Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Metastasis Rev ; 38(1-2): 149-155, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30806853

RESUMEN

The extracellular pH of solid tumors is unequivocally acidic due to a combination of high rates of lactic acid production (a consequence of fermentative glycolytic metabolism) and poor perfusion. This has been documented by us and others in a wide variety of solid tumor models, primarily using magnetic resonance spectroscopic imaging (MRSI). This acidity contributes to tumor progression by inducing genome instability, promoting local invasion and metastases, inhibiting anti-tumor immunity, and conferring resistance to chemo- and radio-therapies. Systemic buffer therapies can neutralize tumor acidity and has been shown to inhibit local invasion and metastasis and improve immune surveillance in a variety of cancer model systems. This review will revisit the causes and consequences of acidosis by summarizing strategies used by cancer cells to adapt to acidosis, and how this acidity associated with carcinogenesis, metastasis, and immune function. Finally, this review will discuss how neutralization of acidity can be used to inhibit carcinogenesis and metastasis and improve anti-cancer immunotherapy.


Asunto(s)
Acidosis/metabolismo , Neoplasias/metabolismo , Bicarbonato de Sodio/farmacología , Acidosis/tratamiento farmacológico , Acidosis/inmunología , Animales , Tampones (Química) , Humanos , Concentración de Iones de Hidrógeno , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología
2.
Mol Pharm ; 13(2): 534-44, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26713599

RESUMEN

Fluorescence molecular imaging can be employed for the development of novel cancer targeting agents. Herein, we investigated the pharmacokinetics (PK) and cellular uptake of Dmt-Tic-Cy5, a delta-opioid receptor (δOR) antagonist-fluorescent dye conjugate, as a tumor-targeting molecular imaging agent. δOR expression is observed normally in the CNS, and pathologically in some tumors, including lung liver and breast cancers. In vitro, in vivo, and ex vivo experiments were conducted to image and quantify the fluorescence signal associated with Dmt-Tic-Cy5 over time using in vitro and intravital fluorescence microscopy and small animal fluorescence imaging of tumor-bearing mice. We observed specific retention of Dmt-Tic-Cy5 in tumors with maximum uptake in δOR-expressing positive tumors at 3 h and observable persistence for >96 h; clearance from δOR nonexpressing negative tumors by 6 h; and systemic clearance from normal organs by 24 h. Live-cell and intravital fluorescence microscopy demonstrated that Dmt-Tic-Cy5 had sustained cell-surface binding lasting at least 24 h with gradual internalization over the initial 6 h following administration. Dmt-Tic-Cy5 is a δOR-targeted agent that exhibits long-lasting and specific signal in δOR-expressing tumors, is rapidly cleared from systemic circulation, and is not retained in non-δOR-expressing tissues. Hence, Dmt-Tic-Cy5 has potential as a fluorescent tumor imaging agent.


Asunto(s)
Carbocianinas/farmacocinética , Neoplasias del Colon/tratamiento farmacológico , Dipéptidos/farmacocinética , Colorantes Fluorescentes/química , Receptores Opioides delta/química , Tetrahidroisoquinolinas/farmacocinética , Animales , Apoptosis , Carbocianinas/administración & dosificación , Proliferación Celular , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Dipéptidos/administración & dosificación , Femenino , Humanos , Técnicas para Inmunoenzimas , Cinética , Ratones , Ratones Desnudos , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacocinética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectroscopía Infrarroja Corta , Tetrahidroisoquinolinas/administración & dosificación , Distribución Tisular , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Biomedicines ; 12(2)2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38398062

RESUMEN

Acidosis is an important immunosuppressive mechanism that leads to tumor growth. Therefore, we investigated the neutralization of tumor acidity to improve immunotherapy response. L-DOS47, a new targeted urease immunoconjugate designed to neutralize tumor acidity, has been well tolerated in phase I/IIa trials. L-DOS47 binds to CEACAM6, a cell-surface protein that is highly expressed in gastrointestinal cancers, allowing urease to cleave endogenous urea into two NH4+ and one CO2, thereby raising local pH. To test the synergetic effect of neutralizing tumor acidity with immunotherapy, we developed a pancreatic orthotopic murine tumor model (KPC961) expressing human CEACAM6. Using chemical exchange saturation transfer-magnetic resonance imaging (CEST-MRI) to measure the tumor extracellular pH (pHe), we confirmed that L-DOS47 raises the tumor pHe from 4 h to 96 h post injection in acidic tumors (average increase of 0.13 units). Additional studies showed that combining L-DOS47 with anti-PD1 significantly increases the efficacy of the anti-PD1 monotherapy, reducing tumor growth for up to 4 weeks.

4.
Mol Pharm ; 10(8): 3175-85, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23763620

RESUMEN

The melanocortin 1 receptor (MC1R) is overexpressed in most melanoma metastases, making it a promising target for imaging of melanomas. In this study, the expression of MC1R in a large fraction of patients with melanoma was confirmed using mRNA and tissue microarray. Here, we have characterized the in vivo tumor and tissue distribution and pharmacokinetics (PK) of uptake and clearance of a MC1R specific peptidomimetic ligand conjugated to a near-infrared fluorescent dye. We propose an interdisciplinary framework to bridge the different time and space scales of ligand-tumor-host interactions: intravital fluorescence microscopy to quantify probe internalization at the cellular level, a xenograft tumor model for whole body pharmacokinetics, and a computational pharmacokinetic model for integration and interpretation of experimental data. Administration of the probe into mice bearing tumors with high and low MC1R expression demonstrated normalized image intensities that correlated with expression levels (p < 0.05). The biodistribution study showed high kidney uptake as early as 30 min postinjection. The PK computational model predicted the presence of receptors in the kidneys with a lower affinity, but at higher numbers than in the tumors. As the mouse kidney is known to express the MC5R, this hypothesis was confirmed by both coinjection of a ligand with higher MC5R affinity compared to MC1R and by injection of lower probe concentrations (e.g., 1 nmol/kg), both leading to decreased kidney accumulation of the MC1R ligand. In addition, through this interdisciplinary approach we could predict the rates of ligand accumulation and clearance into and from organs and tumors, and the amount of injected ligand required to have maximum specific retention in tumors. These predictions have potential to aid in the translation of a targeted agent from lab to the clinic. In conclusion, the characterized MC1R-specific probe has excellent potential for in vivo detection of melanoma metastases. The process of cell-surface marker validation, targeted imaging probe development, and in vitro, in vivo, and in silico characterization described in this study can be generally applied to preclinical development of targeted agents.


Asunto(s)
Melanoma/metabolismo , Sondas Moleculares/metabolismo , Sondas Moleculares/farmacocinética , Receptor de Melanocortina Tipo 1/metabolismo , Animales , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Ratones , Modelos Teóricos , Receptor de Melanocortina Tipo 1/genética , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 83(5): 720-734, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36480167

RESUMEN

Induction of cell death represents a primary goal of most anticancer treatments. Despite the efficacy of such approaches, a small population of "persisters" develop evasion strategies to therapy-induced cell death. While previous studies have identified mechanisms of resistance to apoptosis, the mechanisms by which persisters dampen other forms of cell death, such as pyroptosis, remain to be elucidated. Pyroptosis is a form of inflammatory cell death that involves formation of membrane pores, ion gradient imbalance, water inflow, and membrane rupture. Herein, we investigate mechanisms by which cancer persisters resist pyroptosis, survive, then proliferate in the presence of tyrosine kinase inhibitors (TKI). Lung, prostate, and esophageal cancer persister cells remaining after treatments exhibited several hallmarks indicative of pyroptosis resistance. The inflammatory attributes of persisters included chronic activation of inflammasome, STING, and type I interferons. Comprehensive metabolomic characterization uncovered that TKI-induced pyroptotic persisters display high methionine consumption and excessive taurine production. Elevated methionine flux or exogenous taurine preserved plasma membrane integrity via osmolyte-mediated effects. Increased dependency on methionine flux decreased the level of one carbon metabolism intermediate S-(5'-adenosyl)-L-homocysteine, a determinant of cell methylation capacity. The consequent increase in methylation potential induced DNA hypermethylation of genes regulating metal ion balance and intrinsic immune response. This enabled thwarting TKI resistance by using the hypomethylating agent decitabine. In summary, the evolution of resistance to pyroptosis can occur via a stepwise process of physical acclimation and epigenetic changes without existing or recurrent mutations. SIGNIFICANCE: Methionine enables cancer cells to persist by evading pyroptotic osmotic lysis, which leads to genome-wide hypermethylation that allows persisters to gain proliferative advantages.


Asunto(s)
Neoplasias , Piroptosis , Humanos , Piroptosis/genética , Metionina , Apoptosis , Inflamasomas/metabolismo , Muerte Celular , Racemetionina/farmacología
6.
bioRxiv ; 2023 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-37693389

RESUMEN

Acidosis is an important immunosuppressive mechanism that leads to tumor growth. Therefore, we investigated the neutralization of tumor acidity to improve immunotherapy response. L-DOS47, a new targeted urease immunoconjugate designed to neutralize tumor acidity, has been well tolerated in phase I/IIa trials. L-DOS47 binds CEACAM6, a cell surface protein highly expressed in gastrointestinal cancers, allowing urease to cleave endogenous urea into two NH4+ and one CO2, thereby raising local pH. To test the synergetic effect of neutralizing tumor acidity with immunotherapy, we developed a pancreatic orthotopic murine tumor model (KPC961) expressing human CEACAM6. Our results demonstrate that combining L DOS47 with anti-PD1 significantly increases the efficacy of anti-PD1 monotherapy, reducing tumor growth for up to 4 weeks.

7.
Nat Commun ; 11(1): 4113, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32807791

RESUMEN

The acidic pH of tumors profoundly inhibits effector functions of activated CD8 + T-cells. We hypothesize that this is a physiological process in immune regulation, and that it occurs within lymph nodes (LNs), which are likely acidic because of low convective flow and high glucose metabolism. Here we show by in vivo fluorescence and MR imaging, that LN paracortical zones are profoundly acidic. These acidic niches are absent in athymic Nu/Nu and lymphodepleted mice, implicating T-cells in the acidifying process. T-cell glycolysis is inhibited at the low pH observed in LNs. We show that this is due to acid inhibition of monocarboxylate transporters (MCTs), resulting in a negative feedback on glycolytic rate. Importantly, we demonstrate that this acid pH does not hinder initial activation of naïve T-cells by dendritic cells. Thus, we describe an acidic niche within the immune system, and demonstrate its physiological role in regulating T-cell activation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Ganglios Linfáticos/metabolismo , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Citometría de Flujo , Concentración de Iones de Hidrógeno , Inmunoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Fosfofructoquinasa-1/genética , Fosfofructoquinasa-1/metabolismo
8.
PLoS One ; 14(1): e0211012, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30682160

RESUMEN

99m-Technetium-labeled (99mTc) erythrocyte imaging with planar scintigraphy is widely used for evaluating both patients with occult gastrointestinal bleeding and patients at risk for chemotherapy-induced cardiotoxicity. While a number of alternative radionuclide-based blood pool imaging agents have been proposed, none have yet to achieve widespread clinical use. Here, we present both in vitro and small animal in vivo imaging evidence that the high physiological expression of the glucose transporter GLUT1 on human erythrocytes allows uptake of the widely available radiotracer 2-deoxy-2-(18F)fluoro-D-glucose (FDG), at a rate and magnitude sufficient for clinical blood pool positron emission tomographic (PET) imaging. This imaging technique is likely to be amenable to rapid clinical translation, as it can be achieved using a simple FDG labeling protocol, requires a relatively small volume of phlebotomized blood, and can be completed within a relatively short time period. As modern PET scanners typically have much greater count detection sensitivities than that of commonly used clinical gamma scintigraphic cameras, FDG-labeled human erythrocyte PET imaging may not only have significant advantages over 99mTc-labeled erythrocyte imaging, but may have other novel blood pool imaging applications.


Asunto(s)
Eritrocitos/metabolismo , Fluorodesoxiglucosa F18/farmacología , Transportador de Glucosa de Tipo 1/metabolismo , Marcaje Isotópico , Tomografía de Emisión de Positrones/métodos , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratas , Ratas Endogámicas F344
9.
Cancer Res ; 66(23): 11219-27, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145867

RESUMEN

Tumor cells express HYAL1 hyaluronidase, which degrades hyaluronic acid. HYAL1 expression in bladder cancer cells promotes tumor growth, invasion, and angiogenesis. We previously described five alternatively spliced variants of HYAL1 that encode enzymatically inactive proteins. The HYAL1-v1 variant lacks a 30-amino acid sequence that is present in HYAL1. In this study, we examined whether HYAL1-v1 expression affects bladder cancer growth and invasion by stably transfecting HT1376 bladder cancer cells with a HYAL1-v1 cDNA construct. Although HYAL1-v1 transfectants expressed equivalent levels of enzymatically active HYAL1 protein when compared with vector transfectants, their conditioned medium had 4-fold less hyaluronidase activity due to a noncovalent complex formed between HYAL1 and HYAL1-v1 proteins. HYAL1-v1 transfectants grew 3- to 4-fold slower due to cell cycle arrest in the G(2)-M phase and increased apoptosis. In HYAL1-v1 transfectants, cyclin B1, cdc2/p34, and cdc25c levels were > or =2-fold lower than those in vector transfectants. The increased apoptosis in HYAL1-v1 transfectants was due to the extrinsic pathway involving Fas and Fas-associated death domain up-regulation, caspase-8 activation, and BID cleavage, leading to caspase-9 and caspase-3 activation and poly(ADP-ribose) polymerase cleavage. When implanted in athymic mice, HYAL1-v1-expressing tumors grew 3- to 4-fold slower and tumor weights at day 35 were 3- to 6-fold less than the vector tumors (P < 0.001). Whereas vector tumors were infiltrating and had high mitoses and microvessel density, HYAL1-v1 tumors were necrotic, infiltrated with neutrophils, and showed low mitoses and microvessel density. Therefore, HYAL-v1 expression may negatively regulate bladder tumor growth, infiltration, and angiogenesis.


Asunto(s)
Empalme Alternativo , Hialuronoglucosaminidasa/genética , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Apoptosis/genética , Apoptosis/fisiología , Caspasas/metabolismo , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Expresión Génica/genética , Terapia Genética , Humanos , Hialuronoglucosaminidasa/metabolismo , Immunoblotting , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transfección , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/terapia
10.
Sci Rep ; 8(1): 3638, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29483578

RESUMEN

Targeted therapy has held promise to be a successful anticancer treatment due to its specificity towards tumor cells that express the target receptors. However, not all targeting drugs used in the clinic are equally effective in tumor eradication. To examine which biochemical and biophysical properties of targeted agents are pivotal for their effective distribution inside the tumor and their efficient cellular uptake, we combine mathematical micro-pharmacological modeling with in vivo imaging of targeted human xenograft tumors in SCID mice. The mathematical model calibrated to experimental data was used to explore properties of the targeting ligand (diffusion and affinity) and ligand release schemes (rates and concentrations) with a goal to identify the properties of cells and ligands that enable high receptor saturation. By accounting for heterogeneities typical of in vivo tumors, our model was able to identify cell- and tissue-level barriers to efficient drug uptake. This work provides a base for utilizing experimentally measurable properties of a ligand-targeted agent and patient-specific attributes of the tumor tissue to support the development of novel targeted imaging agents and for improvement in their delivery to individual tumor cells.


Asunto(s)
Modelos Teóricos , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Ratones SCID , Microscopía Fluorescente , Neoplasias Pancreáticas/metabolismo
11.
Int J Oncol ; 31(2): 441-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17611702

RESUMEN

Lysophosphatidic acid (LPA) is an important intercellular signaling molecule involved in a myriad of biological responses. Elevated concentrations of LPA are present in the ascites and plasma of ovarian cancer patients suggesting a role for LPA in the pathophysiology of ovarian cancer. We have demonstrated previously that oleoyl (18:1) LPA at concentrations present in ascites induces the secretion of urokinase plasminogen activator (uPA) from ovarian cancer cells, possibly linking LPA to cellular invasion. In this study we sought to elucidate which signaling pathway(s) are involved in LPA-mediated secretion of uPA from ovarian cancer cells. Specific inhibitors were utilized to determine if interference with the p38(MAPK), p42/44(MAPK), and PI3K pathways functionally blocked LPA-mediated uPA secretion. LPA stimulation of ovarian cancer cells markedly increased the phosphorylation and activity of p38(MAPK), p42/p44(MAPK), and PI3K. Both tyrosine phosphorylation and Src kinase activity were required for optimal activation of signaling by LPA including phosphorylation of p38(MAPK). Inhibition of p38(MAPK) signaling by SB202190 completely abrogated LPA-induced uPA secretion, while inhibition of the p42/44(MAPK) or PI3K pathways with PD98059 or wortmannin and LY294002, respectively, decreased but did not completely block uPA secretion. In contrast, inhibitors of phospholipase D or the p70S6 kinase pathway did not alter LPA-induced uPA secretion. Further, tyrosine phosphorylation and functional Src were required for optimal uPA secretion. Finally, LPA induces uPA secretion from ovarian cancer cells predominantly through the LPA2 receptor, with LPA3 contributing to this process. These results indicate that the p38(MAPK) signaling pathway is required for optimal LPA-dependent uPA secretion from ovarian cancer cells.


Asunto(s)
Lisofosfolípidos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Transporte Biológico , Diferenciación Celular , Línea Celular Tumoral , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas , Neoplasias Ováricas/metabolismo , Fosfolipasa D/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Transducción de Señal
12.
Clin Cancer Res ; 12(19): 5869-78, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17020995

RESUMEN

PURPOSE: We determined the effects of vorinostat [suberoylanilide hydroxamic acid (SAHA)] and/or dasatinib, a dual Abl/Src kinase (tyrosine kinase) inhibitor, on the cultured human (K562 and LAMA-84) or primary chronic myelogenous leukemia (CML) cells, as well as on the murine pro-B BaF3 cells with ectopic expression of the unmutated and kinase domain-mutant forms of Bcr-Abl. EXPERIMENTAL DESIGN: Following exposure to dasatinib and/or vorinostat, apoptosis, loss of clonogenic survival, as well as the activity and levels of Bcr-Abl and its downstream signaling proteins were determined. RESULTS: Treatment with dasatinib attenuated the levels of autophosphorylated Bcr-Abl, p-CrkL, phospho-signal transducer and activator of transcription 5 (p-STAT5), p-c-Src, and p-Lyn; inhibited the activity of Lyn and c-Src; and induced apoptosis of the cultured CML cells. Combined treatment of cultured human CML and BaF3 cells with vorinostat and dasatinib induced more apoptosis than either agent alone, as well as synergistically induced loss of clonogenic survival, which was associated with greater depletion of Bcr-Abl, p-CrkL, and p-STAT5 levels. Cotreatment with dasatinib and vorinostat also attenuated the levels of Bcr-AblE255K and Bcr-AblT315I and induced apoptosis of BaF3 cells with ectopic expression of the mutant forms of Bcr-Abl. Finally, cotreatment of the primary CML cells with vorinostat and dasatinib induced more loss of cell viability and depleted Bcr-Abl or Bcr-AblT315I, p-STAT5, and p-CrkL levels than either agent alone. CONCLUSIONS: As shown here, the preclinical in vitro activity of vorinostat and dasatinib against cultured and primary CML cells supports the in vivo testing of the combination in imatinib mesylate-sensitive and imatinib mesylate-resistant CML cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Ácidos Hidroxámicos/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Piperazinas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Benzamidas , Dasatinib , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/metabolismo , Inhibidores de Histona Desacetilasas , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Factor de Transcripción STAT5/metabolismo , Células Tumorales Cultivadas , Vorinostat , Familia-src Quinasas/metabolismo
13.
Nat Commun ; 6: 8752, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26658462

RESUMEN

Early cancers are avascular and hence, profoundly acidic. Pre-malignant cells must adapt to acidosis to thrive in this hostile microenvironment. Here, we investigate MCF-7 cells that are adapted to grow in acidic conditions using SILAC proteomics and we reveal a significant upregulation of lysosomal proteins. Prominent among these is LAMP2 that functions to protect lysosomal membranes from acid proteolysis. LAMP2 upregulation by acidosis is confirmed both in vitro and in vivo. Furthermore, we show that the depletion of LAMP2 is sufficient to increase acidosis-mediated toxicity. In breast cancer patient samples, there is a high correlation of LAMP2 mRNA and protein expression with progression. We also observe that LAMP2 is located at the plasma membrane in clinical samples and this redistribution is acid-induced in vitro. Our findings suggest a potential adaptive mechanism, wherein cells chronically exposed to an acidic environment translocate lysosomal proteins to their surface, thus protecting the plasmalemma from acid-induced hydrolysis.


Asunto(s)
Membrana Celular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Neoplasias/metabolismo , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Femenino , Humanos , Concentración de Iones de Hidrógeno , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Ratones , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Análisis por Matrices de Proteínas , Proteómica
14.
Front Oncol ; 3: 111, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23717812

RESUMEN

Despite the great progress that has been made in understanding cancer biology and the potential molecular targets for its treatment, the majority of drugs fail in the clinical trials. This may be attributed (at least in part) to the complexity of interstitial drug transport in the patient's body, which is hard to test experimentally. Similarly, recent advances in molecular imaging have led to the development of targeted biomarkers that can predict pharmacological responses to therapeutic interventions. However, both the drug and biomarker molecules need to access the tumor tissue and be taken up into individual cells in concentrations sufficient to exert the desired effect. To investigate the process of drug penetration at the mesoscopic level we developed a computational model of interstitial transport that incorporates the biophysical properties of the tumor tissue, including its architecture and interstitial fluid flow, as well as the properties of the agents. This model is based on the method of regularized Stokeslets to describe the fluid flow coupled with discrete diffusion-advection-reaction equations to model the dynamics of the drugs. Our results show that the tissue cellular porosity and density influence the depth of penetration in a non-linear way, with sparsely packed tissues being traveled through more slowly than the denser tissues. We demonstrate that irregularities in the cell spatial configurations result in the formation of interstitial corridors that are followed by agents leading to the emergence of tissue zones with less exposure to the drugs. We describe how the model can be integrated with in vivo experiments to test the extravasation and penetration of the targeted biomarkers through the tumor tissue. A better understanding of tissue- or compound-specific factors that limit the penetration through the tumors is important for non-invasive diagnoses, chemotherapy, the monitoring of treatment responses, and the detection of tumor recurrence.

15.
Cancer Res ; 73(5): 1524-35, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23288510

RESUMEN

The pH of solid tumors is acidic due to increased fermentative metabolism and poor perfusion. It has been hypothesized that acid pH promotes local invasive growth and metastasis. The hypothesis that acid mediates invasion proposes that H(+) diffuses from the proximal tumor microenvironment into adjacent normal tissues where it causes tissue remodeling that permits local invasion. In the current work, tumor invasion and peritumoral pH were monitored over time using intravital microscopy. In every case, the peritumoral pH was acidic and heterogeneous and the regions of highest tumor invasion corresponded to areas of lowest pH. Tumor invasion did not occur into regions with normal or near-normal extracellular pH. Immunohistochemical analyses revealed that cells in the invasive edges expressed the glucose transporter-1 and the sodium-hydrogen exchanger-1, both of which were associated with peritumoral acidosis. In support of the functional importance of our findings, oral administration of sodium bicarbonate was sufficient to increase peritumoral pH and inhibit tumor growth and local invasion in a preclinical model, supporting the acid-mediated invasion hypothesis. Cancer Res; 73(5); 1524-35. ©2012 AACR.


Asunto(s)
Concentración de Iones de Hidrógeno , Invasividad Neoplásica , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral , Animales , Medicamentos Herbarios Chinos , Humanos , Ratones , Ratones SCID , Modelos Biológicos , Bicarbonato de Sodio/farmacología
16.
Sci Rep ; 3: 1449, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23618955

RESUMEN

To investigate the transition from non-cancerous to metastatic from a physical sciences perspective, the Physical Sciences-Oncology Centers (PS-OC) Network performed molecular and biophysical comparative studies of the non-tumorigenic MCF-10A and metastatic MDA-MB-231 breast epithelial cell lines, commonly used as models of cancer metastasis. Experiments were performed in 20 laboratories from 12 PS-OCs. Each laboratory was supplied with identical aliquots and common reagents and culture protocols. Analyses of these measurements revealed dramatic differences in their mechanics, migration, adhesion, oxygen response, and proteomic profiles. Model-based multi-omics approaches identified key differences between these cells' regulatory networks involved in morphology and survival. These results provide a multifaceted description of cellular parameters of two widely used cell lines and demonstrate the value of the PS-OC Network approach for integration of diverse experimental observations to elucidate the phenotypes associated with cancer metastasis.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Modelos Biológicos , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/fisiopatología , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Movimiento Celular , Tamaño de la Célula , Supervivencia Celular , Simulación por Computador , Humanos
17.
PLoS One ; 7(5): e36894, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22623963

RESUMEN

Mammalian cell function requires timely and accurate transmission of information from the cell membrane (CM) to the nucleus (N). These pathways have been intensively investigated and many critical components and interactions have been identified. However, the physical forces that control movement of these proteins have received scant attention. Thus, transduction pathways are typically presented schematically with little regard to spatial constraints that might affect the underlying dynamics necessary for protein-protein interactions and molecular movement from the CM to the N. We propose messenger protein localization and movements are highly regulated and governed by Coulomb interactions between: 1. A recently discovered, radially directed E-field from the NM into the CM and 2. Net protein charge determined by its isoelectric point, phosphorylation state, and the cytosolic pH. These interactions, which are widely applied in elecrophoresis, provide a previously unknown mechanism for localization of messenger proteins within the cytoplasm as well as rapid shuttling between the CM and N. Here we show these dynamics optimize the speed, accuracy and efficiency of transduction pathways even allowing measurement of the location and timing of ligand binding at the CM--previously unknown components of intracellular information flow that are, nevertheless, likely necessary for detecting spatial gradients and temporal fluctuations in ligand concentrations within the environment. The model has been applied to the RAF-MEK-ERK pathway and scaffolding protein KSR1 using computer simulations and in-vitro experiments. The computer simulations predicted distinct distributions of phosphorylated and unphosphorylated components of this transduction pathway which were experimentally confirmed in normal breast epithelial cells (HMEC).


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Campos Electromagnéticos , Modelos Biológicos , Proteínas/metabolismo , Transducción de Señal/fisiología , Animales , Simulación por Computador , Punto Isoeléctrico , Mamíferos , Fosforilación , Transporte de Proteínas/fisiología
18.
J Cancer Sci Ther ; Suppl 1(4)2011 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-24032073

RESUMEN

BACKGROUND: Malignant tumor cells typically metabolize glucose anaerobically to lactic acid even under normal oxygen tension, a phenomenon called aerobic glycolysis or the Warburg effect. This results in increased acid production and the acidification of the extracellular microenvironment in solid tumors. H+ ions tend to flow along concentration gradients into peritumoral normal tissue causing extracellular matrix degradation and increased tumor cell motility thus promoting invasion and metastasis. We have shown that reducing this acidity with sodium bicarbonate buffer decreases the metastatic fitness of circulating tumor cells in prostate cancer and other cancer models. Mathematical models of the tumor-host dynamics predicted that buffers with a pka around 7 will be more effective in reducing intra- and peri-tumoral acidosis and, thus, and possibly more effective in inhibiting tumor metastasis than sodium bicarbonate which has a pKa around 6. Here we test this prediction the efficacy of free base lysine; a non-bicarbonate/non-volatile buffer with a higher pKa (~10), on prostate tumor metastases model. METHODS: Oxygen consumption and acid production rate of PC3M prostate cancer cells and normal prostate cells were determined using the Seahorse Extracellular Flux (XF-96) analyzer. In vivo effect of 200 mM lysine started four days prior to inoculation on inhibition of metastasis was examined in PC3M-LUC-C6 prostate cancer model using SCID mice. Metastases were followed by bioluminescence imaging. RESULTS: PC3M prostate cancer cells are highly acidic in comparison to a normal prostate cell line indicating that reduction of intra- and perit-tumoral acidosis should inhibit metastases formation. In vivo administration of 200 mM free base lysine increased survival and reduced metastasis. CONCLUSION: PC3M prostate cancer cells are highly glycolytic and produce large amounts of acid when compared to normal prostate cells. Administration of non-volatile buffer decreased growth of metastases and improved survival indicating acidity plays a significant role in growth and invasion in-vivo.

19.
PLoS One ; 6(4): e14790, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21533234

RESUMEN

We investigate a three-dimensional multiscale model of vascular tumour growth, which couples blood flow, angiogenesis, vascular remodelling, nutrient/growth factor transport, movement of, and interactions between, normal and tumour cells, and nutrient-dependent cell cycle dynamics within each cell. In particular, we determine how the domain size, aspect ratio and initial vascular network influence the tumour's growth dynamics and its long-time composition. We establish whether it is possible to extrapolate simulation results obtained for small domains to larger ones, by constructing a large simulation domain from a number of identical subdomains, each subsystem initially comprising two parallel parent vessels, with associated cells and diffusible substances. We find that the subsystem is not representative of the full domain and conclude that, for this initial vessel geometry, interactions between adjacent subsystems contribute to the overall growth dynamics. We then show that extrapolation of results from a small subdomain to a larger domain can only be made if the subdomain is sufficiently large and is initialised with a sufficiently complex vascular network. Motivated by these results, we perform simulations to investigate the tumour's response to therapy and show that the probability of tumour elimination in a larger domain can be extrapolated from simulation results on a smaller domain. Finally, we demonstrate how our model may be combined with experimental data, to predict the spatio-temporal evolution of a vascular tumour.


Asunto(s)
Modelos Anatómicos , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Humanos , Neoplasias/patología , Neovascularización Patológica/metabolismo , Procesos Estocásticos , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Horiz. enferm ; 26(1): 21-28, 2015. tab
Artículo en Español | LILACS, BDENF | ID: biblio-1178054

RESUMEN

INTRODUCCIÓN: los diseños experimentales (DE) podrían ser el futuro de las intervenciones de enfermería, dado que se realizarían manipulando intencionalmente una variable en un grupo experimental versus un control, para mostrar la efectividad del cuidado enfermero. Sin embargo, en la literatura, los DE son minoritarios, es posible que se deba a la falta de financiamiento o preparación metodológica en la formación de estos profesionales. OBJETIVO: Determinar si los artículos publicados en enfermería con diseño experimental cumplen con los requisitos de rigurosidad metodológica. MATERIAL Y MÉTODOS: Diseño retrospectivo y transversal. Se analizaron dos revistas nacionales y tres internacionales, indizadas en LILACS, CINAHL, CUIDEN, PubMed e ISI Web of Science, publicadas del 2008 al 2012. RESULTADOS: Se revisaron 400 artículos de los cuales solo 35 cumplieron con los criterios de selección. En aspectos generales, el 34.3% son escritos por Maestros en ciencias, 28.6% muestra co- elaboración multidisciplinaria, el 31.4 % utilizan conceptos teóricos de enfermería, la temática clínica abarcó 45.7% y el ámbito educativo 51.4%. Respecto a los diseños, el 48. 6% de los artículos son cuasi-experimentales, 22.9% preexperimentales y el 22.9% ensayos clínicos. DISCUSIÓN Y CONCLUSIONES: realizar investigaciones experimentales en enfermería podría reforzar los PLACE, la taxonomía NANDA, NIC, NOC y los modelos teóricos. Esto podría mostrar bajo evidencia científica y rigurosidad metodológica que las intervenciones de enfermería son efectivas. La calidad de la mayoría de los artículos revisados podría mejorar, dado que no cuentan con potencia estadística, tamaño del efecto y el cálculo de la muestra no es claro.


INTRODUCTION. Experimental designs (ED) could be the nursing interventions future, since they would be performed intentionally manipulating a variable in an experimental group vs a control one, aiming to show nurse care effectiveness. However, in the literature, ED are a minority; it is possible this is due the lack of financial fostering or methodological preparation of these professional's upbringing. OBJECTIVE. Determine if the published nursing papers with an experimental design fulfill the methodological rigor requisites. MATERIAL AND METHODS. Retrospective and transversal design. Two national and three international journals published from 2008 to 2012 and indexed in LILACS, CINAHL, CUIDEN, PubMed, and Web of Science were analyzed. RESULTS. Four hundred articles were reviewed, and only 35 out of them fulfill the selection criteria. In general aspects, 34.3% are written by science teachers, 28.6% shows a multidisciplinary collaboration, 31.4% use nursing theoretical concepts, clinical thematic comprised 45.7%, and 51.4% cover the educational scope. Regarding designs, 48.6% are quasi-experimental, 22.9% pre-experimental, and 22.9% are clinical essays. DISCUSSION AND CONCLUSIONS. Performing nursing experimental research could reinforce the PLACE, NANDA taxonomy, NIC, NOC, and the theoretical models. Then so, it could be shown under scientific evidence and methodological rigor that nursing interventions are effective. However, most reviewed articles quality could be enhanced, since they lack statistical potency, effect size, and the sample calculation is unclear.


Asunto(s)
Humanos , Masculino , Femenino , Proyectos de Investigación/normas , Educación en Enfermería , Investigación Científica y Desarrollo Tecnológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA