Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.570
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 699: 149554, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38280308

RESUMEN

In the tumor environment, hypoxia promotes tumor progression, such as cancer cell growth, migration and chemoresistance. This study aimed to evaluate the roles of free fatty acid receptors (FFARs) in the regulation of cancer cell functions under hypoxic conditions, using fibrosarcoma HT1080 cells. HT1080 cells expressed FFAR1, FFAR2 and FFAR3 genes, but not FFAR4 gene. FFAR1, FFAR2 and FFAR3 expression levels in HT1080 cells cultured at 1 % O2 were elevated, compared with 21 % O2. The cell growth activities of HT1080 cells cultured at 21 % O2 were inhibited by acetic acid (AA) and propanoic acid (PA), but not 1 % O2. HT1080 cell motility was markedly reduced by culturing at 1 % O2. The cell growth and motility of HT1080 cells were enhanced by FFAR2 knockdown. The cell viability to cisplatin (CDDP) of HT1080 cells cultured at 1 % O2 was increased, compared with 21 % O2. FFAR2 knockdown suppressed the cell viability to CDDP of HT1080 cells. On the other hand, the cell motility and viability to CDDP of HT1080 cells cultured at 21 % O2 were suppressed by TUG-770. When HT1080 cells were cultured at 1 % O2, the cell motility and viability to CDDP were decreased, correlating with FFAR1 expression level. Moreover, FFAR1 knockdown increased the cell viability to CDDP of HT1080 cells cultured at 1 % O2. These results suggest that FFAR-mediated signaling plays an important role in the modulation of cellular functions of HT1080 cells under hypoxic conditions.


Asunto(s)
Ácidos Grasos no Esterificados , Fibrosarcoma , Humanos , Receptores Acoplados a Proteínas G/metabolismo , Cisplatino/farmacología , Transducción de Señal , Fibrosarcoma/metabolismo , Movimiento Celular
2.
Int J Med Sci ; 21(1): 123-136, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164349

RESUMEN

CREB3 subfamily belongs to the bZIP transcription factor family and comprises five members. Normally they are located on the endoplasmic reticulum (ER) membranes and proteolytically activated through RIP (regulated intramembrane proteolysis) on Golgi apparatus to liberate the N-terminus to serve as transcription factors. CREB3L1 acting as one of them transcriptionally regulates the expressions of target genes and exhibits distinct functions from the other members of CREB3 family in eukaryotes. Physiologically, CREB3L1 involves in the regulation of bone morphogenesis, neurogenesis, neuroendocrine, secretory cell differentiation, and angiogenesis. Pathologically, CREB3L1 implicates in the modulation of osteogenesis imperfecta, low grade fibro myxoid sarcoma (LGFMS), sclerosing epithelioid fibrosarcoma (SEF), glioma, breast cancer, thyroid cancer, and tissue fibrosis. This review summarizes the upstream and downstream regulatory network of CREB3L1 and thoroughly presents our current understanding of CREB3L1 research progress in both physiological and pathological conditions with special focus on the novel findings of CREB3L1 in cancers.


Asunto(s)
Fibrosarcoma , Humanos , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Biomarcadores de Tumor/genética , Proteínas del Tejido Nervioso , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética
3.
Int J Mol Sci ; 25(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38892363

RESUMEN

Autophagy plays a key role in removing protein aggregates and damaged organelles. In addition to its conventional degradative functions, autophagy machinery contributes to the release of cytosolic proteins through an unconventional secretion pathway. In this research, we analyzed autophagy-induced extracellular vesicles (EVs) in HT1080-derived human fibrosarcoma 2FTGH cells using transmission electron microscopy and atomic force microscopy (AFM). We preliminary observed that autophagy induces the formation of a subset of large heterogeneous intracellular vesicular structures. Moreover, AFM showed that autophagy triggering led to a more visible smooth cell surface with a reduced amount of plasma membrane protrusions. Next, we characterized EVs secreted by cells following autophagy induction, demonstrating that cells release both plasma membrane-derived microvesicles and exosomes. A self-forming iodixanol gradient was performed for cell subfractionation. Western blot analysis showed that endogenous LC3-II co-fractionated with CD63 and CD81. Then, we analyzed whether raft components are enriched within EV cargoes following autophagy triggering. We observed that the raft marker GD3 and ER marker ERLIN1 co-fractionated with LC3-II; dual staining by immunogold electron microscopy and coimmunoprecipitation revealed GD3-LC3-II association, indicating that autophagy promotes enrichment of raft components within EVs. Introducing a new brick in the crosstalk between autophagy and the endolysosomal system may have important implications for the knowledge of pathogenic mechanisms, suggesting alternative raft target therapies in diseases in which the generation of EV is active.


Asunto(s)
Autofagia , Vesículas Extracelulares , Humanos , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Línea Celular Tumoral , Microdominios de Membrana/metabolismo , Exosomas/metabolismo , Exosomas/ultraestructura , Tetraspanina 30/metabolismo , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Proteínas Asociadas a Microtúbulos/metabolismo
4.
Int J Mol Sci ; 25(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38791144

RESUMEN

Cellular myxoma is a benign soft tissue tumor frequently associated with GNAS mutation that may morphologically resemble low-grade myxofibrosarcoma. This study aimed to identify the undescribed methylation profile of cellular myxoma and compare it to myxofibrosarcoma. We performed molecular analysis on twenty cellular myxomas and nine myxofibrosarcomas and analyzed the results using the methylation-based DKFZ sarcoma classifier. A total of 90% of the cellular myxomas had GNAS mutations (four loci had not been previously described). Copy number variations were found in all myxofibrosarcomas but in none of the cellular myxomas. In the classifier, none of the cellular myxomas reached the 0.9 threshold. Unsupervised t-SNE analysis demonstrated that cellular myxomas form their own clusters, distinct from myxofibrosarcomas. Our study shows the diagnostic potential and the limitations of molecular analysis in cases where morphology and immunohistochemistry are not sufficient to distinguish cellular myxoma from myxofibrosarcoma, particularly regarding GNAS wild-type tumors. The DKFZ sarcoma classifier only provided a valid prediction for one myxofibrosarcoma case; this limitation could be improved by training the tool with a more considerable number of cases. Additionally, the classifier should be introduced to a broader spectrum of mesenchymal neoplasms, including benign tumors like cellular myxoma, whose distinct methylation pattern we demonstrated.


Asunto(s)
Variaciones en el Número de Copia de ADN , Metilación de ADN , Fibrosarcoma , Mixoma , Humanos , Mixoma/genética , Mixoma/diagnóstico , Mixoma/patología , Fibrosarcoma/genética , Fibrosarcoma/patología , Fibrosarcoma/diagnóstico , Fibrosarcoma/metabolismo , Persona de Mediana Edad , Femenino , Anciano , Masculino , Adulto , Mutación , Diagnóstico Diferencial , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Cromograninas/genética , Anciano de 80 o más Años , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/diagnóstico , Neoplasias de los Tejidos Blandos/patología
5.
Int J Mol Sci ; 25(11)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38892288

RESUMEN

This study demonstrated the anticancer efficacy of chalcones with indole moiety (MIPP, MOMIPP) in fibrosarcoma cells for the first time. The results showed that MIPP and MOMIPP reduced the viability of HT-1080 cells in a concentration-dependent manner. MOMIPP was more active than MIPP in HT-1080 cells, showing lower IC50 values (3.67 vs. 29.90 µM). Both compounds at a concentration of 1 µM induced apoptosis in HT-1080 cells, causing death strictly related to caspase activation, as cell viability was restored when the caspase inhibitor Z-VAD was added. Reactive oxygen species production was approximately 3-fold higher than in control cells, and cotreatment with the inhibitor of mitochondrial ATPase oligomycin diminished this effect. Such effects were also reflected in mitochondrial dysfunction, including decreased membrane potential. Interestingly, the compounds that were studied caused massive vacuolization in HT-1080 cells. Immunocytochemical staining and TEM analysis showed that HT-1080 cells exhibited increased expression of the LC3-II protein and the presence of autophagosomes with a double membrane, respectively. Both compounds induced apoptosis, highlighting a promising link between autophagy and apoptosis. This connection could be a new target for therapeutic strategies to overcome chemoresistance, which is a significant cause of treatment failure and tumour recurrence in fibrosarcoma following traditional chemotherapy.


Asunto(s)
Apoptosis , Autofagia , Chalconas , Fibrosarcoma , Indoles , Especies Reactivas de Oxígeno , Humanos , Apoptosis/efectos de los fármacos , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Autofagia/efectos de los fármacos , Indoles/farmacología , Línea Celular Tumoral , Especies Reactivas de Oxígeno/metabolismo , Chalconas/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Antineoplásicos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
6.
Mol Biol Rep ; 50(11): 8937-8947, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37710072

RESUMEN

Increased hyaluronan deposition (HA) in various cancer tissues, including sarcomas, correlates with disease progression. The receptor for hyaluronic acid-mediated motility (RHAMM) expression is elevated in most human cancers. ß-catenin is a critical downstream mediator of the Wnt signaling pathways, facilitating carcinogenic events characterized by deregulated cell proliferation. We previously showed that low molecular weight (LMW) HA/RHAMM/ß-catenin signaling axis increases HT1080 fibrosarcoma cell growth. Here, focusing on mechanistic aspects and utilizing immunofluorescence and immunoprecipitation, we demonstrate that LMW HA treatment enhanced RHAMM intracellular localization (p ≤ 0.001) and RHAMM/ß-catenin colocalization in HT1080 fibrosarcoma cells (p ≤ 0.05). Downregulating endogenous HA attenuated the association of RHAMM/ß-catenin in HT1080 fibrosarcoma cells (p ≤ 0.0.01). Notably, Axin-2, the key ß-catenin degradation complex component, and RHAMM were demonstrated to form a complex primarily to cell membranes, enhanced by LMW HA (p ≤ 0.01). In contrast, LMW HA attenuated the association of ß-catenin and Axin-2 (p ≤ 0.05). The utilization of FH535, a Wnt signaling inhibitor, showed that LMW HA partially rescued the Wnt-dependent growth of HT1080 cells and restored the expression of Wnt/ß-catenin mediators, cyclin-D1 and c-myc (p ≤ 0.05). B6FS fibrosarcoma cells with different HA metabolism do not respond to the LMW HA growth stimulus (p = NS). The present study identifies a novel LMW HA/RHAMM mechanism in a fibrosarcoma model. LMW HA regulates intracellular RHAMM expression, which acts as a scaffold protein binding ß-catenin and Axin-2 at different cellular compartments to increase ß-catenin expression, transcriptional activity, and fibrosarcoma growth.


Asunto(s)
Fibrosarcoma , Ácido Hialurónico , Humanos , Ácido Hialurónico/farmacología , Proteína Axina/genética , Proteína Axina/metabolismo , beta Catenina/metabolismo , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Proliferación Celular , Fibrosarcoma/metabolismo , Movimiento Celular , Proteínas Portadoras , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo
7.
Diabetologia ; 65(5): 811-828, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35243521

RESUMEN

AIMS/HYPOTHESIS: While pancreatic beta cells have been shown to originate from endocrine progenitors in ductal regions, it remains unclear precisely where beta cells emerge from and which transcripts define newborn beta cells. We therefore investigated characteristics of newborn beta cells extracted by a time-resolved reporter system. METHODS: We established a mouse model, 'Ins1-GFP; Timer', which provides spatial information during beta cell neogenesis with high temporal resolution. Single-cell RNA-sequencing (scRNA-seq) was performed on mouse beta cells sorted by fluorescent reporter to uncover transcriptomic profiles of newborn beta cells. scRNA-seq of human embryonic stem cell (hESC)-derived beta-like cells was also performed to compare newborn beta cell features between mouse and human. RESULTS: Fluorescence imaging of Ins1-GFP; Timer mouse pancreas successfully dissected newly generated beta cells as green fluorescence-dominant cells. This reporter system revealed that, as expected, some newborn beta cells arise close to the ducts (ßduct); unexpectedly, the others arise away from the ducts and adjacent to blood vessels (ßvessel). Single-cell transcriptomic analyses demonstrated five distinct populations among newborn beta cells, confirming spatial heterogeneity of beta cell neogenesis such as high probability of glucagon-positive ßduct, musculoaponeurotic fibrosarcoma oncogene family B (MafB)-positive ßduct and musculoaponeurotic fibrosarcoma oncogene family A (MafA)-positive ßvessel cells. Comparative analysis with scRNA-seq data of mouse newborn beta cells and hESC-derived beta-like cells uncovered transcriptional similarity between mouse and human beta cell neogenesis including microsomal glutathione S-transferase 1 (MGST1)- and synaptotagmin 13 (SYT13)-highly-expressing state. CONCLUSIONS/INTERPRETATION: The combination of time-resolved histological imaging with single-cell transcriptional mapping demonstrated novel features of spatial and transcriptional heterogeneity in beta cell neogenesis, which will lead to a better understanding of beta cell differentiation for future cell therapy. DATA AVAILABILITY: Raw and processed single-cell RNA-sequencing data for this study has been deposited in the Gene Expression Omnibus under accession number GSE155742.


Asunto(s)
Fibrosarcoma , Células Secretoras de Insulina , Transcriptoma , Animales , Diferenciación Celular/genética , Fibrosarcoma/metabolismo , Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/metabolismo , Ratones , Conductos Pancreáticos , ARN
8.
Can J Physiol Pharmacol ; 100(2): 184-191, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34597523

RESUMEN

Apoptosis-inducing factor (AIF) is a mitochondrial flavoprotein which mediates staurosporine (STS) - induced cell death. AIF cleavage and translocation to the cytosol is thought to be calpain-1-dependent as calpain inhibitors reduce AIF proteolysis; however, many calpain inhibitors also inhibit matrix metalloproteinase-2 (MMP-2) activity, an intracellular and extracellular protease implicated in apoptosis. Here we investigated whether MMP-2 activity is affected in response to STS and if it contributes to AIF cleavage. Human fibrosarcoma HT1080 cells were treated with STS (0.1 µM, 0.25-24 h). A significant increase in cellular MMP-2 activity was seen by gelatin zymography after a 6 h STS treatment, prior to induction of cell necrosis. Western blot showed the time-dependent appearance of two forms of AIF (∼60 and 45 kDa) in the cytosol which were significantly increased at 6 h. Surprisingly, knocking down MMP-2 or inhibiting its activity with MMP-2 preferring inhibitors ARP-100 or ONO-4817, or inhibiting calpain activity with ALLM or PD150606, did not prevent the STS-induced increase in cytosolic AIF. These results show that although STS rapidly increases MMP-2 activity, the cytosolic release of AIF may be independent of the proteolytic activities of MMP-2 or calpain.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Estaurosporina/farmacología , Calpaína/metabolismo , Citosol/metabolismo , Humanos , Proteolisis , Células Tumorales Cultivadas
9.
Int J Mol Sci ; 23(5)2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35269801

RESUMEN

A phenyl ethanoid, salidroside (SAL), and two secoiridoids, 8(E)-nuezhenide (NZD) and ligustroside (LIG), were isolated from fruits of Ligustrumjaponicum, used as traditional folk medicine, and their chemical structures were elucidated by the comparison of spectral data with published literature. Matrix metalloproteinases (MMPs) are major enzymes that play crucial roles in the metastasis and invasive behavior of tumors. In particular, MMP-2 and MMP-9, regulated by the MAPK signaling pathways, including p38, ERK and JNK, are known to play a key role in the degradation of the basement membrane. In the present study, the effects of SAL, NZD and LIG on the expression of MMP-2 and -9 were examined in phorbol 12-myristate 13-acetate (PMA)-induced HT 1080 cells. All the compounds significantly lowered the amount of MMP-2 and MMP-9 released, as determined by gelatin zymography and ELISA. In addition, the mRNA and protein expression levels of MMP-2 and MMP-9 were significantly suppressed, as measured by RT-PCR and Western blotting. According to the Western blotting assay, SAL and LIG effectively reduced the expression of MMP-2 in a dose-dependent manner. NZD lowered the expression of MMP-9 in a similar way. The phosphorylation of p38, ERK and JNK was also significantly suppressed by these compounds. These findings suggest that all the compounds regulate the release and expression of MMP-2 and MMP-9 via MAPK signaling pathways.


Asunto(s)
Fibrosarcoma , Ligustrum , Fibrosarcoma/metabolismo , Frutas/metabolismo , Glucósidos , Humanos , Ligustrum/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Fenoles , Piranos , Acetato de Tetradecanoilforbol/farmacología
10.
Angiogenesis ; 24(3): 533-548, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33496909

RESUMEN

Myxofibrosarcoma is genetically complex and lacks effective nonsurgical treatment strategies; thus, elucidation of novel molecular drivers is urgently needed. Reanalyzing public myxofibrosarcoma datasets, we identified mRNA upregulation and recurrent gain of RSF1 and characterized this chromatin remodeling gene. Myxofibrosarcoma cell lines were employed to elucidate the oncogenic mechanisms of RSF1 by genetic manipulation and two IL-1ß-neutralizing antibodies (RD24, P2D7KK), highlighting the regulatory basis and targetability of downstream IL-1ß-mediated angiogenesis. Tumor samples were assessed for RSF1, IL-1ß, and microvascular density (MVD) by immunohistochemistry and for RSF1 gene status by FISH. In vivo, RSF1-silenced and P2D7KK-treated xenografts were analyzed for tumor-promoting effects and the IL-1ß-linked therapeutic relevance of RSF1, respectively. In vitro, RSF1 overexpression promoted invasive and angiogenic phenotypes with a stronger proangiogenic effect. RT-PCR profiling identified IL1B as a top-ranking candidate upregulated by RSF1. RSF1 required hSNF2H and CEBP/ß to cotransactivate the IL1B promoter, which increased the IL1B mRNA level, IL-1ß secretion and angiogenic capacity. Angiogenesis induced by RSF1-upregulated IL-1ß was counteracted by IL1B knockdown and both IL-1ß-neutralizing antibodies. Clinically, RSF1 overexpression was highly associated with RSF1 amplification, IL-1ß overexpression, increased MVD and higher grades (all P ≤ 0.01) and independently predicted shorter disease-specific survival (P = 0.019, hazard ratio: 4.556). In vivo, both RSF1 knockdown and anti-IL-1ß P2D7KK (200 µg twice weekly) enabled significant growth inhibition and devascularization in xenografts. In conclusion, RSF1 overexpression, partly attributable to RSF1 amplification, contributes a novel proangiogenic function by partnering with CEBP/ß to cotransactivate IL1B, highlighting its prognostic, pathogenetic, and therapeutic relevance in myxofibrosarcomas.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Fibrosarcoma/metabolismo , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Interleucina-1beta/metabolismo , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Nucleares/biosíntesis , Transactivadores/biosíntesis , Adenosina Trifosfatasas/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Cromosómicas no Histona/genética , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/genética , Fibrosarcoma/patología , Humanos , Interleucina-1beta/genética , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Proteínas Nucleares/genética , Transactivadores/genética
11.
J Cell Sci ; 132(11)2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31076510

RESUMEN

Interactions between different cell types can induce distinct contact inhibition of locomotion (CIL) responses that are hypothesised to control population-wide behaviours during embryogenesis. However, our understanding of the signals that lead to cell-type specific repulsion and the precise capacity of heterotypic CIL responses to drive emergent behaviours is lacking. Using a new model of heterotypic CIL, we show that fibrosarcoma cells, but not fibroblasts, are actively repelled by epithelial cells in culture. We show that knocking down EphB2 or ERK in fibrosarcoma cells specifically leads to disruption of the repulsion phase of CIL in response to interactions with epithelial cells. We also examine the population-wide effects when these various cell combinations are allowed to interact in culture. Unlike fibroblasts, fibrosarcoma cells completely segregate from epithelial cells and inhibiting their distinct CIL response by knocking down EphB2 or ERK family proteins also disrupts this emergent sorting behaviour. These data suggest that heterotypic CIL responses, in conjunction with processes such as differential adhesion, may aid the sorting of cell populations.


Asunto(s)
Comunicación Celular/fisiología , Inhibición de Contacto/fisiología , Células Epiteliales/fisiología , Fibroblastos/fisiología , Células Madre Mesenquimatosas/fisiología , Células 3T3 , Animales , Línea Celular , Movimiento Celular/fisiología , Separación Celular , Desarrollo Embrionario/fisiología , Quinasas MAP Reguladas por Señal Extracelular/genética , Fibrosarcoma/metabolismo , Humanos , Ratones , Receptor EphB2/genética
12.
Cancer Immunol Immunother ; 70(1): 153-168, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32661685

RESUMEN

The contribution of immune cells in soft tissue sarcomas (STS) is not completely known and understanding their role is very essential for employing immunotherapy strategies. Here, we show that murine fibrosarcoma-conditioned medium promoted total spleen cell proliferation but inhibited T cell responses to mitogenic and allo-antigen-mediated stimulation. This increased proliferation was found to be in B cells resulting in generation of Breg further leading to Treg population. This was found to be the same in vitro and in vivo. The phenotype of these B cells was CD19+CD81+CD27+CD25+PD-L1hi and they secreted both IL-10 and TGF-ß. These tumor evoked Bregs (tBreg), when co-cultured with B depleted T cells, suppressed their proliferation in response to anti-CD3/CD28 stimulation. tBreg-induced suppression of T cell responses was not abrogated by the inhibition or neutralization of IL-10 but by the small molecule inhibitor of TGFß Receptor type I, SB431542. While SB531542 per se was not cytotoxic to tumor cells, administration of SB431542 in tumor-bearing mice (TBM) significantly reduced the tumor burden. In addition, the treatment significantly reduced Treg cells and rescued proliferation of T cells in response to mitogen and allo-antigen. Collectively, our results identify that tumor evoked Breg cells mediate T cell immune suppression through TGFß-mediated pathway and that targeting the Breg-Treg axis can be potentially used as an immunotherapy agent.


Asunto(s)
Linfocitos B Reguladores/efectos de los fármacos , Benzamidas/farmacología , Dioxoles/farmacología , Fibrosarcoma/tratamiento farmacológico , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Linfocitos T Reguladores/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Animales , Antígenos CD/metabolismo , Linfocitos B Reguladores/inmunología , Proliferación Celular/efectos de los fármacos , Femenino , Fibrosarcoma/inmunología , Fibrosarcoma/metabolismo , Factores de Transcripción Forkhead/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Carga Tumoral/inmunología
13.
Mol Pharm ; 18(7): 2574-2585, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34048242

RESUMEN

The biodistribution of molecular imaging probes or tracers mainly depends on the chemical nature of the probe and the preferred metabolization and excretion routes. Small molecules have rather short half-lives while antibodies reside inside the organism for a longer period of time. An excretion via kidneys and bladder is faster than a mainly hepatobiliary elimination. To manipulate the biodistribution behavior of probes, different strategies have been pursued, including utilizing serum albumin as an inherent transport mechanism for small molecules. Here, we modified an existing small molecular fluorescent probe targeted to the endothelin-A receptor (ETAR) with three different albumin-binding moieties to search for an optimal modification strategy. A diphenylcyclohexyl (DPCH) group, a p-iodophenyl butyric acid (IPBA), and a fatty acid (FA) group were attached via amino acid linkers. All three modifications result in transient albumin binding of the developed compounds, as concluded from gel electrophoresis investigations. Spectrophotometric measurements applying variable amounts of bovine, murine, and human serum albumin (BSA, MSA, and HSA) reveal distinct variations of absorption and emission intensities and shifts of their maximum wavelengths. Binding to MSA results in the weakest effects, while binding to HSA leads to the strongest. Cell-based in vitro investigations utilizing ETAR-positive HT-1080 fibrosarcoma and ETAR-negative BT-20 breast adenocarcinoma cells support a retained specific target-binding capacity of the modified compounds and different degrees of unspecific binding. In vivo analysis of a HT-1080 xenograft model in nude mice over the course of 1 week by fluorescence reflectance imaging illustrates noticeable differences between the four examined probes. While the IPBA-modified probe shows the highest absolute signal intensity values, the FA-modified probe exhibits the most favorable tumor-to-organ ratios. In summary, reversible binding to albumin enhances the biological half-life of the designed probes substantially and enables near infrared optical imaging of subcutaneous tumors for several days in vivo. Because the unmodified probe already exhibits reasonable results, the attachment of albumin-binding moieties does not lead to a substantially improved imaging outcome in terms of target-to-background ratios. On the other hand, because the implemented transient albumin binding results in an overall higher amount of probe inside tumor lesions, this strategy might be adaptable for theranostic or therapeutic approaches in a future clinical routine.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibrosarcoma/metabolismo , Colorantes Fluorescentes/metabolismo , Imagen Molecular/métodos , Sondas Moleculares/metabolismo , Receptor de Endotelina A/química , Albúmina Sérica/metabolismo , Animales , Neoplasias de la Mama/patología , Femenino , Fibrosarcoma/patología , Colorantes Fluorescentes/química , Humanos , Ratones , Ratones Desnudos , Sondas Moleculares/química , Imagen Óptica , Albúmina Sérica/química , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Cutan Pathol ; 48(2): 285-289, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32519331

RESUMEN

Dermatofibrosarcoma protuberans (DFSP) is a rare sarcoma of the skin arising from the dermis. Its location is most commonly presented on the trunk of middle-aged adults and rarely on the face. The characteristic genetic aberration in the form of a reciprocal translocation t(17;22)(q21;q13) or a ring fusing the COL1A1 and PDGFB genes is found in 90% of DFSP. We present a case of a 42-year-old man who presented with a DFSP on the left cheek with foci of myxoid-fibrosarcomatous transformation. A conventional chromosomal analysis revealed a complex karyotype without a supernumerary ring chromosome or a linear translocation t(17;22). Comparative genome hybridization and fluorescence in-situ hybridization revealed the fusion of COL1A1 and PDGFB probes inserted in chromosome 15. This is a unique case of DFSP characterized by a rare body location, unique histopathological features, and novel chromosome COL1A1-PDGFB insertion, and may help guide future diagnostic and patient care modalities.


Asunto(s)
Cromosomas Humanos Par 15 , Neoplasias Faciales , Fibrosarcoma , Mutagénesis Insercional , Proteínas de Fusión Oncogénica , Neoplasias Cutáneas , Adulto , Cromosomas Humanos Par 15/genética , Cromosomas Humanos Par 15/metabolismo , Cromosomas Humanos Par 17/genética , Cromosomas Humanos Par 17/metabolismo , Cromosomas Humanos Par 22/genética , Cromosomas Humanos Par 22/metabolismo , Neoplasias Faciales/genética , Neoplasias Faciales/metabolismo , Neoplasias Faciales/patología , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Humanos , Masculino , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Translocación Genética
15.
Pediatr Dev Pathol ; 24(2): 148-153, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33470922

RESUMEN

Sclerosing epithelioid fibrosarcoma (SEF) is a rare variant of fibrosarcoma primarily arising in the deep soft tissue of the extremities and trunk. Despite having the morphologic appearance of a low-grade sarcoma, it generally has an aggressive clinical course with frequent local recurrences and distant metastases. It typically occurs in middle aged adults and is characterized by immunoexpression of MUC4 and recurrent gene fusions, most commonly EWSR1-CREB3L1. We report a primary renal SEF in a 4-year-old male. To our knowledge, this is the youngest patient reported with SEF and the second case of SEF in a pre-adolescent child. It is the eleventh reported case of primary renal SEF in the literature. While SEF arising in visceral organs is rare, the kidney is the most common primary site of any visceral organ. This case demonstrates SEF can occur in pre-adolescents, is an important consideration when evaluating sarcomas in young children, and should be considered in the differential diagnosis for primary renal tumors.


Asunto(s)
Fibrosarcoma/diagnóstico , Neoplasias Renales/diagnóstico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Preescolar , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Fusión Génica , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Mucina 4/metabolismo , Proteínas del Tejido Nervioso/genética , Proteína EWS de Unión a ARN/genética
16.
Carcinogenesis ; 41(5): 656-665, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31276576

RESUMEN

Metabolic changes accompanying a step-wise malignant transformation was investigated using a syngeneic lineage of human fibroblasts. Cell immortalization was associated with minor alterations in metabolism. Consecutive loss of cell cycle inhibition in immortalized cells resulted in increased levels of oxidative phosphorylation (OXPHOS). Overexpression of the H-Ras oncoprotein produced cells forming sarcomas in athymic mice. These transformed cells exhibited increased glucose consumption, glycolysis and a further increase in OXPHOS. Because of the markedly increased OXPHOS in transformed cells, the impact of a transaminase inhibitor, aminooxyacetic acid (AOA), which decreases glutamine influx to the tricarboxylic acid (TCA) cycle, was tested. Indeed, AOA significantly decreased proliferation of malignantly transformed fibroblasts and fibrosarcoma-derived cells in vitro and in vivo. AOA also decreased proliferation of cells susceptible to malignant transformation. Metabolomic studies in normal and transformed cells indicated that, in addition to the anticipated effect on the TCA cycle, AOA decreased production of nucleotides adenosine triphosphate (ATP) and uridine monophosphate. Exogenous nucleotides partially rescued decreased proliferation of the malignant cells treated with AOA. Our data indicate that AOA blocks several metabolic pathways essential for growth of malignant cells. Therefore, OXPHOS may provide important therapeutic targets for treatment of sarcoma.


Asunto(s)
Transformación Celular Neoplásica/patología , Fibroblastos/patología , Fibrosarcoma/patología , Regulación Neoplásica de la Expresión Génica , Genómica/métodos , Metaboloma/efectos de los fármacos , Piel/patología , Ácido Aminooxiacético/farmacología , Animales , Apoptosis , Proliferación Celular , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Glucólisis , Humanos , Ratones , Ratones Desnudos , Fosforilación Oxidativa , Piel/efectos de los fármacos , Piel/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Sci ; 111(2): 429-440, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31808966

RESUMEN

Soft tissue sarcomas (STSs) are a rare cancer type. Almost half are unresponsive to multi-pronged treatment and might therefore benefit from biologically targeted therapy. An emerging target is glycogen synthase kinase (GSK)3ß, which is implicated in various diseases including cancer. Here, we investigated the expression, activity and putative pathological role of GSK3ß in synovial sarcoma and fibrosarcoma, comprising the majority of STS that are encountered in orthopedics. Expression of the active form of GSK3ß (tyrosine 216-phosphorylated) was higher in synovial sarcoma (SYO-1, HS-SY-II, SW982) and in fibrosarcoma (HT1080) tumor cell lines than in untransformed fibroblast (NHDF) cells that are assumed to be the normal mesenchymal counterpart cells. Inhibition of GSK3ß activity by pharmacological agents (AR-A014418, SB-216763) or of its expression by RNA interference suppressed the proliferation of sarcoma cells and their invasion of collagen gel, as well as inducing their apoptosis. These effects were associated with G0/G1-phase cell cycle arrest and decreased expression of cyclin D1, cyclin-dependent kinase (CDK)4 and matrix metalloproteinase 2. Intraperitoneal injection of the GSK3ß inhibitors attenuated the growth of SYO-1 and HT1080 xenografts in athymic mice without obvious detrimental effects. It also mitigated cell proliferation and induced apoptosis in the tumors of mice. This study indicates that increased activity of GSK3ß in synovial sarcoma and fibrosarcoma sustains tumor proliferation and invasion through the cyclin D1/CDK4-mediated pathway and enhanced extracellular matrix degradation. Our results provide a biological basis for GSK3ß as a new and promising therapeutic target for these STS types.


Asunto(s)
Fibrosarcoma/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Indoles/administración & dosificación , Maleimidas/administración & dosificación , Sarcoma Sinovial/tratamiento farmacológico , Tiazoles/administración & dosificación , Urea/análogos & derivados , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Indoles/farmacología , Inyecciones Intraperitoneales , Maleimidas/farmacología , Ratones , Fosforilación/efectos de los fármacos , Interferencia de ARN , Sarcoma Sinovial/genética , Sarcoma Sinovial/metabolismo , Tiazoles/farmacología , Regulación hacia Arriba/efectos de los fármacos , Urea/administración & dosificación , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Biochem Biophys Res Commun ; 532(3): 427-432, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32883524

RESUMEN

Lysophosphatidic acid (LPA) through six subtypes of G protein-coupled LPA receptors (LPA1 to LPA6) mediates a variety of cancer cell functions. The aim of this study was to evaluate the cooperative effects of G12/13 and Gi proteins through LPA2 on cancer cell survival to cisplatin (CDDP). In cell survival assay, cells were treated with CDDP every 24 h for 2 days. The long-term CDDP treated (HT-CDDP) cells established from fibrosarcoma HT1080 cells were pretreated with an LPA2 agonist, GRI-977143. The cell survival rate to CDDP of HT-CDDP cells was significantly increased by GRI-977143. The elevated cell survival to CDDP was suppressed by LPA2 knockdown. Since G12/13 protein stimulates Rho-mediated signaling, RhoA and RhoC knockdown cells were generated from HT1080 cells (HT1080-RhoA and HT1080-RhoC cells, respectively). In the presence of GRI-977143, HT1080-RhoA and HT1080-RhoC cells showed the low cell survival rates to CDDP. On the other hand, Gi protein inhibits adenylyl cyclase (AC) activity. Before cell survival assay, cells were treated with a Gi protein inhibitor, pertussis toxin (PTX) for 24 h. The cell survival rate to CDDP of HT1080 cells was significantly reduced by PTX. Furthermore, when HT1080-RhoA and HT1080-RhoC cells were pretreated with PTX, the cell survival rates to CDDP of both cells were markedly inhibited by PTX. The present results suggest that cooperation of G12/13 and Gi proteins activated by LPA2 enhances the cell survival of HT1080 cells treated with CDDP.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Fibrosarcoma/tratamiento farmacológico , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Receptores del Ácido Lisofosfatídico/genética , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Proteína rhoC de Unión a GTP/metabolismo
19.
Exp Cell Res ; 384(1): 111590, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31479687

RESUMEN

Bacillus Calmette-Guérin (BCG)-activated macrophages (BAMs) have anti-tumor effects, especially on fibrosarcoma cells. However, the mechanism governing this process has not been elucidated to date. TRIM59 is an up-regulated membrane protein expressed on the surface of BAMs. In this study, we found that up-regulated TRIM59 macrophages exhibited excellent growth inhibition on MCA207 fibrosarcoma and induced tumor apoptosis. Moreover, TRIM59 enhanced macrophage infiltration and increased the M1 phenotype macrophages inside the tumor. Furthermore, the cytotoxic T cells and B cells in the spleen and lymphnode have not been affected by TRIM59. These results showed that macrophages expressing TRIM59 exhibited the main cytotoxic effect on tumors. In vitro, we co-cultured TRIM59 up-regulated macrophages fixed with 1% paraformaldehyde or cell culture supernatant and tumor cells. We found that the killing activities of macrophages decreased after treatment with anti-TRIM59 antibody, and the supernatant of TRIM59 up-regulated macrophages had no tumoricidal effect on fibrosarcoma cells, which demonstrated that TRIM59 may be involved in tumoricidal effects via cell-cell contact. In addition, the PI3K-Akt pathway of MCA207 co-cultured with macrophages highly expressing TRIM59 was significantly inhibited, whereas the activation of the PI3K-Akt pathway in MCA207 was not affected after co-culture with TRIM59-CKO macrophages. These results define a vital role of TRIM59 as an anti-tumor effector molecule of BAMs and suggest a new therapeutic target for the treatment of fibrosarcoma.


Asunto(s)
Apoptosis/fisiología , Bacillus/patogenicidad , Fibrosarcoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Animales , Línea Celular Tumoral , Femenino , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mycobacterium bovis/patogenicidad , Fosfatidilinositol 3-Quinasas/metabolismo , Células RAW 264.7 , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
20.
Proc Natl Acad Sci U S A ; 114(46): 12237-12242, 2017 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-29087347

RESUMEN

NIH 3T3 cells grown in conventional Dulbecco's modification of Eagle's basal medium (DME) produce no transformed foci when grown to confluence in 10% calf serum (CS). A few cultures were transformed by ras oncogenes when transfected with DNA from neoplastic cells, but they failed to do so in 80 to 90% of the transfections. However, when they were grown in a medium [molecular, cellular, and developmental biology 402 (MCDB 402)] optimized for their clonal growth in minimal serum, they produced transformed foci without transfection in 10% CS, but not in 2% CS. The first response to growth in MCDB 402 in 2% CS in successive rounds of contact inhibition was uniform increases in saturation density of the population. This was followed by the appearance of transformed foci. A systematic study was made of the dynamics of neoplastic progression in various concentrations of CS in a single round of confluence at 2 and 3 wk, followed by three sequential rounds of confluence in 2% CS for 2 wk. There was a linear relationship between CS concentration and saturation density in the first-round cultures and continuing differences in subsequent cultures. The hyperplastic field of normal-looking cells surrounding transformed foci became increasingly permissive for transformation with serial culture. The dynamics show that epigenetic selection is the major driving force of neoplastic development. Cells from dense foci produced malignant fibrosarcomas in mice, thereby exhibiting a positive relationship between transformation in culture and the development of tumors.


Asunto(s)
Transformación Celular Neoplásica/genética , Medios de Cultivo/farmacología , Epigénesis Genética , Fibrosarcoma/genética , Animales , Bovinos , Recuento de Células , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Medios de Cultivo/química , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Inyecciones , Ratones , Células 3T3 NIH
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA