Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 16.405
1.
Int J Mol Sci ; 25(9)2024 May 02.
Article En | MEDLINE | ID: mdl-38732188

The cytoskeleton plays a pivotal role in maintaining the epithelial phenotype and is vital to several hallmark processes of cancer. Over the past decades, researchers have identified the epithelial protein lost in neoplasm (EPLIN, also known as LIMA1) as a key regulator of cytoskeletal dynamics, cytoskeletal organization, motility, as well as cell growth and metabolism. Dysregulation of EPLIN is implicated in various aspects of cancer progression, such as tumor growth, invasion, metastasis, and therapeutic resistance. Its altered expression levels or activity can disrupt cytoskeletal dynamics, leading to aberrant cell motility and invasiveness characteristic of malignant cells. Moreover, the involvement of EPLIN in cell growth and metabolism underscores its significance in orchestrating key processes essential for cancer cell survival and proliferation. This review provides a comprehensive exploration of the intricate roles of EPLIN across diverse cellular processes in both normal physiology and cancer pathogenesis. Additionally, this review discusses the possibility of EPLIN as a potential target for anticancer therapy in future studies.


Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Cell Movement , Cell Proliferation
2.
Adv Exp Med Biol ; 1452: 1-19, 2024.
Article En | MEDLINE | ID: mdl-38805122

Microtubules are dynamic polymers composed of α- and ß-tubulin heterodimers. Microtubules are universally conserved among eukaryotes and participate in nearly every cellular process, including intracellular trafficking, replication, polarity, cytoskeletal shape, and motility. Due to their fundamental role in mitosis, they represent a classic target of anti-cancer therapy. Microtubule-stabilizing agents currently constitute a component of the most effective regimens for ovarian cancer therapy in both primary and recurrent settings. Unfortunately, the development of resistance continues to present a therapeutic challenge. An understanding of the underlying mechanisms of resistance to microtubule-active agents may facilitate the development of novel and improved approaches to this disease.


Cytoskeleton , Microtubules , Ovarian Neoplasms , Tubulin Modulators , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Female , Microtubules/drug effects , Microtubules/metabolism , Tubulin Modulators/therapeutic use , Tubulin Modulators/pharmacology , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals
3.
Viruses ; 16(5)2024 04 25.
Article En | MEDLINE | ID: mdl-38793550

Rotavirus (RV) replicates within viroplasms, membraneless electron-dense globular cytosolic inclusions with liquid-liquid phase properties. In these structures occur the virus transcription, replication, and packaging of the virus genome in newly assembled double-layered particles. The viroplasms are composed of virus proteins (NSP2, NSP5, NSP4, VP1, VP2, VP3, and VP6), single- and double-stranded virus RNAs, and host components such as microtubules, perilipin-1, and chaperonins. The formation, coalescence, maintenance, and perinuclear localization of viroplasms rely on their association with the cytoskeleton. A stabilized microtubule network involving microtubules and kinesin Eg5 and dynein molecular motors is associated with NSP5, NSP2, and VP2, facilitating dynamic processes such as viroplasm coalescence and perinuclear localization. Key post-translation modifications, particularly phosphorylation events of RV proteins NSP5 and NSP2, play pivotal roles in orchestrating these interactions. Actin filaments also contribute, triggering the formation of the viroplasms through the association of soluble cytosolic VP4 with actin and the molecular motor myosin. This review explores the evolving understanding of RV replication, emphasizing the host requirements essential for viroplasm formation and highlighting their dynamic interplay within the host cell.


Cytoskeleton , Rotavirus , Virus Replication , Rotavirus/physiology , Rotavirus/metabolism , Rotavirus/genetics , Cytoskeleton/metabolism , Cytoskeleton/virology , Humans , Animals , Microtubules/metabolism , Microtubules/virology , Viral Proteins/metabolism , Viral Proteins/genetics , Host-Pathogen Interactions , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Viral Replication Compartments/metabolism , Rotavirus Infections/virology , RNA, Viral/genetics , RNA, Viral/metabolism
4.
Commun Biol ; 7(1): 458, 2024 Apr 15.
Article En | MEDLINE | ID: mdl-38622242

Differentiation of adipose progenitor cells into mature adipocytes entails a dramatic reorganization of the cellular architecture to accommodate lipid storage into cytoplasmic lipid droplets. Lipid droplets occupy most of the adipocyte volume, compressing the nucleus beneath the plasma membrane. How this cellular remodeling affects sub-nuclear structure, including size and number of nucleoli, remains unclear. We describe the morphological remodeling of the nucleus and the nucleolus during in vitro adipogenic differentiation of primary human adipose stem cells. We find that cell cycle arrest elicits a remodeling of nucleolar structure which correlates with a decrease in protein synthesis. Strikingly, triggering cytoskeletal rearrangements mimics the nucleolar remodeling observed during adipogenesis. Our results point to nucleolar remodeling as an active, mechano-regulated mechanism during adipogenic differentiation and demonstrate a key role of the actin cytoskeleton in defining nuclear and nucleolar architecture in differentiating human adipose stem cells.


Adipogenesis , Cytoskeleton , Humans , Cells, Cultured , Cytoskeleton/metabolism , Adipocytes/metabolism , Lipid Droplets/metabolism
5.
PLoS Biol ; 22(4): e3002575, 2024 Apr.
Article En | MEDLINE | ID: mdl-38683844

Muscles undergo developmental transitions in gene expression and alternative splicing that are necessary to refine sarcomere structure and contractility. CUG-BP and ETR-3-like (CELF) family RNA-binding proteins are important regulators of RNA processing during myogenesis that are misregulated in diseases such as Myotonic Dystrophy Type I (DM1). Here, we report a conserved function for Bruno 1 (Bru1, Arrest), a CELF1/2 family homolog in Drosophila, during early muscle myogenesis. Loss of Bru1 in flight muscles results in disorganization of the actin cytoskeleton leading to aberrant myofiber compaction and defects in pre-myofibril formation. Temporally restricted rescue and RNAi knockdown demonstrate that early cytoskeletal defects interfere with subsequent steps in sarcomere growth and maturation. Early defects are distinct from a later requirement for bru1 to regulate sarcomere assembly dynamics during myofiber maturation. We identify an imbalance in growth in sarcomere length and width during later stages of development as the mechanism driving abnormal radial growth, myofibril fusion, and the formation of hollow myofibrils in bru1 mutant muscle. Molecularly, we characterize a genome-wide transition from immature to mature sarcomere gene isoform expression in flight muscle development that is blocked in bru1 mutants. We further demonstrate that temporally restricted Bru1 rescue can partially alleviate hypercontraction in late pupal and adult stages, but it cannot restore myofiber function or correct structural deficits. Our results reveal the conserved nature of CELF function in regulating cytoskeletal dynamics in muscle development and demonstrate that defective RNA processing due to misexpression of CELF proteins causes wide-reaching structural defects and progressive malfunction of affected muscles that cannot be rescued by late-stage gene replacement.


Cytoskeleton , Drosophila Proteins , Drosophila melanogaster , Muscle Development , RNA-Binding Proteins , Sarcomeres , Animals , Sarcomeres/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Muscle Development/genetics , Cytoskeleton/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , RNA Splicing/genetics , Myofibrils/metabolism , Flight, Animal/physiology , Alternative Splicing/genetics , Gene Expression Regulation, Developmental , Muscles/metabolism
6.
Acta Biomater ; 180: 197-205, 2024 May.
Article En | MEDLINE | ID: mdl-38599439

During physiological and pathological processes, cells experience significant morphological alterations with the re-arrangement of cytoskeletal filaments, resulting in anisotropic viscoelasticity. Here, a structure-based cell model is proposed to study the anisotropic viscoelastic mechanical behaviors of living cells. We investigate how cell shape affects its creep responses in longitudinal and perpendicular directions. It is shown that cells exhibit power-law rheological behavior in both longitudinal and perpendicular directions under step stress, with a more solid-like behavior along the longitudinal direction. We reveal that the cell volume and cytoskeletal filament orientation, which have been neglected in most existing models, play a critical role in regulating cellular anisotropic viscoelasticity. The stiffness of the cell in both directions increases linearly with increasing its aspect ratio, due to the decrease of cell volume. Moreover, the increase in the cell's aspect ratio produces the aggregation of cytoskeletal filaments along the longitudinal direction, resulting in higher stiffness in this direction. It is also shown that the increase in cell's aspect ratio corresponds to a process of cellular ordering, which can be quantitatively characterized by the orientational entropy of cytoskeletal filaments. In addition, we present a simple yet robust method to establish the relationship between cell's aspect ratio and cell volume, thus providing a theoretical framework to capture the anisotropic viscoelastic behavior of cells. This study suggests that the structure-based cell models may be further developed to investigate cellular rheological responses to external mechanical stimuli and may be extended to the tissue scale. STATEMENT OF SIGNIFICANCE: The viscoelastic behaviors of cells hold significant importance in comprehending the roles of mechanical forces in embryo development, invasion, and metastasis of cancer cells. Here, a structure-based cell model is proposed to study the anisotropic viscoelastic mechanical behaviors of living cells. Our study highlights the crucial role of previously neglected factors, such as cell volume and cytoskeletal filament orientation, in regulating cellular anisotropic viscoelasticity. We further propose an orientational entropy of cytoskeletal filaments to quantitatively characterize the ordering process of cells with increasing aspect ratios. Moreover, we derived the analytical interrelationships between cell aspect ratio, cell stiffness, cell volume, and cytoskeletal fiber orientation. This study provides a theoretical framework to describe the anisotropic viscoelastic mechanical behavior of cells.


Cytoskeleton , Elasticity , Models, Biological , Anisotropy , Cytoskeleton/metabolism , Viscosity , Rheology , Humans , Cell Size , Stress, Mechanical
7.
PLoS Genet ; 20(4): e1011224, 2024 Apr.
Article En | MEDLINE | ID: mdl-38662776

Cell adhesion requires linkage of transmembrane receptors to the cytoskeleton through intermediary linker proteins. Integrin-based adhesion to the extracellular matrix (ECM) involves large adhesion complexes that contain multiple cytoskeletal adapters that connect to the actin cytoskeleton. Many of these adapters, including the essential cytoskeletal linker Talin, have been shown to contain multiple actin-binding sites (ABSs) within a single protein. To investigate the possible role of having such a variety of ways of linking integrins to the cytoskeleton, we generated mutations in multiple actin binding sites in Drosophila talin. Using this approach, we have been able to show that different actin-binding sites in talin have both unique and complementary roles in integrin-mediated adhesion. Specifically, mutations in either the C-terminal ABS3 or the centrally located ABS2 result in lethality showing that they have unique and non-redundant function in some contexts. On the other hand, flies simultaneously expressing both the ABS2 and ABS3 mutants exhibit a milder phenotype than either mutant by itself, suggesting overlap in function in other contexts. Detailed phenotypic analysis of ABS mutants elucidated the unique roles of the talin ABSs during embryonic development as well as provided support for the hypothesis that talin acts as a dimer in in vivo contexts. Overall, our work highlights how the ability of adhesion complexes to link to the cytoskeleton in multiple ways provides redundancy, and consequently robustness, but also allows a capacity for functional specialization.


Actins , Cell Adhesion , Drosophila Proteins , Drosophila melanogaster , Extracellular Matrix , Integrins , Talin , Talin/metabolism , Talin/genetics , Animals , Cell Adhesion/genetics , Binding Sites , Extracellular Matrix/metabolism , Actins/metabolism , Actins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Integrins/metabolism , Integrins/genetics , Mutation , Protein Binding , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/genetics
8.
EMBO Rep ; 25(5): 2172-2187, 2024 May.
Article En | MEDLINE | ID: mdl-38627564

Cells are equipped with asymmetrically localised and functionally specialised components, including cytoskeletal structures and organelles. Positioning these components to specific intracellular locations in an asymmetric manner is critical for their functionality and affects processes like immune responses, tissue maintenance, muscle functionality, and neurobiology. Here, we provide an overview of strategies to actively move, position, and anchor organelles to specific locations. By conceptualizing the cytoskeletal forces and the organelle-to-cytoskeleton connectivity, we present a framework of active positioning of both membrane-enclosed and membrane-less organelles. Using this framework, we discuss how different principles of force generation and organelle anchorage are utilised by different cells, such as mesenchymal and amoeboid cells, and how the microenvironment influences the plasticity of organelle positioning. Given that motile cells face the challenge of coordinating the positioning of their content with cellular motion, we particularly focus on principles of organelle positioning during migration. In this context, we discuss novel findings on organelle positioning by anchorage-independent mechanisms and their advantages and disadvantages in motile as well as stationary cells.


Cell Movement , Cytoskeleton , Organelles , Organelles/metabolism , Humans , Cytoskeleton/metabolism , Animals
9.
Elife ; 132024 Apr 25.
Article En | MEDLINE | ID: mdl-38661167

Osteoblast adherence to bone surfaces is important for remodeling bone tissue. This study demonstrates that deficiency of TG-interacting factor 1 (Tgif1) in osteoblasts results in altered cell morphology, reduced adherence to collagen type I-coated surfaces, and impaired migration capacity. Tgif1 is essential for osteoblasts to adapt a regular cell morphology and to efficiently adhere and migrate on collagen type I-rich matrices in vitro. Furthermore, Tgif1 acts as a transcriptional repressor of p21-activated kinase 3 (Pak3), an important regulator of focal adhesion formation and osteoblast spreading. Absence of Tgif1 leads to increased Pak3 expression, which impairs osteoblast spreading. Additionally, Tgif1 is implicated in osteoblast recruitment and activation of bone surfaces in the context of bone regeneration and in response to parathyroid hormone 1-34 (PTH 1-34) treatment in vivo in mice. These findings provide important novel insights in the regulation of the cytoskeletal architecture of osteoblasts.


Cytoskeleton , Homeodomain Proteins , Osteoblasts , Repressor Proteins , Signal Transduction , p21-Activated Kinases , Animals , Mice , Cell Adhesion , Cell Movement , Cytoskeleton/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Osteoblasts/metabolism , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/deficiency , Repressor Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/deficiency
10.
J Cell Sci ; 137(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38647525

Cell migration is controlled by the coordinated action of cell adhesion, cytoskeletal dynamics, contractility and cell extrinsic cues. Integrins are the main adhesion receptors to ligands of the extracellular matrix (ECM), linking the actin cytoskeleton to the ECM and enabling cells to sense matrix rigidity and mount a directional cell migration response to stiffness gradients. Most models studied show preferred migration of single cells or cell clusters towards increasing rigidity. This is referred to as durotaxis, and since its initial discovery in 2000, technical advances and elegant computational models have provided molecular level details of stiffness sensing in cell migration. However, modeling has long predicted that, depending on cell intrinsic factors, such as the balance of cell adhesion molecules (clutches) and the motor proteins pulling on them, cells might also prefer adhesion to intermediate rigidity. Recently, experimental evidence has supported this notion and demonstrated the ability of cells to migrate towards lower rigidity, in a process called negative durotaxis. In this Review, we discuss the significant conceptual advances that have been made in our appreciation of cell plasticity and context dependency in stiffness-guided directional cell migration.


Cell Movement , Extracellular Matrix , Cell Movement/physiology , Humans , Animals , Extracellular Matrix/metabolism , Integrins/metabolism , Cell Adhesion , Models, Biological , Cytoskeleton/metabolism
11.
Cell Mol Life Sci ; 81(1): 193, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38652325

The acetylation of α-tubulin on lysine 40 is a well-studied post-translational modification which has been associated with the presence of long-lived stable microtubules that are more resistant to mechanical breakdown. The discovery of α-tubulin acetyltransferase 1 (ATAT1), the enzyme responsible for lysine 40 acetylation on α-tubulin in a wide range of species, including protists, nematodes, and mammals, dates to about a decade ago. However, the role of ATAT1 in different cellular activities and molecular pathways has been only recently disclosed. This review comprehensively summarizes the most recent knowledge on ATAT1 structure and substrate binding and analyses the involvement of ATAT1 in a variety of cellular processes such as cell motility, mitosis, cytoskeletal organization, and intracellular trafficking. Finally, the review highlights ATAT1 emerging roles in human diseases and discusses ATAT1 potential enzymatic and non-enzymatic roles and the current efforts in developing ATAT1 inhibitors.


Acetyltransferases , Microtubule Proteins , Tubulin , Humans , Acetyltransferases/metabolism , Acetyltransferases/chemistry , Tubulin/metabolism , Tubulin/chemistry , Animals , Protein Processing, Post-Translational , Acetylation , Microtubules/metabolism , Mitosis , Cell Movement , Neoplasms/pathology , Neoplasms/enzymology , Neoplasms/metabolism , Cytoskeleton/metabolism
12.
J Cell Biol ; 223(7)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38683248

Nucleocytoplasmic transport (NCT), the facilitated diffusion of cargo molecules between the nucleus and cytoplasm through nuclear pore complexes (NPCs), enables numerous fundamental eukaryotic cellular processes. Ran GTPase uses cellular energy in the direct form of GTP to create a gradient across the nuclear envelope (NE) that drives the majority of NCT. We report here that changes in GTP availability resulting from altered cellular physiology modulate the rate of NCT, as monitored using synthetic and natural cargo, and the dynamics of Ran itself. Cell migration, cell spreading, and/or modulation of the cytoskeleton or its connection to the nucleus alter GTP availability and thus rates of NCT, regulating RNA export and protein synthesis. These findings support a model in which changes in cellular physiology that alter GTP availability can regulate the rate of NCT, impacting fundamental cellular processes that extensively utilize NCT.


Active Transport, Cell Nucleus , Guanosine Triphosphate , ran GTP-Binding Protein , Guanosine Triphosphate/metabolism , ran GTP-Binding Protein/metabolism , ran GTP-Binding Protein/genetics , Humans , Cell Nucleus/metabolism , Cell Movement , Nuclear Pore/metabolism , Nuclear Pore/genetics , Animals , Nuclear Envelope/metabolism , Cytoskeleton/metabolism , Protein Biosynthesis , Cytoplasm/metabolism
13.
Biochem Soc Trans ; 52(2): 517-527, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38572868

Cellular signalling is a complex process and involves cascades of enzymes that, in response to a specific signal, give rise to exact cellular responses. Signalling scaffold proteins organise components of these signalling pathways in space and time to co-ordinate signalling outputs. In this review we introduce a new class of mechanically operated signalling scaffolds that are built into the cytoskeletal architecture of the cell. These proteins contain force-dependent binary switch domains that integrate chemical and mechanical signals to introduce quantised positional changes to ligands and persistent alterations in cytoskeletal architecture providing mechanomemory capabilities. We focus on the concept of spatial organisation, and how the cell organises signalling molecules at the plasma membrane in response to specific signals to create order and distinct signalling outputs. The dynamic positioning of molecules using binary switches adds an additional layer of complexity to the idea of scaffolding. The switches can spatiotemporally organise enzymes and substrates dynamically, with the introduction of ∼50 nm quantised steps in distance between them as the switch patterns change. Together these different types of signalling scaffolds and the proteins engaging them, provide a way for an ordering of molecules that extends beyond current views of the cell.


Cytoskeleton , Signal Transduction , Humans , Cytoskeleton/metabolism , Animals , Mechanotransduction, Cellular , Cell Membrane/metabolism
14.
Mol Biol Cell ; 35(6): ar87, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38656797

Recent findings indicate that Solo, a RhoGEF, is involved in cellular mechanical stress responses. However, the mechanism of actin cytoskeletal remodeling via Solo remains unclear. Therefore, this study aimed to identify Solo-interacting proteins using the BioID, a proximal-dependent labeling method, and elucidate the molecular mechanisms of function of Solo. We identified PDZ-RhoGEF (PRG) as a Solo-interacting protein. PRG colocalized with Solo in the basal area of cells, depending on Solo localization, and enhanced actin polymerization at the Solo accumulation sites. Additionally, Solo and PRG interaction was necessary for actin cytoskeletal remodeling. Furthermore, the purified Solo itself had little or negligible GEF activity, even its GEF-inactive mutant directly activated the GEF activity of PRG through interaction. Moreover, overexpression of the Solo and PRG binding domains, respectively, had a dominant-negative effect on actin polymerization and actin stress fiber formation in response to substrate stiffness. Therefore, Solo restricts the localization of PRG and regulates actin cytoskeletal remodeling in synergy with PRG in response to the surrounding mechanical environment.


Actin Cytoskeleton , Actins , Rho Guanine Nucleotide Exchange Factors , Humans , Actin Cytoskeleton/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Actins/metabolism , PDZ Domains , Protein Binding , Cytoskeleton/metabolism , Animals , HEK293 Cells
15.
Dev Cell ; 59(10): 1333-1344.e4, 2024 May 20.
Article En | MEDLINE | ID: mdl-38579717

Plant morphogenesis relies exclusively on oriented cell expansion and division. Nonetheless, the mechanism(s) determining division plane orientation remain elusive. Here, we studied tissue healing after laser-assisted wounding in roots of Arabidopsis thaliana and uncovered how mechanical forces stabilize and reorient the microtubule cytoskeleton for the orientation of cell division. We identified that root tissue functions as an interconnected cell matrix, with a radial gradient of tissue extendibility causing predictable tissue deformation after wounding. This deformation causes instant redirection of expansion in the surrounding cells and reorientation of microtubule arrays, ultimately predicting cell division orientation. Microtubules are destabilized under low tension, whereas stretching of cells, either through wounding or external aspiration, immediately induces their polymerization. The higher microtubule abundance in the stretched cell parts leads to the reorientation of microtubule arrays and, ultimately, informs cell division planes. This provides a long-sought mechanism for flexible re-arrangement of cell divisions by mechanical forces for tissue reconstruction and plant architecture.


Arabidopsis , Cell Division , Microtubules , Plant Roots , Microtubules/metabolism , Arabidopsis/metabolism , Arabidopsis/cytology , Cell Division/physiology , Plant Roots/metabolism , Plant Roots/cytology , Plant Roots/growth & development , Cytoskeleton/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Biomechanical Phenomena
16.
J Cell Biol ; 223(8)2024 Aug 05.
Article En | MEDLINE | ID: mdl-38668767

The microtubule cytoskeleton consists of microtubule subsets with distinct compositions of microtubule-associated proteins, which instruct the position and traffic of subcellular organelles. In the endocytic pathway, these microtubule-associated cues are poorly understood. Here, we report that in MDCK cells, endosomes with multivesicular body (MVB) and late endosome (LE) markers localize preferentially to microtubules coated with septin GTPases. Compared with early endosomes, CD63-containing MVBs/LEs are largely immotile on septin-coated microtubules. In vitro reconstitution assays revealed that the motility of isolated GFP-CD63 endosomes is directly inhibited by microtubule-associated septins. Quantification of CD63-positive endosomes containing the early endosome antigen (EEA1), the Rab7 effector and dynein adaptor RILP or Rab27a, showed that intermediary EEA1- and RILP-positive GFP-CD63 preferentially associate with septin-coated microtubules. Septin knockdown enhanced GFP-CD63 motility and decreased the percentage of CD63-positive MVBs/LEs with lysobiphosphatidic acid without impacting the fraction of EEA1-positive CD63. These results suggest that MVB maturation involves immobilization on septin-coated microtubules, which may facilitate multivesiculation and/or organelle-organelle contacts.


Microtubules , Multivesicular Bodies , Septins , Animals , Dogs , Madin Darby Canine Kidney Cells , Microtubules/chemistry , Microtubules/metabolism , Multivesicular Bodies/chemistry , Multivesicular Bodies/metabolism , Septins/chemistry , Septins/metabolism , Tetraspanin 30/metabolism , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Endocytosis
17.
Curr Opin Plant Biol ; 79: 102530, 2024 Jun.
Article En | MEDLINE | ID: mdl-38631088

Plant morphogenesis largely depends on the orientation and rate of cell division and elongation, and their coordination at all levels of organization. Despite recent progresses in the comprehension of pathways controlling division plane determination in plant cells, many pieces are missing to the puzzle. For example, we have a partial comprehension of formation, function and evolutionary significance of the preprophase band, a plant-specific cytoskeletal array involved in premitotic setup of the division plane, as well as the role of the nucleus and its connection to the preprophase band of microtubules. Likewise, several modeling studies point to a strong relationship between cell shape and division geometry, but the emergence of such geometric rules from the molecular and cellular pathways at play are still obscure. Yet, recent imaging technologies and genetic tools hold a lot of promise to tackle these challenges and to revisit old questions with unprecedented resolution in space and time.


Cell Division , Plant Cells , Microtubules/metabolism , Cytoskeleton/metabolism , Cell Nucleus/metabolism , Cell Nucleus/genetics
18.
Redox Biol ; 72: 103162, 2024 Jun.
Article En | MEDLINE | ID: mdl-38669864

Protein disulfide isomerases (PDIs) are involved in many intracellular and extracellular processes, including cell adhesion and cytoskeletal reorganisation, but their contribution to the regulation of fenestrations in liver sinusoidal endothelial cells (LSECs) remains unknown. Given that fenestrations are supported on a cytoskeleton scaffold, this study aimed to investigate whether endothelial PDIs regulate fenestration dynamics in primary mouse LSECs. PDIA3 and PDIA1 were found to be the most abundant among PDI isoforms in LSECs. Taking advantage of atomic force microscopy, the effects of PDIA1 or PDIA3 inhibition on the fenestrations in LSECs were investigated using a classic PDIA1 inhibitor (bepristat) and novel aromatic N-sulfonamides of aziridine-2-carboxylic acid derivatives as PDIA1 (C-3389) or PDIA3 (C-3399) inhibitors. The effect of PDIA1 inhibition on liver perfusion was studied in vivo using dynamic contrast-enhanced magnetic resonance imaging. Additionally, PDIA1 inhibitors were examined in vitro in LSECs for effects on adhesion, cytoskeleton organisation, bioenergetics, and viability. Inhibition of PDIA1 with bepristat or C-3389 significantly reduced the number of fenestrations in LSECs, while inhibition of PDIA3 with C-3399 had no effect. Moreover, the blocking of free thiols by the cell-penetrating N-ethylmaleimide, but not by the non-cell-penetrating 4-chloromercuribenzenesulfonate, resulted in LSEC defenestration. Inhibition of PDIA1 did not affect LSEC adhesion, viability, and bioenergetics, nor did it induce a clear-cut rearrangement of the cytoskeleton. However, PDIA1-dependent defenestration was reversed by cytochalasin B, a known fenestration stimulator, pointing to the preserved ability of LSECs to form new pores. Importantly, systemic inhibition of PDIA1 in vivo affected intra-parenchymal uptake of contrast agent in mice consistent with LSEC defenestration. These results revealed the role of intracellular PDIA1 in the regulation of fenestration dynamics in LSECs, and in maintaining hepatic sinusoid homeostasis.


Endothelial Cells , Liver , Protein Disulfide-Isomerases , Animals , Male , Mice , Cell Adhesion , Cells, Cultured , Cytoskeleton/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Enzyme Inhibitors/pharmacology , Liver/metabolism , Liver/cytology , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/antagonists & inhibitors
19.
Cells ; 13(7)2024 Mar 30.
Article En | MEDLINE | ID: mdl-38607046

Membrane nanotubes (NTs) are dynamic communication channels connecting spatially separated cells even over long distances and promoting the transport of different cellular cargos. NTs are also involved in the intercellular spread of different pathogens and the deterioration of some neurological disorders. Transport processes via NTs may be controlled by cytoskeletal elements. NTs are frequently observed membrane projections in numerous mammalian cell lines, including various immune cells, but their functional significance in the 'antibody factory' B cells is poorly elucidated. Here, we report that as active channels, NTs of B-lymphoma cells can mediate bidirectional mitochondrial transport, promoted by the cooperation of two different cytoskeletal motor proteins, kinesin along microtubules and myosin VI along actin, and bidirectional transport processes are also supported by the heterogeneous arrangement of the main cytoskeletal filament systems of the NTs. We revealed that despite NTs and axons being different cell extensions, the mitochondrial transport they mediate may exhibit significant similarities. Furthermore, we found that microtubules may improve the stability and lifespan of B-lymphoma-cell NTs, while F-actin strengthens NTs by providing a structural framework for them. Our results may contribute to a better understanding of the regulation of the major cells of humoral immune response to infections.


Cell Membrane Structures , Lymphoma , Nanotubes , Animals , Cytoskeleton/metabolism , Actins/metabolism , Nanotubes/chemistry , Mitochondria/metabolism , Cytoskeletal Proteins/metabolism , Lymphoma/metabolism , Mammals/metabolism
20.
PLoS One ; 19(4): e0300539, 2024.
Article En | MEDLINE | ID: mdl-38574058

Genetic and pharmacological perturbation of the cytoskeleton enhances the regenerative potential of neurons. This response requires Dual-leucine Zipper Kinase (DLK), a neuronal stress sensor that is a central regulator of axon regeneration and degeneration. The damage and repair aspects of this response are reminiscent of other cellular homeostatic systems, suggesting that a cytoskeletal homeostatic response exists. In this study, we propose a framework for understanding DLK mediated neuronal cytoskeletal homeostasis. We demonstrate that low dose nocodazole treatment activates DLK signaling. Activation of DLK signaling results in a DLK-dependent transcriptional signature, which we identify through RNA-seq. This signature includes genes likely to attenuate DLK signaling while simultaneously inducing actin regulating genes. We identify alterations to the cytoskeleton including actin-based morphological changes to the axon. These results are consistent with the model that cytoskeletal disruption in the neuron induces a DLK-dependent homeostatic mechanism, which we term the Cytoskeletal Stress Response (CSR) pathway.


Actins , Axons , Axons/metabolism , Nocodazole/pharmacology , Actins/metabolism , Leucine Zippers , Nerve Regeneration/physiology , Cytoskeleton/metabolism , Homeostasis , MAP Kinase Kinase Kinases/genetics
...