Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.835
Filter
1.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(4): 886-892, 2024 Jul 20.
Article in Chinese | MEDLINE | ID: mdl-39170023

ABSTRACT

Objective: To investigate the effect of the loss of myeloid-derived growth factor (Mydgf) on the transformation of cardiac fibroblasts into myofibroblasts after myocardial infarction (MI). Methods: Two adult mouse groups, including a wild-type (WT) group and another group with Mydgf knockout (Mydgf-KO), were examined in the study. The mice in these two groups were tested for their cardiac function by measuring left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) (n=10). Quantitative real-time PCR (qRT-PCR) (n=3) was performed to determine the mRNA expression levels of myofibroblast markers, including α-smooth muscle actin (α-SMA), periostin (postn), type Ⅷ collagen (col8al), and connective tissue growth factor (ctgf). Western blot (n=3) was performed to verify the protein expression levels of α-SMA. MI modeling was performed on the WT and the Mydgf-KO mice. Postoperative LVEF and LVFS (n=10) were then measured. The hearts were harvested and Masson staining was performed to determine the infarcted area (n=10). The heart samples of Mydgf-KO and WT mice were collected at d 7 and d 14 after MI, respectively, to verify the expression of myofibroblast markers (n=3). Results: Compared with WT mice, LVEF and LVFS in adult Mydgf-KO mice showed no significant changes (all P>0.05). However, the mRNA levels of α-SMA and postn were upregulated, and α-SMA protein expression was also increased (all P<0.05). After MI, compared with WT mice, LVEF and LVFS in Mydgf-KO mice decreased, and the infarcted area increased significantly (all P<0.05). Furthermore, mRNA levels of α-SMA, col8al, postn, and ctgf were increased in Mydgf-KO mice. In addition, the α-SMA protein expression level was upregulated and α-SMA-positive fibroblasts were increased (P<0.05). Conclusion: Mydgf deletion promotes the transformation of cardiac fibroblasts into myofibroblasts and aggravates myocardial fibrosis after MI.


Subject(s)
Connective Tissue Growth Factor , Fibrosis , Mice, Knockout , Myocardial Infarction , Myofibroblasts , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Mice , Myofibroblasts/metabolism , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Actins/metabolism , Actins/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Myocardium/metabolism , Myocardium/pathology , Mice, Inbred C57BL , Male , Fibroblasts/metabolism
2.
Science ; 385(6711): eado2032, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39172837

ABSTRACT

Clathrin-mediated endocytosis has characteristic features in neuronal dendrites and presynapses, but how membrane proteins are internalized along the axon shaft remains unclear. We focused on clathrin-coated structures and endocytosis along the axon initial segment (AIS) and their relationship to the periodic actin-spectrin scaffold that lines the axonal plasma membrane. A combination of super-resolution microscopy and platinum-replica electron microscopy on cultured neurons revealed that AIS clathrin-coated pits form within "clearings", circular areas devoid of actin-spectrin mesh. Actin-spectrin scaffold disorganization increased clathrin-coated pit formation. Cargo uptake and live-cell imaging showed that AIS clathrin-coated pits are particularly stable. Neuronal plasticity-inducing stimulation triggered internalization of the clathrin-coated pits through polymerization of branched actin around them. Thus, spectrin and actin regulate clathrin-coated pit formation and scission to control endocytosis at the AIS.


Subject(s)
Actins , Axons , Clathrin , Endocytosis , Spectrin , Spectrin/metabolism , Animals , Actins/metabolism , Axons/metabolism , Clathrin/metabolism , Rats , Coated Pits, Cell-Membrane/metabolism , Neuronal Plasticity , Cells, Cultured , Cell Membrane/metabolism , Mice , Neurons/metabolism , Actin Cytoskeleton/metabolism , Clathrin-Coated Vesicles/metabolism
3.
Transl Vis Sci Technol ; 13(8): 22, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39133495

ABSTRACT

Purpose: The purpose of this study was to evaluate the safety and efficacy of topical losartan in the therapeutic treatment of established corneal scaring fibrosis at 1 month after alkali burn in rabbits. Methods: Standardized alkali burns were performed in 1 eye of 24 rabbits with 0.75N NaOH for 15 seconds. Corneas were allowed to heal and develop scaring of the cornea for 1 month. Twelve eyes per group were treated with 50 µL of topical 0.8 mg/mL losartan in balanced salt solution (BSS), pH 7.0, and 12 eyes were treated with vehicle BSS 6 times per day. Six corneas were analyzed at 1 week or 1 month in each group. Standardized slit lamp photographs were obtained at the end point for each cornea and opacity was quantitated using ImageJ. Corneoscleral rims were cryofixed in optimum cutting temperature (OCT) solution and combined duplex immunohistochemistry for myofibroblast marker alpha-smooth muscle actin (α-SMA), mesenchymal cell marker vimentin, and TUNEL assay for apoptosis was performed on all corneas. Results: Topical losartan was effective in the treatment of established stromal fibrosis following alkali burn injury to the rabbit cornea. Stromal myofibroblast density was decreased and stromal cell apoptosis was increased (included both α-SMA-positive myofibroblasts and α-SMA-negative, vimentin-positive cells) at both 1 week and 1 month in the topical losartan-treated compared with vehicle-treated groups. Conclusions: Topical losartan is effective in the treatment of established stromal fibrosis in rabbits. Most myofibroblasts disappear from the stroma within the first month of losartan treatment. Longer treatment with topical losartan is needed to allow time for corneal fibroblast regeneration of the epithelial basement membrane (in coordination with epithelial cells) and the removal of disordered extracellular matrix produced by myofibroblasts.


Subject(s)
Burns, Chemical , Eye Burns , Fibrosis , Losartan , Animals , Rabbits , Losartan/pharmacology , Losartan/administration & dosage , Losartan/therapeutic use , Fibrosis/drug therapy , Burns, Chemical/drug therapy , Burns, Chemical/pathology , Eye Burns/drug therapy , Eye Burns/pathology , Eye Burns/chemically induced , Disease Models, Animal , Apoptosis/drug effects , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Sodium Hydroxide , Corneal Diseases/drug therapy , Corneal Diseases/pathology , Ophthalmic Solutions/therapeutic use , Ophthalmic Solutions/administration & dosage , Cornea/drug effects , Cornea/pathology , In Situ Nick-End Labeling , Myofibroblasts/drug effects , Myofibroblasts/pathology , Actins/metabolism , Male , Corneal Stroma/drug effects , Corneal Stroma/pathology , Corneal Stroma/metabolism , Administration, Topical , Vimentin/metabolism , Wound Healing/drug effects
4.
Sci Rep ; 14(1): 18752, 2024 08 13.
Article in English | MEDLINE | ID: mdl-39138242

ABSTRACT

Subretinal fibrosis is a major untreatable cause of poor outcomes in neovascular age-related macular degeneration. Mouse models of subretinal fibrosis all possess a degree of invasiveness and tissue damage not typical of fibrosis progression. This project characterises JR5558 mice as a model to study subretinal fibrosis. Fundus and optical coherence tomography (OCT) imaging was used to non-invasively track lesions. Lesion number and area were quantified with ImageJ. Retinal sections, wholemounts and Western blots were used to characterise alterations. Subretinal lesions expand between 4 and 8 weeks and become established in size and location around 12 weeks. Subretinal lesions were confirmed to be fibrotic, including various cell populations involved in fibrosis development. Müller cell processes extended from superficial retina into subretinal lesions at 8 weeks. Western blotting revealed increases in fibronectin (4 wk and 8 wk, p < 0.001), CTGF (20 wks, p < 0.001), MMP2 (12 wks and 20 wks p < 0.05), αSMA (12 wks and 20 wks p < 0.05) and GFAP (8 wk and 12 wk, p ≤ 0.01), consistent with our immunofluorescence results. Intravitreal injection of Aflibercept reduced subretinal lesion growth. Our study provides evidence JR5558 mice have subretinal fibrotic lesions that grow between 4 and 8 weeks and confirms this line to be a good model to study subretinal fibrosis development and assess treatment options.


Subject(s)
Disease Models, Animal , Fibrosis , Retina , Tomography, Optical Coherence , Animals , Mice , Tomography, Optical Coherence/methods , Retina/pathology , Retina/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Fibronectins/metabolism , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Connective Tissue Growth Factor/metabolism , Connective Tissue Growth Factor/genetics , Macular Degeneration/pathology , Macular Degeneration/metabolism , Matrix Metalloproteinase 2/metabolism , Intravitreal Injections , Glial Fibrillary Acidic Protein/metabolism , Actins/metabolism , Mice, Inbred C57BL , Recombinant Fusion Proteins
5.
Methods Mol Biol ; 2828: 147-157, 2024.
Article in English | MEDLINE | ID: mdl-39147976

ABSTRACT

Normal-sized cells of Dictyostelium build up a front-tail polarity when they respond to a gradient of chemoattractant. To challenge the polarity-generating system, cells are fused to study the chemotactic response of oversized cells that extend multiple fronts toward the source of attractant. An aspect that can be explored in these cells is the relationship of spontaneously generated actin waves to actin reorganization in response to chemoattractant.


Subject(s)
Chemotaxis , Dictyostelium , Dictyostelium/physiology , Dictyostelium/cytology , Chemotactic Factors/pharmacology , Chemotactic Factors/metabolism , Actins/metabolism , Cell Fusion/methods , Giant Cells/cytology , Giant Cells/metabolism , Cell Polarity
6.
Cell Mol Life Sci ; 81(1): 353, 2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39154297

ABSTRACT

The morphology of dendritic spines, the postsynaptic compartment of most excitatory synapses, decisively modulates the function of neuronal circuits as also evident from human brain disorders associated with altered spine density or morphology. Actin filaments (F-actin) form the backbone of spines, and a number of actin-binding proteins (ABP) have been implicated in shaping the cytoskeleton in mature spines. Instead, only little is known about the mechanisms that control the reorganization from unbranched F-actin of immature spines to the complex, highly branched cytoskeleton of mature spines. Here, we demonstrate impaired spine maturation in hippocampal neurons upon genetic inactivation of cyclase-associated protein 1 (CAP1) and CAP2, but not of CAP1 or CAP2 alone. We found a similar spine maturation defect upon overactivation of inverted formin 2 (INF2), a nucleator of unbranched F-actin with hitherto unknown synaptic function. While INF2 overactivation failed in altering spine density or morphology in CAP-deficient neurons, INF2 inactivation largely rescued their spine defects. From our data we conclude that CAPs inhibit INF2 to induce spine maturation. Since we previously showed that CAPs promote cofilin1-mediated cytoskeletal remodeling in mature spines, we identified them as a molecular switch that control transition from filopodia-like to mature spines.


Subject(s)
Cytoskeletal Proteins , Dendritic Spines , Formins , Hippocampus , Microfilament Proteins , Dendritic Spines/metabolism , Animals , Mice , Formins/metabolism , Formins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Hippocampus/metabolism , Hippocampus/cytology , Cells, Cultured , Neurons/metabolism , Actins/metabolism , Actin Cytoskeleton/metabolism , Mice, Knockout , Humans , Carrier Proteins
7.
Cell Mol Life Sci ; 81(1): 358, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158722

ABSTRACT

Long-term synaptic plasticity is typically associated with morphological changes in synaptic connections. However, the molecular mechanisms coupling functional and structural aspects of synaptic plasticity are still poorly defined. The catalytic activity of type I phosphoinositide-3-kinase (PI3K) is required for specific forms of synaptic plasticity, such as NMDA receptor-dependent long-term potentiation (LTP) and mGluR-dependent long-term depression (LTD). On the other hand, PI3K signaling has been linked to neuronal growth and synapse formation. Consequently, PI3Ks are promising candidates to coordinate changes in synaptic strength with structural remodeling of synapses. To investigate this issue, we targeted individual regulatory subunits of type I PI3Ks in hippocampal neurons and employed a combination of electrophysiological, biochemical and imaging techniques to assess their role in synaptic plasticity. We found that a particular regulatory isoform, p85α, is selectively required for LTP. This specificity is based on its BH domain, which engages the small GTPases Rac1 and Cdc42, critical regulators of the actin cytoskeleton. Moreover, cofilin, a key regulator of actin dynamics that accumulates in dendritic spines after LTP induction, failed to do so in the absence of p85α or when its BH domain was overexpressed as a dominant negative construct. Finally, in agreement with this convergence on actin regulatory mechanisms, the presence of p85α in the PI3K complex determined the extent of actin polymerization in dendritic spines during LTP. Therefore, this study reveals a molecular mechanism linking structural and functional synaptic plasticity through the coordinate action of PI3K catalytic activity and a specific isoform of the regulatory subunits.


Subject(s)
Actin Depolymerizing Factors , Actins , Dendritic Spines , Hippocampus , Long-Term Potentiation , Animals , Dendritic Spines/metabolism , Long-Term Potentiation/physiology , Actins/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Actin Depolymerizing Factors/metabolism , Rats , rac1 GTP-Binding Protein/metabolism , Synapses/metabolism , Polymerization , cdc42 GTP-Binding Protein/metabolism , Neuronal Plasticity/physiology , Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Neurons/metabolism , Signal Transduction , Mice , Cells, Cultured
8.
Cell Mol Life Sci ; 81(1): 354, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158743

ABSTRACT

Mature neurons have stable dendritic architecture, which is essential for the nervous system to operate correctly. The ability to undergo structural plasticity, required to support adaptive processes like memory formation, is still present in mature neurons. It is unclear what molecular and cellular processes control this delicate balance between dendritic structural plasticity and stabilization. Failures in the preservation of optimal dendrite structure due to atrophy or maladaptive plasticity result in abnormal connectivity and are associated with various neurological diseases. Vascular endothelial growth factor D (VEGFD) is critical for the maintenance of mature dendritic trees. Here, we describe how VEGFD affects the neuronal cytoskeleton and demonstrate that VEGFD exerts its effects on dendrite stabilization by influencing the actin cortex and reducing microtubule dynamics. Further, we found that during synaptic activity-induced structural plasticity VEGFD is downregulated. Our findings revealed that VEGFD, acting on its cognate receptor VEGFR3, opposes structural changes by negatively regulating dendrite growth in cultured hippocampal neurons and in vivo in the adult mouse hippocampus with consequences on memory formation. A phosphoproteomic screening identified several regulatory proteins of the cytoskeleton modulated by VEGFD. Among the actin cortex-associated proteins, we found that VEGFD induces dephosphorylation of ezrin at tyrosine 478 via activation of the striatal-enriched protein tyrosine phosphatase (STEP). Activity-triggered structural plasticity of dendrites was impaired by expression of a phospho-deficient mutant ezrin in vitro and in vivo. Thus, VEGFD governs the equilibrium between stabilization and plasticity of dendrites by acting as a molecular brake of structural remodeling.


Subject(s)
Dendrites , Hippocampus , Neuronal Plasticity , Signal Transduction , Animals , Dendrites/metabolism , Mice , Hippocampus/metabolism , Hippocampus/cytology , Mice, Inbred C57BL , Cells, Cultured , Cytoskeleton/metabolism , Male , Neurons/metabolism , Neurons/cytology , Actins/metabolism , Phosphorylation , Microtubules/metabolism
9.
Bull Exp Biol Med ; 177(2): 185-189, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39090469

ABSTRACT

Inflammation plays a crucial role in progression of fibrosis. Epoxyeicosatrienoic acids (EET) have multiple protective effects in different diseases, but their ability to inhibit the development of LPS-induced fibrosis remains unknown. The potential therapeutic effects of 11,12-EET were studied in in vitro model of LPS-induced fibrosis. Mouse embryonic fibroblast cells NIH/3T3 were pre-incubated with 1 µM 11,12-EET and/or a structural analogue and selective EET antagonist 14,15-epoxyeicosa-5(Z)-enoic acid before exposing to LPS. The effect of EET was evaluated by the protein and mRNA expression of NF-κB, collagens I and III, and α-smooth muscle actin by Western blotting and quantitative reverse transcription PCR, respectively. LPS provoked inflammation and fibrosis-like changes accompanied by elevated expression of NF-κB and collagens in NIH/3T3 cells. We also studied the effects of 11,12-EET on the A2AR and PI3K/Akt signaling pathways in intact and LPS-treated NIH/3T3 cells. 11,12-EET prevented inflammation and fibrosis-like changes through up-regulation of A2AR and PI3K/Akt signaling pathways. Our findings demonstrate the potential antifibrotic effects of 11,12-EET, which can be natural antagonists of tissue fibrosis.


Subject(s)
8,11,14-Eicosatrienoic Acid , Fibrosis , Lipopolysaccharides , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Mice , Lipopolysaccharides/toxicity , Signal Transduction/drug effects , NIH 3T3 Cells , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Fibrosis/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , 8,11,14-Eicosatrienoic Acid/analogs & derivatives , 8,11,14-Eicosatrienoic Acid/pharmacology , NF-kappa B/metabolism , Actins/metabolism , Actins/genetics , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology
10.
Nat Commun ; 15(1): 6824, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122694

ABSTRACT

MICAL proteins represent a unique family of actin regulators crucial for synapse development, membrane trafficking, and cytokinesis. Unlike classical actin regulators, MICALs catalyze the oxidation of specific residues within actin filaments to induce robust filament disassembly. The potent activity of MICALs requires tight control to prevent extensive damage to actin cytoskeleton. However, the molecular mechanism governing MICALs' activity regulation remains elusive. Here, we report the cryo-EM structure of MICAL1 in the autoinhibited state, unveiling a head-to-tail interaction that allosterically blocks enzymatic activity. The structure also reveals the assembly of C-terminal domains via a tripartite interdomain interaction, stabilizing the inhibitory conformation of the RBD. Our structural, biochemical, and cellular analyses elucidate a multi-step mechanism to relieve MICAL1 autoinhibition in response to the dual-binding of two Rab effectors, revealing its intricate activity regulation mechanisms. Furthermore, our mutagenesis study of MICAL3 suggests the conserved autoinhibition and relief mechanisms among MICALs.


Subject(s)
Actins , Cryoelectron Microscopy , Mixed Function Oxygenases , Humans , Actins/metabolism , Mixed Function Oxygenases/metabolism , Mixed Function Oxygenases/chemistry , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/chemistry , Protein Binding , Actin Cytoskeleton/metabolism , Models, Molecular , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Protein Domains , Calponins
11.
Sci Rep ; 14(1): 18384, 2024 08 08.
Article in English | MEDLINE | ID: mdl-39117762

ABSTRACT

The fundamental question of how forces are generated in a motile cell, a lamellipodium, and a comet tail is the subject of this note. It is now well established that cellular motility results from the polymerization of actin, the most abundant protein in eukaryotic cells, into an interconnected set of filaments. We portray this process in a continuum mechanics framework, claiming that polymerization promotes a mechanical swelling in a narrow zone around the nucleation loci, which ultimately results in cellular or bacterial motility. To this aim, a new paradigm in continuum multi-physics has been designed, departing from the well-known theory of Larché-Cahn chemo-transport-mechanics. In this note, we set up the theory of network growth and compare the outcomes of numerical simulations with experimental evidence.


Subject(s)
Actins , Cell Movement , Actins/metabolism , Models, Biological , Actin Cytoskeleton/metabolism , Pseudopodia/metabolism , Pseudopodia/physiology , Biomechanical Phenomena , Polymerization
12.
Int J Immunopathol Pharmacol ; 38: 3946320241274225, 2024.
Article in English | MEDLINE | ID: mdl-39140804

ABSTRACT

OBJECTIVES: Tuberostemonine has several biological activity, the aim of study examined the impact of tuberostemonine on the proliferation of TGF-ß1 induced cell model, and its ability to alleviate pulmonary fibrosis stimulated by bleomycin in mice. METHODS: In vitro, we assessed the effect of tuberostemonine (350, 550 and 750 µM) on the proliferation of cells stimulated by TGF-ß1 (10 µg/L), as well as on parameters such as α-SMA vitality, human fibronectin, collagen, and hydroxyproline levels in cells. In vivo, we analyzed inflammation, hydroxyproline, collagen activity and metabolomics in the lungs of mice. Additionally, a comprehensive investigation into the TGF-ß/smad signaling pathway was undertaken, targeting lung tissue as well as HFL cells. RESULTS: Within the confines of an in vitro setup, the tuberostemonine manifested a discerned IC50 of 1.9 mM. Furthermore, a significant reduction of over fifty percent was ascertained in the secretion levels of hydroxyproline, fibronectin, collagen type I, collagen type III and α-SMA. In vivo, tuberostemonine obviously improved the respiratory function percentage over 50% of animal model and decreased the hydroxyproline, lung inflammation and collagen deposition. A prominent decline in TGF-ß/smad pathway functioning was identified within both the internal and external cellular contexts. CONCLUSIONS: Tuberostemonine is considered as a modulator to alleviate fibrosis and may become a new renovation for pulmonary fibrosis.


Subject(s)
Bleomycin , Cell Proliferation , Fibroblasts , Lung , Pulmonary Fibrosis , Signal Transduction , Transforming Growth Factor beta1 , Animals , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Lung/drug effects , Lung/pathology , Lung/metabolism , Humans , Mice , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Hydroxyproline/metabolism , Smad Proteins/metabolism , Mice, Inbred C57BL , Male , Cell Line , Collagen/metabolism , Disease Models, Animal , Fibronectins/metabolism , Actins/metabolism
13.
Elife ; 132024 Aug 15.
Article in English | MEDLINE | ID: mdl-39146380

ABSTRACT

AMPA-type receptors (AMPARs) are rapidly inserted into synapses undergoing plasticity to increase synaptic transmission, but it is not fully understood if and how AMPAR-containing vesicles are selectively trafficked to these synapses. Here, we developed a strategy to label AMPAR GluA1 subunits expressed from their endogenous loci in cultured rat hippocampal neurons and characterized the motion of GluA1-containing vesicles using single-particle tracking and mathematical modeling. We find that GluA1-containing vesicles are confined and concentrated near sites of stimulation-induced structural plasticity. We show that confinement is mediated by actin polymerization, which hinders the active transport of GluA1-containing vesicles along the length of the dendritic shaft by modulating the rheological properties of the cytoplasm. Actin polymerization also facilitates myosin-mediated transport of GluA1-containing vesicles to exocytic sites. We conclude that neurons utilize F-actin to increase vesicular GluA1 reservoirs and promote exocytosis proximal to the sites of synaptic activity.


Subject(s)
Actins , Dendrites , Hippocampus , Neuronal Plasticity , Polymerization , Receptors, AMPA , Animals , Receptors, AMPA/metabolism , Actins/metabolism , Rats , Neuronal Plasticity/physiology , Dendrites/metabolism , Hippocampus/metabolism , Hippocampus/cytology , Protein Transport , Neurons/metabolism , Cells, Cultured , Exocytosis
14.
Proc Natl Acad Sci U S A ; 121(33): e2401816121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39106306

ABSTRACT

Many cytoskeletal networks consist of individual filaments that are organized into elaborate higher-order structures. While it is appreciated that the size and architecture of these networks are critical for their biological functions, much of the work investigating control over their assembly has focused on mechanisms that regulate the turnover of individual filaments through size-dependent feedback. Here, we propose a very different, feedback-independent mechanism to explain how yeast cells control the length of their actin cables. Our findings, supported by quantitative cell imaging and mathematical modeling, indicate that actin cable length control is an emergent property that arises from the cross-linked and bundled organization of the filaments within the cable. Using this model, we further dissect the mechanisms that allow cables to grow longer in larger cells and propose that cell length-dependent tuning of formin activity allows cells to scale cable length with cell length. This mechanism is a significant departure from prior models of cytoskeletal filament length control and presents a different paradigm to consider how cells control the size, shape, and dynamics of higher-order cytoskeletal structures.


Subject(s)
Cytoskeleton , Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolism , Cytoskeleton/metabolism , Actins/metabolism , Actin Cytoskeleton/metabolism , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism
15.
Phys Rev E ; 110(1-1): 014410, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39160984

ABSTRACT

We study the dynamic structure of lipid domain inclusions embedded within a phase-separated reconstituted lipid bilayer in contact with a swarming flow of gliding filamentous actin. Passive circular domains transition into highly deformed morphologies that continuously elongate, rotate, and pinch off into smaller fragments, leading to a dynamic steady state with ≈23× speedup in the relaxation of the intermediate scattering function compared with passive membrane domains driven by purely thermal forces. To corroborate experimental results, we develop a phase-field model of the lipid domains with two-way coupling to the Toner-Tu equations. We report phase domains that become entrained in the chaotic eddy patterns, with oscillating waves of domains that correlate with the dominant wavelengths of the Toner-Tu flow fields.


Subject(s)
Lipid Bilayers , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Actins/metabolism , Models, Biological , Membrane Microdomains/metabolism
16.
Phys Rev E ; 110(1-1): 014406, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39160992

ABSTRACT

The interior of cellular nuclei, the nucleoplasm, is a crowded fluid that is pervaded by protein-decorated DNA polymers, the chromatin. Due to the complex architecture of chromatin and a multitude of associated nonequilibrium processes, e.g., DNA repair, the nucleoplasm can be expected to feature nontrivial material properties and hence anomalous transport phenomena. Here, we have used single-particle tracking on nuclear actin rods to probe such transport phenomena. Our analysis reveals that short actin rods in the nucleus show an intermittent, antipersistent subdiffusion with clear signatures of fractional Brownian motion. Moreover, the diffusive motion is heterogeneous with clear signatures of an intermittent switching of trajectories between at least two different mobilities, most likely due to transient associations with chromatin. In line with this interpretation, hyperosmotic stress is seen to stall the motion of nuclear actin rods, whereas hypo-osmotic conditions yield a reptationlike motion. Our data highlights the heterogeneity of transport in the nucleoplasm that needs to be taken into account for an understanding of nucleoplasmic organization and the mechanobiology of nuclei.


Subject(s)
Actins , Cell Nucleus , Chromatin , Diffusion , Actins/metabolism , Chromatin/metabolism , Cell Nucleus/metabolism , Animals , Models, Biological , Osmotic Pressure
17.
Clin Exp Pharmacol Physiol ; 51(10): e13916, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39155151

ABSTRACT

Bombesin receptor-activated protein (BRAP), encoded by the C6orf89 gene in humans, is expressed in various cells with undefined functions. BC004004, the mouse homologue of C6orf89, has been shown to play a role in bleomycin-induced pulmonary fibrosis through the use of a BC004004 gene knockout mouse (BC004004-/-). In this study, we investigated the potential involvement of BRAP in renal fibrosis using two mouse models: unilateral ureteral obstruction (UUO) and type 2 diabetes mellitus induced by combination of a high-fat diet (HFD) and streptozocin (STZ). BRAP or its homologue was expressed in tubular epithelial cells (TECs) in the kidneys of patients with chronic kidney disease (CKD) and in BC004004+/+ mice. Compared to control mice, BC004004-/- mice exhibited attenuated renal injury and renal fibrosis after UUO or after HFD/STZ treatment. Immunohistochemistry and immunoblot analyses of the kidneys of BC004004+/+ mice after UUO surgery showed a more significant decrease in E-cadherin expression and a more significant increase in both α smooth muscle actin (α-SMA) and vimentin expression compared to BC004004-/- mice. Additionally, stimulation with transforming growth factor-ß1 (TGF-ß1) led to a more significant decrease in E-cadherin expression and a more significant increase in α-SMA and vimentin expression in isolated TECs from BC004004+/+ than in those from BC004004-/- mice. These results suggest that an enhanced epithelial-mesenchymal transition (EMT) process occurred in TECs in BC004004+/+ mice during renal injury, which might contribute to renal fibrosis. The loss of the BRAP homologue in BC004004-/- mice suppressed EMT activation in kidneys and contributed to the suppression of fibrosis during renal injury.


Subject(s)
Fibrosis , Animals , Mice , Male , Humans , Epithelial-Mesenchymal Transition , Mice, Knockout , Ureteral Obstruction/pathology , Ureteral Obstruction/complications , Kidney/pathology , Kidney/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Mice, Inbred C57BL , Cadherins/metabolism , Cadherins/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/genetics
18.
FASEB J ; 38(16): e23889, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39157975

ABSTRACT

Cholestatic liver diseases, such as primary biliary cholangitis (PBC) and primary sclerosing cholangitis (PSC), lead to inflammation and severe hepatic damage with limited therapeutic options. This study assessed the efficacy of the inverse RORγt agonist, GSK805, both in vitro using the hepatic stellate cell-line LX-2 and in vivo using male bile duct-ligated BALB/c mice. In vitro, 0.3 µM GSK805 reduced alpha-smooth muscle actin expression in LX-2 cells. In vivo, GSK805 significantly decreased IL-23R, TNF-α, and IFN-γ expression in cholestatic liver. Despite high concentrations of GSK805 in the liver, no significant reduction in fibrosis was noticed. GSK805 significantly increased aspartate aminotransferase and alanine aminotransferase activity in the blood, while levels of glutamate dehydrogenase, alkaline phosphatase, and bilirubin were not substantially increased. Importantly, GSK805 did neither increase an animal distress score nor substantially reduce body weight, burrowing activity, or nesting behavior. These results suggest that a high liver concentration of GSK805 is achieved by daily oral administration and that this drug modulates inflammation in cholestatic mice without impairing animal well-being.


Subject(s)
Mice, Inbred BALB C , Animals , Mice , Male , Humans , Actins/metabolism , Liver/metabolism , Liver/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Cell Line , Inflammation/metabolism , Inflammation/drug therapy , Cholestasis/metabolism , Cholestasis/drug therapy
19.
Curr Biol ; 34(15): R728-R731, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39106829

ABSTRACT

Cell migration through complex 3D environments relies on the interplay between actin and microtubules. A new study shows that, when cells pass through narrow constrictions, CLASP-dependent microtubule stabilisation at the cell rear controls actomyosin contractility to enable nuclear translocation and preserve cell integrity.


Subject(s)
Cell Movement , Microtubules , Microtubules/metabolism , Microtubules/physiology , Cell Movement/physiology , Actomyosin/metabolism , Actins/metabolism , Animals
SELECTION OF CITATIONS
SEARCH DETAIL