Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
NMR Biomed ; : e5157, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589764

RESUMEN

Cellular senescence is characterized by stable cell cycle arrest. Senescent cells exhibit a senescence-associated secretory phenotype that can promote tumor progression. The aim of our study was to identify specific nuclear magnetic resonance (NMR) spectroscopy-based markers of cancer cell senescence. For metabolic studies, we employed murine liver carcinoma Harvey Rat Sarcoma Virus (H-Ras) cells, in which reactivation of p53 expression induces senescence. Senescent and nonsenescent cell extracts were subjected to high-resolution proton (1H)-NMR spectroscopy-based metabolomics, and dynamic metabolic changes during senescence were analyzed using a magnetic resonance spectroscopy (MRS)-compatible cell perfusion system. Additionally, the ability of intact senescent cells to degrade the extracellular matrix (ECM) was quantified in the cell perfusion system. Analysis of senescent H-Ras cell extracts revealed elevated sn-glycero-3-phosphocholine, myoinositol, taurine, and creatine levels, with decreases in glycine, o-phosphocholine, threonine, and valine. These metabolic findings were accompanied by a greater degradation index of the ECM in senescent H-Ras cells than in control H-Ras cells. MRS studies with the cell perfusion system revealed elevated creatine levels in senescent cells on Day 4, confirming the 1H-NMR results. These senescence-associated changes in metabolism and ECM degradation strongly impact growth and redox metabolism and reveal potential MRS signals for detecting senescent cancer cells in vivo.

2.
NMR Biomed ; 32(10): e4053, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30693605

RESUMEN

Because of the spatial and temporal heterogeneities of cancers, technologies to investigate cancer cells and the consequences of their interactions with abnormal physiological environments, such as hypoxia and acidic extracellular pH, with stromal cells, and with the extracellular matrix, under controlled conditions, are valuable to gain insights into the functioning of cancers. These insights can lead to an understanding of why cancers invade and metastasize, and identify effective treatment strategies. Here we have provided an overview of the applications of MRI/MRS/MRSI to investigate intact perfused cancer cells, their metabolism and invasion, and their interactions with stromal cells and the extracellular matrix.


Asunto(s)
Comunicación Celular , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Neoplasias/metabolismo , Neoplasias/patología , Perfusión , Humanos , Invasividad Neoplásica , Células del Estroma/patología
3.
NMR Biomed ; 31(8): e3936, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29928787

RESUMEN

Elevated phosphoethanolamine (PE) is frequently observed in MRS studies of human cancers and xenografts. The role of PE in cell survival and the molecular causes underlying this increase are, however, relatively underexplored. In this study, we investigated the roles of ethanolamine kinases (Etnk-1 and 2) and choline kinases (Chk-α and ß) in contributing to increased PE in human breast and pancreatic cancer cells. We investigated the effect of silencing Etnk-1 and Etnk-2 on cell viability as a potential therapeutic strategy. Both breast and pancreatic cancer cells showed higher PE compared with their nonmalignant counterparts. We identified Etnk-1 as a major cause of the elevated PE levels in these cancer cells, with little or no contribution from Chk-α, Chk-ß, or Etnk-2. The increase of PE observed in pancreatic cancer cells in culture was replicated in the corresponding tumor xenografts. Downregulation of Etnk-1 with siRNA resulted in cell cytotoxicity that correlated with PE levels in breast and pancreatic cancer cells. Etnk-1 may provide a potential therapeutic target in breast and pancreatic cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Etanolaminas/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Regulación hacia Abajo , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glicerilfosforilcolina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fósforo/química , Fosforilcolina/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
NMR Biomed ; 29(7): 904-11, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27149683

RESUMEN

Lymphatic vessels serve as the primary route for metastatic spread to lymph nodes. However, it is not clear how interactions between cancer cells and lymphatic endothelial cells (LECs), especially within hypoxic microenvironments, affect the invasion of cancer cells. Here, using an MR compatible cell perfusion assay, we investigated the role of LEC-prostate cancer (PCa) cell interaction in the invasion and degradation of the extracellular matrix (ECM) by two human PCa cell lines, PC-3 and DU-145, under normoxia and hypoxia, and determined the metabolic changes that occurred under these conditions. We observed a significant increase in the invasion of ECM by invasive PC-3 cells, but not poorly invasive DU-145 cells when human dermal lymphatic microvascular endothelial cells (HMVEC-dlys) were present. Enhanced degradation of ECM by PC-3 cells in the presence of HMVEC-dlys identified interactions between HMVEC-dlys and PCa cells influencing cancer cell invasion. The enhanced ECM degradation was partly attributed to increased MMP-9 enzymatic activity in PC-3 cells when HMVEC-dlys were in close proximity. Significantly higher uPAR and MMP-9 expression levels observed in PC-3 cells compared to DU-145 cells may be one mechanism for increased invasion and degradation of matrigel by these cells irrespective of the presence of HMVEC-dlys. Hypoxia significantly decreased invasion by PC-3 cells, but this decrease was significantly attenuated when HMVEC-dlys were present. Significantly higher phosphocholine was observed in invasive PC-3 cells, while higher glycerophosphocholine was observed in DU-145 cells. These metabolites were not altered in the presence of HMVEC-dlys. Significantly increased lipid levels and lipid droplets were observed in PC-3 and DU-145 cells under hypoxia reflecting an adaptive survival response to oxidative stress. These results suggest that in vivo, invasive cells in or near lymphatic endothelial cells are likely to be more invasive and degrade the ECM to influence the metastatic cascade. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/patología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Comunicación Celular , Línea Celular Tumoral , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Humanos , Metástasis Linfática , Espectroscopía de Resonancia Magnética/métodos , Masculino , Invasividad Neoplásica
5.
NMR Biomed ; 28(12): 1697-706, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26503172

RESUMEN

High levels of total choline and phosphocholine (PC) are consistently observed in aggressive cancers. Choline kinase (Chk) catalyzes choline phosphorylation to produce PC in phosphatidylcholine (PtdCho) biosynthesis. PtdCho is the most abundant phospholipid in eukaryotic cell membranes and plays a dual role as the structural component of membranes and as a substrate to produce lipid second messengers such as phosphatidic acid and diacylglycerol. Chk-α, but not Chk-ß, is overexpressed in various cancers, and is closely associated with tumor progression and invasiveness. We have previously shown that downregulation of mRNA using small interfering RNA (siRNA) against Chk-α (siRNA-Chk) or Chk short hairpin RNA, and the resultant decrease of Chk-α protein levels, significantly reduced proliferation in breast cancer cells and tumors. A novel potent and selective small-molecule Chk-α inhibitor, V-11-0711, that inhibits the catalytic activity of Chk has recently been developed. Here, we used triple negative MDA-MB-231 and SUM149 breast cancer cells to further investigate the role of Chk-α in cancer, by examining Chk-α protein levels, cell viability/proliferation, choline phospholipid and lipid metabolism, lipid droplet formation, and apoptosis, following treatment with V-11-0711. Under the conditions used in this study, treatment with V-11-0711 significantly decreased PC levels but did not reduce cell viability as long as Chk-α protein and PtdCho levels were not reduced, suggesting that Chk-α protein and PtdCho, but not PC, may be crucial for breast cancer cell survival. These data also support the approach of antitumor strategies that destabilize Chk-α protein or downregulate PtdCho in breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Supervivencia Celular , Colina Quinasa/metabolismo , Fosfatidilcolinas/metabolismo , Fosforilcolina/metabolismo , Línea Celular Tumoral , Humanos , Espectroscopía de Protones por Resonancia Magnética/métodos
6.
Proc Natl Acad Sci U S A ; 109(8): 2736-41, 2012 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-21690342

RESUMEN

Multiple factors including long-term treatment with tamoxifen are involved in the development of selective estrogen receptor (ER) modulator resistance in ERα-positive breast cancer. Many underlying molecular events that confer resistance are known but a unifying theme is yet to be revealed. In this report, we provide evidence that HOXB7 overexpression renders MCF-7 cells resistant to tamoxifen via cross-talk between receptor tyrosine kinases and ERα signaling. HOXB7 is an ERα-responsive gene. Extended treatment of MCF-7 cells with tamoxifen resulted in progressively increasing levels of HOXB7 expression, along with EGFR and EGFR ligands. Up-regulation of EGFR occurs through direct binding of HOXB7 to the EGFR promoter, enhancing transcriptional activity. Finally, higher expression levels of HOXB7 in the tumor significantly correlated with poorer disease-free survival in ERα-positive patients with breast cancer on adjuvant tamoxifen monotherapy. These studies suggest that HOXB7 acts as a key regulator, orchestrating a major group of target molecules in the oncogenic hierarchy. Functional antagonism of HOXB7 could circumvent tamoxifen resistance.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/metabolismo , Proteínas de Homeodominio/metabolismo , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Activación Enzimática/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Humanos , Estimación de Kaplan-Meier , Ratones , Pronóstico , Transducción de Señal/efectos de los fármacos
7.
Front Mol Biosci ; 11: 1354076, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38584702

RESUMEN

Fibroblasts are versatile cells that play a major role in wound healing by synthesizing and remodeling the extracellular matrix (ECM). In cancers, fibroblasts play an expanded role in tumor progression and dissemination, immunosuppression, and metabolic support of cancer cells. In prostate cancer (PCa), fibroblasts have been shown to induce growth and increase metastatic potential. To further understand differences in the functions of human PCa associated fibroblasts (PCAFs) compared to normal prostate fibroblasts (PFs), we investigated the metabolic profile and ECM degradation characteristics of PFs and PCAFs using a magnetic resonance imaging and spectroscopy compatible intact cell perfusion assay. To further understand how PFs and PCAFs respond to hypoxic tumor microenvironments that are often observed in PCa, we characterized the effects of hypoxia on PF and PCAF metabolism, invasion and PD-L1 expression. We found that under normoxia, PCAFs displayed decreased ECM degradation compared to PFs. Under hypoxia, ECM degradation by PFs increased, whereas PCAFs exhibited decreased ECM degradation. Under both normoxia and hypoxia, PCAFs and PFs showed significantly different metabolic profiles. PD-L1 expression was intrinsically higher in PCAFs compared to PFs. Under hypoxia, PD-L1 expression increased in PCAFs but not in PFs. Our data suggest that PCAFs may not directly induce ECM degradation to assist in tumor dissemination, but may instead create an immune suppressive tumor microenvironment that further increases under hypoxic conditions. Our data identify the intrinsic metabolic, ECM degradation and PD-L1 expression differences between PCAFs and PFs under normoxia and hypoxia that may provide novel targets in PCa treatment.

8.
NMR Biomed ; 26(11): 1501-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23775813

RESUMEN

High choline kinase-α (Chk-α) expression is frequently observed in cancer cells, making it a novel target for pharmacological and molecular inhibition. As inhibiting agents are delivered systemically, it is important to determine Chk-α expression levels in endothelial cells that line both normal and tumor vasculature, and the effect of Chk-α downregulation on these cells. Here, we characterized Chk-α expression and the effect of its downregulation in human umbilical vein endothelial cells (HUVECs) relative to MDA-MB-231 human breast cancer cells. We used small interfering RNA (siRNA) to downregulate Chk-α expression. Basal mRNA levels of Chk-α were approximately three-fold lower in HUVECs relative to MDA-MB-231 breast cancer cells. Consistent with the differences in Chk-α protein levels, phosphocholine levels were approximately 10-fold lower in HUVECs relative to MDA-MB-231 cells. Transient transfection with siRNA-Chk resulted in comparable levels of mRNA and protein in MDA-MB-231 breast cancer cells and HUVECs. However, there was a significant reduction in proliferation in MDA-MB-231 cells, but not in HUVECs. No significant difference in CD31 immunostaining was observed in tumor sections obtained from mice injected with control luciferase-short hairpin (sh)RNA or Chk-shRNA lentivirus. These data suggest that systemically delivered agents that downregulate Chk-α in tumors will not affect endothelial cell proliferation during delivery, and further support the development of Chk-α downregulation as a cancer-specific treatment.


Asunto(s)
Colina Quinasa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Animales , Extractos Celulares , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Colina Quinasa/genética , Regulación hacia Abajo , Regulación Enzimológica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Espectroscopía de Resonancia Magnética , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
9.
Cancer Biol Ther ; 24(1): 2184145, 2023 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-37389973

RESUMEN

Vascular endothelial growth factor (VEGF) plays key roles in angiogenesis, vasculogenesis, and wound healing. In cancers, including triple negative breast cancer (TNBC), VEGF has been associated with increased invasion and metastasis, processes that require cancer cells to traverse through the extracellular matrix (ECM) and establish angiogenesis at distant sites. To further understand the role of VEGF in modifying the ECM, we characterized VEGF-mediated changes in the ECM of tumors derived from TNBC MDA-MB-231 cells engineered to overexpress VEGF. We established that increased VEGF expression by these cells resulted in tumors with reduced collagen 1 (Col1) fibers, fibronectin, and hyaluronan. Molecular characterization of tumors identified an increase of MMP1, uPAR, and LOX, and a decrease of MMP2, and ADAMTS1. α-SMA, a marker of cancer associated fibroblasts (CAFs), increased, and FAP-α, a marker of a subset of CAFs associated with immune suppression, decreased with VEGF overexpression. Analysis of human data from The Cancer Genome Atlas Program confirmed mRNA differences for several molecules when comparing TNBC with high and low VEGF expression. We additionally characterized enzymatic changes induced by VEGF overexpression in three different cancer cell lines that clearly identified autocrine-mediated changes, specifically uPAR, in these enzymes. Unlike the increase of Col1 fibers and fibronectin mediated by VEGF during wound healing, in the TNBC model, VEGF significantly reduced key protein components of the ECM. These results further expand our understanding of the role of VEGF in cancer progression and identify potential ECM-related targets to disrupt this progression.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Factor A de Crecimiento Endotelial Vascular , Humanos , Comunicación Autocrina , Matriz Extracelular , Fibronectinas/genética , Neoplasias de la Mama Triple Negativas/genética , Factor A de Crecimiento Endotelial Vascular/genética
10.
Front Oncol ; 13: 1068405, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36937451

RESUMEN

Fibroblast activation protein-α (FAP-α) is a transmembrane serine protease that is attracting significant interest as it is expressed by a subgroup of cancer-associated fibroblasts that play a role in immune suppression and cancer metastasis. FAP-α is also expressed by some cancer cells, such as melanoma, colorectal and breast cancer cells. Triple negative breast cancer (TNBC) is an aggressive cancer that urgently requires identification of novel targets for therapy. To expand our understanding of the functional roles of FAP-α in TNBC we engineered a human TNBC cell line, MDA-MB-231, to stably overexpress FAP-α and characterized changes in metabolism by 1H magnetic resonance spectroscopy, cell proliferation, migration characterized by wound healing, and invasion. FAP-α overexpression resulted in significant alterations in myoinositol, choline metabolites, creatine, and taurine, as well as a significant increase of migration and invasion, although proliferation remained unaltered. The increase of migration and invasion are consistent with the known activities of FAP-α as an exopeptidase and endopeptidase/gelatinase/collagenase in tissue remodeling and repair, and in cell migration. We additionally determined the effects of FAP-α overexpression on the human fibrosarcoma HT1080 cell line that showed increased migration, accompanied by limited changes in metabolism that identified the dependency of the metabolic changes on cell type. These metabolic data identify a previously unknown role of FAP-α in modifying cancer cell metabolism in the TNBC cell line studied here that may provide new insights into its functional roles in cancer progression.

11.
NMR Biomed ; 25(5): 746-54, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21953546

RESUMEN

The expression of cyclooxygenase-2 (COX-2) is observed in approximately 40% of breast cancers. A major product of the COX-2-catalyzed reaction, prostaglandin E(2), is an inflammatory mediator that participates in several biological processes, and influences invasion, vascularization and metastasis. Using noninvasive MRI and MRS, we determined the effect of COX-2 downregulation on the metabolism and invasion of intact poorly differentiated MDA-MB-231 human breast cancer cells stably expressing COX-2 short hairpin RNA. Dynamic tracking of invasion, extracellular matrix degradation and metabolism was performed with an MRI- and MRS-compatible cell perfusion assay under controlled conditions of pH, temperature and oxygenation over the course of 48 h. COX-2-silenced cells exhibited a significant decrease in invasion relative to parental cells that was consistent with the reduced expression of invasion-associated matrix metalloproteinase genes and an increased level of the tissue inhibitor of metalloproteinase-1. We identified, for the first time, a role for COX-2 in mediating changes in choline phospholipid metabolism, and established that choline kinase expression is partly dependent on COX-2 function. COX-2 silencing resulted in a significant decrease in phosphocholine and total choline that was detected by MRS. In addition, a significant increase in lipids, as well as lipid droplet formation, was observed. COX-2 silencing transformed parental cell metabolite patterns to those characteristic of less aggressive cancer cells. These new functional roles of COX-2 may identify new biomarkers and new targets for use in combination with COX-2 targeting to prevent invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Colina/metabolismo , Ciclooxigenasa 2/metabolismo , Fosfolípidos/metabolismo , Fosforilcolina/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Colina Quinasa/metabolismo , Ciclooxigenasa 2/genética , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Invasividad Neoplásica
12.
Cancers (Basel) ; 14(3)2022 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-35158887

RESUMEN

(1) Background: Despite advances in surgical approaches and drug development, ovarian cancer is still a leading cause of death from gynecological malignancies. Patients diagnosed with late-stage disease are treated with aggressive surgical resection and chemotherapy, but recurrence with resistant disease is often observed following treatment. There is a critical need for effective therapy for late-stage ovarian cancer. Photoimmunotherapy (PIT), using an antibody conjugated to a near infrared (NIR) dye, constitutes an effective theranostic strategy to detect and selectively eliminate targeted cell populations. (2) Methods: Here, we are targeting program death ligand 1 (PD-L1) using NIR-PIT in a syngeneic mouse model of ovarian cancer. PD-L1 PIT-mediated cytotoxicity was quantified in RAW264.7 macrophages and ID8-Defb29-VEGF cells in culture, and in vivo with orthotopic ID8-Defb29-VEGF tumors. (3) Results: Treatment efficacy was observed both in vitro and in vivo. (4) Conclusions: Our data highlight the need for further investigations to assess the potential of using NIR-PIT for ovarian cancer therapy to improve the treatment outcome of ovarian cancer.

13.
NMR Biomed ; 23(6): 633-42, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20623626

RESUMEN

A direct correlation exists between increased choline kinase (Chk) expression, and the resulting increase of phosphocholine levels, and histological tumor grade. To better understand the function of Chk and choline phospholipid metabolism in breast cancer we have stably overexpressed one of the two isoforms of Chk-alpha known to be upregulated in malignant cells, in non-invasive MCF-7 human breast cancer cells. Dynamic tracking of cell invasion and cell metabolism were studied with a magnetic resonance (MR) compatible cell perfusion assay. The MR based invasion assay demonstrated that MCF-7 cells overexpressing Chk-alpha (MCF-7-Chk) exhibited an increase of invasion relative to control MCF-7 cells (0.84 vs 0.3). Proton MR spectroscopy studies showed significantly higher phosphocholine and elevated triglyceride signals in Chk overexpressing clones compared to control cells. A test of drug resistance in MCF-7-Chk cells revealed that these cells had an increased resistance to 5-fluorouracil and higher expression of thymidylate synthase compared to control MCF-7 cells. To further characterize increased drug resistance in these cells, we performed rhodamine-123 efflux studies to evaluate drug efflux pumps. MCF-7-Chk cells effluxed twice as much rhodamine-123 compared to MCF-7 cells. Chk-alpha overexpression resulted in MCF-7 human breast cancer cells acquiring an increasingly aggressive phenotype, supporting the role of Chk-alpha in mediating invasion and drug resistance, and the use of phosphocholine as a biomarker of aggressive breast cancers.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Colina Quinasa/metabolismo , Resistencia a Antineoplásicos/fisiología , Isoenzimas/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Colina Quinasa/genética , Femenino , Humanos , Isoenzimas/genética , Espectroscopía de Resonancia Magnética/métodos , Organismos Modificados Genéticamente , Fosforilcolina/metabolismo
14.
Front Oncol ; 10: 599204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33585215

RESUMEN

In magnetic resonance metabolic imaging, signal from the water content is frequently used for normalization to derive quantitative or semi-quantitative values of metabolites in vivo or ex vivo tumors and tissues. Ex vivo high-resolution metabolic characterization of tumors with magnetic resonance spectroscopy (MRS) provides valuable information that can be used to drive the development of noninvasive MRS biomarkers and to identify metabolic therapeutic targets. Variability in the water content between tumor and normal tissue can result in over or underestimation of metabolite concentrations when assuming a constant water content. Assuming a constant water content can lead to masking of differences between malignant and normal tissues both in vivo and ex vivo. There is a critical need to develop biomarkers to detect pancreatic cancer and to develop novel treatments. Our purpose here was to determine the differences in water content between pancreatic tumors and normal pancreatic tissue as well as other organs to accurately quantify metabolic differences when using the water signal for normalization. Our data identify the importance of factoring the differences in water content between tumors and organs. High-resolution proton spectra of tumors and pancreatic tissue extracts normalized to the water signal, assuming similar water content, did not reflect the significantly increased total choline observed in tumors in vivo without factoring the differences in water content. We identified significant differences in the collagen 1 content between Panc1 and BxPC3 pancreatic tumors and the pancreas that can contribute to the differences in water content that were observed.

15.
Curr Cancer Rep ; 2(1): 54-63, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-35814639

RESUMEN

Our purpose was to understand the effects of normoxia or hypoxia on 5-fluorouracil (5-FU) treatment in triple negative breast cancer (TNBC) cells, and characterize the molecular changes in hypoxia inducible factors (HIFs) and cyclooxygenase-2 (COX-2) following treatment. Cell viability and protein levels of HIFs and COX-2 were determined after wild type and HIF silenced MDA-MB-231 cells, and wild type SUM-149 cells, were treated with 5-FU under normoxia or hypoxia. 5-FU reduced cell viability to the same levels irrespective of normoxia or hypoxia. HIF silenced MDA-MB-231 cells showed comparable changes in cell viability, supporting observations that hypoxia and the HIF pathways did not significantly influence cell viability reduction by 5-FU. Our data suggest that HIF-2α accumulation may predispose cancer cells to cell death under hypoxia. SUM-149 cells that have higher COX-2 and HIF-2α following 24 h of hypoxia, were more sensitive to 96 h of hypoxia compared to MDA-MB-231 cells, and were more sensitive to 5-FU than MDA-MB-231 cells. COX-2 levels changed with hypoxia and with 5-FU treatment but patterns were different between the two cell lines. At 96 h, COX-2 increased in both untreated and 5-FU treated cells under hypoxia in MDA-MB-231 cells. In SUM-149 cells, only treatment with 5-FU increased COX-2 at 96 h of hypoxia. Cells that survive hypoxia and 5-FU treatment may exhibit a more aggressive phenotype. Our results support understanding interactions between HIF and COX-2 with chemotherapeutic agents under normoxia and hypoxia, and investigating the use of COX-2 inhibitors in these settings.

16.
Neoplasia ; 22(12): 679-688, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33142234

RESUMEN

Hypoxia is frequently observed in human prostate cancer, and is associated with chemoresistance, radioresistance, metastasis, and castrate-resistance. Our purpose in these studies was to perform hypoxia theranostics by combining in vivo hypoxia imaging and hypoxic cancer cell targeting in a human prostate cancer xenograft. This was achieved by engineering PC3 human prostate cancer cells to express luciferase as well as a prodrug enzyme, yeast cytosine deaminase, under control of hypoxic response elements (HREs). Cancer cells display an adaptive response to hypoxia through the activation of several genes mediated by the binding of hypoxia inducible factors (HIFs) to HRE in the promoter region of target gene that results in their increased transcription. HIFs promote key steps in tumorigenesis, including angiogenesis, metabolism, proliferation, metastasis, and differentiation. HRE-driven luciferase expression allowed us to detect hypoxia in vivo to time the administration of the nontoxic prodrug 5-fluorocytosine that was converted by yeast cytosine deaminase, expressed under HRE regulation, to the chemotherapy agent 5-fluorouracil to target hypoxic cells. Conversion of 5-fluorocytosine to 5-fluorouracil was detected in vivo by 19F magnetic resonance spectroscopy. Morphological and immunohistochemical staining and molecular analyses were performed to characterize tumor microenvironment changes in cancer-associated fibroblasts, cell viability, collagen 1 fiber patterns, and HIF-1α. These studies expand our understanding of the effects of eliminating hypoxic cancer cells on the tumor microenvironment and in reducing stromal cell populations such as cancer-associated fibroblasts.


Asunto(s)
Hipoxia/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Biomarcadores , Hipoxia de la Célula/genética , Línea Celular Tumoral , Supervivencia Celular , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Genes Reporteros , Humanos , Hipoxia/genética , Hipoxia/terapia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Masculino , Ratones , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/terapia , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Neoplasia ; 21(2): 239-246, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30639975

RESUMEN

Metastatic dissemination continues to be a major cause of prostate cancer (PCa) mortality, creating a compelling need to understand factors that play a role in the metastatic cascade. Since hypoxia plays an important role in PCa aggressiveness, we characterized patterns of hypoxia in the primary tumor and metastatic environments of a human PCa xenograft. We previously developed and characterized an imaging strategy based on the hypoxia response element (HRE)-driven expression of long-lived enhanced green fluorescent protein (EGFP) and short-lived luciferase (luc) fused to the oxygen-dependent degradation domain in human PCa PC-3 cells. Both reporter proteins were placed under the transcriptional control of a five-tandem repeat HRE sequence. PC-3 cells also constitutively expressed the tdTomato red fluorescent protein, allowing cancer cell detection in vivo. This "timer" strategy can provide information on the temporal evolution of HIF activity and hypoxia in tumors. Here, for the first time, we performed in vivo and ex vivo imaging of this dual HIF reporter system in PC-3 metastatic tumors implanted orthotopically in the prostate and PC-3 nonmetastatic tumors implanted subcutaneously. We observed distinct patterns of EGFP and luc expression in subcutaneous and orthotopic tumors, and in metastatic nodules, that provide new insights into the presence of hypoxia at primary and metastatic tumor sites, and of the role of hypoxia in metastasis.


Asunto(s)
Hipoxia/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Microambiente Tumoral , Animales , Biomarcadores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Xenoinjertos , Humanos , Hipoxia/genética , Masculino , Ratones , Imagen Molecular , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias de la Próstata/diagnóstico por imagen , Neoplasias de la Próstata/genética , Microambiente Tumoral/genética
18.
ACS Appl Mater Interfaces ; 11(8): 7850-7861, 2019 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-30707559

RESUMEN

Biomimetic nanoparticles (NPs) combine the flexibility and reproducibility of synthetic materials with the functionality of biological materials. Here, we developed and characterized biomimetic poly(lactic- co-glycolic acid) (PLGA) NPs coated with human cancer cell membrane fractions (CCMFs) to form CCMF-coated PLGA (CCMF-PLGA) NPs. We evaluated the ability of these CCMF-PLGA NPs to disrupt cancer cell-stromal cell interactions and to induce an immune response. Western blot analysis verified the plasma membrane purity of CCMFs. Confocal fluorescence microscopy and flow cytometry confirmed the presence of intact membrane-associated proteins including CXCR4 and CD44 following membrane derivation and coating. CCMFs and CCMF-PLGA NPs were capable of inhibiting cancer cell migration toward human mammary fibroblasts. Intravenous injection of CCMF-PLGA NPs significantly reduced experimental metastasis in vivo. Following immunization of Balb/c mice, near-infrared fluorescence imaging confirmed the migration of NPs to proximal draining lymph nodes (LNs). A higher percentage of CD8+ and CD4+ cytotoxic T-lymphocyte populations was observed in spleens and LNs of CCMF-PLGA NP-immunized mice. Splenocytes isolated from CCMF-PLGA NP-immunized mice had the highest number of interferon gamma-producing T-cells as detected by the ELISpot assay. CCMF-PLGA NPs hold promise for disrupting cancer cell-stromal cell interactions and for priming the immune system in cancer immunotherapy.


Asunto(s)
Materiales Biomiméticos/uso terapéutico , Membrana Celular/química , Neoplasias Pulmonares/prevención & control , Nanopartículas/química , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/farmacología , Neoplasias de la Mama/patología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Receptores de Hialuranos/química , Interferón gamma/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos BALB C , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Receptores CXCR4/química
19.
Mol Cancer Res ; 5(5): 435-42, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17510310

RESUMEN

Cyclooxygenases (COX) are rate-limiting enzymes involved in the conversion of PLA(2)-mobilized arachidonic acid into prostaglandins and thromboxanes. COX-2 is a key mediator of inflammation during both physiologic and pathologic responses to endogenous stimuli and infectious agents. Its overexpression has been detected in different cancers, including that of the breast. Using RNA interference, we have reduced the expression of COX-2 in the highly malignant breast cancer cell line MDA-MB-231 below detectable levels in response to interleukin-1 beta or 12-O-tetradecanoylphorbol-13-acetate treatment. Microarray analysis showed that COX-2 silencing resulted in the loss of mRNA expression of several oncogenic markers, such as matrix metalloproteinase-1, chemokine (C-X-C motif) receptor 4, and interleukin-11, which have been correlated with poor disease outcome, and in the up-regulation of antimetastatic transcripts, such as thrombospondin-1 and Epstein-Barr-Induced 3. Cells lacking COX-2 were less able to invade reconstituted extracellular matrix than parental cells in vitro. Consistent with these changes, loss of COX-2 resulted in the abolition or the significant delay of tumor onset when the cells were injected in the mammary fat pad of severe combined immunodeficient mice. Finally, silencing of COX-2 resulted in the inhibition of metastasis to the lungs of severe combined immunodeficient mice after intravenous injection. These data show that silencing of COX-2 abolishes the metastatic potential of MDA-MB-231 cells in vivo.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/metabolismo , Silenciador del Gen , Animales , Neoplasias de la Mama/genética , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Células Clonales , Femenino , Humanos , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Transcripción Genética
20.
J Med Chem ; 51(12): 3572-82, 2008 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-18517259

RESUMEN

We previously observed that bacterial cytosine deaminase (bCD) conjugated with multimodal imaging reporter labeled poly-L-lysine (PLL) demonstrated high therapeutic efficacy in an enzyme/prodrug cancer therapeutic strategy. To understand the role of polycationic PLL in the cellular uptake of bCD-PLL conjugate, two control molecules, bCD-BF, without the PLL moiety, and bCD-AcPLL, with all positive charges in PLL neutralized, were prepared. bCD-PLL demonstrated about 50 times higher cellular uptake than that of control molecules in human breast MDA-MB-231 cancer cells. Internalized bCD-PLL demonstrated high enzymatic stability in cell cultures as indicated by significant cytotoxicity after addition of prodrug, whereas no obvious cytotoxicity was detected by control molecules. These data indicate that conjugated PLL not only provides a multivalent modification platform to facilitate the delivery of a high payload of imaging reporters or targeting moieties without compromising enzymatic activity but also enhances therapeutic efficacy by accelerating the intracellular uptake of prodrug-activating enzyme.


Asunto(s)
Antineoplásicos/metabolismo , Proteínas Bacterianas/metabolismo , Citosina Desaminasa/metabolismo , Polilisina/metabolismo , Profármacos/metabolismo , Proteínas Bacterianas/química , Neoplasias de la Mama , Línea Celular Tumoral , Células Cultivadas , Citosina Desaminasa/química , Endocitosis , Células Endoteliales/metabolismo , Femenino , Citometría de Flujo , Fluorescencia , Humanos , Cinética , Microscopía Confocal , Polilisina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA