier des cellules qui se déplacent et se développent dans des matrices denses. La signalisation croisée entre les entrées mécaniques extracellulaires et le noyau cellulaire sont des composants essentiels dans la modulation de la fonction des cellules qui naviguent dans des microenvironnements encombrés. De plus, il existe un lien entre les régimes de déploiement de l'enveloppe nucléaire et les différents comportements cellulaires, allant des cascades de signalisation jusquaux perturbations et dommages cellulaires.


Neoplasms , Nuclear Envelope , Humans , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Cell Movement , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Tumor Microenvironment
5.
Plant Cell ; 35(12): 4284-4303, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-37738557

The nucleoskeleton forms a filamentous meshwork under the nuclear envelope and contributes to the regulation of nuclear shape and gene expression. To understand how the Arabidopsis (Arabidopsis thaliana) nucleoskeleton physically connects to the nuclear periphery in plants, we investigated the Arabidopsis nucleoskeleton protein KAKU4 and sought for functional regions responsible for its localization at the nuclear periphery. We identified 3 conserved peptide motifs within the N-terminal region of KAKU4, which are required for intermolecular interactions of KAKU4 with itself, interaction with the nucleoskeleton protein CROWDED NUCLEI (CRWN), localization at the nuclear periphery, and nuclear elongation in differentiated tissues. Unexpectedly, we find these motifs to be present also in NUP82 and NUP136, 2 plant-specific nucleoporins from the nuclear pore basket. We further show that NUP82, NUP136, and KAKU4 have a common evolutionary history predating nonvascular land plants with KAKU4 mainly localizing outside the nuclear pore suggesting its divergence from an ancient nucleoporin into a new nucleoskeleton component. Finally, we demonstrate that both NUP82 and NUP136, through their shared N-terminal motifs, interact with CRWN and KAKU4 proteins revealing the existence of a physical continuum between the nuclear pore and the nucleoskeleton in plants.


Arabidopsis Proteins , Arabidopsis , Nuclear Pore/genetics , Nuclear Pore/metabolism , Arabidopsis/genetics , Arabidopsis/metabolism , Amino Acid Motifs , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Nuclear Matrix/metabolism
6.
J Cell Biol ; 222(8)2023 08 07.
Article En | MEDLINE | ID: mdl-37440179

Eukaryotic cell division involves the segregation of chromosomes between two daughter cells and must be coordinated with extensive rearrangement of their nuclear envelopes. In this issue, Saik et al. (2023 J. Cell Biol. https://doi.org/10.1083/jcb.202208137) show that a SUMOylation cascade at the inner nuclear membrane elevates the levels of phosphatidic acid, a key phospholipid precursor, to support the need for nuclear membrane expansion during mitosis.


Nuclear Envelope , Phosphatidic Acids , Sumoylation , Chromosomes , Mitosis , Nuclear Envelope/genetics
7.
PLoS Genet ; 19(7): e1010837, 2023 07.
Article En | MEDLINE | ID: mdl-37428798

Spermatozoa in animal species are usually highly elongated cells with a long motile tail attached to a head that contains the haploid genome in a compact and often elongated nucleus. In Drosophila melanogaster, the nucleus is compacted two hundred-fold in volume during spermiogenesis and re-modeled into a needle that is thirty-fold longer than its diameter. Nuclear elongation is preceded by a striking relocalization of nuclear pore complexes (NPCs). While NPCs are initially located throughout the nuclear envelope (NE) around the spherical nucleus of early round spermatids, they are later confined to one hemisphere. In the cytoplasm adjacent to this NPC-containing NE, the so-called dense complex with a strong bundle of microtubules is assembled. While this conspicuous proximity argued for functional significance of NPC-NE and microtubule bundle, experimental confirmation of their contributions to nuclear elongation has not yet been reported. Our functional characterization of the spermatid specific Mst27D protein now resolves this deficit. We demonstrate that Mst27D establishes physical linkage between NPC-NE and dense complex. The C-terminal region of Mst27D binds to the nuclear pore protein Nup358. The N-terminal CH domain of Mst27D, which is similar to that of EB1 family proteins, binds to microtubules. At high expression levels, Mst27D promotes bundling of microtubules in cultured cells. Microscopic analyses indicated co-localization of Mst27D with Nup358 and with the microtubule bundles of the dense complex. Time-lapse imaging revealed that nuclear elongation is accompanied by a progressive bundling of microtubules into a single elongated bundle. In Mst27D null mutants, this bundling process does not occur and nuclear elongation is abnormal. Thus, we propose that Mst27D permits normal nuclear elongation by promoting the attachment of the NPC-NE to the microtubules of the dense complex, as well as the progressive bundling of these microtubules.


Drosophila Proteins , Nuclear Pore , Male , Animals , Nuclear Pore/genetics , Nuclear Pore/metabolism , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Microtubules/metabolism , Spermatogenesis/genetics , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
8.
FEBS Lett ; 597(15): 1946-1956, 2023 08.
Article En | MEDLINE | ID: mdl-37339935

The nuclear envelope (NE) encloses the genetic material and functions in chromatin organization and stability. In Saccharomyces cerevisiae, the NE is bound to the ribosomal DNA (rDNA), highly repeated and transcribed, thus prone to genetic instability. While tethering limits instability, it simultaneously triggers notable NE remodeling. We posit here that NE remodeling may contribute to genome integrity maintenance. The NE importance in genome expression, structure, and integrity is well recognized, yet studies mostly focus on peripheral proteins and nuclear pores, not on the membrane itself. We recently characterized a NE invagination drastically obliterating the rDNA, which we propose here as a model to probe if and how membranes play an active role in genome stability preservation.


Nuclear Envelope , Nuclear Pore , Humans , Nuclear Envelope/genetics , Nuclear Pore/genetics , Nuclear Pore/metabolism , Genomic Instability , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism
9.
J Cell Biol ; 222(6)2023 06 05.
Article En | MEDLINE | ID: mdl-37154843

Nuclear pore complexes (NPCs) are embedded in the nuclear envelope and built from ∼30 different nucleoporins (Nups) in multiple copies, few are integral membrane proteins. One of these transmembrane nucleoporins, Ndc1, is thought to function in NPC assembly at the fused inner and outer nuclear membranes. Here, we show a direct interaction of Ndc1's transmembrane domain with Nup120 and Nup133, members of the pore membrane coating Y-complex. We identify an amphipathic helix in Ndc1's C-terminal domain binding highly curved liposomes. Upon overexpression, this amphipathic motif is toxic and dramatically alters the intracellular membrane organization in yeast. Ndc1's amphipathic motif functionally interacts with related motifs in the C-terminus of the nucleoporins Nup53 and Nup59, important for pore membrane binding and interconnecting NPC modules. The essential function of Ndc1 can be suppressed by deleting the amphipathic helix from Nup53. Our data indicate that nuclear membrane and presumably NPC biogenesis depends on a balanced ratio between amphipathic motifs in diverse nucleoporins.


Nuclear Envelope , Nuclear Pore Complex Proteins , Saccharomyces cerevisiae Proteins , Cell Membrane/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/chemistry
10.
Gene ; 870: 147423, 2023 Jun 20.
Article En | MEDLINE | ID: mdl-37044185

Lamin B2 (LMNB2), on the inner side of the nuclear envelope, constitutes the nuclear skeleton by connecting with other nuclear proteins. LMNB2 is involved in a wide range of nuclear functions, including DNA replication and stability, regulation of chromatin, and nuclear stiffness. Moreover, LMNB2 regulates several cellular processes, such as tissue development, cell cycle, cellular proliferation and apoptosis, chromatin localization and stability, and DNA methylation. Besides, the influence of abnormal expression and mutations of LMNB2 has been gradually discovered in cancers and laminopathies. Therefore, this review summarizes the recent advances of LMNB2-associated biological roles in physiological or pathological conditions, with a particular emphasis on cancers and laminopathies, as well as the potential mechanism of LMNB2 in related cancers.


Lamin Type B , Laminopathies , Neoplasms , Nuclear Proteins , Humans , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Lamin Type A/genetics , Lamin Type A/metabolism , Lamin Type B/genetics , Lamin Type B/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics , Laminopathies/metabolism , Neoplasms/metabolism
11.
J Cell Biol ; 222(7)2023 07 03.
Article En | MEDLINE | ID: mdl-37042812

The nuclear envelope (NE) is important in maintaining genome organization. The role of lipids in communication between the NE and telomere regulation was investigated, including how changes in lipid composition impact gene expression and overall nuclear architecture. Yeast was treated with the non-metabolizable lysophosphatidylcholine analog edelfosine, known to accumulate at the perinuclear ER. Edelfosine induced NE deformation and disrupted telomere clustering but not anchoring. Additionally, the association of Sir4 at telomeres decreased. RNA-seq analysis showed altered expression of Sir-dependent genes located at sub-telomeric (0-10 kb) regions, consistent with Sir4 dispersion. Transcriptomic analysis revealed that two lipid metabolic circuits were activated in response to edelfosine, one mediated by the membrane sensing transcription factors, Spt23/Mga2, and the other by a transcriptional repressor, Opi1. Activation of these transcriptional programs resulted in higher levels of unsaturated fatty acids and the formation of nuclear lipid droplets. Interestingly, cells lacking Sir proteins displayed resistance to unsaturated-fatty acids and edelfosine, and this phenotype was connected to Rap1.


Nuclear Envelope , Silent Information Regulator Proteins, Saccharomyces cerevisiae , Telomere , Membrane Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Phospholipid Ethers/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Telomere/genetics , Telomere/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Genes (Basel) ; 14(4)2023 03 23.
Article En | MEDLINE | ID: mdl-37107534

The nuclear envelope (NE) in eukaryotic cells is essential to provide a protective compartment for the genome. Beside its role in connecting the nucleus with the cytoplasm, the NE has numerous important functions including chromatin organization, DNA replication and repair. NE alterations have been linked to different human diseases, such as laminopathies, and are a hallmark of cancer cells. Telomeres, the ends of eukaryotic chromosomes, are crucial for preserving genome stability. Their maintenance involves specific telomeric proteins, repair proteins and several additional factors, including NE proteins. Links between telomere maintenance and the NE have been well established in yeast, in which telomere tethering to the NE is critical for their preservation and beyond. For a long time, in mammalian cells, except during meiosis, telomeres were thought to be randomly localized throughout the nucleus, but recent advances have uncovered close ties between mammalian telomeres and the NE that play important roles for maintaining genome integrity. In this review, we will summarize these connections, with a special focus on telomere dynamics and the nuclear lamina, one of the main NE components, and discuss the evolutionary conservation of these mechanisms.


Nuclear Envelope , Telomere , Animals , Humans , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Telomere/genetics , Telomere/metabolism , DNA Replication/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Meiosis , Mammals/genetics , Mammals/metabolism
13.
Cardiovasc Res ; 119(11): 2074-2088, 2023 09 05.
Article En | MEDLINE | ID: mdl-37067297

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.


Nuclear Envelope , Nuclear Proteins , Animals , Humans , Mice , DNA Damage , Heart , Mutation , Myocytes, Cardiac/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Proteins/genetics
14.
Hum Genet ; 142(4): 531-541, 2023 Apr.
Article En | MEDLINE | ID: mdl-36933034

One of the most severe forms of infertility in humans, caused by gametogenic failure, is non-obstructive azoospermia (NOA). Approximately, 20-30% of men with NOA may have single-gene mutations or other genetic variables that cause this disease. While a range of single-gene mutations associated with infertility has been identified in prior whole-exome sequencing (WES) studies, current insight into the precise genetic etiology of impaired human gametogenesis remains limited. In this paper, we described a proband with NOA who experienced hereditary infertility. WES analyses identified a homozygous variant in the SUN1 (Sad1 and UNC84 domain containing 1) gene [c. 663C > A: p.Tyr221X] that segregated with infertility. SUN1 encodes a LINC complex component essential for telomeric attachment and chromosomal movement. Spermatocytes with the observed mutations were incapable of repairing double-strand DNA breaks or undergoing meiosis. This loss of SUN1 functionality contributes to significant reductions in KASH5 levels within impaired chromosomal telomere attachment to the inner nuclear membrane. Overall, our results identify a potential genetic driver of NOA pathogenesis and provide fresh insight into the role of the SUN1 protein as a regulator of prophase I progression in the context of human meiosis.


Azoospermia , Nuclear Envelope , Male , Humans , Nuclear Envelope/genetics , Azoospermia/pathology , Microtubule-Associated Proteins/genetics , Spermatocytes/metabolism , Spermatocytes/pathology , Telomere/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
15.
J Cell Biol ; 222(5)2023 05 01.
Article En | MEDLINE | ID: mdl-36946995

Cytoplasmic dynein-driven movement of chromosomes during prophase I of mammalian meiosis is essential for synapsis and genetic exchange. Dynein connects to chromosome telomeres via KASH5 and SUN1 or SUN2, which together span the nuclear envelope. Here, we show that KASH5 promotes dynein motility in vitro, and cytosolic KASH5 inhibits dynein's interphase functions. KASH5 interacts with a dynein light intermediate chain (DYNC1LI1 or DYNC1LI2) via a conserved helix in the LIC C-terminal, and this region is also needed for dynein's recruitment to other cellular membranes. KASH5's N-terminal EF-hands are essential as the interaction with dynein is disrupted by mutation of key calcium-binding residues, although it is not regulated by cellular calcium levels. Dynein can be recruited to KASH5 at the nuclear envelope independently of dynactin, while LIS1 is essential for dynactin incorporation into the KASH5-dynein complex. Altogether, we show that the transmembrane protein KASH5 is an activating adaptor for dynein and shed light on the hierarchy of assembly of KASH5-dynein-dynactin complexes.


Cell Cycle Proteins , Cytoplasmic Dyneins , Dynactin Complex , Microtubule-Associated Proteins , Animals , Calcium/metabolism , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Dynactin Complex/genetics , Dynactin Complex/metabolism , Mammals/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Telomere/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism
16.
Nat Commun ; 14(1): 1135, 2023 02 28.
Article En | MEDLINE | ID: mdl-36854718

Partitioning of active gene loci to the nuclear envelope (NE) is a mechanism by which organisms increase the speed of adaptation and metabolic robustness to fluctuating resources in the environment. In the yeast Saccharomyces cerevisiae, adaptation to nutrient depletion or other stresses, manifests as relocalization of active gene loci from nucleoplasm to the NE, resulting in more efficient transport and translation of mRNA. The mechanism by which this partitioning occurs remains a mystery. Here, we demonstrate that the yeast inositol depletion-responsive gene locus INO1 partitions to the nuclear envelope, driven by local histone acetylation-induced polymer-polymer phase separation from the nucleoplasmic phase. This demixing is consistent with recent evidence for chromatin phase separation by acetylation-mediated dissolution of multivalent histone association and fits a physical model where increased bending stiffness of acetylated chromatin polymer causes its phase separation from de-acetylated chromatin. Increased chromatin spring stiffness could explain nucleation of transcriptional machinery at active gene loci.


Chromatin , Nuclear Envelope , Saccharomyces cerevisiae , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Histones/chemistry , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Histone Acetyltransferases/metabolism , Biopolymers/chemistry , Biopolymers/metabolism
17.
Sci Adv ; 9(6): eadd1453, 2023 02 10.
Article En | MEDLINE | ID: mdl-36753547

Interactions between chromosomes and LINC (linker of nucleoskeleton and cytoskeleton) complexes in the nuclear envelope (NE) promote homolog pairing and synapsis during meiosis. By tethering chromosomes to cytoskeletal motors, these connections lead to processive chromosome movements along the NE. This activity is usually mediated by telomeres, but in the nematode Caenorhabditis elegans, special chromosome regions called "pairing centers" (PCs) have acquired this meiotic function. Here, we identify a previously uncharacterized meiosis-specific NE protein, MJL-1 (MAJIN-Like-1), that is essential for interactions between PCs and LINC complexes in C. elegans. Mutations in MJL-1 eliminate active chromosome movements during meiosis, resulting in nonhomologous synapsis and impaired homolog pairing. Fission yeast and mice also require NE proteins to connect chromosomes to LINC complexes. Extensive similarities in the molecular architecture of meiotic chromosome-NE attachments across eukaryotes suggest a common origin and/or functions of this architecture during meiosis.


Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Mice , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Meiosis/genetics , Telomere/genetics , Telomere/metabolism , Chromosome Pairing , Membrane Proteins/metabolism
18.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article En | MEDLINE | ID: mdl-36768299

For the past several years, fundamental research on Sigma-1R (S1R) protein has unveiled its necessity for maintaining proper cellular homeostasis through modulation of calcium and lipid exchange between the endoplasmic reticulum (ER) and mitochondria, ER-stress response, and many other mechanisms. Most of these processes, such as ER-stress response and autophagy, have been associated with neuroprotective roles. In fact, improving these mechanisms using S1R agonists was beneficial in several brain disorders including neurodegenerative diseases. In this review, we will examine S1R subcellular localization and describe S1R-associated biological activity within these specific compartments, i.e., the Mitochondrion-Associated ER Membrane (MAM), ER-Lipid Droplet (ER-LD) interface, ER-Plasma Membreane (ER-PM) interface, and the Nuclear Envelope (NE). We also discussed how the dysregulation of these pathways contributes to neurodegenerative diseases, while highlighting the cellular mechanisms and key binding partners engaged in these processes.


Endoplasmic Reticulum , Mitochondria , Neurodegenerative Diseases , Neuroprotection , Receptors, sigma , Humans , Autophagy/genetics , Autophagy/physiology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Mitochondria/genetics , Mitochondria/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neuroprotection/genetics , Neuroprotection/physiology , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Receptors, sigma/genetics , Receptors, sigma/metabolism , Sigma-1 Receptor
19.
Trends Cell Biol ; 33(1): 18-29, 2023 01.
Article En | MEDLINE | ID: mdl-35778326

The nuclear envelope (NE) is central to the architecture of eukaryotic cells, both as a physical barrier separating the nucleus from the cytoplasm and as gatekeeper of selective transport between them. However, in open mitosis, the NE fragments to allow for spindle formation and segregation of chromosomes, resulting in intermixing of nuclear and cytoplasmic soluble fractions. Recent studies have shed new light on the mechanisms driving reinstatement of soluble proteome homeostasis following NE reformation in daughter cells. Here, we provide an overview of how mitotic cells confront this challenge to ensure continuity of basic cellular functions across generations and elaborate on the implications for the proteasome - a macromolecular machine that functions in both cytoplasmic and nuclear compartments.


Cell Nucleus , Proteostasis , Humans , Cell Nucleus/metabolism , Mitosis , Cytoplasm/metabolism , Nuclear Envelope/genetics
20.
Hum Mol Genet ; 32(2): 177-191, 2023 01 06.
Article En | MEDLINE | ID: mdl-35925868

Mutations in LMNA, the gene encoding A-type lamins, cause laminopathies-diseases of striated muscle and other tissues. The aetiology of laminopathies has been attributed to perturbation of chromatin organization or structural weakening of the nuclear envelope (NE) such that the nucleus becomes more prone to mechanical damage. The latter model requires a conduit for force transmission to the nucleus. NE-associated Linker of Nucleoskeleton and Cytoskeleton (LINC) complexes are one such pathway. Using clustered regularly interspaced short palindromic repeats to disrupt the Nesprin-1 KASH (Klarsicht, ANC-1, Syne Homology) domain, we identified this LINC complex protein as the predominant NE anchor for microtubule cytoskeleton components, including nucleation activities and motor complexes, in mouse cardiomyocytes. Loss of Nesprin-1 LINC complexes resulted in loss of microtubule cytoskeleton proteins at the nucleus and changes in nuclear morphology and positioning in striated muscle cells, but with no overt physiological defects. Disrupting the KASH domain of Nesprin-1 suppresses Lmna-linked cardiac pathology, likely by reducing microtubule cytoskeleton activities at the nucleus. Nesprin-1 LINC complexes thus represent a potential therapeutic target for striated muscle laminopathies.


Laminopathies , Muscle, Striated , Animals , Mice , Microtubule Proteins/metabolism , Nuclear Proteins/metabolism , Membrane Proteins/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Nuclear Matrix/genetics , Microtubules/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Intermediate Filament Proteins/metabolism , Muscle, Striated/metabolism , Laminopathies/metabolism
...