Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
J Hepatol ; 64(1): 128-34, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26341825

RESUMEN

BACKGROUND & AIMS: NKp46(+) cells are major effector cells in the pathogenesis of hepatic ischemia reperfusion injury (IRI). Nevertheless, the precise role of unconventional subsets like the IL-22-producing NKp46(+) cells (NK22) remains unknown. The purpose of this study was to examine the role of NK22 cells in IRI in transplantation, particularly with respect to regulation by the transcription factor ROR-gamma-t (RORγt). METHODS: To explore the role of NK22 cells in IRI in the absence of adaptive immunity, B6.RORγt-(gfp/wt)-reporter and B6.RORγt-(gfp/gfp)-knockout (KO) mice on a Rag KO background underwent 90min partial warm ischemia, followed by 24h of reperfusion. RESULTS: Rag KO mice that possess fully functional NKp46(+) cells, and Rag-common-γ-chain-double-KO (Rag-γc-DKO) mice that lack T, B and NKp46(+) cells, were used as controls. We found that Rag-γc-DKO mice lacking NK22 cells show more severe levels of hepatocellular damage (GPT, histological injury) when compared to both Rag-RORγt-reporter and Rag KO mice that possess NK22 cells. Importantly, Rag-RORγt-reporter and Rag KO mice undergoing IRI expressed high protein levels of both IL-22 and GFP (RORγt), suggesting a protective role for RORγt(+) NK22 cells in IRI. Therefore, we tested the hypothesis that RORγt critically protects from IRI through the induction of hepatic NK22 cells by studying Rag-Rorγt-DKO mice under IRI conditions. We found that the lack of RORγt(+) NK22 cells in Rag-Rorγt-DKO mice significantly enhanced IR-induced hepatocellular injury, a phenotype that could be reversed upon adoptive transfer of Rag-Rorγt-reporter NK22 cells into DKO mice. CONCLUSIONS: RORγt(+) NK22 cells play an important protective role in IRI in mice.


Asunto(s)
Antígenos Ly/fisiología , Interleucinas/biosíntesis , Hígado/irrigación sanguínea , Receptor 1 Gatillante de la Citotoxidad Natural/fisiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Daño por Reperfusión/prevención & control , Animales , Antígenos Ly/análisis , Proteínas de Homeodominio/fisiología , Interferón gamma/biosíntesis , Células Asesinas Naturales/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor 1 Gatillante de la Citotoxidad Natural/análisis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/análisis , Daño por Reperfusión/inmunología , Interleucina-22
2.
J Immunol ; 192(4): 1954-61, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24415778

RESUMEN

We studied the developmental and functional mechanisms behind NK cell-mediated antitumor responses against metastatic colorectal carcinoma (CRC) in mice. In particular, we focused on investigating the significance of T-box transcription factors and the immunotherapeutic relevance of IL-15 in the development and function of tumor-reactive NK cells. Pulmonary CRC metastases were experimentally seeded via an adoptive i.v. transfer of luciferase-expressing CT26 CRC cells that form viewable masses via an in vivo imaging device; genetically deficient mice were used to dissect the antitumor effects of developmentally different NK cell subsets. IL-15 precomplexed to IL-15 receptor-α was used in immunotherapy experiments. We found that mice deficient for the T-box transcription factor T-bet lack terminally differentiated antitumor CD27(low)KLRG1(+) NK cells, leading to a terminal course of rapid-onset pulmonary CRC metastases. The importance of this NK cell subset for effective antitumor immunity was shown by adoptively transferring purified CD27(low)KLRG1(+) NK cells into T-bet-deficient mice and, thereby, restoring immunity against lung metastasis formation. Importantly, immunity to metastasis formation could also be restored in T-bet-deficient recipients by treating mice with IL-15 precomplexed to IL-15 receptor-α, which induced the development of eomesodermin(+)KLRG1(+) NK cells from existing NK cell populations. Thus, contingent upon their T-bet-dependent development and activation status, NK cells can control metastatic CRC in mice, which is highly relevant for the development of immunotherapeutic approaches in the clinic.


Asunto(s)
Neoplasias Colorrectales/patología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/secundario , Receptores Inmunológicos/metabolismo , Proteínas de Dominio T Box/genética , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Proteínas de Homeodominio/genética , Inmunoterapia , Interferón gamma/genética , Interleucina-15/metabolismo , Células Asesinas Naturales/citología , Lectinas Tipo C , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Perforina , Proteínas Citotóxicas Formadoras de Poros/genética , Receptores de Interleucina-15/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
3.
J Immunol ; 193(5): 2600-8, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25057005

RESUMEN

Administering immunoregulatory cells to patients as medicinal agents is a potentially revolutionary approach to the treatment of immunologically mediated diseases. Presently, there are no satisfactory, clinically applicable methods of tracking human cells in patients with adequate spatial resolution and target cell specificity over a sufficient period of time. Laser ablation-inductively coupled plasma mass spectrometry (LA-ICP-MS) represents a potential solution to the problem of detecting very rare cells in tissues. In this article, this exquisitely sensitive technique is applied to the tracking of gold-labeled human regulatory macrophages (Mregs) in immunodeficient mice. Optimal conditions for labeling Mregs with 50-nm gold particles were investigated by exposing Mregs in culture to variable concentrations of label: Mregs incubated with 3.5 × 10(9) particles/ml for 1 h incorporated an average of 3.39 × 10(8) Au atoms/cell without loss of cell viability. Analysis of single, gold-labeled Mregs by LA-ICP-MS registered an average of 1.9 × 10(5) counts/cell. Under these conditions, 100% labeling efficiency was achieved, and label was retained by Mregs for ≥36 h. Gold-labeled Mregs adhered to glass surfaces; after 24 h of culture, it was possible to colabel these cells with human-specific (154)Sm-tagged anti-HLA-DR or (174)Yb-tagged anti-CD45 mAbs. Following injection into immunodeficient mice, signals from gold-labeled human Mregs could be detected in mouse lung, liver, and spleen for at least 7 d by solution-based inductively coupled plasma mass spectrometry and LA-ICP-MS. These promising results indicate that LA-ICP-MS tissue imaging has great potential as an analytical technique in immunology.


Asunto(s)
Oro/farmacología , Rayos Láser , Antígenos Comunes de Leucocito/inmunología , Pulmón , Espectrometría de Masas/instrumentación , Monocitos , Animales , Anticuerpos Monoclonales de Origen Murino , Xenoinjertos , Humanos , Antígenos Comunes de Leucocito/química , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos NOD , Monocitos/citología , Monocitos/inmunología , Monocitos/trasplante
4.
J Immunol ; 191(1): 480-7, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23740948

RESUMEN

An emerging body of evidence suggests a pivotal role of CD3(+) T cells in mediating early ischemia reperfusion injury (IRI). However, the precise phenotype of T cells involved and the mechanisms underlying such T cell-mediated immune responses in IRI, as well as their clinical relevance, are poorly understood. In this study, we investigated early immunological events in a model of partial warm hepatic IRI in genetically targeted mice to study the precise pathomechanistic role of RORγt(+) T cells. We found that unconventional CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells are the major RORγt-expressing effector cells in hepatic IRI that play a mechanistic role by being the main source of IRI-mediating IL-17A. We further show that unconventional IRI-mediating T cells are contingent on RORγt, as highlighted by the fact that a genetic deficiency for RORγt, or its therapeutic antagonization via digoxin, is protective against hepatic IRI. Therefore, identification of CD27(-)γδTCR(+) and CD4(-)CD8(-) double-negative T cells as the major source of IL-17A via RORγt in hepatic IRI opens new therapeutic options to improve liver transplantation outcomes.


Asunto(s)
Hepatitis Animal/inmunología , Hepatitis Animal/patología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Daño por Reperfusión/inmunología , Daño por Reperfusión/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Animales , Modelos Animales de Enfermedad , Genes Reporteros , Hepatitis Animal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/biosíntesis , Daño por Reperfusión/metabolismo , Subgrupos de Linfocitos T/metabolismo , Factores de Tiempo
5.
J Immunol ; 191(8): 4440-6, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24058178

RESUMEN

Although Th1, Th2, and Th17 cells are thought to be major effector cells in adaptive alloimmune responses, their respective contribution to allograft rejection remains unclear. To precisely address this, we used mice genetically modified for the Th1 and Th17 hallmark transcription factors T-bet and RORγt, respectively, which allowed us to study the alloreactive role of each subset in an experimental transplant setting. We found that in a fully mismatched heterotopic mouse heart transplantation model, T cells deficient for T-bet (prone to Th17 differentiation) versus RORγt (prone to Th1 differentiation) rejected allografts at a more accelerated rate, indicating a predominance of Th17- over Th1-driven alloimmunity. Importantly, T cells doubly deficient for both T-bet and RORγt differentiated into alloreactive GATA-3-expressing Th2 cells, which promptly induced allograft rejection characterized by a Th2-type intragraft expression profile and eosinophilic infiltration. Mechanistically, Th2-mediated allograft rejection was contingent on IL-4, as its neutralization significantly prolonged allograft survival by reducing intragraft expression of Th2 effector molecules and eosinophilic allograft infiltration. Moreover, under IL-4 neutralizing conditions, alloreactive double-deficient T cells upregulated Eomesodermin (Eomes) and IFN-γ, but not GATA-3. Thus, in the absence of T-bet and RORγt, Eomes may salvage Th1-mediated alloimmunity that underlies IL-4 neutralization-resistant allograft rejection. We summarize that, whereas Th17 cells predictably promote allograft rejection, IL-4-producing GATA-3(+) Th2 cells, which are generally thought to protect allogeneic transplants, may actually be potent facilitators of organ transplant rejection in the absence of T-bet and RORγt. Moreover, Eomes may rescue Th1-mediated allograft rejection in the absence of IL-4, T-bet, and RORγt.


Asunto(s)
Aloinjertos/inmunología , Rechazo de Injerto/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular , Eosinófilos/inmunología , Factor de Transcripción GATA3/biosíntesis , Trasplante de Corazón/efectos adversos , Interferón gamma/biosíntesis , Interleucina-4/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/deficiencia , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Proteínas de Dominio T Box/biosíntesis , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética
6.
Artif Organs ; 39(9): 782-7, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25845704

RESUMEN

Thrombosis inside the membrane oxygenator (MO) is a critical complication during venovenous extracorporeal membrane oxygenation (ECMO). The aim of this study was to prove if thrombotic clots manifest within the MO when D-dimer levels are elevated over a long-term period. Heparin-coated polymethylpentene MOs (n = 13) were exchanged due to high plasma D-dimer levels. Clot volume was calculated using multidetector computed tomography (MDCT). Coagulation parameters and MO function were analyzed before and after MO exchange. Before MO exchange, D-dimer levels increased significantly in each patient (11.5 [6.5-15.5] mg/L to 35.0 [34-35] mg/L, P ≤ 0.001). High levels of D-dimers were tolerated for 1 to 6 days. Additionally, fibrinogen concentration (n = 8) and platelet count decreased (n = 8). Within 48 h after exchange, D-dimer levels decreased significantly (n = 11, 12 [8-16] mg/L, P = 0.004). Fibrinogen concentration and platelet counts increased. Clots were found in all MOs in the inlet part of the device. Clot volume (16-106 cm(3) ) did not correlate with MO support time but increased significantly when high D-dimer levels were accepted for >2 days. An increase or high levels of D-dimers in absence of other explaining pathology during ECMO therapy reflected coagulation activity within the MO. Evidence of clots within the MO at high D-dimer levels and decrease after exchange underline the relevance of D-dimer testing during ECMO treatment. Besides, surveillance of MOs during ongoing ECMO therapy will help to predict clot formation, and to avoid system-induced coagulation disorders as well as critical situations.


Asunto(s)
Coagulación Sanguínea , Oxigenación por Membrana Extracorpórea/efectos adversos , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Oxigenadores de Membrana/efectos adversos , Trombosis/diagnóstico , Trombosis/etiología , Diseño de Equipo , Oxigenación por Membrana Extracorpórea/instrumentación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Trombosis/patología
7.
J Immunol ; 188(10): 4931-9, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22504651

RESUMEN

ß-Defensins are known for their antimicrobial activity and belong to the molecular barrier of the innate immune system against invading pathogens. In addition, it has been shown that some members of the ß-defensin superfamily have the capacity to promote local innate inflammatory and systemic adaptive immune responses, mediated in part by the interaction with CCR6. We found that mouse ß-defensin 14 (mBD14, Defb14), a newly identified member of the mouse ß-defensin superfamily, is expressed in mouse fibrosarcoma tumor tissue. Tumor cells overexpressing mBD14 demonstrated enhanced solid tumor growth in syngeneic C57BL/6 mice concomitant with increased vascularization of these tumors. Furthermore, mBD14-overexpressing tumors demonstrated increased expression of proangiogenic MIP-2 (CXCL2) ex vivo. In contrast, vascular endothelial growth factor expression was not affected. Cellular analysis of tumor-infiltrating leukocytes revealed a significant increase of CCR6(+) B220(+) lymphocytes in solid tumors derived from mBD14-overexpressing tumor cells. Enhanced tumor growth of mBD14-overexpressing fibrosarcomas was abolished in CCR6-deficient mice, which was paralleled by decreased infiltration of CCR6(+) B220(+) lymphocytes, indicating the requirement of CCR6 expression on host cells. Previously, the interaction of activated, LTαß(+), lymphocytes with lymphotoxin ß-receptor-expressing fibrosarcoma tumor cells has been identified as a new CXCL2-dependent proangiogenic pathway. Coexpression of a soluble lymphotoxin ß-receptor:Ig fusion protein, an inhibitor of CXCL2-dependent angiogenesis, in mBD14-overexpressing fibrosarcoma tumor cells abolished enhanced solid tumor growth. Thus, we conclude that mBD14 expression by tumor-infiltrating host cells results in the chemoattraction of CCR6(+) B220(+) lymphocytes, which in turn initiates a proangiogenic pathway leading to enhanced angiogenesis and organized tumor tissue development.


Asunto(s)
Fibrosarcoma/inmunología , Fibrosarcoma/patología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Receptores CCR6/fisiología , beta-Defensinas/fisiología , Animales , Femenino , Fibrosarcoma/metabolismo , Antígenos Comunes de Leucocito/biosíntesis , Antígenos Comunes de Leucocito/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Receptores CCR6/biosíntesis , Células Tumorales Cultivadas , beta-Defensinas/biosíntesis
8.
J Immunother Cancer ; 11(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37880183

RESUMEN

BACKGROUND & AIMS: Intratumoral lactate accumulation and acidosis impair T-cell function and antitumor immunity. Interestingly, expression of the lactate transporter monocarboxylate transporter (MCT) 4, but not MCT1, turned out to be prognostic for the survival of patients with rectal cancer, indicating that single MCT4 blockade might be a promising strategy to overcome glycolysis-related therapy resistance. METHODS: To determine whether blockade of MCT4 alone is sufficient to improve the efficacy of immune checkpoint blockade (ICB) therapy, we examined the effects of the selective MCT1 inhibitor AZD3965 and a novel MCT4 inhibitor in a colorectal carcinoma (CRC) tumor spheroid model co-cultured with blood leukocytes in vitro and the MC38 murine CRC model in vivo in combination with an antibody against programmed cell death ligand-1(PD-L1). RESULTS: Inhibition of MCT4 was sufficient to reduce lactate efflux in three-dimensional (3D) CRC spheroids but not in two-dimensional cell-cultures. Co-administration of the MCT4 inhibitor and ICB augmented immune cell infiltration, T-cell function and decreased CRC spheroid viability in a 3D co-culture model of human CRC spheroids with blood leukocytes. Accordingly, combination of MCT4 and ICB increased intratumoral pH, improved leukocyte infiltration and T-cell activation, delayed tumor growth, and prolonged survival in vivo. MCT1 inhibition exerted no further beneficial impact. CONCLUSIONS: These findings demonstrate that single MCT4 inhibition represents a novel therapeutic approach to reverse lactic-acid driven immunosuppression and might be suitable to improve ICB efficacy.


Asunto(s)
Neoplasias Colorrectales , Inhibidores de Puntos de Control Inmunológico , Animales , Humanos , Ratones , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Glucólisis , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores
9.
Biochim Biophys Acta ; 1803(4): 435-42, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20116405

RESUMEN

Inhibition of mTORC1 with the mTOR inhibitor rapamycin may lead to an induction of Akt phosphorylation in cancer cells via mTORC2 activation. Using gastric and pancreatic cancer cells, we further investigated this paradoxical signaling response and found that rapamycin additionally up-regulates both IGF-IR and Her2 expression. Using RNAi for down-regulating RICTOR, this induction of receptor kinase expression was identified to be mediated via an mTORC2-induced Akt activation. Moreover, mTORC2 inhibition reduced the phosphorylation of GSK-3 and NF-kappaB, and significantly impaired cancer cell motility. In conclusion, inhibition of mTORC2 may abrogate unfavorable signaling effects of mTOR inhibitors, hence providing a novel rationale for therapy.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 1/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Transcripción/fisiología , Western Blotting , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , FN-kappa B/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Proteína Asociada al mTOR Insensible a la Rapamicina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Sirolimus/farmacología , Neoplasias Gástricas/patología
10.
J Exp Med ; 217(1)2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31658985

RESUMEN

Cancer fatalities result from metastatic dissemination and therapy resistance, both processes that depend on signals from the tumor microenvironment. To identify how invasion and resistance programs cooperate, we used intravital microscopy of orthotopic sarcoma and melanoma xenografts. We demonstrate that these tumors invade collectively and that, specifically, cells within the invasion zone acquire increased resistance to radiotherapy, rapidly normalize DNA damage, and preferentially survive. Using a candidate-based approach to identify effectors of invasion-associated resistance, we targeted ß1 and αVß3/ß5 integrins, essential extracellular matrix receptors in mesenchymal tumors, which mediate cancer progression and resistance. Combining radiotherapy with ß1 or αV integrin monotargeting in invading tumors led to relapse and metastasis in 40-60% of the cohort, in line with recently failed clinical trials individually targeting integrins. However, when combined, anti-ß1/αV integrin dual targeting achieved relapse-free radiosensitization and prevented metastatic escape. Collectively, invading cancer cells thus withstand radiotherapy and DNA damage by ß1/αVß3/ß5 integrin cross-talk, but efficient radiosensitization can be achieved by multiple integrin targeting.


Asunto(s)
Adhesión Celular/fisiología , Integrinas/metabolismo , Invasividad Neoplásica/patología , Neoplasias/metabolismo , Neoplasias/patología , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Daño del ADN/fisiología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia/patología , Microambiente Tumoral/fisiología
11.
Mol Cancer Ther ; 6(11): 2868-78, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025273

RESUMEN

We recently showed that inhibition of heat shock protein 90 (Hsp90) decreases tumor growth and angiogenesis in gastric cancer through interference with oncogenic signaling pathways. However, controversy still exists about the antimetastatic potential of Hsp90 inhibitors. Moreover, in vitro studies suggested that blocking Hsp90 could overcome p53-mediated resistance of cancer cells to oxaliplatin. We therefore hypothesized that blocking oncogenic signaling with a Hsp90 inhibitor would impair metastatic behavior of colon cancer cells and also improve the efficacy of oxaliplatin in vivo. Human colon cancer cells (HCT116, HT29, and SW620) and the Hsp90 inhibitor 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17-DMAG) were used for experiments. In vitro, 17-DMAG substantially inhibited phosphorylation of epidermal growth factor receptor, c-Met, and focal adhesion kinase, overall resulting in a significant decrease in cancer cell invasiveness. Importantly, 17-DMAG led to an up-regulation of the transcription factor activating transcription factor-3, a tumor suppressor and antimetastatic factor, on mRNA and protein levels. In a cell death ELISA, 17-DMAG markedly induced apoptosis in both p53-wt and p53-deficient cells. In vivo, 17-DMAG significantly reduced tumor growth and vascularization. Furthermore, blocking Hsp90 reduced hepatic tumor burden and metastatic nodules in an experimental model of hepatic colon cancer growth. Importantly, combining oxaliplatin with 17-DMAG in vivo significantly improved growth inhibitory and proapoptotic effects on p53-deficient cells, compared with either substance alone. In conclusion, inhibition of Hsp90 abrogates the invasive properties of colon cancer cells and modulates the expression of the antimetastatic factor activating transcription factor-3. Hence, targeting Hsp90 could prove valuable for treatment of advanced colorectal cancer by effectively inhibiting colon cancer growth and hepatic metastasis and improving the efficacy of oxaliplatin.


Asunto(s)
Neoplasias del Colon/patología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Hepáticas/secundario , Compuestos Organoplatinos/farmacología , Proteína p53 Supresora de Tumor/deficiencia , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Receptores ErbB/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica/prevención & control , Compuestos Organoplatinos/uso terapéutico , Oxaliplatino , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Res ; 78(10): 2513-2523, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29510993

RESUMEN

Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy.Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. Cancer Res; 78(10); 2513-23. ©2018 AACR.


Asunto(s)
Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/metabolismo , Proliferación Celular/efectos de los fármacos , Ácido Cítrico/metabolismo , Gluconatos/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Ácidos Grasos/biosíntesis , Glucólisis/fisiología , Humanos , Masculino , Ratones , Transportadores de Anión Orgánico , Próstata/citología , Próstata/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética
13.
Transplantation ; 83(5): 607-14, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17353782

RESUMEN

BACKGROUND: Identifying immunosuppressive agents with antitumor effects could help address the problem of posttransplant malignancy. Here we tested for potential inhibitory effects of mycophenolate mofetil (MMF) on tumors in vitro and in vivo. METHODS: Mouse CT26 colon adenocarcinoma, B16 melanoma, and human TMK1 gastric adenocarcinoma cells were tested for in vitro growth in the presence of MMF. In vitro angiogenesis was tested with a rat aortic-ring assay. Tumor cells were implanted into dorsal skinfold chambers (DSFC) to assess in vivo angiogenesis. Subcutaneous tumor growth was determined in mice receiving MMF. RESULTS: MMF caused a dose-dependent reduction in tumor cell numbers in vitro, starting between 0.1 to 1 microM. Vessel sprouting from aortic rings was markedly blocked by similar concentrations of MMF. In vivo, however, DSFC results showed a marginal reduction in CT26 tumor angiogenesis with MMF doses of 40 or 80 mg/kg/day, although MMF did inhibit TMK1 vascularity. Moreover, 80 mg/kg/day MMF did not reduce subcutaneous CT26 tumor volumes, but did slightly inhibit B16 and TMK1 expansion. Interestingly, the mycophenolic acid (MPA) blood level 2 hr after 80 mg/kg/day MMF bolus dosing was near 14 mg/L, but decreased dramatically thereafter, suggesting a drug availability issue. Indeed, intermittent 2-hr MMF pulses in tumor-cell cultures substantially reduced the antiproliferative effect of MPA. CONCLUSION: MMF strongly inhibits tumor cell growth and angiogenesis in vitro, but only marginally inhibits tumors in vivo. These contrasting results may relate to drug availability, where intermittent exposure of tumor cells to immunosuppressive doses of MMF substantially reduce its potential antitumor effects.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Ácido Micofenólico/análogos & derivados , Neovascularización Patológica/prevención & control , Adenocarcinoma , Inhibidores de la Angiogénesis/farmacocinética , Animales , Antineoplásicos/farmacocinética , Aorta , Disponibilidad Biológica , Línea Celular Tumoral , Neoplasias del Colon , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiología , Ácido Micofenólico/farmacocinética , Ácido Micofenólico/farmacología , Neoplasias Gástricas , Venas Umbilicales
14.
Front Immunol ; 8: 248, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28337200

RESUMEN

Cytotoxic T lymphocytes and NK cells play an important role in eliminating malignant tumor cells and the number and activity of tumor-infiltrating T cells represent a good marker for tumor prognosis. Based on these findings, immunotherapy, e.g., checkpoint blockade, has received considerable attention during the last couple of years. However, for the majority of patients, immune control of their tumors is gray theory as malignant cells use effective mechanisms to outsmart the immune system. Increasing evidence suggests that changes in tumor metabolism not only ensure an effective energy supply and generation of building blocks for tumor growth but also contribute to inhibition of the antitumor response. Immunosuppression in the tumor microenvironment is often based on the mutual metabolic requirements of immune cells and tumor cells. Cytotoxic T and NK cell activation leads to an increased demand for glucose and amino acids, a well-known feature shown by tumor cells. These close metabolic interdependencies result in metabolic competition, limiting the proliferation, and effector functions of tumor-specific immune cells. Moreover, not only nutrient restriction but also tumor-driven shifts in metabolite abundance and accumulation of metabolic waste products (e.g., lactate) lead to local immunosuppression, thereby facilitating tumor progression and metastasis. In this review, we describe the metabolic interplay between immune cells and tumor cells and discuss tumor cell metabolism as a target structure for cancer therapy. Metabolic (re)education of tumor cells is not only an approach to kill tumor cells directly but could overcome metabolic immunosuppression in the tumor microenvironment and thereby facilitate immunotherapy.

15.
Transplantation ; 82(6): 741-8, 2006 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-17006319

RESUMEN

BACKGROUND: Development of cancer in transplant recipients may be influenced by different immunosuppressive agents. Recent publications suggest that rapamycin (RAPA), or possibly mycophenolate mofetil (MMF), may reduce established tumor growth; however, experimental data is lacking for de novo cancer prevention. METHODS: We tested the effects of long-term immunosuppression on spontaneous tumor formation in p53 knock-out mice. Mice received no treatment, or were given RAPA, MMF, or cyclosporine (CsA) starting on week nine after birth, with the experimental endpoint being week 29. RESULTS: All (9/9) untreated mice developed clinically evident tumors before week 26, as confirmed by histology (6 lymphomas, 2 sarcomas, 1 lymphoma+sarcoma). All CsA-treated mice (9/9) also developed clinical tumors before the endpoint (7 lymphomas, 1 sarcoma, 1 lymphoma+sarcoma). With MMF, 7/10 mice showed clinical evidence of tumor before the experimental endpoint (4 lymphomas, 2 sarcomas, 1 lymphoma+sarcoma), however, histologic tissue analysis revealed that the remaining three mice had subclinical cancer (3 lymphomas). In contrast, RAPA treatment resulted in only three mice with clinical tumors (all lymphomas), with histology revealing subclinical lymphomas in three additional mice, but no evidence of cancer in four animals. Statistically, cancer development was decreased with RAPA treatment (P=0.002), but was not affected with either MMF or CsA (P>0.10). CONCLUSION: These experiments are the first to show immunosuppression under RAPA can reduce spontaneous de novo cancer associated with p53 mutations. Although neither CsA nor MMF treatment affects p53-associated tumor incidence, MMF may have some tendency to reduce clinical tumor appearance.


Asunto(s)
Genes p53 , Inmunosupresores/farmacología , Neoplasias Experimentales/genética , Animales , Anticarcinógenos/farmacología , Ciclosporina/farmacología , Supervivencia sin Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/prevención & control , Sirolimus/farmacología
16.
Transplantation ; 81(11): 1589-95, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16770249

RESUMEN

BACKGROUND: Recent reports suggest that mesenchymal stem cells (MSCs) have immunomodulatory properties. Mesenchymal stem cells can suppress the immune response toward alloantigen in vitro by inhibiting T cell proliferation in mixed-lymphocyte reactions (MLRs). However, relatively little has been reported regarding the immunomodulative potential of MSCs in vivo. Herein the authors confirm the immunomodulatory effects of rat MSCs in vitro and tested for tolerogenic features in a model of allogeneic heart transplantation. METHODS: Mesenchymal stem cells were obtained from bone marrow aspirates of male Lewis rats (major histocompatibility complex [MHC] haplotype RT1) and ACI rats (RT1). Lewis MSCs were cocultured with ACI spleen cells to reveal direct effects of MSCs on lymphocytes. In addition, MSCs were added to MLRs between ACI T cells as responders and irradiated Lewis spleen cells as stimulators. Finally, MSCs were administered in an allogeneic heart transplantation model at different doses (with and without cyclosporin A [CsA]). RESULTS: Mesenchymal stem cells appeared with typical spindle-shaped morphology in culture and readily differentiated into adipocytes when exposed to differentiation media. Mesenchymal stem cells expressed MHC class I, but not class II or costimulatory molecules. In vitro, MSCs phagocytosed ACI spleen cells. When introduced into an MLR, donor MSCs suppressed the proliferation of stimulated T cells. However, in vivo, MSC injection did not prolong allograft survival. In addition, concurrent treatment with low-dose CsA and MSCs accelerated allograft rejection. CONCLUSIONS: The present data confirm previous reports suggesting that MSCs have immunomodulatory properties in vitro. However, their tolerogenic properties in vivo must be questioned, as MSC injections were not only ineffective at prolonging allograft survival, but tended to promote rejection.


Asunto(s)
Trasplante de Corazón/inmunología , Células Madre Mesenquimatosas/inmunología , Trasplante Homólogo/inmunología , Adipocitos/citología , Adipocitos/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Ciclosporina/farmacología , Rechazo de Injerto/fisiopatología , Antígenos de Histocompatibilidad Clase I/análisis , Inmunosupresores/farmacología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Antígenos de Histocompatibilidad Menor , Ratas , Ratas Endogámicas ACI , Ratas Endogámicas Lew , Bazo/citología , Bazo/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/fisiología , Tolerancia al Trasplante/fisiología
17.
Cell Metab ; 24(5): 657-671, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27641098

RESUMEN

Elevated lactate dehydrogenase A (LDHA) expression is associated with poor outcome in tumor patients. Here we show that LDHA-associated lactic acid accumulation in melanomas inhibits tumor surveillance by T and NK cells. In immunocompetent C57BL/6 mice, tumors with reduced lactic acid production (Ldhalow) developed significantly slower than control tumors and showed increased infiltration with IFN-γ-producing T and NK cells. However, in Rag2-/-γc-/- mice, lacking lymphocytes and NK cells, and in Ifng-/- mice, Ldhalow and control cells formed tumors at similar rates. Pathophysiological concentrations of lactic acid prevented upregulation of nuclear factor of activated T cells (NFAT) in T and NK cells, resulting in diminished IFN-γ production. Database analyses revealed negative correlations between LDHA expression and T cell activation markers in human melanoma patients. Our results demonstrate that lactic acid is a potent inhibitor of function and survival of T and NK cells leading to tumor immune escape.


Asunto(s)
Vigilancia Inmunológica , Células Asesinas Naturales/inmunología , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/biosíntesis , Melanoma/inmunología , Linfocitos T/inmunología , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Recuento de Células , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Citocinas/biosíntesis , Glucólisis/efectos de los fármacos , Humanos , Vigilancia Inmunológica/efectos de los fármacos , Interferón gamma/farmacología , Isoenzimas/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Lactato Deshidrogenasa 5 , Ácido Láctico/farmacología , Masculino , Melanoma/patología , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Fenotipo , Lactato de Sodio/farmacología , Linfocitos T/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
18.
Int J Oncol ; 27(5): 1197-206, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16211213

RESUMEN

Eph-receptor tyrosine kinases (Eph-RTKs) and their membrane-bound receptor-like ligands, the ephrins, represent a cell-cell signaling system that directs cellular migration during development. Differential expression in cancer suggests similar roles in tumor progression. We have previously shown that ephrin-B2 mRNA is overexpressed in advanced malignant melanomas (MM). In this study, immunohistochemistry revealed a most prominent expression of ephrin-B2 in the invasive front of advanced MM. Therefore, we addressed the question of whether ephrin-B2 signaling modulates MM cell migration and matrix interaction. Using a wild-type ephrin-B2-negative B16 mouse MM subclone we show that overexpression of ephrin-B2 leads to the formation of multiple lamellipodia, enhanced polymerisation of actin fibers, and induction of focal adhesion complexes with constitutive activation of focal adhesion kinase. Consequently, ephrin-B2-overexpressing B16 cells display a significant increase of beta1-integrin-mediated attachment to matrix components, preferentially laminin and fibronectin. As a further effect of ephrin-B2 overexpression, we observed an accelerated migration in both Boyden chamber invasion experiments as well as in in vitro scratch-wound assays. We conclude that ephrin-B2 can act as a major modulator of cell migration and matrix interactions of MM cells, which possibly contributes to the expansion and metastatic spread of MM in vivo.


Asunto(s)
Efrina-B2/biosíntesis , Matriz Extracelular/fisiología , Melanoma/patología , Neoplasias Cutáneas/patología , Animales , Adhesión Celular , Movimiento Celular , Perfilación de la Expresión Génica , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia/fisiopatología , Seudópodos/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
19.
Clin Cancer Res ; 10(6): 2109-19, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15041732

RESUMEN

PURPOSE: Despite current chemotherapies, pancreatic cancer remains an uncontrollable, rapidly progressive disease. Here, we tested an approach combining a recently described antiangiogenic drug, rapamycin, with standard gemcitabine cytotoxic therapy on human pancreatic tumor growth. EXPERIMENTAL DESIGN: Tumor growth was assessed in rapamycin and gemcitabine-treated nude mice orthotopically injected with metastatic L3.6pl human pancreatic cancer cells. H&E staining was performed on tumors, along with Ki67 staining for cell proliferation and immunohistochemical terminal deoxynucleotidyl transferase-mediated nick end labeling and CD31 analysis. Rapamycin-treated tumor vessels were also directly examined in dorsal skin-fold chambers for blood flow after thrombosis induction. Cell death in human umbilical vein endothelial cells was assessed by flow cytometry after annexin-V staining. RESULTS: Rapamycin therapy alone inhibited tumor growth and metastasis more than gemcitabine, with remarkable long-term tumor control when the drugs were combined. Mechanistically, H&E analysis revealed tumor vessel endothelium damage and thrombosis with rapamycin treatment. Indeed, dorsal skin-fold chamber analysis of rapamycin-treated tumors showed an increased susceptibility of tumor-specific vessels to thrombosis. Furthermore, terminal deoxynucleotidyl transferase-mediated nick end labeling/CD31 double staining of orthotopic tumors demonstrated apoptotic endothelial cells with rapamycin treatment, which also occurred with human umbilical vein endothelial cells in vitro. In contrast, gemcitabine was not antiangiogenic and, despite its known cytotoxicity, did not reduce proliferation in orthotopic tumors; nevertheless, rapamycin did reduce tumor proliferation. CONCLUSIONS: Our data suggest a novel mechanism whereby rapamycin targets pancreatic tumor endothelium for destruction and thrombosis. We propose that rapamycin-based vascular targeting not only reduces tumor vascularization, it decreases the number of proliferating tumor cells to be destroyed by gemcitabine, thus introducing a new, clinically feasible strategy against pancreatic cancer.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Desoxicitidina/análogos & derivados , Endotelio Vascular/patología , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/patología , Sirolimus/uso terapéutico , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Muerte Celular , División Celular/efectos de los fármacos , Desoxicitidina/uso terapéutico , Endotelio Vascular/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante Heterólogo , Gemcitabina
20.
Clin Cancer Res ; 10(5): 1843-52, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15014039

RESUMEN

PURPOSE: Colorectal neoplasms remain a leading cause of cancer-related deaths. A recognized weakness of conventional 5-fluorouracil (5-FU) therapy relates to expression of the intracellular enzyme, thymidine phosphorylase (TP). Although TP promotes 5-FU cytotoxicity, TP-derived 2-deoxy-D-ribose (dRib) counterproductively stimulates tumor angiogenesis. Here, the newly discovered antiangiogenic drug rapamycin was combined with 5-FU to counteract the potential escape mechanism of dRib-induced angiogenesis. EXPERIMENTAL DESIGN: Orthotopic tumor growth was assessed in rapamycin and 5-FU-treated BALB/c mice with TP-expressing CT-26 colon adenocarcinoma cells. To examine liver metastasis, green-fluorescent protein-transfected CT-26 cells were visualized by fluorescence microscopy after intraportal injection. Cell counting and Ki67 staining were used to determine in vitro and in vivo cell expansion, respectively. In vitro angiogenic effects of dRib were assessed with endothelial cell migration and aortic ring assays. Western blotting detected dRib effects on p70/S6 kinase activation. RESULTS: Rapamycin treatment of mice bearing orthotopic tumors inhibited tumor growth more than did 5-FU, and mice treated with both drugs typically developed no tumors. In the liver metastasis assay, combination therapy blocked metastatic expansion of solitary tumor cells. Interestingly, complex drug activities were suggested by tumor-cell proliferation being more sensitive to 5-FU than to rapamycin in vitro, but more sensitive to rapamycin in vivo. With regard to angiogenesis, dRib-induced endothelial cell migration and aortic ring formation were completely abrogated by rapamycin, correlating with blockage of dRib-induced p70/S6 kinase activation in endothelial cells. CONCLUSIONS: Inhibition of dRib-induced angiogenesis with rapamycin counteracts a potential TP-based escape mechanism for colorectal cancer under 5-FU therapy, introducing a novel, clinically feasible, combination treatment option for this common neoplasm.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Desoxirribosa/farmacología , Fluorouracilo/uso terapéutico , Neovascularización Patológica/prevención & control , Sirolimus/farmacología , Timidina Fosforilasa/metabolismo , Adenocarcinoma/irrigación sanguínea , Animales , Antimetabolitos Antineoplásicos/uso terapéutico , Aorta Torácica/fisiología , Neoplasias del Colon/irrigación sanguínea , Modelos Animales de Enfermedad , Inmunosupresores , Neoplasias Hepáticas/prevención & control , Neoplasias Hepáticas/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Músculo Liso Vascular/fisiología , Ratas , Ratas Endogámicas ACI
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA