Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.124
Filtrar
1.
Pathol Res Pract ; 226: 153596, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34481213

RESUMEN

Growth arrest specific 6 (GAS6) encodes a protein that serves as a ligand for AXL receptor tyrosine kinase and stimulates cell proliferation. Notably, an antisense RNA, namely GAS6-AS1 is transcribed from chromosome 13q34, near GAS6 gene. In vitro functional experiments have demonstrated that GAS6-AS1 can promote proliferation, migration and invasive properties of transformed cells through enhancing entry into S-phase. Notably, mechanistic investigations have shown that GAS6-AS1 can regulate expression of GAS6 at the transcriptional or translational stages through constructing a RNA-RNA duplex, thus enhancing expression of AXL and inducing AXL signaling. Both GAS6 and its antisense transcript contribute in the pathogenesis of human malignancies. In the current review, we provide a summary of studies that appraised the role of these genes in the carcinogenesis.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , ARN sin Sentido/fisiología , Animales , Carcinogénesis/metabolismo , Proliferación Celular/fisiología , Humanos , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Tirosina Quinasa del Receptor Axl
2.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33947152

RESUMEN

Previously, we have revealed that the miR-130 family (miR-130b, miR-301a, and miR-301b) functions as an oncomiR in bladder cancer. The pharmacological inhibition of the miR-130 family molecules by the seed-targeting strategy with an 8-mer tiny locked nucleic acid (LNA) inhibits the growth, migration, and invasion of bladder cancer cells by repressing stress fiber formation. Here, we searched for a functionally advanced target sequence with LNA for the miR-130 family with low cytotoxicity and found LNA #9 (A(L)^i^i^A(L)^T(L)^T(L)^G(L)^5(L)^A(L)^5(L)^T(L)^G) as a candidate LNA. LNA #9 inhibited cell growth in vitro and in an in vivo orthotopic bladder cancer model. Proteome-wide tyrosine phosphorylation analysis suggested that the miR-130 family upregulates a wide range of receptor tyrosine kinases (RTKs) signaling via the expression of phosphorylated Src (pSrcTyr416). SILAC-based proteome analysis and a luciferase assay identified protein tyrosine phosphatase non-receptor type 1 (PTPN1), which is implicated as a negative regulator of multiple signaling pathways downstream of RTKs as a target gene of the miR-130 family. The miR-130-targeted LNA increased and decreased PTPN1 and pSrcTyr416 expressions, respectively. PTPN1 knockdown led to increased tumor properties (cell growth, invasion, and migration) and increased pSrcTyr416 expression in bladder cancer cells, suggesting that the miR-130 family upregulates multiple RTK signaling by targeting PTPN1 and subsequent Src activation in bladder cancer. Thus, our newly designed miR-130 family targeting LNA could be a promising nucleic acid therapeutic agent for bladder cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , MicroARNs/antagonistas & inhibidores , Proteínas de Neoplasias/fisiología , Oligonucleótidos/uso terapéutico , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , ARN Neoplásico/antagonistas & inhibidores , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Carcinoma de Células Transicionales/tratamiento farmacológico , Carcinoma de Células Transicionales/genética , Carcinoma de Células Transicionales/metabolismo , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Ratones , MicroARNs/genética , ARN Neoplásico/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Recombinantes/metabolismo , Regulación hacia Arriba , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Heart Lung Transplant ; 40(6): 435-446, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846079

RESUMEN

Cardiac Allograft Vasculopathy (CAV) is a leading contributor to late transplant rejection. Although implicated, the mechanisms by which bone marrow-derived cells promote CAV remain unclear. Emerging evidence implicates the cell surface receptor tyrosine kinase AXL to be elevated in rejecting human allografts. AXL protein is found on multiple cell types, including bone marrow-derived myeloid cells. The causal role of AXL from this compartment and during transplant is largely unknown. This is important because AXL is a key regulator of myeloid inflammation. Utilizing experimental chimeras deficient in the bone marrow-derived Axl gene, we report that Axl antagonizes cardiac allograft survival and promotes CAV. Flow cytometric and histologic analyses of Axl-deficient transplant recipients revealed reductions in both allograft immune cell accumulation and vascular intimal thickness. Co-culture experiments designed to identify cell-intrinsic functions of Axl uncovered complementary cell-proliferative pathways by which Axl promotes CAV-associated inflammation. Specifically, Axl-deficient myeloid cells were less efficient at increasing the replication of both antigen-specific T cells and vascular smooth muscle cells (VSMCs), the latter a key hallmark of CAV. For the latter, we discovered that Axl-was required to amass the VSMC mitogen Platelet-Derived Growth Factor. Taken together, our studies reveal a new role for myeloid Axl in the progression of CAV and mitogenic crosstalk. Inhibition of AXL-protein, in combination with current standards of care, is a candidate strategy to prolong cardiac allograft survival.


Asunto(s)
Células de la Médula Ósea/patología , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Trasplante de Corazón/efectos adversos , Músculo Liso Vascular/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Ecocardiografía , Citometría de Flujo , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/metabolismo , Supervivencia de Injerto , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Músculo Liso Vascular/patología , Miocitos Cardíacos/patología , Miocitos del Músculo Liso , Proteínas Proto-Oncogénicas/biosíntesis , ARN/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Trasplante Homólogo , Tirosina Quinasa del Receptor Axl
4.
Leuk Res ; 94: 106383, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32460059

RESUMEN

Metformin is an anti-diabetic drug known to have anticancer activity by inhibiting mechanistic target of rapamycin (mTOR); however, other molecular mechanisms may also be involved. In this study, we examined the effects of metformin on the activity of receptor tyrosine kinases of the TAM (TYRO3, AXL, and MERTK) family, which have important roles in leukemia cell growth. The results indicated that metformin suppressed the in vitro growth of four leukemia cell lines, OCI/AML2, OCI/AML3, THP-1, and K562, in a dose-dependent manner, which corresponded to the downregulation of the expression and phosphorylation of AXL and inhibition of its downstream targets such as phosphorylation of STAT3. Furthermore, metformin augmented the suppressive effects of a small-molecule AXL inhibitor TP-0903 on the growth of OCI/AML3 and K562 cells and prevented doxorubicin-induced AXL activation in K562 cells, which induces chemoresistance in leukemia cells, thus potentiating doxorubicin anti-proliferative effects. Given that metformin also downregulated expression of TYRO3 and phosphorylation of MERTK, these findings indicate that anti-leukemic effects exerted by metformin could be partly due to the inhibition of TAM kinases. Thus, metformin has a clinical potential for patients with leukemia cells positive for AXL and the other TAM proteins as well as activated mTOR.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia , Metformina/farmacología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Humanos , Células K562 , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Leucemia/patología , Células THP-1 , Tirosina Quinasa del Receptor Axl
5.
J Neuroinflammation ; 17(1): 68, 2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075650

RESUMEN

BACKGROUND: CD8+ T lymphocytes are critical mediators of neuroinflammatory diseases. Understanding the mechanisms that govern the function of this T cell population is crucial to better understanding central nervous system autoimmune disease pathology. We recently identified a novel population of highly cytotoxic c-Met-expressing CD8+ T lymphocytes and found that hepatocyte growth factor (HGF) limits effective murine cytotoxic T cell responses in cancer models. Here, we examined the role of c-Met-expressing CD8+ T cells by using a MOG35-55 T cell-mediated EAE model. METHODS: Mice were subcutaneously immunized with myelin oligodendrocyte glycoprotein peptide (MOG)35-55 in complete Freund's adjuvant (CFA). Peripheral and CNS inflammation was evaluated at peak disease and chronic phase, and c-Met expression by CD8 was evaluated by flow cytometry and immunofluorescence. Molecular, cellular, and killing function analysis were performed by real-time PCR, ELISA, flow cytometry, and killing assay. RESULTS: In the present study, we observed that a fraction of murine effector CD8+ T cells expressed c-Met receptor (c-Met+CD8+) in an experimental autoimmune encephalitis (EAE) model. Phenotypic and functional analysis of c-Met+CD8+ T cells revealed that they recognize the encephalitogenic epitope myelin oligodendrocyte glycoprotein37-50. We demonstrated that this T cell population produces higher levels of interferon-γ and granzyme B ex vivo and that HGF directly restrains the cytolytic function of c-Met+CD8+ T cells in cell-mediated cytotoxicity reactions CONCLUSIONS: Altogether, our findings suggest that the HGF/c-Met pathway could be exploited to modulate CD8+ T cell-mediated neuroinflammation.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Secuencia de Aminoácidos , Animales , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Femenino , Adyuvante de Freund/toxicidad , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/toxicidad , Proteínas Tirosina Quinasas Receptoras/genética
6.
Pathol Res Pract ; 215(11): 152662, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31575452

RESUMEN

Small molecule inhibitors such as Larotrectinib have been recently approved in the treatment of patients with fusions of the neurotrophic-tropomyosin receptor tyrosine kinase (NTRK) genes 1-3. These genomic rearrangements have been reported across different tumor subtypes with a high prevalence in rare tumors. However, in gastric and esophageal adenocarcinoma (AGE) NTRK fusions have also been described in a subset of Asian patients. In order to study the prevalence of this alteration in Caucasian patients with AGE we performed immunohistochemistry for pan-NTRK in 438 formalin-fixed paraffin embedded (FFPE) tumor samples. While we found NTRK expression in gastric glands and tumor adjacent nerve tissue, we did not detect this marker in the tumor compartment. Based on our findings NTRK fusions do not seem to play a role in the molecularpathology of Caucasian AEG patients, so that other treatment options are required.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Esofágicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Neoplasias Gástricas/genética , Adenocarcinoma/enzimología , Adulto , Anciano , Neoplasias Esofágicas/enzimología , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Fusión de Oncogenes , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Neoplasias Gástricas/enzimología
7.
Prostate ; 79(12): 1422-1438, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31269290

RESUMEN

BACKGROUND: We previously identified a panel of five microRNAs (miRNAs) associated with biochemical recurrence and metastasis following prostatectomy from prostate cancer patients using next-generation sequencing-based whole miRNome sequencing and quantitative polymerase chain reaction-based validation analysis. In this study, we examined the mechanism of action of miR-139-5p, one of the downregulated miRNAs identified in the panel. METHODS: Using a cohort of 585 patients treated with radical prostatectomy, we examined the prognostic significance of miR-139 (dichotomized around the median) using the Kaplan-Meier method and Cox proportional hazard models. We validated these results using The Cancer Genome Atlas (TCGA) data. We created cell lines that overexpressed miR-139 to confirm its targets as well as examine pathways through which miR-139 may function using cell-based assays. RESULTS: Low miR-139 expression was significantly associated with a variety of prognostic factors in prostate cancer, including Gleason score, pathologic stage, margin positivity, and lymph node status. MiR-139 expression was associated with prognosis: the cumulative incidence of biochemical recurrence and metastasis were significantly lower among patients with high miR-139 expression (P = .0004 and .038, respectively). Validation in the TCGA data set showed a significant association between dichotomized miR-139 expression and biochemical recurrence (odds ratio, 0.52; 95% confidence interval, 0.33-0.82). Overexpression of miR-139 in prostate cancer cells led to a significant reduction in cell proliferation and migration compared with control cells, with cells arrested in G2 of cell cycle. IGF1R and AXL were identified as potential targets of miR-139 based on multiple miRNA-binding sites in 3'-untranslated regions of both the genes and their association with prostate cancer growth pathways. Luciferase assays verified AXL and IGF1R as direct targets of miR-139. Furthermore, immunoblotting of prostate cancer cells demonstrated IGF1R and AXL protein expression were inhibited by miR-139 treatment, which was reversed by the addition of miR-139 antagomir. Examination of the molecular mechanism of growth inhibition by miR-139 revealed the downregulation of activated AKT and cyclin D1, with upregulation of the CDK inhibitor p21. CONCLUSIONS: miR-139 is associated with improved prognosis in patients with localized prostate cancer, which may be mediated through downregulation of IGF1R and/or AXL and associated signaling pathway components.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , MicroARNs/biosíntesis , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptor IGF Tipo 1/metabolismo , Anciano , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Próstata/patología , Neoplasias de la Próstata/patología , Transducción de Señal , Tirosina Quinasa del Receptor Axl
8.
Haematologica ; 104(10): 2017-2027, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30923103

RESUMEN

Internal tandem duplication in Fms-like tyrosine kinase 3 (FLT3-ITD) is the most frequent mutation observed in acute myeloid leukemia (AML) and correlates with poor prognosis. FLT3 tyrosine kinase inhibitors are promising for targeted therapy. Here, we investigated mechanisms dampening the response to the FLT3 inhibitor quizartinib, which is specific to the hematopoietic niche. Using AML primary samples and cell lines, we demonstrate that convergent signals from the hematopoietic microenvironment drive FLT3-ITD cell resistance to quizartinib through the expression and activation of the tyrosine kinase receptor AXL. Indeed, cytokines sustained phosphorylation of the transcription factor STAT5 in quizartinib-treated cells, which enhanced AXL expression by direct binding of a conserved motif in its genomic sequence. Likewise, hypoxia, another well-known hematopoietic niche hallmark, also enhanced AXL expression. Finally, in a xenograft mouse model, inhibition of AXL significantly increased the response of FLT3-ITD cells to quizartinib exclusively within a bone marrow environment. These data highlight a new bypass mechanism specific to the hematopoietic niche that hampers the response to quizartinib through combined upregulation of AXL activity. Targeting this signaling offers the prospect of a new therapy to eradicate resistant FLT3-ITD leukemic cells hidden within their specific microenvironment, thereby preventing relapses from FLT3-ITD clones.


Asunto(s)
Benzotiazoles/farmacología , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda/metabolismo , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Factor de Transcripción STAT5/metabolismo , Microambiente Tumoral , Tirosina Quinasa 3 Similar a fms/metabolismo , Hipoxia de la Célula , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Factor de Transcripción STAT5/genética , Regulación hacia Arriba/efectos de los fármacos , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa del Receptor Axl
9.
Pathol Int ; 69(2): 94-96, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30707464

RESUMEN

Anti-TRK targeted therapies offer opportunities to treat patients with advanced NTRK1/2/3-rearranged cancers. Beyond NTRK-rearranged secretory breast carcinomas, little is known about NTRK rearrangements and the expression of TRK proteins in non-secretory breast carcinomas. We search for TRK proteins expressions using pan-TRK immunohistochemistry and NTRK1, NTRK2 and NTRK3 rearrangements using fluorescent in situ hybridization (FISH) tests in a set of tissue microarray included breast carcinomas. Only 1/339 invasive breast carcinomas, the only example of secretory subtype, was positive using pan-TRK immunohistochemistry and harboured a NTRK-rearrangement (NTRK1 positive FISH test). According to our results, druggable NTRK rearrangements and related-TRK proteins expression are not encountered in non-secretory breast carcinomas.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinoma/metabolismo , Carcinoma/patología , Femenino , Reordenamiento Génico , Humanos , Persona de Mediana Edad
10.
Cell Prolif ; 52(1): e12511, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30311693

RESUMEN

OBJECTIVES: Aptamer sgc8c is a short DNA sequence that can target protein tyrosine kinase 7 (PTK7), which was overexpressed on many tumour cells. This study aimed to fabricate a novelty DNA nanostructure drug delivery system target on PTK7-positive cells-CCRF-CEM (human T-cell ALL). METHODS: Aptamer-modified tetrahedron DNA was synthesized through one-step thermal annealing process. The sgc8c-TDNs (s-TDNs) loading DOX complexes were applied to investigate the effect to PTK7-negative and -positive cells. RESULTS: When s-TDN:DOX acted on PTK7-positive and -negative cells respectively, the complexes exhibited specific toxic effect on PTK7-positive cells but not on PTK7-negative Ramos cells in vitro research. CONCLUSIONS: In this work, we successfully constructed a PTK7-targeting aptamer-guided DNA tetrahedral nanostructure (s-TDN) as a drug delivery system via a facile one-pot synthesis method. The results showed that s-TDN:DOX exhibited enhanced cytotoxicity against PTK7-positive CCRF-CEM cells, with a minor effect against PTK7-negative Ramos cells. Hence, this functionalized TDNs drug delivery system displayed its potential application in targeting PTK7-positive tumour T-cell acute lymphoblastic leukaemia.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Moléculas de Adhesión Celular/biosíntesis , ADN/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Nanoestructuras/química , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Secuencia de Bases/genética , Transporte Biológico/fisiología , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Proteínas Tirosina Quinasas Receptoras/genética
11.
Gynecol Oncol ; 152(2): 408-415, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30448261

RESUMEN

OBJECTIVE: The objective of this study was to analyze the expression level and clinical role of soluble AXL (sAXL) in cancers affecting the serosal surfaces, with focus on ovarian carcinoma. METHODS: sAXL protein expression by ELISA was analyzed in 572 effusion supernatants, including 424 peritoneal, 147 pleural and 1 pericardial specimens. RESULTS: sAXL was overexpressed in peritoneal effusions compared to pleural and pericardial specimens (p < 0.001). sAXL levels were additionally significantly higher in effusions from patients with ovarian carcinoma, malignant mesothelioma and breast carcinoma compared to specimens from patients with other cancers (predominantly carcinomas of lung, gastrointestinal or uterine corpus/cervix origin) or benign reactive effusions (p < 0.001). sAXL was further overexpressed in high-grade serous carcinoma (HGSC; n = 373) compared to low-grade serous carcinoma (LGSC; n = 32; p = 0.036). In HGSC, sAXL levels were significantly lower in post-chemotherapy effusions compared to primary diagnosis pre-chemotherapy specimens (p = 0.002). sAXL levels in HGSC were unrelated to chemoresponse at diagnosis, progression-free survival or overall survival. Levels were similarly unrelated to survival in LGSC and breast carcinoma. CONCLUSIONS: sAXL is widely expressed in malignant effusions, particularly in ovarian and breast carcinoma and in malignant mesothelioma. sAXL is overexpressed in HGSC compared to LGSC and its levels are lower following exposure to chemotherapy. However, sAXL levels are not informative of chemoresponse or survival.


Asunto(s)
Líquido Ascítico/enzimología , Neoplasias de la Mama/enzimología , Cistadenocarcinoma Seroso/enzimología , Neoplasias Pulmonares/enzimología , Mesotelioma/enzimología , Neoplasias Ováricas/enzimología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Líquido Ascítico/efectos de los fármacos , Líquido Ascítico/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/patología , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Mesotelioma/tratamiento farmacológico , Mesotelioma/patología , Mesotelioma Maligno , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Adulto Joven , Tirosina Quinasa del Receptor Axl
12.
Gynecol Oncol ; 151(2): 337-344, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30190114

RESUMEN

OBJECTIVE: Paclitaxel, a microtubule inhibitor, is subject to tumor resistance while treating high-grade serous ovarian and uterine cancer. This study aims to directly compare the effects of SQ1274, a novel microtubule inhibitor that binds to the colchicine-binding site on tubulin, and paclitaxel in high-grade serous ovarian and uterine cancer cell lines both in vitro and in vivo. METHODS: We assessed the sensitivity of ovarian (OVCAR8) and uterine (ARK1) cancer cell lines to SQ1274 and paclitaxel using XTT assays. We used western blot and quantitative real-time PCR to analyze changes in AXL RNA and protein expression by SQ1274 and paclitaxel. Differences in cell-cycle arrest and apoptosis were investigated using flow cytometry. Finally, we treated ovarian and uterine xenograft models with vehicle, paclitaxel, or SQ1274. RESULTS: First, we demonstrate that SQ1274 has a much lower IC50 than paclitaxel in both ARK1 (1.26 nM vs. 15.34 nM, respectively) and OVCAR8 (1.34 nM vs. 10.29 nM, respectively) cancer cell lines. Second, we show SQ1274 decreases both RNA and protein expression of AXL. Third, we show that SQ1274 causes increased cell-cycle arrest and apoptosis compared to paclitaxel. Finally, we report that SQ1274 more effectively inhibits tumor growth in vivo compared to paclitaxel. CONCLUSIONS: SQ1274 presents as a viable alternative to paclitaxel for treating ovarian and uterine cancer. This study supports the development of SQ1274 as a chemotherapeutic to treat ovarian and uterine cancer.


Asunto(s)
Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Moduladores de Tubulina/farmacología , Animales , Apoptosis/efectos de los fármacos , Carcinoma Epitelial de Ovario , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
13.
Gene ; 674: 170-177, 2018 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-29953919

RESUMEN

Ribosome: machinery in control of messenger RNA's (mRNAs) and several ribosomal proteins are in the small and large subunit of the ribosome. Various aspects of ribosomal proteins have related to cell growth, cell cycle, and diseases. Ribosomal protein L10A (RpL10A) in shrimp and fruit fly has been demonstrated to play a role in oogenesis. Interestingly, deletion RpL10A gene (RpL10Ab-/-) germ line clone of the fruit fly showed a loss of follicle cells surrounding the egg chamber, but nurse cells appeared normal. This phenotype is reminiscent of insulin receptor mutants (InR-/-). In contrast, over-expression of RpL10A in the eyes of the fruit flies resulted in abnormal ommatidia with a loss of red pigment in the center of the eyes. In this study, the abnormal rearrangements of nuclei and lack of cell membranes in those eyes were demonstrated. Furthermore, the expression of InR gene and the InR protein were extensively increased as determined by real-time PCR and immunohistochemistry, respectively. In addition, some insulin signaling mediators were also detected. The Akt and FOXO proteins were highly phosphorylated in the RpL10A over-expressed mutant. The results revealed that RpL10A induced over-expression of the insulin receptor and consequently activated in insulin signaling pathway which affects cell proliferation and we suggest RpL10A stimulates cell proliferation via the insulin signaling pathway.


Asunto(s)
Insulina/fisiología , Proteínas Ribosómicas/metabolismo , Transducción de Señal , Animales , Metabolismo de los Hidratos de Carbono , Membrana Celular/ultraestructura , Núcleo Celular/ultraestructura , Proteínas de Drosophila/biosíntesis , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Ojo/ultraestructura , Fosforilación , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Ribosómicas/genética
14.
Sci Rep ; 8(1): 8519, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29867084

RESUMEN

Protein tyrosine kinase 7 (PTK7), also known as colon carcinoma kinase 4 (CCK-4), is a member of the catalytically defective receptor protein tyrosine kinase family and is upregulated in various cancers, where it is known to act as either an oncoprotein or a tumor suppressor. To understand the contrasting roles of PTK7 in tumorigenesis, we analyzed the tumorigenic characteristics of esophageal squamous cell carcinoma (ESCC) cells with low levels of endogenous PTK7 expression (TE-5 and TE-14 cells) and high levels of expression (TE-6 and TE-10 cells) after transfections with a PTK7 expression vector. PTK7 overexpression increased the proliferation of TE-5 and TE-14 cells but decreased the proliferation of TE-6 and TE-10 cells. In the ESCC cells, proliferation, migration, and invasion were initially increased and then decreased according to PTK7 expression levels, which were mirrored by initial increases and then decreases in the tyrosine phosphorylation of cellular proteins and phosphorylation of Src, Akt, and ERK. In ESCC patients included in The Cancer Genome Atlas database, those with higher PTK7 mRNA levels had a longer overall survival and lower relative risk than those with lower PTK7 mRNA levels. These results demonstrate that PTK7 biphasically regulates tumorigenesis in ESCC.


Asunto(s)
Carcinogénesis/metabolismo , Carcinoma de Células Escamosas/enzimología , Moléculas de Adhesión Celular/biosíntesis , Neoplasias Esofágicas/epidemiología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Carcinogénesis/genética , Carcinogénesis/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Proliferación Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Humanos , Proteínas de Neoplasias/genética , Proteínas Tirosina Quinasas Receptoras/genética
15.
Life Sci ; 202: 103-109, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29604268

RESUMEN

AIMS: Existing evidence emphasize the role of mitochondrial dysfunction in sarcopenia which is revealed as loss of skeletal muscle mass and neuromuscular junction remodeling. We assessed the effect of low-intensity aerobic training along with blood flow restriction on muscle hypertrophy index, muscle-specific kinase (MuSK), a pivotal protein of the neuromuscular junction and Peroxisome proliferator-activated receptor gamma co-activator 1-alpha (PGC-1α) in aged male rats. MAIN METHODS: Animals groups were control (CTL), sham (Sh), leg blood flow restriction (BFR), exercise (Ex), sham + exercise (Sh + Ex), and BFR plus exercise (BFR + Ex) groups. The exercise groups were trained with low intensity exercise for 10 weeks. 48 h after the last training session, animals were sacrificed under anesthesia. Soleus and EDL muscles were isolated, hypertrophy index was estimated and MuSK and PGC-1α were measured by western blot method. KEY FINDINGS: Hypertrophy index enhanced in soleus and Extensor digitorum longus (EDL) muscles of BFR + Ex group (P < 0.01 versus CTL and Sh groups, and P < 0.001 versus other groups). The MuSK protein of soleus and EDL muscles increased in BFR + Ex group (P < 0.01 and P < 0.001, respectively) in comparison with CTL and Sh groups. In BFR + Ex group, the PGC-1α protein increased in both soleus and EDL (P < 0.001 compared to other groups). Also the PGC-1α of soleus muscle was higher in Ex and Sh + Ex groups versus CTL and Sh groups (P < 0.05). SIGNIFICANCE: Findings suggest that low endurance exercise plus BFR improves the MuSK and hypertrophy index of both slow and fast muscles of elderly rats probably through the rise of PGC-1α expression.


Asunto(s)
Fibras Musculares de Contracción Rápida/metabolismo , Fibras Musculares de Contracción Lenta/metabolismo , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/crecimiento & desarrollo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/fisiología , Condicionamiento Físico Animal/fisiología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Aerobiosis , Envejecimiento/fisiología , Animales , Miembro Posterior/irrigación sanguínea , Hipertrofia , Masculino , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/genética , Flujo Sanguíneo Regional
16.
Mol Med Rep ; 17(5): 6253-6260, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29488611

RESUMEN

MicroRNAs (miRNAs) are small non­coding RNAs, which are critical in a diverse range of biological processes, including development, differentiation, homeostasis, and in the formation of diseases by accelerating and/or inhibiting the translation of mRNAs. The present study aimed to examine the potential role of miRNA (miR)­205­5p in the developmental process of colorectal cancer (CRC) through protein­tyrosine kinase 7 (PTK7). Initially, TargetScan was used to predict the miRNA target sites in the sequence of the PTK7 3'­untranslated region. It was then found that the mRNA expression level of miR­205­5p was lower in CRC cells, determined using reverse transcription­quantitative polymerase chain reaction analysis, and there was a negative correlation between miR­205­5p and PTK7 in CRC tissues. It was also found that miR­205­5p regulated the gene transcription of PTK7, determined using a luciferase reporter assay. The results of RT­qPCR and western blot analyses in human colorectal cancer revealed that miR­205­5p suppressed the expression of PTK7. Finally, it was revealed that miR­205­5p restricted the proliferation ability of CRC cells through inhibiting PTK7, which was determined using colony forming and 3­(4,5­dimethylthiazol­2­yl)­2,5­diphenyltetrazolium bromide assays. miR­205­5p accelerated cell apoptosis through inhibiting PTK7, demonstrated using Annexin V­FITC/propidium iodide staining. The results of a Transwell assay indicated that miR­205­5p inhibited the migration and invasion abilities of CRC cells through inhibiting PTK7. Therefore, miR­205­5p is involved in the proliferation, migration and invasion of CRC through inhibiting PTK7.


Asunto(s)
Moléculas de Adhesión Celular/biosíntesis , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/biosíntesis , ARN Neoplásico/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Humanos , MicroARNs/genética , Invasividad Neoplásica , Proteínas de Neoplasias/genética , ARN Neoplásico/genética , Proteínas Tirosina Quinasas Receptoras/genética
17.
Neurochem Int ; 118: 233-241, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29572052

RESUMEN

Malignant glioma is an aggressive type of brain tumor with poor prognosis and mostly incurable. Although cisplatin is used for adjuvant chemotherapy against glioma, intrinsic and acquired resistance restricts the application of cisplatin. Long noncoding RNA (lncRNA) DANCR is reported to regulate the differentiation and progression of several cancers. However, whether DANCR participates in cisplatin resistance of glioma is still unknown. In this study, we found that DANCR expression was negatively correlated with cisplatin sensitivity in glioma cells. Gain-of and loss-of function assays revealed that DNACR attenuated cisplatin-induced cell proliferation inhibition in vitro and xenograft growth suppression in vivo. Furthermore, DNACR also attenuated cisplatin-induced cell apoptosis in vitro and in vivo. Mechanistically, we found that DANCR upregulated AXL via competitively binding miR-33a-5p, miR-33b-5p, miR-1-3p, miR-206, and miR-613. Through upregulating AXL, DANCR activated PI3K/Akt/NF-κB signaling pathway in glioma cells. Inhibiting AXL/PI3K/Akt/NF-κB signaling pathway reversed the effects of DANCR on cisplatin resistance. In conclusion, we identified a cisplatin-resistance associated lncRNA DANCR. DANCR promotes cisplatin resistance via activating AXL/PI3K/Akt/NF-κB signaling pathway in glioma. Our data suggested that DANCR would be a potential biomarker for predicting cisplatin sensitivity and a therapeutic target for enhancing cisplatin efficacy in glioma.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Cisplatino/farmacología , Resistencia a Antineoplásicos/fisiología , Glioma/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , ARN Largo no Codificante/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Animales , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Línea Celular Tumoral , Cisplatino/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Glioma/tratamiento farmacológico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
18.
J Med Econ ; 21(6): 577-586, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29458286

RESUMEN

AIMS: To assess the cost-effectiveness of first-line ceritinib vs crizotinib and platinum doublet chemotherapy for anaplastic lymphoma kinase (ALK)-positive metastatic non-small cell lung cancer (NSCLC) from a US third-party payer's perspective. MATERIALS AND METHODS: A partitioned survival model with three health states (stable disease, progressive disease, death) was developed over a 20-year time horizon. Ceritinib's efficacy inputs (progression-free and overall survival) were estimated from ASCEND-4; parametric survival models extrapolated data beyond the trial period. The relative efficacy of ceritinib vs chemotherapy was obtained from ASCEND-4, the relative efficacy of ceritinib vs crizotinib was estimated using a matching-adjusted indirect comparison based on ASCEND-4 and PROFILE 1014. Drug acquisition, treatment administration, adverse event management, and medical costs were obtained from publicly available databases and the literature, and inflated to 2016 US dollars. Treatment-specific stable-state utilities were derived from trials and progressive-state utility from the literature. Incremental costs per quality-adjusted life year (QALY) were estimated for ceritinib vs each comparator. Cost-effectiveness was assessed based on US willingness-to-pay thresholds. Deterministic and probabilistic sensitivity analyses were performed to test model robustness. RESULTS: In the base case, first-line ceritinib was associated with total direct costs of $299,777 and 3.28 QALYs (from 4.61 life years gained [LYG]) over 20 years. First-line crizotinib and chemotherapy were associated with 2.73 and 2.41 QALYs, 3.92 and 3.53 LYG, and $263,172 and $228,184 total direct costs, respectively. The incremental cost per QALY gained was $66,064 for ceritinib vs crizotinib and $81,645 for ceritinib vs chemotherapy. In the first 2 years following treatment initiation, ceritinib dominated crizotinib by conferring greater health benefits at reduced total costs. Results were robust to deterministic and probabilistic sensitivity analyses. LIMITATIONS: In the absence of head-to-head trials, an indirect comparison method was used. CONCLUSIONS: Ceritinib is cost-effective compared to crizotinib and chemotherapy in the treatment of previously untreated ALK-positive metastatic NCSLC in the US.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Sulfonas/uso terapéutico , Quinasa de Linfoma Anaplásico , Antineoplásicos/efectos adversos , Antineoplásicos/economía , Protocolos de Quimioterapia Combinada Antineoplásica/economía , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Costo de Enfermedad , Análisis Costo-Beneficio , Crizotinib , Supervivencia sin Enfermedad , Humanos , Modelos Econométricos , Pirazoles/efectos adversos , Pirazoles/economía , Piridinas/efectos adversos , Piridinas/economía , Pirimidinas/efectos adversos , Pirimidinas/economía , Años de Vida Ajustados por Calidad de Vida , Sulfonas/efectos adversos , Sulfonas/economía , Análisis de Supervivencia , Estados Unidos
19.
Fetal Pediatr Pathol ; 37(2): 117-125, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29336647

RESUMEN

BACKGROUND: Inflammatory myofibroblastic tumor (IMT) is an uncommon mesenchymal lesion composed of myofibroblastic and fibroblastic spindle cells, accompanied by inflammatory infiltration. IMT may occur in the tongue. Five cases have been previously reported at this site. CASE REPORT: An inflammatory myofibroblastic tumor arose in the tongue of a 10 month old infant, confirmed by anaplastic lymphoma kinase (ALK) immunohistochemical staining and the clinical response to the tyrosine kinase inhibitor. CONCLUSIONS: IMT can occur in the tongue. This report highlights the differential diagnosis of IMT. ALK staining is both a helpful diagnostic marker and a predictive marker for targeted therapy in this tumor type.


Asunto(s)
Neoplasias de Tejido Muscular/patología , Neoplasias de la Lengua/patología , Quinasa de Linfoma Anaplásico , Biomarcadores de Tumor/análisis , Humanos , Lactante , Inflamación/patología , Masculino , Proteínas Tirosina Quinasas Receptoras/análisis , Proteínas Tirosina Quinasas Receptoras/biosíntesis
20.
Cancer Res Treat ; 50(2): 495-505, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28546523

RESUMEN

PURPOSE: In this study, anaplastic lymphoma kinase (ALK) mutation and amplification, ALK protein expression, loss of the nuclear alpha thalassemia/mental retardation syndrome X-linked (ATRX) protein, and telomerase reverse transcriptase (TERT) protein expressionwere studied to investigate potential correlations between these molecular characteristics and clinical features or outcomes in neuroblastoma. MATERIALS AND METHODS: Seventy-two patients were enrolled in this study. Polymerase chain reaction amplification and direct sequencing were used for mutation analysis. ALK and MYCN amplifications were detected by fluorescence in situ hybridization. Protein expressionwas evaluated by immunohistochemical (IHC) staining. RESULTS: ALK mutation was found in only two patients (4.1%); ALK amplification was not detected. ALK positivity, loss of nuclear ATRX protein, TERT positivity by IHC were detected in 40 (55.6%), nine (13.0%), and 42 (59.2%) patients, respectively. The incidence of ALK expression increased in accordance with increasing tumor stage (p=0.001) and risk group (p < 0.001). The relapse rate was significantly higher in ALK+ patients compared to that of other patients (47.5% vs. 11.3%, p=0.007). However, there was no significant difference in relapse rate when the survival analysis was confined to the high-risk patients. CONCLUSION: Although ALK mutation was rare and no amplification was observed, ALK protein expression was found in a significant number of patients and was correlated with advanced stage and high-risk neuroblastoma. ALK protein expression could be considered as a marker related to the aggressive neuroblastoma, but it was not the independent prognostic factor for the outcome.


Asunto(s)
Neuroblastoma/enzimología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Adolescente , Quinasa de Linfoma Anaplásico , Niño , Preescolar , Femenino , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Masculino , Neuroblastoma/genética , Neuroblastoma/patología , Pronóstico , Proteínas Tirosina Quinasas Receptoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA