Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33.852
Filter
1.
Bone Res ; 12(1): 35, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849345

ABSTRACT

DNAX-associated protein 12 kD size (DAP12) is a dominant immunoreceptor tyrosine-based activation motif (ITAM)-signaling adaptor that activates costimulatory signals essential for osteoclastogenesis. Although several DAP12-associated receptors (DARs) have been identified in osteoclasts, including triggering receptor expressed on myeloid cells 2 (TREM-2), C-type lectin member 5 A (CLEC5A), and sialic acid-binding Ig-like lectin (Siglec)-15, their precise role in the development of osteoclasts and bone remodeling remain poorly understood. In this study, mice deficient in Trem-2, Clec5a, Siglec-15 were generated. In addition, mice double deficient in these DAR genes and FcεRI gamma chain (FcR)γ, an alternative ITAM adaptor to DAP12, were generated. Bone mass analysis was conducted on all mice. Notably, Siglec-15 deficient mice and Siglec-15/FcRγ double deficient mice exhibited mild and severe osteopetrosis respectively. In contrast, other DAR deficient mice showed normal bone phenotype. Likewise, osteoclasts from Siglec-15 deficient mice failed to form an actin ring, suggesting that Siglec-15 promotes bone resorption principally by modulating the cytoskeletal organization of osteoclasts. Furthermore, biochemical analysis revealed that Sigelc-15 activates macrophage colony-stimulating factor (M-CSF)-induced Ras-associated protein-1 (RAP1)/Ras-related C3 botulinum toxin substrate 1 (Rac1) pathway through formation of a complex with p130CAS and CrkII, leading to cytoskeletal remodeling of osteoclasts. Our data provide genetic and biochemical evidence that Siglec-15 facilitates M-CSF-induced cytoskeletal remodeling of the osteoclasts.


Subject(s)
Macrophage Colony-Stimulating Factor , Osteoclasts , Signal Transduction , rap1 GTP-Binding Proteins , Animals , Osteoclasts/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/genetics , rap1 GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/genetics , Mice , Cytoskeleton/metabolism , Mice, Knockout , Mice, Inbred C57BL , Membrane Proteins/metabolism , Membrane Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Immunoglobulins
2.
Open Biol ; 14(6): 240041, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835242

ABSTRACT

Platelets are blood cells derived from megakaryocytes that play a central role in regulating haemostasis and vascular integrity. The microtubule cytoskeleton of megakaryocytes undergoes a critical dynamic reorganization during cycles of endomitosis and platelet biogenesis. Quiescent platelets have a discoid shape maintained by a marginal band composed of microtubule bundles, which undergoes remarkable remodelling during platelet activation, driving shape change and platelet function. Disrupting or enhancing this process can cause platelet dysfunction such as bleeding disorders or thrombosis. However, little is known about the molecular mechanisms underlying the reorganization of the cytoskeleton in the platelet lineage. Recent studies indicate that the emergence of a unique platelet tubulin code and specific pathogenic tubulin mutations cause platelet defects and bleeding disorders. Frequently, these mutations exhibit dominant negative effects, offering valuable insights into both platelet disease mechanisms and the functioning of tubulins. This review will highlight our current understanding of the role of the microtubule cytoskeleton in the life and death of platelets, along with its relevance to platelet disorders.


Subject(s)
Blood Platelets , Cytoskeleton , Megakaryocytes , Microtubules , Humans , Blood Platelets/metabolism , Megakaryocytes/metabolism , Megakaryocytes/cytology , Cytoskeleton/metabolism , Microtubules/metabolism , Tubulin/metabolism , Tubulin/genetics , Animals , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/genetics , Blood Platelet Disorders/pathology , Mutation
3.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838015

ABSTRACT

O-GlcNAcase (OGA) is the only human enzyme that catalyzes the hydrolysis (deglycosylation) of O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation) from numerous protein substrates. OGA has broad implications in many challenging diseases including cancer. However, its role in cell malignancy remains mostly unclear. Here, we report that a cancer-derived point mutation on the OGA's noncatalytic stalk domain aberrantly modulates OGA interactome and substrate deglycosylation toward a specific set of proteins. Interestingly, our quantitative proteomic studies uncovered that the OGA stalk domain mutant preferentially deglycosylated protein substrates with +2 proline in the sequence relative to the O-GlcNAcylation site. One of the most dysregulated substrates is PDZ and LIM domain protein 7 (PDLIM7), which is associated with the tumor suppressor p53. We found that the aberrantly deglycosylated PDLIM7 suppressed p53 gene expression and accelerated p53 protein degradation by promoting the complex formation with E3 ubiquitin ligase MDM2. Moreover, deglycosylated PDLIM7 significantly up-regulated the actin-rich membrane protrusions on the cell surface, augmenting the cancer cell motility and aggressiveness. These findings revealed an important but previously unappreciated role of OGA's stalk domain in protein substrate recognition and functional modulation during malignant cell progression.


Subject(s)
Cytoskeleton , LIM Domain Proteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Cytoskeleton/metabolism , Acetylglucosamine/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Line, Tumor , Glycosylation , Hydrolysis , Mutation , Cell Movement , Antigens, Neoplasm , Hyaluronoglucosaminidase , Histone Acetyltransferases
4.
Science ; 384(6700): eadk5511, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843314

ABSTRACT

Fundamental limits of cellular deformations, such as hyperextension of a living cell, remain poorly understood. Here, we describe how the single-celled protist Lacrymaria olor, a 40-micrometer cell, is capable of reversibly and repeatably extending its necklike protrusion up to 1200 micrometers in 30 seconds. We discovered a layered cortical cytoskeleton and membrane architecture that enables hyperextensions through the folding and unfolding of cellular-scale origami. Physical models of this curved crease origami display topological singularities, including traveling developable cones and cytoskeletal twisted domain walls, which provide geometric control of hyperextension. Our work unravels how cell geometry encodes behavior in single cells and provides inspiration for geometric control in microrobotics and deployable architectures.


Subject(s)
Cytoskeleton , Cell Membrane , Single-Cell Analysis
5.
Science ; 384(6700): 1064-1065, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843349

ABSTRACT

Lacrymaria olor cytoskeleton and membrane "origami" enables rapid cell hyperextension.


Subject(s)
Cell Membrane , Cytoskeleton , Cell Membrane/metabolism
6.
World J Surg Oncol ; 22(1): 152, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849867

ABSTRACT

BACKGROUND: Although sorafenib has been consistently used as a first-line treatment for advanced hepatocellular carcinoma (HCC), most patients will develop resistance, and the mechanism of resistance to sorafenib needs further study. METHODS: Using KAS-seq technology, we obtained the ssDNA profiles within the whole genome range of SMMC-7721 cells treated with sorafenib for differential analysis. We then intersected the differential genes obtained from the analysis of hepatocellular carcinoma patients in GSE109211 who were ineffective and effective with sorafenib treatment, constructed a PPI network, and obtained hub genes. We then analyzed the relationship between the expression of these genes and the prognosis of hepatocellular carcinoma patients. RESULTS: In this study, we identified 7 hub ERGs (ACTB, CFL1, ACTG1, ACTN1, WDR1, TAGLN2, HSPA8) related to drug resistance, and these genes are associated with the cytoskeleton. CONCLUSIONS: The cytoskeleton is associated with sorafenib resistance in hepatocellular carcinoma. Using KAS-seq to analyze the early changes in tumor cells treated with drugs is feasible for studying the drug resistance of tumors, which provides reference significance for future research.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Drug Resistance, Neoplasm , Liver Neoplasms , Sorafenib , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Prognosis , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Cytoskeleton/drug effects , Cytoskeleton/pathology , Cytoskeleton/metabolism , Biomarkers, Tumor/genetics , Tumor Cells, Cultured , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Profiling
7.
Commun Biol ; 7(1): 708, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851810

ABSTRACT

Robotically assisted proteomics provides insights into the regulation of multiple proteins achieving excellent spatial resolution. However, developing an effective method for spatially resolved quantitative proteomics of formalin fixed paraffin embedded tissue (FFPE) in an accessible and economical manner remains challenging. We introduce non-robotic In-insert FFPE proteomics approach, combining glass insert FFPE tissue processing with spatial quantitative data-independent mass spectrometry (DIA). In-insert approach identifies 450 proteins from a 5 µm thick breast FFPE tissue voxel with 50 µm lateral dimensions covering several tens of cells. Furthermore, In-insert approach associated a keratin series and moesin (MOES) with prolactin-induced protein (PIP) indicating their prolactin and/or estrogen regulation. Our data suggest that PIP is a spatial biomarker for hormonally triggered cytoskeletal remodeling, potentially useful for screening hormonally affected hotspots in breast tissue. In-insert proteomics represents an alternative FFPE processing method, requiring minimal laboratory equipment and skills to generate spatial proteotype repositories from FFPE tissue.


Subject(s)
Biomarkers , Cytoskeleton , Paraffin Embedding , Proteomics , Tissue Fixation , Humans , Proteomics/methods , Cytoskeleton/metabolism , Female , Biomarkers/metabolism , Tissue Fixation/methods , Prolactin/metabolism , Formaldehyde/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Membrane Transport Proteins
8.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732188

ABSTRACT

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.


Subject(s)
Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/pathology , Animals , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism , Cell Movement , Cell Proliferation
9.
Science ; 384(6699): eadd6260, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38815015

ABSTRACT

Abnormal calcium signaling is a central pathological component of Alzheimer's disease (AD). Here, we describe the identification of a class of compounds called ReS19-T, which are able to restore calcium homeostasis in cell-based models of tau pathology. Aberrant tau accumulation leads to uncontrolled activation of store-operated calcium channels (SOCCs) by remodeling septin filaments at the cell cortex. Binding of ReS19-T to septins restores filament assembly in the disease state and restrains calcium entry through SOCCs. In amyloid-ß and tau-driven mouse models of disease, ReS19-T agents restored synaptic plasticity, normalized brain network activity, and attenuated the development of both amyloid-ß and tau pathology. Our findings identify the septin cytoskeleton as a potential therapeutic target for the development of disease-modifying AD treatments.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Calcium , Disease Models, Animal , Homeostasis , Neuroprotective Agents , Septins , tau Proteins , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Septins/metabolism , Mice , Calcium/metabolism , tau Proteins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Amyloid beta-Peptides/metabolism , Humans , Neuronal Plasticity/drug effects , Calcium Signaling/drug effects , Calcium Channels/metabolism , Cytoskeleton/metabolism , Cytoskeleton/drug effects
10.
Exp Cell Res ; 439(1): 114059, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38705228

ABSTRACT

Filopodia are thin, actin-rich projection from the plasma membrane that promote cancer cell invasion and migration. Sex-determining region Y-related high-mobility group-box 4 (SOX4) is a crucial transcription factor that plays a role in the development and metastasis of colorectal cancer (CRC). However, the involvement of SOX4 in cytoskeleton remodeling in CRC remains unknown. For the first time, we demonstrate that SOX4 is a potent regulator of filopodia formation in CRC cells. Overexpression of SOX4 protein enhances both migration and invasion ability of HCT116, and CACO2 cells, which is relevant to the metastasis. Furthermore, through phalloidin staining, cytoskeleton re-assembly was observed in SOX4-modified cell lines. Enhanced expression of SOX4 increased the number and length of filopodia on cell surface. In contrast, silencing SOX4 in SW620 cells with higher endogenous expression of SOX4, impeded the filopodia formation. Moreover, SOX4 was found to be positively regulating the expression of central regulators of actin cytoskeleton - N-Wiskott-Aldrich syndrome protein (N-WASP); WAVE2; Actin related proteins, ARP2 and ARP3. Inhibiting the N-WASP/ARP2/3 pathway diminishes the filopodia formation and the migration of CRC cells. These results indicate the crucial role of SOX4 in the regulation of filopodia formation mediated by N-WASP/ARP2/3 pathway in CRC cells.


Subject(s)
Actin-Related Protein 2-3 Complex , Cell Movement , Colorectal Neoplasms , Cytoskeleton , Pseudopodia , SOXC Transcription Factors , Wiskott-Aldrich Syndrome Protein, Neuronal , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , SOXC Transcription Factors/metabolism , SOXC Transcription Factors/genetics , Cell Movement/genetics , Actin-Related Protein 2-3 Complex/metabolism , Actin-Related Protein 2-3 Complex/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , Cytoskeleton/metabolism , Pseudopodia/metabolism , Caco-2 Cells , Signal Transduction , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , HCT116 Cells , Actin Cytoskeleton/metabolism
11.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38738619

ABSTRACT

Synaptic development requires multiple signaling pathways to ensure successful connections. Transmembrane receptors are optimally positioned to connect the synapse and the rest of the neuron, often acting as synaptic organizers to synchronize downstream events. One such organizer, the LDL receptor-related protein LRP4, is a cell surface receptor that has been most well-studied postsynaptically at mammalian neuromuscular junctions. Recent work, however, identified emerging roles, but how LRP4 acts as a presynaptic organizer and the downstream mechanisms of LRP4 are not well understood. Here, we show that LRP4 functions presynaptically at Drosophila neuromuscular synapses, acting in motoneurons to instruct pre- and postsynaptic development. Loss of presynaptic LRP4 results in multiple defects, impairing active zone organization, synapse growth, physiological function, microtubule organization, synaptic ultrastructure and synapse maturation. We further demonstrate that LRP4 promotes most aspects of presynaptic development via a downstream SR-protein kinase, SRPK79D. These data demonstrate a function for presynaptic LRP4 as a peripheral synaptic organizer, highlight a downstream mechanism conserved with its CNS function in Drosophila, and underscore previously unappreciated but important developmental roles for LRP4 in cytoskeletal organization, synapse maturation and active zone organization.


Subject(s)
Cytoskeleton , Drosophila Proteins , Neuromuscular Junction , Synapses , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Neuromuscular Junction/metabolism , Synapses/metabolism , Cytoskeleton/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Motor Neurons/metabolism , Drosophila , Neurons/metabolism , Neurons/cytology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction
12.
Genes (Basel) ; 15(5)2024 May 16.
Article in English | MEDLINE | ID: mdl-38790262

ABSTRACT

Intermediate filaments (IFs) are integral components of the cytoskeleton which provide cells with tissue-specific mechanical properties and are involved in a plethora of cellular processes. Unfortunately, due to their intricate architecture, the 3D structure of the complete molecule of IFs has remained unresolved. Even though most of the rod domain structure has been revealed by means of crystallographic analyses, the flanked head and tail domains are still mostly unknown. Only recently have studies shed light on head or tail domains of IFs, revealing certainsecondary structures and conformational changes during IF assembly. Thus, a deeper understanding of their structure could provide insights into their function.


Subject(s)
Intermediate Filaments , Protein Domains , Intermediate Filaments/metabolism , Intermediate Filaments/genetics , Intermediate Filaments/chemistry , Humans , Animals , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Cytoskeleton , Models, Molecular
13.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793550

ABSTRACT

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.


Subject(s)
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
14.
Ecotoxicol Environ Saf ; 279: 116502, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38788563

ABSTRACT

BACKGROUND: Despite the known reproductive toxicity induced by triptolide (TP) exposure, the regulatory mechanism underlying testicular vacuolization injury caused by TP remains largely obscure. METHODS: Male mice were subjected to TP at doses of 15, 30, and 60 µg/kg for 35 consecutive days. Primary Sertoli cells were isolated from 20-day-old rat testes and exposed to TP at concentrations of 0, 40, 80, 160, 320, and 640 nM. A Biotin tracer assay was conducted to assess the integrity of the blood-testis barrier (BTB). Transepithelial electrical resistance (TER) assays were employed to investigate BTB function in primary Sertoli cells. Histological structures of the testes and epididymides were stained with hematoxylin and eosin (H&E). The expression and localization of relevant proteins or pathways were assessed through Western blotting or immunofluorescence staining. RESULTS: TP exposure led to dose-dependent testicular injuries, characterized by a decreased organ coefficient, reduced sperm concentration, and the formation of vacuolization damage. Furthermore, TP exposure disrupted BTB integrity by reducing the expression levels of tight junction (TJ) proteins in the testes without affecting basal ectoplasmic specialization (basal ES) proteins. Through the TER assay, we identified that a TP concentration of 160 nM was optimal for elucidating BTB function in primary Sertoli cells, correlating with reductions in TJ protein expression. Moreover, TP exposure induced changes in the distribution of the BTB and cytoskeleton-associated proteins in primary Sertoli cells. By activating the AKT/mTOR signaling pathway, TP exposure disturbed the balance between mTORC1 and mTORC2, ultimately compromising BTB integrity in Sertoli cells. CONCLUSION: This investigation sheds light on the impacts of TP exposure on testes, elucidating the mechanism by which TP exposure leads to testicular vacuolization injury and offering valuable insights into comprehending the toxic effects of TP exposure on testes.


Subject(s)
Blood-Testis Barrier , Cytoskeleton , Diterpenes , Epoxy Compounds , Phenanthrenes , Proto-Oncogene Proteins c-akt , Sertoli Cells , Signal Transduction , TOR Serine-Threonine Kinases , Testis , Male , Animals , Sertoli Cells/drug effects , Sertoli Cells/pathology , Diterpenes/toxicity , Phenanthrenes/toxicity , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Testis/drug effects , Testis/pathology , Epoxy Compounds/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Mice , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/pathology , Cytoskeleton/drug effects , Rats , Vacuoles/drug effects , Rats, Sprague-Dawley
15.
Mol Biol Cell ; 35(7): ar91, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38758658

ABSTRACT

Mechanical cues from the tissue microenvironment, such as the stiffness of the extracellular matrix, modulate cellular forms and functions. As numerous studies have shown, this modulation depends on the stiffness-dependent remodeling of cytoskeletal elements. In contrast, very little is known about how the intracellular organelles such as mitochondria respond to matrix stiffness and whether their form, function, and localization change accordingly. Here, we performed an extensive quantitative characterization of mitochondrial morphology, subcellular localization, dynamics, and membrane tension on soft and stiff matrices. This characterization revealed that while matrix stiffness affected all these aspects, matrix stiffening most distinctively led to an increased perinuclear clustering of mitochondria. Subsequently, we could identify the matrix stiffness-sensitive perinuclear localization of filamin as the key factor dictating this perinuclear clustering. The perinuclear and peripheral mitochondrial populations differed in their motility on soft matrix but surprisingly they did not show any difference on stiff matrix. Finally, perinuclear mitochondrial clustering appeared to be crucial for the nuclear localization of RUNX2 and hence for priming human mesenchymal stem cells towards osteogenesis on a stiff matrix. Taken together, we elucidate a dependence of mitochondrial localization on matrix stiffness, which possibly enables a cell to adapt to its microenvironment.


Subject(s)
Extracellular Matrix , Mesenchymal Stem Cells , Mitochondria , Humans , Extracellular Matrix/metabolism , Mitochondria/metabolism , Mesenchymal Stem Cells/metabolism , Cytoskeleton/metabolism , Filamins/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Cell Nucleus/metabolism , Osteogenesis/physiology , Cell Differentiation/physiology
16.
Adv Exp Med Biol ; 1452: 1-19, 2024.
Article in English | MEDLINE | ID: mdl-38805122

ABSTRACT

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.


Subject(s)
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
17.
J Cell Sci ; 137(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38563209

ABSTRACT

Actin is well known for its cytoskeletal functions, where it helps to control and maintain cell shape and architecture, as well as regulating cell migration and intracellular cargo transport, among others. However, actin is also prevalent in the nucleus, where genome-regulating roles have been described, including it being part of chromatin-remodeling complexes. More recently, with the help of advances in microscopy techniques and specialized imaging probes, direct visualization of nuclear actin filament dynamics has helped elucidate new roles for nuclear actin, such as in cell cycle regulation, DNA replication and repair, chromatin organization and transcriptional condensate formation. In this Cell Science at a Glance article, we summarize the known signaling events driving the dynamic assembly of actin into filaments of various structures within the nuclear compartment for essential genome functions. Additionally, we highlight the physiological role of nuclear F-actin in meiosis and early embryonic development.


Subject(s)
Actins , Cell Nucleus , Actins/metabolism , Cell Nucleus/metabolism , Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Cell Cycle
18.
Cells ; 13(7)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38607046

ABSTRACT

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.


Subject(s)
Cell Membrane Structures , Lymphoma , Nanotubes , Animals , Cytoskeleton/metabolism , Actins/metabolism , Nanotubes/chemistry , Mitochondria/metabolism , Cytoskeletal Proteins/metabolism , Lymphoma/metabolism , Mammals/metabolism
19.
Nat Commun ; 15(1): 3198, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609383

ABSTRACT

In order to shape a tissue, individual cell-based mechanical forces have to be integrated into a global force pattern. Over the last decades, the importance of actomyosin contractile arrays, which are the key constituents of various morphogenetic processes, has been established for many tissues. Recent studies have demonstrated that the microtubule cytoskeleton mediates folding and elongation of the epithelial sheet during Drosophila morphogenesis, placing microtubule mechanics on par with actin-based processes. While these studies establish the importance of both cytoskeletal systems during cell and tissue rearrangements, a mechanistic understanding of their functional hierarchy is currently missing. Here, we dissect the individual roles of these two key generators of mechanical forces during epithelium elongation in the developing Drosophila wing. We show that wing extension, which entails columnar-to-cuboidal cell shape remodeling in a cell-autonomous manner, is driven by anisotropic cell expansion caused by the remodeling of the microtubule cytoskeleton from apico-basal to planarly polarized. Importantly, cell and tissue elongation is not associated with Myosin activity. Instead, Myosin II exhibits a homeostatic role, as actomyosin contraction balances polarized microtubule-based forces to determine the final cell shape. Using a reductionist model, we confirm that pairing microtubule and actomyosin-based forces is sufficient to recapitulate cell elongation and the final cell shape. These results support a hierarchical mechanism whereby microtubule-based forces in some epithelial systems prime actomyosin-generated forces.


Subject(s)
Actomyosin , Microtubules , Animals , Actin Cytoskeleton , Cytoskeleton , Drosophila
20.
Int J Mol Sci ; 25(7)2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38612685

ABSTRACT

Endometriosis is a complex gynecological disease that affects more than 10% of women in their reproductive years. While surgery can provide temporary relief from women's pain, symptoms often return in as many as 75% of cases within two years. Previous literature has contributed to theories about the development of endometriosis; however, the exact pathogenesis and etiology remain elusive. We conducted a preliminary investigation into the influence of primary endometrial cells (ECs) on the development and progression of endometriosis. In vitro studies, they were involved in inducing Lipopolysaccharide (LPS) in rat-isolated primary endometrial cells, which resulted in increased nuclear factor-kappa B (NF-κB) and vascular endothelial growth factor (VEGF) mRNA gene expression (quantitative polymerase chain reaction analysis, qPCR) and protein expression (western blot analysis). Additionally, in vivo studies utilized autogenic and allogeneic transplantations (rat to rat) to investigate endometriosis-like lesion cyst size, body weight, protein levels (immunohistochemistry), and mRNA gene expression. These studies demonstrated that estrogen upregulates the gene and protein regulation of cytoskeletal (CK)-18, transforming growth factor-ß (TGF-ß), VEGF, and tumor necrosis factor (TNF)-α, particularly in the peritoneum. These findings may influence cell proliferation, angiogenesis, fibrosis, and inflammation markers. Consequently, this could exacerbate the occurrence and progression of endometriosis.


Subject(s)
Endometriosis , Female , Humans , Animals , Rats , Vascular Endothelial Growth Factor A/genetics , Cell Proliferation , Cytoskeleton , RNA, Messenger
SELECTION OF CITATIONS
SEARCH DETAIL
...