Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.379
Filtrar
1.
Cancer Med ; 13(18): e70212, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39300946

RESUMEN

INTRODUCTION: Cells are covered with a glycan surface layer that is referred to as the glycocalyx (GCX). It has been reported that the formation of the GCX is promoted on cancer cells and is associated with tumor growth and metastasis. Heparan sulfate proteoglycan glypican-1 (GPC1) is a core protein of the GCX that is overexpressed in esophageal squamous cell carcinoma (ESCC) and is involved in the development and progression of cancer cells. The purpose of the present study is to analyze the utility of GPC1 as a new biomarker ralated to glycocalyx that reflects therapeutic effect and prognosis of ESCC. METHODS: We measured the concentration of GPC1 protein in preoperative plasma from advanced esophageal cancer patients and examined its relationships with clinicopathological factors and therapeutic efficacy, and the effects of extracellular GPC1 were investigated. RESULTS: The following clinical factors were significantly correlated with the preoperative high GPC1 concentration: male, tumor size ≥30 mm, venous invasion, pT factor ≥2, pStage ≥3, residual tumor, and distant metastatic recurrence. Both overall and recurrence-free survival were significantly worse in the high GPC1 group. Extracellular GPC1 protein concentration reflected intracellular GPC1 expression. Furthermore, we examined the effects of extracellular recombinant human (rh)GPC1 on ESCC cells, and found that extracellular rhGPC1 affects cell motility, including migration and invasion. CONCLUSIONS: These results demonstrated the utility of extracellular GPC1 as a biomarker, which can be assayed from a less invasive blood sample-based liquid biopsy. Extracellular GPC1 protein plays a role in both tumor cell motility and cancer progression. Thus, plasma GPC1 is a useful biomarker for esophageal cancer progression and may be a potential candidate of therapeutic target.


Asunto(s)
Biomarcadores de Tumor , Progresión de la Enfermedad , Neoplasias Esofágicas , Glipicanos , Humanos , Glipicanos/metabolismo , Glipicanos/genética , Glipicanos/sangre , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/mortalidad , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/sangre , Masculino , Femenino , Pronóstico , Persona de Mediana Edad , Biomarcadores de Tumor/metabolismo , Anciano , Línea Celular Tumoral , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/sangre , Carcinoma de Células Escamosas de Esófago/cirugía , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/mortalidad , Movimiento Celular , Estadificación de Neoplasias
3.
Talanta ; 280: 126766, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39191106

RESUMEN

Glypican-1 (GPC-1) protein-positive small extracellular vesicles (GPC-1+-sEV) have been proposed as potential biomarkers for early diagnosis of pancreatic cancer. In this study, we present an integrated real-time isolation and detection platform (IRTIDP) to capture and analyze GPC-1+-sEV directly from sera of pancreatic cancer patients. First, CD63 antibody-modified metal-organic framework (MOF) materials were utilized to enrich sEVs with a capture efficiency of 93.93 %. Second, a SERS probe was constructed by Raman reporter 4-MBA and GPC-1 antibody modified SERS active silver nanoparticles (AgNPs), which formed a sandwich complex structure of "MOFs@GPC-1+-sEV@AgNPs-4-MBA" with MOFs-enriched sEVs. The IRTSDP can complete the capture and detection process within 35 min, with a detection limit for 1 GPC-1+-sEV/µL, and linear range between 105∼109 GPC-1+-sEV/mL. Furthermore, this approach has been applied to quantify serum sEV GPC-1 in clinical pancreatic cancer patients. Based on the SERS intensity analysis, pancreatic cancer patients can be distinguished from pancreatic cystadenoma patients and healthy individuals effectively using this innovative platform that provides highly specific and sensitive means for early diagnosis of pancreatic cancer as well as other tumor types.


Asunto(s)
Vesículas Extracelulares , Glipicanos , Nanopartículas del Metal , Neoplasias Pancreáticas , Humanos , Glipicanos/sangre , Glipicanos/inmunología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/sangre , Vesículas Extracelulares/química , Nanopartículas del Metal/química , Espectrometría Raman/métodos , Plata/química , Estructuras Metalorgánicas/química , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/análisis , Límite de Detección , Tetraspanina 30
4.
Nat Commun ; 15(1): 7141, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39164224

RESUMEN

Novel chimeric antigen receptor (CAR) T-cell approaches are needed to improve therapeutic efficacy in solid tumors. High-risk neuroblastoma is an aggressive pediatric solid tumor that expresses cell-surface GPC2 and GD2 with a tumor microenvironment infiltrated by CD16a-expressing innate immune cells. Here we engineer T-cells to express a GPC2-directed CAR and simultaneously secrete a bispecific innate immune cell engager (BiCE) targeting both GD2 and CD16a. In vitro, GPC2.CAR-GD2.BiCE T-cells induce GPC2-dependent cytotoxicity and secrete GD2.BiCE that promotes GD2-dependent activation of antitumor innate immunity. In vivo, GPC2.CAR-GD2.BiCE T-cells locally deliver GD2.BiCE and increase intratumor retention of NK-cells. In mice bearing neuroblastoma patient-derived xenografts and reconstituted with human CD16a-expressing immune cells, GD2.BiCEs enhance GPC2.CAR antitumor efficacy. A CAR.BiCE strategy should be considered for tumor histologies where antigen escape limits CAR efficacy, especially for solid tumors like neuroblastoma that are infiltrated by innate immune cells.


Asunto(s)
Gangliósidos , Inmunidad Innata , Inmunoterapia Adoptiva , Células Asesinas Naturales , Neuroblastoma , Receptores Quiméricos de Antígenos , Linfocitos T , Neuroblastoma/inmunología , Neuroblastoma/terapia , Neuroblastoma/patología , Animales , Humanos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Ratones , Gangliósidos/inmunología , Línea Celular Tumoral , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto , Glipicanos/inmunología , Glipicanos/metabolismo , Microambiente Tumoral/inmunología , Femenino
5.
Clin Exp Med ; 24(1): 204, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39196390

RESUMEN

The application of CAR-T cells in solid tumors poses several challenges, including poor T cell homing ability, limited infiltration of T cells and an immunosuppressive tumor environment. In this study, we developed a novel approach to address these obstacles by designing GPC3-specific CAR-T cell that co-express IL-21 and CXCL9 (21 × 9 GPC3 CAR-T cells) and blocking the PD-1 expression on it. The proliferation, cell phenotype, cytokine secretion and cell migration of indicated CAR-T cells were evaluated in vitro. The cytotoxic activities of genetically engineered CAR-T cells were accessed in vitro and in vivo. Compared to conventional GPC3 CAR-T cells, the 21 × 9 GPC3 CAR-T cells demonstrated superior proliferation, cytokine secretion and chemotaxis capabilities in vitro. Furthermore, when combined with PD-1 blockade, the 21 × 9 GPC3 CAR-T cells exhibited enhanced proliferation, cytokine secretion and enrichment of effector T cells such as CTL, NKT and TEM cells. In xenograft tumor models, the PD-1 blocked 21 × 9 GPC3 CAR-T cells effectively suppressed HCC xenograft growth and increased T cell infiltration. Overall, our study successfully generated GPC3 CAR-T cells expressing both IL-21 and CXCL9, demonstrated that combining PD-1 blockade can further enhance CAR-T cell function by promoting proliferation, cytokine secretion, chemotaxis and antitumor activity. These findings present a hopeful and potentially effective strategy for GPC3-positive HCC patients.


Asunto(s)
Carcinoma Hepatocelular , Quimiocina CXCL9 , Glipicanos , Inmunoterapia Adoptiva , Interleucinas , Neoplasias Hepáticas , Receptor de Muerte Celular Programada 1 , Receptores Quiméricos de Antígenos , Glipicanos/inmunología , Glipicanos/metabolismo , Glipicanos/antagonistas & inhibidores , Glipicanos/genética , Interleucinas/metabolismo , Interleucinas/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/tratamiento farmacológico , Animales , Humanos , Inmunoterapia Adoptiva/métodos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Ratones , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proliferación Celular , Línea Celular Tumoral
6.
Cytokine Growth Factor Rev ; 79: 29-38, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39191624

RESUMEN

Hepatocellular carcinoma (HCC) is a leading contributor to cancer-related deaths worldwide and presents significant challenges in diagnosis and treatment due to its heterogeneous nature. The discovery of biomarkers has become crucial in addressing these challenges, promising early detection, precise diagnosis, and personalized treatment plans. Key biomarkers, such as alpha fetoprotein (AFP) glypican 3 (GPC3) and des gamma carboxy prothrombin (DCP) have shown potential in improving clinical results. Progress in proteomic technologies, including next-generation sequencing (NGS), mass spectrometry, and liquid biopsies detecting circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), has deepened our understanding of HCC's molecular landscape. Immunological markers, like PD-L1 expression and tumor-infiltrating lymphocytes (TILs), also play a crucial role in guiding immunotherapy decisions. Despite these advancements, challenges remain in biomarker validation, standardization, integration into clinical practice, and cost-related barriers. Emerging technologies like single-cell sequencing and machine learning offer promising avenues for further exploration. Continued investment in research and collaboration among researchers, healthcare providers, and policymakers is vital to harness the potential of biomarkers fully, ultimately revolutionizing HCC management and improving patient outcomes through personalized treatment approaches.


Asunto(s)
Biomarcadores de Tumor , Carcinoma Hepatocelular , Neoplasias Hepáticas , Medicina de Precisión , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/terapia , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/terapia , Medicina de Precisión/métodos , Pronóstico , Células Neoplásicas Circulantes/patología , Glipicanos , Proteómica/métodos
7.
Funct Integr Genomics ; 24(4): 127, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39014225

RESUMEN

Prostate cancer is a major medical problem for men worldwide. Advanced prostate cancer is currently incurable. Recently, much attention was paid to the role of GPC2 in the field of oncology. Nevertheless, there have been no investigations of GPC2 and its regulatory mechanism in prostate cancer. Here, we revealed a novel action of GPC2 and a tumor promoting mechanism in prostate cancer. GPC2 was upregulated in prostate cancer tissues and cell lines. Higher expression of GPC2 was correlated with higher Gleason score, lymphatic metastasis, and worse overall survival in prostate cancer patients. Decreased expression of GPC2 inhibited cell proliferation, migration, and invasion in prostate cancer, whereas GPC2 overexpression promoted these properties. Mechanistically, GPC2 promoted the activation of PI3K/AKT signaling pathway through MDK. The rescue assay results in prostate cancer cells demonstrated that overexpression of MDK could attenuate GPC2 knockdown induced inactivation of PI3K/AKT signaling and partly reverse GPC2 knockdown induced inhibition of cell proliferation, migration, and invasion. In all, our study identified GPC2 as an oncogene in prostate cancer. GPC2 promoted prostate cancer cell proliferation, migration, and invasion via MDK-mediated activation of PI3K/AKT signaling pathway. GPC2 might be a promising prognosis predictor and potential therapeutic target in prostate cancer.


Asunto(s)
Movimiento Celular , Proliferación Celular , Glipicanos , Fosfatidilinositol 3-Quinasas , Neoplasias de la Próstata , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Masculino , Humanos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Línea Celular Tumoral , Glipicanos/metabolismo , Glipicanos/genética , Regulación Neoplásica de la Expresión Génica , Progresión de la Enfermedad
8.
Front Biosci (Landmark Ed) ; 29(7): 268, 2024 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-39082348

RESUMEN

Liver cancer, primarily hepatocellular carcinoma (HCC), is the second leading cause of cancer-related deaths globally. It is typically characterized by rapid progression, poor prognosis, and high mortality rates. Given these challenges, the search for molecular targets aiding early diagnosis and targeted therapy remains imperative. Glypican 3 (GPC3), a cell-surface glycoprotein, emerges as a promising candidate for addressing HCC Overexpressed in HCC tissues; GPC3 is a credible immunohistochemical marker for liver cancer diagnosis and a potential marker for liquid biopsy through soluble GPC3 in serum. Various immunotherapies targeting GPC3 have been developed, including vaccines, anti-GPC3 immunotoxins, and chimeric antigen receptor-modified cells. This review comprehensively covers the structure, physicochemical properties, biological functions, and clinical applications of GPC3. It explores diagnostic and treatment strategies centered around GPC3, offering hope for improved early detection and targeted therapies in the challenging landscape of HCC.


Asunto(s)
Biomarcadores de Tumor , Carcinoma Hepatocelular , Glipicanos , Inmunoterapia , Neoplasias Hepáticas , Glipicanos/inmunología , Glipicanos/metabolismo , Humanos , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Inmunoterapia/métodos , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/inmunología , Biomarcadores de Tumor/sangre , Terapia Molecular Dirigida/métodos , Medicina de Precisión/métodos , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico
9.
Zhonghua Zhong Liu Za Zhi ; 46(7): 686-695, 2024 Jul 23.
Artículo en Chino | MEDLINE | ID: mdl-39034804

RESUMEN

Objective: To investigate the immunophenotypic and molecular biological characteristics of patients with elevated serum alpha-fetoprotein (AFP) and enteroblastic differentiated gastric adenocarcinoma (GAED). Methods: The clinicopathological data of 13 patients with elevated serum AFP and GAED admitted to Shanxi Cancer Hospital from 2018 to 2020 were collected. Immunohistochemistry (IHC) and next-generation sequencing (NGS) were used to analyze the immune markers and molecular biological characteristics of the pathological tissues of the patients. Kaplan-Meier method and log rank test were used for survival analysis. Results: Among the 13 patients with GAED, 12 were male and 1 was female, aged 41-70 years, with a median age of 64 years. The lesions were mainly located in the gastric antrum (5 cases) and gastric body (4 cases). IHC results showed that the tumor embryonic protein (AFP, SALL4, GPC3), intestinal epithelial differentiation protein (CDX-2, CD10), and some original intestinal epithelial phenotype markers (OCT3/4, Claudin6) were expressed in the tumor tissues. Combined application of multiple markers can reduce the rate of missed diagnosis. Among the 13 patients, 12 had at least one mutation (1 mutation: 1 case, 2-5 mutations: 3 cases, 6-15 mutations: 8 cases), and 1 case was not detected. The gene with the highest mutation frequency was TP53 (10 cases), and other mutant genes included EPHB1 (3 cases), ATRX (2 cases), EPHA5 (2 cases), GATA3 (2 cases), LRP1B (2 cases) and MAP2K4 (2 cases) were also detected. Three of the 13 patients had structural variations, which were C14orf177-GNAS, AIM1-FGFR3, and EPHA6-ROS1 gene rearrangements. All 13 patients had copy number variation, and 11 patients had copy number variation of more than 2 genes. The common amplification genes were IRS2 (5 cases), PTEN (5 cases), GNAS (4 cases), CCNE1 (3 cases), CEBPA (3 cases), PCK1 (3 cases) and ERBB2 (2 cases). The common deletion genes were SOX2 (5 cases) and MYC (5 cases). Among the 13 patients, 4 died, and 2 of the dead patients had liver metastasis. There were 4 patients with disease-free survival and 5 patients with disease progression, including 3 cases of abdominal metastasis and 2 cases of liver metastasis. The 3-year survival rate of patients was 65.9 %, and the 3-year progression-free survival rate was 30.7 %. Gene LRP1B point mutation was associated with poor prognosis (P<0.001). There was no significant improvement in the prognosis of patients treated with immunotherapy compared with those treated with chemotherapy alone (P=0.595), but the prognosis of patients treated with postoperative chemotherapy or postoperative chemotherapy plus immunotherapy was better than that of patients treated with surgery alone (P<0.05). Conclusions: Elevated serum AFP with GAED is a highly invasive tumor with unique molecular characteristics, often accompanied by multiple molecular events. TP53 mutation is the most common type of gene mutation. In addition, some cases are accompanied by HER2 amplification and gene rearrangement.


Asunto(s)
Adenocarcinoma , Neoplasias Gástricas , alfa-Fetoproteínas , Humanos , Masculino , alfa-Fetoproteínas/metabolismo , Femenino , Persona de Mediana Edad , Neoplasias Gástricas/patología , Neoplasias Gástricas/sangre , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Anciano , Adulto , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/sangre , Adenocarcinoma/metabolismo , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/sangre , Diferenciación Celular , Mutación , Factor de Transcripción CDX2/metabolismo , Factor de Transcripción CDX2/genética , Glipicanos
10.
Sci Rep ; 14(1): 15870, 2024 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-38982153

RESUMEN

Recent studies indicate that Glypican 1 (GPC-1) is aberrantly expressed and plays a key role in certain cancers, but little is known in the hepatocellular carcinoma. Raw data from TCGA, GTEx and TIMER databases were utilized to comprehensively analyze GPC-1 expression landscape in pan-cancer, and the biological function of GPC-1 was investigated in liver cancer cells. The results revealed that GPC-1 is highly expressed in HCC, negatively correlated with survival, and also positively correlated with immune infiltration and clinical stage. Furthermore, GPC-1 promoted cell proliferation and inhibited apoptosis in the HCC cell lines. WGCNA analysis and HCCDB database revealed that Akt acted as a key molecule related to GPC-1, influencing biological functions and regulating cell malignant behaviors via the AKT signaling pathway. In conclusion, our findings provide a relatively comprehensive understanding of the oncogenic role of GPC-1 in HCC, implying that GPC-1 could serve as an innovative therapeutic target.


Asunto(s)
Carcinoma Hepatocelular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glipicanos , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Glipicanos/metabolismo , Glipicanos/genética , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Línea Celular Tumoral , Apoptosis/genética , Transducción de Señal , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Biomolecules ; 14(7)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-39062482

RESUMEN

Glypican-4 belongs to a group of poorly understood adipokines, with potential importance in people with metabolic syndrome, especially in groups of patients with glucose metabolism disorder. This study aimed to assess the effect of physical activity on serum glypican-4 and irisin levels and total antioxidant status (TAS) in plasma and saliva in women with metabolic syndrome (MetS). Seventy-two Caucasian women aged 25-60 were included in the study (36 women with MetS and 36 women without MetS (control group, CONTR)). The glypican-4 and irisin concentrations, total antioxidant status, glycemia, lipid profile, anthropometric parameters, and blood pressure were analyzed before and after 28 days of controlled physical activity. Serum glypican-4 and plasma TAS levels were higher (p = 0.006 and p = 0.043, respectively) on the 28th day than on the first day of the study only in the CONTR group. In the MetS group, 28 days of physical activity caused a reduction in body fat mass (p = 0.049) without changes in glypican-4, irisin, or TAS levels. In both groups, glypican-4 levels correlated positively with irisin levels and negatively with Waist-Hip Ratio (WHR), while irisin levels correlated positively with High-Density Lipoprotein Cholesterol (HDL-C) levels and negatively with waist circumference (WC) and WHR values on the 28th day of the study. To summarize, a 28-day moderate training, accompanied by a reduction in body fat mass, stabilized glypican-4 levels and TAS in female patients with MetS.


Asunto(s)
Antioxidantes , Ejercicio Físico , Fibronectinas , Glipicanos , Síndrome Metabólico , Humanos , Síndrome Metabólico/sangre , Síndrome Metabólico/metabolismo , Femenino , Fibronectinas/sangre , Adulto , Glipicanos/sangre , Glipicanos/metabolismo , Persona de Mediana Edad , Antioxidantes/metabolismo , Relación Cintura-Cadera
12.
Int J Nanomedicine ; 19: 7015-7031, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011387

RESUMEN

Purpose: The incidence of hepatocellular carcinoma (HCC) is continuously increasing, and the mortality rate remains high. Thus, more effective strategies are needed to improve the treatment of HCC. Methods: In this study, we report the use of a visualized glypican-3 (GPC3)-targeting nanodelivery system (named GC-NBs) in combination with sonodynamic therapy (SDT) to enhance the therapeutic efficacy for treating HCC. The obtained nanodelivery system could actively target hepatocellular carcinoma cells and achieve ultrasound imaging through phase changes into nanobubbles under low-intensity ultrasound irradiation. Meanwhile, the released chlorine e6 (Ce6) after the nanobubbles collapse could lead to the generation of reactive oxygen species (ROS) under ultrasound irradiation to induce SDT. Results: Both in vitro and in vivo experiments have shown that GC-NBs can accumulate in tumour areas and achieve sonodynamic antitumour therapy under the navigation action of glypican-3-antibody (GPC3-Ab). Furthermore, in vitro and in vivo experiments did not show significant biological toxicity of the nanodelivery system. Moreover, GC-NBs can be imaged with ultrasound, providing personalized treatment monitoring. Conclusion: GC-NBs enable a visualized antitumour strategy from a targeted sonodynamic perspective by combining tumour-specific targeting and stimuli-responsive controlled release into a single system.


Asunto(s)
Carcinoma Hepatocelular , Glipicanos , Neoplasias Hepáticas , Terapia por Ultrasonido , Glipicanos/metabolismo , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Humanos , Terapia por Ultrasonido/métodos , Ratones , Línea Celular Tumoral , Clorofilidas , Especies Reactivas de Oxígeno/metabolismo , Ratones Endogámicos BALB C , Células Hep G2 , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Ultrasonografía/métodos , Nanopartículas/química
13.
Neuroscience ; 553: 121-127, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-38992568

RESUMEN

INTRODUCTION: Alzheimer's disease (AD) is characterized by accumulation of amyloid beta (Aß) and hyperphosphorylated tau (Tau-P) in the brain. Aß enhances the activity of kinases involved in the formation of Tau-P. Phosphorylation at Thr 181 determines the propagation of multiple tau phosphorylations. Aß is derived from the amyloid precursor protein (APP). Cleavage of APP by ß-secretase also initiates release of heparan sulfate (HS) from the proteoglycan glypican-1 (GPC1). OBJECTIVES: In this study, we have explored possible connections between GPC1 expression, HS release, APP processing and Tau-P formation in human neural stem cells. METHODS: GPC1 formation was suppressed by using CRISPR/Cas9 and increased by using a vector encoding GPC1. HS release from GPC1 was increased by growing cells in medium containing Arg and ascorbate. Effects were monitored by immunofluorescence microscopy and slot immunoblotting using antibodies/antisera recognizing Aß, GPC1, HS released from GPC1, total Tau, and Tau phosphorylated at Thr-181, 217 or 231. The latter have been used as blood biomarkers for AD. RESULTS: Suppression of GPC1 expression resulted in increased phosphorylation at Thr 181 and Thr 217. When GPC1 was overexpressed, phosphorylation at Thr 217 decreased. Stimulation of HS release from GPC1 diminished tau phosphorylation at all of the three Thr positions, while expression of GPC1 was unaffected. Simultaneous stimulation of HS release and APP processing by the cytokine TNF-α also suppressed tau phosphorylation. CONCLUSION: The increased release of GPC1-derived HS may interfere with Aß formation and/or Aß interaction with tau.


Asunto(s)
Péptidos beta-Amiloides , Glipicanos , Células-Madre Neurales , Proteínas tau , Humanos , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Glipicanos/metabolismo , Células-Madre Neurales/metabolismo , Fosforilación/fisiología , Proteínas tau/metabolismo
14.
Turk Kardiyol Dern Ars ; 52(5): 307-314, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38982813

RESUMEN

OBJECTIVE: Myocardial infarction is associated with right ventricular (RV) remodeling. Glypican-6 (GPC6), a member of the membrane proteoglycan family, plays a significant role in cardiac remodeling. This study aims to determine if GPC6 can predict RV remodeling after percutaneous coronary intervention (PCI) in patients with non-ST segment elevation myocardial infarction (NSTEMI). METHODS: The study enrolled 164 consecutive patients with NSTEMI and controls. It compared baseline plasma GPC6 levels, echocardiography, and laboratory parameters between the RV remodeling and non-RV remodeling groups with NSTEMI. Echocardiographic data were measured at baseline and at six months. RESULTS: GPC6 levels were higher in the NSTEMI group 11.06 ng/mL (4.61-18.17) vs. 5.98 ng/mL (3.81-9.83) compared to the control group in the initial phase. RV remodeling, defined as a ≥ 20% increase in RV end-diastolic area (RV EDA), was observed in 23 patients (30%). After six months, RV EDA increased significantly from baseline 18.68 ± 1.20 cm2 vs. 24.91 ± 1.08 cm2, P < 0.001. GPC6 was a significant independent predictor of RV remodeling (hazard ratio [HR]: 1.546, 95% confidence interval [CI]: 1.056-2.245, P < 0.001). Receiver operating characteristic curve (ROC) analyses showed that GPC6 values > 15.5 ng/mL (area under the curve [AUC] = 0.828, sensitivity: 70%, specificity: 74%, P < 0.001) were strong predictors of RV remodeling. CONCLUSION: NSTEMI patients should be closely monitored for RV remodeling. GPC6 appears useful in detecting RV remodeling following NSTEMI in patients undergoing PCI.


Asunto(s)
Glipicanos , Infarto del Miocardio sin Elevación del ST , Intervención Coronaria Percutánea , Remodelación Ventricular , Humanos , Masculino , Femenino , Glipicanos/sangre , Remodelación Ventricular/fisiología , Persona de Mediana Edad , Infarto del Miocardio sin Elevación del ST/sangre , Infarto del Miocardio sin Elevación del ST/fisiopatología , Ecocardiografía , Anciano , Estudios de Casos y Controles , Biomarcadores/sangre , Curva ROC
15.
Sheng Wu Gong Cheng Xue Bao ; 40(7): 2258-2269, 2024 Jul 25.
Artículo en Chino | MEDLINE | ID: mdl-39044589

RESUMEN

This study aims to prepare bacterial outer membrane vesicles (OMVs) with anti-glypican-3 (GPC3) single-chain antibody and analyze their targeting effects on Hep G2 hepatocellular carcinoma (HCC) cells and tissue. The recombinant plasmid pET28a-Hbp-hGC 33-scFv was constructed by ligating Hbp-hGC 33-scFv to pET28a. Western blotting was employed to determine the prokaryotic expression of the fusion protein Hbp-hGC 33-scFv, on the basis of which the optimal induction conditions were determined. Hbp-hGC 33-OMVs secreted from the recombinant expressing strains were collected by ultrafiltration concentration and then characterized. The localization of Hbp-hGC 33-scFv in bacteria and Hbp-hGC 33-OMVs was analyzed by immune electron microscopy. The binding of Hbp-hGC 33-scFv to Hep G2 cells was observed by immunofluorescence. The Hep G2 tumor-bearing mouse model was established, and the targeted retention of Hbp-hGC 33-OMVs in the tumor site of mice was observed by a fluorescence imaging system in vivo. The results showed that the actual molecular weight of the fusion protein was 175.3 kDa, and the optimal induction conditions were as follows: OD600=0.5, IPTG added at a final concentration of 0.5 mmol/L, and overnight induction at 16 ℃. The prepared Hbp-hGC 33-OMVs were irregular spherical structures with an average particle size of (112.3±4.6) nm, expressing OmpC, OmpA, and the fusion protein Hbp-hGC 33-scFv. The Hbp-hGC 33-OMVs prepared in this study demonstrated stronger ability of binding to Hep G2 cells than the wild-type OMVs (P=0.008). All the data indicated that Hbp-hGC 33-OMVs with anti-GPC3 single-chain antibody were successfully prepared and could be used for research on the targeted therapy of hepatocellular carcinoma.


Asunto(s)
Membrana Externa Bacteriana , Carcinoma Hepatocelular , Glipicanos , Neoplasias Hepáticas , Anticuerpos de Cadena Única , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/química , Animales , Ratones , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Membrana Externa Bacteriana/metabolismo , Membrana Externa Bacteriana/inmunología , Células Hep G2 , Glipicanos/inmunología , Glipicanos/metabolismo , Glipicanos/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/biosíntesis , Escherichia coli/genética , Escherichia coli/metabolismo , Sistemas de Liberación de Medicamentos , Ratones Desnudos
16.
Front Med ; 18(4): 708-720, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38833102

RESUMEN

CD39 serves as a crucial biomarker for neoantigen-specific CD8+ T cells and is associated with antitumor activity and exhaustion. However, the relationship between CD39 expression levels and the function of chimeric antigen receptor T (CAR-T) cells remains controversial. This study aimed to investigate the role of CD39 in the functional performance of CAR-T cells against hepatocellular carcinoma (HCC) and explore the therapeutic potential of CD39 modulators, such as mitochondrial division inhibitor-1 (mdivi-1), or knockdown CD39 through short hairpin RNA. Our findings demonstrated that glypican-3-CAR-T cells with moderate CD39 expression exhibited a strong antitumor activity, while high and low levels of CD39 led to an impaired cellular function. Methods modulating the proportion of CD39 intermediate (CD39int)-phenotype CAR-T cells such as mdivi-1 and CD39 knockdown enhanced and impaired T cell function, respectively. The combination of mdivi-1 and CD39 knockdown in CAR-T cells yielded the highest proportion of infiltrated CD39int CAR-T cells and demonstrated a robust antitumor activity in vivo. In conclusion, this study revealed the crucial role of CD39 in CAR-T cell function, demonstrated the potential therapeutic efficacy of combining mdivi-1 with CD39 knockdown in HCC, and provided a novel treatment strategy for HCC patients in the field of cellular immunotherapy.


Asunto(s)
Apirasa , Carcinoma Hepatocelular , Glipicanos , Inmunoterapia Adoptiva , Neoplasias Hepáticas , Receptores Quiméricos de Antígenos , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/genética , Glipicanos/inmunología , Glipicanos/genética , Glipicanos/metabolismo , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/genética , Apirasa/metabolismo , Apirasa/genética , Humanos , Animales , Ratones , Inmunoterapia Adoptiva/métodos , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Línea Celular Tumoral , Antígenos CD/genética , Antígenos CD/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Clin Cancer Res ; 30(16): 3578-3591, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38864848

RESUMEN

PURPOSE: Retinoblastoma is the most common intraocular malignancy in children. Although new chemotherapeutic approaches have improved ocular salvage rates, novel therapies are required for patients with refractory intraocular and metastatic disease. Chimeric antigen receptor (CAR) T cells targeting glypican-2 (GPC2) are a potential new therapeutic strategy. EXPERIMENTAL DESIGN: GPC2 expression and its regulation by the E2F1 transcription factor were studied in retinoblastoma patient samples and cellular models. In vitro, we performed functional studies comparing GPC2 CAR T cells with different costimulatory domains (4-1BB and CD28). In vivo, the efficacy of local and systemic administration of GPC2 CAR T cells was evaluated in intraocular and leptomeningeal human retinoblastoma xenograft models. RESULTS: Retinoblastoma tumors, but not healthy retinal tissues, expressed cell surface GPC2, and this tumor-specific expression was driven by E2F1. GPC2-directed CARs with 4-1BB costimulation (GPC2.BBz) were superior to CARs with CD28 stimulatory domains (GPC2.28z), efficiently inducing retinoblastoma cell cytotoxicity and enhancing T-cell proliferation and polyfunctionality. In vivo, GPC2.BBz CARs had enhanced persistence, which led to significant tumor regression compared with either control CD19 or GPC2.28z CARs. In intraocular models, GPC2.BBz CAR T cells efficiently trafficked to tumor-bearing eyes after intravitreal or systemic infusions, significantly prolonging ocular survival. In central nervous system (CNS) retinoblastoma models, intraventricular or systemically administered GPC2.BBz CAR T cells were activated in retinoblastoma-involved CNS tissues, resulting in robust tumor regression with substantially extended overall mouse survival. CONCLUSIONS: GPC2-directed CAR T cells are effective against intraocular and CNS metastatic retinoblastomas.


Asunto(s)
Glipicanos , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Retinoblastoma , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Animales , Retinoblastoma/inmunología , Retinoblastoma/patología , Retinoblastoma/terapia , Ratones , Receptores Quiméricos de Antígenos/inmunología , Glipicanos/inmunología , Glipicanos/antagonistas & inhibidores , Inmunoterapia Adoptiva/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/terapia , Neoplasias del Sistema Nervioso Central/inmunología , Neoplasias del Sistema Nervioso Central/secundario , Neoplasias del Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Femenino
20.
Anal Chem ; 96(26): 10705-10713, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38910291

RESUMEN

Circulating tumor cells (CTCs) serve as important biomarkers in the liquid biopsy of hepatocellular carcinoma (HCC). Herein, a homogeneous dual fluorescence indicators aptasensing strategy is described for CTCs in HCC, with the core assistance of a steric hindrance-mediated enzymatic reaction. CTCs in the sample could specifically bind to a 5'-biotin-modified glypican-3 (GPC3) aptamer and remove the steric hindrance formed by the biotin-streptavidin system. This influences the efficiency of the terminal deoxynucleotidyl transferase enzymatic reaction. Then, methylene blue (MB) was introduced to react with the main product poly cytosine (polyC) chain, and trivalent cerium ion (Ce3+) was added to react with the byproduct pyrophosphate to form fluorescent pyrophosphate cerium coordination polymeric nanoparticles. Finally, the CTCs were quantified by dual fluorescence indicators analysis. Under optimized conditions, the linear range was 5 to 104 cells/mL, and the limits of detection reached 2 cells/mL. Then, 40 clinical samples (15 healthy and 25 HCC patients) were analyzed. The receiver operating characteristic curve analysis revealed an area under the curve of 0.96, a sensitivity of 92%, and a specificity of 100%. Therefore, this study established a sensitive and accurate CTCs sensing system for clinical HCC patients, promoting early tumor diagnosis.


Asunto(s)
Aptámeros de Nucleótidos , Carcinoma Hepatocelular , Colorantes Fluorescentes , Neoplasias Hepáticas , Células Neoplásicas Circulantes , Humanos , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/metabolismo , Células Neoplásicas Circulantes/patología , Células Neoplásicas Circulantes/metabolismo , Aptámeros de Nucleótidos/química , Colorantes Fluorescentes/química , Glipicanos/metabolismo , Técnicas Biosensibles
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA