Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 796
Filtrer
1.
Oncogene ; 43(34): 2564-2577, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39020072

RÉSUMÉ

Perineural invasion (PNI) is an adverse prognostic feature of pancreatic ductal adenocarcinoma (PDAC). However, the understanding of the interactions between tumors and neural signaling within the tumor microenvironment is limited. In the present study, we found that MUC21 servers as an independent risk factor for poor prognosis in PDAC. Furthermore, we demonstrated that MUC21 promoted the metastasis and PNI of PDAC cells by activating JNK and inducing epithelial-mesenchymal transition (EMT). Mechanistically, glial cell-derived neurotrophic factor, secreted by Schwann cells, phosphorylates the intracellular domain S543 of MUC21 via CDK1 in PDAC cells, facilitating the interaction between MUC21 and RAC2. This interaction leads to membrane anchoring and activation of RAC2, which in turn activates the JNK/ZEB1/EMT axis, ultimately enhancing the metastasis and PNI of PDAC cells. Our results present a novel mechanism of PNI, suggesting that MUC21 is a potential prognostic marker and therapeutic target for PDAC.


Sujet(s)
Carcinome du canal pancréatique , Transition épithélio-mésenchymateuse , Invasion tumorale , Tumeurs du pancréas , , Humains , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/génétique , Phosphorylation , Animaux , Lignée cellulaire tumorale , Souris , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/métabolisme , Carcinome du canal pancréatique/génétique , Facteur neurotrophique dérivé des cellules gliales/métabolisme , Protéines G rac/métabolisme , Protéines G rac/génétique , Pronostic , Métastase tumorale , Mâle , Femelle , Souris nude
2.
J Cell Mol Med ; 28(11): e18473, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847477

RÉSUMÉ

Bladder cancer is one of the most prevalent cancers worldwide, and its morbidity and mortality rates have been increasing over the years. However, how RAC family small GTPase 3 (RAC3) affects the proliferation, migration and invasion of cisplatin-resistant bladder cancer cells remains unclear. Bioinformatics techniques were used to investigate the expression of RAC3 in bladder cancer tissues. Influences of RAC3 in the grade, stage, distant metastasis, and survival rate of bladder cancer were also examined. Analysis of the relationship between RAC3 expression and the immune microenvironment (TIME), genomic mutations, and stemness index. In normal bladder cancer cells (T24, 5637, and BIU-87) and cisplatin-resistant bladder cancer cells (BIU-87-DDP), the expression of RAC3 was detected separately with Western blotting. Plasmid transfection was used to overexpress or silence the expression of RAC3 in bladder cancer cells resistant to cisplatin (BIU-87-DDP). By adding activators and inhibitors, the activities of the JNK/MAPK signalling pathway were altered. Cell viability, invasion, and its level of apoptosis were measured in vitro using CCK-8, transwell, and flow cytometry. The bioinformatics analyses found RAC3 levels were elevated in bladder cancer tissues and were associated with a poor prognosis in bladder cancer. RAC3 in BIU-87-DDP cells expressed a higher level than normal bladder cancer cells. RAC3 overexpression promoted BIU-87-DDP proliferation. The growth of BIU-87-DDP cells slowed after the knockdown of RAC3, and RAC3 may have had an impact on the activation of the JNK/MAPK pathway.


Sujet(s)
Apoptose , Mouvement cellulaire , Prolifération cellulaire , Cisplatine , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Invasion tumorale , Tumeurs de la vessie urinaire , Protéines G rac , Humains , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/métabolisme , Tumeurs de la vessie urinaire/traitement médicamenteux , Cisplatine/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Protéines G rac/métabolisme , Protéines G rac/génétique , Apoptose/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Femelle , Mâle , Adulte d'âge moyen , Microenvironnement tumoral , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
3.
Bone Res ; 12(1): 35, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849345

RÉSUMÉ

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


Sujet(s)
Facteur de stimulation des colonies de macrophages , Ostéoclastes , Transduction du signal , Protéines G rap1 , Animaux , Ostéoclastes/métabolisme , Facteur de stimulation des colonies de macrophages/métabolisme , Facteur de stimulation des colonies de macrophages/génétique , Protéines G rap1/métabolisme , Protéines G rap1/génétique , Souris , Cytosquelette/métabolisme , Souris knockout , Souris de lignée C57BL , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Protéines G rac/métabolisme , Protéines G rac/génétique , Glycoprotéines membranaires/métabolisme , Glycoprotéines membranaires/génétique , Récepteurs immunologiques/métabolisme , Récepteurs immunologiques/génétique , Immunoglobulines
4.
Curr Biol ; 34(11): 2387-2402.e5, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38776905

RÉSUMÉ

The C. elegans hermaphrodite distal tip cell (DTC) leads gonadogenesis. Loss-of-function mutations in a C. elegans ortholog of the Rac1 GTPase (ced-10) and its GEF complex (ced-5/DOCK180, ced-2/CrkII, ced-12/ELMO) cause gonad migration defects related to directional sensing; we discovered an additional defect class of gonad bifurcation in these mutants. Using genetic approaches, tissue-specific and whole-body RNAi, and in vivo imaging of endogenously tagged proteins and marked cells, we find that loss of Rac1 or its regulators causes the DTC to fragment as it migrates. Both products of fragmentation-the now-smaller DTC and the membranous patch of cellular material-localize important stem cell niche signaling (LAG-2 ligand) and migration (INA-1/integrin subunit alpha) factors to their membranes, but only one retains the DTC nucleus and therefore the ability to maintain gene expression over time. The enucleate patch can lead a bifurcating branch off the gonad arm that grows through germ cell proliferation. Germ cells in this branch differentiate as the patch loses LAG-2 expression. While the nucleus is surprisingly dispensable for aspects of leader cell function, it is required for stem cell niche activity long term. Prior work found that Rac1-/-;Rac2-/- mouse erythrocytes fragment; in this context, our new findings support the conclusion that maintaining a cohesive but deformable cell is a conserved function of this important cytoskeletal regulator.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Mouvement cellulaire , Gonades , Organogenèse , Transduction du signal , Animaux , Caenorhabditis elegans/génétique , Caenorhabditis elegans/croissance et développement , Gonades/métabolisme , Gonades/croissance et développement , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Organogenèse/génétique , Protéines G rac/métabolisme , Protéines G rac/génétique
5.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38732116

RÉSUMÉ

Hypertension is a pervasive and widespread health condition that poses a significant risk factor for cardiovascular disease, which includes conditions such as heart attack, stroke, and heart failure. Despite its widespread occurrence, the exact cause of hypertension remains unknown, and the mechanisms underlying the progression from prehypertension to hypertension require further investigation. Recent proteomic studies have shown promising results in uncovering potential biomarkers related to disease development. In this study, serum proteomic data collected from Qatar Biobank were analyzed to identify altered protein expression between individuals with normal blood pressure, prehypertension, and hypertension and to elucidate the biological pathways contributing to this disease. The results revealed a cluster of proteins, including the SRC family, CAMK2B, CAMK2D, TEC, GSK3, VAV, and RAC, which were markedly upregulated in patients with hypertension compared to those with prehypertension (fold change ≥ 1.6 or ≤-1.6, area under the curve ≥ 0.8, and q-value < 0.05). Pathway analysis showed that the majority of these proteins play a role in actin cytoskeleton remodeling. Actin cytoskeleton reorganization affects various biological processes that contribute to the maintenance of blood pressure, including vascular tone, endothelial function, cellular signaling, inflammation, fibrosis, and mechanosensing. Therefore, the findings of this study suggest a potential novel role of actin cytoskeleton-related proteins in the progression from prehypertension to hypertension. The present study sheds light on the underlying pathological mechanisms involved in hypertension and could pave the way for new diagnostic and therapeutic approaches for the treatment of this disease.


Sujet(s)
Cytosquelette d'actine , Hypertension artérielle , Protéomique , Femelle , Humains , Mâle , Cytosquelette d'actine/métabolisme , Marqueurs biologiques , Pression sanguine , Hypertension artérielle/métabolisme , Préhypertension/métabolisme , Protéome/métabolisme , Protéomique/méthodes , Glycogen Synthase Kinase 3/génétique , Glycogen Synthase Kinase 3/métabolisme , Protéines proto-oncogènes c-vav/génétique , Protéines proto-oncogènes c-vav/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme
6.
Childs Nerv Syst ; 40(5): 1597-1602, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38214746

RÉSUMÉ

Pathogenic variants in RAC3 cause a neurodevelopmental disorder with brain malformations and craniofacial dysmorphism, called NEDBAF. This gene encodes a small GTPase, which plays a critical role in neurogenesis and neuronal migration. We report a 31 weeks of gestation fetus with triventricular dilatation, and temporal and perisylvian polymicrogyria, without cerebellar, brainstem, or callosal anomalies. Trio whole exome sequencing identified a RAC3 (NM_005052.3, GRCh38) probably pathogenic de novo variant c.276 T>A p.(Asn92Lys). Eighteen patients harboring 13 different and essentially de novo missense RAC3 variants were previously reported. All the patients presented with corpus callosum malformations. Gyration disorders, ventriculomegaly (VM), and brainstem and cerebellar malformations have frequently been described. The only previous prenatal case associated with RAC3 variant presented with complex brain malformations, mainly consisting of midline and posterior fossa anomalies. We report the second prenatal case of NEDBAF presenting an undescribed pattern of cerebral anomalies, including VM and polymicrogyria, without callosal, cerebellar, or brainstem malformations. All neuroimaging data were reviewed to clarify the spectrum of cerebral malformations.


Sujet(s)
Hydrocéphalie , Malformations du système nerveux , Polymicrogyrie , Grossesse , Femelle , Humains , Diagnostic prénatal , Agénésie du corps calleux , Mutation faux-sens , Protéines G rac/génétique
7.
Infect Immun ; 92(2): e0038023, 2024 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-38168666

RÉSUMÉ

Macrophages act as a first line of defense against pathogens. Against Aspergillus fumigatus, a fungus with pathogenic potential in immunocompromised patients, macrophages can phagocytose fungal spores and inhibit spore germination to prevent the development of tissue-invasive hyphae. However, the cellular pathways that macrophages use to accomplish these tasks and any roles macrophages have later in infection against invasive forms of fungi are still not fully known. Rac-family Rho GTPases are signaling hubs for multiple cellular functions in leukocytes, including cell migration, phagocytosis, reactive oxygen species (ROS) generation, and transcriptional activation. We therefore aimed to further characterize the function of macrophages against A. fumigatus in an in vivo vertebrate infection model by live imaging of the macrophage behavior in A. fumigatus-infected rac2 mutant zebrafish larvae. While Rac2-deficient zebrafish larvae are susceptible to A. fumigatus infection, Rac2 deficiency does not impair macrophage migration to the infection site, interaction with and phagocytosis of spores, spore trafficking to acidified compartments, or spore killing. However, we reveal a role for Rac2 in macrophage-mediated inhibition of spore germination and control of invasive hyphae. Re-expression of Rac2 under a macrophage-specific promoter rescues the survival of A. fumigatus-infected rac2 mutant larvae through increased control of germination and hyphal growth. Altogether, we describe a new role for macrophages against extracellular hyphal growth of A. fumigatus and report that the function of the Rac2 Rho GTPase in macrophages is required for this function.


Sujet(s)
Aspergillose , Danio zébré , Animaux , Humains , Danio zébré/microbiologie , dGTPases , Macrophages/microbiologie , Phagocytose , Aspergillose/microbiologie , Aspergillus fumigatus/physiologie , Spores fongiques , Protéines G rac/génétique , Protéines de poisson-zèbre/génétique
8.
Blood ; 143(15): 1476-1487, 2024 04 11.
Article de Anglais | MEDLINE | ID: mdl-38194689

RÉSUMÉ

ABSTRACT: Mutations in the small Rho-family guanosine triphosphate hydrolase RAC2, critical for actin cytoskeleton remodeling and intracellular signal transduction, are associated with neonatal severe combined immunodeficiency (SCID), infantile neutrophilic disorder resembling leukocyte adhesion deficiency (LAD), and later-onset combined immune deficiency (CID). We investigated 54 patients (23 previously reported) from 37 families yielding 15 novel RAC2 missense mutations, including one present only in homozygosity. Data were collected from referring physicians and literature reports with updated clinical information. Patients were grouped by presentation: neonatal SCID (n = 5), infantile LAD-like disease (n = 5), or CID (n = 44). Disease correlated to RAC2 activity: constitutively active RAS-like mutations caused neonatal SCID, dominant-negative mutations caused LAD-like disease, whereas dominant-activating mutations caused CID. Significant T- and B-lymphopenia with low immunoglobulins were seen in most patients; myeloid abnormalities included neutropenia, altered oxidative burst, impaired neutrophil migration, and visible neutrophil macropinosomes. Among 42 patients with CID with clinical data, upper and lower respiratory infections and viral infections were common. Twenty-three distinct RAC2 mutations, including 15 novel variants, were identified. Using heterologous expression systems, we assessed downstream effector functions including superoxide production, p21-activated kinase 1 binding, AKT activation, and protein stability. Confocal microscopy showed altered actin assembly evidenced by membrane ruffling and macropinosomes. Altered protein localization and aggregation were observed. All tested RAC2 mutant proteins exhibited aberrant function; no single assay was sufficient to determine functional consequence. Most mutants produced elevated superoxide; mutations unable to support superoxide formation were associated with bacterial infections. RAC2 mutations cause a spectrum of immune dysfunction, ranging from early onset SCID to later-onset combined immunodeficiencies depending on RAC2 activity. This trial was registered at www.clinicaltrials.gov as #NCT00001355 and #NCT00001467.


Sujet(s)
Déficits immunitaires , Déficit d'adhérence leucocytaire , Maladies d'immunodéficience primaire , Immunodéficience combinée grave , Humains , Nouveau-né , Déficits immunitaires/génétique , Déficits immunitaires/métabolisme , Granulocytes neutrophiles/métabolisme , Maladies d'immunodéficience primaire/génétique , Maladies d'immunodéficience primaire/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/métabolisme , , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/métabolisme , Superoxydes/métabolisme
9.
Nucleic Acids Res ; 52(3): 1387-1403, 2024 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-38015468

RÉSUMÉ

While the majority of circRNAs are formed from infrequent back-splicing of exons from protein coding genes, some can be produced at quite high level and in a regulated manner. We describe the regulation, biogenesis and function of circDOCK1(2-27), a large, abundant circular RNA that is highly regulated during epithelial-mesenchymal transition (EMT) and whose formation depends on the epithelial splicing regulator ESRP1. CircDOCK1(2-27) synthesis in epithelial cells represses cell motility both by diverting transcripts from DOCK1 mRNA production to circRNA formation and by direct inhibition of migration by the circRNA. HITS-CLIP analysis and CRISPR-mediated deletions indicate ESRP1 controls circDOCK1(2-27) biosynthesis by binding a GGU-containing repeat region in intron 1 and detaining its splicing until Pol II completes its 157 kb journey to exon 27. Proximity-dependent biotinylation (BioID) assay suggests ESRP1 may modify the RNP landscape of intron 1 in a way that disfavours communication of exon 1 with exon 2, rather than physically bridging exon 2 to exon 27. The X-ray crystal structure of RNA-bound ESRP1 qRRM2 domain reveals it binds to GGU motifs, with the guanines embedded in clamp-like aromatic pockets in the protein.


Sujet(s)
Épissage alternatif , ARN circulaire , Protéines de liaison à l'ARN , Protéines G rac , ARN/génétique , ARN/métabolisme , Épissage des ARN , ARN circulaire/génétique , Humains , Lignée cellulaire tumorale , Protéines de liaison à l'ARN/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme
10.
Proc Natl Acad Sci U S A ; 120(52): e2310221120, 2023 Dec 26.
Article de Anglais | MEDLINE | ID: mdl-38109551

RÉSUMÉ

The 21kD GTPase Rac is an evolutionarily ancient regulator of cell shape and behavior. Rac2 is predominantly expressed in hematopoietic cells where it is essential for survival and motility. The hyperactivating mutation Rac2E62K also causes human immunodeficiency, although the mechanism remains unexplained. Here, we report that in Drosophila, hyperactivating Rac stimulates ovarian cells to cannibalize neighboring cells, destroying the tissue. We then show that hyperactive Rac2E62K stimulates human HL60-derived macrophage-like cells to engulf and kill living T cell leukemia cells. Primary mouse Rac2+/E62K bone-marrow-derived macrophages also cannibalize primary Rac2+/E62K T cells due to a combination of macrophage hyperactivity and T cell hypersensitivity to engulfment. Additionally, Rac2+/E62K macrophages non-autonomously stimulate wild-type macrophages to engulf T cells. Rac2E62K also enhances engulfment of target cancer cells by chimeric antigen receptor-expressing macrophages (CAR-M) in a CAR-dependent manner. We propose that Rac-mediated cell cannibalism may contribute to Rac2+/E62K human immunodeficiency and enhance CAR-M cancer immunotherapy.


Sujet(s)
Déficits immunitaires , Tumeurs , Récepteurs chimériques pour l'antigène , Animaux , Souris , Humains , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/métabolisme , Cannibalisme , Macrophages/métabolisme , Déficits immunitaires/génétique , Mort cellulaire
11.
Turk J Ophthalmol ; 53(6): 343-348, 2023 12 21.
Article de Anglais | MEDLINE | ID: mdl-38014881

RÉSUMÉ

Objectives: To determine the roles of small GTP-binding proteins Rac1, Rac2, and Rac3 expression in pterygial tissue and to compare these expressions with normal conjunctival tissue. Materials and Methods: Seventy-eight patients with primary pterygium were enrolled. Healthy conjunctival graft specimens obtained during pterygium surgery were used as control tissue. The real-time polymerase chain reaction method on the BioMark HD dynamic array system was utilized in genomic mRNA for the gene expression analysis. Protein expressions were analyzed using western blot and immunohistochemical methods. Results: RAC1, RAC2, and RAC3 gene expressions in pterygial tissues were not markedly elevated when compared to the control specimens (p>0.05). As a very low level of RAC1 gene expression was observed, further protein expression analysis was performed for the Rac2 and Rac3 proteins. Western blot and immunohistochemical analysis of Rac2 and Rac3 protein expression revealed no significant differences between pterygial and healthy tissues (p>0.05). Conclusion: This is the first study to identify the contribution of Rac proteins in pterygium. Our results indicate that the small GTP-binding protein Rac may not be involved in pterygium pathogenesis.


Sujet(s)
Ptérygion , Humains , Ptérygion/chirurgie , Ptérygion/génétique , Ptérygion/métabolisme , Conjonctive/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Technique de Western
12.
J Cell Sci ; 136(19)2023 10 01.
Article de Anglais | MEDLINE | ID: mdl-37737020

RÉSUMÉ

The Rho family GTPases Rac and Rho play critical roles in transmitting mechanical information contained within the extracellular matrix (ECM) to the cell. Rac and Rho have well-described roles in regulating stiffness-dependent actin remodeling, proliferation and motility. However, much less is known about the relative roles of these GTPases in stiffness-dependent transcription, particularly at the genome-wide level. Here, we selectively inhibited Rac and Rho in mouse embryonic fibroblasts cultured on deformable substrata and used RNA sequencing to elucidate and compare the contribution of these GTPases to the early transcriptional response to ECM stiffness. Surprisingly, we found that the stiffness-dependent activation of Rac was dominant over Rho in the initial transcriptional response to ECM stiffness. We also identified activating transcription factor 3 (ATF3) as a major target of stiffness- and Rac-mediated signaling and show that ATF3 repression by ECM stiffness helps to explain how the stiffness-dependent activation of Rac results in the induction of cyclin D1.


Sujet(s)
Facteur de transcription ATF-3 , Fibroblastes , Animaux , Souris , Facteur de transcription ATF-3/génétique , Matrice extracellulaire/métabolisme , Fibroblastes/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéines G rho/métabolisme , Transduction du signal
13.
J Cell Mol Med ; 27(16): 2385-2397, 2023 08.
Article de Anglais | MEDLINE | ID: mdl-37386813

RÉSUMÉ

Endometrial cancer (EC) is one of the most common gynaecological malignant tumours with a high incidence, leading to urgent demands for exploring novel carcinogenic mechanisms and developing rational therapeutic strategies. The rac family of small GTPase 3 (RAC3) functions as an oncogene in various human malignant tumours and plays an important role in tumour development. However, the critical roles of RAC3 in the progression of EC need further investigation. Based on TCGA, single-cell RNA-Seq, CCLE and clinical specimens, we revealed that the RAC3 was specifically distributed in EC tumour cells compared to normal tissues and functioned as an independent diagnostic marker with a high area under curve (AUC) score. Meanwhile, the RAC3 expression in EC tissues was also correlated with a poor prognosis. In detail, the high levels of RAC3 in EC tissues were reversely associated with CD8+ T cell infiltration and orchestrated an immunosuppressive microenvironment. Furthermore, RAC3 accelerated tumour cell proliferation and inhibited its apoptosis, without impacting cell cycle stages. Importantly, silencing RAC3 improved the sensitivity of EC cells to chemotherapeutic drugs. In this paper, we revealed that RAC3 was predominantly expressed in EC and significantly correlated with the progression of EC via inducing immunosuppression and regulating tumour cell viability, providing a novel diagnostic biomarker and a promising strategy for sensitizing chemotherapy to EC.


Sujet(s)
Tumeurs de l'endomètre , Femelle , Humains , Pronostic , Tumeurs de l'endomètre/traitement médicamenteux , Tumeurs de l'endomètre/génétique , Prolifération cellulaire , Division cellulaire , Marqueurs biologiques , Microenvironnement tumoral/génétique , Protéines G rac/génétique , Protéines G rac/métabolisme
14.
J Mol Med (Berl) ; 101(7): 843-854, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37204479

RÉSUMÉ

Rac small GTPases play important roles during embryonic development of the inner ear; however, little is known regarding their function in cochlear hair cells (HCs) after specification. Here, we revealed the localization and activation of Racs in cochlear HCs using GFP-tagged Rac plasmids and transgenic mice expressing a Rac1-fluorescence resonance energy transfer (FRET) biosensor. Furthermore, we employed Rac1-knockout (Rac1-KO, Atoh1-Cre;Rac1flox/flox) and Rac1 and Rac3 double KO (Rac1/Rac3-DKO, Atoh1-Cre;Rac1flox/flox;Rac3-/-) mice, under the control of the Atoh1 promoter. However, both Rac1-KO and Rac1/Rac3-DKO mice exhibited normal cochlear HC morphology at 13 weeks of age and normal hearing function at 24 weeks of age. No hearing vulnerability was observed in young adult (6-week-old) Rac1/Rac3-DKO mice even after intense noise exposure. Consistent with prior reports, the results from Atoh1-Cre;tdTomato mice confirmed that the Atoh1 promoter became functional only after embryonic day 14 when the sensory HC precursors exit the cell cycle. Taken together, these findings indicate that although Rac1 and Rac3 contribute to the early development of sensory epithelia in cochleae, as previously shown, they are dispensable for the maturation of cochlear HCs in the postmitotic state or for hearing maintenance following HC maturation. KEY MESSAGES: Mice with Rac1 and Rac3 deletion were generated after HC specification. Knockout mice exhibit normal cochlear hair cell morphology and hearing. Racs are dispensable for hair cells in the postmitotic state after specification. Racs are dispensable for hearing maintenance after HC maturation.


Sujet(s)
Protéines G rac , Protéine G rac1 , Animaux , Souris , Protéines G rac/génétique , Protéines G rac/métabolisme , Souris knockout , Protéine G rac1/génétique , Protéine G rac1/métabolisme , Cellules ciliées auditives/métabolisme , Souris transgéniques
15.
Eur J Hum Genet ; 31(7): 805-814, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37059841

RÉSUMÉ

RAC1 is a member of the Rac/Rho GTPase subfamily within the RAS superfamily of small GTP-binding proteins, comprising 3 paralogs playing a critical role in actin cytoskeleton remodeling, cell migration, proliferation and differentiation. De novo missense variants in RAC1 are associated with a rare neurodevelopmental disorder (MRD48) characterized by DD/ID and brain abnormalities coupled with a wide range of additional features. Structural and functional studies have documented either a dominant negative or constitutively active behavior for a subset of mutations. Here, we describe two individuals with previously unreported de novo missense RAC1 variants. We functionally demonstrate their pathogenicity proving a gain-of-function (GoF) effect for both. By reviewing the clinical features of these two individuals and the previously published MRD48 subjects, we further delineate the clinical profile of the disorder, confirming its phenotypic variability. Moreover, we compare the main features of MRD48 with the neurodevelopmental disease caused by GoF variants in the paralog RAC3, highlighting similarities and differences. Finally, we review all previously reported variants in RAC proteins and in the closely related CDC42, providing an updated overview of the spectrum and hotspots of pathogenic variants affecting these functionally related GTPases.


Sujet(s)
Troubles du développement neurologique , Protéine G rac1 , Humains , Protéine G rac1/génétique , Protéine G rac1/composition chimique , Protéine G rac1/métabolisme , Protéines G rac/génétique , Mutation , Troubles du développement neurologique/génétique , Mutation faux-sens
16.
Proc Natl Acad Sci U S A ; 120(11): e2220825120, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36897976

RÉSUMÉ

Macroendocytosis comprising phagocytosis and macropinocytosis is an actin-driven process regulated by small GTPases that depend on the dynamic reorganization of the membrane that protrudes and internalizes extracellular material by cup-shaped structures. To effectively capture, enwrap, and internalize their targets, these cups are arranged into a peripheral ring or ruffle of protruding actin sheets emerging from an actin-rich, nonprotrusive zone at its base. Despite extensive knowledge of the mechanism driving actin assembly of the branched network at the protrusive cup edge, which is initiated by the actin-related protein (Arp) 2/3 complex downstream of Rac signaling, our understanding of actin assembly in the base is still incomplete. In the Dictyostelium model system, the Ras-regulated formin ForG was previously shown to specifically contribute to actin assembly at the cup base. Loss of ForG is associated with a strongly impaired macroendocytosis and a 50% reduction in F-actin content at the base of phagocytic cups, in turn indicating the presence of additional factors that specifically contribute to actin formation at the base. Here, we show that ForG synergizes with the Rac-regulated formin ForB to form the bulk of linear filaments at the cup base. Consistently, combined loss of both formins virtually abolishes cup formation and leads to severe defects of macroendocytosis, emphasizing the relevance of converging Ras- and Rac-regulated formin pathways in assembly of linear filaments in the cup base, which apparently provide mechanical support to the entire structure. Remarkably, we finally show that active ForB, unlike ForG, additionally drives phagosome rocketing to aid particle internalization.


Sujet(s)
Phagosomes , Dictyostelium , Formines/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéines G ras/génétique , Protéines G ras/métabolisme , Transduction du signal , Phagosomes/métabolisme , Actines/métabolisme
17.
J Cell Sci ; 136(6)2023 03 15.
Article de Anglais | MEDLINE | ID: mdl-36744839

RÉSUMÉ

Rho GTPases, among them Rac1 and Rac3, are major transducers of extracellular signals and are involved in multiple cellular processes. In cortical interneurons, the neurons that control the balance between excitation and inhibition of cortical circuits, Rac1 and Rac3 are essential for their development. Ablation of both leads to a severe reduction in the numbers of mature interneurons found in the murine cortex, which is partially due to abnormal cell cycle progression of interneuron precursors and defective formation of growth cones in young neurons. Here, we present new evidence that upon Rac1 and Rac3 ablation, centrosome, Golgi complex and lysosome positioning is significantly perturbed, thus affecting both interneuron migration and axon growth. Moreover, for the first time, we provide evidence of altered expression and localization of the two-pore channel 2 (TPC2) voltage-gated ion channel that mediates Ca2+ release. Pharmacological inhibition of TPC2 negatively affected axonal growth and migration of interneurons. Our data, taken together, suggest that TPC2 contributes to the severe phenotype in axon growth initiation, extension and interneuron migration in the absence of Rac1 and Rac3.


Sujet(s)
Canaux calciques , Interneurones , Protéines G rac , Protéine G rac1 , Animaux , Souris , Cônes de croissance/métabolisme , Interneurones/métabolisme , Neurones/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/génétique , Protéine G rac1/métabolisme , Canaux calciques/génétique , Canaux calciques/métabolisme
18.
J Med Genet ; 60(3): 223-232, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-35595279

RÉSUMÉ

BACKGROUND: RAC3 encodes a Rho family small GTPase that regulates the behaviour and organisation of actin cytoskeleton and intracellular signal transduction. Variants in RAC3 can cause a phenotypically heterogeneous neurodevelopmental disorder with structural brain anomalies and dysmorphic facies. The pathomechanism of this recently discovered genetic disorder remains unclear. METHODS: We investigated an early adolescent female with intellectual disability, drug-responsive epilepsy and white matter abnormalities. Through exome sequencing, we identified the novel de novo variant (NM_005052.3): c.83T>C (p.Phe28Ser) in RAC3. We then examined the pathophysiological significance of the p.F28S variant in comparison with the recently reported disease-causing p.Q61L variant, which results in a constitutively activated version of RAC3. RESULTS: In vitro analyses revealed that the p.F28S variant was spontaneously activated by substantially increased intrinsic GTP/GDP-exchange activity and bound to downstream effectors tested, such as PAK1 and MLK2. The variant suppressed the differentiation of primary cultured hippocampal neurons and caused cell rounding with lamellipodia. In vivo analyses using in utero electroporation showed that acute expression of the p.F28S variant caused migration defects of excitatory neurons and axon growth delay during corticogenesis. Notably, defective migration was rescued by a dominant negative version of PAK1 but not MLK2. CONCLUSION: Our results indicate that RAC3 is critical for brain development and the p.F28S variant causes morphological and functional defects in cortical neurons, likely due to the hyperactivation of PAK1.


Sujet(s)
Déficience intellectuelle , Troubles du développement neurologique , Adolescent , Humains , Femelle , Mutation gain de fonction , Troubles du développement neurologique/génétique , Neurogenèse , Déficience intellectuelle/génétique , Différenciation cellulaire , Protéines G rac/génétique , Protéines G rac/métabolisme
19.
Exp Neurol ; 361: 114316, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36586552

RÉSUMÉ

Rac1, a member of small Rho GTPases, is involved in diverse cellular processes in neuronal cells. Rac1 plays especially important roles during development, and its roles have been extensively studied using Rac1-deficient mice. Rac3, a close homolog of Rac1, is ubiquitously expressed in the nervous system and may therefore compensate for Rac1 in Rac1-deficient cells. Exploration of the roles of Rac in neurons may therefore be difficult. We thus deleted both Rac1 and Rac3 in cortical neurons. Rac-deficient cerebral cortices formed slightly hypoplastic but almost normally layered structures at birth, but cortical neurons underwent apoptosis soon after birth. Rac-deficient cortical neurons had poor survivability and there was reduction in the length and the number of neurites in vitro. Activation of Pak1, a downstream effector of Rac, in Rac-deficient cortical neurons rescued the survivability in vitro. Pak1-activated Rac-deficient neurons had numerous dendrites, but no axons. Restoration of p35, a regulator of Cdk5, partly rescued the survivability of Rac-deficient neurons both in vitro and in vivo. Expression of p35 also partly rescued the length and the number of neurites in Rac-deficient neurons in vitro. Rac was shown to be indispensable for the survival of cortical neurons, and Pak1 and Cdk5/p35 work as downstream effectors of Rac to promote neuronal survival.


Sujet(s)
Protéines G rac , Animaux , Souris , Axones/métabolisme , Neurites , Neurones/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/génétique , Protéine G rac1/métabolisme
20.
Sci Rep ; 12(1): 14848, 2022 09 01.
Article de Anglais | MEDLINE | ID: mdl-36050459

RÉSUMÉ

Granule neurons are the most common cell type in the cerebellum. They are generated in the external granule layer and migrate inwardly, forming the internal granule layer. Small Rho GTPases play various roles during development of the nervous system and may be involved in generation, differentiation and migration of granule neurons. We deleted Rac1, a member of small Rho GTPases, by GFAP-Cre driver in cerebellar granule neurons and Bergmann glial cells. Rac1flox/flox; Cre mice showed impaired migration and slight reduction in the number of granule neurons in the internal granule layer. Deletion of both Rac1 and Rac3 resulted in almost complete absence of granule neurons. Rac-deficient granule neurons differentiated into p27 and NeuN-expressing post mitotic neurons, but died before migration to the internal granule layer. Loss of Rac3 has little effect on granule neuron development. Rac1flox/flox; Rac3+/-; Cre mice showed intermediate phenotype between Rac1flox/flox; Cre and Rac1flox/flox; Rac3-/-; Cre mice in both survival and migration of granule neurons. Rac3 itself seems to be unimportant in the development of the cerebellum, but has some roles in Rac1-deleted granule neurons. Conversely, overall morphology of Rac1+/flox; Rac3-/-; Cre cerebella was normal. One allele of Rac1 is therefore thought to be sufficient to promote development of cerebellar granule neurons.


Sujet(s)
Cervelet , Neurogenèse , Protéines G rac , Protéine G rac1 , Animaux , Mort cellulaire , Mouvement cellulaire , Cervelet/métabolisme , Souris , Souris knockout , Neurogenèse/physiologie , Névroglie/métabolisme , Neurones/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/génétique , Protéine G rac1/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE