Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 468
Filtrer
1.
Cell Death Dis ; 15(8): 622, 2024 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-39187490

RÉSUMÉ

GC (Gastric cancer) is one of the most common malignant tumours, with over 95% of gastric cancer patients being adenocarcinoma and most gastric cancer patients having no apparent symptoms in the early stages. Finding biomarkers for early screening of gastric cancer and exploring new targets for gastric cancer treatment are urgent problems to be solved in the treatment of gastric cancer, with significant clinical outcomes for the survival rate of gastric cancer patients. The AAA+ family ATPase thyroid hormone receptor-interacting protein 13 (TRIP13) has been reported to play an essential role in developing various tumours. However, the biological function and molecular mechanism of TRIP13 in gastric cancer remain unclear. This study confirms that TRIP13 is highly expressed in gastric cancer tissue samples and that TRIP13 participates in the proliferation, migration, invasion in vitro, and tumourigenesis and metastasis in vivo of gastric cancer cells. Mechanistically, this study confirms that TRIP13 directly interacts with DDX21 and stabilises its expression by restraining its ubiquitination degradation, thereby promoting gastric cancer progression. Additionally, histone deacetylase 1 (HDAC1) is an upstream factor of TRIP13, which could target the TRIP13 promoter region to promote the proliferation, migration, and invasion of gastric cancer cells. These results indicate that TRIP13 serve is a promising biomarker for the treating of gastric cancer patients, and the HDAC1-TRIP13/DDX21 axis might provide a solid theoretical basis for clinical treatment of gastric cancer patients.


Sujet(s)
ATPases associated with diverse cellular activities , Mouvement cellulaire , Prolifération cellulaire , DEAD-box RNA helicases , Évolution de la maladie , Régulation de l'expression des gènes tumoraux , Tumeurs de l'estomac , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , DEAD-box RNA helicases/métabolisme , DEAD-box RNA helicases/génétique , Histone Deacetylase 1/métabolisme , Histone Deacetylase 1/génétique , Souris de lignée BALB C , Souris nude , Invasion tumorale , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Ubiquitination
2.
Proc Natl Acad Sci U S A ; 121(34): e2315759121, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39145935

RÉSUMÉ

Ubiquitination status of proliferating cell nuclear antigen (PCNA) is crucial for regulating DNA lesion bypass. After the resolution of fork stalling, PCNA is subsequently deubiquitinated, but the underlying mechanism remains undefined. We found that the N-terminal domain of ATAD5 (ATAD5-N), the largest subunit of the PCNA-unloading complex, functions as a scaffold for Ub-PCNA deubiquitination. ATAD5 recognizes DNA-loaded Ub-PCNA through distinct DNA-binding and PCNA-binding motifs. Furthermore, ATAD5 forms a heterotrimeric complex with UAF1-USP1 deubiquitinase, facilitating the deubiquitination of DNA-loaded Ub-PCNA. ATAD5 also enhances the Ub-PCNA deubiquitination by USP7 and USP11 through specific interactions. ATAD5 promotes the distinct deubiquitination process of UAF1-USP1, USP7, and USP11 for poly-Ub-PCNA. Additionally, ATAD5 mutants deficient in UAF1-binding had increased sensitivity to DNA-damaging agents. Our results ultimately reveal that ATAD5 and USPs cooperate to efficiently deubiquitinate Ub-PCNA prior to its release from the DNA in order to safely deactivate the DNA repair process.


Sujet(s)
ATPases associated with diverse cellular activities , Protéines de liaison à l'ADN , Antigène nucléaire de prolifération cellulaire , Ubiquitin thiolesterase , Ubiquitin-specific peptidase 7 , Ubiquitination , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Antigène nucléaire de prolifération cellulaire/métabolisme , Antigène nucléaire de prolifération cellulaire/génétique , Humains , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Ubiquitin thiolesterase/métabolisme , Ubiquitin thiolesterase/génétique , Ubiquitin-specific peptidase 7/métabolisme , Ubiquitin-specific peptidase 7/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Thiolester hydrolases/métabolisme , Thiolester hydrolases/génétique , Ubiquitine/métabolisme , Altération de l'ADN , Liaison aux protéines , Ubiquitin-specific proteases
3.
Nat Commun ; 15(1): 7092, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39154037

RÉSUMÉ

Mammalian TIP60 is a multi-functional enzyme with histone acetylation and histone dimer exchange activities. It plays roles in diverse cellular processes including transcription, DNA repair, cell cycle control, and embryonic development. Here we report the cryo-electron microscopy structures of the human TIP60 complex with the core subcomplex and TRRAP module refined to 3.2-Å resolution. The structures show that EP400 acts as a backbone integrating the motor module, the ARP module, and the TRRAP module. The RUVBL1-RUVBL2 hexamer serves as a rigid core for the assembly of EP400 ATPase and YL1 in the motor module. In the ARP module, an ACTL6A-ACTB heterodimer and an extra ACTL6A make hydrophobic contacts with EP400 HSA helix, buttressed by network interactions among DMAP1, EPC1, and EP400. The ARP module stably associates with the motor module but is flexibly tethered to the TRRAP module, exhibiting a unique feature of human TIP60. The architecture of the nucleosome-bound human TIP60 reveals an unengaged nucleosome that is located between the core subcomplex and the TRRAP module. Our work illustrates the molecular architecture of human TIP60 and provides architectural insights into how this complex is bound by the nucleosome.


Sujet(s)
Cryomicroscopie électronique , Lysine acetyltransferase 5 , Humains , Lysine acetyltransferase 5/métabolisme , Lysine acetyltransferase 5/composition chimique , Lysine acetyltransferase 5/génétique , Nucléosomes/métabolisme , Nucléosomes/ultrastructure , Nucléosomes/composition chimique , Helicase/métabolisme , Helicase/composition chimique , Modèles moléculaires , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/composition chimique , ATPases associated with diverse cellular activities/génétique , Protéines de transport/métabolisme , Protéines de transport/composition chimique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/composition chimique , Liaison aux protéines , Multimérisation de protéines , Protéines contenant un bromodomaine , Protéines adaptatrices de la transduction du signal
4.
Science ; 385(6712): eadp7114, 2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39116259

RÉSUMÉ

Endoplasmic reticulum (ER) stress induces the repression of protein synthesis throughout the cell. Attempts to understand how localized stress leads to widespread repression have been limited by difficulties in resolving translation rates at the subcellular level. Here, using live-cell imaging of reporter mRNA translation, we unexpectedly found that during ER stress, active translation at mitochondria was significantly protected. The mitochondrial protein ATPase family AAA domain-containing protein 3A (ATAD3A) interacted with protein kinase RNA-like endoplasmic reticulum kinase (PERK) and mediated this effect on localized translation by competing for binding with PERK's target, eukaryotic initiation factor 2 (eIF2). PERK-ATAD3A interactions increased during ER stress, forming mitochondria-ER contact sites. Furthermore, ATAD3A binding attenuated local PERK signaling and rescued the expression of some mitochondrial proteins. Thus, PERK-ATAD3A interactions can control translational repression at a subcellular level, mitigating the impact of ER stress on the cell.


Sujet(s)
ATPases associated with diverse cellular activities , Stress du réticulum endoplasmique , Facteur-2 d'initiation eucaryote , Protéines membranaires , Protéines mitochondriales , Biosynthèse des protéines , eIF-2 Kinase , Humains , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , eIF-2 Kinase/métabolisme , Réticulum endoplasmique/métabolisme , Facteur-2 d'initiation eucaryote/métabolisme , Cellules HEK293 , Cellules HeLa , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Liaison aux protéines , ARN messager/métabolisme , ARN messager/génétique , Transduction du signal , Protéines membranaires/génétique , Protéines membranaires/métabolisme
5.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39201707

RÉSUMÉ

Multiple myeloma is the second most hematological cancer. RUVBL1 and RUVBL2 form a subcomplex of many chromatin remodeling complexes implicated in cancer progression. As an inhibitor specific to the RUVBL1/2 complex, CB-6644 exhibits remarkable anti-tumor activity in xenograft models of Burkitt's lymphoma and multiple myeloma (MM). In this work, we defined transcriptional signatures corresponding to CB-6644 treatment in MM cells and determined underlying epigenetic changes in terms of chromatin accessibility. CB-6644 upregulated biological processes related to interferon response and downregulated those linked to cell proliferation in MM cells. Transcriptional regulator inference identified E2Fs as regulators for downregulated genes and MED1 and MYC as regulators for upregulated genes. CB-6644-induced changes in chromatin accessibility occurred mostly in non-promoter regions. Footprinting analysis identified transcription factors implied in modulating chromatin accessibility in response to CB-6644 treatment, including ATF4/CEBP and IRF4. Lastly, integrative analysis of transcription responses to various chemical compounds of the molecular signature genes from public gene expression data identified CB-5083, a p97 inhibitor, as a synergistic candidate with CB-6644 in MM cells, but experimental validation refuted this hypothesis.


Sujet(s)
ATPases associated with diverse cellular activities , Helicase , Régulation de l'expression des gènes tumoraux , Myélome multiple , Humains , Myélome multiple/génétique , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Helicase/génétique , Helicase/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/antagonistes et inhibiteurs , Protéines de transport/génétique , Protéines de transport/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Assemblage et désassemblage de la chromatine/effets des médicaments et des substances chimiques , Épigenèse génétique/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique
6.
Gene ; 928: 148767, 2024 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-39013483

RÉSUMÉ

BACKGROUND: Zellweger Syndrome (ZS), or cerebrohepatorenal syndrome, is a rare disorder due to PEX gene mutations affecting peroxisome function. While PEX6 coding mutations are known to cause ZS, the impact of noncoding mutations is less clear. METHODS: A Chinese neonate and his family were subjected to whole exome sequencing (WES) and bioinformatics to assess variant pathogenicity. A minigene assay was also performed for detailed splicing variant analysis. RESULTS: WES identified compound heterozygous PEX6 variants: c.315G>A (p. Trp105Ter) and c.2095-3 T>G. Minigene assays indicated that the latter variant led to abnormal mRNA splicing and the loss of exon 11 in PEX6 expression, potentially causing nonsense-mediated mRNA decay (NMD) or truncated protein structure. CONCLUSION: The study suggests that PEX6: c.2095-3 T>G might be a genetic contributor to the patient's condition, broadening the known mutation spectrum of PEX6. These insights lay groundwork for potential gene therapy for such variants.


Sujet(s)
ATPases associated with diverse cellular activities , Introns , Syndrome de Zellweger , Humains , Syndrome de Zellweger/génétique , Nouveau-né , Mâle , ATPases associated with diverse cellular activities/génétique , , Mutation , Épissage des ARN , Protéines membranaires/génétique , Pedigree , Asiatiques/génétique , Péroxines/génétique , Chine , Femelle , Peuples d'Asie de l'Est
7.
Sci Rep ; 14(1): 15740, 2024 07 08.
Article de Anglais | MEDLINE | ID: mdl-38977862

RÉSUMÉ

Genome replication is frequently impeded by highly stable DNA secondary structures, including G-quadruplex (G4) DNA, that can hinder the progression of the replication fork. Human WRNIP1 (Werner helicase Interacting Protein 1) associates with various components of the replication machinery and plays a crucial role in genome maintenance processes. However, its detailed function is still not fully understood. Here we show that human WRNIP1 interacts with G4 structures and provide evidence for its contribution to G4 processing. The absence of WRNIP1 results in elevated levels of G4 structures, DNA damage and chromosome aberrations following treatment with PhenDC3, a G4-stabilizing ligand. Additionally, we establish a functional and physical relationship between WRNIP1 and the PIF1 helicase in G4 processing. In summary, our results suggest that WRNIP1 aids genome replication and maintenance by regulating G4 processing and this activity relies on Pif1 DNA helicase.


Sujet(s)
Helicase , Réplication de l'ADN , G-quadruplexes , Humains , Helicase/métabolisme , Altération de l'ADN , Aberrations des chromosomes , Protéines de transport/métabolisme , Protéines de transport/génétique , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique
8.
Cell Rep ; 43(8): 114473, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39024102

RÉSUMÉ

Mitochondria require the constant import of nuclear-encoded proteins for proper functioning. Impaired protein import not only depletes mitochondria of essential factors but also leads to toxic accumulation of un-imported proteins outside the organelle. Here, we investigate the consequences of impaired mitochondrial protein import in human cells. We demonstrate that un-imported proteins can clog the mitochondrial translocase of the outer membrane (TOM). ATAD1, a mitochondrial ATPase, removes clogged proteins from TOM to clear the entry gate into the mitochondria. ATAD1 interacts with both TOM and stalled proteins, and its knockout results in extensive accumulation of mitochondrial precursors as well as decreased protein import. Increased ATAD1 expression contributes to improved fitness of cells with inefficient mitochondrial protein import. Overall, we demonstrate the importance of the ATAD1 quality control pathway in surveilling protein import and its contribution to cellular health.


Sujet(s)
ATPases associated with diverse cellular activities , Mitochondries , Protéines mitochondriales , Transport des protéines , Humains , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Mitochondries/métabolisme , Cellules HeLa , Protéines de transport de la membrane mitochondriale/métabolisme , Protéines de transport de la membrane mitochondriale/génétique , Protéines du complexe d'import des protéines précurseurs mitochondriales , Adenosine triphosphatases/métabolisme , Cellules HEK293 , Membranes mitochondriales/métabolisme
9.
PLoS Pathog ; 20(6): e1012300, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38900818

RÉSUMÉ

The AAA-type ATPase VPS4 is recruited by proteins of the endosomal sorting complex required for transport III (ESCRT-III) to catalyse membrane constriction and membrane fission. VPS4A accumulates at the cytoplasmic viral assembly complex (cVAC) of cells infected with human cytomegalovirus (HCMV), the site where nascent virus particles obtain their membrane envelope. Here we show that VPS4A is recruited to the cVAC via interaction with pUL71. Sequence analysis, deep-learning structure prediction, molecular dynamics and mutagenic analysis identify a short peptide motif in the C-terminal region of pUL71 that is necessary and sufficient for the interaction with VPS4A. This motif is predicted to bind the same groove of the N-terminal VPS4A Microtubule-Interacting and Trafficking (MIT) domain as the Type 2 MIT-Interacting Motif (MIM2) of cellular ESCRT-III components, and this viral MIM2-like motif (vMIM2) is conserved across ß-herpesvirus pUL71 homologues. However, recruitment of VPS4A by pUL71 is dispensable for HCMV morphogenesis or replication and the function of the conserved vMIM2 during infection remains enigmatic. VPS4-recruitment via a vMIM2 represents a previously unknown mechanism of molecular mimicry in viruses, extending previous observations that herpesviruses encode proteins with structural and functional homology to cellular ESCRT-III components.


Sujet(s)
Cytomegalovirus , Complexes de tri endosomique requis pour le transport , Mimétisme moléculaire , Vacuolar Proton-Translocating ATPases , Assemblage viral , Humains , Complexes de tri endosomique requis pour le transport/métabolisme , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Cytomegalovirus/métabolisme , Cytomegalovirus/génétique , Cytomegalovirus/physiologie , Assemblage viral/physiologie , Infections à cytomégalovirus/virologie , Infections à cytomégalovirus/métabolisme , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Protéines virales/métabolisme , Protéines virales/génétique
10.
Proc Natl Acad Sci U S A ; 121(27): e2314702121, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38916997

RÉSUMÉ

Enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles (cerebral ventriculomegaly), the cardinal feature of congenital hydrocephalus (CH), is increasingly recognized among patients with autism spectrum disorders (ASD). KATNAL2, a member of Katanin family microtubule-severing ATPases, is a known ASD risk gene, but its roles in human brain development remain unclear. Here, we show that nonsense truncation of Katnal2 (Katnal2Δ17) in mice results in classic ciliopathy phenotypes, including impaired spermatogenesis and cerebral ventriculomegaly. In both humans and mice, KATNAL2 is highly expressed in ciliated radial glia of the fetal ventricular-subventricular zone as well as in their postnatal ependymal and neuronal progeny. The ventriculomegaly observed in Katnal2Δ17 mice is associated with disrupted primary cilia and ependymal planar cell polarity that results in impaired cilia-generated CSF flow. Further, prefrontal pyramidal neurons in ventriculomegalic Katnal2Δ17 mice exhibit decreased excitatory drive and reduced high-frequency firing. Consistent with these findings in mice, we identified rare, damaging heterozygous germline variants in KATNAL2 in five unrelated patients with neurosurgically treated CH and comorbid ASD or other neurodevelopmental disorders. Mice engineered with the orthologous ASD-associated KATNAL2 F244L missense variant recapitulated the ventriculomegaly found in human patients. Together, these data suggest KATNAL2 pathogenic variants alter intraventricular CSF homeostasis and parenchymal neuronal connectivity by disrupting microtubule dynamics in fetal radial glia and their postnatal ependymal and neuronal descendants. The results identify a molecular mechanism underlying the development of ventriculomegaly in a genetic subset of patients with ASD and may explain persistence of neurodevelopmental phenotypes in some patients with CH despite neurosurgical CSF shunting.


Sujet(s)
Cils vibratiles , Hydrocéphalie , Microtubules , Animaux , Femelle , Humains , Mâle , Souris , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/métabolisme , Trouble du spectre autistique/génétique , Trouble du spectre autistique/anatomopathologie , Trouble du spectre autistique/métabolisme , Cils vibratiles/métabolisme , Cils vibratiles/anatomopathologie , Épendyme/métabolisme , Épendyme/anatomopathologie , Hydrocéphalie/génétique , Hydrocéphalie/anatomopathologie , Hydrocéphalie/métabolisme , Katanine/métabolisme , Katanine/génétique , Microtubules/métabolisme , Neurones/métabolisme , Cellules pyramidales/métabolisme , Cellules pyramidales/anatomopathologie
11.
Proc Natl Acad Sci U S A ; 121(24): e2404383121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38843184

RÉSUMÉ

Transcription is extremely important for cellular processes but can be hindered by RNA polymerase II (RNAPII) pausing and stalling. Cockayne syndrome protein B (CSB) promotes the progression of paused RNAPII or initiates transcription-coupled nucleotide excision repair (TC-NER) to remove stalled RNAPII. However, the specific mechanism by which CSB initiates TC-NER upon damage remains unclear. In this study, we identified the indispensable role of the ARK2N-CK2 complex in the CSB-mediated initiation of TC-NER. The ARK2N-CK2 complex is recruited to damage sites through CSB and then phosphorylates CSB. Phosphorylation of CSB enhances its binding to stalled RNAPII, prolonging the association of CSB with chromatin and promoting CSA-mediated ubiquitination of stalled RNAPII. Consistent with this finding, Ark2n-/- mice exhibit a phenotype resembling Cockayne syndrome. These findings shed light on the pivotal role of the ARK2N-CK2 complex in governing the fate of RNAPII through CSB, bridging a critical gap necessary for initiating TC-NER.


Sujet(s)
Syndrome de Cockayne , Helicase , Enzymes de réparation de l'ADN , Réparation de l'ADN , Protéines liant le poly-adp-ribose , RNA polymerase II , Enzymes de réparation de l'ADN/métabolisme , Enzymes de réparation de l'ADN/génétique , RNA polymerase II/métabolisme , RNA polymerase II/génétique , Protéines liant le poly-adp-ribose/métabolisme , Protéines liant le poly-adp-ribose/génétique , Humains , Animaux , Souris , Helicase/métabolisme , Helicase/génétique , Syndrome de Cockayne/génétique , Syndrome de Cockayne/métabolisme , Transcription génétique , Phosphorylation , Casein Kinase II/métabolisme , Casein Kinase II/génétique , Souris knockout , Altération de l'ADN , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Chromatine/métabolisme , Ubiquitination , Réparation par excision
12.
Cancer Res ; 84(17): 2856-2872, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-38900944

RÉSUMÉ

The N6-methyladenosine (m6A) RNA-binding protein YTHDF1 is frequently overexpressed in colorectal cancer and drives chemotherapeutic resistance. To systematically identify druggable targets in colorectal cancer with high expression of YTHDF1, this study used a CRISPR/Cas9 screening strategy that revealed RUVBL1 and RUVBL2 as putative targets. RUVBL1/2 were overexpressed in primary colorectal cancer samples and represented independent predictors of poor patient prognosis. Functionally, loss of RUVBL1/2 preferentially impaired the growth of YTHDF1-high colorectal cancer cells, patient-derived primary colorectal cancer organoids, and subcutaneous xenografts. Mechanistically, YTHFD1 and RUVBL1/2 formed a positive feedforward circuit to accelerate oncogenic translation. YTHDF1 bound to m6A-modified RUVBL1/2 mRNA to promote translation initiation and protein expression. Coimmunoprecipitation and mass spectrometry identified that RUVBL1/2 reciprocally interacted with YTHDF1 at 40S translation initiation complexes. Consequently, RUVBL1/2 depletion stalled YTHDF1-driven oncogenic translation and nascent protein biosynthesis, leading to proliferative arrest and apoptosis. Ribosome sequencing revealed that RUVBL1/2 loss impaired the activation of MAPK, RAS, and PI3K-AKT signaling induced by YTHDF1. Finally, the blockade of RUVBL1/2 by the pharmacological inhibitor CB6644 or vesicle-like nanoparticle-encapsulated siRNAs preferentially arrested the growth of YTHDF1-expressing colorectal cancer in vitro and in vivo. Our findings show that RUVBL1/2 are potential prognostic markers and druggable targets that regulate protein translation in YTHDF1-high colorectal cancer. Significance: RUVBL1/2 inhibition is a therapeutic strategy to abrogate YTHDF1-driven oncogenic translation and overcome m6A dysregulation in colorectal cancer.


Sujet(s)
ATPases associated with diverse cellular activities , Adénosine , Tumeurs colorectales , Helicase , Protéines de liaison à l'ARN , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/traitement médicamenteux , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Animaux , Souris , Helicase/génétique , Helicase/métabolisme , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/antagonistes et inhibiteurs , Adénosine/analogues et dérivés , Adénosine/métabolisme , Carcinogenèse/génétique , Protéines de transport/métabolisme , Protéines de transport/génétique , Biosynthèse des protéines , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Prolifération cellulaire , Souris nude , Tests d'activité antitumorale sur modèle de xénogreffe , Pronostic
13.
Adv Biol Regul ; 93: 101041, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38909398

RÉSUMÉ

ATPase family AAA domain containing protein 3, commonly known as ATAD3 is a versatile mitochondrial protein that is involved in a large number of pathways. ATAD3 is a transmembrane protein that spans both the inner mitochondrial membrane and outer mitochondrial membrane. It, therefore, functions as a connecting link between the mitochondrial lumen and endoplasmic reticulum facilitating their cross-talk. ATAD3 contains an N-terminal domain which is amphipathic in nature and is inserted into the membranous space of the mitochondria, while the C-terminal domain is present towards the lumen of the mitochondria and contains the ATPase domain. ATAD3 is known to be involved in mitochondrial biogenesis, cholesterol transport, hormone synthesis, apoptosis and several other pathways. It has also been implicated to be involved in cancer and many neurological disorders making it an interesting target for extensive studies. This review aims to provide an updated comprehensive account of the role of ATAD3 in the mitochondria especially in lipid transport, mitochondrial-endoplasmic reticulum interactions, cancer and inhibition of mitophagy.


Sujet(s)
ATPases associated with diverse cellular activities , Protéines membranaires , Mitochondries , Protéines mitochondriales , Tumeurs , Humains , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Animaux , Mitochondries/métabolisme , Mitochondries/génétique , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Tumeurs/métabolisme , Tumeurs/génétique , Tumeurs/anatomopathologie , Membranes mitochondriales/métabolisme , Réticulum endoplasmique/métabolisme , Mitophagie
14.
Cell Death Dis ; 15(5): 346, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38769124

RÉSUMÉ

Exploring novel diagnostic and therapeutic biomarkers is extremely important for osteosarcoma. YME1 Like 1 ATPase (YME1L), locating in the mitochondrial inner membrane, is key in regulating mitochondrial plasticity and metabolic activity. Its expression and potential functions in osteosarcoma are studied in the present study. We show that YME1L mRNA and protein expression is significantly elevated in osteosarcoma tissues derived from different human patients. Moreover, its expression is upregulated in various primary and immortalized osteosarcoma cells. The Cancer Genome Atlas database results revealed that YME1L overexpression was correlated with poor overall survival and poor disease-specific survival in sarcoma patients. In primary and immortalized osteosarcoma cells, silencing of YME1L through lentiviral shRNA robustly inhibited cell viability, proliferation, and migration. Moreover, cell cycle arrest and apoptosis were detected in YME1L-silenced osteosarcoma cells. YME1L silencing impaired mitochondrial functions in osteosarcoma cells, causing mitochondrial depolarization, oxidative injury, lipid peroxidation and DNA damage as well as mitochondrial respiratory chain complex I activity inhibition and ATP depletion. Contrarily, forced YME1L overexpression exerted pro-cancerous activity and strengthened primary osteosarcoma cell proliferation and migration. YME1L is important for Akt-S6K activation in osteosarcoma cells. Phosphorylation of Akt and S6K was inhibited after YME1L silencing in primary osteosarcoma cells, but was strengthened with YME1L overexpression. Restoring Akt-mTOR activation by S473D constitutively active Akt1 mitigated YME1L shRNA-induced anti-osteosarcoma cell activity. Lastly, intratumoral injection of YME1L shRNA adeno-associated virus inhibited subcutaneous osteosarcoma xenograft growth in nude mice. YME1L depletion, mitochondrial dysfunction, oxidative injury, Akt-S6K inactivation, and apoptosis were detected in YME1L shRNA-treated osteosarcoma xenografts. Together, overexpressed YME1L promotes osteosarcoma cell growth, possibly by maintaining mitochondrial function and Akt-mTOR activation.


Sujet(s)
Tumeurs osseuses , Prolifération cellulaire , Souris nude , Ostéosarcome , Animaux , Femelle , Humains , Mâle , Souris , Apoptose/génétique , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Souris de lignée BALB C , Mitochondries/métabolisme , Protéines mitochondriales/métabolisme , Protéines mitochondriales/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme
15.
Gut ; 73(9): 1509-1528, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-38821858

RÉSUMÉ

OBJECTIVE: The hallmark oncogene MYC drives the progression of most tumours, but direct inhibition of MYC by a small-molecule drug has not reached clinical testing. MYC is a transcription factor that depends on several binding partners to function. We therefore explored the possibility of targeting MYC via its interactome in pancreatic ductal adenocarcinoma (PDAC). DESIGN: To identify the most suitable targets among all MYC binding partners, we constructed a targeted shRNA library and performed screens in cultured PDAC cells and tumours in mice. RESULTS: Unexpectedly, many MYC binding partners were found to be important for cultured PDAC cells but dispensable in vivo. However, some were also essential for tumours in their natural environment and, among these, the ATPases RUVBL1 and RUVBL2 ranked first. Degradation of RUVBL1 by the auxin-degron system led to the arrest of cultured PDAC cells but not untransformed cells and to complete tumour regression in mice, which was preceded by immune cell infiltration. Mechanistically, RUVBL1 was required for MYC to establish oncogenic and immunoevasive gene expression identifying the RUVBL1/2 complex as a druggable vulnerability in MYC-driven cancer. CONCLUSION: One implication of our study is that PDAC cell dependencies are strongly influenced by the environment, so genetic screens should be performed in vitro and in vivo. Moreover, the auxin-degron system can be applied in a PDAC model, allowing target validation in living mice. Finally, by revealing the nuclear functions of the RUVBL1/2 complex, our study presents a pharmaceutical strategy to render pancreatic cancers potentially susceptible to immunotherapy.


Sujet(s)
ATPases associated with diverse cellular activities , Carcinome du canal pancréatique , Helicase , Tumeurs du pancréas , Protéines proto-oncogènes c-myc , Animaux , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Souris , Humains , Helicase/génétique , Helicase/métabolisme , Carcinome du canal pancréatique/génétique , Carcinome du canal pancréatique/anatomopathologie , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Lignée cellulaire tumorale , Protéines de transport/métabolisme , Protéines de transport/génétique
16.
PLoS Biol ; 22(4): e3002327, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38687820

RÉSUMÉ

Mutations in the human AAA-ATPase VPS4 isoform, VPS4A, cause severe neurodevelopmental defects and congenital dyserythropoietic anemia (CDA). VPS4 is a crucial component of the endosomal sorting complex required for transport (ESCRT) system, which drives membrane remodeling in numerous cellular processes, including receptor degradation, cell division, and neural pruning. Notably, while most organisms encode for a single VPS4 gene, human cells have 2 VPS4 paralogs, namely VPS4A and VPS4B, but the functional differences between these paralogs is mostly unknown. Here, we set out to investigate the role of the human VPS4 paralogs in cytokinetic abscission using a series of knockout cell lines. We found that VPS4A and VPS4B hold both overlapping and distinct roles in abscission. VPS4A depletion resulted in a more severe abscission delay than VPS4B and was found to be involved in earlier stages of abscission. Moreover, VPS4A and a monomeric-locked VPS4A mutant bound the abscission checkpoint proteins CHMP4C and ANCHR, while VPS4B did not, indicating a regulatory role for the VPS4A isoform in abscission. Depletion of VTA1, a co-factor of VPS4, disrupted VPS4A-ANCHR interactions and accelerated abscission, suggesting that VTA1 is also involved in the abscission regulation. Our findings reveal a dual role for VPS4A in abscission, one that is canonical and can be compensated by VPS4B, and another that is regulatory and may be delivered by its monomeric form. These observations provide a potential mechanistic explanation for the neurodevelopmental defects and other related disorders reported in VPS4A-mutated patients with a fully functional VPS4B paralog.


Sujet(s)
ATPases associated with diverse cellular activities , Cytocinèse , Complexes de tri endosomique requis pour le transport , Vacuolar Proton-Translocating ATPases , Humains , Complexes de tri endosomique requis pour le transport/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Vacuolar Proton-Translocating ATPases/métabolisme , Vacuolar Proton-Translocating ATPases/génétique , Cellules HeLa , Isoformes de protéines/métabolisme , Isoformes de protéines/génétique
17.
Ophthalmic Genet ; 45(4): 351-362, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38664000

RÉSUMÉ

PURPOSE: This cross-sectional study describes the ophthalmological and general phenotype of 10 patients from six different families with a comparatively mild form of Zellweger spectrum disorder (ZSD), a rare peroxisomal disorder. METHODS: Ophthalmological assessment included best-corrected visual acuity (BCVA), perimetry, microperimetry, ophthalmoscopy, fundus photography, spectral-domain optical coherence tomography (SD-OCT), and fundus autofluorescence (FAF) imaging. Medical records were reviewed for medical history and systemic manifestations of ZSD. RESULTS: Nine patients were homozygous for c.2528 G > A (p.Gly843Asp) variants in PEX1 and one patient was compound heterozygous for c.2528 G>A (p.Gly843Asp) and c.2097_2098insT (p.Ile700TyrfsTer42) in PEX1. Median age was 22.6 years (interquartile range (IQR): 15.9 - 29.9 years) at the most recent examination, with a median symptom duration of 22.1 years. Symptom onset was variable with presentations of hearing loss (n = 7) or nyctalopia/reduced visual acuity (n = 3) at a median age of 6 months (IQR: 1.9-8.3 months). BCVA (median of 0.8 logMAR; IQR: 0.6-0.9 logMAR) remained stable over 10.8 years and all patients were hyperopic. Fundus examination revealed a variable retinitis pigmentosa (RP)-like phenotype with rounded hyperpigmentations as most prominent feature in six out of nine patients. Electroretinography, visual field measurements, and microperimetry further established the RP-like phenotype. Multimodal imaging revealed significant intraretinal fluid cavities on SD-OCT and a remarkable pattern of hyperautofluorescent abnormalities on FAF in all patients. CONCLUSION: This study highlights the ophthalmological phenotype resembling RP with moderate to severe visual impairment in patients with mild ZSD. These findings can aid ophthalmologists in diagnosing, counselling, and managing patients with mild ZSD.


Sujet(s)
Phénotype , Tomographie par cohérence optique , Acuité visuelle , Syndrome de Zellweger , Humains , Mâle , Femelle , Adulte , Adolescent , Acuité visuelle/physiologie , Études transversales , Syndrome de Zellweger/génétique , Syndrome de Zellweger/diagnostic , Syndrome de Zellweger/physiopathologie , Jeune adulte , Péroxines/génétique , Ophtalmoscopie , ATPases associated with diverse cellular activities/génétique , Mutation , Champs visuels/physiologie , Tests du champ visuel , Électrorétinographie , Pedigree , Protéines membranaires
18.
Cell Death Dis ; 15(4): 259, 2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-38609375

RÉSUMÉ

Radiotherapy effectiveness in breast cancer is limited by radioresistance. Nevertheless, the mechanisms behind radioresistance are not yet fully understood. RUVBL1 and RUVBL2, referred to as RUVBL1/2, are crucial AAA+ ATPases that act as co-chaperones and are connected to cancer. Our research revealed that RUVBL1, also known as pontin/TIP49, is excessively expressed in MMTV-PyMT mouse models undergoing radiotherapy, which is considered a murine spontaneous breast-tumor model. Our findings suggest that RUVBL1 enhances DNA damage repair and radioresistance in breast cancer cells both in vitro and in vivo. Mechanistically, we discovered that DTL, also known as CDT2 or DCAF2, which is a substrate adapter protein of CRL4, promotes the ubiquitination of RUVBL1 and facilitates its binding to RUVBL2 and transcription cofactor ß-catenin. This interaction, in turn, attenuates its binding to acetyltransferase Tat-interacting protein 60 (TIP60), a comodulator of nuclear receptors. Subsequently, ubiquitinated RUVBL1 promotes the transcriptional regulation of RUVBL1/2-ß-catenin on genes associated with the non-homologous end-joining (NHEJ) repair pathway. This process also attenuates TIP60-mediated H4K16 acetylation and the homologous recombination (HR) repair process. Expanding upon the prior study's discoveries, we exhibited that the ubiquitination of RUVBL1 by DTL advances the interosculation of RUVBL1/2-ß-catenin. And, it then regulates the transcription of NHEJ repair pathway protein. Resulting in an elevated resistance of breast cancer cells to radiation therapy. From the aforementioned, it is evident that targeting DTL-RUVBL1/2-ß-catenin provides a potential radiosensitization approach when treating breast cancer.


Sujet(s)
Tumeurs mammaires de l'animal , bêta-Caténine , Animaux , Souris , ATPases associated with diverse cellular activities/génétique , bêta-Caténine/génétique , Helicase/génétique , Régulation de l'expression des gènes , Ubiquitine , Ubiquitination , Protéines nucléaires
19.
Cell Cycle ; 23(3): 233-247, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38551450

RÉSUMÉ

Colorectal cancer (CRC) poses a significant challenge in terms of treatment due to the prevalence of radiotherapy resistance. However, the underlying mechanisms responsible for radio-resistance in CRC have not been thoroughly explored. This study aimed to shed light on the role of human coilin interacting nuclear ATPase protein (hCINAP) in radiation-resistant HT-29 and SW480 CRC cells (HT-29-IR and SW480-IR) and investigate its potential implications. Firstly, radiation-resistant CRC cell lines were established by subjecting HT-29 and SW480 cells to sequential radiation exposure. Subsequent analysis revealed a notable increase in hCINAP expression in radiation-resistant CRC cells. To elucidate the functional role of hCINAP in radio-resistance, knockdown experiments were conducted. Remarkably, knockdown of hCINAP resulted in an elevation of reactive oxygen species (ROS) generation upon radiation treatment and subsequent activation of apoptosis mediated by mitochondria. These observations indicate that hCINAP depletion enhances the radiosensitivity of CRC cells. Conversely, when hCINAP was overexpressed, it was found to enhance the radio-resistance of CRC cells. This suggests that elevated hCINAP expression contributes to the development of radio-resistance. Further investigation revealed an interaction between hCINAP and ATPase family AAA domain containing 3A (ATAD3A). Importantly, ATAD3A was identified as an essential factor in hCINAP-mediated radio-resistance. These findings establish the involvement of hCINAP and its interaction with ATAD3A in the regulation of radio-resistance in CRC cells. Overall, the results of this study demonstrate that upregulating hCINAP expression may improve the survival of radiation-exposed CRC cells. Understanding the intricate molecular mechanisms underlying hCINAP function holds promise for potential strategies in targeted radiation therapy for CRC. These findings emphasize the importance of further research to gain a comprehensive understanding of hCINAP's precise molecular mechanisms and explore its potential as a therapeutic target in overcoming radio-resistance in CRC. By unraveling the complexities of hCINAP and its interactions, novel therapeutic approaches may be developed to enhance the efficacy of radiation therapy and improve outcomes for CRC patients.


Sujet(s)
ATPases associated with diverse cellular activities , Apoptose , Tumeurs colorectales , Techniques de knock-down de gènes , Radiotolérance , Espèces réactives de l'oxygène , Humains , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/métabolisme , Tumeurs colorectales/radiothérapie , Radiotolérance/génétique , Apoptose/effets des radiations , Apoptose/génétique , Espèces réactives de l'oxygène/métabolisme , ATPases associated with diverse cellular activities/métabolisme , ATPases associated with diverse cellular activities/génétique , Lignée cellulaire tumorale , Rayonnement ionisant , Mitochondries/métabolisme , Mitochondries/effets des radiations , Cellules HT29
20.
Trends Genet ; 40(6): 526-539, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38485608

RÉSUMÉ

Proliferating cell nuclear antigen (PCNA) is a eukaryotic replicative DNA clamp. Furthermore, DNA-loaded PCNA functions as a molecular hub during DNA replication and repair. PCNA forms a closed homotrimeric ring that encircles the DNA, and association and dissociation of PCNA from DNA are mediated by clamp-loader complexes. PCNA must be actively released from DNA after completion of its function. If it is not released, abnormal accumulation of PCNA on chromatin will interfere with DNA metabolism. ATAD5 containing replication factor C-like complex (RLC) is a PCNA-unloading clamp-loader complex. ATAD5 deficiency causes various DNA replication and repair problems, leading to genome instability. Here, we review recent progress regarding the understanding of the action mechanisms of PCNA unloading complex in DNA replication/repair pathways.


Sujet(s)
Réparation de l'ADN , Réplication de l'ADN , Mammifères , Antigène nucléaire de prolifération cellulaire , Réplication de l'ADN/génétique , Antigène nucléaire de prolifération cellulaire/génétique , Antigène nucléaire de prolifération cellulaire/métabolisme , Réparation de l'ADN/génétique , Animaux , Humains , Mammifères/génétique , Chromatine/génétique , Chromatine/métabolisme , Instabilité du génome/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , ADN/génétique , ADN/métabolisme , ATPases associated with diverse cellular activities/génétique , ATPases associated with diverse cellular activities/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE