Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.694
Filter
1.
J Cell Mol Med ; 28(19): e70143, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39383106

ABSTRACT

Sad and UNC84 domain 1 (SUN1) is a kind of nuclear envelope protein with established involvement in cellular processes, including nuclear motility and meiosis. SUN1 plays an intriguing role in human adipose-derived stem cells (hASCs) differentiation; however, this role remains largely undefined. This study was undertaken to investigate the role of SUN1 in hASCs differentiation, as well as its underlying mechanisms. Employing siRNAs, we selectively downregulated SUN1 and CD36 expression. Microtubules were depolymerized using nocodazole, and PPARγ was activated using rosiglitazone. Western blotting was performed to quantify SUN1, PPARγ, α-tubulin, CD36, OPN, and adiponectin protein expression levels. Alkaline phosphatase and Oil red O staining were used to assess osteogenesis and adipogenesis, respectively. Downregulated SUN1 expression increased osteogenesis and decreased adipogenesis in hASCs, concomitant with upregulated α-tubulin expression and downregulated CD36 expression, alongside reduced nuclear localization of PPARγ. Microtubule depolymerization increased CD36 expression. Rescue experiments indicated that microtubule depolymerization counteracted the downregulated SUN1-induced phenotypic changes. This study demonstrates that SUN1 influences the differentiation of hASCs towards osteogenic and adipogenic lineages, indicating its essential role in cell fate.


Subject(s)
Adipogenesis , Adipose Tissue , CD36 Antigens , Cell Differentiation , Osteogenesis , PPAR gamma , Stem Cells , Tubulin , Humans , Adipogenesis/genetics , CD36 Antigens/metabolism , CD36 Antigens/genetics , Osteogenesis/genetics , Tubulin/metabolism , Stem Cells/metabolism , Stem Cells/cytology , PPAR gamma/metabolism , PPAR gamma/genetics , Adipose Tissue/cytology , Adipose Tissue/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Gene Expression Regulation , Cells, Cultured , Nuclear Proteins
2.
Cell Mol Life Sci ; 81(1): 416, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39367928

ABSTRACT

Neurons are dependent on efficient quality control mechanisms to maintain cellular homeostasis and function due to their polarization and long-life span. Autophagy is a lysosomal degradative pathway that provides nutrients during starvation and recycles damaged and/or aged proteins and organelles. In neurons, autophagosomes constitutively form in distal axons and at synapses and are trafficked retrogradely to the cell soma to fuse with lysosomes for cargo degradation. How the neuronal autophagy pathway is organized and controlled remains poorly understood. Several presynaptic endocytic proteins have been shown to regulate both synaptic vesicle recycling and autophagy. Here, by combining electron, fluorescence, and live imaging microscopy with biochemical analysis, we show that the neuron-specific protein APache, a presynaptic AP-2 interactor, functions in neurons as an important player in the autophagy process, regulating the retrograde transport of autophagosomes. We found that APache colocalizes and co-traffics with autophagosomes in primary cortical neurons and that induction of autophagy by mTOR inhibition increases LC3 and APache protein levels at synaptic boutons. APache silencing causes a blockade of autophagic flux preventing the clearance of p62/SQSTM1, leading to a severe accumulation of autophagosomes and amphisomes at synaptic terminals and along neurites due to defective retrograde transport of TrkB-containing signaling amphisomes along the axons. Together, our data identify APache as a regulator of the autophagic cycle, potentially in cooperation with AP-2, and hypothesize that its dysfunctions contribute to the early synaptic impairments in neurodegenerative conditions associated with impaired autophagy.


Subject(s)
Autophagosomes , Autophagy , Axonal Transport , Neurons , Autophagosomes/metabolism , Autophagy/physiology , Animals , Neurons/metabolism , Axonal Transport/physiology , Mice , Cells, Cultured , TOR Serine-Threonine Kinases/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Sequestosome-1 Protein/metabolism , Receptor, trkB/metabolism , Signal Transduction , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Presynaptic Terminals/metabolism
3.
Planta ; 260(5): 110, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352582

ABSTRACT

MAIN CONCLUSION: The GhEB1C gene of the EB1 protein family functions as microtubule end-binding protein and may be involved in the regulation of microtubule-related pathways to enhance resistance to Verticillium wilt. The expression of GhEB1C is induced by SA, also contributing to Verticillium wilt resistance. Cotton, as a crucial cash and oil crop, faces a significant threat from Verticillium wilt, a soil-borne disease induced by Verticillium dahliae, severely impacting cotton growth and development. Investigating genes associated with resistance to Verticillium wilt is paramount. We identified and performed a phylogenetic analysis on members of the EB1 family associated with Verticillium wilt in this work. GhEB1C was discovered by transcriptome screening and was studied for its function in cotton defense against V. dahliae. The RT-qPCR analysis revealed significant expression of the GhEB1C gene in cotton leaves. Subsequent localization analysis using transient expression demonstrated cytoplasmic localization of GhEB1C. VIGS experiments indicated that silencing of the GhEB1C gene significantly increased susceptibility of cotton to V. dahliae. Comparative RNA-seq analysis showed that GhEB1C silenced plants exhibited altered microtubule-associated protein pathways and flavonogen-associated pathways, suggesting a role for GhEB1C in defense mechanisms. Overexpression of tobacco resulted in enhanced resistance to V. dahliae as compared to wild-type plants. Furthermore, our investigation into the relationship between the GhEB1C gene and plant disease resistance hormones salicylic axid (SA) and jasmonic acid (JA) revealed the involvement of GhEB1C in the regulation of the SA pathway. In conclusion, our findings demonstrate that GhEB1C plays a crucial role in conferring immunity to cotton against Verticillium wilt, providing valuable insights for further research on plant adaptability to pathogen invasion.


Subject(s)
Disease Resistance , Gossypium , Phylogeny , Plant Diseases , Plant Proteins , Gossypium/genetics , Gossypium/microbiology , Gossypium/immunology , Plant Diseases/microbiology , Plant Diseases/genetics , Plant Diseases/immunology , Disease Resistance/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Gene Expression Regulation, Plant , Ascomycota/physiology , Ascomycota/pathogenicity , Salicylic Acid/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Plant Leaves/microbiology , Plant Leaves/genetics , Plant Leaves/immunology , Oxylipins/metabolism , Verticillium/physiology , Cyclopentanes/metabolism
4.
Int J Mol Sci ; 25(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39273545

ABSTRACT

Cytopathology induced by methamphetamine (METH) is reminiscent of degenerative disorders such as Parkinson's disease, and it is characterized by membrane organelles arranged in tubulo-vesicular structures. These areas, appearing as clusters of vesicles, have never been defined concerning the presence of specific organelles. Therefore, the present study aimed to identify the relative and absolute area of specific membrane-bound organelles following a moderate dose (100 µM) of METH administered to catecholamine-containing PC12 cells. Organelles and antigens were detected by immunofluorescence, and they were further quantified by plain electron microscopy and in situ stoichiometry. This analysis indicated an increase in autophagosomes and damaged mitochondria along with a decrease in lysosomes and healthy mitochondria. Following METH, a severe dissipation of hallmark proteins from their own vesicles was measured. In fact, the amounts of LC3 and p62 were reduced within autophagy vacuoles compared with the whole cytosol. Similarly, LAMP1 and Cathepsin-D within lysosomes were reduced. These findings suggest a loss of compartmentalization and confirm a decrease in the competence of cell clearing organelles during catecholamine degeneration. Such cell entropy is consistent with a loss of energy stores, which routinely govern appropriate subcellular compartmentalization.


Subject(s)
Autophagosomes , Lysosomes , Methamphetamine , Methamphetamine/pharmacology , Animals , PC12 Cells , Rats , Lysosomes/metabolism , Lysosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/drug effects , Autophagy/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Cathepsin D/metabolism , Microtubule-Associated Proteins/metabolism
5.
Viruses ; 16(9)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39339932

ABSTRACT

Enteroviruses are a genus of small RNA viruses that are responsible for approximately one billion global infections annually. These infections range in severity from the common cold and flu-like symptoms to more severe diseases, such as viral myocarditis, pancreatitis, and neurological disorders, that continue to pose a global health challenge with limited therapeutic strategies currently available. In the current study, we sought to understand the interaction between coxsackievirus B3 (CVB3), which is a model enterovirus, and macrophage cells, as there is limited understanding of how this virus interacts with macrophage innate immune cells. Our study demonstrated that CVB3 can robustly activate macrophages without apparent viral replication in these cells. We also showed that myeloid cells lacked the viral entry receptor coxsackievirus and adenovirus receptor (CAR). However, the expression of exogenous CAR in RAW264.7 macrophages was unable to overcome the viral replication deficit. Interestingly, the CAR expression was associated with altered inflammatory responses during prolonged infection. Additionally, we identified the autophagy protein LC3 as a novel stimulus for macrophage activation. These findings provide new insights into the mechanisms of CVB3-induced macrophage activation and its implications for viral pathogenesis.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enterovirus B, Human , Macrophage Activation , Macrophages , Virus Internalization , Animals , Humans , Mice , Autophagy , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Coxsackievirus Infections/virology , Coxsackievirus Infections/immunology , Enterovirus B, Human/physiology , Macrophages/virology , Macrophages/immunology , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , RAW 264.7 Cells , Virus Replication
6.
Proc Natl Acad Sci U S A ; 121(37): e2402817121, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39236246

ABSTRACT

Autophagy of glycogen (glycophagy) is crucial for the maintenance of cellular glucose homeostasis and physiology in mammals. STBD1 can serve as an autophagy receptor to mediate glycophagy by specifically recognizing glycogen and relevant key autophagic factors, but with poorly understood mechanisms. Here, we systematically characterize the interactions of STBD1 with glycogen and related saccharides, and determine the crystal structure of the STBD1 CBM20 domain with maltotetraose, uncovering a unique binding mode involving two different oligosaccharide-binding sites adopted by STBD1 CBM20 for recognizing glycogen. In addition, we demonstrate that the LC3-interacting region (LIR) motif of STBD1 can selectively bind to six mammalian ATG8 family members. We elucidate the detailed molecular mechanism underlying the selective interactions of STBD1 with ATG8 family proteins by solving the STBD1 LIR/GABARAPL1 complex structure. Importantly, our cell-based assays reveal that both the STBD1 LIR/GABARAPL1 interaction and the intact two oligosaccharide binding sites of STBD1 CBM20 are essential for the effective association of STBD1, GABARAPL1, and glycogen in cells. Finally, through mass spectrometry, biochemical, and structural modeling analyses, we unveil that STBD1 can directly bind to the Claw domain of RB1CC1 through its LIR, thereby recruiting the key autophagy initiation factor RB1CC1. In all, our findings provide mechanistic insights into the recognitions of glycogen, ATG8 family proteins, and RB1CC1 by STBD1 and shed light on the potential working mechanism of STBD1-mediated glycophagy.


Subject(s)
Autophagy-Related Protein 8 Family , Autophagy , Glycogen , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Autophagy/physiology , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Protein 8 Family/genetics , Autophagy-Related Protein 8 Family/chemistry , Binding Sites , Crystallography, X-Ray , Glycogen/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Models, Molecular , Protein Binding
7.
Curr Biol ; 34(19): 4476-4494.e11, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-39276769

ABSTRACT

Kinesin and dynein-dynactin motors move endosomes and other vesicles bidirectionally along microtubules, a process mainly studied under in vitro conditions. Here, we provide a physiological bidirectional transport model following color-coded, endogenously tagged transport-related proteins as they move through a crowded cellular environment. Late endosomes (LEs) surf bidirectionally on Protrudin-enriched endoplasmic reticulum (ER) membrane contact sites, while hopping and gliding along microtubules and bypassing cellular obstacles, such as mitochondria. During bidirectional transport, late endosomes do not switch between opposing Rab7 GTPase effectors, RILP and FYCO1, or their associated dynein and KIF5B motor proteins, respectively. In the endogenous setting, far fewer motors associate with endosomal membranes relative to effectors, implying coordination of transport with other aspects of endosome physiology through GTPase-regulated mechanisms. We find that directionality of transport is provided in part by various microtubule-associated proteins (MAPs), including MID1, EB1, and CEP169, which recruit Lis1-activated dynein motors to microtubule plus ends for transport of early and late endosomal populations. At these microtubule plus ends, activated dynein motors encounter the dynactin subunit p150glued and become competent for endosomal capture and minus-end movement in collaboration with membrane-associated Rab7-RILP. We show that endosomes surf over the ER through the crowded cell and move bidirectionally under the control of MAPs for motor activation and through motor replacement and capture by endosomal anchors.


Subject(s)
Endosomes , Microtubules , Endosomes/metabolism , Humans , Microtubules/metabolism , Dyneins/metabolism , Biological Transport , Microtubule-Associated Proteins/metabolism , HeLa Cells , Endoplasmic Reticulum/metabolism , Kinesins/metabolism , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , Protein Transport
8.
Mol Biol Cell ; 35(10): br19, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39259768

ABSTRACT

Microtubules are dynamic cytoskeletal polymers essential for cell division, motility, and intracellular transport. Microtubule dynamics are characterized by dynamic instability-the ability of individual microtubules to switch between phases of growth and shrinkage. Dynamic instability can be explained by the GTP-cap model, suggesting that a "cap" of GTP-tubulin subunits at the growing microtubule end has a stabilizing effect, protecting against microtubule catastrophe-the switch from growth to shrinkage. Although the GTP-cap is thought to protect the growing microtubule end, whether the GTP-cap size affects microtubule stability in cells is not known. Notably, microtubule end-binding proteins, EBs, recognize the nucleotide state of tubulin and display comet-like localization at growing microtubule ends, which can be used as a proxy for the GTP-cap. Here, we employ high spatiotemporal resolution imaging to compare the relationship between EB comet size and microtubule dynamics in interphase LLC-PK1 cells to that measured in vitro. Our data reveal that the GTP-cap size in cells scales with the microtubule growth rate in the same way as in vitro. However, we find that microtubule ends in cells can withstand transition to catastrophe even after the EB comet is lost. Thus, our findings suggest that the presence of the GTP-cap is not the determinant of microtubule end stability in cells.


Subject(s)
Guanosine Triphosphate , Microtubule-Associated Proteins , Microtubules , Tubulin , Microtubules/metabolism , Guanosine Triphosphate/metabolism , Tubulin/metabolism , Animals , Microtubule-Associated Proteins/metabolism , Swine , LLC-PK1 Cells , Interphase/physiology
9.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39347717

ABSTRACT

Lysosomes, essential for intracellular degradation and recycling, employ damage-control strategies such as lysophagy and membrane repair mechanisms to maintain functionality and cellular homeostasis. Our study unveils migratory autolysosome disposal (MAD), a response to lysosomal damage where cells expel LAMP1-LC3 positive structures via autolysosome exocytosis, requiring autophagy machinery, SNARE proteins, and cell migration. This mechanism, crucial for mitigating lysosomal damage, underscores the role of cell migration in lysosome damage control and facilitates the release of small extracellular vesicles, highlighting the intricate relationship between cell migration, organelle quality control, and extracellular vesicle release.


Subject(s)
Autophagy , Cell Movement , Lysosomes , Lysosomes/metabolism , Humans , Exocytosis , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , SNARE Proteins/metabolism , SNARE Proteins/genetics , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Animals , Extracellular Vesicles/metabolism , HeLa Cells , Lysosomal-Associated Membrane Protein 1
10.
FASEB J ; 38(19): e70059, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39331575

ABSTRACT

White matter hyperintensity (WMH) is strongly correlated with age-related dementia and hypertension, but its pathogenesis remains obscure. Genome-wide association studies identified TRIM47 at the 17q25 locus as a top genetic risk factor for WMH formation. TRIM family is a class of E3 ubiquitin ligase with pivotal functions in autophagy, which is critical for brain endothelial cell (ECs) remodeling during hypertension. We hypothesize that TRIM47 regulates autophagy and its loss-of-function disturbs cerebrovasculature. Based on transcriptomics and immunohistochemistry, TRIM47 is found highly expressed by brain ECs in human and mouse, and its transcription is upregulated by artificially induced autophagy while downregulated in hypertension-like conditions. Using in silico simulation, immunocytochemistry and super-resolution microscopy, we predicted a highly conserved binding site between TRIM47 and the LIR (LC3-interacting region) motif of LC3B. Importantly, pharmacological autophagy induction increased Trim47 expression on mouse ECs (b.End3) culture, while silencing Trim47 significantly increased autophagy with ULK1 phosphorylation induction, transcription, and vacuole formation. Together, we demonstrate that TRIM47 is an endogenous inhibitor of autophagy in brain ECs, and such TRIM47-mediated regulation connects genetic and physiological risk factors for WMH formation but warrants further investigation.


Subject(s)
Autophagy , Brain , Endothelial Cells , Animals , Mice , Humans , Endothelial Cells/metabolism , Brain/metabolism , Brain/pathology , White Matter/metabolism , White Matter/pathology , Risk Factors , Male , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
J Cell Biol ; 223(12)2024 Dec 02.
Article in English | MEDLINE | ID: mdl-39316454

ABSTRACT

Subcortical heterotopia is a cortical malformation associated with epilepsy, intellectual disability, and an excessive number of cortical neurons in the white matter. Echinoderm microtubule-associated protein like 1 (EML1) mutations lead to subcortical heterotopia, associated with abnormal radial glia positioning in the cortical wall, prior to malformation onset. This perturbed distribution of proliferative cells is likely to be a critical event for heterotopia formation; however, the underlying mechanisms remain unexplained. This study aimed to decipher the early cellular alterations leading to abnormal radial glia. In a forebrain conditional Eml1 mutant model and human patient cells, primary cilia and centrosomes are altered. Microtubule dynamics and cell cycle kinetics are also abnormal in mouse mutant radial glia. By rescuing microtubule formation in Eml1 mutant embryonic brains, abnormal radial glia delamination and heterotopia volume were significantly reduced. Thus, our new model of subcortical heterotopia reveals the causal link between Eml1's function in microtubule regulation and cell position, both critical for correct cortical development.


Subject(s)
Centrosome , Microtubule-Associated Proteins , Microtubules , Prosencephalon , Animals , Centrosome/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Prosencephalon/metabolism , Prosencephalon/pathology , Prosencephalon/embryology , Microtubules/metabolism , Mice , Cilia/metabolism , Cilia/pathology , Mutation/genetics , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Cell Cycle/genetics
12.
J Cell Sci ; 137(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39257379

ABSTRACT

Association of tau (encoded by Mapt) with microtubules causes them to be labile, whereas association of MAP6 with microtubules causes them to be stable. As axons differentiate and grow, tau and MAP6 segregate from one another on individual microtubules, resulting in the formation of stable and labile domains. The functional significance of the yin-yang relationship between tau and MAP6 remains speculative, with one idea being that such a relationship assists in balancing morphological stability with plasticity. Here, using primary rodent neuronal cultures, we show that tau depletion has opposite effects compared to MAP6 depletion on the rate of neuronal development, the efficiency of growth cone turning, and the number of neuronal processes and axonal branches. Opposite effects to those seen with tau depletion were also observed on the rate of neuronal migration, in an in vivo assay, when MAP6 was depleted. When tau and MAP6 were depleted together from neuronal cultures, the morphological phenotypes negated one another. Although tau and MAP6 are multifunctional proteins, our results suggest that the observed effects on neuronal development are likely due to their opposite roles in regulating microtubule stability.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Neurons , tau Proteins , tau Proteins/metabolism , Animals , Neurons/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Neurogenesis , Rats , Cells, Cultured , Axons/metabolism , Mice , Cell Movement , Growth Cones/metabolism
13.
Zhen Ci Yan Jiu ; 49(8): 821-828, 2024 Aug 25.
Article in English, Chinese | MEDLINE | ID: mdl-39318311

ABSTRACT

OBJECTIVES: To observe the effects of electroacupuncture (EA) on the morphological changes of intervertebral disc tissues, apoptosis of nucleus pulposus cells, and the protein expression of Unc-51 like autophagy-activated kinase 1 (ULK1), homologous series of yeast Atg6 (Beclin1), and light chain protease complication 3 type (LC3) in nucleus pulposus tissue of cervical spondylosis rabbits, so as to explore the role of cellular autophagy in EA treatment of cervical spondylosis. METHODS: A total of 24 New Zealand white rabbits were randomly divided into blank, model and EA groups, with 8 rabbits in each group. In the EA group, both sides of the cervical (C)3-C6 "Jiaji" (EX-B2) were stimulated by EA (2 Hz/100 Hz, 1 mA) for 25 min, once daily for 5 days in a course, with a 2-day interval between courses, totaling 4 treatment courses. X-ray was used to assess cervical spine radiographic changes and evaluate radiographic scores;transmission electron microscopy was used to observe ultrastructural changes in nucleus pulposus cells;HE staining was used to observe morphological changes of intervertebral disc tissues and conduct pathological scoring;TUNEL staining was used to observe apoptosis rate of nucleus pulposus cells;Western blot was performed to detect protein expression levels of ULK1, Beclin1, and LC3 in nucleus pulposus tissue. RESULTS: Compared with the blank group, rabbits in the model group showed significantly higher cervical spine radiographic scores (P<0.01), higher pathological scores of intervertebral disc tissues (P<0.05), increased apoptosis rate of nucleus pulposus cells (P<0.01), and decreased expression levels of ULK1, Beclin1, and LC3Ⅱ proteins in nucleus pulposus tissue (P<0.05). Compared with the model group, the EA group showed significantly lower pathological scores of intervertebral discs (P<0.05), lower apoptosis rate of nucleus pulposus cells (P<0.01), and higher protein expression levels of ULK1, Beclin1, and LC3Ⅱ in nucleus pulposus tissue (P<0.01). Rabbits in the blank control group exhibited generally normal organelle structures in nucleus pulposus tissues with few autophagic vacuoles, indicative of early stages of autophagy;while those in the model group showed disrupted organelle structures with cytoplasmic condensation and those in the EA group exhibited autophagosomes with double-membrane structures in nucleus pulposus tissues. CONCLUSIONS: EA promotes the expression of ULK1, Beclin1, and LC3Ⅱ proteins in nucleus pulposus tissues, reduces apoptosis of nucleus pulposus cells, and improves intervertebral disc degeneration.


Subject(s)
Acupuncture Points , Autophagy , Electroacupuncture , Nucleus Pulposus , Spondylosis , Animals , Rabbits , Nucleus Pulposus/metabolism , Spondylosis/therapy , Spondylosis/metabolism , Spondylosis/genetics , Humans , Male , Apoptosis , Beclin-1/metabolism , Beclin-1/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Autophagy-Related Protein-1 Homolog/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Female , Cervical Vertebrae/metabolism , Intervertebral Disc/metabolism
14.
Phytomedicine ; 134: 155946, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39276538

ABSTRACT

BACKGROUND: Bronchial asthma is a chronic condition characterized by airway inflammation and remodeling, which pose complex pathophysiological challenges. Autophagy has been identified as a practical strategy to regulate inflammation and remodeling processes in chronic inflammatory diseases with pathological characteristics, such as asthma. PF (Paeoniflorin) is a potential new autophagy regulatory compound. Previous studies have reported that PF can inhibit airway inflammation to alleviate allergic asthma, but whether this is mediated through the regulation of autophagy and the molecular mechanism of action remains unclear. PURPOSE: The aim of this study was to evaluate the inhibitory effect of natural small molecule PF on asthma by regulating epithelial autophagy. METHODS: The rat asthma model was established through intraperitoneal injection of OVA and aluminum hydroxide suspension, followed by atomized inhalation of OVA for a period of two weeks. Following treatment with PF, histopathology was observed using Masson and H&E staining, while airway Max Rrs was evaluated using a pulmonary function apparatus. Levels of inflammatory cells in BALF were detected using a blood cell analyzer, and levels of inflammatory factors in BALF were detected through Elisa. Expressions of p-PRAS40 and p-Raptor were observed through immunohistochemistry, and levels of Beclin1 and LC3B were observed through immunofluorescence. The structure and quantity of autophagosomes and autophagolysosomal were observed through TEM. An autophagy model of 16HBE cells was established after treatment with 10ng/mL IL13 for 30 minutes. PRAS40 (AKT1S1) overexpression and mutation of PF and Raptor binding site (K207M& L302I& Q417H) were introduced in 16HBE cells. Autophagy in cells was measured by mFRP-GFP-LC3 ADV fluorescent tracer. The binding sites of PF and Raptor were analyzed using the Autodock Tool. The p-mTOR, p-Raptor, p-PRAS40, LC3II/LC3I were detected through Western blot, and interaction between PRAS40-Raptor and Raptor-mTOR was detected through Co-IP. RESULTS: The results showed that PF effectively reduced airway inflammation, improved airway pathological changes and remodeling, and maintained lung function. Additionally, PF was found to reverse excessive autophagy in airway epithelial cells. Interestingly, PF activated the mTORC1 subunit PRAS40 and Raptor in airway epithelial cells by regulating their phosphorylation. PRAS40 is an endogenous mTOR inhibitor that promotes autophagy. PF competitively binds Raptor to PRAS40, promoting Raptor-mTOR interactions to activate mTORC1, an outcome that can be reversed by PRAS40 overexpression and site-specific amino acid codon mutations in Raptor. CONCLUSION: These findings suggest that PF intervention and inhibition of PRAS40-Raptor interaction are effective treatments for bronchial asthma. By activating mTORC1, PF effectively reverses excessive autophagy in airway epithelial cells, leading to improved airway function and reduced inflammation.


Subject(s)
Asthma , Autophagy , Epithelial Cells , Glucosides , Mechanistic Target of Rapamycin Complex 1 , Monoterpenes , Animals , Humans , Male , Rats , Asthma/drug therapy , Autophagy/drug effects , Beclin-1/metabolism , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Epithelial Cells/drug effects , Glucosides/pharmacology , Mechanistic Target of Rapamycin Complex 1/metabolism , Microtubule-Associated Proteins/metabolism , Monoterpenes/pharmacology , Ovalbumin , Rats, Sprague-Dawley , Regulatory-Associated Protein of mTOR/metabolism
15.
PeerJ ; 12: e18118, 2024.
Article in English | MEDLINE | ID: mdl-39346063

ABSTRACT

Microtubule (MT) consists of α-tubulin and ß-tubulin. The dynamic instability regulated by various microtubule associated proteins (MAPs) is essential for MT functions. To analyze the interaction between tubulin/MT and MAP in vivo, we usually need tubulin and MAP co-expressed. Here, we constructed a dual-transgene vector expressing rice (Oryza sativa) α-tubulin and MAP simultaneously. To construct this vector, plant expression vector pCambia1301 was used as the plasmid backbone and Gibson assembly cloning technology was used. We first fused and cloned the GFP fragment, α-tubulin open reading frame (ORF), and NOS terminator into the vector pCambia1301 to construct the p35S::GFP-α-tubulin vector that expressed GFP-α-tubulin fusion protein. Subsequently, we fused and cloned the CaMV 35S promoter, mCherry fragment, and NOS terminator into the p35S::GFP-α-tubulin vector to generate the universal dual-transgene expression vector (p35S::GFP-α-tubulin-p35S::mCherry vector). With the p35S::GFP-α-tubulin-p35S::mCherry vector, MAP ORF can be cloned into the site of 5' or 3' terminus of mCherry to co-express GFP-α-tubulin and MAP-mCherry/mCherry-MAP. To validate the availability and universality of the dual-transgene expression vector, a series of putative rice MAP genes including GL7, OsKCBP, OsCLASP, and OsMOR1 were cloned into the vector respectively, transformed into Agrobacterium tumefaciens strain, and expressed in Nicotiana benthamiana leaves. The results indicated that all of the MAPs were co-expressed with α-tubulin and localized to MTs, validating the availability and universality of the vector and that GL7, OsKCBP, OsCLASP, and OsMOR1 might be MAPs. The application of the co-expression vector constructed by us would facilitate studies on the interaction between tubulin/MT and MAP in tobacco transient expression systems or transgenic rice.


Subject(s)
Genetic Vectors , Microtubule-Associated Proteins , Oryza , Tubulin , Oryza/genetics , Oryza/metabolism , Tubulin/genetics , Tubulin/metabolism , Genetic Vectors/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Plants, Genetically Modified/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Microtubules/metabolism , Microtubules/genetics
16.
Acta Neuropathol ; 148(1): 45, 2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39305312

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease with a mean survival time of three years. The 97% of the cases have TDP-43 nuclear depletion and cytoplasmic aggregation in motor neurons. TDP-43 prevents non-conserved cryptic exon splicing in certain genes, maintaining transcript stability, including ATG4B, which is crucial for autophagosome maturation and Microtubule-associated proteins 1A/1B light chain 3B (LC3B) homeostasis. In ALS mice (G93A), Atg4b depletion worsens survival rates and autophagy function. For the first time, we observed an elevation of LC3ylation in the CNS of both ALS patients and atg4b-/- mouse spinal cords. Furthermore, LC3ylation modulates the distribution of ATG3 across membrane compartments. Antisense oligonucleotides (ASOs) targeting cryptic exon restore ATG4B mRNA in TARDBP knockdown cells. We further developed multi-target ASOs targeting TDP-43 binding sequences for a broader effect. Importantly, our ASO based in peptide-PMO conjugates show brain distribution post-IV administration, offering a non-invasive ASO-based treatment avenue for neurodegenerative diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Autophagy-Related Proteins , Cysteine Endopeptidases , DNA-Binding Proteins , Microtubule-Associated Proteins , Animals , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Humans , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Mice , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Male , Spinal Cord/metabolism , Spinal Cord/pathology , Autophagy/physiology , Mice, Knockout , RNA Splicing/genetics , Female , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Oligonucleotides, Antisense/pharmacology
17.
Int Immunopharmacol ; 142(Pt B): 112862, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39306889

ABSTRACT

Osteosarcoma, a highly aggressive bone cancer, poses significant treatment challenges. This study investigates a novel approach utilizing induced pluripotent stem cells (iPSCs) engineered with the FGFR3-TACC3 fusion gene to generate cytotoxic T lymphocytes (CTLs) targeting osteosarcoma. The aim was to assess the efficacy of iPSC-derived CTLs in combating osteosarcoma progression. Abnormal expression of the FGFR3-TACC3 fusion gene was confirmed in osteosarcoma samples. iPSCs were successfully modified to express the fusion gene and were then differentiated into CTLs. In vitro experiments demonstrated that these modified CTLs effectively killed osteosarcoma cells, induced apoptosis, and inhibited migration and invasion. Findings were validated in in vivo experiments. This study suggests that iPSC-derived CTLs targeting FGFR3-TACC3 hold promise for personalized immunotherapy against osteosarcoma.


Subject(s)
Bone Neoplasms , Induced Pluripotent Stem Cells , Microtubule-Associated Proteins , Osteosarcoma , Receptor, Fibroblast Growth Factor, Type 3 , T-Lymphocytes, Cytotoxic , Osteosarcoma/therapy , Osteosarcoma/genetics , Osteosarcoma/immunology , Humans , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 3/metabolism , T-Lymphocytes, Cytotoxic/immunology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/therapy , Bone Neoplasms/immunology , Cell Line, Tumor , Induced Pluripotent Stem Cells/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Cell Differentiation , Mice , Oncogene Proteins, Fusion/genetics , Apoptosis/genetics , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Xenograft Model Antitumor Assays , Cell Movement/genetics
18.
Science ; 385(6715): 1366-1375, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39298589

ABSTRACT

Faithful chromosome segregation requires biorientation, where the pair of kinetochores on the chromosome establish bipolar microtubule attachment. The integrity of the kinetochore, a macromolecular complex built on centromeric DNA, is required for biorientation, but components sufficient for biorientation remain unknown. Here, we show that tethering the outer kinetochore heterodimer NDC80-NUF2 to the surface of apolar microbeads establishes their biorientation-like state in mouse cells. NDC80-NUF2 microbeads align at the spindle equator and self-correct alignment errors. The alignment is associated with stable bipolar microtubule attachment and is independent of the outer kinetochore proteins SPC24-SPC25, KNL1, the Mis12 complex, inner kinetochore proteins, and Aurora. Larger microbeads align more rapidly, suggesting a size-dependent biorientation mechanism. This study demonstrates a biohybrid kinetochore design for synthetic biorientation of microscale particles in cells.


Subject(s)
Cell Cycle Proteins , Chromosome Segregation , Kinetochores , Microspheres , Microtubule-Associated Proteins , Microtubules , Spindle Apparatus , Animals , Mice , Cell Cycle Proteins/metabolism , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Kinetochores/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubules/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Spindle Apparatus/metabolism
19.
Nat Commun ; 15(1): 7967, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261498

ABSTRACT

Microtubule-based vesicle trafficking usually relies upon kinesin and dynein motors and few reports describe microtubule polymerisation driving directional vesicle trafficking. Here we show that Arabidopsis END BINDING1b (EB1b), a microtubule plus-end binding protein, directly interacts with SYP121, a SNARE protein that mediates the trafficking of the K+ channel KAT1 and its distribution to the plasma membrane (PM) in Arabidopsis guard cells. Knockout of AtEB1b and its homologous proteins results in a modest but significant change in the distribution of KAT1 and SYP121 in guard cells and consequently delays light-induced stomatal opening. Live-cell imaging reveals that a portion of SYP121-associated endomembrane compartments co-localise with AtEB1b at the growing ends of microtubules, trafficking along with the growth of microtubules for targeting to the PM. Our study reveals a mechanism of vesicle trafficking driven by microtubule growth, which is involved in the redistribution of PM proteins to modulate guard cell movement.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Cell Membrane , Microtubule-Associated Proteins , Microtubules , Plant Stomata , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis/physiology , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Microtubules/metabolism , Plant Stomata/metabolism , Plant Stomata/physiology , Cell Membrane/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Protein Transport , Katanin/metabolism , Katanin/genetics , Cell Movement , Cell Cycle Proteins
20.
PLoS Genet ; 20(9): e1011373, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226307

ABSTRACT

Although centrosomes help organize spindles in most cell types, oocytes of most species lack these structures. During acentrosomal spindle assembly in C. elegans oocytes, microtubule minus ends are sorted outwards away from the chromosomes where they form poles, but then these outward forces must be balanced to form a stable bipolar structure. Simultaneously, microtubule dynamics must be precisely controlled to maintain spindle length and organization. How forces and dynamics are tuned to create a stable bipolar structure is poorly understood. Here, we have gained insight into this question through studies of ZYG-8, a conserved doublecortin-family kinase; the mammalian homolog of this microtubule-associated protein is upregulated in many cancers and has been implicated in cell division, but the mechanisms by which it functions are poorly understood. We found that ZYG-8 depletion from oocytes resulted in overelongated spindles with pole and midspindle defects. Importantly, experiments with monopolar spindles revealed that ZYG-8 depletion led to excess outward forces within the spindle and suggested a potential role for this protein in regulating the force-generating motor BMK-1/kinesin-5. Further, we found that ZYG-8 is also required for proper microtubule dynamics within the oocyte spindle and that kinase activity is required for its function during both meiosis and mitosis. Altogether, our findings reveal new roles for ZYG-8 in oocytes and provide insights into how acentrosomal spindles are stabilized to promote faithful meiosis.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Microtubules , Oocytes , Spindle Apparatus , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Microtubules/metabolism , Microtubules/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Oocytes/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Centrosome/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL