Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Vis ; 28: 192-202, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36274817

RESUMEN

Purpose: Retinoblastoma (RB) is a malignant childhood intraocular tumor. Current treatment options for RB have undesirable side effects. A comprehensive understanding of gene expression in human RB is essential for the development of safe and effective new therapies. Methods: We reviewed published microarray and RNA sequencing studies in which gene expression profiles were compared between human RB and normal retina tissues. We investigated the expression of genes of interest using quantitative reverse transcription PCR. We examined the activities of cloned promoter DNA fragments with luciferase assay. Results: Dopachrome tautomerase (DCT) was among the most overexpressed genes in RB in published studies. We found that DCT was highly expressed in six of 13 samples microdissected from Thai RB tissues. Expression of DCT was absent or barely detected in retina tissues, various human ocular cells, and major organs. We also demonstrated that the -657 to +411 DCT promoter fragment efficiently directs RB cell-specific transcription of the luciferase reporter gene in cell lines. Conclusions: The present work highlights that DCT is one of the most RB-specific genes. The regulatory elements required for this cell-specific gene expression are likely located within its proximal promoter.


Asunto(s)
Oxidorreductasas Intramoleculares , Neoplasias de la Retina , Retinoblastoma , Niño , Humanos , Regulación Neoplásica de la Expresión Génica , Genes de Retinoblastoma/genética , Oxidorreductasas Intramoleculares/genética , Oxidorreductasas Intramoleculares/metabolismo , Neoplasias de la Retina/genética , Retinoblastoma/genética , Retinoblastoma/patología
2.
Breast Cancer Res Treat ; 186(1): 25-36, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33389403

RESUMEN

PURPOSE: Treatment of breast cancer (BC) by standard methods is effective in the early stage, but ineffective in the advanced stage of disease. To develop an adoptive T cell therapy for advanced and severe BC, we generated fourth-generation chimeric antigen receptor (CAR) T cells targeting folate receptor alpha antigen (FRα) expressed on BC cells, and preclinically evaluated their anti-BC activities. METHODS: The fourth-generation FRα-CAR T cells containing extracellular FRα-specific single-chain variable fragment (scFv) and three intracellular costimulatory domains (CD28, 4-1BB, and CD27) linked to CD3ζ were generated using a lentiviral system, and then were evaluated for their anti-BC activities in two-dimensional and three-dimensional (spheroid) cultures. RESULTS: When our fourth-generation FRα-CAR T cells were cocultured with FRα-expressing MDA-MB-231 BC cell line at an effector to target ratio of 20:1, these CAR T cells specifically lysed 88.7 ± 10.6% of the target cells. Interestingly, the cytotoxic lysis of FRα-CAR T cells was more pronounced in target cells with higher surface FRα expression. This specific cytotoxicity of the CAR T cells was not observed when cocultured with FRα-negative MCF10A normal breast-like cell line at the same ratio (34.3 ± 4.7%). When they were cocultured with MDA-MD-231 spheroid, the FRα-CAR T cells exhibited antitumor activity marked with spheroid size reduction and breakage. CONCLUSION: This proof-of-concept study thus shows the feasibility of using these fourth-generation FRα-CAR T cells for adoptive T cell therapy in BC.


Asunto(s)
Neoplasias de la Mama , Receptores Quiméricos de Antígenos , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Tratamiento Basado en Trasplante de Células y Tejidos , Femenino , Receptor 1 de Folato/genética , Humanos , Inmunoterapia Adoptiva , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Phytother Res ; 32(8): 1546-1554, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29672960

RESUMEN

Thai herbal antipyretic 22 formula (APF22), a polyherbal formula, has been traditionally used to treat dermatologic problems including hyperpigmentation. Exposure of the skin to ultraviolet A (UVA) causes abnormal melanin production induced by photooxidative stress. This study thus aimed to investigate the protective effects of APF22 extracts and phenolic compounds, ferulic acid (FA), and gallic acid (GA; used as positive control and reference compounds), on melanogenesis through modulation of nuclear factor E2-related factor 2 (Nrf2) signaling and antioxidant defenses in mouse melanoma (B16F10) cells exposed to UVA. Our results revealed that the APF22 extracts, FA, and GA reduced melanin synthesis as well as activity and protein levels of tyrosinase in UVA-irradiated B16F10 cells. Moreover, APF22 extracts and both FA and GA were able to activate Nrf2-antioxidant response element signaling and promote antioxidant defenses including glutathione, catalase, glutathione peroxidase, and the glutathione-S-transferase at both mRNA and enzyme activity levels in irradiated cells. In conclusion, APF22 extracts suppressed UVA-mediated melanogenesis in B16F10 cells possibly via redox mechanisms involving activation of Nrf2 signaling and upregulation of antioxidant defenses. Moreover, pharmacological action of the APF22 extracts may be attributed to the phenolic compounds, FA, and GA, probably serving as the APF22's active compounds.


Asunto(s)
Antipiréticos/farmacología , Melaninas/biosíntesis , Extractos Vegetales/farmacología , Rayos Ultravioleta , Animales , Elementos de Respuesta Antioxidante , Antioxidantes/metabolismo , Catalasa/metabolismo , Línea Celular Tumoral , Ácidos Cumáricos/farmacología , Ácido Gálico/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Melanoma Experimental , Ratones , Monofenol Monooxigenasa/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Fenoles/farmacología , Piel/efectos de los fármacos , Tailandia
4.
Mol Vis ; 22: 342-51, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27122965

RESUMEN

PURPOSE: Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous group of inherited retinal degenerations characterized by progressive loss of photoreceptor cells and RPE functions. More than 70 causative genes are known to be responsible for RP. This study aimed to identify the causative gene in a patient from a consanguineous family with childhood-onset severe retinal dystrophy. METHODS: To identify the defective gene, whole exome sequencing was performed. Candidate causative variants were selected and validated using Sanger sequencing. Segregation analysis of the causative gene was performed in additional family members. To verify that the mutation has an effect on protein synthesis, an expression vector containing the first ten amino acids of the mutant protein fused with the DsRed2 fluorescent protein was constructed and transfected into HEK293T cells. Expression of the fusion protein in the transfected cells was measured using fluorescence microscopy. RESULTS: By filtering against public variant databases, a novel homozygous missense mutation (c.3G>A) localized in the start codon of the MERTK gene was detected as a potentially pathogenic mutation for autosomal recessive RP. The c.3G>A mutation cosegregated with the disease phenotype in the family. No expression of the first ten amino acids of the MerTK mutant fused with the DsRed2 fluorescent protein was detected in HEK293T cells, indicating that the mutation affects the translation initiation site of the gene that may lead to loss of function of the MerTK signaling pathway. CONCLUSIONS: We report a novel missense mutation (c.3G>A, p.0?) in the MERTK gene that causes severe vision impairment in a patient. Taken together with previous reports, our results expand the spectrum of MERTK mutations and extend our understanding of the role of the MerTK protein in the pathogenesis of retinitis pigmentosa.


Asunto(s)
Codón Iniciador/genética , Mutación Missense , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Retinitis Pigmentosa/genética , Adulto , Consanguinidad , Exoma/genética , Angiografía con Fluoresceína , Células HEK293 , Humanos , Masculino , Linaje , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/metabolismo , Análisis de Secuencia de ADN , Tomografía de Coherencia Óptica , Transfección , Tirosina Quinasa c-Mer
5.
J Med Assoc Thai ; 98(6): 549-54, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26219158

RESUMEN

BACKGROUND: Polymorphisms in cytochrome P450 2C9 and vitamin K epoxide reductase complex, subunit 1 genes (CYP2C9 and VKORC1, respectively) were previously shown to affect the warfarin dose required in anticoagulant therapy of deep vein thrombosis (DVT). However, little is known about the role of these genetic variants in the Thai population. OBJECTIVE: To identify the effect of CYP2C9 and VKORC1 genetic variants on warfarin dosage in the Thai population with DVT. MATERIAL AND METHOD: Genotyping of CYP2C9 (*2 and *3) and VKORC1 promoter (-1 639G>A) variants were carried out in 97 Thai DVT patients receiving constant warfarin therapy and with a stable international normalized ratio using real-time PCR assays. RESULTS: VKORC1 AA, GA, and GG genotype frequencies were found to be 49.5%, 46.4%, and 4.1%, respectively, while those of CYP2C9 genotypes were 88.7% for *1/*1 and 11.3%for *1/*3. The CYP2C9*2 variant was not present in the patients studied. The mean daily warfarin dose required to maintain a therapeutic INR differed significantly according to VKORC1 genotype, with 3.6 mg/day required for AA, 4.7 mg/day for GA, and 7.4 mg/day for GG (p-value < 0.001). The CYP2C9 genotype did not significantly affect the warfarin dosage requirement (p-value = 0.29). CONCLUSION: These findings underline the impact of VKORC1 genotypes on the wide variation in warfarin maintenance dosing in Thai patients with DVT.


Asunto(s)
Anticoagulantes/administración & dosificación , Trombosis de la Vena/tratamiento farmacológico , Vitamina K Epóxido Reductasas/genética , Warfarina/administración & dosificación , Adulto , Anciano , Pueblo Asiatico/genética , Citocromo P-450 CYP2C9/genética , Femenino , Variación Genética , Genotipo , Humanos , Relación Normalizada Internacional , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Reacción en Cadena en Tiempo Real de la Polimerasa , Trombosis de la Vena/genética
6.
Exp Mol Pathol ; 96(1): 85-91, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24370549

RESUMEN

Mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene predict benefit from tyrosine kinase inhibitors in patients suffering from non-small-cell lung cancer. In this study, we developed a fast, simple, cost-effective and highly sensitive assay for detection of five clinically important EGFR mutations in exon 19 (2235_2249del and 2236_2250del), exon 20 (C2369T) and exon 21 (T2573G and c.2573_2574 TG > GT). We designed EGFR mutation detection assays by combining allele-specific PCR amplification with the detection of SYBR Green I fluorescence, and optimized PCR conditions to specifically amplify mutant alleles. These one-step assays were able to detect the mutations at levels as low as 1.5 mutant copies in a DNA sample. Commercially available probe-based allele-specific PCR exhibited relatively poor performance when detecting very low copies of mutated DNA, especially in exon 19 and 20. Our assays offered dramatically less reagent cost than that of the commercial kit and generated results in less than 90 min after DNA extraction. These protocols can also be applied to conventional thermal cyclers followed by gel electrophoresis detection.


Asunto(s)
Análisis Mutacional de ADN/métodos , Receptores ErbB/genética , Neoplasias Pulmonares/diagnóstico , Proteínas Mutantes/genética , Mutación/genética , Reacción en Cadena de la Polimerasa/métodos , Alelos , Humanos , Neoplasias Pulmonares/genética
7.
Int Immunopharmacol ; 129: 111631, 2024 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-38359664

RESUMEN

The treatment of breast cancer (BC) remains a formidable challenge due to the emergence of drug resistance, necessitating the exploration of innovative strategies. Chimeric antigen receptor (CAR)-T cell therapy, a groundbreaking approach in hematologic malignancies, is actively under investigation for its potential application in solid tumors, including BC. Trophoblast cell surface antigen 2 (Trop2) has emerged as a promising immunotherapeutic target in various cancers and is notably overexpressed in BC. To enhance therapeutic efficacy in BC, a fourth-generation CAR (CAR4) construct was developed. This CAR4 design incorporates an anti-Trop2 single-chain variable fragment (scFv) fused with three costimulatory domains -CD28/4-1BB/CD27, and CD3ζ. Comparative analysis with the conventional second-generation CAR (CAR2; 28ζ) revealed that anti-Trop2 CAR4 T cells exhibited heightened cytotoxicity and interferon-gamma (IFN-γ) production against Trop2-expressing MCF-7 cells. Notably, anti-Trop2 CAR4-T cells demonstrated superior long-term cytotoxic functionality and proliferative capacity. Crucially, anti-Trop2 CAR4-T cells displayed specific cytotoxicity against Trop2-positive BC cells (MDA-MB-231, HCC70, and MCF-7) in both two-dimensional (2D) and three-dimensional (3D) culture systems. Following antigen-specific killing, these cells markedly secreted interleukin-2 (IL-2), tumor necrosis factor-alpha (TNF-α), IFN-γ, and Granzyme B compared to non-transduced T cells. This study highlights the therapeutic potential of anti-Trop2 CAR4-T cells in adoptive T cell therapy for BC, offering significant promise for the advancement of BC treatment strategies.


Asunto(s)
Neoplasias de la Mama , Receptores de Antígenos de Linfocitos T , Humanos , Femenino , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T , Inmunoterapia Adoptiva/métodos , Interferón gamma/metabolismo , Línea Celular Tumoral
8.
Pharmaceuticals (Basel) ; 17(2)2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38399413

RESUMEN

Cholangiocarcinoma (CCA) is a difficult-to-treat cancer, with limited therapeutic options and surgery being the only curative treatment. Standard chemotherapy involves gemcitabine-based therapies combined with cisplatin, oxaliplatin, capecitabine, or 5-FU with a dismal prognosis for most patients. Receptor tyrosine kinases (RTKs) are aberrantly expressed in CCAs encompassing potential therapeutic opportunity. Hence, 112 RTK inhibitors were screened in KKU-M213 cells, and ceritinib, an approved targeted therapy for ALK-fusion gene driven cancers, was the most potent candidate. Ceritinib's cytotoxicity in CCA was assessed using MTT and clonogenic assays, along with immunofluorescence, western blot, and qRT-PCR techniques to analyze gene expression and signaling changes. Furthermore, the drug interaction relationship between ceritinib and cisplatin was determined using a ZIP synergy score. Additionally, spheroid and xenograft models were employed to investigate the efficacy of ceritinib in vivo. Our study revealed that ceritinib effectively killed CCA cells at clinically relevant plasma concentrations, irrespective of ALK expression or mutation status. Ceritinib modulated multiple signaling pathways leading to the inhibition of the PI3K/Akt/mTOR pathway and activated both apoptosis and autophagy. Additionally, ceritinib and cisplatin synergistically reduced CCA cell viability. Our data show ceritinib as an effective treatment of CCA, which could be potentially explored in the other cancer types without ALK mutations.

9.
Cancers (Basel) ; 15(4)2023 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-36831441

RESUMEN

Colorectal cancers (CRC) with KRAS mutations (KRASmut) are frequently included in consensus molecular subtype 3 (CMS3) with profound metabolic deregulation. We explored the transcriptomic impact of KRASmut, focusing on the tumor microenvironment (TME) and pathways beyond metabolic deregulation. The status of KRASmut in patients with CRC was investigated and overall survival (OS) was compared with wild-type KRAS (KRASwt). Next, we identified CMS, and further investigated differentially expressed genes (DEG) of KRASmut and distinctive pathways. Lastly, we used spatially resolved gene expression profiling to define the effect of KRASmut in the TME regions of CMS3-classified CRC tissues. CRC patients with KRASmut were mainly enriched in CMS3. Their specific enrichments of immune gene signatures in immunosuppressive TME were associated with worse OS. Activation of TGFß signaling by KRASmut was related to reduced pro-inflammatory and cytokine gene signatures, leading to suppression of immune infiltration. Digital spatial profiling in TME regions of KRASmut CMS3-classified tissues suggested up-regulated genes, CD40, CTLA4, ARG1, STAT3, IDO, and CD274, that could be characteristic of immune suppression in TME. This study may help to depict the complex transcriptomic profile of KRASmut in immunosuppressive TME. Future studies and clinical trials in CRC patients with KRASmut should consider these transcriptional landscapes.

10.
Front Oncol ; 12: 813009, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35242708

RESUMEN

BACKGROUND: Current guidelines recommend anti-epidermal growth factor receptor monoclonal antibodies (anti-EGFR Ab) as first-line treatment only in patients with left-sided RAS wild type (RASwt) metastatic colorectal cancer (mCRC). However, there are no guideline recommendations specific to tumor sidedness in subsequent-line treatment. This study aimed to investigate the effect of primary tumor location on second- or later-line treatment outcomes in patients with KRASwt mCRC. METHODS: Medical records of patients diagnosed with mCRC at 3 academic centers in Thailand (Siriraj, Chulalongkorn, and Ramathibodi hospital) between 2008 and 2019 were retrospectively reviewed. Patients with KRASwt mCRC who received anti-EGFR Ab in second- or later-line treatment were included. The impact of tumor sidedness on progression-free survival (PFS) was determined using Kaplan-Meier method, and those results were compared using log-rank test. RESULTS: Among the 2,102 patients who had KRAS analysis data, 1,130 (54%) patients had KRASwt. Of those, 413 patients received anti-EGFR Ab in second- or later-line treatment. One hundred and sixty-two of 413 (39%) patients had extended RAS analysis. Seventy (17%) patients had right-sided tumors. Two hundred and thirty-eight (58%) patients received anti-EGFR Ab in the third line, and 132 (32%) patients and 43 (10%) patients were treated in the second and more than third line, respectively. Single-agent irinotecan was the most commonly used backbone chemotherapy (303/413, 73%). Patients with right-sided tumors had non-significantly inferior PFS compared to patients with left-sided tumors (median PFS: 5.7 months (mo), 95% confidence interval [CI]: 3.9-7.5 vs. 7.5 mo, 95% CI 6.5-8.5; p=0.17). Subgroup analysis showed no difference in PFS when stratified by treatment lines. Patient with right-sided tumors had significantly inferior OS compared to patients with left-sided tumors (median OS: 23.3 mo vs. 29.9 mo; p=0.005). CONCLUSIONS: To date, this is the largest real world data of the effect of primary tumor location on anti-EGFR Ab which demonstrated that tumor sidedness has no significant impact on treatment outcomes in KRASwt mCRC patients receiving second- or later-line therapy. Our findings do not support the utility of tumor sidedness for treatment selection in these settings. We confirmed that patients with right-sided tumors had significantly worse survival.

11.
Sci Rep ; 12(1): 2167, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35140316

RESUMEN

Despite the development of predictive biomarkers to shape treatment paradigms and outcomes, de novo EGFR TKI resistance advanced non-small cell lung cancer (NSCLC) remains an issue of concern. We explored clinical factors in 332 advanced NSCLC who received EGFR TKI and molecular characteristics through 65 whole exome sequencing of various EGFR TKI responses including; de novo (progression within 3 months), intermediate response (IRs) and long-term response (LTRs) (durability > 2 years). Uncommon EGFR mutation subtypes were significantly variable enriched in de novo resistance. The remaining sensitizing EGFR mutation subtypes (exon 19 del and L858R) accounted for 75% of de novo resistance. Genomic landscape analysis was conducted, focusing in 10 frequent oncogenic signaling pathways with functional contributions; cell cycle, Hippo, Myc, Notch, Nrf2, PI-3-Kinase/Akt, RTK-RAS, TGF-ß, p53 and ß-catenin/Wnt signaling. Cell cycle pathway was the only significant alteration pathway among groups with the FDR p-value of 6 × 10-4. We found only significant q-values of < 0.05 in 7 gene alterations; CDK6, CCNE1, CDK4, CCND3, MET, FGFR4 and HRAS which enrich in de novo resistance [range 36-73%] compared to IRs/LTRs [range 4-22%]. Amplification of CDK4/6 was significant in de novo resistance, contrary to IRs and LTRs (91%, 27.9% and 0%, respectively). The presence of co-occurrence CDK4/6 amplification correlated with poor disease outcome with HR of progression-free survival of 3.63 [95% CI 1.80-7.31, p-value < 0.001]. The presence of CDK4/6 amplification in pretreatment specimen serves as a predictive biomarker for de novo resistance in sensitizing EGFR mutation.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Anciano , Biomarcadores , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Femenino , Amplificación de Genes , Genes erbB-1 , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Resultado del Tratamiento
12.
Sci Rep ; 11(1): 6276, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737613

RESUMEN

Current treatments for cholangiocarcinoma (CCA) are largely unsuccessful due to late diagnosis at advanced stage, leading to high mortality rate. Consequently, improved therapeutic approaches are urgently needed. Chimeric antigen receptor (CAR) T cell therapy is a newly potential therapy that can recognize specific surface antigen without major histocompatibility complex (MHC) restriction. Mucin 1 (MUC1) is an attractive candidate antigen as it is highly expressed and associated with poor prognosis and survival in CCA. We, therefore, set forth to create the fourth-generation CAR (CAR4) construct containing anti-MUC1-single-chain variable fragment (scFv) and three co-stimulatory domains (CD28, CD137, and CD27) linked to CD3ζ and evaluate anti-MUC1-CAR4 T cells in CCA models. Compared to untransduced T cells, anti-MUC1-CAR4 T cells produced increased levels of TNF-α, IFN-γ and granzyme B when exposed to MUC1-expressing KKU-100 and KKU-213A CCA cells (all p < 0.05). Anti-MUC1-CAR4 T cells demonstrated specific killing activity against KKU-100 (45.88 ± 7.45%, p < 0.05) and KKU-213A cells (66.03 ± 3.14%, p < 0.001) at an effector to target ratio of 5:1, but demonstrated negligible cytolytic activity against immortal cholangiocytes. Furthermore, the anti-MUC1-CAR4 T cells could effectively disrupt KKU-213A spheroids. These activities of anti-MUC1-CAR4 T cells supports the development of this approach as an adoptive T cell therapeutic strategy for CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/terapia , Trasplante de Células/métodos , Colangiocarcinoma/inmunología , Colangiocarcinoma/terapia , Inmunoterapia Adoptiva/métodos , Mucina-1/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Técnicas de Cocultivo , Citocinas/biosíntesis , Células HEK293 , Humanos , Células MCF-7 , Mucina-1/metabolismo , Receptores Quiméricos de Antígenos/genética , Anticuerpos de Cadena Única/inmunología , Esferoides Celulares/inmunología , Transfección , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
13.
Front Oncol ; 11: 802876, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35117999

RESUMEN

Second-generation anti-CD19-chimeric antigen receptor T cells (anti-CD19-CAR2 T cells) are effective for treating B-cell malignancies; however, anti-CD19-CAR2 T cells can induce human anti-mouse immune responses because anti-CD19 single-chain variable fragment (scFv) in the CAR molecules is derived from a murine FMC63 (mFMC63) monoclonal antibody. Consequently, the persistence of mFMC63-CAR2 T cells and their therapeutic efficiency in patients are decreased, which results in tumor relapse. In an attempt to remedy this shortcoming, we generated a new anti-CD19-CAR T cells containing fully human anti-CD19 scFv (Hu1E7-CAR4 T cells) to pre-clinically evaluate and compare with mFMC63-CAR4 T cells. The human anti-CD19 scFv (Hu1E7) was isolated from a human scFv phage display library and fused to the hinge region of CD8α, the transmembrane domain of CD28, three intracellular costimulatory domains (CD28, 4-1BB, and CD27), and a CD3ζ signaling domain (28BB27ζ). Compared to mFMC63-CAR2 T cells (BBζ) and mFMC63-CAR3 (BB27ζ), the mFMC63-CAR4 T cells (28BB27ζ) exerted superior anti-tumor activity against Raji (CD19+) target cell. The Hu1E7-CAR4 and mFMC63-CAR4 T cells demonstrated comparable cytotoxicity and proliferation. Interestingly, compared to mFMC63-CAR4 T cells, the Hu1E7-CAR4 T cells secreted lower levels of cytokines (IFN-γ and TNF-α), which may be due to the lower binding affinity of Hu1E7-CAR4 T cells. These findings demonstrated the successfulness in creation of a new CAR T cells containing a novel fully human-derived scFv specific to CD19+ cancer cells. In vivo studies are needed to further compare the anti-tumor efficacy and safety of Hu1E7-CAR4 T cells and mFMC63-CAR4 T cells.

14.
Oncol Rep ; 46(6)2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34651665

RESUMEN

Breast cancer cell lines are widely used as an in vitro system with which to study the mechanisms underlying biological and chemotherapeutic resistance. In the present study, two novel breast cancer cell lines designated as PC­B­142CA and PC­B­148CA were successfully established from HER2­positive and triple­negative (TN) breast cancer tissues. The cell lines were characterized by cytokeratin (CK), α­smooth muscle actin (α­SMA), fibroblast­activation protein (FAP) and programmed death­ligand 1 (PD­L1). Cell proliferation was assessed using a colony formation assay, an MTS assay, 3­dimensional (3­D) spheroid and 3­D organoid models. Wound healing and Transwell migration assays were used to explore the cell migration capability. The responses to doxorubicin (DOX) and paclitaxel (PTX) were evaluated by 3­D spheroids. The results showed that the PC­B­142CA and PC­B­148CA cell lines were α­SMA­negative, FAP­negative, CK­positive and PD­L1­positive. Both cell lines were adherent with the ability of 3­D­multicellular spheroid and organoid formations; invadopodia were found in the spheroids/organoids of only PC­B­148CA. PC­B­142CA had a faster proliferative but lower metastatic rate compared to PC­B­148CA. Compared to MDA­MB­231, a commercial TN breast cancer cell line, PC­B­148CA had a similar CD44+/CD24­ stemness property (96.90%), whereas only 8.75% were found in PC­B­142CA. The mutations of BRCA1/2, KIT, PIK3CA, SMAD4, and TP53 were found in PC­B­142CA cells related to the resistance of several drugs, whereas PC­B­148CA had mutated BRCA2, NRAS and TP53. In conclusion, PC­B­142CA can serve as a novel HER2­positive breast cancer cell line for drug resistance studies; while PC­B­148CA is a novel TN breast cancer cell line suitable for metastatic and stemness­related properties.


Asunto(s)
Línea Celular Tumoral/patología , Fragmentos de Péptidos , Receptor ErbB-2 , Neoplasias de la Mama Triple Negativas/patología , Movimiento Celular , Proliferación Celular , Doxorrubicina/farmacología , Femenino , Humanos , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Organoides/patología , Paclitaxel/farmacología , Esferoides Celulares/patología , Células Tumorales Cultivadas/patología
15.
Mol Genet Genomic Med ; 8(7): e1256, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32329244

RESUMEN

BACKGROUND: To review the performance of noninvasive prenatal screening (NIPS) using targeted single-nucleotide polymorphisms (SNPs) approach in mixed-risk Thai women. METHODS: Retrospective analysis of data for detection of trisomy 21 (T21), 18 (T18), 13 (T13), monosomy X (XO), other sex chromosome aneuploidies (SCA), and triploidy/vanishing twins (VT) from a single commercial laboratory. RESULTS: Mean (±SD) gestational age and maternal weight were 13.2 (±2.1) weeks and 125.7 (±22.4) pounds, respectively (n = 8,572). From 462/8,572 (5.4%) no-calls; 1/462 (0.2%) was uninformative SNPs, and 1/462 chose amniocentesis. Redraw settled 323/460 (70%) samples with low fetal fraction (FF); and 8,434/8,572 (98.4%) were finally reportable, with 131 high risks (1.6%). The median (min-max) FF of reportable (n = 8,434) and unreportable samples (n = 137) samples were 10.5% (2.6-37.9) and 3.8% (1-14.1), respectively (p < .05). Fetal karyotypes were available in 106/131 (80.9%) and 52/138 (37.7%) high risk and repeated no-calls, respectively. The positive predictive values (PPVs) for T21 (n = 47), T18 (n = 15), T13 (n = 7), XO (n = 8), other SCA (n = 7), and triploidy/VT were 94%, 100%, 58.3%, 66.7%, 70%, and 57.1%, respectively. None of repeated no-calls had aneuploidies. CONCLUSION: SNP-based NIPS has high PPVs for T21 and T18. Although the proprietary SNPs library is not population-specific, uninformative SNPs are uncommon.


Asunto(s)
Trastornos de los Cromosomas/diagnóstico , Pruebas Prenatales no Invasivas/normas , Polimorfismo de Nucleótido Simple , Adulto , Trastornos de los Cromosomas/genética , Femenino , Humanos , Pruebas Prenatales no Invasivas/métodos , Embarazo , Sensibilidad y Especificidad , Tailandia
16.
Int Immunopharmacol ; 89(Pt B): 107069, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33242709

RESUMEN

Current treatment of cholangiocarcinoma (CCA) - a lethal bile duct cancer - is ineffective because the disease is usually diagnosed at late and advanced stage. Thus, a novel therapeutic modality is urgently required. Fourth-generation chimeric antigen receptor (CAR4) T cells was created to target CD133, a well-known cancer stem cell marker, that is highly expressed and associates with cancer progression. The anti-CD133-CAR4 T cells showed high efficacy against CD133-expressing CCA cells. Tumour cell lysis occurred in a dose- and CD133 antigen-dependent manner, and significantly higher, up to 57.59% ± 9.62 at effector to target ratio of 5:1 in a CCA cell line - KKU-213A cells, compared to mock control (p = 0.008). Similarly, significant IFN-γ (p = 0.011) and TNF-α (p = 0.002) upregulation was observed upon tumour treatment. The effectiveness of our anti-CD133-CAR4 T cells will be beneficial not only for CD133-expressing CCA, but also for other CD133-expressing tumours. This study may guide future in vivo study and clinical trials.


Asunto(s)
Antígeno AC133/metabolismo , Neoplasias de los Conductos Biliares/terapia , Colangiocarcinoma/terapia , Inmunoterapia Adoptiva , Células Madre Neoplásicas/metabolismo , Anticuerpos de Cadena Única/metabolismo , Linfocitos T/metabolismo , Antígeno AC133/inmunología , Neoplasias de los Conductos Biliares/inmunología , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Antígenos CD28/genética , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Complejo CD3/genética , Complejo CD3/inmunología , Complejo CD3/metabolismo , Línea Celular Tumoral , Colangiocarcinoma/inmunología , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Humanos , Interferón gamma/metabolismo , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Esferoides Celulares , Linfocitos T/inmunología , Linfocitos T/trasplante , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Int Immunopharmacol ; 78: 106006, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31780372

RESUMEN

Cholangiocarcinoma (CCA) can resist chemotherapy resulting in treatment failure. Gemcitabine, a chemotherapeutic drug, can sensitize cancer cells to become susceptible to cytotoxic T-lymphocytes (CTLs). We, therefore, hypothesized that a combination of gemcitabine and CTLs would be more effective for CCA treatment than individual therapy. To test this hypothesis, we conducted an in vitro study using gemcitabine combined with CTLs to treat gemcitabine-resistant CCA (KKU-213) cells. KKU-213 cells were pretreated with gemcitabine and tested for killing by CTLs activated by dendritic cells that were prepared by three different methods, including: (i) monocyte-derived dendritic cells pulsed with cancer cell lysate (Mo-DC + Lys), (ii) self-differentiated dendritic cells pulsed with cancer-cell lysate (SD-DC + Lys), and (iii) SD-DC presenting tumor-associated antigen PRKAR1A (SD-DC-PR). KKU-213 cells pretreated with gemcitabine were killed by CTLs activated by either SD-DC + Lys or SD-DC-PR more efficiently than those activated by Mo-DC + Lys. Furthermore, KKU-213 cells pretreated with a low dose (2 µM) of gemcitabine significantly enhanced the cytotoxic activity of CTLs activated by either SD-DC + Lys or SD-DC-PR at all evaluated effector (E) to target cell (T) ratios. At an E:T ratio of 5:1, KKU-213 cells pretreated with gemcitabine enhanced the cytotoxic activity of CLTs by approximately 2.5-fold (greater than 50% cell death) compared to untreated condition. The upregulation of HLA class I upon pretreatment of KKU-213 cells with gemcitabine may suggest a mechanism that leads to alteration of the antigen presentation process to promote CTL functions. These findings support the concept of combination therapy for overcoming chemo-resistant CCA.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/terapia , Colangiocarcinoma/terapia , Desoxicitidina/análogos & derivados , Linfocitos T Citotóxicos , Línea Celular Tumoral , Células Dendríticas/inmunología , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Humanos , Gemcitabina
18.
Hum Vaccin Immunother ; 16(10): 2318-2327, 2020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-31976810

RESUMEN

Cholangiocarcinoma (CCA) is an aggressive tumor that is associated with high rates of recurrence and mortality. This is due, in part, to the fact that CCA cells and their microenvironment secrete immunosuppressive cytokines, transforming growth factor-ß (TGF-ß) and interleukin-10 (IL-10), that inhibit dendritic cell (DC) functions, which, in turn, results in the decreased anti-tumor activity of T-cells. We hypothesized that the TGF-ß receptor and IL-10 blockade on dendritic cells would improve DC function, thereby allowing improved activation of T cells against CCA cells. To test our hypothesis, we generated self-differentiated DCs (SD-DCs) via transduction of human peripheral blood monocytes with lentivirus expressing IL-4 and GM-CSF. SD-DCs were transduced with a second lentivirus containing short-hairpin RNAs (shRNAs) to knock-down TGF-ßRII and IL-10RA mRNAs. Immunoblot confirmed the reduced expression levels of TGF-ß and IL-10 receptors in both SD-DCs that were transduced with a single and/or combination of lentiviruses containing shRNAs. SD-DCs were thereafter pulsed with tumor antigens extracted from CCA cell lines in an effort to activate DC function. MHC class II (HLA-DR) and co-stimulatory molecules (CD40 and CD86) on SD-DCs were upregulated to levels comparable to those on DCs generated by the conventional method. Suppression of TGF-ß and IL-10 receptors on SD-DCs influenced the effector T-cells to produce IFN-γ, which enhanced their ability to kill CCA cells. The preparation of adoptive effector T-cells holds the potential of becoming a novel therapy for cellular immunotherapy in CCA.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/terapia , Conductos Biliares Intrahepáticos , Diferenciación Celular , Colangiocarcinoma/genética , Colangiocarcinoma/terapia , Células Dendríticas , Humanos , Recurrencia Local de Neoplasia , ARN Interferente Pequeño/genética , Receptores de Interleucina-10 , Linfocitos T , Factor de Crecimiento Transformador beta , Microambiente Tumoral
19.
Mol Endocrinol ; 22(7): 1658-72, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18436566

RESUMEN

Carbohydrate response element-binding protein (ChREBP) is a basic helix-loop-helix/leucine zipper transcription factor that binds to the carbohydrate response element in the promoter of certain lipogenic and glycolytic genes. High glucose can activate ChREBP by releasing an intramolecular inhibition within the glucose-sensing module (GSM) that occurs in low glucose. We report here that the glucose response of GSM is mediated by cooperation between five conserved submodules known as Mondo conserved regions (MCRs) I through V within GSM. Deletion of individual MCRs leads to complete (for MCR II, III, and IV) or partial (MCR I) loss of glucose response of ChREBP. MCR IV is necessary and sufficient for inhibiting the transcriptional activity of ChREBP under low glucose. The roles of MCR II and III in glucose response of ChREBP are independent of and distinct from their function in controlling subcellular localization. We further demonstrate that, instead of inhibiting ChREBP activity as would be predicted from its cytoplasmic retentive function, 14-3-3 binding with MCR III is essential for the glucose responsiveness of ChREBP. The interaction between 14-3-3 and ChREBP is constitutive, indicating a permissive role of 14-3-3 in the glucose response of ChREBP. We further uncovered an unconventional 14-3-3 binding motif (residues 116-135) lacking phosphor-serine/threonine within MCR III, a predicted alpha-helix highly conserved in all Mondo proteins. We conclude that individual subdomains in the GSM (MCR I through V) play diverse but crucial roles in cooperation with essential trans-acting cofactors such as 14-3-3 proteins to mediate the glucose response of ChREBP.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proteínas 14-3-3/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/química , Glucosa/química , Glucosa/metabolismo , Glutatión Transferasa/metabolismo , Proteínas Luminiscentes/química , Microscopía Fluorescente , Modelos Genéticos , Datos de Secuencia Molecular , Ratas , Retroviridae/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
20.
J Gastrointest Oncol ; 10(3): 437-444, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31183193

RESUMEN

BACKGROUND: The colorectal cancer (CRC) tumorigenesis is driving by genetic alterations leading to changes in protein expression such as p53. The p53 is frequently expressed in CRC and its association with clinicopathological features is still controversial. Moreover, accumulated evidence suggests that both p53 and nuclear exporter protein, exportin 1 (XPO1), are working in reciprocal manner may lead to loss of p53 nuclear localization and enhance cancer progression through hyperactive nuclear export. Accordingly, the present study aimed to evaluate the expression of p53 in CRC Yemeni patients and to explore the association between the p53 and XPO1 coexpression in relation to clinicopathological features. METHODS: A series of 40 formalin fixed paraffin embedded (FFPE) tissue blocks taken from CRC patients that diagnosed as adenocarcinoma were prospectively collected and then analyzed for p53 and XPO1 expression by immunohistochemistry (IHC). The patients and tumor clinicopathological characteristics were retrieved from the histopathology reports and the P value <0.05 were considered statistically significant. RESULTS: The p53 expression was observed in 60% (24/40) of CRC tumor samples. Significantly, the p53 expression was noted in 72.4% (21/29) of the left side compared to 27.3% (3/11) of the right side colon tumors (P=0.014). Furthermore, p53 expression was positively and significantly correlated with well-but not moderate- or poorly-differentiated tumors (P=0.023). No significant difference was observed between the p53 expression and age, gender and tumor size. Regarding the XPO1 expression, the p53 expression didn't show an association with XPO1 expression. The coexpression of p53 and XPO1 analysis revealed that 100% (11/11) tumors with negative p53 and positive XPO1 coexpression was noted with lymph node metastasis with significant difference (P=0.003) and more frequently observed in moderate-or poorly- differentiated tumors. CONCLUSIONS: The loss of p53 accompanied with increased XPO1 expressions was associated with the progression of histopathological features of CRC Yemeni patients. Further studies are needed to elucidate the p53 genetic mutations in relation to the XPO1 coexpression in CRC prognosis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA