Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Aesthet Surg J ; 44(4): 363-372, 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38078367

RESUMO

BACKGROUND: Breast implants have always been composed of a silicone elastomer envelope filled with either silicone gel or saline. Breast implant illness (BII) is a set of symptoms that has previously been linked to the leakage of silicone particles from the implants into the body. OBJECTIVES: Our research aimed to quantify the number of silicone particles present in the capsules of breast implants available in North America. METHODS: Thirty-five periprosthetic capsules were sampled and analyzed, and silicone particles were counted and measured. The capsule surface area was then measured and utilized to calculate particle density and total number of silicone particles. RESULTS: Eighty-five percent of capsules analyzed from silicone gel implants contained silicone, with an average of 62 particles per mm3 of capsular tissue. These implants had approximately 1 million silicone particles per capsule. In contrast, none of the saline implant capsules contained silicone. Capsules from macrotextured tissue expanders contained fewer and larger silicone particles. CONCLUSIONS: Silicone gel implants presented silicone particle bleeding into the periprosthetic capsule, totaling on average 1 million silicone particles per capsule. On the other hand, no silicone particle bleeding was observed from saline breast implants. These data suggest that particle bleeding comes from the inner silicone gel, and not from the smooth outer silicone shell. Previous studies have reported the presence of breast implant illness in patients with both silicone- and saline-filled implants. Therefore, our data suggest that silicone migration is not the sole cause of BII.


Assuntos
Implante Mamário , Implantes de Mama , Humanos , Implantes de Mama/efeitos adversos , Géis de Silicone/efeitos adversos , Mama/cirurgia , Implante Mamário/efeitos adversos , Solução Salina , América do Norte
3.
ACS Appl Bio Mater ; 6(3): 1173-1184, 2023 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-36795958

RESUMO

We designed, synthesized, and characterized a Raman nanoprobe made of dye-sensitized single-walled carbon nanotubes (SWCNTs) that can selectively target biomarkers of breast cancer cells. The nanoprobe is composed of Raman-active dyes encapsulated inside a SWCNT, whose surface is covalently grafted with poly(ethylene glycol) (PEG) at a density of ∼0.7% per carbon. Using α-sexithiophene- and ß-carotene-derived nanoprobes covalently bound to an antibody, either anti-E-cadherin (E-cad) or anti-keratin-19 (KRT19), we prepared two distinct nanoprobes that specifically recognize biomarkers on breast cancer cells. Immunogold experiments and transmission electron microscopy (TEM) images are first used to guide the synthesis protocol for higher PEG-antibody attachment and biomolecule loading capacity. The duplex of nanoprobes was then applied to target E-cad and KRT19 biomarkers in T47D and MDA-MB-231 breast cancer cell lines. Hyperspectral imaging of specific Raman bands allows for simultaneous detection of this nanoprobe duplex on target cells without the need for additional filters or subsequent incubation steps. Our results confirm the high reproducibility of the nanoprobe design for duplex detection and highlight the potential of Raman imaging for advanced biomedical applications in oncology.


Assuntos
Nanotubos de Carbono , Neoplasias , Reprodutibilidade dos Testes , Biomarcadores , Polietilenoglicóis , Anticorpos , Fenótipo
4.
J Allergy Clin Immunol ; 151(1): 247-259.e7, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35973477

RESUMO

BACKGROUND: Familial hemophagocytic lymphohistiocytosis is a life-threatening hyperinflammatory disease caused by genetic defects in the granule-mediated cytotoxic pathway. Success of hematopoietic cell transplantation, the only cure, is correlated with the extent of disease control before transplantation. Unfortunately, disease refractoriness and toxicities to standard chemotherapy-based regimens are fatal in a fraction of patients. Novel targeted immunotherapies, such as IFN-γ blocking antibodies or ruxolitinib, a Janus kinase (JAK) 1/2 inhibitor, are promising but only partially effective at controlling disease. OBJECTIVE: We asked whether combinations of cytokine-targeted therapies, using antibodies or JAK inhibitor, work synergistically to counteract HLH. METHODS: Genetically predisposed mice were infected and treated with distinct combinations of immunotherapies. Disease outcome was monitored and compared to monotherapies. RESULTS: We showed that inhibiting IL-6 or IL-18 signaling in combination with IFN-γ blockade or ruxolitinib did not increase disease control compared to anti-IFN-γ antibodies or ruxolitinib monotherapies. In contrast, clinically relevant doses of ruxolitinib combined with low doses of anti-IFN-γ blocking antibodies corrected cytopenias, prevented overt neutrophilia, limited cytokinemia, and resolved HLH immunopathology and symptomatology. CONCLUSIONS: Our findings demonstrate that IFN-γ blockade and ruxolitinib act synergistically to suppress HLH progression. This supports the use of combined cytokine-targeted therapies as a bridge to hematopoietic cell transplantation in severe familial hemophagocytic lymphohistiocytosis.


Assuntos
Linfo-Histiocitose Hemofagocítica , Animais , Camundongos , Linfo-Histiocitose Hemofagocítica/tratamento farmacológico , Anticorpos Bloqueadores/uso terapêutico , Interferon gama/genética , Citocinas/metabolismo
5.
Cancers (Basel) ; 14(21)2022 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-36358871

RESUMO

Estrogen receptor alpha (ERα) expression in ~2/3 breast tumors selects patients for hormonal therapies. Tumors negative for ERα but positive for the progesterone receptor (PR, encoded by PGR) have also been candidates for ER-targeting therapies, as PR expression may reflect undetected ER activity. Conversely, PR- status in ER+ tumors predicts a worse therapeutic response. Our analysis of breast tumor transcriptome datasets, however, revealed that in tumors with lower PGR expression, the clinical PR status does not correlate accurately with the expression of ESR1 or of ER target genes, including PGR itself. We identified carbonic anhydrase 12 (CA12) as an estrogen target gene better correlated with ESR1 than PGR, reflecting CA12 regulation by both ERα and the luminal factor and upstream ESR1 regulator GATA3. Immunostaining supported strong positive correlations at the protein level with ERα and GATA3 in a cohort of 118 tumors. Most ER+PR- tumors expressed CAXII at levels similar to those of ER+PR+ tumors, consistent with observations in tumor transcriptome datasets and with active estrogenic signaling in some ER+PR- breast cancer cell lines. The few ER-PR+ tumors did not express CAXII or the other luminal markers FOXA1 and GATA3. Overall, CAXII is a luminal marker that can help interpret ER status in single ER/PR positive tumors.

6.
Cell Mol Biol (Noisy-le-grand) ; 68(9): 113-124, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36905266

RESUMO

Apelin-13 is an adipokine known for its growth-inducing effects on human breast cancer cells in an estrogen-containing environment. However, the response of these cells to apelin-13 in the absence of estrogen and its association with the expression of the apelin receptor (APLNR) has not yet been investigated. In the present study, we show that the breast cancer cell line MCF-7 expresses the APLNR as shown by immunofluorescence and flow cytometry, under conditions of ER starvation and that culture of these cells in the presence of apelin-13 results in an increased growth rate and a diminished autophagy flux.  Moreover, the binding of APLNR by apelin-13 resulted in an increased growth rate (assayed by AlamarBlue) and a diminished autophagy flux (monitored by Lysotracker Green). The latter observations were reversed in the presence of exogenous estrogen. Finally, apelin-13 induces the deactivation of the apoptotic kinase AMPK. Taken together, our results show that APLNR signaling in breast cancer cells is functional and prevents tumor growth under conditions of estrogen starvation. They furthermore suggest an alternative mechanism of estrogen-independent tumor growth thereby positioning the APLNR-AMPK axis as a novel pathway and a possible therapeutic target in endocrine resistance of breast cancer cells.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Apelina , Neoplasias da Mama/patologia , Receptores de Apelina , Estrogênios , Células MCF-7 , Autofagia , Linhagem Celular Tumoral
7.
Cancers (Basel) ; 13(20)2021 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-34680248

RESUMO

The impact of epithelial-mesenchymal transition (EMT) signature on the immune infiltrate present in the breast cancer tumor microenvironment (TME) is still poorly understood. Since there is mounting interest in the use of immunotherapy for the treatment of subsets of breast cancer patients, it is of major importance to understand the fundamental tumor characteristics which dictate the inter-tumor heterogeneity in immune landscapes. We aimed to assess the impact of EMT-related markers on the nature and magnitude of the inflammatory infiltrate present in breast cancer TME and their association with the clinicopathological parameters. Tissue microarrays were constructed from 144 formalin-fixed paraffin-embedded invasive breast cancer tumor samples. The protein expression patterns of Snail, Twist, ZEB1, N-cadherin, Vimentin, GRHL2, E-cadherin, and EpCAM were examined by immunohistochemistry (IHC). The inflammatory infiltrate in the TME was assessed semi-quantitatively on hematoxylin and eosin (H&E)-stained whole sections and was characterized using IHC. The inflammatory infiltrate was more intense in poorly differentiated carcinomas and triple-negative carcinomas in which the expression of E-cadherin and GRHL2 was reduced, while EpCAM was overexpressed. Most EMT-related markers correlated with plasma cell infiltration of the TME. Taken together, our findings reveal that the EMT signature might impact the immune response in the TME.

8.
J Immunol ; 207(6): 1599-1615, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34408010

RESUMO

GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1ß and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.


Assuntos
Proteínas de Ligação a DNA/deficiência , Histona Desmetilases/metabolismo , Proteínas Mutantes/metabolismo , Transdução de Sinais/genética , Síndrome de Resposta Inflamatória Sistêmica/sangue , Fatores de Transcrição/deficiência , Animais , Transplante de Medula Óssea/métodos , Linfócitos T CD8-Positivos/imunologia , Citocinas/sangue , Citocinas/genética , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Técnicas de Introdução de Genes , Histonas/metabolismo , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Ligação Proteica , Síndrome de Resposta Inflamatória Sistêmica/genética , Síndrome de Resposta Inflamatória Sistêmica/imunologia , Fatores de Transcrição/genética
9.
Oncogene ; 40(12): 2243-2257, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33649538

RESUMO

Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein ß-arrestin2 (ß-arr2) regulates tumor suppressor p53 by counteracting Mdm2. ß-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. ß-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. ß-arr2 can be SUMOylated, but no information is available on how SUMO may regulate ß-arr2 nucleocytoplasmic shuttling. While we found ß-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and ß-arr2, via a SUMO interaction motif (SIM), that is required for ß-arr2 cytonuclear trafficking. This SIM promotes association of ß-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective ß-arr2 nuclear entry. Mutation of the SIM inhibits ß-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a ß-arr2 SIM nuclear entry checkpoint, coupled with active ß-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.


Assuntos
Proteínas Ativadoras de GTPase/genética , Neoplasias/genética , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína SUMO-1/genética , Proteína Supressora de Tumor p53/genética , beta-Arrestina 2/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Sinais de Exportação Nuclear/genética , Transdução de Sinais/genética , Sumoilação/genética
10.
Curr Oncol ; 28(1): 593-605, 2021 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-33498159

RESUMO

BACKGROUND: Studies have shown that aggressive treatment of non-small cell lung cancer (NSCLC) with oligometastatic disease improves the overall survival (OS) compared to a palliative approach and some immunotherapy checkpoint inhibitors, such as anti-programmed cell death ligand 1 (PD-L1), anti-programmed cell death protein 1 (PD-1), and T-Lymphocyte-associated antigen 4 (CTLA-4) inhibitors are now part of the standard of care for advanced NSCLC. However, the prognostic impact of PD-L1 expression in the oligometastatic setting remains unknown. METHODS: Patients with oligometastatic NSCLC were identified from the patient database of the Centre hospitalier de l'Université de Montréal (CHUM). "Oligometastatic disease" definition chosen is one synchronous metastasis based on the M1b staging of the eight IASLC (The International Association for the Study of Lung Cancer) Classification (within sixth months of diagnosis) or up to three cerebral metastasis based on the methodology of the previous major phase II randomized study of Gomez et al. We compared the OS between patients receiving aggressive treatment at both metastatic and primary sites (Group A) and patients receiving non-aggressive treatment (Group B). Subgroup analysis was performed using tumor PD-L1 expression. RESULTS: Among 643 metastatic NSCLC patients, we identified 67 patients with oligometastasis (10%). Median follow-up was 13.3 months. Twenty-nine patients (43%) received radical treatment at metastatic and primary sites (Group A), and 38 patients (57%) received non-aggressive treatment (Group B). The median OS (mOS) of Group A was significantly longer than for Group B (26 months vs. 5 months, p = 0.0001). Median progression-free survival (mPFS) of Group A was superior than Group B (17.5 months vs. 3.4 months, p = 0.0001). This difference was still significant when controlled for primary tumor staging: stage I (p = 0.316), stage II (p = 0.024), and stage III (p = 0.001). In the cohort of patients who were not treated with PD-L1 inhibitors, PD-L1 expression negatively correlated with mOS. CONCLUSIONS: Aggressive treatments of oligometastatic NSCLC significantly improve mOS and mPFS compared to a more palliative approach. PD-L1 expression is a negative prognostic factor which suggests a possible role for immunotherapy in this setting.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antígeno B7-H1/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Estadiamento de Neoplasias , Prognóstico
11.
J Immunol ; 205(5): 1268-1280, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32690660

RESUMO

Regulation of MHC class I (MHC I) expression has been studied almost exclusively in hematolymphoid cells. We report that thymic epithelial cells (TECs), particularly the medullary TECs, constitutively express up to 100-fold more cell surface MHC I proteins than epithelial cells (ECs) from the skin, colon, and lung. Differential abundance of cell surface MHC I in primary ECs is regulated via transcription of MHC I and of genes implicated in the generation of MHC I-binding peptides. Superior MHC I expression in TECs is unaffected by deletion of Ifnar1 or Ifngr1, but is lessened by deletion of Aire, Ifnlr1, Stat1, or Nlrc5, and is driven mainly by type III IFN produced by medullary TECs. Ifnlr1 -/- mice show impaired negative selection of CD8 thymocytes and, at 9 mo of age, present autoimmune manifestations. Our study shows unanticipated variation in MHC I expression by ECs from various sites and provides compelling evidence that superior expression of MHC I in TECs is crucial for proper thymocyte education.


Assuntos
Células Epiteliais/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Interferons/imunologia , Receptores de Interferon/imunologia , Timo/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Timócitos/imunologia , Interferon lambda
12.
Nat Commun ; 11(1): 3701, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32709883

RESUMO

Despite its importance in human cancers, including colorectal cancers (CRC), oncogenic KRAS has been extremely challenging to target therapeutically. To identify potential vulnerabilities in KRAS-mutated CRC, we characterize the impact of oncogenic KRAS on the cell surface of intestinal epithelial cells. Here we show that oncogenic KRAS alters the expression of a myriad of cell-surface proteins implicated in diverse biological functions, and identify many potential surface-accessible therapeutic targets. Cell surface-based loss-of-function screens reveal that ATP7A, a copper-exporter upregulated by mutant KRAS, is essential for neoplastic growth. ATP7A is upregulated at the surface of KRAS-mutated CRC, and protects cells from excess copper-ion toxicity. We find that KRAS-mutated cells acquire copper via a non-canonical mechanism involving macropinocytosis, which appears to be required to support their growth. Together, these results indicate that copper bioavailability is a KRAS-selective vulnerability that could be exploited for the treatment of KRAS-mutated neoplasms.


Assuntos
Neoplasias Colorretais/metabolismo , Cobre/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Disponibilidade Biológica , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , ATPases Transportadoras de Cobre/metabolismo , Feminino , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Mutação
13.
Am J Transplant ; 20(3): 726-738, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31729155

RESUMO

Tertiary lymphoid structures (TLS) accumulate at sites of chronic injury where they function as an ectopic germinal center, fostering local autoimmune responses. Vascular injury leads to the release of endothelial-derived apoptotic exosome-like vesicles (ApoExo) that contribute to rejection in transplanted organs. The purpose of the study was to evaluate the impact of ApoExo on TLS formation in a model of vascular allograft rejection. Mice transplanted with an allogeneic aortic transplant were injected with ApoExo. The formation of TLS was significantly increased by ApoExo injection along with vascular remodeling and increased levels of antinuclear antibodies and anti-perlecan/LG3 autoantibodies. ApoExo also enhanced allograft infiltration by γδT17 cells. Recipients deficient in γδT cells showed reduced TLS formation and lower autoantibodies levels following ApoExo injection. ApoExo are characterized by proteasome activity, which can be blocked by bortezomib. Bortezomib treated ApoExo reduced the recruitment of γδT17 cells to the allograft, lowered TLS formation, and reduced autoantibody production. This study identifies vascular injury-derived extracellular vesicles (ApoExo), as initiators of TLS formation and demonstrates the pivotal role of γδT17 in coordinating TLS formation and autoantibody production. Finally, our results suggest proteasome inhibition with bortezomib as a potential option for controlling TLS formation in rejected allografts.


Assuntos
Vesículas Extracelulares , Estruturas Linfoides Terciárias , Aloenxertos , Animais , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Transplante Homólogo
14.
Ann Surg Oncol ; 26(13): 4337-4345, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31605348

RESUMO

BACKGROUND: The sentinel node biopsy following neoadjuvant chemotherapy (SN FNAC) study has shown that in node-positive (N+) breast cancer, sentinel node biopsy (SNB) can be performed following neoadjuvant chemotherapy (NAC), with a low false negative rate (FNR = 8.4%). A secondary endpoint of the SN FNAC study was to determine whether axillary ultrasound (AxUS) could predict axillary pathological complete response (ypN0) and increase the accuracy of SNB. METHODS: The SN FNAC trial is a study of patients with biopsy-proven N+ breast cancer who underwent SNB followed by completion node dissection. All patients had AxUS following NAC and the axillary nodes were classified as either positive (AxUS+) or negative (AxUS-). AxUS was compared with the final axillary pathology results. RESULTS: There was no statistical difference in the baseline characteristics of patients with AxUS+ versus those with AxUS-. Overall, 82.5% (47/57) of AxUS+ patients had residual positive lymph nodes (ypN+) at surgery and 53.8% (42/78) of AxUS- patients had ypN+. Post NAC AxUS sensitivity was 52.8%, specificity 78.3%, and negative predictive value 46.2%. AxUS FNR was 47.2%, versus 8.4% for SNB. If post-NAC AxUS- was used to select patients for SNB, FNR would decrease from 8.4 to 2.7%. However, using post-NAC AxUS in addition to SNB as an indication for ALND would have led to unnecessary ALND in 7.8% of all patients. CONCLUSION: AxUS is not appropriate as a standalone staging procedure, and SNB itself is sufficient to assess the axilla post NAC in patients who present with N+ breast cancer.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Carcinoma Lobular/patologia , Linfonodos/patologia , Terapia Neoadjuvante/métodos , Ultrassonografia Mamária/métodos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Axila , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Carcinoma Ductal de Mama/diagnóstico por imagem , Carcinoma Ductal de Mama/tratamento farmacológico , Carcinoma Lobular/diagnóstico por imagem , Carcinoma Lobular/tratamento farmacológico , Quimioterapia Adjuvante , Feminino , Seguimentos , Humanos , Linfonodos/diagnóstico por imagem , Linfonodos/efeitos dos fármacos , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Biópsia de Linfonodo Sentinela
15.
Neoplasia ; 21(10): 1015-1035, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31476594

RESUMO

Breast cancer is a heterogeneous disease comprising the estrogen receptor (ER)-positive luminal subtype which is subdivided into luminal A and luminal B and ER-negative breast cancer which includes the triple-negative subtype. This study has four aims: 1) to examine whether Minichromosome Maintenance (MCM)2, MCM4, and MCM6 can be used as markers to differentiate between luminal A and luminal B subtypes; 2) to study whether MCM2, MCM4, and MCM6 are highly expressed in triple-negative breast cancer, as there is an urgent need to search for surrogate markers in this aggressive subtype, for drug development purposes; 3) to compare the prognostic values of these markers in predicting relapse-free survival; and 4) to compare the three approaches used for scoring the protein expression of these markers by immunohistochemistry (IHC). MCM2, MCM4, MCM6, and MKI67 mRNA expression was first studied using in silico analysis of available breast cancer datasets. We next used IHC to evaluate their protein expression on tissue microarrays using three scoring methods. MCM2, MCM4, and MCM6 can help in distinction between luminal A and luminal B whose therapeutic management and clinical outcomes are different. MCM2, MCM4, MCM6, and Ki-67 are highly expressed in breast cancer of high histological grades that comprise clinically aggressive tumors such as luminal B, HER2-positive, and triple-negative subtypes. Low transcript expression of these markers is associated with increased probability of relapse-free survival. A positive relationship exists among the three scoring methods of each of the four markers. An independent validation cohort is needed to confirm their clinical utility.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/metabolismo , Componente 2 do Complexo de Manutenção de Minicromossomo/metabolismo , Componente 4 do Complexo de Manutenção de Minicromossomo/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Biologia Computacional/métodos , Progressão da Doença , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Componente 2 do Complexo de Manutenção de Minicromossomo/genética , Componente 4 do Complexo de Manutenção de Minicromossomo/genética , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Análise de Sobrevida
16.
Br J Cancer ; 119(5): 580-590, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30078843

RESUMO

BACKGROUND: Distinguishing lung adenocarcinoma (ADC) from squamous cell carcinoma (SCC) has a tremendous therapeutic implication. Sometimes, the commonly used immunohistochemistry (IHC) markers fail to discriminate between them, urging for the identification of new diagnostic biomarkers. METHODS: We performed IHC on tissue microarrays from two cohorts of lung cancer patients to analyse the expression of beta-arrestin-1, beta-arrestin-2 and clinically used diagnostic markers in ADC and SCC samples. Logistic regression models were applied for tumour subtype prediction. Parallel reaction monitoring (PRM)-based mass spectrometry was used to quantify beta-arrestin-1 in plasma from cancer patients and healthy donors. RESULTS: Beta-arrestin-1 expression was significantly higher in ADC versus SCC samples. Beta-arrestin-1 displayed high sensitivity, specificity and negative predictive value. Its usefulness in an IHC panel was also shown. Plasma beta-arrestin-1 levels were considerably higher in lung cancer patients than in healthy donors and were higher in patients who later experienced a progressive disease than in patients showing complete/partial response following EGFR inhibitor therapy. CONCLUSIONS: Our data identify beta-arrestin-1 as a diagnostic marker to differentiate ADC from SCC and indicate its potential as a plasma biomarker for non-invasive diagnosis of lung cancer. Its utility to predict response to EGFR inhibitors is yet to be confirmed.


Assuntos
Adenocarcinoma de Pulmão/diagnóstico , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/diagnóstico , Neoplasias Pulmonares/diagnóstico , Regulação para Cima , beta-Arrestina 1/metabolismo , Adenocarcinoma de Pulmão/sangue , Adenocarcinoma de Pulmão/metabolismo , Biomarcadores Tumorais/sangue , Carcinoma de Células Escamosas/sangue , Carcinoma de Células Escamosas/metabolismo , Estudos de Casos e Controles , Diagnóstico Diferencial , Progressão da Doença , Detecção Precoce de Câncer , Regulação Neoplásica da Expressão Gênica , Humanos , Modelos Logísticos , Neoplasias Pulmonares/sangue , Neoplasias Pulmonares/metabolismo , Valor Preditivo dos Testes , Análise Serial de Tecidos , beta-Arrestina 1/sangue
17.
Cell Rep ; 23(5): 1476-1490, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29719259

RESUMO

AXL is activated by its ligand GAS6 and is expressed in triple-negative breast cancer cells. In the current study, we report AXL expression in HER2-positive (HER2+) breast cancers where it correlates with poor patient survival. Using murine models of HER2+ breast cancer, Axl, but not its ligand Gas6, was found to be essential for metastasis. We determined that AXL is required for intravasation, extravasation, and growth at the metastatic site. We found that AXL is expressed in HER2+ cancers displaying epithelial-to-mesenchymal transition (EMT) signatures where it contributes to sustain EMT. Interfering with AXL in a patient-derived xenograft (PDX) impaired transforming growth factor ß (TGF-ß)-induced cell invasion. Last, pharmacological inhibition of AXL specifically decreased the metastatic burden of mice developing HER2+ breast cancer. Our data identify AXL as a potential anti-metastatic co-therapeutic target for the treatment of HER2+ breast cancers.


Assuntos
Neoplasias da Mama/mortalidade , Transição Epitelial-Mesenquimal , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptor ErbB-2/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Xenoenxertos , Humanos , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Receptor ErbB-2/genética , Receptor Tirosina Quinase Axl
19.
Cancer Res ; 78(9): 2191-2204, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29440170

RESUMO

Metabolic reprogramming is a hallmark of cancer that includes increased glucose uptake and accelerated aerobic glycolysis. This phenotype is required to fulfill anabolic demands associated with aberrant cell proliferation and is often mediated by oncogenic drivers such as activated BRAF. In this study, we show that the MAPK-activated p90 ribosomal S6 kinase (RSK) is necessary to maintain glycolytic metabolism in BRAF-mutated melanoma cells. RSK directly phosphorylated the regulatory domain of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 2 (PFKFB2), an enzyme that catalyzes the synthesis of fructose-2,6-bisphosphate during glycolysis. Inhibition of RSK reduced PFKFB2 activity and glycolytic flux in melanoma cells, suggesting an important role for RSK in BRAF-mediated metabolic rewiring. Consistent with this, expression of a phosphorylation-deficient mutant of PFKFB2 decreased aerobic glycolysis and reduced the growth of melanoma in mice. Together, these results indicate that RSK-mediated phosphorylation of PFKFB2 plays a key role in the metabolism and growth of BRAF-mutated melanomas.Significance: RSK promotes glycolytic metabolism and the growth of BRAF-mutated melanoma by driving phosphorylation of an important glycolytic enzyme. Cancer Res; 78(9); 2191-204. ©2018 AACR.


Assuntos
Melanoma/genética , Fosfofrutoquinase-2/genética , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Proliferação de Células/genética , Reprogramação Celular/genética , Glucose/metabolismo , Glicólise/genética , Células HeLa , Humanos , Melanoma/metabolismo , Melanoma/patologia , Fosforilação
20.
Otolaryngol Head Neck Surg ; 158(4): 721-728, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29436285

RESUMO

Objectives Endolymphatic sac (ELS) pathophysiology in Ménière's disease (MD) remains poorly understood. We identified from the literature a group of proteins expressed on the ELS and involved in endolymph volume regulation: aquaporin-2 (AQP2), vasopressin receptor V2R, sodium potassium chloride cotransporter 2 (NKCC2), and transient receptor potential cation channel V4 (TRPV4). Our objective was to determine whether their ELS expression was altered in MD, to better understand the pathophysiology of endolymphatic hydrops. Study Design Prospective case-control study. Setting Tertiary care center. Subjects Twenty-four patients with definite MD undergoing endolymphatic duct blockage surgery were recruited, as well as 23 controls with no history of MD undergoing surgery for vestibular schwannoma (VS). Methods ELS biopsies and blood samples for plasma arginine vasopressin (AVP) were obtained. Immunohistochemistry for AQP2, V2R, NKCC2, and TRPV4 was performed. Slides were scanned digitally for highly sensitive pixel density analysis by specialized software (VIS; Visiopharm). Results Global scores generated by the software represent total and relative protein expression density of 3 staining intensity levels, exclusively on ELS epithelium. AQP2 expression density was significantly elevated in MD compared to VS ( P = .003). There was no significant difference in plasma AVP, V2R, NKCC2, and TRPV4 expression. Conclusion This original study evaluates simultaneous in situ expression of AQP2, V2R, NKCC2, and TRPV4 on the human ELS in MD, with a control group. Our results show only AQP2 upregulation on the ELS of patients with MD. We suggest a constitutively increased expression of AQP2 in MD, independent of its regulatory axis (AVP-V2R). Acquired regulator sequence mutations could support this model.


Assuntos
Aquaporina 2/metabolismo , Saco Endolinfático/metabolismo , Doença de Meniere/metabolismo , Doença de Meniere/fisiopatologia , Receptores de Vasopressinas/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Canais de Cátion TRPV/metabolismo , Biópsia , Estudos de Casos e Controles , Estudos Transversais , Feminino , Humanos , Imuno-Histoquímica , Masculino , Doença de Meniere/cirurgia , Pessoa de Meia-Idade , Estudos Prospectivos , Quebeque , Software
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...