Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 610
Filter
1.
Nat Rev Mol Cell Biol ; 18(4): 229-245, 2017 04.
Article in English | MEDLINE | ID: mdl-28120913

ABSTRACT

As a compartment border, the nuclear envelope (NE) needs to serve as both a protective membrane shell for the genome and a versatile communication interface between the nucleus and the cytoplasm. Despite its important structural role in sheltering the genome, the NE is a dynamic and highly adaptable boundary that changes composition during differentiation, deforms in response to mechanical challenges, can be repaired upon rupture and even rapidly disassembles and reforms during open mitosis. NE remodelling is fundamentally involved in cell growth, division and differentiation, and if perturbed can lead to devastating diseases such as muscular dystrophies or premature ageing.


Subject(s)
Cell Differentiation , Mitosis , Nuclear Envelope/physiology , Active Transport, Cell Nucleus , Animals , Capsid/metabolism , Cell Differentiation/physiology , Cell Movement , Humans , Neutrophils/metabolism
2.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article in English | MEDLINE | ID: mdl-34290138

ABSTRACT

Nuclear envelope budding (NEB) is a recently discovered alternative pathway for nucleocytoplasmic communication distinct from the movement of material through the nuclear pore complex. Through quantitative electron microscopy and tomography, we demonstrate how NEB is evolutionarily conserved from early protists to human cells. In the yeast Saccharomyces cerevisiae, NEB events occur with higher frequency during heat shock, upon exposure to arsenite or hydrogen peroxide, and when the proteasome is inhibited. Yeast cells treated with azetidine-2-carboxylic acid, a proline analog that induces protein misfolding, display the most dramatic increase in NEB, suggesting a causal link to protein quality control. This link was further supported by both localization of ubiquitin and Hsp104 to protein aggregates and NEB events, and the evolution of these structures during heat shock. We hypothesize that NEB is part of normal cellular physiology in a vast range of species and that in S. cerevisiae NEB comprises a stress response aiding the transport of protein aggregates across the nuclear envelope.


Subject(s)
Azetidinecarboxylic Acid/toxicity , Heat-Shock Response , Nuclear Envelope/physiology , Protein Folding , Proteostasis/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/growth & development , Arsenites/toxicity , Hydrogen Peroxide/toxicity , Nuclear Envelope/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Sodium Compounds/toxicity , Ubiquitin/metabolism , Ubiquitination
3.
PLoS Biol ; 17(8): e3000388, 2019 08.
Article in English | MEDLINE | ID: mdl-31398189

ABSTRACT

Methods for measuring the properties of individual cells within their native 3D environment will enable a deeper understanding of embryonic development, tissue regeneration, and tumorigenesis. However, current methods for segmenting nuclei in 3D tissues are not designed for situations in which nuclei are densely packed, nonspherical, or heterogeneous in shape, size, or texture, all of which are true of many embryonic and adult tissue types as well as in many cases for cells differentiating in culture. Here, we overcome this bottleneck by devising a novel method based on labelling the nuclear envelope (NE) and automatically distinguishing individual nuclei using a tree-structured ridge-tracing method followed by shape ranking according to a trained classifier. The method is fast and makes it possible to process images that are larger than the computer's memory. We consistently obtain accurate segmentation rates of >90%, even for challenging images such as mid-gestation embryos or 3D cultures. We provide a 3D editor and inspector for the manual curation of the segmentation results as well as a program to assess the accuracy of the segmentation. We have also generated a live reporter of the NE that can be used to track live cells in 3 dimensions over time. We use this to monitor the history of cell interactions and occurrences of neighbour exchange within cultures of pluripotent cells during differentiation. We provide these tools in an open-access user-friendly format.


Subject(s)
Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Pattern Recognition, Automated/methods , Algorithms , Animals , Cell Nucleus/physiology , Fluorescent Dyes , Humans , Indoles , Lamin Type B , Nuclear Envelope/metabolism , Nuclear Envelope/physiology
4.
Hum Mol Genet ; 28(3): 351-371, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30239736

ABSTRACT

Laminopathies are diseases caused by dominant mutations in the human LMNA gene encoding A-type lamins. Lamins are intermediate filaments that line the inner nuclear membrane, provide structural support for the nucleus and regulate gene expression. Drosophila melanogaster models of skeletal muscle laminopathies were developed to investigate the pathological defects caused by mutant lamins and identify potential therapeutic targets. Human disease-causing LMNA mutations were modeled in Drosophila Lamin C (LamC) and expressed in indirect flight muscle (IFM). IFM-specific expression of mutant, but not wild-type LamC, caused held-up wings indicative of myofibrillar defects. Analyses of the muscles revealed cytoplasmic aggregates of nuclear envelope (NE) proteins, nuclear and mitochondrial dysmorphology, myofibrillar disorganization and up-regulation of the autophagy cargo receptor p62. We hypothesized that the cytoplasmic aggregates of NE proteins trigger signaling pathways that alter cellular homeostasis, causing muscle dysfunction. In support of this hypothesis, transcriptomics data from human muscle biopsy tissue revealed misregulation of the AMP-activated protein kinase (AMPK)/4E-binding protein 1 (4E-BP1)/autophagy/proteostatic pathways. Ribosomal protein S6K (S6K) messenger RNA (mRNA) levels were increased and AMPKα and mRNAs encoding downstream targets were decreased in muscles expressing mutant LMNA relative controls. The Drosophila laminopathy models were used to determine if altering the levels of these factors modulated muscle pathology. Muscle-specific over-expression of AMPKα and down-stream targets 4E-BP, Forkhead box transcription factors O (Foxo) and Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), as well as inhibition of S6K, suppressed the held-up wing phenotype, myofibrillar defects and LamC aggregation. These findings provide novel insights on mutant LMNA-based disease mechanisms and identify potential targets for drug therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Lamins/genetics , Lamins/physiology , AMP-Activated Protein Kinases/physiology , Animals , Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Lamin Type A/genetics , Lamin Type A/metabolism , Membrane Proteins/genetics , Models, Animal , Muscle, Skeletal/physiology , Mutation , Nuclear Envelope/metabolism , Nuclear Envelope/physiology , Peptide Initiation Factors/metabolism , Peptide Initiation Factors/physiology , Phenotype , Signal Transduction
5.
PLoS Comput Biol ; 16(10): e1008300, 2020 10.
Article in English | MEDLINE | ID: mdl-33035221

ABSTRACT

Large nuclear deformations during migration through confined spaces have been associated with nuclear membrane rupture and DNA damage. However, the stresses associated with nuclear damage remain unclear. Here, using a quasi-static plane strain finite element model, we map evolution of nuclear shape and stresses during confined migration of a cell through a deformable matrix. Plastic deformation of the nucleus observed for a cell with stiff nucleus transiting through a stiffer matrix lowered nuclear stresses, but also led to kinking of the nuclear membrane. In line with model predictions, transwell migration experiments with fibrosarcoma cells showed that while nuclear softening increased invasiveness, nuclear stiffening led to plastic deformation and higher levels of DNA damage. In addition to highlighting the advantage of nuclear softening during confined migration, our results suggest that plastic deformations of the nucleus during transit through stiff tissues may lead to bending-induced nuclear membrane disruption and subsequent DNA damage.


Subject(s)
Cell Movement/physiology , Cell Nucleus/physiology , Models, Biological , Cell Line, Tumor , DNA Damage , Finite Element Analysis , Humans , Nuclear Envelope/physiology
6.
Physiology (Bethesda) ; 33(1): 39-49, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29212891

ABSTRACT

Recent observations in laminopathy patient cells and cancer cells have revealed that the nuclear envelope (NE) can transiently rupture during interphase. NE rupture leads to an uncoordinated exchange of nuclear and cytoplasmic material, thereby deregulating cellular homeostasis. Moreover, concurrently inflicted DNA damage could prime rupture-prone cells for genome instability. Thus, NE rupture may represent a novel pathogenic mechanism that has far-reaching consequences for cell and organism physiology.


Subject(s)
Nuclear Envelope/physiology , Active Transport, Cell Nucleus , Animals , DNA Damage , Disease/etiology , Humans , Lamins/physiology , Stress, Mechanical
7.
Proc Natl Acad Sci U S A ; 113(32): E4671-80, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27462105

ABSTRACT

Inflammasomes are critical sensors that convey cellular stress and pathogen presence to the immune system by activating inflammatory caspases and cytokines such as IL-1ß. The nature of endogenous stress signals that activate inflammasomes remains unclear. Here we show that an inhibitor of the HIV aspartyl protease, Nelfinavir, triggers inflammasome formation and elicits an IL-1R-dependent inflammation in mice. We found that Nelfinavir impaired the maturation of lamin A, a structural component of the nuclear envelope, thereby promoting the release of DNA in the cytosol. Moreover, deficiency of the cytosolic DNA-sensor AIM2 impaired Nelfinavir-mediated inflammasome activation. These findings identify a pharmacologic activator of inflammasome and demonstrate the role of AIM2 in detecting endogenous DNA release upon perturbation of nuclear envelope integrity.


Subject(s)
Inflammasomes/drug effects , Nelfinavir/pharmacology , Nuclear Envelope/drug effects , Animals , CARD Signaling Adaptor Proteins/physiology , Caspase 1/metabolism , DNA/metabolism , Inflammasomes/physiology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Nuclear Envelope/physiology , Receptors, Interleukin-1/physiology
8.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28356536

ABSTRACT

Upon herpes simplex virus 1 (HSV-1) infection, the CD98 heavy chain (CD98hc) is redistributed around the nuclear membrane (NM), where it promotes viral de-envelopment during the nuclear egress of nucleocapsids. In this study, we attempted to identify the factor(s) involved in CD98hc accumulation and demonstrated the following: (i) the null mutation of HSV-1 UL34 caused specific dispersion throughout the cytoplasm of CD98hc and the HSV-1 de-envelopment regulators, glycoproteins B and H (gB and gH); (ii) as observed with CD98hc, gB, and gH, wild-type HSV-1 infection caused redistribution of the endoplasmic reticulum (ER) markers calnexin and ERp57 around the NM, whereas the UL34-null mutation caused cytoplasmic dispersion of these markers; (iii) the ER markers colocalized efficiently with CD98hc, gB, and gH in the presence and absence of UL34 in HSV-1-infected cells; (iv) at the ultrastructural level, wild-type HSV-1 infection caused ER compression around the NM, whereas the UL34-null mutation caused cytoplasmic dispersion of the ER; and (v) the UL34-null mutation significantly decreased the colocalization efficiency of lamin protein markers of the NM with CD98hc and gB. Collectively, these results indicate that HSV-1 infection causes redistribution of the ER around the NM, with resulting accumulation of ER-associated CD98hc, gB, and gH around the NM and that UL34 is required for ER redistribution, as well as for efficient recruitment to the NM of the ER-associated de-envelopment factors. Our study suggests that HSV-1 induces remodeling of the global ER architecture for recruitment of regulators mediating viral nuclear egress to the NM.IMPORTANCE The ER is an important cellular organelle that exists as a complex network extending throughout the cytoplasm. Although viruses often remodel the ER to facilitate viral replication, information on the effects of herpesvirus infections on ER morphological integrity is limited. Here, we showed that HSV-1 infection led to compression of the global ER architecture around the NM, resulting in accumulation of ER-associated regulators associated with nuclear egress of HSV-1 nucleocapsids. We also identified HSV-1 UL34 as a viral factor that mediated ER remodeling. Furthermore, we demonstrated that UL34 was required for efficient targeting of these regulators to the NM. To our knowledge, this is the first report showing that a herpesvirus remodels ER global architecture. Our study also provides insight into the mechanism by which the regulators for HSV-1 nuclear egress are recruited to the NM, where this viral event occurs.


Subject(s)
Endoplasmic Reticulum/ultrastructure , Endoplasmic Reticulum/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Viral Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Calnexin/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Fusion Regulatory Protein-1/metabolism , Herpesvirus 1, Human/chemistry , Humans , Mutation , Nuclear Envelope/physiology , Nuclear Envelope/virology , Nucleocapsid/metabolism , Protein Binding , Vero Cells , Viral Proteins/genetics , Virus Assembly , Virus Release , Virus Replication
9.
Biochem Soc Trans ; 46(3): 669-681, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29784648

ABSTRACT

Nesprins (nuclear envelope spectrin repeat proteins) are a family of multi-isomeric scaffolding proteins. Nesprins form the LInker of Nucleoskeleton-and-Cytoskeleton (LINC) complex with SUN (Sad1p/UNC84) domain-containing proteins at the nuclear envelope, in association with lamin A/C and emerin, linking the nucleoskeleton to the cytoskeleton. The LINC complex serves as both a physical linker between the nuclear lamina and the cytoskeleton and a mechanosensor. The LINC complex has a broad range of functions and is involved in maintaining nuclear architecture, nuclear positioning and migration, and also modulating gene expression. Over 80 disease-related variants have been identified in SYNE-1/2 (nesprin-1/2) genes, which result in muscular or central nervous system disorders including autosomal dominant Emery-Dreifuss muscular dystrophy, dilated cardiomyopathy and autosomal recessive cerebellar ataxia type 1. To date, 17 different nesprin mouse lines have been established to mimic these nesprin-related human diseases, which have provided valuable insights into the roles of nesprin and its scaffold LINC complex in a tissue-specific manner. In this review, we summarise the existing nesprin mouse models, compare their phenotypes and discuss the potential mechanisms underlying nesprin-associated diseases.


Subject(s)
Disease Models, Animal , Heart Diseases/physiopathology , Muscular Diseases/physiopathology , Nerve Tissue Proteins/physiology , Nuclear Envelope/physiology , Animals , Heart Diseases/genetics , Humans , Mice , Muscular Diseases/genetics , Nerve Tissue Proteins/genetics , Phenotype
10.
Biochem Soc Trans ; 46(2): 311-320, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29487227

ABSTRACT

Nesprins (nuclear envelope spectrin repeat proteins) are multi-isomeric scaffolding proteins. Nesprin-1 and -2 are highly expressed in skeletal and cardiac muscles and together with SUN (Sad1p/UNC84) domain-containing proteins form the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex at the nuclear envelope in association with lamin A/C and emerin. Mutations in nesprin-1/2 have been found in patients with autosomal dominant Emery-Dreifuss muscular dystrophy (EDMD) as well as dilated cardiomyopathy (DCM). Several lines of evidence indicate that compromised LINC complex function is the critical step leading to muscle disease. Here, we review recent advances in our understanding of the functions of nesprin-1/2 in the LINC complex and mechanistic insights into how mutations in nesprin-1/2 lead to nesprin-related muscle diseases, in particular DCM and EDMD.


Subject(s)
Muscle Development , Muscle Proteins/physiology , Muscular Diseases/physiopathology , Nuclear Envelope/physiology , Nuclear Proteins/physiology , Animals , Disease Models, Animal , Humans , Muscle Proteins/genetics , Muscular Dystrophies/physiopathology , Nuclear Proteins/genetics , Phenotype
11.
Mem Inst Oswaldo Cruz ; 113(6): e170531, 2018.
Article in English | MEDLINE | ID: mdl-29924141

ABSTRACT

BACKGROUND Eukaryotic ribonucleoprotein (RNP) granules are important for the regulation of RNA fate. RNP granules exist in trypanosomatids; however, their roles in controlling gene expression are still not understood. XRNA is a component of granules in Trypanosoma brucei but has not been investigated in Trypanosoma cruzi. OBJECTIVES This study aimed to investigate the TcXRNA dynamic assembly and its interaction with RNP components under conditions that affect the mRNA availability. METHODS We used in vitro metacyclogenesis of T. cruzi to observe changes in RNP granules during the differentiation process. TcXRNA expression was analysed by Western blot and immunofluorescence. Colocalisation assays were performed to investigate the interaction of TcXRNA with other RNP components. FINDINGS TcXRNA is constantly present during metacyclogenesis and is localised in cytoplasmic granules. TcXRNA does not colocalise with TcDHH1 and TcCAF1 granules in the cytoplasm. However, TcXRNA granules colocalise with mRNP granules at the nuclear periphery when mRNA processing is inhibited. MAIN CONCLUSIONS TcXRNA plays a role in mRNA metabolism as a component of mRNP granules whose assembly is dependent on mRNA availability. TcXRNA granules colocalise with distinct RNP granules at the nuclear periphery, suggesting that the perinuclear region is a regulatory compartment in T. cruzi mRNA metabolism.


Subject(s)
Cytoplasmic Granules/genetics , Protozoan Proteins/genetics , RNA, Protozoan/genetics , Ribonucleoproteins/genetics , Trypanosoma cruzi/cytology , Blotting, Western , Cytoplasmic Granules/physiology , Fluorescent Antibody Technique , Nuclear Envelope/physiology , Protozoan Proteins/physiology , RNA, Protozoan/physiology , Ribonucleoproteins/physiology , Trypanosoma cruzi/genetics
12.
J Cell Sci ; 128(6): 1090-6, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25653391

ABSTRACT

The inner nuclear membrane proteins emerin and LEMD2 have both overlapping and separate functions in regulation of nuclear organization, gene expression and cell differentiation. We report here that emerin (EMR-1) and LEM domain protein 2 (LEM-2) are expressed in all tissues throughout Caenorhaditis elegans development but their relative distribution differs between cell types. The ratio of EMR-1 to LEM-2 is particularly high in contractile tissues, intermediate in neurons and hypodermis and lowest in intestine and germ line. We find that LEM-2 is recruited earlier than EMR-1 to reforming nuclear envelopes, suggesting the presence of separate mitotic membrane compartments and specific functions of each protein. Concordantly, we observe that nuclei of lem-2 mutant embryos, but not of emr-1 mutants, have reduced nuclear circularity. Finally, we uncover a so-far-unknown role of LEM-2 in nuclear separation and anchoring of microtubule organizing centers.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Cell Nucleus/physiology , Cell Nucleus/ultrastructure , Membrane Proteins/metabolism , Nuclear Envelope/physiology , Nuclear Proteins/metabolism , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/genetics , Cell Cycle Proteins , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Membrane Proteins/genetics , Nuclear Proteins/genetics
13.
Bioessays ; 37(8): 862-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26060083

ABSTRACT

It is now well accepted that defined architectural compartments within the cell nucleus can regulate the transcriptional activity of chromosomal domains within their vicinity. However, it is generally unclear how these compartments are formed. The nuclear periphery has received a great deal of attention as a repressive compartment that is implicated in many cellular functions during development and disease. The inner nuclear membrane, the nuclear lamina, and associated proteins compose the nuclear periphery and together they interact with proximal chromatin creating a repressive environment. A new study by Harr et al. identifies specific protein-DNA interactions and epigenetic states necessary to re-position chromatin to the nuclear periphery in a cell-type specific manner. Here, we review concepts in gene positioning within the nucleus and current accepted models of dynamic gene repositioning within the nucleus during differentiation. This study highlights that myriad pathways lead to nuclear organization.


Subject(s)
Heterochromatin/physiology , Nuclear Envelope/physiology , Animals , Cell Differentiation , Epigenesis, Genetic , Heterochromatin/ultrastructure , Histones/metabolism , Humans , Nuclear Envelope/ultrastructure , Protein Processing, Post-Translational
14.
J Biomech Eng ; 139(2)2017 02 01.
Article in English | MEDLINE | ID: mdl-27918797

ABSTRACT

Biophysical stimuli presented to cells via microenvironmental properties (e.g., alignment and stiffness) or external forces have a significant impact on cell function and behavior. Recently, the cell nucleus has been identified as a mechanosensitive organelle that contributes to the perception and response to mechanical stimuli. However, the specific mechanotransduction mechanisms that mediate these effects have not been clearly established. Here, we offer a comprehensive review of the evidence supporting (and refuting) three hypothetical nuclear mechanotransduction mechanisms: physical reorganization of chromatin, signaling at the nuclear envelope, and altered cytoskeletal structure/tension due to nuclear remodeling. Our goal is to provide a reference detailing the progress that has been made and the areas that still require investigation regarding the role of nuclear mechanotransduction in cell biology. Additionally, we will briefly discuss the role that mathematical models of cell mechanics can play in testing these hypotheses and in elucidating how biophysical stimulation of the nucleus drives changes in cell behavior. While force-induced alterations in signaling pathways involving lamina-associated polypeptides (LAPs) (e.g., emerin and histone deacetylase 3 (HDAC3)) and transcription factors (TFs) located at the nuclear envelope currently appear to be the most clearly supported mechanism of nuclear mechanotransduction, additional work is required to examine this process in detail and to more fully test alternative mechanisms. The combination of sophisticated experimental techniques and advanced mathematical models is necessary to enhance our understanding of the role of the nucleus in the mechanotransduction processes driving numerous critical cell functions.


Subject(s)
Cell Nucleus/physiology , Cytoskeleton/physiology , Mechanotransduction, Cellular/physiology , Models, Biological , Animals , Chromatin/physiology , Compressive Strength/physiology , Computer Simulation , Elastic Modulus/physiology , Humans , Nuclear Envelope/physiology , Stress, Mechanical
15.
Semin Cell Dev Biol ; 29: 164-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24508913

ABSTRACT

Mutations in genes encoding widely expressed nuclear envelope proteins often lead to diseases that manifest in specific tissues. Lamina-associated polypeptide 1 (LAP1) is an integral protein of the inner nuclear membrane that is expressed in most cells and tissues. Within the nuclear envelope, LAP1 interacts physically with lamins, torsinA and emerin, suggesting it may serve as a key node for transducing signals across the inner nuclear membrane. Indeed, recent in vivo studies in genetically modified mice strongly support functional links between LAP1 and both torsinA (in neurons) and emerin (in muscle). These studies suggest that tissue-selective diseases caused by mutations in genes encoding nuclear envelope proteins may result, at least in part, from the selective disruption of discrete nuclear envelope protein complexes.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscular Dystrophies/genetics , Nuclear Envelope/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Animals , Carrier Proteins/metabolism , Cytoskeletal Proteins , Humans , Lamins/metabolism , Mice , Molecular Chaperones/metabolism , Muscular Dystrophies/pathology , Mutation , Signal Transduction
16.
EMBO J ; 31(21): 4179-90, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23034402

ABSTRACT

The microtubule motor protein kinesin-5 (Eg5) provides an outward force on centrosomes, which drives bipolar spindle assembly. Acute inhibition of Eg5 blocks centrosome separation and causes mitotic arrest in human cells, making Eg5 an attractive target for anti-cancer therapy. Using in vitro directed evolution, we show that human cells treated with Eg5 inhibitors can rapidly acquire the ability to divide in the complete absence of Eg5 activity. We have used these Eg5-independent cells to study alternative mechanisms of centrosome separation. We uncovered a pathway involving nuclear envelope (NE)-associated dynein that drives centrosome separation in prophase. This NE-dynein pathway is essential for bipolar spindle assembly in the absence of Eg5, but also functions in the presence of full Eg5 activity, where it pulls individual centrosomes along the NE and acts in concert with Eg5-dependent outward pushing forces to coordinate prophase centrosome separation. Together, these results reveal how the forces are produced to drive prophase centrosome separation and identify a novel mechanism of resistance to kinesin-5 inhibitors.


Subject(s)
Centrosome/physiology , Dyneins/metabolism , Kinesins/metabolism , Mitosis/physiology , Nuclear Envelope/physiology , Prophase/physiology , Spindle Apparatus/physiology , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Colony-Forming Units Assay , Dyneins/genetics , Flow Cytometry , HeLa Cells , Humans , Kinesins/antagonists & inhibitors , Kinesins/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Small Interfering/genetics
17.
Biochem Biophys Res Commun ; 470(1): 35-40, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26721436

ABSTRACT

Permeabilization of cell membranes occurs upon exposure to a threshold absorbed dose (AD) of nanosecond pulsed electric fields (nsPEF). The ultimate, physiological bioeffect of this exposure depends on the type of cultured cell and environment, indicating that cell-specific pathways and structures are stimulated. Here we investigate 10 and 600 ns duration PEF effects on Chinese hamster ovary (CHO) cell nuclei, where our hypothesis is that pulse disruption of the nuclear envelope membrane leads to observed cell death and decreased viability 24 h post-exposure. To observe short-term responses to nsPEF exposure, CHO cells have been stably transfected with two fluorescently-labeled proteins known to be sequestered for cellular chromosomal function within the nucleus - histone-2b (H2B) and proliferating cell nuclear antigen (PCNA). H2B remains associated with chromatin after nsPEF exposure, whereas PCNA leaks out of nuclei permeabilized by a threshold AD of 10 and 600 ns PEF. A downturn in 24 h viability, measured by MTT assay, is observed at the number of pulses required to induce permeabilization of the nucleus.


Subject(s)
Apoptosis/radiation effects , Cell Membrane Permeability/physiology , Cell Membrane Permeability/radiation effects , Electroporation/methods , Nuclear Envelope/physiology , Nuclear Envelope/radiation effects , Animals , Apoptosis/physiology , CHO Cells , Cell Survival/physiology , Cell Survival/radiation effects , Cricetinae , Cricetulus , Dose-Response Relationship, Radiation , Electromagnetic Fields , Radiation Dosage
18.
Bioessays ; 36(7): 665-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24848719

ABSTRACT

During mitosis, cells comprehensively restructure their interior to promote the faithful inheritance of DNA and cytoplasmic contents. In metazoans, this restructuring entails disassembly of the nuclear envelope, redistribution of its components into the endoplasmic reticulum (ER) and eventually nuclear envelope reassembly around the segregated chromosomes. The microtubule cytoskeleton has recently emerged as a critical regulator of mitotic nuclear envelope and ER dynamics. Microtubules and associated molecular motors tear open the nuclear envelope in prophase and remove nuclear envelope remnants from chromatin. Additionally, two distinct mechanisms of microtubule-based regulation of ER dynamics operate later in mitosis. First, association of the ER with microtubules is reduced, preventing invasion of ER into the spindle area, and second, organelle membrane is actively cleared from metaphase chromosomes. However, we are only beginning to understand the role of microtubules in shaping and distributing ER and other organelles during mitosis.


Subject(s)
Endoplasmic Reticulum/physiology , Microtubules/physiology , Mitosis/physiology , Nuclear Envelope/physiology , Animals , Chromatin Assembly and Disassembly , Cytoskeleton/physiology , Humans
19.
Eukaryot Cell ; 14(4): 345-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25527523

ABSTRACT

Sarcolemmal membrane-associated protein (SLMAP) is a tail-anchored protein involved in fundamental cellular processes, such as myoblast fusion, cell cycle progression, and chromosomal inheritance. Further, SLMAP misexpression is associated with endothelial dysfunctions in diabetes and cancer. SLMAP is part of the conserved striatin-interacting phosphatase and kinase (STRIPAK) complex required for specific signaling pathways in yeasts, filamentous fungi, insects, and mammals. In filamentous fungi, STRIPAK was initially discovered in Sordaria macrospora, a model system for fungal differentiation. Here, we functionally characterize the STRIPAK subunit PRO45, a homolog of human SLMAP. We show that PRO45 is required for sexual propagation and cell-to-cell fusion and that its forkhead-associated (FHA) domain is essential for these processes. Protein-protein interaction studies revealed that PRO45 binds to STRIPAK subunits PRO11 and SmMOB3, which are also required for sexual propagation. Superresolution structured-illumination microscopy (SIM) further established that PRO45 localizes to the nuclear envelope, endoplasmic reticulum, and mitochondria. SIM also showed that localization to the nuclear envelope requires STRIPAK subunits PRO11 and PRO22, whereas for mitochondria it does not. Taken together, our study provides important insights into fundamental roles of the fungal SLMAP homolog PRO45 and suggests STRIPAK-related and STRIPAK-unrelated functions.


Subject(s)
Endoplasmic Reticulum/physiology , Membrane Proteins/physiology , Mitochondria/physiology , Nuclear Envelope/physiology , Sordariales/cytology , Amino Acid Sequence , Fungal Proteins/physiology , Hyphae/cytology , Models, Biological , Molecular Sequence Data , Protein Binding , Signal Transduction
20.
J Cell Sci ; 126(Pt 17): 4048-58, 2013 09 01.
Article in English | MEDLINE | ID: mdl-23813954

ABSTRACT

Skeletal muscle fibres are very large and elongated. In response to excitation there must be a rapid and uniform release of Ca(2+) throughout for contraction. To ensure a uniform spread of excitation throughout the fibre to all the Ca(2+) release sites, the muscle internalizes the plasma membrane, to form the tubular (t-) system. Hence the t-system forms a complex and dense network throughout the fibre that is responsible for excitation-contraction coupling and other signalling mechanisms. However, we currently do not have a very detailed view of this membrane network because of limitations in previously used imaging techniques to visualize it. In this study we serially imaged fluorescent dye trapped in the t-system of fibres from rat and toad muscle using the confocal microscope, and deconvolved and reconstructed these images to produce the first three-dimensional reconstructions of large volumes of the vertebrate t-system. These images showed complex arrangements of tubules that have not been described previously and also allowed the association of the t-system with cellular organelles to be visualized. There was a high density of tubules close to the nuclear envelope because of the close and parallel alignment of the long axes of the myofibrils and the nuclei. Furthermore local fluorescence intensity variations from sub-resolution tubules were converted to tubule diameters. Mean diameters of tubules were 85.9±6.6 and 91.2±8.2 nm, from rat and toad muscle under isotonic conditions, respectively. Under osmotic stress the distribution of tubular diameters shifted significantly in toad muscle only, with change specifically occurring in the transverse but not longitudinal tubules.


Subject(s)
Cell Membrane/metabolism , Muscle Contraction/physiology , Myofibrils/physiology , Animals , Bufo marinus , Calcium/metabolism , Fluorescent Dyes , Nuclear Envelope/physiology , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL