Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Cancer Res ; 80(16): 3292-3304, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32561533

RESUMEN

Although new generations of EGFR-tyrosine kinase inhibitors (EGFR-TKI) have been developed for the treatment of patients with non-small cell lung cancer (NSCLC) with EGFR-mutant tumors, TKI resistance often returns as a result of additional EGFR mutations. In addition to seeking for next-generation EGFR-TKI, developing novel EGFR-targeting strategies may hold the key to overcome the vicious cycle of TKI resistance. Endocan is known as a receptor tyrosine kinase ligand enhancer in tumorigenesis, but the impact of endocan on EGFR-driven NSCLC progression remains unknown. In this study, higher endocan levels were found in lung tumors compared with cancer-free tissues and correlated with poor prognosis in patients with NSCLC harboring mutant EGFR; circulating endocan levels were also significantly higher in patients with mutant EGFR. Endocan facilitated EGFR signaling via direct binding and enhancing of the EGF-EGFR interaction and supported the growth of tumors driven by mutated EGFR. Activated EGFR in turn upregulated expression of endocan via JAK/STAT3 and ERK/ELK cascades, thus forming a positive regulatory loop of endocan-EGFR signaling. On the basis of the binding region between endocan and EGFR, we designed therapeutic peptides and demonstrated promising therapeutic effects in xenografts harboring EGFR mutations including TKI-resistant T790M. Together, our findings highlight the novel interaction between endocan and EGFR and new opportunities to effectively target endocan-EGFR regulatory axis in patients with TKI-resistant NSCLC. SIGNIFICANCE: Endocan is a novel and critical regulator of EGF/EGFR signaling and serves as an alternative target of EGFR-TKI resistance in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Proteoglicanos/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expresión Génica , Genes ras/genética , Xenoinjertos , Humanos , Quinasas Janus/metabolismo , Pulmón/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Pronóstico , Inhibidores de Proteínas Quinasas/farmacología , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , ARN Mensajero/metabolismo , Receptor Cross-Talk , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba , Fosfatasas cdc25/metabolismo
2.
Neoplasia ; 22(2): 86-97, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31896526

RESUMEN

While recent research points to the importance of glycans in cancer immunity, knowledge on functional mechanisms is lacking. In lung carcinoma among other tumors, anti-tumor immunity is suppressed; and while some recent therapies boost T-cell mediated immunity by targeting immune-checkpoint pathways, robust responses are uncommon. Augmenting tumor antigen-specific immune responses by endogenous dendritic cells (DCs) is appealing from a specificity standpoint, but challenging. Here, we show that restricting a heparan sulfate (HS) loss-of-function mutation in the HS sulfating enzyme Ndst1 to predominantly conventional DCs (Ndst1f/f CD11cCre+ mutation) results in marked inhibition of Lewis lung carcinoma growth along with increased tumor-associated CD8+ T cells. In mice deficient in a major DC HS proteoglycan (syndecan-4), splenic CD8+ T cells showed increased anti-tumor cytotoxic responses relative to controls. Studies examining Ndst1f/f CD11cCre + mutants revealed that mutation was associated with an increase in anti-tumor cytolysis using either splenic CD8+ T cells or tumor-infiltrating (TIL) CD8+ T cells purified ex-vivo, and tested in pooled effector-to-target cytolytic assays against tumor cells from respective animals. On glycan compositional analysis, HS purified from Ndst1f/f CD11cCre + mutant DCs had reduced overall sulfation, including reduced sulfation of a tri-sulfated disaccharide species that was intriguingly abundant on wildtype DC HS. Interestingly, antigen presentation in the context of major histocompatibility complex class-I (MHC-I) was enhanced in mutant DCs, with more striking effects in the setting of HS under-sulfation, pointing to a likely regulatory role by sulfated glycans at the antigen/MHC-I - T-cell interface; and possibly future opportunities to improve antigen-specific T cell responses by immunologic targeting of HS proteoglycans in cancer.


Asunto(s)
Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Complejo Mayor de Histocompatibilidad/genética , Polisacáridos/genética , Proteoglicanos/genética , Sulfotransferasas/genética , Animales , Antígeno CD11c/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Células Dendríticas/inmunología , Células Dendríticas/patología , Heparitina Sulfato/farmacología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Mutación con Pérdida de Función/genética , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Polisacáridos/antagonistas & inhibidores , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/inmunología , Linfocitos T/inmunología , Linfocitos T/patología
3.
Semin Cancer Biol ; 62: 48-67, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31336150

RESUMEN

Over the past few decades, understanding how tumor cells evade the immune system and their communication with their tumor microenvironment, has been the subject of intense investigation, with the aim of developing new cancer immunotherapies. The current therapies against cancer such as monoclonal antibodies against checkpoint inhibitors, adoptive T-cell transfer, cytokines, vaccines, and oncolytic viruses have managed to improve the clinical outcome of the patients. However, in some tumor entities, the response is limited and could benefit from the identification of novel therapeutic targets. It is known that tumor-extracellular matrix interplay and matrix remodeling are necessary for anti-tumor and pro-tumoral immune responses. Proteoglycans are dominant components of the extracellular matrix and are a highly heterogeneous group of proteins characterized by the covalent attachment of a specific linear carbohydrate chain of the glycosaminoglycan type. At cell surfaces, these molecules modulate the expression and activity of cytokines, chemokines, growth factors, adhesion molecules, and function as signaling co-receptors. By these mechanisms, proteoglycans influence the behavior of cancer cells and their microenvironment during the progression of solid tumors and hematopoietic malignancies. In this review, we discuss why cell surface proteoglycans are attractive pharmacological targets in cancer, and we present current and recent developments in cancer immunology and immunotherapy utilizing proteoglycan-targeted strategies.


Asunto(s)
Membrana Celular/metabolismo , Neoplasias/metabolismo , Proteoglicanos/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor , Metabolismo de los Hidratos de Carbono , Membrana Celular/inmunología , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Matriz Extracelular , Glicosaminoglicanos , Humanos , Inmunomodulación/efectos de los fármacos , Inmunoterapia , Terapia Molecular Dirigida , Neoplasias/etiología , Neoplasias/inmunología , Neoplasias/terapia , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/inmunología
4.
Toxicol Appl Pharmacol ; 386: 114827, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31734320

RESUMEN

Endothelial-mesenchymal transition (EndMT) is a frequent event in endothelial dysfunction, which is associated with pulmonary arterial hypertension (PAH). MiR-181 family members exert diverse effects in multiple biological processes. However, the relationships between miR-181b-5p (miR-181b) and EndMT in PAH are not well understood. In this study, Sprague-Dawley (SD) rats were injected with monocrotaline (MCT) to establish PAH model, and primary rat pulmonary arterial endothelial cells (rPAECs) were treated with TNF-α, TGFß1 and IL-1ß in combination to induce EndMT (I-EndMT). Then we explored miR-181b expression and examined its functional role in PAH. Our data showed that miR-181b was down-expressed in PAH, and its overexpression attenuated the hemodynamics, pulmonary vascular hypertrophy, right ventricular remodeling and EndMT process in MCT-induced PAH rats. In I-EndMT rPAECs, we observed that inducing miR-181b reversed the decrease of endothelial markers and increase of mesenchymal markers. However, knockdown of miR-181b induced similar effects to EndMT. In addition, endocan and TGFBR1 levels were also increased in EndMT, which were negatively regulated by miR-181b. Luciferase activity results indicated that endocan and TGFBR1 were direct target genes of miR-181b. In summary, our findings firstly demonstrate that the beneficial effect of miR-181b on PAH may be associated with endocan/TGFBR1-mediated EndMT, providing a new insight into the diagnosis and treatment of PAH.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , MicroARNs/uso terapéutico , Monocrotalina/farmacología , Proteoglicanos/antagonistas & inhibidores , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Masculino , MicroARNs/farmacología , Hipertensión Arterial Pulmonar/inducido químicamente , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Cell Physiol Biochem ; 48(6): 2429-2440, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30121667

RESUMEN

BACKGROUND/AIMS: The chondroitin sulfate proteoglycan serglycin (SRGN), a hematopoietic cell granule proteoglycan, has been implicated in promoting tumor metastasis; however, the underlying mechanisms remain to be elucidated. The present study aimed to investigate the SRGN gene expression and its regulation as downstream signaling of hypoxia-inducible transcription factor 1 alpha (HIF-1α) in colorectal cancer (CRC) cells and tissues. METHODS: The expression of SRGN was analyzed in CRC specimens for its correlation with progression and metastasis. Using chromatin-immunoprecipitation (ChIP), quantitative real-time PCR, Western blot, and transwell assay, the functional role and underlying mechanism of SRGN in CRC metastasis were elucidated. Thus, this study provides evidence of a critical role of SRGN in metastatic progression of CRC. RESULTS: Our results indicated that SRGN overexpression was significantly associated with poor prognosis in CRC specimens. SRGN overexpression promoted CRC cell migration and invasion in vitro; however, SRGN depletion exhibited contrasting effects. Mechanistic investigations revealed that HIF-1α regulated SRGN transcription via physically binding to a hypoxia response element in its promoter region. CONCLUSIONS: In conclusion, we demonstrated that dysregulated HIF-1α/SRGN signaling promotes CRC progression and metastasis. SRGN may serve as a potential candidate therapeutic target for metastatic CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteoglicanos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Anciano , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Persona de Mediana Edad , Pronóstico , Regiones Promotoras Genéticas , Unión Proteica , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Tasa de Supervivencia , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Proteínas de Transporte Vesicular/genética
6.
Invest Ophthalmol Vis Sci ; 59(2): 930-939, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29450540

RESUMEN

Purpose: Ocular neovascularization (NV) is a pathologic process characterized by the proliferation and infiltration of various types of cells such as RPE, glial, and endothelial cells, which interact with proangiogenic factors and inflammatory cytokines. Endocan is known to be enriched in retinal endothelial tip cells under hypoxia, but the effect of endocan on ocular NV progression is largely unknown. In this study, we investigated the role of endocan in the ocular NV pathologic process and the possible mechanisms involved. Methods: In the eyes of mice with oxygen-induced retinopathy (OIR); choroidal NV (CNV); and rhodopsin promoter (rho)/VEGF transgenic mice, endocan expression was assessed by quantitative real-time PCR (RT-PCR) and Western blot. In vivo, a specific functional antibody was used to neutralize endocan and ocular NV levels were evaluated by RT-PCR, Western blot and immunostaining of flat-mounts. In vitro, the effect of endocan on human retinal microvascular endothelial cell (HREC) tube formation was observed using a routine method. Results: Endocan was significantly elevated in these three experimental mice models. Endocan blockade with the neutralizer intravitreal injection not only suppressed the area of retinal, choroidal and subretinal NV, but also resulted in a decrease in several angiogenesis-associated molecules. Recombinant endocan protein (rhEndocan) was found to induce tube formation on HRECs directly. Conclusions: The current data suggest that endocan is a potential therapeutic or an additional target for retinal and subretinal NV diseases.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Neovascularización Coroidal/prevención & control , Modelos Animales de Enfermedad , Proteoglicanos/antagonistas & inhibidores , Neovascularización Retiniana/prevención & control , Animales , Western Blotting , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Técnica del Anticuerpo Fluorescente Indirecta , Regulación de la Expresión Génica/fisiología , Inyecciones Intravítreas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Neoplasias/farmacología , Proteoglicanos/genética , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/farmacología , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Vasos Retinianos/citología
7.
Biochem Biophys Res Commun ; 494(1-2): 82-87, 2017 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-29050936

RESUMEN

TGF-ß type III receptor (TßRIII) is a co-receptor for TGFß family members required for high-affinity binding of these ligands to their receptors, potentiating their cellular functions. TGF-ßs, Bone Morphogenetic Proteins (BMP2/4) and Inhibins/Activins regulate different checkpoints during T cell differentiation. We have previously reported that TßRIII modulates T cell development by protecting developing thymocytes from apoptosis, however the role of this co-receptor in peripheral lymphocytes still remains elusive. Here we describe a detailed characterization of TßRIII expression in murine and human lymphocyte subpopulations demonstrating that this co-receptor is significantly expressed in T but not B lymphocytes and among them, preferentially expressed on naïve and central memory T cells. TßRIII was upregulated after TCR stimulation, in parallel to other early activation markers. In contrast, natural and induced Tregs downregulated TßRIII in association with FoxP3 upregulation. Finally, anti-TßRIII blocking experiments demonstrated that TßRIII promotes TGFß-dependent iTreg conversion in vitro, and suggest that this co-receptor may be involved in modulating peripheral T cell tolerance and could be considered as a potential target to boost T cell immune responses.


Asunto(s)
Proteoglicanos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Humanos , Memoria Inmunológica , Técnicas In Vitro , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/metabolismo , Regulación hacia Arriba
8.
Biol Trace Elem Res ; 176(2): 270-277, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27565798

RESUMEN

Keloids are fibroproliferative disorders characterized by the overabundant deposition of extracellular matrix (ECM), especially collagen and overgrowth of scar tissue in response to cutaneous injury. In this study, we isolated a selenium (Se)-containing polysaccharide (Se-ZGTP-I) from Ziyang green tea and explored its potential therapeutic effects on keloid fibroblasts formation. 3-(4,5-Dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and annexin V/propidium iodide (PI) staining assays demonstrated that Se-ZGTP-I or neuron-glia 2 (NG2) short hairpin RNA (shRNA) significantly inhibited proliferation of human keloid fibroblasts via induction of apoptosis. Besides, the activation of caspase-3 and the subsequent cleavage of poly (ADP-ribose) polymerase (PARP) were observed in keloid fibroblasts following Se-ZGTP-I (200 and 400 µg/ml) or NG2 shRNA treatment. Moreover, Western blotting analysis showed that treatment of keloid fibroblasts with Se-ZGTP-I (200 and 400 µg/ml) or NG2 shRNA resulted in an increase of pro-apoptotic protein Bax expression and a decrease in expression levels of anti-apoptotic protein Bcl-2 and NG2. In addition, type I collagen biosynthesis and protein expression in keloid fibroblasts following TGF-ß1 stimulation were decreased by Se-ZGTP-I (200 and 400 µg/ml) or NG2 shRNA management. Current findings imply that Se-ZGTP-I has a therapeutic potential to intervene and prevent keloid formation and other fibrotic diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Colágeno/biosíntesis , Mitocondrias/efectos de los fármacos , Polisacáridos/farmacología , Proteoglicanos/antagonistas & inhibidores , Selenio/farmacología , Té/química , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Proteína X Asociada a bcl-2/metabolismo , Antígenos , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Polisacáridos/química , Polisacáridos/aislamiento & purificación , Selenio/química , Factor de Crecimiento Transformador beta1/metabolismo
9.
Drug Des Devel Ther ; 10: 2311-21, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27486308

RESUMEN

Prostate cancer is the most diagnosed noncutaneous cancer and ranks as the second leading cause of cancer-related deaths in American males. Metastasis is the primary cause of prostate cancer mortality. Survival rate is only 28% for metastatic patients, but is nearly 100% for patients with localized prostate cancers. Molecular mechanisms that underlie this malignancy remain obscure, and this study investigated the role of SPARC/osteonectin, cwcv, and kazal-like domain proteoglycan 1 (SPOCK1) in prostate cancer progression. Initially, we found that SPOCK1 expression was significantly higher in prostate cancer tissues relative to noncancerous tissues. In particular, SPOCK1 expression was also markedly high in metastatic tissues compared with nonmetastatic cancerous tissues. SPOCK1 expression knockdown by specific short hairpin RNA in PC3 cells was significantly inhibited, whereas SPOCK1 overexpression in RWPE-1 cells promoted cell viability, colony formation in vitro, and tumor growth in vivo. Moreover, the SPOCK1 knockdown in PC3 cells was associated with cell cycle arrest in G0/G1 phase, while the SPOCK1 overexpression in RWPE-1 cells induced cell cycle arrest in S phase. The SPOCK1 knockdown in PC3 cells even increased cell apoptosis. SPOCK1 modulation was also observed to affect cancerous cell proliferation and apoptotic processes in the mouse model of prostate cancer. Additionally, the SPOCK1 knockdown decreased, whereas the SPOCK1 overexpression increased cell migration and invasion abilities in vitro. Injection of SPOCK1-depleted PC3 cells significantly decreased metastatic nodules in mouse lungs. These findings suggest that SPOCK1 is a critical mediator of tumor growth and metastasis in prostate cancer.


Asunto(s)
Neoplasias de la Próstata/metabolismo , Proteoglicanos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias de la Próstata/patología , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , ARN Interferente Pequeño/farmacología , Células Tumorales Cultivadas
10.
Reprod Sci ; 23(3): 302-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26423601

RESUMEN

Uterine leiomyoma are a common benign pelvic tumors composed of modified smooth muscle cells and a large amount of extracellular matrix (ECM). The proteoglycan composition of the leiomyoma ECM is thought to affect pathophysiology of the disease. To test this hypothesis, we examined the abundance (by immunoblotting) and expression (by quantitative real-time polymerase chain reaction) of the proteoglycans biglycan, decorin, and versican in leiomyoma and normal myometrium and determined whether expression is affected by steroid hormones and menstrual phase. Leiomyoma and normal myometrium were collected from women (n = 17) undergoing hysterectomy or myomectomy. In vitro studies were performed on immortalized leiomyoma (UtLM) and normal myometrial (hTERT-HM) cells with and without exposure to estradiol and progesterone. In leiomyoma tissue, abundance of decorin messenger RNA (mRNA) and protein were 2.6-fold and 1.4-fold lower, respectively, compared with normal myometrium. Abundance of versican mRNA was not different between matched samples, whereas versican protein was increased 1.8-fold in leiomyoma compared with myometrium. Decorin mRNA was 2.4-fold lower in secretory phase leiomyoma compared with proliferative phase tissue. In UtLM cells, progesterone decreased the abundance of decorin mRNA by 1.3-fold. Lower decorin expression in leiomyoma compared with myometrium may contribute to disease growth and progression. As decorin inhibits the activity of specific growth factors, its reduced level in the leiomyoma cell microenvironment may promote cell proliferation and ECM deposition. Our data suggest that decorin expression in leiomyoma is inhibited by progesterone, which may be a mechanism by which the ovarian steroids affect leiomyoma growth and disease progression.


Asunto(s)
Decorina/biosíntesis , Leiomioma/metabolismo , Miometrio/metabolismo , Proteoglicanos/biosíntesis , Neoplasias Uterinas/metabolismo , Adulto , Línea Celular Transformada , Línea Celular Tumoral , Decorina/antagonistas & inhibidores , Estradiol/farmacología , Femenino , Humanos , Leiomioma/fisiopatología , Persona de Mediana Edad , Miometrio/efectos de los fármacos , Miometrio/fisiopatología , Progesterona/farmacología , Promegestona/farmacología , Proteoglicanos/antagonistas & inhibidores , Neoplasias Uterinas/fisiopatología
11.
Endocrine ; 52(3): 641-51, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26662185

RESUMEN

Dopamine agonists (DAs) are the first-line treatment of prolactinomas. They function through the dopamine 2 receptor (D2R) in the tumor cells. Endocan, also called endothelial cell-specific molecule-1 (ESM1), has been described as a marker of neoangiogenesis. However, whether ESM1 promotes the resistance of prolactinomas to DA therapy is largely unknown. In our study, 25 patients with prolactinomas were divided into resistant- and sensitive- groups according to the clinical response to bromocriptine. We found that ESM1-microvessel density of resistant prolactinomas was significantly higher than that of sensitive prolactinomas (47.9 ± 11.6, n = 8, vs 13.1 ± 2.8, n = 17, p = 0.0006), indicating that ESM1 was a DA resistance-related gene. Immunostaining showed that ESM1 was expressed in tumor vessels and sporadic tumor cells, and ESM1 was overlapped with the Smooth Muscle Actin (SMA) and von Willebrand Factor (VWF) in the tumor vessels. Silencing of ESM1 markedly suppressed the viability of GH3 and MMQ cells in vitro, and furthermore, significantly increased the sensitivity of GH3 and MMQ cells to DA treatment. Additionally, silencing of ESM1 down-regulated the angiogenesis-associated genes, such as VEGFR2, FGF2, CD34, CD31, VWF, and EGFR. Knockdown of ESM1 decreased endothelial tube formation of HUVECs, and significantly increased the sensitivity of HUVECs to Avastin treatment. Therefore, we first demonstrate that DA resistance-related ESM1 promotes the angiogenesis and tumor cells growth of prolactinomas, suggesting that ESM1 may be a novel therapeutic target for prolactinomas.


Asunto(s)
Agonistas de Dopamina/uso terapéutico , Resistencia a Antineoplásicos/genética , Proteínas de Neoplasias/fisiología , Neovascularización Patológica/genética , Neoplasias Hipofisarias/patología , Prolactinoma/patología , Proteoglicanos/fisiología , Adolescente , Adulto , Anciano , Animales , Bromocriptina/farmacología , Bromocriptina/uso terapéutico , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Agonistas de Dopamina/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Hipofisarias/irrigación sanguínea , Neoplasias Hipofisarias/genética , Prolactinoma/irrigación sanguínea , Prolactinoma/genética , Proteoglicanos/antagonistas & inhibidores , Ratas , Adulto Joven
12.
PLoS One ; 10(9): e0137311, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26340347

RESUMEN

The NG2 proteoglycan is characteristically expressed by oligodendrocyte progenitor cells (OPC) and also by aggressive brain tumours highly resistant to chemo- and radiation therapy. Oligodendrocyte-lineage cells are particularly sensitive to stress resulting in cell death in white matter after hypoxic or ischemic insults of premature infants and destruction of OPC in some types of Multiple Sclerosis lesions. Here we show that the NG2 proteoglycan binds OMI/HtrA2, a mitochondrial serine protease which is released from damaged mitochondria into the cytosol in response to stress. In the cytosol, OMI/HtrA2 initiates apoptosis by proteolytic degradation of anti-apoptotic factors. OPC in which NG2 has been downregulated by siRNA, or OPC from the NG2-knockout mouse show an increased sensitivity to oxidative stress evidenced by increased cell death. The proapoptotic protease activity of OMI/HtrA2 in the cytosol can be reduced by the interaction with NG2. Human glioma expressing high levels of NG2 are less sensitive to oxidative stress than those with lower NG2 expression and reducing NG2 expression by siRNA increases cell death in response to oxidative stress. Binding of NG2 to OMI/HtrA2 may thus help protect cells against oxidative stress-induced cell death. This interaction is likely to contribute to the high chemo- and radioresistance of glioma.


Asunto(s)
Antígenos/metabolismo , Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Proteínas Mitocondriales/metabolismo , Proteoglicanos/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Antígenos/genética , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Cerebelo/patología , Citosol/efectos de los fármacos , Citosol/metabolismo , Glioblastoma/genética , Glioblastoma/patología , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Estrés Oxidativo , Cultivo Primario de Células , Unión Proteica , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal
13.
Biochim Biophys Acta ; 1855(2): 276-300, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25829250

RESUMEN

Proteoglycans control numerous normal and pathological processes, among which are morphogenesis, tissue repair, inflammation, vascularization and cancer metastasis. During tumor development and growth, proteoglycan expression is markedly modified in the tumor microenvironment. Altered expression of proteoglycans on tumor and stromal cell membranes affects cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. Despite the high complexity and heterogeneity of breast cancer, the rapid evolution in our knowledge that proteoglycans are among the key players in the breast tumor microenvironment suggests their potential as pharmacological targets in this type of cancer. It has been recently suggested that pharmacological treatment may target proteoglycan metabolism, their utilization as targets for immunotherapy or their direct use as therapeutic agents. The diversity inherent in the proteoglycans that will be presented herein provides the potential for multiple layers of regulation of breast tumor behavior. This review summarizes recent developments concerning the biology of selected proteoglycans in breast cancer, and presents potential targeted therapeutic approaches based on their novel key roles in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neovascularización Patológica/genética , Proteoglicanos/biosíntesis , Investigación Biomédica Traslacional , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/uso terapéutico , Transducción de Señal/genética , Microambiente Tumoral/genética
14.
Exp Cell Res ; 335(1): 82-90, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25882497

RESUMEN

After CNS injury, astrocytes and mesenchymal cells attempt to restore the disrupted glia limitans by secreting proteoglycans and extracellular matrix proteins (ECMs), forming the so-called glial scar. Although the glial scar is important in sealing the lesion, it is also a physical and functional barrier that prevents axonal regeneration. The synthesis of secretory proteins in the RER is under the control of the initiation factor of translation eIF2α. Inhibiting the synthesis of secretory proteins by increasing the phosphorylation of eIF2α, might be a pharmacologically efficient way of reducing proteoglycans and other profibrotic proteins present in the glial scar. Salubrinal, a neuroprotective drug, decreased the expression and secretion of proteoglycans and other profibrotic proteins induced by EGF or TGFß, maintaining eIF2α phosphorylated. Besides, Salubrinal also reduced the transcription of proteoglycans and other profibrotic proteins, suggesting that it induced the degradation of non-translated mRNA. In a model in vitro of the glial scar, cortical neurons grown on cocultures of astrocytes and fibroblasts with TGFß treated with Salubrinal, showed increased neurite outgrowth compared to untreated cells. Our results suggest that Salubrinal may be considered of therapeutic value facilitating axonal regeneration, by reducing overproduction and secretion of proteoglycans and profibrotic protein inhibitors of axonal growth.


Asunto(s)
Corteza Cerebral/fisiología , Cinamatos/farmacología , Proteínas de la Matriz Extracelular/biosíntesis , Regeneración Nerviosa/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Proteoglicanos/antagonistas & inhibidores , Tiourea/análogos & derivados , Animales , Astrocitos/metabolismo , Células Cultivadas , Corteza Cerebral/lesiones , Técnicas de Cocultivo , Fibroblastos/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Neuritas/fisiología , Neuroglía/metabolismo , Fosforilación , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteoglicanos/biosíntesis , ARN Mensajero/metabolismo , Tiourea/farmacología , Factor de Crecimiento Transformador beta/farmacología
15.
Theranostics ; 5(5): 530-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25767619

RESUMEN

NG2/CSPG4 is an unusual cell-membrane integral proteoglycan widely recognized to be a prognostic factor, a valuable tool for ex vivo and non-invasive molecular diagnostics and, by virtue of its tight association with malignancy, a tantalizing therapeutic target in several tumour types. Although the biology behind its involvement in cancer progression needs to be better understood, implementation of NG2/CSPG4 in the routine clinical practice is attainable and has the potential to contribute to an improved individualized management of cancer patients. In this context, its polymorphic nature seems to be particularly valuable in the effort to standardize informative diagnostic procedures and consolidate forcible immunotherapeutic treatment strategies. We discuss here the underpinnings for this potential and highlight the benefits of taking advantage of the intra-tumour and inter-patient variability in the regulation of NG2/CSPG4 expression. We envision that NG2/CSPG4 may effectively be exploited in therapeutic interventions aimed at averting resistance to target therapy agents and at interfering with secondary lesion formation and/or tumour recurrence.


Asunto(s)
Antígenos/análisis , Pruebas Diagnósticas de Rutina/métodos , Inmunoterapia/métodos , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/análisis , Humanos
16.
Cell Signal ; 27(3): 453-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25499979

RESUMEN

During embryogenesis, the epicardium undergoes proliferation, migration, and differentiation into several cardiac cell types which contribute to the coronary vessels. This process requires epithelial to mesenchymal transition (EMT) and directed cellular invasion. The Type III Transforming Growth Factor-beta Receptor (TGFßR3) is required for epicardial cell invasion and coronary vessel development. Using primary epicardial cells derived from Tgfbr3(+/+) and Tgfbr3(-/-) mouse embryos, high-molecular weight hyaluronan (HMWHA) stimulated cellular invasion and filamentous (f-actin) polymerization are detected in Tgfbr3(+/+) cells, but not in Tgfbr3(-/-) cells. Furthermore, HMWHA-stimulated cellular invasion and f-actin polymerization in Tgfbr3(+/+) epicardial cells are dependent on Src kinase. Src activation in HMWHA-stimulated Tgfbr3(-/-) epicardial cells is not detected in response to HMWHA. RhoA and Rac1 also fail to activate in response to HMWHA in Tgfbr3(-/-) cells. These events coincide with defective f-actin formation and deficient cellular invasion. Finally, a T841A activating substitution in TGFßR3 drives ligand-independent Src activation. Collectively, these data define a TGFßR3-Src-RhoA/Rac1 pathway that is essential for hyaluronan-directed cell invasion in epicardial cells.


Asunto(s)
Ácido Hialurónico/farmacología , Pericardio/efectos de los fármacos , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Familia-src Quinasas/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Arrestina/química , Arrestina/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Transición Epitelial-Mesenquimal , Ratones , Neuropéptidos/metabolismo , Pericardio/citología , Pericardio/metabolismo , Unión Proteica , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
17.
Cell Res ; 25(2): 157-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25547119

RESUMEN

As a gram-positive, spore-forming anaerobic bacillus, Clostridium difficile (C. difficile) is responsible for severe and fatal pseudomembranous colitis, and poses the most urgent antibiotic resistance threat worldwide. Epidemic C. difficile is the leading cause of antibiotic-associated diarrhoea globally, especially diarrhoea due to the emergence of hypervirulent strains associated with high mortality and morbidity. TcdB, one of the key virulence factors secreted by this bacterium, enters host cells through a poorly understood mechanism to elicit its pathogenic effect. Here we report the first identification of the TcdB cellular receptor, chondroitin sulfate proteoglycan 4 (CSPG4). CSPG4 was initially isolated from a whole-genome human shRNAmir library screening, and its role was confirmed by both TALEN- and CRISPR/Cas9-mediated gene knockout in human cells. CSPG4 is critical for TcdB binding to the cell surface, inducing cytoskeleton disruption and cell death. A direct interaction between the N-terminus of CSPG4 and the C-terminus of TcdB was confirmed, and the soluble peptide of the toxin-binding domain of CSPG4 could protect cells from the action of TcdB. Notably, the complete loss of CSPG4/NG2 decreased TcdB-triggered interleukin-8 induction in mice without significantly affecting animal mortality. Based on both the in vitro and in vivo studies, we propose a dual-receptor model for TcdB endocytosis. The discovery of the first TcdB receptor reveals a previously unsuspected role for CSPG4 and provides a new therapeutic target for the treatment of C. difficile infection.


Asunto(s)
Antígenos/metabolismo , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Proteoglicanos/metabolismo , Animales , Antígenos/química , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Secuencia de Bases , Infecciones por Clostridium/metabolismo , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/patología , Citoesqueleto/metabolismo , Endocitosis , Técnicas de Inactivación de Genes , Células HEK293 , Células HT29 , Células HeLa , Humanos , Interleucina-8/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Estructura Terciaria de Proteína , Proteoglicanos/antagonistas & inhibidores , Proteoglicanos/química , ARN Interferente Pequeño/metabolismo
18.
Arthritis Res Ther ; 16(6): 484, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25465897

RESUMEN

INTRODUCTION: The objective of this study was to investigate the possible role of UDP-glucose dehydrogenase (UGDH) in osteoarthritis (OA) and uncover whether, furthermore how interleukin-1beta (IL-1ß) affects UGDH gene expression. METHODS: UGDH specific siRNAs were applied to determine the role of UGDH in proteoglycan (PG) synthesis in human articular chondrocytes. Protein levels of UGDH and Sp1 in human and rat OA cartilage were detected. Then, human primary chondrocytes were treated with IL-1ß to find out whether and how IL-1ß could regulate the gene expression of UGDH and its trans-regulators, that is Sp1, Sp3 and c-Krox. Finally, p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580 and stress-activated protein kinase/c-Jun N-terminal kinase (SAP/JNK) inhibitor SP600125 were used to pick out the pathway that mediated the IL-1ß-modulated PGs synthesis and gene expression of UGDH, Sp1, Sp3 and c-Krox. RESULTS: UGDH specific siRNAs markedly inhibited UGDH mRNA and protein expression, and thus led to an obvious suppression of PGs synthesis in human articular chondrocytes. UGDH protein level in human and rat OA cartilage were much lower than the corresponding controls and negatively correlated to the degree of OA. Decrease in Sp1 protein level was also observed in human and rat OA cartilage respectively. Meanwhile, IL-1ß suppressed UGDH gene expression in human articular chondrocytes in the late phase, which also modulated gene expression of Sp1, Sp3 and c-Krox and increased both Sp3/Sp1 and c-Krox/Sp1 ratio. Moreover, the inhibition of SAP/JNK and p38 MAPK pathways both resulted in an obvious attenuation of the IL-1ß-induced suppression on the UGDH gene expression. CONCLUSIONS: UGDH is essential in the PGs synthesis of articular chondrocytes, while the suppressed expression of UGDH might probably be involved in advanced OA, partly due to the modulation of p38 MAPK and SAP/JNK pathways and its trans-regulators by IL-1ß.


Asunto(s)
Cartílago Articular/metabolismo , Condrocitos/metabolismo , Osteoartritis/metabolismo , Proteoglicanos/biosíntesis , Uridina Difosfato Glucosa Deshidrogenasa/biosíntesis , Anciano , Animales , Cartílago Articular/patología , Células Cultivadas , Condrocitos/patología , Femenino , Humanos , Persona de Mediana Edad , Osteoartritis/patología , Proteoglicanos/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Ratas , Ratas Wistar , Uridina Difosfato Glucosa Deshidrogenasa/antagonistas & inhibidores
19.
PLoS One ; 9(9): e108414, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25268630

RESUMEN

There are currently no established radiological parameters that predict response to immunotherapy. We hypothesised that multiparametric, longitudinal magnetic resonance imaging (MRI) of physiological parameters and pharmacokinetic models might detect early biological responses to immunotherapy for glioblastoma targeting NG2/CSPG4 with mAb9.2.27 combined with natural killer (NK) cells. Contrast enhanced conventional T1-weighted MRI at 7±1 and 17±2 days post-treatment failed to detect differences in tumour size between the treatment groups, whereas, follow-up scans at 3 months demonstrated diminished signal intensity and tumour volume in the surviving NK+mAb9.2.27 treated animals. Notably, interstitial volume fraction (ve), was significantly increased in the NK+mAb9.2.27 combination therapy group compared mAb9.2.27 and NK cell monotherapy groups (p = 0.002 and p = 0.017 respectively) in cohort 1 animals treated with 1 million NK cells. ve was reproducibly increased in the combination NK+mAb9.2.27 compared to NK cell monotherapy in cohort 2 treated with increased dose of 2 million NK cells (p<0.0001), indicating greater cell death induced by NK+mAb9.2.27 treatment. The interstitial volume fraction in the NK monotherapy group was significantly reduced compared to mAb9.2.27 monotherapy (p<0.0001) and untreated controls (p = 0.014) in the cohort 2 animals. NK cells in monotherapy were unable to kill the U87MG cells that highly expressed class I human leucocyte antigens, and diminished stress ligands for activating receptors. A significant association between apparent diffusion coefficient (ADC) of water and ve in combination NK+mAb9.2.27 and NK monotherapy treated tumours was evident, where increased ADC corresponded to reduced ve in both cases. Collectively, these data support histological measures at end-stage demonstrating diminished tumour cell proliferation and pronounced apoptosis in the NK+mAb9.2.27 treated tumours compared to the other groups. In conclusion, ve was the most reliable radiological parameter for detecting response to intralesional NK cellular therapy.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Inmunoterapia Adoptiva , Células Asesinas Naturales/trasplante , Proteoglicanos/antagonistas & inhibidores , Animales , Antígenos/genética , Antígenos/inmunología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Difusión , Modelos Animales de Enfermedad , Líquido Extracelular/química , Líquido Extracelular/efectos de los fármacos , Líquido Extracelular/metabolismo , Femenino , Expresión Génica , Glioblastoma/genética , Glioblastoma/inmunología , Glioblastoma/patología , Aumento de la Imagen , Inyecciones Intralesiones , Células Asesinas Naturales/inmunología , Imagen por Resonancia Magnética , Masculino , Terapia Molecular Dirigida , Proteoglicanos/genética , Proteoglicanos/inmunología , Ratas , Ratas Desnudas , Carga Tumoral/efectos de los fármacos
20.
Oncotarget ; 4(9): 1527-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24127551

RESUMEN

Glioblastoma (GBM) is the most malignant brain tumor where patients' survival is only 14.6 months, despite multimodal therapy with debulking surgery, concurrent chemotherapy and radiotherapy. There is an urgent, unmet need for novel, effective therapeutic strategies for this devastating disease. Although several immunotherapies are under development for the treatment of GBM patients, the use of natural killer (NK) cells is still marginal despite this being a promising approach to treat cancer. In regard of our knowledge on the role of NG2/CSPG4 in promoting GBM aggressiveness we investigated the potential of an innovative immunotherapeutic strategy combining mAb9.2.27 against NG2/CSPG4 and NK cells in preclinical animal models of GBM. Multiple immune escape mechanisms maintain the tumor microenvironment in an anti-inflammatory state to promote tumor growth, however, the distinct roles of resident microglia versus recruited macrophages is not elucidated. We hypothesized that exploiting the cytokine release capabilities of activated (NK) cells to reverse the anti-inflammatory axis combined with mAb9.2.27 targeting the NG2/CSPG4 may favor tumor destruction by editing pro-GBM immune responses. Combination treatment with NK+mAb9.2.27 diminished tumor growth that was associated with reduced tumor proliferation, increased cellular apoptosis and prolonged survival compared to vehicle and monotherapy controls. The therapeutic efficacy was mediated by recruitment of CCR2low macrophages into the tumor microenvironment, increased ED1 and MHC class II expression on microglia that might render them competent for GBM antigen presentation, as well as elevated IFN-γ and TNF-α levels in the cerebrospinal fluid compared to controls. Depletion of systemic macrophages by liposome-encapsulated clodronate decreased the CCR2low macrophages recruited to the brain and abolished the beneficial outcomes. Moreover, mAb9.2.27 reversed tumor-promoting effects of patient-derived tumor-associated macrophage/microglia(TAM) ex vivo.Taken together, these findings indicate thatNK+mAb9.2.27 treatment may be an amenable therapeutic strategy to treat NG2/CSPG4 expressing GBMs. We provide a novel conceptual approach of combination immunotherapy for glioblastoma. The results traverse beyond the elucidation of NG2/CSPG4 as a therapeutic target, but demonstrate a proof of concept that this antibody may hold potential for the treatment of GBM by activation of tumor infiltrated microglia/macrophages.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Células Asesinas Naturales/inmunología , Proteoglicanos/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Procesos de Crecimiento Celular/inmunología , Glioblastoma/inmunología , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/patología , Persona de Mediana Edad , Proteoglicanos/inmunología , Ratas , Análisis de Supervivencia , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...