Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 274: 116217, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38489904

RESUMEN

The aim of this study is to conduct a thorough evaluation of the association between Benzophenone-3 (BP-3) exposure and OA, offering critical insights into the underlying mechanisms involved. The National Health and Nutrition Examination Survey (NHANES) database was utilized to investigate the correlation between BP-3 and osteoarthritis. Proteomic sequencing from clinical sample and the PharmMapper online tool were employed to predict the biological target of BP-3. Cellular molecular assays and transfection studies were performed to verify the prediction from bioinformatics analyses. Through cross-sectional analysis of the NHANES database, we identified BP-3 as a risk factor for OA development. The results of proteomic sequencing showed that Secreted Protein Acidic and Rich in Cysteine (SPARC) was significantly elevated in the area of damage compared to the undamaged area. SPARC was also among the potential biological targets of BP-3 predicted by the online program. Through in vitro cell experiments, we further determined that the toxicological effects of BP-3 may be due to SPARC, which elevates intracellular GPX4 levels, activates the glutathione system, and promotes lipid peroxidation to mitigate ferroptosis. Inhibiting SPARC expression has been shown to reduce inflammation and ferroptosis in OA contexts. This research provides an expansive understanding of BP-3's influence on osteoarthritis development. We have identified SPARC as a potent target for combating chondrocyte ferroptosis in BP-3-associated osteoarthritis.


Asunto(s)
Benzofenonas , Ferroptosis , Osteoartritis , Osteonectina , Humanos , Benzofenonas/metabolismo , Benzofenonas/toxicidad , Biología Computacional , Estudios Transversales , Ferroptosis/efectos de los fármacos , Encuestas Nutricionales , Osteoartritis/inducido químicamente , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Osteonectina/metabolismo , Proteómica
2.
Biochem Biophys Res Commun ; 558: 134-140, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33910127

RESUMEN

Previous studies have shown that secreted protein acidic and rich in cysteine (SPARC) proteins can inhibit the development of cancer cells in various ways, such as by inhibiting angiogenesis and inhibiting cell proliferation. In fact, SPARC proteins may have an effect on the chemoresistance of gastric cancer cells to 5-Fluorouracil (5-FU), which needs further research in the future. Therefore, the purpose of this study was to explore the relationship between SPARC proteins and the chemosensitivity of gastric cancer cells to 5-FU. In vitro, after SPARC protein levels were regulated by plasmid, siRNA and human recombinant SPARC protein transfection in MGC-803, SGC-7901 and BGC-823 cells, we detected epithelial-mesenchymal transition (EMT), apoptosis markers and cell viability after 5-FU treatment. In vivo, we implanted BGC-823 cells with stable SPARC overexpression into nude mice. Tumour size was measured to assess the effect of SPARC protein on tumour formation and 5-FU chemosensitivity. In SGC-7901 and BGC-823 cells, both endogenous and exogenous upregulation of SPARC protein levels decreased cell viability, destroyed cytoskeletal F-actin, inhibited cell migration, and downregulated a series of transcription factors to inhibit cell EMT; it also upregulated cell apoptosis-related proteins to promote cell apoptosis. However, we obtained opposite results in SPARC knockdown MGC-803 cells. In vivo, compared with the control group, the group engrafted with BGC-823 cells stably overexpressing SPARC had a significant smaller tumour size. After 5-FU treatment, the new tumour gradually decreased in size. Our results show that the SPARC protein could enhance 5-FU chemosensitivity in gastric cancer cell lines by inhibiting EMT and promoting cell apoptosis.


Asunto(s)
Fluorouracilo/farmacología , Osteonectina/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Actinas/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Interferente Pequeño/genética , Neoplasias Gástricas/patología , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Mol Life Sci ; 75(6): 1001-1012, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28993833

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is among the most devastating human malignancies, with approximately 20-30% of PDAC patients receiving the surgical resection with curative intent. Although many studies have focused on finding ideal "drug chaperones" that facilitate and/or potentiate the effects of gemcitabine (GEM) in pancreatic cancer, a significant benefit in overall survival could not be demonstrated for any of these combination therapies in PDAC. Given that pancreatic cancer is characterized by desmoplasia and the dual biological roles of stroma in pancreatic cancer, we reassess the importance of stroma in GEM-based therapeutic approaches in light of current findings. This review is focused on understanding the role of stromal components in the extrinsic resistance to GEM and whether anti-stroma therapies have a positive effect on the GEM delivery. This work contributes to the development of novel and promising combination GEM-based regimens that have achieved significant survival benefits for the patients with pancreatic cancer.


Asunto(s)
Albúminas/uso terapéutico , Anilidas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Regulación Neoplásica de la Expresión Génica , Paclitaxel/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Piridinas/uso terapéutico , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/cirugía , Desoxicitidina/uso terapéutico , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Humanos , Ácido Hialurónico/metabolismo , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Osteonectina/metabolismo , Pancreatectomía , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/patología , Análisis de Supervivencia , Resultado del Tratamiento , Gemcitabina
4.
J Surg Oncol ; 116(1): 7-15, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28605029

RESUMEN

The tumor microenvironment in pancreatic cancer is a complex balance of pro- and anti-tumor components. The dense desmoplasia consists of immune cells, extracellular matrix, growth factors, cytokines, and cancer associated fibroblasts (CAF) or pancreatic stellate cells (PSC). There are a multitude of targets including hyaluronan, angiogenesis, focal adhesion kinase (FAK), connective tissue growth factor (CTGF), CD40, chemokine (C-X-C motif) receptor 4 (CXCR-4), immunotherapy, and Vitamin D. The developing clinical therapeutics will be reviewed.


Asunto(s)
Neoplasias Pancreáticas/patología , Microambiente Tumoral , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Inhibidores de la Angiogénesis/farmacología , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Fibroblastos Asociados al Cáncer/patología , Carcinogénesis , Transformación Celular Neoplásica , Ensayos Clínicos como Asunto , Factor de Crecimiento del Tejido Conjuntivo/antagonistas & inhibidores , Proteína-Tirosina Quinasas de Adhesión Focal/antagonistas & inhibidores , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Ácido Hialurónico/metabolismo , Inmunoterapia , Macrófagos/patología , Células Mieloides/patología , Neutrófilos/patología , Osteonectina/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Células Estrelladas Pancreáticas/patología , Vitamina D/farmacología
5.
PLoS One ; 11(9): e0162698, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27622658

RESUMEN

TGFBI has been shown to sensitize ovarian cancer cells to the cytotoxic effects of paclitaxel via an integrin receptor-mediated mechanism that modulates microtubule stability. Herein, we determine that TGFBI localizes within organized fibrillar structures in mesothelial-derived ECM. We determined that suppression of SPARC expression by shRNA decreased the deposition of TGFBI in mesothelial-derived ECM, without affecting its overall protein expression or secretion. Conversely, overexpression of SPARC increased TGFBI deposition. A SPARC-YFP fusion construct expressed by the Met5a cell line co-localized with TGFBI in the cell-derived ECM. Interestingly, in vitro produced SPARC was capable of precipitating TGFBI from cell lysates dependent on an intact SPARC carboxy-terminus with in vitro binding assays verifying a direct interaction. The last 37 amino acids of SPARC were shown to be required for the TGFBI interaction while expression of a SPARC-YFP construct lacking this region (aa 1-256) did not interact and co-localize with TGFBI in the ECM. Furthermore, ovarian cancer cells have a reduced motility and decreased response to the chemotherapeutic agent paclitaxel when plated on ECM derived from mesothelial cells lacking SPARC compared to control mesothelial-derived ECM. In conclusion, SPARC regulates the fibrillar ECM deposition of TGFBI through a novel interaction, subsequently influencing cancer cell behavior.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Proteínas de la Matriz Extracelular/metabolismo , Osteonectina/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Paclitaxel/farmacología , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células Epiteliales/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibronectinas/metabolismo , Humanos , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
6.
Int J Oncol ; 48(5): 1765-71, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26983777

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most clinically challenging cancers to manage. An estimated 48,960 people will be diagnosed with pancreatic cancer in 2015, of that population, 94% are projected to perish within 5 years. These dismal survival rates can be attributed, in part, to an advanced diagnosis occurring in 80% of cases. The heterogeneous and dynamic microenvironment of pancreatic cancer, and the lack of both specific risk factors and efficacious screening tools contribute to the challenge of diagnosing pancreatic cancer in its early stages. These clinical challenges have directed research into the unique characteristics that define PDAC. Recently, there has been an increased focus on the interaction of tumor cells with their microenvironment in the hope of identifying new therapeutic targets. One of the most promising avenues in this new vein of research is targeting protein communication between the cancer cells and the extracellular matrix. The secreted protein acidic and rich in cysteine (SPARC) is one such extracellular matrix protein that has shown potential as a therapeutic target due to its influence on PDAC invasion and metastasis. In this review, we discuss the complex interaction of SPARC with PDAC cells and its potential to guide treatment and eventually improve the survival of patients diagnosed with this devastating disease.


Asunto(s)
Carcinoma Ductal Pancreático/diagnóstico , Osteonectina/antagonistas & inhibidores , Osteonectina/análisis , Neoplasias Pancreáticas/diagnóstico , Carcinoma Ductal Pancreático/metabolismo , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica , Humanos , Osteonectina/metabolismo , Neoplasias Pancreáticas/metabolismo , Pronóstico , Análisis de Supervivencia , Microambiente Tumoral
7.
Mol Med Rep ; 12(2): 2926-32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25936899

RESUMEN

Bufalin is an active compound in the traditional Chinese medicine Chan Su, which has been shown to induce apoptosis in a range of cancer cell types. However, certain gastric cancer cells are known to be resistant to bufalin. Intracellular secreted protein acidic and rich in cysteine (SPARC) regulates proliferation and apoptosis. This study aimed to evaluate the role of SPARC in bufalin-induced apoptosis in SGC7901 and MGC803 gastric cancer cells. SGC7901 cells with high SPARC expression were more resistant to bufalin than MGC803 cells with low SPARC expression. This resistance was significantly reversed by small interfering (si)RNA-mediated knockdown of SPARC. Furthermore, it was shown that SPARC negatively regulated bufalin-induced intrinsic apoptosis by protecting mitochondrial integrity, decreasing the release of cytoplasmic cytochrome c and increasing the ratio of Bcl-2/Bax. In addition, SPARC overcame bufalin-induced G2/M phase arrest by increasing levels of Cyclin B1 and Cyclin A protein expression. SPARC also activated cellular survival signals, including Src and Akt, but not extracellular signal-regulated kinase. This study demonstrated that SPARC antagonizes bufalin-induced apoptosis via inhibition of the intrinsic apoptosis pathway, inhibition of cell cycle arrest and activation of certain pathways involved in proliferation. This provides novel evidence for SPARC as a potential target by which to sensitize gastric cancer cells to bufalin.


Asunto(s)
Apoptosis/efectos de los fármacos , Bufanólidos/toxicidad , Osteonectina/metabolismo , Bufanólidos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Ciclina B1/metabolismo , Citocromos c/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Medicina Tradicional China , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/metabolismo , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proteína X Asociada a bcl-2/metabolismo , Familia-src Quinasas/metabolismo
8.
Cell Rep ; 9(1): 104-117, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25284788

RESUMEN

Metastatic colonization of distant organs underpins the majority of human-cancer-related deaths, including deaths from head and neck squamous cell carcinoma (HNSCC). We report that miR-203, a miRNA that triggers differentiation in multilayered epithelia, inhibits multiple postextravasation events during HNSCC lung metastasis. Inducible reactivation of miR-203 in already established lung metastases reduces the overall metastatic burden. Using an integrated approach, we reveal that miR-203 inhibits metastasis independently of its effects on differentiation. In vivo genetic reconstitution experiments show that miR-203 inhibits lung metastasis by suppressing the prometastatic activities of three factors involved in cytoskeletal dynamics (LASP1), extracellular matrix remodeling (SPARC), and cell metabolism (NUAK1). Expression of miR-203 and its downstream effectors correlates with HNSCC overall survival outcomes, indicating the therapeutic potential of targeting this signaling axis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , MicroARNs/genética , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proteínas del Citoesqueleto/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/metabolismo , Xenoinjertos , Humanos , Proteínas con Dominio LIM/antagonistas & inhibidores , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Metástasis de la Neoplasia , Osteonectina/antagonistas & inhibidores , Pronóstico , Proteínas Quinasas , Proteínas Represoras/antagonistas & inhibidores , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello
9.
Cell Rep ; 8(6): 1905-1918, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25242334

RESUMEN

Circulating tumor cells (CTCs) are shed from primary tumors into the bloodstream, mediating the hematogenous spread of cancer to distant organs. To define their composition, we compared genome-wide expression profiles of CTCs with matched primary tumors in a mouse model of pancreatic cancer, isolating individual CTCs using epitope-independent microfluidic capture, followed by single-cell RNA sequencing. CTCs clustered separately from primary tumors and tumor-derived cell lines, showing low-proliferative signatures, enrichment for the stem-cell-associated gene Aldh1a2, biphenotypic expression of epithelial and mesenchymal markers, and expression of Igfbp5, a gene transcript enriched at the epithelial-stromal interface. Mouse as well as human pancreatic CTCs exhibit a very high expression of stromal-derived extracellular matrix (ECM) proteins, including SPARC, whose knockdown in cancer cells suppresses cell migration and invasiveness. The aberrant expression by CTCs of stromal ECM genes points to their contribution of microenvironmental signals for the spread of cancer to distant organs.


Asunto(s)
Matriz Extracelular/genética , Regulación Neoplásica de la Expresión Génica , Células Neoplásicas Circulantes/metabolismo , Neoplasias Pancreáticas/patología , Familia de Aldehído Deshidrogenasa 1 , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Movimiento Celular , Matriz Extracelular/metabolismo , Humanos , Ratones , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Osteonectina/metabolismo , Neoplasias Pancreáticas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Análisis de Secuencia de ARN , Células Tumorales Cultivadas
10.
J Clin Invest ; 124(4): 1512-24, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24590286

RESUMEN

Aberrant expression of the secreted protein, acidic, cysteine-rich (osteonectin) (SPARC) gene, which encodes a matricellular protein that participates in normal tissue remodeling, is associated with a variety of diseases including cancer, but the contribution of SPARC to malignant growth remains controversial. We previously reported that SPARC was among the most upregulated genes in cytogenetically normal acute myeloid leukemia (CN-AML) patients with gene-expression profiles predictive of unfavorable outcome, such as mutations in isocitrate dehydrogenase 2 (IDH2-R172) and overexpression of the oncogenes brain and acute leukemia, cytoplasmic (BAALC) and v-ets erythroblastosis virus E26 oncogene homolog (ERG). In contrast, SPARC was downregulated in CN-AML patients harboring mutations in nucleophosmin (NPM1) that are associated with favorable prognosis. Based on these observations, we hypothesized that SPARC expression is clinically relevant in AML. Here, we found that SPARC overexpression is associated with adverse outcome in CN-AML patients and promotes aggressive leukemia growth in murine models of AML. In leukemia cells, SPARC expression was mediated by the SP1/NF-κB transactivation complex. Furthermore, secreted SPARC activated the integrin-linked kinase/AKT (ILK/AKT) pathway, likely via integrin interaction, and subsequent ß-catenin signaling, which is involved in leukemia cell self-renewal. Pharmacologic inhibition of the SP1/NF-κB complex resulted in SPARC downregulation and leukemia growth inhibition. Together, our data indicate that evaluation of SPARC expression has prognosticative value and SPARC is a potential therapeutic target for AML.


Asunto(s)
Leucemia Mieloide Aguda/etiología , Osteonectina/fisiología , Adolescente , Adulto , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , FN-kappa B/metabolismo , Nucleofosmina , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Pronóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Adulto Joven , beta Catenina/metabolismo
11.
Small ; 10(9): 1790-8, 2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24510544

RESUMEN

Efficient and safe delivery systems for siRNA therapeutics remain a challenge. Elevated secreted protein, acidic, and rich in cysteine (SPARC) protein expression is associated with tissue scarring and fibrosis. Here we investigate the feasibility of encapsulating SPARC-siRNA in the bilayers of layer-by-layer (LbL) nanoparticles (NPs) with poly(L-arginine) (ARG) and dextran (DXS) as polyelectrolytes. Cellular binding and uptake of LbL NPs as well as siRNA delivery were studied in FibroGRO cells. siGLO-siRNA and SPARC-siRNA were efficiently coated onto hydroxyapatite nanoparticles. The multilayered NPs were characterized with regard to particle size, zeta potential and surface morphology using dynamic light scattering and transmission electron microscopy. The SPARC-gene silencing and mRNA levels were analyzed using ChemiDOC western blot technique and RT-PCR. The multilayer SPARC-siRNA incorporated nanoparticles are about 200 nm in diameter and are efficiently internalized into FibroGRO cells. Their intracellular fate was also followed by tagging with suitable reporter siRNA as well as with lysotracker dye; confocal microscopy clearly indicates endosomal escape of the particles. Significant (60%) SPARC-gene knock down was achieved by using 0.4 pmole siRNA/µg of LbL NPs in FibroGRO cells and the relative expression of SPARC mRNA reduced significantly (60%) against untreated cells. The cytotoxicity as evaluated by xCelligence real-time cell proliferation and MTT cell assay, indicated that the SPARC-siRNA-loaded LbL NPs are non-toxic. In conclusion, the LbL NP system described provides a promising, safe and efficient delivery platform as a non-viral vector for siRNA delivery that uses biopolymers to enhance the gene knock down efficiency for the development of siRNA therapeutics.


Asunto(s)
Silenciador del Gen , Técnicas de Transferencia de Gen , Nanopartículas/química , Osteonectina/genética , ARN Interferente Pequeño/metabolismo , Endocitosis , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Humanos , Espacio Intracelular/metabolismo , Masculino , Nanopartículas/ultraestructura , Proteínas de Neoplasias/metabolismo , Osteonectina/antagonistas & inhibidores , Osteonectina/biosíntesis , Osteonectina/ultraestructura , Tamaño de la Partícula , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas Ribosómicas/metabolismo , Electricidad Estática
12.
Cancer Metastasis Rev ; 32(3-4): 585-602, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23690170

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) stands as the poorest prognostic tumor of the digestive tract, with a 5-year survival rate of less than 5%. Therapeutic options for unresectable PDAC are extremely limited and there is a pressing need for expanded therapeutic approaches to improve current options available with gemcitabine-based regimens. With PDAC displaying one of the most prominent desmoplastic stromal reactions of all carcinomas, recent research has focused on the microenvironment surrounding PDAC cells. Secreted protein acid and rich in cysteine (SPARC), which is overexpressed in PDAC, may display tumor suppressor functions in several cancers (e.g., in colorectal, ovarian, prostate cancers, and acute myelogenous leukemia) but also appears to be overexpressed in other tumor types (e.g., breast cancer, melanoma, and glioblastoma). The apparent contradictory functions of SPARC may yield inhibition of angiogenesis via inhibition of vascular endothelial growth factor, while promoting epithelial-to-mesenchymal transition and invasion through matrix metalloprotease expression. This feature is of particular interest in PDAC where SPARC overexpression in the stroma stands along with inhibition of angiogenesis and promotion of cancer cell invasion and metastasis. Several therapeutic strategies to deplete stromal tissue have been developed. In this review, we focused on key preclinical and clinical data describing the role of SPARC in PDAC biology, the properties, and mechanisms of delivery of drugs that interact with SPARC and discuss the proof-of-concept clinical trials using nab-paclitaxel.


Asunto(s)
Adenocarcinoma/metabolismo , Osteonectina/metabolismo , Neoplasias Pancreáticas/metabolismo , Células del Estroma/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Albúminas/farmacología , Albúminas/uso terapéutico , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Expresión Génica , Humanos , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/mortalidad , Pronóstico
13.
Int Endod J ; 46(2): 160-8, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22889382

RESUMEN

AIM: To investigate the expression of two endoplasmic reticulum (ER)-resident key chaperone proteins, ERdj5 and BiP, under the influence of resinous monomers and its relationship with the inhibition of mineralization caused by the monomer 2-hydroxyethyl methacrylate (HEMA). METHODOLOGY: The ERdj5 and BiP expression was studied in vitro, in primary human pulp cell cultures after treatment with three different HEMA concentrations at different time periods. Subsequently, the expression of both the odontoblast markers dentine sialoprotein (DSP) and osteonectin (OSN) was studied in human pulp cells under the same conditions. RESULTS: The ERdj5 and BiP expression was upregulated in the pulp cells. DSP and OSN were largely dispersed in the cytoplasm in control cell cultures but accumulated in a perinuclear area after exposure to HEMA. Their expression levels were not affected. CONCLUSIONS: The increased expression of ERdj5 and BiP may reflect activation of ER stress. DSP and OSN accumulation into the cells may lead to their secretion arrest and inhibition of dentine matrix formation. These events may elucidate the mechanism by which HEMA inhibits the mineralization process.


Asunto(s)
Pulpa Dental/efectos de los fármacos , Estrés del Retículo Endoplásmico , Metacrilatos/efectos adversos , Chaperonas Moleculares/metabolismo , Odontoblastos/efectos de los fármacos , Estrés Fisiológico , Calcificación de Dientes/efectos de los fármacos , Adolescente , Células Cultivadas , Pulpa Dental/citología , Análisis del Estrés Dental , Chaperón BiP del Retículo Endoplásmico , Matriz Extracelular/efectos de los fármacos , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Odontoblastos/metabolismo , Osteonectina/antagonistas & inhibidores , Osteonectina/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Sialoglicoproteínas/antagonistas & inhibidores , Sialoglicoproteínas/metabolismo
14.
J Dent Res ; 91(3): 268-74, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22180568

RESUMEN

Bisphosphonates are therapeutic agents in the treatment of post-menopausal osteoporosis. Although they have been associated with delayed healing in injured tissues, inappropriate femoral fractures, and osteonecrosis of the jaw (ONJ), the pathophysiological mechanisms involved are not clear. Our hypothesis is that alendronate, a member of the N-containing bisphosphonates, indirectly inhibits osteoblast function through the coupling of osteoclasts to osteoblasts by ephrinB-EphB interaction. We found that alendronate increased gene and protein expression of ephrinB1 and EphB1, as well as B3, in femurs of adult mice injected with alendronate (10 µg/100 g/wk) for 8 weeks. Alendronate suppressed the expression of bone sialoprotein (BSP) and osteonectin in both femurs and bone marrow osteoblastic cells of mice. After elimination of pre-osteoclasts from bone marrow cells, alendronate did not affect osteoblast differentiation, indicating the need for pre-osteoclasts for alendronate's effects. Alendronate stimulated EphB1 and EphB3 protein expression in osteoblasts, whereas it enhanced ephrinB1 protein in pre-osteoclasts. In addition, a reverse signal by ephrinB1 inhibited osteoblast differentiation and suppressed BSP gene expression. Thus, alendronate, through its direct effects on the pre-osteoclast, appears to regulate expression of ephrinB1, which regulates and acts through the EphB1, B3 receptors on the osteoblast to suppress osteoblast differentiation.


Asunto(s)
Alendronato/farmacología , Conservadores de la Densidad Ósea/farmacología , Remodelación Ósea/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Efrina-B1/metabolismo , Receptores de la Familia Eph/metabolismo , Animales , Células Cultivadas , Colágeno Tipo I/antagonistas & inhibidores , Regulación de la Expresión Génica , Sialoproteína de Unión a Integrina/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteonectina/antagonistas & inhibidores
15.
PLoS One ; 6(11): e26390, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22069448

RESUMEN

SPARC, a matricellular protein with tumor suppressor properties in certain human cancers, was initially identified in a genome-wide analysis of differentially expressed genes in chemotherapy resistance. Its exciting new role as a potential chemosensitizer arises from its ability to augment the apoptotic cascade, although the exact mechanisms are unclear. This study further examines the mechanism by which SPARC may be promoting apoptosis and identifies a smaller peptide analogue with greater chemosensitizing and tumor-regressing properties than the native protein. We examined the possibility that the apoptosis-enhancing activity of SPARC could reside within one of its three biological domains (N-terminus (NT), the follistatin-like (FS), or extracellular (EC) domains), and identified the N-terminus as the region with its chemosensitizing properties. These results were not only confirmed by studies utilizing stable cell lines overexpressing the different domains of SPARC, but as well, with a synthetic 51-aa peptide spanning the NT-domain. It revealed that the NT-domain induced a significantly greater reduction in cell viability than SPARC, and that it enhanced the apoptotic cascade via its activation of caspase 8. Moreover, in chemotherapy resistant human colon, breast and pancreatic cancer cells, its chemosensitizing properties also depended on its ability to dissociate Bcl2 from caspase 8. These observations translated to clinically significant findings in that, in-vivo, mouse tumor xenografts overexpressing the NT-domain of SPARC had significantly greater sensitivity to chemotherapy and tumor regression, even when compared to the highly-sensitive SPARC-overexpressing tumors. Our results identified an interplay between the NT-domain, Bcl2 and caspase 8 that helps augment apoptosis and as a consequence, a tumor's response to therapy. This NT-domain of SPARC and its 51-aa peptide are highly efficacious in modulating and enhancing apoptosis, thereby conferring greater chemosensitivity to resistant tumors. Our findings provide additional insight into mechanisms involved in chemotherapy resistance and a potential novel therapeutic that specifically targets this devastating phenomenon.


Asunto(s)
Neoplasias de la Mama/prevención & control , Caspasa 8/metabolismo , Neoplasias Colorrectales/prevención & control , Resistencia a Antineoplásicos , Osteonectina/metabolismo , Neoplasias Pancreáticas/prevención & control , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Caspasa 8/genética , Inhibidores de Caspasas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Fluorouracilo/uso terapéutico , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Ratones , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas
16.
Cancer Res ; 71(14): 4748-57, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21616936

RESUMEN

Tumor progression is associated with the release of signaling substances from the primary tumor into the bloodstream. Tumor-derived cytokines are known to promote the mobilization and the recruitment of cells from the bone marrow, including endothelial progenitor cells (EPC). Here, we examined whether such paracrine influence could also influence the capacity of EPC to interfere with circulating metastatic cells. We therefore consecutively injected EPC prestimulated by tumor-conditioned medium (EPC-CM) and luciferase-expressing B16 melanoma cells to mice. A net decrease in metastases spreading (vs. nonstimulated EPC) led us to carry out a 2-dimensional difference gel electrophoresis (2D-DIGE) proteomic study to identify possible mediators of EPC-driven protection. Among 33 proteins exhibiting significant changes in expression, secreted protein, acidic and rich in cysteine (SPARC) presented the highest induction after EPC exposure to CM. We then showed that contrary to control EPC, SPARC-silenced EPC were not able to reduce the extent of metastases when injected with B16 melanoma cells. Using adhesion tests and the hanging drop assay, we further documented that cell-cell interactions between EPC-CM and melanoma cells were promoted in a SPARC-dependent manner. This interaction led to the engulfment of melanoma cells by EPC-CM, a process prevented by SPARC silencing and mimicked by recombinant SPARC. Finally, we showed that contrary to melanoma cells, the prometastatic human breast cancer cell line MDA-MB231-D3H2 reduced SPARC expression in human EPC and stimulated metastases spreading. Our findings unravel the influence of tumor cells on EPC phenotypes through a SPARC-driven accentuation of macrophagic capacity associated with limitations to metastatic spread.


Asunto(s)
Comunicación Celular/fisiología , Células Endoteliales/trasplante , Melanoma Experimental/secundario , Melanoma Experimental/terapia , Osteonectina/antagonistas & inhibidores , Trasplante de Células Madre/métodos , Células Madre/citología , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Medios de Cultivo Condicionados , Endocitosis/fisiología , Células Endoteliales/citología , Femenino , Humanos , Masculino , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Osteonectina/biosíntesis
18.
J Exp Clin Cancer Res ; 29: 59, 2010 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-20525171

RESUMEN

BACKGROUND: Secreted protein acidic and rich in cysteine (SPARC) plays a key role in the development of many tissues and organ types. Aberrant SPARC expression was found in a wide variety of human cancers, contributes to tumor development. Because SPARC was found to be overexpressed in human gastric cancer tissue, we therefore to explore the expression of SPARC in gastric cancer lines and the carcinogenic mechanisms. METHODS: SPARC expression was evaluated in a panel of human gastric cancer cell lines. MGC803 and HGC 27 gastric cancer cell lines expressing high level of SPARC were transiently transfected with SPARC-specific small interfering RNAs and subsequently evaluated for effects on invasion and proliferation. RESULTS: Small interfering RNA-mediated knockdown of SPARC in MGC803 and HGC 27 gastric cancer cells dramatically decreased their invasion. Knockdown of SPARC was also observed to significantly increase the apoptosis of MGC803 and HGC 27 gastric cancer cells compared with control transfected group. CONCLUSIONS: Our data showed that downregulating of SPARC inhibits invasion and growth of human gastric cancer cells. Thus, targeting of SPARC could be an effective therapeutic approach against gastric cancer.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Osteonectina/metabolismo , ARN Interferente Pequeño/farmacología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Apoptosis , Western Blotting , Adhesión Celular , Ciclo Celular , Regulación hacia Abajo , Humanos , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Células Tumorales Cultivadas
19.
Arthritis Res Ther ; 12(2): R60, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20359365

RESUMEN

INTRODUCTION: SPARC is a matricellular protein, which, along with other extracellular matrix components including collagens, is commonly over-expressed in fibrotic diseases. The purpose of this study was to examine whether inhibition of SPARC can regulate collagen expression in vitro and in vivo, and subsequently attenuate fibrotic stimulation by bleomycin in mouse skin and lungs. METHODS: In in vitro studies, skin fibroblasts obtained from a Tgfbr1 knock-in mouse (TBR1CA; Cre-ER) were transfected with SPARC siRNA. Gene and protein expressions of the Col1a2 and the Ctgf were examined by real-time RT-PCR and Western blotting, respectively. In in vivo studies, C57BL/6 mice were induced for skin and lung fibrosis by bleomycin and followed by SPARC siRNA treatment through subcutaneous injection and intratracheal instillation, respectively. The pathological changes of skin and lungs were assessed by hematoxylin and eosin and Masson's trichrome stains. The expression changes of collagen in the tissues were assessed by real-time RT-PCR and non-crosslinked fibrillar collagen content assays. RESULTS: SPARC siRNA significantly reduced gene and protein expression of collagen type 1 in fibroblasts obtained from the TBR1CA; Cre-ER mouse that was induced for constitutively active TGF-beta receptor I. Skin and lung fibrosis induced by bleomycin was markedly reduced by treatment with SPARC siRNA. The anti-fibrotic effect of SPARC siRNA in vivo was accompanied by an inhibition of Ctgf expression in these same tissues. CONCLUSIONS: Specific inhibition of SPARC effectively reduced fibrotic changes in vitro and in vivo. SPARC inhibition may represent a potential therapeutic approach to fibrotic diseases.


Asunto(s)
Fibroblastos/patología , Pulmón/patología , Osteonectina/genética , Fibrosis Pulmonar/patología , ARN Interferente Pequeño/genética , Piel/patología , Animales , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Expresión Génica , Técnicas de Sustitución del Gen , Silenciador del Gen , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteonectina/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , ARN Interferente Pequeño/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Piel/efectos de los fármacos , Piel/metabolismo , Transfección
20.
Cancer Biol Ther ; 9(7): 507-13, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20215880

RESUMEN

Krüppel-Like Factor 4 (KLF4) functions as a tumor suppressor in some cancers, but its molecular mechanism is not clear. Our recent study also showed that the expression of KLF4 is dramatically reduced in primary lung cancer tissues. To investigate the possible role of KLF4 in lung cancer, we stably transfected KLF4 into cells from lung cancer cell lines H322 and A549 to determine the cells' invasion ability. Our results showed that ectopic expression of KLF4 extensively suppressed lung cancer cell invasion in Matrigel. This effect was independent of KLF4-mediated p21 up-regulation because ectopic expression of p21 had minimal effect on cell invasion. Our analysis of the expression of 12 genes associated with cell invasion in parental, vector-transfected, and KLF4-transfected cells showed that ectopic expression of KLF4 resulted in extensively repressed expression of secreted protein acidic and rich in cysteine (SPARC), an extracellular matrix protein that plays a role in tumor development and metastasis. Knockdown of SPARC expression in H322 and A549 cells led to suppression of cell invasion, comparable to that observed in KLF4-transfected cells. Moreover, retrovirus-mediated restoration of SPARC expression in KLF4-transfected cells abrogated KLF4-induced anti-invasion activity. Together, our results indicate that KLF4 inhibits lung cancer cell invasion by suppressing SPARC gene expression.


Asunto(s)
Movimiento Celular , Regulación Neoplásica de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Osteonectina/metabolismo , Transfección , Western Blotting , Adhesión Celular , Proliferación Celular , Colágeno/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Combinación de Medicamentos , Humanos , Factor 4 Similar a Kruppel , Laminina/metabolismo , Invasividad Neoplásica , Osteonectina/antagonistas & inhibidores , Osteonectina/genética , Proteoglicanos/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...