Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.379
Filtrer
1.
Cell Mol Biol Lett ; 29(1): 124, 2024 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-39333841

RÉSUMÉ

BACKGROUND: The involvement of tetraspanins in cancer development has been widely implicated. In this study, the function and molecular mechanisms of tetraspanin 3 (TSPAN3) in non-small cell lung cancer (NSCLC) cells were explored. METHODS: Tissue samples from patients diagnosed with NSCLC were analyzed by immunohistochemistry, western blotting, and real-time polymerase chain reaction (PCR) to indicate the involvement of TSPAN3 in cancer progression. In the meantime, we also performed exhaustive mechanistic studies using A549 and H460 cells in vitro through a variety of methods including western blotting, real-time PCR, immunofluorescent staining, coimmunoprecipitation, cell proliferation assay, and nocodazole (NZ) washout assay. Proper statistical analysis was implemented wherever necessary in this study. RESULTS: TSPAN3 was found to be highly expressed in lung cancer cells and tissues. Moreover, high levels of TSPAN3 positively correlated with poor differentiation, lymph node involvement, advanced pathological tumor-node-metastasis stage, and poor prognosis in patients with NSCLC. TSPAN3 showed potential to promote the proliferation of NSCLC cells in vitro and in vivo. Specifically, TSPAN3 was found to interact with ß1 integrin via the LEL domain, thereby facilitating the sorting of ß1 integrin into Rab11a endosomes and promoting ß1 integrin recycling and upregulation. CONCLUSIONS: Our findings reveal TSPAN3 may represent a potentially valuable therapeutic target for NSCLC.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Antigènes CD29 , Tumeurs du poumon , Tétraspanines , Humains , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/génétique , Prolifération cellulaire/génétique , Tétraspanines/métabolisme , Tétraspanines/génétique , Antigènes CD29/métabolisme , Antigènes CD29/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Femelle , Mâle , Lignée cellulaire tumorale , Adulte d'âge moyen , Animaux , Cellules A549 , Souris nude , Endosomes/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines G rab/métabolisme , Protéines G rab/génétique , Souris , Souris de lignée BALB C
2.
Gut Microbes ; 16(1): 2400575, 2024.
Article de Anglais | MEDLINE | ID: mdl-39312647

RÉSUMÉ

Enteropathogenic E. coli (EPEC) is a Gram-negative bacterial pathogen that causes persistent diarrhea. Upon attachment to the apical plasma membrane of the intestinal epithelium, the pathogen translocates virulence proteins called effectors into the infected cells. These effectors hijack numerous host processes for the pathogen's benefit. Therefore, studying the mechanisms underlying their action is crucial for a better understanding of the disease. We show that translocated EspH interacts with multiple host Rab GTPases. AlphaFold predictions and site-directed mutagenesis identified glutamic acid and lysine at positions 37 and 41 as Rab interacting residues in EspH. Mutating these sites abolished the ability of EspH to inhibit Akt and mTORC1 signaling, lysosomal exocytosis, and bacterial invasion. Knocking out the endogenous Rab8a gene expression highlighted the involvement of Rab8a in Akt/mTORC1 signaling and lysosomal exocytosis. A phosphoinositide binding domain with a critical tyrosine was identified in EspH. Mutating the tyrosine abolished the localization of EspH at infection sites and its capacity to interact with the Rabs. Our data suggest novel EspH-dependent mechanisms that elicit immune signaling and membrane trafficking during EPEC infection.


Sujet(s)
Membrane cellulaire , Escherichia coli entéropathogène , Protéines G rab , Humains , Protéines G rab/métabolisme , Protéines G rab/génétique , Membrane cellulaire/métabolisme , Escherichia coli entéropathogène/métabolisme , Escherichia coli entéropathogène/génétique , Phosphatidyl inositols/métabolisme , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Infections à Escherichia coli/microbiologie , Infections à Escherichia coli/métabolisme , Liaison aux protéines , Lysosomes/métabolisme , Transduction du signal , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Exocytose , Interactions hôte-pathogène , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique
3.
J Cell Biol ; 223(11)2024 Nov 04.
Article de Anglais | MEDLINE | ID: mdl-39325073

RÉSUMÉ

Deleterious mutations in the lipopolysaccharide responsive beige-like anchor protein (LRBA) gene cause severe childhood immune dysregulation. The complexity of the symptoms involving multiple organs and the broad range of unpredictable clinical manifestations of LRBA deficiency complicate the choice of therapeutic interventions. Although LRBA has been linked to Rab11-dependent trafficking of the immune checkpoint protein CTLA-4, its precise cellular role remains elusive. We show that LRBA, however, only slightly colocalizes with Rab11. Instead, LRBA is recruited by members of the small GTPase Arf protein family to the TGN and to Rab4+ endosomes, where it controls intracellular traffic. In patient-derived fibroblasts, loss of LRBA led to defects in the endosomal pathway promoting the accumulation of enlarged endolysosomes and lysosome secretion. Thus, LRBA appears to regulate flow through the endosomal system on Rab4+ endosomes. Our data strongly suggest functions of LRBA beyond CTLA-4 trafficking and provide a conceptual framework to develop new therapies for LRBA deficiency.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Endosomes , Homéostasie , Lysosomes , Protéines G rab , Protéines G rab4 , Humains , Endosomes/métabolisme , Lysosomes/métabolisme , Protéines G rab4/métabolisme , Protéines G rab4/génétique , Protéines G rab/métabolisme , Protéines G rab/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Transport des protéines , Fibroblastes/métabolisme , Facteurs d'ADP-ribosylation/métabolisme , Facteurs d'ADP-ribosylation/génétique , Réseau trans-golgien/métabolisme , Cellules HeLa , Cellules HEK293 , Facteur-1 d'ADP-ribosylation
4.
Traffic ; 25(9): e12956, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39313937

RÉSUMÉ

Limited nutrient availability in the tumor microenvironment can cause the rewiring of signaling and metabolic networks to confer cancer cells with survival advantages. We show here that the limitation of glucose, glutamine and serum from the culture medium resulted in the survival of a population of cancer cells with high viability and capacity to form tumors in vivo. These cells also displayed a remarkable increase in the abundance and size of lysosomes. Moreover, lysosomes were located mainly in the perinuclear region in nutrient-limited cells; this translocation was mediated by a rapid post-transcriptional increase in the key endolysosomal trafficking protein Rab7a. The acidic lysosomes in nutrient-limited cells could trap weakly basic drugs such as doxorubicin, mediating resistance of the cells to the drug, which could be partially reversed with the lysosomal inhibitor bafilomycin A1. An in vivo chorioallantoic membrane (CAM) assay indicated a remarkable decrease in microtumor volume when nutrient-limited cells were treated with 5-Fluorouracil (5-FU) and bafilomycin A1 compared to cells treated with either agent alone. Overall, our data indicate the activation of complementary pathways with nutrient limitation that can enable cancer cells to survive, proliferate and acquire drug resistance.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Lysosomes , Macrolides , Protéines G rab , Protéines Rab7 liant le GTP , Lysosomes/métabolisme , Humains , Protéines Rab7 liant le GTP/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Protéines G rab/métabolisme , Protéines G rab/génétique , Macrolides/pharmacologie , Nutriments/métabolisme , Lignée cellulaire tumorale , Fluorouracil/pharmacologie , Animaux , Doxorubicine/pharmacologie , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/traitement médicamenteux
5.
Cell Death Dis ; 15(9): 655, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39242574

RÉSUMÉ

Studies have indicated that RAB17 expression levels are associated with tumor malignancy, and RAB17 is more highly expressed in endometrial cancer (EC) tissues than in peritumoral tissues. However, the roles and potential mechanisms of RAB17 in EC remain undefined. The present study confirmed that the expression of RAB17 facilitates EC progression by suppressing cellular ferroptosis-like alterations. Mechanistically, RAB17 attenuated ferroptosis in EC cells by inhibiting transferrin receptor (TFRC) protein expression in a ubiquitin proteasome-dependent manner. Because EC is a blood-deprived tumor with a poor energy supply, the relationship between RAB17 and hypoglycemia was investigated. RAB17 expression was increased in EC cells incubated in low-glucose medium. Moreover, low-glucose medium limited EC cell ferroptosis and promoted EC progression through the RAB17-TFRC axis. The in vitro results were corroborated by in vivo studies and clinical data. Overall, the present study revealed that increased RAB17 promotes the survival of EC cells during glucose deprivation by inhibiting the onset of TFRC-dependent ferroptosis.


Sujet(s)
Évolution de la maladie , Tumeurs de l'endomètre , Ferroptose , Récepteurs à la transferrine , Protéines G rab , Animaux , Femelle , Humains , Souris , Antigènes CD , Lignée cellulaire tumorale , Tumeurs de l'endomètre/anatomopathologie , Tumeurs de l'endomètre/métabolisme , Tumeurs de l'endomètre/génétique , Ferroptose/génétique , Glucose/métabolisme , Souris nude , Protéines G rab/métabolisme , Protéines G rab/génétique , Récepteurs à la transferrine/métabolisme , Récepteurs à la transferrine/génétique
6.
Nat Commun ; 15(1): 8434, 2024 Sep 29.
Article de Anglais | MEDLINE | ID: mdl-39343966

RÉSUMÉ

The leucine-rich repeat kinase 2 (LRRK2) phosphorylates a subset of RAB GTPases, and their phosphorylation levels are elevated by Parkinson's disease (PD)-linked mutations of LRRK2. However, the precise function of the LRRK2-regulated RAB GTPase in the brain remains to be elucidated. Here, we identify RAB12 as a robust LRRK2 substrate in the mouse brain through phosphoproteomics profiling and solve the structure of RAB12-LRRK2 protein complex through Cryo-EM analysis. Mechanistically, RAB12 cooperates with LRRK2 to inhibit primary ciliogenesis and regulate centrosome homeostasis in astrocytes through enhancing the phosphorylation of RAB10 and recruiting RILPL1, while the functions of RAB12 require a direct interaction with LRRK2 and LRRK2 activity. Furthermore, the ciliary and centrosome defects caused by the PD-linked LRRK2-G2019S mutation are prevented by Rab12 deletion in astrocytes. Thus, our study reveals a physiological function of the RAB12-LRRK2 complex in regulating ciliogenesis and centrosome homeostasis. The RAB12-LRRK2 structure offers a guidance in the therapeutic development of PD by targeting the RAB12-LRRK2 interaction.


Sujet(s)
Astrocytes , Centrosome , Cils vibratiles , Homéostasie , Leucine-rich repeat serine-threonine protein kinase-2 , Protéines G rab , Animaux , Astrocytes/métabolisme , Leucine-rich repeat serine-threonine protein kinase-2/métabolisme , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Protéines G rab/métabolisme , Protéines G rab/génétique , Cils vibratiles/métabolisme , Souris , Centrosome/métabolisme , Humains , Phosphorylation , Maladie de Parkinson/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/anatomopathologie , Souris knockout , Mutation , Encéphale/métabolisme , Souris de lignée C57BL , Cellules HEK293
7.
J Cell Biol ; 223(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-39259305

RÉSUMÉ

How are Rab GTPases regulated during lysosome-related organelle (LRO) biogenesis? Li et al. (https://doi.org/10.1083/jcb.202402016) identify LYSMD proteins as crucial activators of Rab32-family GTPases in LRO development, shedding light on the previously ambiguous mechanisms governing Rab functionality in this process.


Sujet(s)
Lysosomes , Biogenèse des organelles , Protéines G rab , Lysosomes/métabolisme , Protéines G rab/métabolisme , Protéines G rab/génétique , Humains , Animaux , Organites/métabolisme
8.
Sci Rep ; 14(1): 20400, 2024 09 02.
Article de Anglais | MEDLINE | ID: mdl-39223234

RÉSUMÉ

To select the core target (RAB13) in sepsis patients' peripheral blood and investigate its molecular functions and possible mechanisms. The peripheral blood of sepsis patients (n = 21) and healthy individuals (n = 9) within 24 h after admission were collected for RNA-seq, and differential gene screening was performed by iDEP online analysis software (P < 0.01; log2FC ≥ 2) and enrichment analysis, the potential core target RAB13 was screened out. The association between RAB13 expression and sepsis severity was explored using multiple datasets in the GEO database, and survival analysis was conducted. Subsequently, peripheral blood mononuclear cells (PBMCs) from sepsis and control groups were isolated, and 10 × single-cell sequencing was used to identify the main RAB13-expressing cell types. Finally, LPS was used to stimulate THP1 cells to construct a sepsis model to explore the function and possible mechanism of RAB13. We found that RAB13 was a potential core target, and RAB13 expression level was positively associated with sepsis severity and negatively correlated with survival based on multiple public datasets. A single-cell sequencing indicated that RAB13 is predominantly localized in monocytes. Cell experiments validated that RAB13 is highly expressed in sepsis, and the knockdown of RAB13 promotes the polarization of macrophages towards the M2 phenotype. This mechanism may be associated with the ECM-receptor interaction signaling pathway. The upregulation of RAB13 in sepsis patients promotes the polarization of M2-like macrophages and correlates positively with the severity of sepsis.


Sujet(s)
Macrophages , Sepsie , Protéines G rab , Humains , Sepsie/métabolisme , Sepsie/génétique , Sepsie/anatomopathologie , Protéines G rab/métabolisme , Protéines G rab/génétique , Macrophages/métabolisme , Mâle , Femelle , Adulte d'âge moyen , Cellules THP-1 , Sujet âgé , Études cas-témoins , Agranulocytes/métabolisme , Lipopolysaccharides/pharmacologie
9.
EMBO J ; 43(18): 3948-3967, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39103493

RÉSUMÉ

Lysosomes play a pivotal role in coordinating macromolecule degradation and regulating cell growth and metabolism. Despite substantial progress in identifying lysosomal signaling proteins, understanding the pathways that synchronize lysosome functions with changing cellular demands remains incomplete. This study uncovers a role for TANK-binding kinase 1 (TBK1), well known for its role in innate immunity and organelle quality control, in modulating lysosomal responsiveness to nutrients. Specifically, we identify a pool of TBK1 that is recruited to lysosomes in response to elevated amino acid levels. This lysosomal TBK1 phosphorylates Rab7 on serine 72. This is critical for alleviating Rab7-mediated inhibition of amino acid-dependent mTORC1 activation. Furthermore, a TBK1 mutant (E696K) associated with amyotrophic lateral sclerosis and frontotemporal dementia constitutively accumulates at lysosomes, resulting in elevated Rab7 phosphorylation and increased mTORC1 activation. This data establishes the lysosome as a site of amino acid regulated TBK1 signaling that is crucial for efficient mTORC1 activation. This lysosomal pool of TBK1 has broader implications for lysosome homeostasis, and its dysregulation could contribute to the pathogenesis of ALS-FTD.


Sujet(s)
Acides aminés , Lysosomes , Complexe-1 cible mécanistique de la rapamycine , Protein-Serine-Threonine Kinases , Protéines Rab7 liant le GTP , Humains , Acides aminés/métabolisme , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Démence frontotemporale/métabolisme , Démence frontotemporale/génétique , Démence frontotemporale/anatomopathologie , Cellules HEK293 , Lysosomes/métabolisme , Complexe-1 cible mécanistique de la rapamycine/métabolisme , Complexe-1 cible mécanistique de la rapamycine/génétique , Phosphorylation , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protéines G rab/métabolisme , Protéines G rab/génétique , Transduction du signal
10.
J Virol ; 98(9): e0059924, 2024 Sep 17.
Article de Anglais | MEDLINE | ID: mdl-39136459

RÉSUMÉ

Herpes simplex virus 1 (HSV-1) is an alpha herpesvirus that infects a majority of the world population. The mechanisms and cellular host factors involved in the intracellular transport and exocytosis of HSV-1 particles are not fully understood. To elucidate these late steps in the replication cycle, we developed a live-cell fluorescence microscopy assay of HSV-1 virion intracellular trafficking and exocytosis. This method allows us to track individual virus particles and identify the precise moment and location of particle exocytosis using a pH-sensitive reporter. We show that HSV-1 uses the host cell's post-Golgi secretory pathway during egress. The small GTPase, Rab6, binds to nascent secretory vesicles at the trans-Golgi network and plays important, but non-essential, roles in vesicle traffic and exocytosis at the plasma membrane, therefore making it a useful marker of the Golgi and post-Golgi secretory pathway. We show that HSV-1 particles colocalize with Rab6a in the region of the Golgi, cotraffic with Rab6a to the cell periphery, and undergo exocytosis from Rab6a vesicles. Consistent with previous reports, we find that HSV-1 particles accumulate at preferential egress sites in infected cells. The secretory pathway mediates this preferential/polarized egress, since Rab6a vesicles accumulate near the plasma membrane similarly in uninfected cells. These data suggest that, following particle envelopment, HSV-1 egress follows a pre-existing cellular secretory pathway to exit infected cells rather than novel, virus-induced mechanisms. IMPORTANCE: Herpes simplex virus 1 (HSV-1) infects a majority of people. It establishes a life-long latent infection and occasionally reactivates, typically causing characteristic oral or genital lesions. Rarely in healthy natural hosts, but more commonly in zoonotic infections and in elderly, newborn, or immunocompromised patients, HSV-1 can cause severe herpes encephalitis. The precise cellular mechanisms used by HSV-1 remain an important area of research. In particular, the egress pathways that newly assembled virus particles use to exit from infected cells are unclear. In this study, we used fluorescence microscopy to visualize individual virus particles exiting from cells and found that HSV-1 particles use the pre-existing cellular secretory pathway.


Sujet(s)
Exocytose , Appareil de Golgi , Herpèsvirus humain de type 1 , Voie de sécrétion , Libération de particules virales , Protéines G rab , Herpèsvirus humain de type 1/physiologie , Herpèsvirus humain de type 1/métabolisme , Protéines G rab/métabolisme , Humains , Animaux , Appareil de Golgi/métabolisme , Appareil de Golgi/virologie , Cellules Vero , Réseau trans-golgien/métabolisme , Réseau trans-golgien/virologie , Chlorocebus aethiops , Herpès/virologie , Herpès/métabolisme , Virion/métabolisme , Cellules HeLa , Membrane cellulaire/métabolisme , Membrane cellulaire/virologie
11.
Curr Biol ; 34(18): 4170-4183.e9, 2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-39181128

RÉSUMÉ

Proteome maintenance in contracting skeletal and cardiac muscles depends on the chaperone-regulating protein BAG3. Reduced BAG3 activity leads to muscle weakness and heart failure in animal models and patients. BAG3 and its chaperone partners recognize mechanically damaged muscle proteins and initiate their disposal through chaperone-assisted selective autophagy (CASA). However, molecular details of the force-dependent regulation of BAG3 have remained elusive so far. Here, we demonstrate that mechanical stress triggers the dephosphorylation of BAG3 in human muscle and in isolated cells. We identify force-regulated phospho-switches in BAG3 that control CASA complex assembly and CASA activity. Differential proteomics reveal RAB GTPases, which organize membrane traffic and fusion, as dephosphorylation-dependent interactors of BAG3. In fact, RAB7A and RAB11B are shown here to be essential for CASA in skeletal muscle cells. Moreover, BAG3 dephosphorylation is also observed upon induction of mitophagy, suggesting an involvement of the cochaperone in the RAB7A-dependent autophagic engulfment of damaged mitochondria in exercised muscle. Cooperation of BAG3 with RAB7A relies on a direct interaction of both proteins, which is regulated by the nucleotide state of the GTPase and by association with the autophagosome membrane protein LC3B. Finally, we provide evidence that BAG3 and RAB7A also cooperate in non-muscle cells and propose that overactivation of CASA in RAB7A-L129F patients contributes to the loss of peripheral neurons in Charcot-Marie-Tooth neuropathy.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Protéines régulatrices de l'apoptose , Protéines G rab , Protéines Rab7 liant le GTP , Humains , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Phosphorylation , Protéines G rab/métabolisme , Protéines G rab/génétique , Protéines Rab7 liant le GTP/métabolisme , Homéostasie protéique , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Muscles squelettiques/métabolisme , Autophagie/physiologie , Animaux , Souris , Transport des protéines
12.
Dev Cell ; 59(17): 2287-2301.e6, 2024 Sep 09.
Article de Anglais | MEDLINE | ID: mdl-39111309

RÉSUMÉ

Autophagy is a universal degradation system in eukaryotic cells. In plants, although autophagosome biogenesis has been extensively studied, the mechanism of how autophagosomes are transported to the vacuole for degradation remains largely unexplored. In this study, we demonstrated that upon autophagy induction, Arabidopsis homotypic fusion and protein sorting (HOPS) subunit VPS41 converts first from condensates to puncta, then to ring-like structures, termed VPS41-associated phagic vacuoles (VAPVs), which enclose autophagy-related gene (ATG)8s for vacuolar degradation. This process is initiated by ADP ribosylation factor (ARF)-like GTPases ARLA1s and occurs concurrently with autophagy progression through coupling with the synaptic-soluble N-ethylmaleimide-sensitive factor attachment protein rmleceptor (SNARE) proteins. Unlike in other eukaryotes, autophagy degradation in Arabidopsis is largely independent of the RAB7 pathway. By contrast, dysfunction in the condensates-to-VAPVs conversion process impairs autophagosome structure and disrupts their vacuolar transport, leading to a significant reduction in autophagic flux and plant survival rate. Our findings suggest that the conversion pathway might be an integral part of the autophagy program unique to plants.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Autophagosomes , Autophagie , Vacuoles , Arabidopsis/métabolisme , Arabidopsis/génétique , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Vacuoles/métabolisme , Autophagosomes/métabolisme , Protéines du transport vésiculaire/métabolisme , Protéines du transport vésiculaire/génétique , Famille de la protéine-8 associée à l'autophagie/métabolisme , Famille de la protéine-8 associée à l'autophagie/génétique , Protéines SNARE/métabolisme , Protéines SNARE/génétique , Protéines Rab7 liant le GTP , Protéines G rab/métabolisme , Protéines G rab/génétique
13.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39125997

RÉSUMÉ

The transmembrane protein ß-amyloid precursor protein (APP) is central to the pathophysiology of Alzheimer's disease (AD). The ß-amyloid hypothesis posits that aberrant processing of APP forms neurotoxic ß-amyloid aggregates, which lead to the cognitive impairments observed in AD. Although numerous additional factors contribute to AD, there is a need to better understand the synaptic function of APP. We have found that Drosophila APP-like (APPL) has both shared and non-shared roles at the synapse with Kismet (Kis), a chromatin helicase binding domain (CHD) protein. Kis is the homolog of CHD7 and CHD8, both of which are implicated in neurodevelopmental disorders including CHARGE Syndrome and autism spectrum disorders, respectively. Loss of function mutations in kis and animals expressing human APP and BACE in their central nervous system show reductions in the glutamate receptor subunit, GluRIIC, the GTPase Rab11, and the bone morphogenetic protein (BMP), pMad, at the Drosophila larval neuromuscular junction (NMJ). Similarly, processes like endocytosis, larval locomotion, and neurotransmission are deficient in these animals. Our pharmacological and epistasis experiments indicate that there is a functional relationship between Kis and APPL, but Kis does not regulate appl expression at the larval NMJ. Instead, Kis likely influences the synaptic localization of APPL, possibly by promoting rab11 transcription. These data identify a potential mechanistic connection between chromatin remodeling proteins and aberrant synaptic function in AD.


Sujet(s)
Précurseur de la protéine bêta-amyloïde , Protéines de Drosophila , Jonction neuromusculaire , Protéines G rab , Animaux , Jonction neuromusculaire/métabolisme , Protéines de Drosophila/métabolisme , Protéines de Drosophila/génétique , Protéines G rab/métabolisme , Protéines G rab/génétique , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique , Drosophila melanogaster/métabolisme , Drosophila melanogaster/génétique , Transmission synaptique , Synapses/métabolisme , Récepteurs au glutamate/métabolisme , Récepteurs au glutamate/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Humains , Helicase/métabolisme , Helicase/génétique , Protéines membranaires , Protéines de tissu nerveux , Protéines à homéodomaine , Récepteurs ionotropes du glutamate
14.
Cell Calcium ; 123: 102945, 2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39191091

RÉSUMÉ

Orai1 is a plasma membrane Ca2+ channel involved in store operated calcium entry (SOCE). SOCE can regulate cell growth, exocytosis, gene expression and inflammation. We previously found that short palate lung and nasal epithelial clone 1's (SPLUNC1) sixth α-helix (α6) bound Orai1 to inhibit SOCE. SPLUNC1 was not proteolytically stable, so we developed ELD607, an 11 amino acid peptide based on SPLUNC1's α6 region which was more stable and more potent than SPLUNC1/α6. Here, we studied ELD607's mechanism of action. We overexpressed either Orai1-HA or Orai1-YFP in HEK293T cells to probe ELD607-Orai1 interactions by confocal microscopy. We also measured changes in Fluo-4 fluorescence in a multiplate reader as a marker of cytoplasmic Ca2+ levels. ELD607 internalized Orai1 independently of STIM1. Both 15 min and 3 h exposure to ELD607 similarly depleted Orai1 in the plasma membrane. However, 3 h exposure to ELD607 yielded greater inhibition of SOCE. ELD607 continued to colocalize with Orai1 after internalization and this process was dependent on the presence of the ubiquitin ligase NEDD4.2. Similarly, ELD607 increased the colocalization between Orai1 and ubiquitin. ELD607 also increased the colocalization between Orai1 and Rab5 and 7, but not Rab11, suggesting that Orai1 trafficked through early and late but not recycling endosomes. Finally, ELD607 caused Orai1, but not Orai2, Orai3, or STIM1 to traffic to lysosomes. We conclude that ELD607 rapidly binds to Orai1 and works in an identical fashion as full length SPLUNC1 by internalizing Orai1 and sending it to lysosomes, leading to a decrease in SOCE.


Sujet(s)
Calcium , Lysosomes , Protéine ORAI1 , Humains , Calcium/métabolisme , Canaux calciques/métabolisme , Membrane cellulaire/métabolisme , Cellules HEK293 , Lysosomes/métabolisme , Protéine ORAI1/métabolisme , Peptides/métabolisme , Peptides/pharmacologie , Transport des protéines , Protéines G rab/métabolisme , Protéines G rab5/métabolisme , Molécule-1 d'interaction stromale/métabolisme
15.
Oncogene ; 43(37): 2768-2780, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39138263

RÉSUMÉ

Peritoneal metastasis is one of the most common risk factors contributing to the poor prognosis of gastric cancer. We previously reported that extracellular vesicles from gastric cancer cells could facilitate peritoneal metastasis. However, their impact on gastric cancer-induced peritoneal metastasis under hypoxic conditions remains unclear. This study aims to elucidate how hypoxia-resistant gastric cancer cell-derived extracellular vesicles affect the peritoneal metastasis of normoxic gastric cancer cells. Proteomic analysis revealed elevated levels of Caveolin1 and Laminin ß2 in hypoxia-resistant gastric cancer cells and their corresponding extracellular vesicles. Importantly, Caveolin1 was found to play a central role in mediating Laminin ß2 sorting into extracellular vesicles derived from hypoxia-resistant gastric cancer cells, and subsequently, extracellular vesicle-associated Laminin ß2 promoted peritoneal metastasis in normoxic gastric cancer cells by activating the AKT pathway. Further investigation confirmed that Caveolin1 activation by Rho-related Coiled-coil kinase 1-mediated phosphorylation of Y14 residue is a key factor facilitating Laminin ß2 sorting into extracellular vesicles. Moreover, Y14 phosphorylated- Caveolin1 enhanced Laminin ß2 sorting by activating Rab11. Finally, our study demonstrated that a combined assessment of plasma extracellular vesicle-associated Caveolin1 and extracellular vesicle-associated Laminin ß2 could provide an accurate predictive tool for peritoneal metastasis occurrence in gastric cancer.


Sujet(s)
Cavéoline-1 , Vésicules extracellulaires , Tumeurs du péritoine , Tumeurs de l'estomac , Protéines G rab , rho-Associated Kinases , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Humains , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/métabolisme , Animaux , rho-Associated Kinases/métabolisme , Vésicules extracellulaires/métabolisme , Souris , Protéines G rab/métabolisme , Protéines G rab/génétique , Lignée cellulaire tumorale , Transduction du signal , Mâle , Femelle
16.
J Microbiol ; 62(8): 581-590, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39212865

RÉSUMÉ

Most bacteria will use their toxins to interact with the host cell, causing damage to the cell and then escaping from it. When bacteria enter the cell, they will be transported via the endosomal pathway. Rab GTPases are involved in bacterial transport as major components of endosomes that bind to their downstream effector proteins. The bacteria manipulate some Rab GTPases, escape the cell, and get to survive. In this review, we will focus on summarizing the many processes of how bacteria manipulate Rab GTPases to control their escape.


Sujet(s)
Bactéries , Endosomes , Interactions hôte-pathogène , Protéines G rab , Protéines G rab/métabolisme , Bactéries/métabolisme , Bactéries/enzymologie , Bactéries/génétique , Endosomes/métabolisme , Humains , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Transport des protéines , Animaux , Transport biologique
17.
PLoS One ; 19(8): e0308982, 2024.
Article de Anglais | MEDLINE | ID: mdl-39213285

RÉSUMÉ

Breast cancer is the most frequently diagnosed cancer worldwide, and the incidence rate has increased enormously over the last three decades. Rab proteins are members of the Rab GTPase superfamily. The aberrant function of these proteins leads to the development of tumors. Mentha longifolia var. asiatica and Zygophyllum arabicum have been known for their therapeutic potential for ages. The present study aimed to synthesize ZnO nanoparticles encapsulated with the extracts of M. longifolia var. asiatica and Z. arabicum and evaluating their therapeutic potential against breast cancer, targeting the Rab22A gene and its protein. UV-Vis spectrophotometer showed characteristic absorbance peaks at 295 nm and 345 nm for Z. arabicum and M. longifolia var. asiatica ZnONPs, respectively. The FTIR bands of Z. arabicum nanoparticles suggested the presence of aldehydes, alcohols, and polyols whereas bands of M. longifolia var. asiatica ZnONPs suggested the presence of carboxyl groups, hydroxyl groups, alkynes, and amines. SEM revealed the size of Z. arabicum ZnO NPs to be 25 ± 4 nm with a spherical shape as compared to nanoparticles of M. longifolia var. asiatica having a size of 35 ± 6 nm with a hexagonal shape. EDX determined the elemental composition of both particles. The cytotoxicity of both plant extracts and respective NPs was determined against the MCF-7 breast cancer cell line, which was found to be significant with an IC50 value of 51.68 µM for Z. arabicum and 88.02 µM for M. longifolia var. asiatica ZnO compared to plant extracts (64.01 µM and 107.9 µM for Z. arabicum and M. longifolia var. asiatica). The gene expression and protein levels of Rab22A were decreased in nanoparticle-treated cells as compared to the control group. The apoptotic role of synthesized nanoparticles against the MCF-7 cell line was also determined by the expression of apoptotic pathway genes and proteins (bax, caspase 3, caspase 8 and caspase 9). All samples showed significant apoptotic activity by activating intrinsic and extrinsic pathway genes. The activity of Z. arabicum was more eminent as compared to M. longifolia var. asiatica which was evident by the greater expression of studied genes and proteins as determined by Real-time qPCR and ELISA. This is the first-ever report describing the comparative analysis of the efficacy of Z. arabicum and M. longifolia var. asiatica ZnONPs against breast cancer.


Sujet(s)
Tumeurs du sein , Mentha , Extraits de plantes , Oxyde de zinc , Protéines G rab , Humains , Oxyde de zinc/pharmacologie , Oxyde de zinc/composition chimique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Protéines G rab/métabolisme , Protéines G rab/génétique , Mentha/composition chimique , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Cellules MCF-7 , Nanoparticules/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Nanoparticules métalliques/composition chimique
18.
Front Immunol ; 15: 1403008, 2024.
Article de Anglais | MEDLINE | ID: mdl-39192986

RÉSUMÉ

Background: As one of the most common cancer, colorectal cancer (CRC) is with high morbidity and mortality. Peritoneal metastasis (PM) is a fatal state of CRC, and few patients may benefit from traditional therapies. There is a complex interaction between PM and immune cell infiltration. Therefore, we aimed to determine biomarkers associated with colorectal cancer peritoneal metastasis (CRCPM) and their relationship with immune cell infiltration. Methods: By informatic analysis, differently expressed genes (DEGs) were selected and hub genes were screened out. RAB13, one of the hub genes, was identificated from public databases and validated in CRC tissues. The ESTIMATE, CEBERSORT and TIMER algorithms were applied to analyze the correlation between RAB13 and immune infiltration in CRC. RAB13's expression in different cells were analyzed at the single-cell level in scRNA-Seq. The Gene Set Enrichment Analysis (GSEA) was performed for RAB13 enrichment and further confirmed. Using oncoPredict algorithm, RAB13's impact on drug sensitivity was evaluated. Results: High RAB13 expression was identified in public databases and led to a poor prognosis. RAB13 was found to be positively correlated with the macrophages and other immune cells infiltration and from scRNA-Seq, RAB13 was found to be located in CRC cells and macrophages. GSEA revealed that high RAB13 expression enriched in a various of biological signaling, and oncoPredict algorithm showed that RAB13 expression was correlated with paclitaxel sensitivity. Conclusion: Our study indicated clinical role of RAB13 in CRC-PM, suggesting its potential as a therapeutic target in the future.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs colorectales , Tumeurs du péritoine , Protéines G rab , Humains , Protéines G rab/génétique , Protéines G rab/métabolisme , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/immunologie , Tumeurs colorectales/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/génétique , Tumeurs du péritoine/immunologie , Régulation de l'expression des gènes tumoraux , Pronostic , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Femelle , Mâle , Analyse de profil d'expression de gènes
19.
Nat Commun ; 15(1): 6824, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39122694

RÉSUMÉ

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.


Sujet(s)
Actines , Cryomicroscopie électronique , Mixed function oxygenases , Humains , Actines/métabolisme , Mixed function oxygenases/métabolisme , Mixed function oxygenases/composition chimique , Protéines des microfilaments/métabolisme , Protéines des microfilaments/génétique , Protéines des microfilaments/composition chimique , Liaison aux protéines , Cytosquelette d'actine/métabolisme , Modèles moléculaires , Protéines G rab/métabolisme , Protéines G rab/génétique , Protéines du cytosquelette/métabolisme , Protéines du cytosquelette/composition chimique , Protéines du cytosquelette/génétique , Domaines protéiques ,
20.
Cell Rep ; 43(8): 114598, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39126651

RÉSUMÉ

Endosomal Toll-like receptors (eTLRs) are essential for the sensing of non-self through RNA and DNA detection. Here, using spatiotemporal analysis of vesicular dynamics, super-resolution microscopy studies, and functional assays, we show that endomembrane defects associated with the deficiency of the small GTPase Rab27a cause delayed eTLR ligand recognition, defective early signaling, and impaired cytokine secretion. Rab27a-deficient neutrophils show retention of eTLRs in amphisomes and impaired ligand internalization. Extracellular signal-regulated kinase (ERK) signaling and ß2-integrin upregulation, early responses to TLR7 and TLR9 ligands, are defective in Rab27a deficiency. CpG-stimulated Rab27a-deficient neutrophils present increased tumor necrosis factor alpha (TNF-α) secretion and decreased secretion of a selected group of mediators, including interleukin (IL)-10. In vivo, CpG-challenged Rab27a-null mice show decreased production of type I interferons (IFNs) and IFN-γ, and the IFN-α secretion defect is confirmed in Rab27a-null plasmacytoid dendritic cells. Our findings have significant implications for immunodeficiency, inflammation, and CpG adjuvant vaccination.


Sujet(s)
Cytokines , Récepteur-9 de type Toll-like , Protéines rab27 liant le GTP , Animaux , Protéines rab27 liant le GTP/métabolisme , Protéines rab27 liant le GTP/génétique , Souris , Cytokines/métabolisme , Récepteur-9 de type Toll-like/métabolisme , Récepteur-9 de type Toll-like/déficit , Protéines G rab/métabolisme , Protéines G rab/déficit , Protéines G rab/génétique , Récepteur de type Toll-7/métabolisme , Récepteur de type Toll-7/déficit , Récepteur de type Toll-7/génétique , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Endosomes/métabolisme , Souris de lignée C57BL , Souris knockout , Facteur de nécrose tumorale alpha/métabolisme , Acides nucléiques/métabolisme , Transduction du signal , Interféron gamma/métabolisme , Glycoprotéines membranaires
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE