Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Cell Int ; 24(1): 50, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38291394

RESUMEN

BACKGROUND: Although a combination of immune checkpoint inhibitors (ICIs) is recommended as the first line treatment option for metastatic renal cell carcinoma (mRCC), several immune-related adverse events (irAEs) occur, especially hepatitis. We explored the therapeutic benefits and safety profile of combining oncolytic vaccinia virus, JX-594, with a programmed cell death protein-1 (PD-1) inhibitor. METHODS: We used early-stage and advanced-stage orthotopic murine mRCC models developed by our group. PD-1 inhibitor monotherapy or a PD-1 inhibitor combined with either JX-594 or a cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitor were systemically injected through the peritoneum. An immunofluorescence analysis was performed to analyze the tumor immune microenvironment (TIME). irAEs were assessed in terms of hepatitis. RESULTS: In the early-stage mRCC model mice, the combination of JX-594 and a PD-1 inhibitor significantly decreased the primary tumor size and number of lung nodules, compared with the ICI combination, but the JX-594 and PD-1 inhibitor combination and ICI combination did not differ significantly in the advanced-stage mRCC model mice. The JX-594 and PD-1 inhibitor combination induced tumor-suppressing TIME changes in both the early- and advanced-stage mRCC models. Furthermore, mice treated with the ICI combination had significantly greater hepatic injuries than those treated with the JX-594 and PD-1 inhibitor combination which was evaluated in early-stage mRCC model. CONCLUSIONS: The JX-594 and PD-1 inhibitor combination effectively reduced primary tumors and the metastatic burden, similar to ICI combination therapy, through dynamic remodeling of the TIME. Furthermore, hepatitis was significantly decreased in the JX-594 and PD-1 inhibitor combination group, suggesting the potential benefit of that combination for reducing ICI-induced toxicity.

2.
Adv Exp Med Biol ; 1187: 245-266, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33983582

RESUMEN

Cancer stem cells are a subpopulation of cancer cells responsible for the most demanding and aggressive cancer cell phenotypes: therapy resistance, a self-protective feature of stem cells; distant metastasis, requiring anchorage independence for survival in the circulation; and recurrence, which is related to the dormant-active cycling of stem cells. Normal tissues are composed of parenchymal cells, supportive connective components, and cellular disposal systems for removing the products of physiological wear and tear. Cancer stem cells develop from normal counterparts and progressively interact with their microenvironments, modifying and conditioning the cancer microenvironment. Cancer-associated myeloid cells constitute a major element of the cancer microenvironment. During the process of carcinogenesis, cancer stem cells and their intimately associated myeloid cells mutually interact and evolve, such that the cancer cells potentiate the activity of the myeloid cells and, in return, the myeloid cells increase cancer stem cell characteristics. Normal myeloid cells function as key accessory cells to maintain homeostasis in normal tissues and organs; in cancers, these cells co-evolve with the malignant parenchymal cells and are involved in every aspect of cancer cell biology, including proliferation, invasion, distant metastasis, and the development of resistance to therapy. In this way, cancer-associated myeloid cells provide two of the key hallmarks of cancer: evasion of immune destruction and cancer-promoting inflammation.


Asunto(s)
Recurrencia Local de Neoplasia , Células Madre Neoplásicas , Carcinogénesis , Humanos , Células Mieloides , Microambiente Tumoral
3.
BMC Cancer ; 16(1): 724, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27609180

RESUMEN

BACKGROUND: We previously reported that IL-6 and transglutaminase 2 (TG2) were expressed in more aggressive basal-like breast cancer cells, and TG2 and IL-6 expression gave these cells stem-cell-like phenotypes, increased invasive ability, and increased metastatic potential. In the present study, the underlying mechanism by which IL-6 production is induced in luminal-type breast cancer cells was evaluated, and TG2 overexpression, IL-1ß stimulation, and IL-6 expression were found to give cancerous cells a hormone-independent phenotype. METHODS: Luminal-type breast cancer cells (MCF7 cells) were stably transfected with TG2. To evaluate the requirement for IL-6 neogenesis, MCF7 cells were stimulated with various cytokines. To evaluate tumorigenesis, cancer cells were grown in a three-dimensional culture system and grafted into the mammary fat pads of NOD/scid/IL-2Rγ(-/-) mice. RESULTS: IL-1ß induced IL-6 production in TG2-expressing MCF7 cells through an NF-kB-, PI3K-, and JNK-dependent mechanism. IL-1ß increased stem-cell-like phenotypes, invasiveness, survival in a three-dimensional culture model, and estrogen-independent tumor growth of TG2-expressing MCF7 cells, which was attenuated by either anti-IL-6 or anti-IL-1ß antibody treatment. CONCLUSION: Within the inflammatory tumor microenvironment, IL-1ß increases luminal-type breast cancer cell aggressiveness by stimulating IL-6 production through a TG2-dependent mechanism.


Asunto(s)
Neoplasias de la Mama/patología , Estrógenos/metabolismo , Proteínas de Unión al GTP/genética , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Transglutaminasas/genética , Animales , Neoplasias de la Mama/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Femenino , Proteínas de Unión al GTP/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos NOD , Invasividad Neoplásica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transducción de Señal , Transglutaminasas/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(31): 12673-8, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23858470

RESUMEN

Leptin is a pivotal regulator of energy and glucose homeostasis, and defects in leptin signaling result in obesity and diabetes. The ATP-sensitive potassium (K(ATP)) channels couple glucose metabolism to insulin secretion in pancreatic ß-cells. In this study, we provide evidence that leptin modulates pancreatic ß-cell functions by promoting K(ATP) channel translocation to the plasma membrane via AMP-activated protein kinase (AMPK) signaling. K(ATP) channels were localized mostly to intracellular compartments of pancreatic ß-cells in the fed state and translocated to the plasma membrane in the fasted state. This process was defective in leptin-deficient ob/ob mice, but restored by leptin treatment. We discovered that the molecular mechanism of leptin-induced AMPK activation involves canonical transient receptor potential 4 and calcium/calmodulin-dependent protein kinase kinase ß. AMPK activation was dependent on both leptin and glucose concentrations, so at optimal concentrations of leptin, AMPK was activated sufficiently to induce K(ATP) channel trafficking and hyperpolarization of pancreatic ß-cells in a physiological range of fasting glucose levels. There was a close correlation between phospho-AMPK levels and ß-cell membrane potentials, suggesting that AMPK-dependent K(ATP) channel trafficking is a key mechanism for regulating ß-cell membrane potentials. Our results present a signaling pathway whereby leptin regulates glucose homeostasis by modulating ß-cell excitability.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Células Secretoras de Insulina/metabolismo , Leptina/metabolismo , Potenciales de la Membrana/fisiología , Transducción de Señal/fisiología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Glucosa/metabolismo , Homeostasis/fisiología , Células Secretoras de Insulina/citología , Leptina/genética , Ratones , Ratones Obesos , Transporte de Proteínas/fisiología , ATPasa Intercambiadora de Sodio-Potasio/genética , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
5.
Breast Cancer Res Treat ; 154(1): 13-22, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26438141

RESUMEN

We aimed to identify the factors affecting the successful tumor engraftment in breast cancer patient-derived xenograft (PDX) models. Further, we investigated the prognostic significance and the functional importance of the PDX engraftment-related genes in triple-negative breast cancers (TNBC). The clinico-pathologic features of 81 breast cancer patients whose tissues were used for PDX transplantation were analyzed to identify the factors affecting the PDX engraftment. A gene signature associated with the PDX engraftment was discovered and its clinical importance was tested in a publicly available dataset and in vitro assays. Nineteen out of 81 (23.4 %) transplanted tumors were successfully engrafted into the PDX models. The engraftment rate was highest in TNBC when compared to other subtypes (p = 0.001) and in recurrent or chemotherapy-resistant tumors compared to newly diagnosed primary tumors (p = 0.024). PDX tumors originated from the TNBC cases showed more rapid tumor growth in mice. Gene expression profiling showed that down-regulation of genes involved in the tumor-immune interaction was significantly associated with the successful PDX engraftment. The engraftment gene signature was associated with worse survival outcome when tested in publicly available mRNA datasets of TNBC cases. Among the engraftment-related genes, PHLDA2, TKT, and P4HA2 showed high expression in triple-negative breast cancer cell lines, and siRNA-based gene silencing resulted in reduced cell invasion and proliferation in vitro. Our results show that the PDX engraftment may reflect the aggressive phenotype in breast cancer. Genes associated with the PDX engraftment may provide a novel prognostic biomarker and therapeutic targets in TNBC.


Asunto(s)
Proliferación Celular/genética , Pronóstico , Neoplasias de la Mama Triple Negativas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Prolil Hidroxilasas/biosíntesis , Prolil Hidroxilasas/genética , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/genética
6.
Cell Immunol ; 292(1-2): 94-101, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25460084

RESUMEN

We evaluated the therapeutic effect of human adipose tissue-derived mesenchymal stem cells (hAd-MSCs) in a SKG arthritis model, a relevant animal model for human rheumatoid arthritis. hAd-MSCs were administered intraperitoneally into the mice for five consecutive days from on day 12 or 34 after arthritis induction, when the average clinical score was 0.5 or 5, respectively. They remarkably suppressed arthritis when administered on day 12. Disease suppression was correlated with reduction of pro-inflammatory cytokines and with increased levels of TGF-ß and IL-10 from splenocytes. However, when hAd-MSCs were administered on day 34, the clinical scores were not improved, the histopathological scores were aggravated, and cytokine profiles were differed. Thus, hAd-MSCs showed paradoxical effects, according to the disease phase when they were administered. These suggest that the same cells acted differently depending on the disease progress, and cautions should be paid for safe and effective use of MSCs.


Asunto(s)
Tejido Adiposo/inmunología , Artritis Experimental/inmunología , Células Madre Mesenquimatosas/inmunología , Animales , Células Cultivadas , Humanos , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Ratones Endogámicos BALB C , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/inmunología
7.
J Immunol ; 188(1): 230-7, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22116827

RESUMEN

Naive T cells undergo robust proliferation in lymphopenic conditions, whereas they remain quiescent in steady-state conditions. However, a mechanism by which naive T cells are kept from proliferating under steady-state conditions remains unclear. In this study, we report that memory CD4 T cells are able to limit naive T cell proliferation within lymphopenic hosts by modulating stimulatory functions of dendritic cells (DC). The inhibition was mediated by IL-27, which was primarily expressed in CD8(+) DC subsets as the result of memory CD4 T cell-DC interaction. IL-27 appeared to be the major mediator of inhibition, as naive T cells deficient in IL-27R were resistant to memory CD4 T cell-mediated inhibition. Finally, IL-27-mediated regulation of T cell proliferation was also observed in steady-state conditions as well as during Ag-mediated immune responses. We propose a new model for maintaining peripheral T cell homeostasis via memory CD4 T cells and CD8(+) DC-derived IL-27 in vivo.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígenos CD8 , Proliferación Celular , Células Dendríticas/inmunología , Regulación de la Expresión Génica/inmunología , Memoria Inmunológica , Interleucinas/inmunología , Modelos Inmunológicos , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Comunicación Celular/genética , Comunicación Celular/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/genética , Homeostasis/genética , Homeostasis/inmunología , Interleucinas/biosíntesis , Interleucinas/genética , Ratones , Ratones Noqueados
8.
Breast Cancer Res ; 15(5): R79, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24021059

RESUMEN

INTRODUCTION: Tumor cell interactions with the microenvironment, especially those of bone-marrow-derived myeloid cells, are important in various aspects of tumor metastasis. Myeloid-derived suppressor cells (MDSCs) have been suggested to constitute tumor-favoring microenvironments. In this study, we elucidated a novel mechanism by which the MDSCs can mediate spontaneous distant metastasis of breast cancer cells. METHODS: Murine breast cancer cells, 4T1 and EMT6, were orthotopically grafted into the mammary fat pads of syngeneic BALB/c mice. CD11b(+)Gr-1(+) MDSCs in the spleen, liver, lung and primary tumor mass were analyzed. To evaluate the role of MDSCs in the distant metastasis, MDSCs were depleted or reconstituted in tumor-bearing mice. To evaluate whether MDSCs in the metastasizing tumor microenvironment affect breast cancer cell behavior, MDSCs and cancer cells were co-cultivated. To investigate the role of MDSCs in in vivo metastasis, we blocked the interactions between MDSCs and cancer cells. RESULTS: Using a murine breast cancer cell model, we showed that murine breast cancer cells with high IL-6 expression recruited more MDSCs and that the metastasizing capacity of cancer cells paralleled MDSC recruitment in tumor-bearing mice. Metastasizing, but not non-metastasizing, tumor-derived factors induced MDSCs to increase IL-6 production and full activation of recruited MDSCs occurred in the primary tumor site and metastatic organ in the vicinity of metastasizing cancer cells, but not in lymphoid organs. In addition, tumor-expanded MDSCs expressed Adam-family proteases, which facilitated shedding of IL-6 receptor, thereby contributing to breast cancer cell invasiveness and distant metastasis through IL-6 trans-signaling. The critical role of IL-6 trans-signaling was confirmed in both the afferent and efferent pathways of metastasis. CONCLUSION: In this study, we showed that metastasizing cancer cells induced higher MDSCs infiltration and prompted them to secret exaggerated IL-6 as well as soluble IL-6Ra, which, in turn, triggered a persistent increase of pSTAT3 in tumor cells. This potential tumor-MDSC axis involving IL-6 trans-signaling directly affected breast cancer cell aggressiveness, leading to spontaneous metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Subunidad alfa del Receptor de Interleucina-6/metabolismo , Interleucina-6/metabolismo , Células Mieloides/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/inmunología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Subunidad alfa del Receptor de Interleucina-6/sangre , Ratones , Células Mieloides/inmunología , Metástasis de la Neoplasia , Fosforilación , Factor de Transcripción STAT3/metabolismo , Carga Tumoral , Microambiente Tumoral/inmunología
9.
J Exp Med ; 204(5): 1025-36, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17470642

RESUMEN

IRAK4 is a member of IL-1 receptor (IL-1R)-associated kinase (IRAK) family and has been shown to play an essential role in Toll-like receptor (TLR)-mediated signaling. We recently generated IRAK4 kinase-inactive knock-in mice to examine the role of kinase activity of IRAK4 in TLR-mediated signaling pathways. The IRAK4 kinase-inactive knock-in mice were completely resistant to lipopolysaccharide (LPS)- and CpG-induced shock, due to impaired TLR-mediated induction of proinflammatory cytokines and chemokines. Although inactivation of IRAK4 kinase activity did not affect the levels of TLR/IL-1R-mediated nuclear factor kappaB activation, a reduction of LPS-, R848-, and IL-1-mediated mRNA stability contributed to the reduced cytokine and chemokine production in bone marrow-derived macrophages from IRAK4 kinase-inactive knock-in mice. Both TLR7- and TLR9-mediated type I interferon production was abolished in plasmacytoid dendritic cells isolated from IRAK4 knock-in mice. In addition, influenza virus-induced production of interferons in plasmacytoid DCs was also dependent on IRAK4 kinase activity. Collectively, our results indicate that IRAK4 kinase activity plays a critical role in TLR-dependent immune responses.


Asunto(s)
Inmunidad Innata/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , Animales , Northern Blotting , Western Blotting , Citocinas/metabolismo , Cartilla de ADN , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Virus de la Influenza A/inmunología , Quinasas Asociadas a Receptores de Interleucina-1/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Respir Res ; 14: 35, 2013 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-23496815

RESUMEN

BACKGROUND: Transglutaminase 2 (TG2) is a post-translational protein-modifying enzyme that catalyzes the transamidation reaction, producing crosslinked or polyaminated proteins. Increased TG2 expression and activity have been reported in various inflammatory conditions, such as rheumatoid arthritis, inflammation-associated pulmonary fibrosis, and autoimmune encephalitis. In particular, TG2 from epithelial cells is important during the initial inflammatory response in the lung. In this study, we evaluated the role of TG2 in the pathogenesis of allergic asthma, particularly whether TG2 affects initial activation signaling leading to Th2 differentiation against antigens. METHODS: We induced allergic asthma by ovalbumin sensitization and intranasal challenge in wild-type (WT) BALB/c and TG2-deficient mice. Broncheoalveolar lavage fluid cells and intracellular cytokine production were analyzed by flow cytometry. Interleukin (IL)-33 and TG2 expression in lung epithelial cells was detected by confocal microscopy. RESULTS: Airway responsiveness was attenuated in TG2-deficient mice compared to that in the WT control. In addition, recruitment of eosinophils and Th2 and Th17 differentiation decreased in TG2-deficient mice. Treatment with cysteamine, a transglutaminase inhibitor, also reduced airway hypersensitivity, inflammatory cell recruitment, and T helper cell differentiation. TG2-deficient mice showed reduced IL-33 expression following induction of allergic asthma compared to those in the WT control. CONCLUSIONS: We found that pulmonary epithelial cells damaged by allergens triggered TG2-mediated IL-33 expression leading to type 2 responses by recruiting both innate and adaptive arms of the immune system.


Asunto(s)
Alérgenos/inmunología , Asma/inmunología , Proteínas de Unión al GTP/metabolismo , Interleucinas/metabolismo , Mucosa Respiratoria/inmunología , Células Th2/inmunología , Transglutaminasas/metabolismo , Animales , Asma/metabolismo , Asma/patología , Líquido del Lavado Bronquioalveolar , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/inmunología , Expresión Génica , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-33 , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Proteína Glutamina Gamma Glutamiltransferasa 2 , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Transducción de Señal , Células Th2/metabolismo , Transglutaminasas/inmunología
11.
J Immunother Cancer ; 11(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36717184

RESUMEN

BACKGROUND: Oncolytic virus immunotherapy has revolutionized cancer immunotherapy by efficiently inducing both oncolysis and systemic immune activation. Locoregional administration has been used for oncolytic virus therapy, but its applications to deep-seated cancers have been limited. Although systemic delivery of the oncolytic virus would maximize viral immunotherapy's potential, this remains a hurdle due to the rapid removal of the administered virus by the complement and innate immune system. Infected cells produce some vaccinia viruses as extracellular enveloped virions, which evade complement attack and achieve longer survival by expressing host complement regulatory proteins (CRPs) on the host-derived envelope. Here, we generated SJ-600 series oncolytic vaccinia viruses that can mimic complement-resistant extracellular enveloped virions by incorporating human CRP CD55 on the intracellular mature virion (IMV) membrane. METHODS: The N-terminus of the human CD55 protein was fused to the transmembrane domains of the six type I membrane proteins of the IMV; the resulting recombinant viruses were named SJ-600 series viruses. The SJ-600 series viruses also expressed human granulocyte-macrophage colony-stimulating factor (GM-CSF) to activate dendritic cells. The viral thymidine kinase (J2R) gene was replaced by genes encoding the CD55 fusion proteins and GM-CSF. RESULTS: SJ-600 series viruses expressing human CD55 on the IMV membrane showed resistance to serum virus neutralization. SJ-607 virus, which showed the highest CD55 expression and the highest resistance to serum complement-mediated lysis, exhibited superior anticancer activity in three human cancer xenograft models, compared with the control Pexa-Vec (JX-594) virus, after single-dose intravenous administration. The SJ-607 virus administration elicited neutralizing antibody formation in two immunocompetent mouse strains like the control JX-594 virus. Remarkably, we found that the SJ-607 virus evades neutralization by vaccinia virus-specific antibodies. CONCLUSION: Our new oncolytic vaccinia virus platform, which expresses human CD55 protein on its membrane, prolonged viral survival by protecting against complement-mediated lysis and by evading neutralization by vaccinia virus-specific antibodies; this may provide a continuous antitumor efficacy until a complete remission has been achieved. Such a platform may expand the target cancer profile to include deep-seated cancers and widespread metastatic cancers.


Asunto(s)
Neoplasias , Virus Oncolíticos , Humanos , Ratones , Animales , Virus Vaccinia/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/terapia , Neoplasias/patología , Administración Intravenosa
12.
J Biol Chem ; 286(27): 24036-45, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21602274

RESUMEN

Lysophosphatidic acid (LPA) is a bioactive phospholipid that affects various biological functions, such as cell proliferation, migration, and survival, through LPA receptors. Among them, the motility of cancer cells is an especially important activity for invasion and metastasis. Recently, AMP-activated protein kinase (AMPK), an energy-sensing kinase, was shown to regulate cell migration. However, the specific role of AMPK in cancer cell migration is unknown. The present study investigated whether LPA could induce AMPK activation and whether this process was associated with cell migration in ovarian cancer cells. We found that LPA led to a striking increase in AMPK phosphorylation in pathways involving the phospholipase C-ß3 (PLC-ß3) and calcium/calmodulin-dependent protein kinase kinase ß (CaMKKß) in SKOV3 ovarian cancer cells. siRNA-mediated knockdown of AMPKα1, PLC-ß3, or (CaMKKß) impaired the stimulatory effects of LPA on cell migration. Furthermore, we found that knockdown of AMPKα1 abrogated LPA-induced activation of the small GTPase RhoA and ezrin/radixin/moesin proteins regulating membrane dynamics as membrane-cytoskeleton linkers. In ovarian cancer xenograft models, knockdown of AMPK significantly decreased peritoneal dissemination and lung metastasis. Taken together, our results suggest that activation of AMPK by LPA induces cell migration through the signaling pathway to cytoskeletal dynamics and increases tumor metastasis in ovarian cancer.


Asunto(s)
Movimiento Celular , Lisofosfolípidos/metabolismo , Neoplasias Ováricas/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Línea Celular Tumoral , Membrana Celular/genética , Membrana Celular/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Activación Enzimática , Femenino , Humanos , Lisofosfolípidos/genética , Metástasis de la Neoplasia , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Receptores del Ácido Lisofosfatídico/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
13.
Clin Immunol ; 145(2): 122-32, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001131

RESUMEN

The increased activity of transglutaminase 2 (TG2) in various inflammatory and fibrotic conditions results in the development of numerous disease processes. Experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis, is an inflammatory and demyelinating disease of the central nervous system and is mediated by many inflammatory cytokines and mediators. We examined the role of TG2 in encephalitogenic CD4(+) T cell responses and EAE development using mice lacking TG2 (TG2(-/-)). TG2(-/-) mice showed decreased disease severity as compared with wild-type (WT) mice. Treatment with cysteamine, a TG2 inhibitor, ameliorated disease severity in WT mice. Exacerbated disability in WT mice resulted from the increased infiltration of cytokine-producing CD4(+) T cells and sustained expression of inflammatory cytokines and mediators in the lesion. The increased number of IL-17- and IFN-γ-producing cells in the spinal cord resulted from peripheral expansion of these cells after immunization with myelin-derived antigen. In vitro differentiation of WT and TG2(-/-) splenocytes revealed that proliferation and activation-induced cell death did not differ, but differentiation into IL-17- or IFN-γ-producing cells was increased in WT mice. Adoptive transfer experiments revealed that pathogenic CD4(+) T cell differentiation and disease progression were caused by both the T cell-intrinsic and -extrinsic effects of TG2. This study is the first to report a pathogenic role for TG2 in the EAE progress and suggests that therapeutic targeting of TG2 may be effective against multiple sclerosis.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteínas de Unión al GTP/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Transglutaminasas/antagonistas & inhibidores , Traslado Adoptivo , Animales , Antígenos/inmunología , Antígenos/farmacología , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Proliferación Celular/efectos de los fármacos , Cisteamina/farmacología , Cisteamina/uso terapéutico , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Proteínas de Unión al GTP/deficiencia , Proteínas de Unión al GTP/genética , Inflamación/enzimología , Inflamación/inmunología , Inflamación/patología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Ratones , Ratones Noqueados , Vaina de Mielina/inmunología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Índice de Severidad de la Enfermedad , Transducción de Señal , Médula Espinal/efectos de los fármacos , Médula Espinal/enzimología , Médula Espinal/inmunología , Transglutaminasas/deficiencia , Transglutaminasas/genética
14.
Eur J Immunol ; 41(2): 392-402, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21268009

RESUMEN

Although NKT cells have been implicated in diverse immunomodulatory responses, the effector mechanisms underlying the NKT cell-mediated regulation of pathogenic T helper cells are not well understood. Here, we show that invariant NKT cells inhibited the differentiation of CD4(+) T cells into Th17 cells both in vitro and in vivo. The number of IL-17-producing CD4(+) T cells was reduced following co-culture with purified NK1.1(+) TCR(+) cells from WT, but not from CD1d(-/-) or Jα18(-/-) , mice. Co-cultured NKT cells from either cytokine-deficient (IL-4(-/-) , IL-10(-/-) , or IFN-γ(-/-) ) or WT mice efficiently inhibited Th17 differentiation. The contact-dependent mechanisms of NKT cell-mediated regulation of Th17 differentiation were confirmed using transwell co-culture experiments. On the contrary, the suppression of Th1 differentiation was dependent on IL-4 derived from the NKT cells. The in vivo regulatory capacity of NKT cells on Th17 cells was confirmed using an experimental autoimmune uveitis model induced with human IRBP(1-20) (IRBP, interphotoreceptor retinoid-binding protein) peptide. NKT cell-deficient mice (CD1d(-/-) or Jα18(-/-) ) demonstrated an increased disease severity, which was reversed by the transfer of WT or cytokine-deficient (IL-4(-/-) , IL-10(-/-) , or IFN-γ(-/-) ) NKT cells. Our results indicate that invariant NKT cells inhibited autoimmune uveitis predominantly through the cytokine-independent inhibition of Th17 differentiation.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Diferenciación Celular/inmunología , Células T Asesinas Naturales/inmunología , Células Th17/inmunología , Uveítis/inmunología , Traslado Adoptivo , Animales , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Comunicación Celular/inmunología , Recuento de Células , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Ojo/patología , Proteínas del Ojo/inmunología , Interleucina-4/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células T Asesinas Naturales/metabolismo , Células T Asesinas Naturales/patología , Células T Asesinas Naturales/trasplante , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Proteínas de Unión al Retinol/inmunología , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/metabolismo , Uveítis/genética , Uveítis/metabolismo , Uveítis/patología , Vacunación
15.
Biomedicines ; 10(1)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35052851

RESUMEN

Immune checkpoint inhibitors and tyrosine kinase inhibitors are the first-line treatment for metastatic renal cell carcinoma (mRCC), but their benefits are limited to specific patient subsets. Here, we aimed to evaluate the therapeutic efficacy of JX-594 (pexastimogene devacirepvec, Pexa-vec) monotherapy by systemic injection in comparison with sunitinib monotherapy in metastatic orthotopic RCC murine models. Two highly metastatic orthotopic RCC models were developed to compare the treatment efficacy in the International Metastatic RCC Database Consortium favorable-risk and intermediate- or poor-risk groups. JX-594 was systemically injected through the peritoneum, whereas sunitinib was orally administered. Post-treatment, tumor microenvironment (TME) remodeling was determined using immunofluorescence analysis. Systemic JX-594 monotherapy injection demonstrated therapeutic benefit in both early- and advanced-stage mRCC models. Sunitinib monotherapy significantly reduced the primary tumor burden and number of lung metastases in the early-stage, but not in the advanced-stage mRCC model. Systemic JX-594 delivery remodeled the primary TME and lung metastatic sites by increasing tumor-infiltrating CD4/8+ T cells and dendritic cells. Systemic JX-594 monotherapy demonstrated significantly better therapeutic outcomes compared with sunitinib monotherapy in both early- and advanced-stage mRCCs by converting cold tumors into hot tumors. Sunitinib monotherapy effectively suppressed primary tumor growth and lung metastasis in early-stage mRCC.

16.
Biomedicines ; 10(4)2022 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-35453555

RESUMEN

Oncolytic virotherapy has garnered attention as an antigen-agnostic therapeutic cancer vaccine that induces cancer-specific T cell responses without additional antigen loading. As anticancer immune responses are compromised by a lack of antigenicity and chronic immunosuppressive microenvironments, an effective immuno-oncology modality that converts cold tumors into hot tumors is crucial. To evaluate the immune-activating characteristics of oncolytic vaccinia virus (VACV; JX-594, pexastimogene devacirepvec), diverse murine syngeneic cancer models with different tissue types and immune microenvironments were used. Intratumorally administered mJX-594, a murine variant of JX-594, potently increased CD8+ T cells, including antigen-specific cancer CD8+ T cells, and decreased immunosuppressive cells irrespective of tissue type or therapeutic efficacy. Remodeling of tumors into inflamed ones by mJX-594 led to a response to combined anti-PD-1 treatment, but not to mJX-594 or anti-PD-1 monotherapy. mJX-594 treatment increased T cell factor 1-positive stem-like T cells among cancer-specific CD8+ T cells, and anti-PD-1 combination treatment further increased proliferation of these cells, which was important for therapeutic efficacy. The presence of functional cancer-specific CD8+ T cells in the spleen and bone marrow for an extended period, which proliferated upon encountering cancer antigen-loaded splenic dendritic cells, further indicated that long-term durable anticancer immunity was elicited by oncolytic VACV.

17.
Breast Cancer Res ; 13(5): R96, 2011 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-21967801

RESUMEN

INTRODUCTION: Inflammation has been implicated in cancer aggressiveness. As transglutaminase 2 (TG2), which has been associated with inflammatory signaling, has been suggested to play a role in tumor behavior, we propose that TG2 may be an important linker inducing interleukin (IL)-6-mediated cancer-cell aggressiveness, including distant hematogenous metastasis. METHODS: To investigate the role for TG2 and IL-6, TG2-knocked-down and IL-6-knocked-down cancer cells were generated by using shRNA. Human breast cancer cell xenograft model in highly immunocompromised mice and human advanced breast cancer primary tumor tissue microarrays were used in this study. RESULTS: IL-6 production in human breast cancer cells was dependent on their TG2 expression level. In vitro tumor-sphere formation was dependent on TG2 and downstream IL-6 production from cancer cells. Primary tumor growth in the mammary fat pads and distant hematogenous metastasis into the lung was also dependent on TG2 and downstream IL-6 expression levels. The effect of TG2 expression on human breast cancer distant metastasis was investigated by analyzing a tissue microarray of primary tumors from 412 patients with their clinical data after 7 years. TG2 expression in primary tumor tissue was inversely correlated with recurrence-free survival (P = 0.019) and distant metastasis-free survival (DMFS) (P = 0.006) in patients with advanced breast cancer. Furthermore, by using public datasets that included a total of 684 breast cancer patients, we found that the combined high expression of TG2 and IL-6 was associated with shorter DMFS, compared with the high expression of IL-6 only (P = 0.013). CONCLUSIONS: We provide evidence that TG2 is an important link in IL-6-mediated tumor aggressiveness, and that TG2 could be an important mediator of distant metastasis, both in a xenograft animal model and in patients with advanced breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Interleucina-6/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo , Animales , Neoplasias de la Mama/mortalidad , Supervivencia Celular/genética , Supervivencia sin Enfermedad , Femenino , Proteínas de Unión al GTP , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-6/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Proteína Glutamina Gamma Glutamiltransferasa 2 , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
J Korean Med Sci ; 26(10): 1270-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22022177

RESUMEN

Secondary lymphoid tissue chemokine (SLC), which is expressed in T cell zones of secondary lymphoid organs, including the spleen and lymph nodes, strongly recruits both T lymphocytes and mature dendritic cells. As appropriate interaction of tumor-specific T cells and mature dendritic cells, equipped with tumor antigens, is a prerequisite for effective T cell immunity against established tumors, we mobilized lymphocytes and dendritic cells to tumor sites by intratumoral injection of secondary lymphoid tissue chemokine-Fc (SLC-Fc) fusion protein using the B16F10 murine melanoma model. Activation of dendritic cells, another prerequisite for the effective activation of naïve tumor-specific T cells, was achieved by the addition of immunostimulatory cytosine-phosphorothioate-guanine oligodeoxynucleotide (CpG-ODN) into the tumor site. Intratumoral administration of SLC-Fc or CpG-ODN revealed antitumor effects against B16F10 murine melanoma grown in the subcutaneous space. Co-treatment of SLC-Fc and CpG-ODN displayed synergistic effects in reducing the tumor size. The synergistic antitumor effect in co-treatment group was correlated with the synergistic/additive increase in the infiltration of CD4(+) T cells and CD11c(+) dendritic cells in the tumor mass compared to the single treatment groups. These results suggest that the combined use of chemokines and adjuvant molecules may be a possible strategy in clinical tumor immunotherapy.


Asunto(s)
Quimiocina CCL21/administración & dosificación , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Oligodesoxirribonucleótidos/administración & dosificación , Animales , Antígeno CD11c/inmunología , Linfocitos T CD4-Positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL21/farmacología , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inmunoterapia , Inyecciones Intralesiones , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/farmacología , Linfocitos T/inmunología , Linfocitos T/metabolismo
19.
Immun Inflamm Dis ; 9(3): 871-882, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33945658

RESUMEN

BACKGROUND: Transglutaminase 2 (TG2), a multifunctional calcium-dependent acyltransferase, is upregulated in asthmatic airways and reported to play a role in the pathogenesis of allergic asthma. However, the underlying mechanism is not fully understood. OBJECTIVE: To investigate the role of TG2 in alternative activation of alveolar macrophages by using murine asthma model. METHODS: TG2 expression was assessed in induced sputum of 21 asthma patients and 19 healthy controls, and lung tissue of ovalbumin (OVA)-induced murine asthma model. To evaluate the role of TG2 in asthma, we developed an OVA asthma model in both TG2 null and wild-type mice. The expression of M2 macrophage markers was measured by fluorescence-activated cell sorting (FACS) after OVA sensitization and challenge. To evaluate the effect of TG2 inhibition in vitro, interleukin 4 (IL-4) or IL-13-stimulated expression of M2 macrophage markers was measured in CRL-2456 cells in the presence and absence of a TG2 inhibitor. RESULTS: The expression of both TG2 and M2 markers was increased in the sputum of asthmatics compared with that of healthy controls. The expression of TG2 was increased in macrophages of OVA mice. Airway hyperresponsiveness, and the number of inflammatory cells, including eosinophils, was significantly reduced in TG2 null mice compared with wild-type mice. Enhanced expression of M2 markers in OVA mice was normalized by TG2 knockout. IL-4 or IL-13-stimulated expression of M2 markers in alveolar macrophages was also attenuated by TG2 inhibitor treatment in vitro. CONCLUSION: Our results suggest that TG2-mediated modulation of alveolar macrophage polarization plays important roles in the pathogenesis of asthma.


Asunto(s)
Asma , Macrófagos Alveolares , Animales , Humanos , Inflamación , Pulmón , Activación de Macrófagos , Ratones
20.
Immunology ; 129(4): 567-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20102412

RESUMEN

The maturation of antigen-presenting dendritic cells (DCs) serves as an important determinant for the regulation of immunity, and overall immune response. We hypothesized that a reduced immune response to donor alloantigens and improved allograft survival could be induced by pre-treating recipients with bone-marrow-derived donor-strain fixed immature DCs (FIDCs). Donor-strain-derived mature and immature DCs were fixed before grafting to ensure that they possessed a stable immunogenic phenotype. The fixed mature DCs effectively induced allogeneic T-cell proliferation in recipients, whereas FIDCs were unable to elicit an allogeneic T-cell response. T cells that had previously been exposed to FIDCs maintained naïve phenotypes and were unable to extensively divide after injection into lethally irradiated donor-strain mice. The pre-treatment of recipients with donor-strain FIDCs markedly prolonged the survival of islet as well as skin allografts. However, T-cell hyporesponsiveness induced by FIDC injection was abrogated by the depletion of CD4(+) CD25(+) T cells. Consequently, FIDC-induced T-cell hyporesponsiveness could reflect anergy rather than specific deletion. Our findings suggest that FIDCs of donor strain could be used to induce long-term graft survival.


Asunto(s)
Células Dendríticas/inmunología , Isoantígenos/inmunología , Donantes de Tejidos , Animales , Células de la Médula Ósea/inmunología , Proliferación Celular , Femenino , Formaldehído/química , Supervivencia de Injerto/inmunología , Antígenos H-2/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Fenotipo , Linfocitos T Reguladores/inmunología , Fijación del Tejido , Tolerancia al Trasplante , Trasplante Homólogo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA