Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Front Oncol ; 13: 1258474, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38304030

RESUMEN

Several breast cancer (BC) patients showed urinary tract infection after adjuvant trastuzumab plus paclitaxel, but no case of urothelial injury has been reported. In this case, we report a 47-year-old female patient with stage I invasive ductal carcinoma in the left breast presenting with urothelial injury after the combination of trastuzumab and paclitaxel. Initially, the patient was highly suspected of having urinary tract infection as she showed abdominal and low back pain, as well as urinary irritation symptoms and hematuria. Unfortunately, the conditions were not attenuated after anti-infection therapy. Contrast-enhanced CT showed extensive exudation and edema in the bilateral renal pelvis, ureter, and bladder, together with dilatation and effusion in the renal pelvis and ureter. Cystoscopy showed extensive congestion, edema, and erosion in the bladder epithelium. Pathological analysis demonstrated slight thinning or even loss in the uroepithelial cell layer and interstitial congestion. In addition, there was growth arrest in the epithelial cells. Immunohistochemistry indicated HER2 expression in the urothelial cells. Finally, the patient was diagnosed with urothelial injury after combination of paclitaxel and trastuzumab. The symptoms were spontaneously cured with no administration of any antibiotics in the 3-month follow-up.

2.
Oncol Lett ; 20(2): 1270-1280, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32724368

RESUMEN

Long non-coding RNAs (lncRNAs) are functional components of the human genome. Recent studies have demonstrated that lncRNAs play essential roles in tumorigenesis, and are involved in cell proliferation, apoptosis, migration and invasion in several types of tumor, including lung cancer. However, the clinical relevance of lncRNA expression in lung cancer remains unknown. The aim of the present study was to investigate the expression pattern of RFPL3 antisense (RFPL3S) and its associations with clinicopathological characteristics in patients with lung cancer. Whether RFPL3S can act as a potential prognostic biomarker for lung cancer was also investigated. RFPL3S expression in tumor samples and cells was assessed using the Oncomine database and the Cancer Cell Line Encyclopedia, respectively. Based on Kaplan-Meier Plotter analyses, the prognostic values of RFPL3S were further evaluated. It was revealed that RFPL3S was highly expressed in lung cancer tissues when compared with normal tissues and was significantly associated with pN factor, pTNM stage and Ki-67 labeling index. In the survival analyses, increased RFPL3S expression was associated with poor survival and was inversely associated with first progression in all patients. These results indicate that RFPL3S may be of clinical significance and may act as a prognostic biomarker in lung cancer.

3.
Int Immunopharmacol ; 56: 128-134, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29414642

RESUMEN

The knowledge about B7-H2 expression in tumor is growing, but many questions remain unresolved. Especially in human tumor microenvironment, little studies were done. To explore the expression and clinical significance of B7-H2 on T cells in colorectal cancer microenvironment, fresh tumor tissues and paired non-tumor tissues collected from 25 patients with colorectal cancer were made to research B7-H2 expression on the infiltrating T cells including CD8+ T cells and CD4+ T cells. Also, tumor bearing mice were sacrificed on day 5, day 10, day 15, day 20, day 25 and flow cytometry was used to analyze B7-H2 expression on CD8+ T cells and CD4+ T cells in mouse tumors and spleens. Then, it was found that B7-H2 expression on CD8+ T cells in patients' tumor tissues was significantly higher than in non-tumor tissues. The expression of B7-H2 on CD8+ T cells in tumor microenvironment was significantly higher in patients with age ≤60 years old and the stage I-II. The expression level of B7-H2 on CD8+ T cells in mouse tumors and spleens both reached the highest level at the early stage of inoculation (on day 5), decreased to the lowest level on day 10 and day 15 separately, and then gradually increased. In mouse spleens, B7-H2 expression on CD8+ T cells was all significantly higher than on CD4+ T cells in five time periods. So, in this study, it was found that B7-H2 expression on CD8+ T cells in tumor microenvironment was closely related to the progression of colorectal cancer.


Asunto(s)
Factores de Edad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Animales , Línea Celular Tumoral , Separación Celular , Neoplasias Colorrectales/patología , Femenino , Citometría de Flujo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Microambiente Tumoral
4.
J BUON ; 23(6): 1862-1866, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30610814

RESUMEN

PURPOSE: Esophageal cancer (EC) is the 9th most common carcinoma worldwide with poor prognosis. Specific biomarkers can help predicting the development of esophageal squamous cell carcinoma (ESCC), which can improve the assessment of prognosis. This study aimed to explore long noncoding RNA (lncRNA) ANRIL expression and its potential value in ESCC prognosis. METHODS: Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was utilized to detect lncRNA ANRIL expression in 50 pairs of ESCC and matched normal samples in order to explore the role of lncRNA ANRIL in ESCC. Moreover, the association was investigated between clinical characteristics of ESCC and the expression level of ANRIL. RESULTS: Disease-free survival (DFS) and overall survival (OS) were significantly shorter in ESCC patients with higher expression level of lncRNA ANRIL. ESCC tissues examined showed an obvious increment in ANRIL expression when compared to normal tissues. Furthermore, ANRIL was positively related to lymph nodes metastasis, TNM stage and tumor clinical stage. Moreover, upregulated ANRIL expression was remarkably associated with shorter survival in ESCC patients,which was also an independent prognostic factor for both OS and DFS. CONCLUSIONS: This study suggested that lncRNA ANRIL could be a potential oncogene of ESCC. ANRIL expression might be served as another potential therapeutic target and prognostic biomarker for ESCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/secundario , Neoplasias Esofágicas/patología , Esofagectomía/mortalidad , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/cirugía , Estudios de Casos y Controles , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/cirugía , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Regulación hacia Arriba
5.
Immunopharmacol Immunotoxicol ; 39(3): 124-130, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28303727

RESUMEN

CONTEXT: Shikonin is a kind of naphthoquinone compound found mainly in Lithospermum erythrorhizon Sieb,et Zucc. Previous studies have shown that Shikonin has anti-tumor, anti-inflammatory and extensive pharmacological effects. According to new studies, Shikonin could also modulate the immune system function, but the effect to NK (nature killer) cells is yet unknown. OBJECTIVE: To investigate the effect and mechanism of Shikonin on NK cells proliferation and cytotoxicity to colon cancer cell line (Caco-2). METHODS: The proliferation and cytotoxicity of NK cells cultured with Shikonin were detected with CCK-8 assay. The expressions of perforin, GranB and IFN-γ were examined with FCM. The content of TNF-alpha was disclosed with ELISA kit. p-ERK1/2 and p-Akt expression of NK cells were detected with western blot. RESULTS: With CCK-8 assay, it is found that Shikonin could significantly enhance NK cells proliferation and cytotoxicity to colon cancer cells. With FCM assay, it is found that Shikonin could improve the expression of perforin and GranB in a dose-dependent manner. Shikonin had no effect on TNF-alpha and IFN-γ expression. In mechanism, the study shows that Shikonin could enhance the expression of p-ERK1/2 and p-Akt. CONCLUSIONS: Shikonin enhances NK cells proliferation and cytotoxicity via the improvement of perforin, GranB, p-ERK1/2 and p-Akt expression.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/inmunología , Inmunidad Celular/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Naftoquinonas/farmacología , Células CACO-2 , Neoplasias del Colon/patología , Humanos , Células Asesinas Naturales , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 1 Activada por Mitógenos/inmunología , Proteína Quinasa 3 Activada por Mitógenos/inmunología , Proteínas Proto-Oncogénicas c-akt/inmunología
6.
Oncotarget ; 8(3): 4572-4581, 2017 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-27992364

RESUMEN

MicroRNAs (miRNAs) play critical roles in variousbiological processes,including malignancy. Here, we demonstrated that miR-30e levels were markedly reduced in human lung carcinoma specimens in comparisonwith adjacent normal tissues.In addition, miR-30eamounts were starkly lower in the resistant PC9/gefitinib (PC9G) cancer cells compared with PC9 cells. Meanwhile, miR-30eoverexpression inPC9G cells resulted in reduced cell proliferation and migration,reversing drug resistance to gefitinib.Conversely,miR-30e silencing in PC9 cells increased proliferation as well as migration, and conferred resistance to gefitinib.Moreover, HOXA1, which was identified asa new miR-30etarget, plays important roles in regulating cell fate, early developmental patterns and organogenesis.Importantly, miR-30ealso inhibited PC9G growth in vivo. Taken together, these findings demonstrated that miR-30eshould be considered a tumor suppressor miRNA, which could be used in treatinghuman lung cancer.


Asunto(s)
Antineoplásicos/administración & dosificación , Resistencia a Antineoplásicos , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/patología , MicroARNs/genética , Quinazolinas/administración & dosificación , Factores de Transcripción/genética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Ratones , Trasplante de Neoplasias , Quinazolinas/farmacología
7.
Clin Immunol ; 136(3): 329-37, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20472506

RESUMEN

Recent studies and our current data demonstrated the deficits in the numbers and/or functions of the CD4(+)CD25(+)Foxp3(+) Treg cells in the patients with autoimmune diseases, indicating that restoration of Treg cells in these patients could be a potential therapeutic approach. Here, we demonstrated that CD4(+)CD25(+)Foxp3(+) Treg cells can be purified, activated and expanded from peripheral blood of patients with immune-mediated diseases, to a similar degree to those from healthy donors. Within 3weeks, Treg cells from most patients could be expanded ex vivo 100-2000 fold and maintained their phenotypic characteristics. Furthermore, ex vivo expanded Treg cells displayed potent and enhanced in vitro suppressive activities inhibiting T effector cell proliferation compared to Treg cells freshly purified from the same patients. The expanded Treg cells with enhanced biological function may provide an opportunity to restore the proper balance of immunity and tolerance, suggesting the potential of using Treg cell therapy for treatment of immune-mediated diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/terapia , Inflamación/inmunología , Inflamación/terapia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Artritis Reumatoide/inmunología , Artritis Reumatoide/terapia , Asma/inmunología , Asma/terapia , Estudios de Casos y Controles , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/terapia , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/terapia , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Técnicas In Vitro , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/terapia , Activación de Linfocitos , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/terapia , Autotolerancia , Trasplante Autólogo
8.
Immunol Rev ; 219: 118-42, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17850486

RESUMEN

The dendritic cell (DC) system of antigen-presenting cells controls immunity and tolerance. DCs initiate and regulate immune responses in a manner that depends on signals they receive from microbes and their cellular environment. They allow the immune system to make qualitatively distinct responses against different microbial infections. DCs are composed of subsets that express different microbial receptors and express different surface molecules and cytokines. Our studies lead us to propose that interstitial (dermal) DCs preferentially activate humoral immunity, whereas Langerhans cells preferentially induce cellular immunity. Alterations of the DC system result in diseases such as autoimmunity, allergy, and cancer. Conversely, DCs can be exploited for vaccination, and novel vaccines that directly target DCs in vivo are being designed.


Asunto(s)
Autoinmunidad , Citocinas/metabolismo , Células Dendríticas/inmunología , Inmunidad , Infecciones/inmunología , Neoplasias/inmunología , Animales , Presentación de Antígeno , Citocinas/inmunología , Células Dendríticas/metabolismo , Humanos , Inmunidad Innata , Lectinas Tipo C/inmunología , Lectinas Tipo C/metabolismo , Proteínas Adaptadoras de Señalización NOD/inmunología , Proteínas Adaptadoras de Señalización NOD/metabolismo , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
9.
Eur J Immunol ; 37(9): 2657-67, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17683111

RESUMEN

Dendritic cells (DC) have a unique capacity to present external antigens to CD8(+) T cells, i.e. cross-presentation. However, it is not fully established whether the ability to cross-presentation is restricted to a unique subset of DC in humans. Here, we show that two major myeloid DC subsets, i.e. Langerhans cells (LC) and interstitial DC (Int-DC), have the ability to cross-present antigens to CD8(+) T cells in vitro. LC and Int-DC were obtained from DC generated by culturing human CD34(+)-hematopoietic progenitor cells with GM-CSF, FLT3-L, and TNF-alpha (CD34-DC). Both DC subsets were able to capture necrotic/apoptotic allogeneic melanoma cells and present antigens to CD8(+) T cells, resulting in efficient priming of naive CD8(+) T cells into CTL capable of killing melanoma cells. Strikingly, a single stimulation with either subset (LC or Int-DC) or total CD34-DC loaded with necrotic/apoptotic melanoma cells was sufficient to activate melanoma-specific memory CD8(+) T cells obtained from patients with metastatic melanoma to become effective CTL. Thus, this study provides the rationale to use CD34-DC loaded with necrotic/apoptotic allogeneic melanoma cells in a clinical trial.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Células de Langerhans/inmunología , Melanoma/inmunología , Linfocitos T Citotóxicos/inmunología , Antígenos CD34/inmunología , Antígenos CD34/metabolismo , Apoptosis , Diferenciación Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Humanos , Activación de Linfocitos , Melanoma/patología , Necrosis/patología
10.
J Immunol ; 176(4): 2134-41, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16455969

RESUMEN

Dendritic cells (DCs) loaded with killed allogeneic melanoma cells can cross-prime naive CD8(+) T cells to differentiate into melanoma-specific CTLs in 3-wk cultures. In this study we show that DCs loaded with killed melanoma cells that were heated to 42 degrees C before killing are more efficient in cross-priming of naive CD8(+) T cells than DCs loaded with unheated killed melanoma cells. The enhanced cross-priming was demonstrated by several parameters: 1) induction of naive CD8(+) T cell differentiation in 2-wk cultures, 2) enhanced killing of melanoma peptide-pulsed T2 cells, 3) enhanced killing of HLA-A*0201(+) melanoma cells in a standard 4-h chromium release assay, and 4) enhanced capacity to prevent tumor growth in vitro in a tumor regression assay. Two mechanisms might explain the hyperthermia-induced enhanced cross-priming. First, heat-treated melanoma cells expressed increased levels of 70-kDa heat shock protein (HSP70), and enhanced cross-priming could be reproduced by overexpression of HSP70 in melanoma cells transduced with HSP70 encoding lentiviral vector. Second, hyperthermia resulted in the increased transcription of several tumor Ag-associated Ags, including MAGE-B3, -B4, -A8, and -A10. Thus, heat treatment of tumor cells permits enhanced cross-priming, possibly via up-regulation of both HSPs and tumor Ag expression.


Asunto(s)
Reactividad Cruzada/inmunología , Hipertermia Inducida , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Antígenos de Neoplasias/metabolismo , Apoptosis , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP70 de Choque Térmico/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patología , Datos de Secuencia Molecular , Linfocitos T Citotóxicos/química , Linfocitos T Citotóxicos/metabolismo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA