Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 18 de 18
Filtrer
1.
Aesthetic Plast Surg ; 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724640

RÉSUMÉ

Lipomas, benign adipose tissue tumors, are a common occurrence but currently, the options for their treatment are limited, with surgical excision being the most frequently used management pathway. This scenario can often lead to unsatisfactory cosmetic results and significant patient discomfort. This paper introduces a novel technique, percutaneous microwave ablation with liposuction, to address these challenges. The innovative procedure aims to enhance patient satisfaction, minimize post-operative discomfort, and improve aesthetic outcomes. The technique involves two key steps: (1) the application of percutaneous microwave ablation to selectively disrupt the lipoma cells, followed by (2) a targeted liposuction procedure to remove the ablated lipoma tissue. Our approach optimizes the removal of the lipoma and preserves the surrounding healthy tissue, reducing the risk of local recurrence and improving the cosmetic result. The use of preoperative ultrasound imaging allows for precise localization and delineation of the lipoma, aiding in the planning and execution of the procedure. This novel approach to lipoma treatment is reliable, associated with minimal morbidity, and consistently yields effective results. Additionally, it provides a new perspective on lipoma management, potentially changing the paradigm of current treatment approaches.Level of Evidence IV This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .

2.
Cancer Res Commun ; 3(6): 1067-1077, 2023 06.
Article de Anglais | MEDLINE | ID: mdl-37377614

RÉSUMÉ

The arginine methyltransferase CARM1 exhibits high expression levels in several human cancers, with the trend also observed in ovarian cancer. However, therapeutic approaches targeting tumors that overexpress CARM1 have not been explored. Cancer cells exploit metabolic reprogramming such as fatty acids for their survival. Here we report that CARM1 promotes monounsaturated fatty acid synthesis and fatty acid reprogramming represents a metabolic vulnerability for CARM1-expressing ovarian cancer. CARM1 promotes the expression of genes encoding rate-limiting enzymes of de novo fatty acid metabolism such as acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FASN). In addition, CARM1 upregulates stearoyl-CoA desaturase 1 (SCD1) that produces monounsaturated fatty acid by desaturation. Thus, CARM1 enhances de novo fatty acids synthesis which was subsequently utilized for synthesis of monounsaturated fatty acids. Consequently, inhibition of SCD1 suppresses the growth of ovarian cancer cells in a CARM1 status-dependent manner, which was rescued by the addition of monounsaturated fatty acids. Consistently, CARM1-expressing cells were more tolerant to the addition of saturated fatty acids. Indeed, SCD1 inhibition demonstrated efficacy against ovarian cancer in both orthotopic xenograft and syngeneic mouse models in a CARM1-dependent manner. In summary, our data show that CARM1 reprograms fatty acid metabolism and targeting SCD1 through pharmacological inhibition can serve as a potent therapeutic approach for CARM1-expressing ovarian cancers. Significance: CARM1 reprograms fatty acid metabolism transcriptionally to support ovarian cancer growth by producing monounsaturated fatty acids, supporting SCD1 inhibition as a rational strategy for treating CARM1-expressing ovarian cancer.


Sujet(s)
Acides gras , Tumeurs de l'ovaire , Animaux , Souris , Humains , Femelle , Acides gras/métabolisme , Acyl-(acyl-carrier-protein)desaturase/génétique , Tumeurs de l'ovaire/génétique , Acides gras monoinsaturés/métabolisme
3.
Cancer Cell ; 41(4): 740-756.e10, 2023 04 10.
Article de Anglais | MEDLINE | ID: mdl-36963401

RÉSUMÉ

ARID1A, encoding a subunit of the SWI/SNF complex, is mutated in ∼50% of clear cell ovarian carcinoma (OCCC) cases. Here we show that inhibition of the mevalonate pathway synergizes with immune checkpoint blockade (ICB) by driving inflammasome-regulated immunomodulating pyroptosis in ARID1A-inactivated OCCCs. SWI/SNF inactivation downregulates the rate-limiting enzymes in the mevalonate pathway such as HMGCR and HMGCS1, which creates a dependence on the residual activity of the pathway in ARID1A-inactivated cells. Inhibitors of the mevalonate pathway such as simvastatin suppresses the growth of ARID1A mutant, but not wild-type, OCCCs. In addition, simvastatin synergizes with anti-PD-L1 antibody in a genetic OCCC mouse model driven by conditional Arid1a inactivation and in a humanized immunocompetent ARID1A mutant patient-derived OCCC mouse model. Our data indicate that inhibition of the mevalonate pathway simultaneously suppresses tumor cell growth and boosts antitumor immunity by promoting pyroptosis, which synergizes with ICB in suppressing ARID1A-mutated cancers.


Sujet(s)
Carcinomes , Tumeurs de l'ovaire , Humains , Femelle , Souris , Animaux , Acide mévalonique , Pyroptose , Protéines nucléaires/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Mutation , Protéines de liaison à l'ADN/génétique , Facteurs de transcription/génétique
4.
Front Surg ; 10: 1283573, 2023.
Article de Anglais | MEDLINE | ID: mdl-38259977

RÉSUMÉ

Background: There has been a substantial increase in incidence of thyroid cancer globally over the past three decades, emphasizing the necessity for efficient surgical management. Surgical intervention requires meticulous lymphatic dissection; however, it is challenging to both accurately identify lymph nodes and preserve the surrounding structures. We investigated the role of carbon nanoparticles in endoscopic thyroid cancer surgery to improve surgical effects and reduce postoperative complications. Methods: Chinese and English literature databases from inception to May 2023 were searched based on inclusion criteria, and data were extracted independently by two investigators. STATA software was used for data analysis. Results: A comprehensive systematic review and meta-analysis were conducted with 13 publications (9 randomized and 4 non-randomized controlled trials). The results demonstrated that the application of carbon nanoparticles in thyroid surgery led to an increase in the number of retrieved lymph nodes and identification of metastatic lymph nodes. Furthermore, it considerably reduced the rate of improper parathyroidectomy and the incidence of postoperative hypocalcemia. Conclusion: The application of carbon nanoparticles can effectively improve the effects of surgical treatment, can enhance the identification of intraoperative lymph nodes, reduce postoperative complications, and protect the integrity and function of the parathyroid gland. Systematic Review Registration: www.crd.york.ac.uk/PROSPERO, identifier, CRD42023420504.

5.
Cancer Immunol Res ; 10(8): 1028-1038, 2022 08 03.
Article de Anglais | MEDLINE | ID: mdl-35726891

RÉSUMÉ

The extent to which effector CD8+ T cells infiltrate into tumors is one of the major predictors of clinical outcome for patients with epithelial ovarian cancer (EOC). Immune cell infiltration into EOC is a complex process that could be affected by the epigenetic makeup of the tumor. Here, we have demonstrated that a lysine 4 histone H3 (H3K4) demethylase, (lysine-specific demethylase 5A; KDM5A) impairs EOC infiltration by immune cells and inhibits antitumor immune responses. Mechanistically, we found that KDM5A silenced genes involved in the antigen processing and presentation pathway. KDM5A inhibition restored the expression of genes involved in the antigen-presentation pathway in vitro and promoted antitumor immune responses mediated by CD8+ T cells in vivo in a syngeneic EOC mouse model. A negative correlation between expression of KDM5A and genes involved in the antigen processing and presentation pathway such as HLA-A and HLA-B was observed in the majority of cancer types. In summary, our results establish KDM5A as a regulator of CD8+ T-cell infiltration of tumors and demonstrate that KDM5A inhibition may provide a novel therapeutic strategy to boost antitumor immune responses.


Sujet(s)
Présentation d'antigène , Tumeurs de l'ovaire , Protéine-2 de liaison à la protéine du rétinoblastome , Animaux , Régulation négative , Femelle , Humains , Immunité , Lysine/métabolisme , Souris , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/immunologie , Protéine-2 de liaison à la protéine du rétinoblastome/métabolisme
6.
Cancer Immunol Res ; 9(12): 1413-1424, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34848497

RÉSUMÉ

The tumor immune microenvironment is influenced by the epigenetic landscape of the tumor. Here, we have identified the SETDB1-TRIM28 complex as a critical suppressor of antitumor immunity. An epigenetic CRISPR-Cas9 screen of 1,218 chromatin regulators identified TRIM28 as a suppressor of PD-L1 expression. We then revealed that expression of the SETDB1-TRIM28 complex negatively correlated with infiltration of effector CD8+ T cells. Inhibition of SETDB1-TRIM28 simultaneously upregulated PD-L1 and activated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) innate immune response pathway to increase infiltration of CD8+ T cells. Mechanistically, SETDB1-TRIM28 inhibition led to micronuclei formation in the cytoplasm, which is known to activate the cGAS-STING pathway. Thus, SETDB1-TRIM28 inhibition bridges innate and adaptive immunity. Indeed, SETDB1 knockout enhanced the antitumor effects of immune checkpoint blockade with anti-PD-L1 in a mouse model of ovarian cancer in a cGAS-dependent manner. Our findings establish the SETDB1-TRIM28 complex as a regulator of antitumor immunity and demonstrate that its loss activates cGAS-STING innate immunity to boost the antitumor effects of immune checkpoint blockade.


Sujet(s)
Épigénomique/méthodes , Histone-lysine N-methyltransferase/métabolisme , Immunité innée/immunologie , Immunothérapie/méthodes , Tumeurs de l'ovaire/génétique , Protéine-28 à motif tripartite/métabolisme , Animaux , Lignée cellulaire tumorale , Prolifération cellulaire , Femelle , Humains , Souris , Transfection , Microenvironnement tumoral
7.
Cancer Res ; 81(20): 5325-5335, 2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34548333

RÉSUMÉ

The SWI/SNF chromatin-remodeling complex is frequently altered in human cancers. For example, the SWI/SNF component ARID1A is mutated in more than 50% of ovarian clear cell carcinomas (OCCC), for which effective treatments are lacking. Here, we report that ARID1A transcriptionally represses the IRE1α-XBP1 axis of the endoplasmic reticulum (ER) stress response, which confers sensitivity to inhibition of the IRE1α-XBP1 pathway in ARID1A-mutant OCCC. ARID1A mutational status correlated with response to inhibition of the IRE1α-XBP1 pathway. In a conditional Arid1aflox/flox/Pik3caH1047R genetic mouse model, Xbp1 knockout significantly improved survival of mice bearing OCCCs. Furthermore, the IRE1α inhibitor B-I09 suppressed the growth of ARID1A-inactivated OCCCs in vivo in orthotopic xenograft, patient-derived xenograft, and the genetic mouse models. Finally, B-I09 synergized with inhibition of HDAC6, a known regulator of the ER stress response, in suppressing the growth of ARID1A-inactivated OCCCs. These studies define the IRE1α-XBP1 axis of the ER stress response as a targetable vulnerability for ARID1A-mutant OCCCs, revealing a promising therapeutic approach for treating ARID1A-mutant ovarian cancers. SIGNIFICANCE: These findings indicate that pharmacological inhibition of the IRE1α-XBP1 pathway alone or in combination with HDAC6 inhibition represents an urgently needed therapeutic strategy for ARID1A-mutant ovarian cancers.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protéines de liaison à l'ADN/génétique , Stress du réticulum endoplasmique , Endoribonucleases/antagonistes et inhibiteurs , Mutation , Tumeurs de l'ovaire/anatomopathologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Facteurs de transcription/génétique , Protéine-1 liant la boite X/antagonistes et inhibiteurs , Adénocarcinome à cellules claires/traitement médicamenteux , Adénocarcinome à cellules claires/génétique , Adénocarcinome à cellules claires/métabolisme , Adénocarcinome à cellules claires/anatomopathologie , Animaux , Apoptose , Prolifération cellulaire , Protéines de liaison à l'ADN/physiologie , Endoribonucleases/génétique , Endoribonucleases/métabolisme , Endoribonucleases/physiologie , Femelle , Régulation de l'expression des gènes tumoraux , Histone deacetylase 6/antagonistes et inhibiteurs , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Souris , Souris knockout , Souris nude , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/métabolisme , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/physiologie , Facteurs de transcription/physiologie , Cellules cancéreuses en culture , Protéine-1 liant la boite X/génétique , Protéine-1 liant la boite X/métabolisme , Protéine-1 liant la boite X/physiologie , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Nat Commun ; 12(1): 5321, 2021 09 07.
Article de Anglais | MEDLINE | ID: mdl-34493732

RÉSUMÉ

CARM1 is often overexpressed in human cancers including in ovarian cancer. However, therapeutic approaches based on CARM1 expression remain to be an unmet need. Cancer cells exploit adaptive responses such as the endoplasmic reticulum (ER) stress response for their survival through activating pathways such as the IRE1α/XBP1s pathway. Here, we report that CARM1-expressing ovarian cancer cells are selectively sensitive to inhibition of the IRE1α/XBP1s pathway. CARM1 regulates XBP1s target gene expression and directly interacts with XBP1s during ER stress response. Inhibition of the IRE1α/XBP1s pathway was effective against ovarian cancer in a CARM1-dependent manner both in vitro and in vivo in orthotopic and patient-derived xenograft models. In addition, IRE1α inhibitor B-I09 synergizes with immune checkpoint blockade anti-PD1 antibody in an immunocompetent CARM1-expressing ovarian cancer model. Our data show that pharmacological inhibition of the IRE1α/XBP1s pathway alone or in combination with immune checkpoint blockade represents a therapeutic strategy for CARM1-expressing cancers.


Sujet(s)
Carcinome épithélial de l'ovaire/thérapie , Endoribonucleases/génétique , Tumeurs de l'ovaire/thérapie , Récepteur-1 de mort cellulaire programmée/génétique , Protein-Serine-Threonine Kinases/génétique , Protein-arginine N-methyltransferases/génétique , Protéine-1 liant la boite X/génétique , Animaux , Anticorps monoclonaux/pharmacologie , Séquence nucléotidique , Benzopyranes/pharmacologie , Carcinome épithélial de l'ovaire/génétique , Carcinome épithélial de l'ovaire/immunologie , Carcinome épithélial de l'ovaire/anatomopathologie , Lignée cellulaire tumorale , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Endoribonucleases/antagonistes et inhibiteurs , Endoribonucleases/immunologie , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Hymécromone/analogues et dérivés , Hymécromone/pharmacologie , Inhibiteurs de points de contrôle immunitaires , Souris , Thérapie moléculaire ciblée , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/immunologie , Tumeurs de l'ovaire/anatomopathologie , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Récepteur-1 de mort cellulaire programmée/immunologie , Liaison aux protéines , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/immunologie , Protein-arginine N-methyltransferases/antagonistes et inhibiteurs , Protein-arginine N-methyltransferases/immunologie , Transduction du signal , Protéine-1 liant la boite X/antagonistes et inhibiteurs , Protéine-1 liant la boite X/immunologie , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Nat Cancer ; 2(2): 189-200, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-34085048

RÉSUMÉ

Alterations in components of the SWI/SNF chromatin-remodeling complex occur in ~20% of all human cancers. For example, ARID1A is mutated in up to 62% of clear cell ovarian carcinoma (OCCC), a disease currently lacking effective therapies. Here we show that ARID1A mutation creates a dependence on glutamine metabolism. SWI/SNF represses glutaminase (GLS1) and ARID1A inactivation upregulates GLS1. ARID1A inactivation increases glutamine utilization and metabolism through the tricarboxylic acid cycle to support aspartate synthesis. Indeed, glutaminase inhibitor CB-839 suppresses the growth of ARID1A mutant, but not wildtype, OCCCs in both orthotopic and patient-derived xenografts. In addition, glutaminase inhibitor CB-839 synergizes with immune checkpoint blockade anti-PDL1 antibody in a genetic OCCC mouse model driven by conditional Arid1a inactivation. Our data indicate that pharmacological inhibition of glutaminase alone or in combination with immune checkpoint blockade represents an effective therapeutic strategy for cancers involving alterations in the SWI/SNF complex such as ARID1A mutations.


Sujet(s)
Adénocarcinome à cellules claires , Tumeurs de l'ovaire , Adénocarcinome à cellules claires/traitement médicamenteux , Animaux , Protéines de liaison à l'ADN/génétique , Femelle , Glutaminase/génétique , Glutamine/usage thérapeutique , Humains , Inhibiteurs de points de contrôle immunitaires , Souris , Protéines nucléaires/génétique , Tumeurs de l'ovaire/traitement médicamenteux , Facteurs de transcription/génétique
10.
Nat Cell Biol ; 23(4): 355-365, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-33795874

RÉSUMÉ

Methyltransferase-like 3 (METTL3) and 14 (METTL14) are core subunits of the methyltransferase complex that catalyses messenger RNA N6-methyladenosine (m6A) modification. Despite the expanding list of m6A-dependent functions of the methyltransferase complex, the m6A-independent function of the METTL3 and METTL14 complex remains poorly understood. Here we show that genome-wide redistribution of METTL3 and METTL14 transcriptionally drives the senescence-associated secretory phenotype (SASP) in an m6A-independent manner. METTL14 is redistributed to the enhancers, whereas METTL3 is localized to the pre-existing NF-κB sites within the promoters of SASP genes during senescence. METTL3 and METTL14 are necessary for SASP. However, SASP is not regulated by m6A mRNA modification. METTL3 and METTL14 are required for both the tumour-promoting and immune-surveillance functions of senescent cells, which are mediated by SASP in vivo in mouse models. In summary, our results report an m6A-independent function of the METTL3 and METTL14 complex in transcriptionally promoting SASP during senescence.


Sujet(s)
Vieillissement/génétique , Vieillissement de la cellule/génétique , Methyltransferases/génétique , Adénosine/analogues et dérivés , Adénosine/génétique , Animaux , Méthylation de l'ADN/génétique , Génome/génétique , Souris , Facteur de transcription NF-kappa B/génétique , ARN messager/génétique
11.
J Invest Dermatol ; 141(6): 1564-1572.e4, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33333124

RÉSUMÉ

The SWI/SNF chromatin remodeler family includes the BAF and PBAF complexes. ARID2, encoding a PBAF complex subunit, is frequently mutated in melanoma independently of BRAF/RAS mutations. Emerging evidence shows that SWI/SNF complexes regulate tumor immunity; for instance, the loss of PBRM1, another PBAF complex subunit, enhances susceptibility to immune checkpoint inhibitors in melanoma. Notably, ARID2 mutations are more frequent in melanoma than PBRM1 mutations. However, the role of ARID2 as a modulator of tumor immunity remains unclear. In this study, we show that ARID2 knockout sensitizes melanoma to immune checkpoint inhibitors. Anti‒PD-L1 treatment restricts tumor growth in mice bearing ARID2-knockout melanoma cells, correlating with an increase in the infiltration of cytotoxic CD8+ T cells. Furthermore, ARID2 deficiency leads to signal transducer and activator of transcription 1 upregulation, which subsequently causes increased expression of T-cell‒attracting chemokines such as CXCL9, CXCL10, and CCL5. These results demonstrate that ARID2 is an immunomodulator and a potential biomarker that indicates immune checkpoint inhibitor effectiveness in patients with melanoma.


Sujet(s)
Résistance aux médicaments antinéoplasiques/génétique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Mélanome/traitement médicamenteux , Tumeurs cutanées/traitement médicamenteux , Facteurs de transcription/déficit , Animaux , Modèles animaux de maladie humaine , Techniques de knock-out de gènes , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Mélanome/génétique , Mélanome/immunologie , Mélanome/anatomopathologie , Souris , Mutation , Transduction du signal/génétique , Transduction du signal/immunologie , Tumeurs cutanées/génétique , Tumeurs cutanées/immunologie , Tumeurs cutanées/anatomopathologie , Facteurs de transcription/génétique
12.
Nat Commun ; 11(1): 908, 2020 02 19.
Article de Anglais | MEDLINE | ID: mdl-32075966

RÉSUMÉ

Cyclic cGMP-AMP synthase (cGAS) is a pattern recognition cytosolic DNA sensor that is essential for cellular senescence. cGAS promotes inflammatory senescence-associated secretory phenotype (SASP) through recognizing cytoplasmic chromatin during senescence. cGAS-mediated inflammation is essential for the antitumor effects of immune checkpoint blockade. However, the mechanism by which cGAS recognizes cytoplasmic chromatin is unknown. Here we show that topoisomerase 1-DNA covalent cleavage complex (TOP1cc) is both necessary and sufficient for cGAS-mediated cytoplasmic chromatin recognition and SASP during senescence. TOP1cc localizes to cytoplasmic chromatin and TOP1 interacts with cGAS to enhance the binding of cGAS to DNA. Retention of TOP1cc to cytoplasmic chromatin depends on its stabilization by the chromatin architecture protein HMGB2. Functionally, the HMGB2-TOP1cc-cGAS axis determines the response of orthotopically transplanted ex vivo therapy-induced senescent cells to immune checkpoint blockade in vivo. Together, these findings establish a HMGB2-TOP1cc-cGAS axis that enables cytoplasmic chromatin recognition and response to immune checkpoint blockade.


Sujet(s)
Vieillissement de la cellule/immunologie , ADN topoisomérases de type I/métabolisme , Protéine HMG2/métabolisme , Nucleotidyltransferases/métabolisme , Animaux , Antigène CD274/immunologie , Lignée cellulaire , Chromatine/immunologie , Chromatine/métabolisme , Cytosol/immunologie , Cytosol/métabolisme , ADN/immunologie , ADN/métabolisme , Altération de l'ADN/immunologie , ADN topoisomérases de type I/génétique , Techniques de knock-down de gènes , Protéine HMG2/génétique , Humains , Inflammation , Souris , Souris de lignée C57BL , Mutation , Tumeurs/immunologie , Nucleotidyltransferases/génétique , Liaison aux protéines , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Sci Rep ; 10(1): 1821, 2020 02 04.
Article de Anglais | MEDLINE | ID: mdl-32020017

RÉSUMÉ

Chemoresistance is one of the leading causes of mortality in breast cancer (BC). Understanding the molecules regulating chemoresistance is critical in order to combat chemoresistant BC. Drug efflux pump ABCB1 is overexpressed in chemoresistant neoplasms where it effluxes various chemotherapeutic agents from cells. Because it is expressed in normal and cancerous cells alike, attempts at targeting ABCB1 directly have failed due to low specificity and disruption of normal tissue. A proposed method to inhibit ABCB1 is to target its cancer-specific, upstream regulators, mitigating damage to normal tissue. Few such cancer-specific upstream regulators have been described. Here we characterize ROR1 as an upstream regulator of ABCB1. ROR1 is highly expressed during development but not expressed in normal adult tissue. It is however highly expressed in several cancers. ROR1 is overexpressed in chemoresistant BC where it correlates with poor therapy response and tumor recurrence. Our data suggests, ROR1 inhibition sensitizes BC cells to chemo drugs. We also show ROR1 regulates ABCB1 stability and transcription via MAPK/ERK and p53. Validating our overall findings, inhibition of ROR1 directly correlated with decreased efflux of chemo-drugs from cells. Overall, our results highlight ROR1's potential as a therapeutic target for multidrug resistant malignancies.


Sujet(s)
Tumeurs du sein/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Sous-famille B de transporteurs à cassette liant l'ATP/métabolisme , Antinéoplasiques/métabolisme , Antinéoplasiques/usage thérapeutique , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Immunoprécipitation de la chromatine , Doxorubicine/métabolisme , Doxorubicine/usage thérapeutique , Femelle , Technique d'immunofluorescence , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Humains , Système de signalisation des MAP kinases , Réaction de polymérisation en chaine en temps réel
14.
Cancer Cell ; 37(2): 157-167.e6, 2020 02 10.
Article de Anglais | MEDLINE | ID: mdl-32004442

RÉSUMÉ

In response to DNA double-strand breaks, MAD2L2-containing shieldin complex plays a critical role in the choice between homologous recombination (HR) and non-homologous end-joining (NHEJ)-mediated repair. Here we show that EZH2 inhibition upregulates MAD2L2 and sensitizes HR-proficient epithelial ovarian cancer (EOC) to poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor in a CARM1-dependent manner. CARM1 promotes MAD2L2 silencing by driving the switch from the SWI/SNF complex to EZH2 through methylating the BAF155 subunit of the SWI/SNF complex on the MAD2L2 promoter. EZH2 inhibition upregulates MAD2L2 to decrease DNA end resection, which increases NHEJ and chromosomal abnormalities, ultimately causing mitotic catastrophe in PARP inhibitor treated HR-proficient cells. Significantly, EZH2 inhibitor sensitizes CARM1-high, but not CARM-low, EOCs to PARP inhibitors in both orthotopic and patient-derived xenografts.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Recombinaison homologue/effets des médicaments et des substances chimiques , Tumeurs de l'ovaire/traitement médicamenteux , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Antinéoplasiques/usage thérapeutique , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Réparation de l'ADN par jonction d'extrémités/effets des médicaments et des substances chimiques , Antienzymes/usage thérapeutique , Femelle , Humains , Tumeurs de l'ovaire/génétique , Protein-arginine N-methyltransferases/effets des médicaments et des substances chimiques , Réparation de l'ADN par recombinaison/effets des médicaments et des substances chimiques
15.
Nat Commun ; 10(1): 4067, 2019 09 06.
Article de Anglais | MEDLINE | ID: mdl-31492885

RÉSUMÉ

ARID1A inactivation causes mitotic defects. Paradoxically, cancers with high ARID1A mutation rates typically lack copy number alterations (CNAs). Here, we show that ARID1A inactivation causes defects in telomere cohesion, which selectively eliminates gross chromosome aberrations during mitosis. ARID1A promotes the expression of cohesin subunit STAG1 that is specifically required for telomere cohesion. ARID1A inactivation causes telomere damage that can be rescued by STAG1 expression. Colony formation capability of single cells in G2/M, but not G1 phase, is significantly reduced by ARID1A inactivation. This correlates with an increase in apoptosis and a reduction in tumor growth. Compared with ARID1A wild-type tumors, ARID1A-mutated tumors display significantly less CNAs across multiple cancer types. Together, these results show that ARID1A inactivation is selective against gross chromosome aberrations through causing defects in telomere cohesion, which reconciles the long-standing paradox between the role of ARID1A in maintaining mitotic integrity and the lack of genomic instability in ARID1A-mutated cancers.


Sujet(s)
Instabilité du génome , Mutation , Protéines nucléaires/génétique , Tumeurs de l'ovaire/génétique , Télomère/génétique , Facteurs de transcription/génétique , Animaux , Apoptose/génétique , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire , Lignée cellulaire tumorale , Protéines chromosomiques nonhistones/génétique , Protéines chromosomiques nonhistones/métabolisme , Variations de nombre de copies de segment d'ADN , Protéines de liaison à l'ADN , Femelle , Humains , Souris de lignée NOD , Souris knockout , Souris SCID , Souris transgéniques , Protéines nucléaires/métabolisme , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/anatomopathologie , Télomère/métabolisme , Facteurs de transcription/métabolisme , Transplantation hétérologue/méthodes , Charge tumorale/génétique , Cohesins
16.
FEBS Lett ; 592(2): 244-255, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29251779

RÉSUMÉ

Nucleophosmin (NPM1) is a multifunctional nucleolar protein. Here, we analyze the role of NPM1 in gene expression using our previous microarray data and find a relationship between NPM1 and interferon (IFN)-γ-inducible genes. We show that NPM1 selectively regulates the expression of a subset of IFN-γ-inducible genes and directly binds to two important transcription factors in the type II IFN pathway: signal transducer and activator of transcription 1 and interferon regulatory factor 1 (IRF1). Furthermore, NPM1 is found to regulate the IFN-γ-inducible promoter activity of major histocompatibility complex class II transactivator (CIITA), and mutation of the IRF1-binding site on the CIITA promoter abolishes the effect of NPM1. Our results suggest a novel mechanism for IFN-γ-mediated gene expression by NPM1.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Interféron gamma/pharmacologie , Protéines nucléaires/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Cellules HEK293 , Cellules HeLa , Humains , Facteur-1 de régulation d'interféron/génétique , Protéines nucléaires/génétique , Nucléophosmine , Régions promotrices (génétique) , Facteur de transcription STAT-1/génétique , Transactivateurs/génétique
17.
Nucleic Acids Res ; 45(7): 3707-3723, 2017 04 20.
Article de Anglais | MEDLINE | ID: mdl-28003476

RÉSUMÉ

NPM1/nucleophosmin is frequently overexpressed in various tumors, although the oncogenic role of NPM1 remains unclear. Here we revealed the link between NPM1 and nuclear factor-κB (NF-κB), a master regulator of inflammation. We found that NPM1 knockdown decreased NF-κB-mediated transcription of selected target genes by decreasing the recruitment of NF-κB p65 to the gene promoters. NPM1 is directly associated with the DNA binding domain of p65 to enhance its DNA binding activity without being a part of the DNA-NF-κB complex. This result suggests that NF-κB requires the chaperone-like function of NPM1 for DNA binding. Furthermore, we demonstrated that NPM1 was required for efficient inflammatory gene expression induced by tumor necrosis factor alpha (TNF-α) and lipopolysaccharide in fibroblasts and macrophages. The NF-κB-mediated invasion of breast cancer cells was significantly decreased by NPM1 knockdown. Our study suggests a novel mechanistic insight into the NF-κB-mediated transcription and an oncogenic role of NPM1 in both tumor cells and the tumor micro-environment through the regulation of NF-κB.


Sujet(s)
Régulation de l'expression des gènes , Facteur de transcription NF-kappa B/métabolisme , Protéines nucléaires/métabolisme , Transcription génétique , Animaux , Cellules cultivées , ADN/métabolisme , Cellules HeLa , Humains , Souris de lignée C57BL , Protéines nucléaires/physiologie , Nucléophosmine , Liaison aux protéines , Facteur de transcription RelA/métabolisme
18.
Biochem Biophys Res Commun ; 480(4): 702-708, 2016 11 25.
Article de Anglais | MEDLINE | ID: mdl-27983985

RÉSUMÉ

NPM1/nucleophosmin is a multifunctional and oligomeric phosphoprotein. A number of observations have suggested that changes in the oligomer formation of NPM1 could influence its biological functions, especially its oncogenic functions. To understand the functional meaning of oligomerization of NPM1/nucleophosmin, we have established a novel method to monitor protein oligomerization in cells. We utilized the split synthetic Renilla luciferase protein fragment-assisted complementation (SRL-PFAC) bioluminescence activity and observed the change of NPM1 oligomer levels under various cell culture conditions. Our study provides a method for systematic characterization of NPM1 oligomer formation changes and for screening inhibitors of NPM1 oligomerization.


Sujet(s)
Protéines nucléaires/métabolisme , Nucléoplasmines/métabolisme , Sites de fixation , Dimérisation , Cellules HEK293 , Cellules HeLa , Humains , Microscopie de fluorescence , Nucléophosmine , Liaison aux protéines , Cartographie d'interactions entre protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE