Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 161
Filtrer
1.
J Biol Chem ; : 107601, 2024 Jul 24.
Article de Anglais | MEDLINE | ID: mdl-39059493

RÉSUMÉ

Ubiquitination plays a crucial role in cellular homeostasis by regulating the degradation, localization, and activity of proteins, ensuring proper cell function and balance. Among E3 ubiquitin ligases, WWP1 is implicated in cell proliferation, survival and apoptosis. Notably WWP1 is frequently amplified in breast cancer and associated with poor prognosis. Here we identify the protein CYYR1 that had previously no assigned function, as a regulator of WWP1 activity and stability. We show that CYYR1 binds to the WW domains of the E3 ubiquitin ligase WWP1 through its PPxY motifs. This interaction triggers K63-linked auto-ubiquitination and subsequent degradation of WWP1. We furthermore demonstrate that CYYR1 localizes to late endosomal vesicles and directs poly-ubiquitinated WWP1 toward lysosomal degradation through binding to ANKRD13A. Moreover, we found that CYYR1 expression attenuates breast cancer cell growth in anchorage-dependent and independent colony formation assays in a PPxY-dependent manner. Finally, we highlight that CYYR1 expression is significantly decreased in breast cancer and is associated with beneficial clinical outcome. Taken together our study suggests tumor suppressor properties for CYYR1 through regulation of WWP1 auto-ubiquitination and lysosomal degradation.

2.
Elife ; 132024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38921824

RÉSUMÉ

While often undetected and untreated, persistent seasonal asymptomatic malaria infections remain a global public health problem. Despite the presence of parasites in the peripheral blood, no symptoms develop. Disease severity is correlated with the levels of infected red blood cells (iRBCs) adhering within blood vessels. Changes in iRBC adhesion capacity have been linked to seasonal asymptomatic malaria infections, however how this is occurring is still unknown. Here, we present evidence that RNA polymerase III (RNA Pol III) transcription in Plasmodium falciparum is downregulated in field isolates obtained from asymptomatic individuals during the dry season. Through experiments with in vitro cultured parasites, we have uncovered an RNA Pol III-dependent mechanism that controls pathogen proliferation and expression of a major virulence factor in response to external stimuli. Our findings establish a connection between P. falciparum cytoadhesion and a non-coding RNA family transcribed by Pol III. Additionally, we have identified P. falciparum Maf1 as a pivotal regulator of Pol III transcription, both for maintaining cellular homeostasis and for responding adaptively to external signals. These results introduce a novel perspective that contributes to our understanding of P. falciparum virulence. Furthermore, they establish a connection between this regulatory process and the occurrence of seasonal asymptomatic malaria infections.


Sujet(s)
Paludisme à Plasmodium falciparum , Plasmodium falciparum , RNA polymerase III , Plasmodium falciparum/génétique , Plasmodium falciparum/pathogénicité , Plasmodium falciparum/enzymologie , Virulence , RNA polymerase III/métabolisme , RNA polymerase III/génétique , Humains , Paludisme à Plasmodium falciparum/parasitologie , Érythrocytes/parasitologie , Protéines de protozoaire/métabolisme , Protéines de protozoaire/génétique , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Adhérence cellulaire , Régulation de l'expression des gènes
3.
Nat Cell Biol ; 26(7): 1093-1109, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38886558

RÉSUMÉ

Extracellular vesicles such as exosomes are now recognized as key players in intercellular communication. Their role is influenced by the specific repertoires of proteins and lipids, which are enriched when they are generated as intraluminal vesicles (ILVs) in multivesicular endosomes. Here we report that a key component of small extracellular vesicles, the tetraspanin CD63, sorts cholesterol to ILVs, generating a pool that can be mobilized by the NPC1/2 complex, and exported via exosomes to recipient cells. In the absence of CD63, cholesterol is retrieved from the endosomes by actin-dependent vesicular transport, placing CD63 and cholesterol at the centre of a balance between inward and outward budding of endomembranes. These results establish CD63 as a lipid-sorting mechanism within endosomes, and show that ILVs and exosomes are alternative providers of cholesterol.


Sujet(s)
Cholestérol , Endosomes , Exosomes , Antigène CD63 , Antigène CD63/métabolisme , Cholestérol/métabolisme , Exosomes/métabolisme , Endosomes/métabolisme , Humains , Animaux , Protéine NPC1 , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Transport biologique , Actines/métabolisme , Souris
4.
Dev Cell ; 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38834071

RÉSUMÉ

Normal cells coordinate proliferation and differentiation by precise tuning of gene expression based on the dynamic shifts of the epigenome throughout the developmental timeline. Although non-mutational epigenetic reprogramming is an emerging hallmark of cancer, the epigenomic shifts that occur during the transition from normal to malignant cells remain elusive. Here, we capture the epigenomic changes that occur during tumorigenesis in a prototypic embryonal brain tumor, medulloblastoma. By comparing the epigenomes of the different stages of transforming cells in mice, we identify nuclear factor I family of transcription factors, known to be cell fate determinants in development, as oncogenic regulators in the epigenomes of precancerous and cancerous cells. Furthermore, genetic and pharmacological inhibition of NFIB validated a crucial role of this transcription factor by disrupting the cancer epigenome in medulloblastoma. Thus, this study exemplifies how epigenomic changes contribute to tumorigenesis via non-mutational mechanisms involving developmental transcription factors.

5.
Nat Commun ; 15(1): 4023, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38740816

RÉSUMÉ

Abscission is the final stage of cytokinesis, which cleaves the intercellular bridge (ICB) connecting two daughter cells. Abscission requires tight control of the recruitment and polymerization of the Endosomal Protein Complex Required for Transport-III (ESCRT-III) components. We explore the role of post-translational modifications in regulating ESCRT dynamics. We discover that SMYD2 methylates the lysine 6 residue of human CHMP2B, a key ESCRT-III component, at the ICB, impacting the dynamic relocation of CHMP2B to sites of abscission. SMYD2 loss-of-function (genetically or pharmacologically) causes CHMP2B hypomethylation, delayed CHMP2B polymerization and delayed abscission. This is phenocopied by CHMP2B lysine 6 mutants that cannot be methylated. Conversely, SMYD2 gain-of-function causes CHMP2B hypermethylation and accelerated abscission, specifically in cells undergoing cytokinetic challenges, thereby bypassing the abscission checkpoint. Additional experiments highlight the importance of CHMP2B methylation beyond cytokinesis, namely during ESCRT-III-mediated HIV-1 budding. We propose that lysine methylation signaling fine-tunes the ESCRT-III machinery to regulate the timing of cytokinetic abscission and other ESCRT-III dependent functions.


Sujet(s)
Cytocinèse , Complexes de tri endosomique requis pour le transport , Histone-lysine N-methyltransferase , Humains , Complexes de tri endosomique requis pour le transport/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Cellules HeLa , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Lysine/métabolisme , Méthylation , Maturation post-traductionnelle des protéines
6.
Nat Cancer ; 5(7): 1082-1101, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38816660

RÉSUMÉ

Dose-limiting toxicity poses a major limitation to the clinical utility of targeted cancer therapies, often arising from target engagement in nonmalignant tissues. This obstacle can be minimized by targeting cancer dependencies driven by proteins with tissue-restricted and/or tumor-restricted expression. In line with another recent report, we show here that, in acute myeloid leukemia (AML), suppression of the myeloid-restricted PIK3CG/p110γ-PIK3R5/p101 axis inhibits protein kinase B/Akt signaling and compromises AML cell fitness. Furthermore, silencing the genes encoding PIK3CG/p110γ or PIK3R5/p101 sensitizes AML cells to established AML therapies. Importantly, we find that existing small-molecule inhibitors against PIK3CG are insufficient to achieve a sustained long-term antileukemic effect. To address this concern, we developed a proteolysis-targeting chimera (PROTAC) heterobifunctional molecule that specifically degrades PIK3CG and potently suppresses AML progression alone and in combination with venetoclax in human AML cell lines, primary samples from patients with AML and syngeneic mouse models.


Sujet(s)
Phosphatidylinositol 3-kinases de classe Ib , Leucémie aigüe myéloïde , Protéines proto-oncogènes c-akt , Transduction du signal , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/génétique , Humains , Animaux , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Phosphatidylinositol 3-kinases de classe Ib/génétique , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique , Protéolyse/effets des médicaments et des substances chimiques , Femelle , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/usage thérapeutique
7.
Sci Rep ; 14(1): 11242, 2024 05 16.
Article de Anglais | MEDLINE | ID: mdl-38755230

RÉSUMÉ

The interaction of Plasmodium falciparum-infected red blood cells (iRBCs) with the vascular endothelium plays a crucial role in malaria pathology and disease. KAHRP is an exported P. falciparum protein involved in iRBC remodelling, which is essential for the formation of protrusions or "knobs" on the iRBC surface. These knobs and the proteins that are concentrated within them allow the parasites to escape the immune response and host spleen clearance by mediating cytoadherence of the iRBC to the endothelial wall, but this also slows down blood circulation, leading in some cases to severe cerebral and placental complications. In this work, we have applied genetic and biochemical tools to identify proteins that interact with P. falciparum KAHRP using enhanced ascorbate peroxidase 2 (APEX2) proximity-dependent biotinylation and label-free shotgun proteomics. A total of 30 potential KAHRP-interacting candidates were identified, based on the assigned fragmented biotinylated ions. Several identified proteins have been previously reported to be part of the Maurer's clefts and knobs, where KAHRP resides. This study may contribute to a broader understanding of P. falciparum protein trafficking and knob architecture and shows for the first time the feasibility of using APEX2-proximity labelling in iRBCs.


Sujet(s)
Érythrocytes , Plasmodium falciparum , Protéomique , Protéines de protozoaire , Érythrocytes/parasitologie , Érythrocytes/métabolisme , Plasmodium falciparum/métabolisme , Protéines de protozoaire/métabolisme , Humains , Protéomique/méthodes , Paludisme à Plasmodium falciparum/parasitologie , Paludisme à Plasmodium falciparum/métabolisme , DNA-(apurinic or apyrimidinic site) lyase/métabolisme , Ascorbate peroxidases/métabolisme , Liaison aux protéines , Biotinylation , Endonucleases , Peptides , Protéines , Enzymes multifonctionnelles
8.
Nat Commun ; 15(1): 3602, 2024 Apr 29.
Article de Anglais | MEDLINE | ID: mdl-38684700

RÉSUMÉ

Glioblastoma (GBM) is a highly lethal type of cancer. GBM recurrence following chemoradiation is typically attributed to the regrowth of invasive and resistant cells. Therefore, there is a pressing need to gain a deeper understanding of the mechanisms underlying GBM resistance to chemoradiation and its ability to infiltrate. Using a combination of transcriptomic, proteomic, and phosphoproteomic analyses, longitudinal imaging, organotypic cultures, functional assays, animal studies, and clinical data analyses, we demonstrate that chemoradiation and brain vasculature induce cell transition to a functional state named VC-Resist (vessel co-opting and resistant cell state). This cell state is midway along the transcriptomic axis between proneural and mesenchymal GBM cells and is closer to the AC/MES1-like state. VC-Resist GBM cells are highly vessel co-opting, allowing significant infiltration into the surrounding brain tissue and homing to the perivascular niche, which in turn induces even more VC-Resist transition. The molecular and functional characteristics of this FGFR1-YAP1-dependent GBM cell state, including resistance to DNA damage, enrichment in the G2M phase, and induction of senescence/stemness pathways, contribute to its enhanced resistance to chemoradiation. These findings demonstrate how vessel co-option, perivascular niche, and GBM cell plasticity jointly drive resistance to therapy during GBM recurrence.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Humains , Animaux , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/traitement médicamenteux , Tumeurs du cerveau/génétique , Lignée cellulaire tumorale , Souris , Chimioradiothérapie/méthodes , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Radiotolérance , Protéines de signalisation YAP/métabolisme , Encéphale/métabolisme , Encéphale/anatomopathologie , Protéomique
9.
J Med Chem ; 67(8): 6189-6206, 2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38577779

RÉSUMÉ

Identification of intracellular targets of anticancer drug candidates provides key information on their mechanism of action. Exploiting the ability of the anticancer (C∧N)-chelated half-sandwich iridium(III) complexes to covalently bind proteins, click chemistry with a bioorthogonal azido probe was used to localize a phenyloxazoline-chelated iridium complex within cells and profile its interactome at the proteome-wide scale. Proteins involved in protein folding and actin cytoskeleton regulation were identified as high-affinity targets. Upon iridium complex treatment, the folding activity of Heat Shock Protein HSP90 was inhibited in vitro and major cytoskeleton disorganization was observed. A wide array of imaging and biochemical methods validated selected targets and provided a multiscale overview of the effects of this complex on live human cells. We demonstrate that it behaves as a dual agent, inducing both electrophilic and oxidative stresses in cells that account for its cytotoxicity. The proposed methodological workflow can open innovative avenues in metallodrug discovery.


Sujet(s)
Antinéoplasiques , Complexes de coordination , Iridium , Stress oxydatif , Humains , Iridium/composition chimique , Iridium/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Complexes de coordination/pharmacologie , Complexes de coordination/composition chimique , Complexes de coordination/synthèse chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Protéines du choc thermique HSP90/métabolisme , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Protéines du choc thermique HSP90/composition chimique , Chimie click
11.
Sci Rep ; 14(1): 6777, 2024 03 21.
Article de Anglais | MEDLINE | ID: mdl-38514682

RÉSUMÉ

Extracellular matrix (ECM) is an intricate structure providing the microenvironment niche that influences stem cell differentiation. This study aimed to investigate the efficacy of decellularized ECM derived from human dental pulp stem cells (dECM_DPSCs) and gingival-derived mesenchymal stem cells (dECM_GSCs) as an inductive scaffold for osteogenic differentiation of GSCs. The proteomic analysis demonstrated that common and signature matrisome proteins from dECM_DPSCs and dECM_GSCs were related to osteogenesis/osteogenic differentiation. RNA sequencing data from GSCs reseeded on dECM_DPSCs revealed that dECM_DPSCs upregulated genes related to the Hippo and Wnt signaling pathways in GSCs. In the inhibitor experiments, results revealed that dECM_DPSCs superiorly promoted GSCs osteogenic differentiation, mainly mediated through Hippo and Wnt signaling. The present study emphasizes the promising translational application of dECM_DPSCs as a bio-scaffold rich in favorable regenerative microenvironment for tissue engineering.


Sujet(s)
Ostéogenèse , Voie de signalisation Wnt , Humains , Ostéogenèse/génétique , Protéomique , Pulpe dentaire , Matrice extracellulaire/métabolisme , Différenciation cellulaire , Cellules souches/métabolisme , Prolifération cellulaire , Cellules cultivées
12.
J Cell Biol ; 223(3)2024 03 04.
Article de Anglais | MEDLINE | ID: mdl-38386112

RÉSUMÉ

The small G-protein CDC42 is an evolutionary conserved polarity protein and a key regulator of polarized cell functions, including directed cell migration. In vertebrates, alternative splicing gives rise to two CDC42 proteins: the ubiquitously expressed isoform (CDC42u) and the brain isoform (CDC42b), which only differ in their carboxy-terminal sequence, including the CAAX motif essential for their association with membranes. We show that these divergent sequences do not directly affect the range of CDC42's potential binding partners but indirectly influence CDC42-driven signaling by controlling the subcellular localization of the two isoforms. In astrocytes and neural precursors, which naturally express both variants, CDC42u associates with the leading-edge plasma membrane of migrating cells, where it recruits the Par6-PKCζ complex to fulfill its polarity function. In contrast, CDC42b mainly localizes to intracellular membrane compartments, where it regulates N-WASP-mediated endocytosis. Both CDC42 isoforms contribute their specific functions to promote the chemotaxis of neural precursors, demonstrating that their expression pattern is decisive for tissue-specific cell behavior.


Sujet(s)
Épissage alternatif , Astrocytes , Mouvement cellulaire , Protéine G cdc42 , Animaux , Astrocytes/cytologie , Isoformes de protéines/génétique , Rats , Protéine G cdc42/génétique , Membrane cellulaire
14.
Eur Urol ; 85(5): 483-494, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-37380559

RÉSUMÉ

BACKGROUND: Molecular understanding of muscle-invasive (MIBC) and non-muscle-invasive (NMIBC) bladder cancer is currently based primarily on transcriptomic and genomic analyses. OBJECTIVE: To conduct proteogenomic analyses to gain insights into bladder cancer (BC) heterogeneity and identify underlying processes specific to tumor subgroups and therapeutic outcomes. DESIGN, SETTING, AND PARTICIPANTS: Proteomic data were obtained for 40 MIBC and 23 NMIBC cases for which transcriptomic and genomic data were already available. Four BC-derived cell lines harboring FGFR3 alterations were tested with interventions. INTERVENTION: Recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), second mitochondrial-derived activator of caspases mimetic (birinapant), pan-FGFR inhibitor (erdafitinib), and FGFR3 knockdown. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Proteomic groups from unsupervised analyses (uPGs) were characterized using clinicopathological, proteomic, genomic, transcriptomic, and pathway enrichment analyses. Additional enrichment analyses were performed for FGFR3-mutated tumors. Treatment effects on cell viability for FGFR3-altered cell lines were evaluated. Synergistic treatment effects were evaluated using the zero interaction potency model. RESULTS AND LIMITATIONS: Five uPGs, covering both NMIBC and MIBC, were identified and bore coarse-grained similarity to transcriptomic subtypes underlying common features of these different entities; uPG-E was associated with the Ta pathway and enriched in FGFR3 mutations. Our analyses also highlighted enrichment of proteins involved in apoptosis in FGFR3-mutated tumors, not captured through transcriptomics. Genetic and pharmacological inhibition demonstrated that FGFR3 activation regulates TRAIL receptor expression and sensitizes cells to TRAIL-mediated apoptosis, further increased by combination with birinapant. CONCLUSIONS: This proteogenomic study provides a comprehensive resource for investigating NMIBC and MIBC heterogeneity and highlights the potential of TRAIL-induced apoptosis as a treatment option for FGFR3-mutated bladder tumors, warranting a clinical investigation. PATIENT SUMMARY: We integrated proteomics, genomics, and transcriptomics to refine molecular classification of bladder cancer, which, combined with clinical and pathological classification, should lead to more appropriate management of patients. Moreover, we identified new biological processes altered in FGFR3-mutated tumors and showed that inducing apoptosis represents a new potential therapeutic option.


Sujet(s)
Tumeurs de la vessie n'infiltrant pas le muscle , Protéogénomique , Tumeurs de la vessie urinaire , Humains , Protéomique , Ligands , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/anatomopathologie , Apoptose , Facteur de nécrose tumorale alpha , Ligand TRAIL/génétique , Ligand TRAIL/pharmacologie , Récepteur de type 3 des facteurs de croissance fibroblastique/génétique
15.
EMBO J ; 42(24): e113590, 2023 Dec 11.
Article de Anglais | MEDLINE | ID: mdl-38073509

RÉSUMÉ

Cells secrete extracellular vesicles (EVs) and non-vesicular extracellular (nano)particles (NVEPs or ENPs) that may play a role in intercellular communication. Tumor-derived EVs have been proposed to induce immune priming of antigen presenting cells or to be immuno-suppressive agents. We suspect that such disparate functions are due to variable compositions in EV subtypes and ENPs. We aimed to characterize the array of secreted EVs and ENPs of murine tumor cell lines. Unexpectedly, we identified virus-like particles (VLPs) from endogenous murine leukemia virus in preparations of EVs produced by many tumor cells. We established a protocol to separate small EVs from VLPs and ENPs. We compared their protein composition and analyzed their functional interaction with target dendritic cells. ENPs were poorly captured and did not affect dendritic cells. Small EVs specifically induced dendritic cell death. A mixed large/dense EV/VLP preparation was most efficient to induce dendritic cell maturation and antigen presentation. Our results call for systematic re-evaluation of the respective proportions and functions of non-viral EVs and VLPs produced by murine tumors and their contribution to tumor progression.


Sujet(s)
Rétrovirus endogènes , Vésicules extracellulaires , Tumeurs , Animaux , Souris , Vésicules extracellulaires/métabolisme , Lignée cellulaire tumorale , Différenciation cellulaire , Cellules dendritiques , Tumeurs/métabolisme
17.
Mol Cell ; 83(20): 3659-3668.e10, 2023 10 19.
Article de Anglais | MEDLINE | ID: mdl-37832547

RÉSUMÉ

The integrity of the nuclear envelope (NE) is essential for maintaining the structural stability of the nucleus. Rupture of the NE has been frequently observed in cancer cells, especially in the context of mechanical challenges, such as physical confinement and migration. However, spontaneous NE rupture events, without any obvious physical challenges to the cell, have also been described. The molecular mechanism(s) of these spontaneous NE rupture events remain to be explored. Here, we show that DNA damage and subsequent ATR activation leads to NE rupture. Upon DNA damage, lamin A/C is phosphorylated in an ATR-dependent manner, leading to changes in lamina assembly and, ultimately, NE rupture. In addition, we show that cancer cells with intrinsic DNA repair defects undergo frequent events of DNA-damage-induced NE rupture, which renders them extremely sensitive to further NE perturbations. Exploiting this NE vulnerability could provide a new angle to complement traditional, DNA-damage-based chemotherapy.


Sujet(s)
Lamine A , Enveloppe nucléaire , Enveloppe nucléaire/métabolisme , Lamine A/génétique , Lamine A/métabolisme , Phosphorylation , Altération de l'ADN , ADN/métabolisme , Noyau de la cellule/métabolisme
18.
Nature ; 621(7978): 415-422, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-37674080

RÉSUMÉ

DNA double-strand breaks (DSBs) are deleterious lesions that challenge genome integrity. To mitigate this threat, human cells rely on the activity of multiple DNA repair machineries that are tightly regulated throughout the cell cycle1. In interphase, DSBs are mainly repaired by non-homologous end joining and homologous recombination2. However, these pathways are completely inhibited in mitosis3-5, leaving the fate of mitotic DSBs unknown. Here we show that DNA polymerase theta6 (Polθ) repairs mitotic DSBs and thereby maintains genome integrity. In contrast to other DSB repair factors, Polθ function is activated in mitosis upon phosphorylation by Polo-like kinase 1 (PLK1). Phosphorylated Polθ is recruited by a direct interaction with the BRCA1 C-terminal domains of TOPBP1 to mitotic DSBs, where it mediates joining of broken DNA ends. Loss of Polθ leads to defective repair of mitotic DSBs, resulting in a loss of genome integrity. This is further exacerbated in cells that are deficient in homologous recombination, where loss of mitotic DSB repair by Polθ results in cell death. Our results identify mitotic DSB repair as the underlying cause of synthetic lethality between Polθ and homologous recombination. Together, our findings reveal the critical importance of mitotic DSB repair in the maintenance of genome integrity.


Sujet(s)
Cassures double-brin de l'ADN , Réparation de l'ADN , DNA-directed DNA polymerase , Mitose , Protein-Serine-Threonine Kinases , Humains , Protéine BRCA1/métabolisme , Protéines du cycle cellulaire/métabolisme , Mort cellulaire/génétique , DNA-directed DNA polymerase/composition chimique , DNA-directed DNA polymerase/métabolisme , Recombinaison homologue/génétique , Phosphorylation , Protein-Serine-Threonine Kinases/métabolisme , Mutations synthétiques létales , ,
19.
Sci Adv ; 9(37): eadd9084, 2023 09 15.
Article de Anglais | MEDLINE | ID: mdl-37703363

RÉSUMÉ

The mechanistic target of rapamycin complex 1 (mTORC1) is part of the amino acid sensing machinery that becomes activated on the endolysosomal surface in response to nutrient cues. Branched actin generated by WASH and Arp2/3 complexes defines endolysosomal microdomains. Here, we find mTORC1 components in close proximity to endolysosomal actin microdomains. We investigated for interactors of the mTORC1 lysosomal tether, RAGC, by proteomics and identified multiple actin filament capping proteins and their modulators. Perturbation of RAGC function affected the size of endolysosomal actin, consistent with a regulation of actin filament capping by RAGC. Reciprocally, the pharmacological inhibition of actin polymerization or alteration of endolysosomal actin obtained upon silencing of WASH or Arp2/3 complexes impaired mTORC1 activity. Mechanistically, we show that actin is required for proper association of RAGC and mTOR with endolysosomes. This study reveals an unprecedented interplay between actin and mTORC1 signaling on the endolysosomal system.


Sujet(s)
Actines , Transduction du signal , Complexe-1 cible mécanistique de la rapamycine , Cytosquelette d'actine , Lysosomes
20.
Sci Rep ; 13(1): 14960, 2023 09 11.
Article de Anglais | MEDLINE | ID: mdl-37696912

RÉSUMÉ

In this work, we investigated the oncogenic role of Streptococcus gallolyticus subsp. gallolyticus (SGG), a gut bacterium associated with colorectal cancer (CRC). We showed that SGG UCN34 accelerates colon tumor development in a chemically induced CRC murine model. Full proteome and phosphoproteome analysis of murine colons chronically colonized by SGG UCN34 revealed that 164 proteins and 725 phosphorylation sites were differentially regulated. Ingenuity Pathway Analysis (IPA) indicates a pro-tumoral shift specifically induced by SGG UCN34, as ~ 90% of proteins and phosphoproteins identified were associated with digestive cancer. Comprehensive analysis of the altered phosphoproteins using ROMA software revealed up-regulation of several cancer hallmark pathways such as MAPK, mTOR and integrin/ILK/actin, affecting epithelial and stromal colonic cells. Importantly, an independent analysis of protein arrays of human colon tumors colonized with SGG showed up-regulation of PI3K/Akt/mTOR and MAPK pathways, providing clinical relevance to our findings. To test SGG's capacity to induce pre-cancerous transformation of the murine colonic epithelium, we grew ex vivo organoids which revealed unusual structures with compact morphology. Taken together, our results demonstrate the oncogenic role of SGG UCN34 in a murine model of CRC associated with activation of multiple cancer-related signaling pathways.


Sujet(s)
Tumeurs du côlon , Streptococcus gallolyticus sous-espèce gallolyticus , Humains , Animaux , Souris , Modèles animaux de maladie humaine , Phosphatidylinositol 3-kinases , Protéomique , Sérine-thréonine kinases TOR , Phosphoprotéines , Protéome , Transduction du signal
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE