Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lab Chip ; 24(14): 3470-3479, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38896021

RESUMEN

Liver cancer, characterized as a kind of malignant tumor within the digestive system, poses great health harm, and immune escape stands out as an important reason for its occurrence and development. Chemokines, pivotal in guiding immune cells' migration, is necessary to initiate and deliver an effective anti-tumor immune response. Therefore, understanding the chemotactic environment and identifying chemokines that regulate recruitment of immune cells to the tumor microenvironment (TME) are critical to improve current immunotherapy interventions. Herein, we report a well-defined inverse opal scaffold generated with a microfluidic emulsion template for the construction of a vascularized liver tumor model, offering insights into immune cells' recruitment. Due to the excellent 3D porous morphology of the inverse opal scaffold, human hepatocellular carcinoma cells can aggregate in the pores of the scaffold to form uniform multicellular tumor spheroids. More attractively, the vascularized liver tumor model can be achieved by constructing a 3D co-culture system involving endothelial cells and hepatocellular carcinoma cells. The results demonstrate that the 3D co-cultured tumor cells increase the neutrophil chemokines remarkably and recruit neutrophils to tumor tissues, then promote tumor progression. This approach opens a feasible avenue for realizing a vascularized liver tumor model with a reliable immune microenvironment close to that of a solid tumor of liver cancer.


Asunto(s)
Técnicas de Cocultivo , Neoplasias Hepáticas , Microambiente Tumoral , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/patología , Técnicas Analíticas Microfluídicas/instrumentación , Dispositivos Laboratorio en un Chip , Quimiocinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Línea Celular Tumoral , Andamios del Tejido/química , Células Hep G2 , Esferoides Celulares
2.
ACS Biomater Sci Eng ; 10(6): 3792-3805, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38814749

RESUMEN

Most antimicrobials treat wound infections by an oxidation effect, which is induced by the generation of reactive oxygen species (ROS). However, the potential harm of the prolonged high level of ROS should not be ignored. In this study, we presented a novel cascade-reaction nanoparticle, Ir@Cu/Zn-MOF, to effectively regulate the ROS level throughout the healing progress of the infected wound. The nanoparticles consisted of a copper/zinc-modified metal-organic framework (Cu/Zn-MOF) serving as the external structure and an inner core composed of Ir-PVP NPs, which were achieved through a process known as "bionic mineralization". The released Cu2+ and Zn2+ from the shell structure contributed to the production of ROS, which acted as antimicrobial agents during the initial stage. With the disintegration of the shell, the Ir-PVP NP core was gradually released, exhibiting the property of multiple antioxidant enzyme activities, thereby playing an important role in clearing excessive ROS and alleviating oxidative stress. In a full-layer infected rat wound model, Ir@Cu/Zn-MOF nanoparticles presented exciting performance in promoting wound healing by clearing the bacteria and accelerating neovascularization as well as collagen deposition. This study provided a promising alternative for the repair of infected wounds.


Asunto(s)
Cobre , Estructuras Metalorgánicas , Nanopartículas , Especies Reactivas de Oxígeno , Cicatrización de Heridas , Zinc , Especies Reactivas de Oxígeno/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Cobre/química , Cobre/farmacología , Zinc/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Ratas , Infección de Heridas/tratamiento farmacológico , Infección de Heridas/microbiología , Infección de Heridas/patología , Infección de Heridas/metabolismo , Ratas Sprague-Dawley , Antibacterianos/farmacología , Antibacterianos/química , Antibacterianos/uso terapéutico , Masculino , Staphylococcus aureus/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Antioxidantes/farmacología , Antioxidantes/química
3.
Cancer Immunol Res ; 11(4): 501-514, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36758174

RESUMEN

Neutrophils act as a "double-edged sword" in the tumor microenvironment by either supporting or suppressing tumor progression. Thus, eliciting a neutrophil antitumor response remains challenging. Here, we showed that tumor cell-derived microparticles induced by methotrexate (MTX-MP) acts as an immunotherapeutic agent to activate neutrophils, increasing the tumor-killing effect of the cells and augmenting T-cell antitumor responses. We found that lactate induced tumor-associated neutrophils to elevate expression of programmed cell death protein 1 (PD-1) and that PD-1+ neutrophils had the properties of N2 neutrophils and suppressed T-cell activation through PD-1/programmed death-ligand 1 (PD-L1) signaling. By performing ex vivo experiments, we found that MTX-MPs-activated neutrophils had reduced surface expression of PD-1 as a result of PD-1 internalization and degradation in the lysosomes, leading to the cells showing a decreased capacity to suppress T-cell responses. In addition, we also found that MTX-MP-activated neutrophils released neutrophil elastase which could kill tumor cells and disrupt tumor stroma, leading to increased T-cell infiltration. Furthermore, using a combination of anti-PD-L1 and MTX-MPs, we observed that long-term survival increased in a mouse model of lung cancer. Collectively, these findings highlight the potential use of a combination of anti-PD-L1 and MTX-MPs to enhance the therapeutic effect of anti-PD-L1 alone.


Asunto(s)
Micropartículas Derivadas de Células , Neoplasias Pulmonares , Animales , Ratones , Linfocitos T/metabolismo , Metotrexato/farmacología , Metotrexato/metabolismo , Neutrófilos/metabolismo , Micropartículas Derivadas de Células/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Antígeno B7-H1/metabolismo , Microambiente Tumoral , Línea Celular Tumoral
4.
Signal Transduct Target Ther ; 8(1): 22, 2023 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-36658134

RESUMEN

Macrophages in tumors (tumor-associated macrophages, TAMs), a major population within most tumors, play key homeostatic functions by stimulating angiogenesis, enhancing tumor cell growth, and suppressing antitumor immunity. Resetting TAMs by simple, efficacious and safe approach(s) is highly desirable to enhance antitumor immunity and attenuate tumor cell malignancy. Previously, we used tumor cell-derived microparticles to package chemotherapeutic drugs (drug-MPs), which resulted in a significant treatment outcome in human malignant pleural effusions via neutrophil recruitments, implicating that drug-MPs might reset TAMs, considering the inhibitory effects of M2 macrophages on neutrophil recruitment and activation. Here, we show that drug-MPs can function as an antitumor immunomodulator by resetting TAMs with M1 phenotype and IFN-ß release. Mechanistically, drug molecules in tumor MPs activate macrophage lysosomal P450 monooxygenases, resulting in superoxide anion formation, which further amplifies lysosomal ROS production and pH value by activating lysosomal NOX2. Consequently, lysosomal Ca2+ signaling is activated, thus polarizing macrophages towards M1. Meanwhile, the drug molecules are delivered from lysosomes into the nucleus where they activate DNA sensor hnRNPA2B1 for IFN-ß production. This lysosomal-nuclear machinery fully arouses the antitumor activity of macrophages by targeting both lysosomal pH and the nuclear innate immunity. These findings highlight that drug-MPs can act as a new immunotherapeutic approach by revitalizing antitumor activity of macrophages. This mechanistic elucidation can be translated to treat malignant ascites by drug-MPs combined with PD-1 blockade.


Asunto(s)
Antineoplásicos , Micropartículas Derivadas de Células , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B , Macrófagos , Humanos , Antineoplásicos/farmacología , Línea Celular Tumoral , Lisosomas , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo
5.
Br J Cancer ; 127(12): 2108-2117, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36229578

RESUMEN

BACKGROUND: Pancreatic cancer is among the most common malignant tumours, and effective therapeutic strategies are still lacking. While Corynoxine (Cory) can induce autophagy in neuronal cells, it remains unclear whether Cory has anti-tumour activities against pancreatic cancer. METHODS: Two pancreatic cancer cell lines, Patu-8988 and Panc-1, were used. Effects of Cory were evaluated by cell viability analysis, EdU staining, TUNEL assay, colony formation assay, and flow cytometry. Quantitative PCR and Western blot were performed to analyse mRNA and protein levels, respectively. In vivo anti-tumour efficacy of Cory was determined by a xenograft model. RESULTS: Cory treatment inhibited cell proliferation, induced endoplasmic reticulum (ER) stress, and triggered apoptosis in the pancreatic cancer cell lines. CHOP knockdown-mediated inhibition of ER stress alleviated the Cory-induced apoptosis but showed a limited effect on cell viability. Cory induced cell death partially via promoting reactive oxygen species (ROS) production and activating p38 signalling. Pretreatment with ROS scavenger N-acetylcysteine and p38 inhibitor SB203580 relieved the Cory-induced inhibition on cell growth. Cory remarkably blocked pancreatic tumour growth in vivo. CONCLUSIONS: Cory exerts an anti-tumour effect on pancreatic cancer primarily via ROS-p38-mediated cytostatic effects. Cory may serve as a promising therapeutic agent for pancreatic cancer.


Asunto(s)
Citostáticos , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico
7.
Cancer Immunol Res ; 8(9): 1193-1205, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661094

RESUMEN

Malignant pleural effusion (MPE) is a frequent complication of various cancers and often leads to a poor quality of life, prognosis, and life expectancy, and its management remains palliative. New approaches that can effectively treat MPE are highly desirable. Here, we show that methotrexate (MTX)-packaging tumor cell-derived microparticles (MTX-MP) act as an effective immunotherapeutic agent to treat patients with MPE by mobilizing and activating neutrophils. We find that MTX-MP perfusion via a pleural catheter elicits the recruitment of neutrophils in patients through macrophage-released CXCL1 and CXCL2. By performing ex vivo experiments, we find that the recruited neutrophils are activated and release reactive oxygen species (ROS) and neutrophil extracellular trap (NET) to kill tumor cells. Neutrophil-released NETs were also able to seal off the damaged endothelium, facilitating MPE resolution in vitro and in tumor-bearing mice. These findings reveal the potential for use of cell-derived materials to package drugs as an immunotherapeutic agent against MPE.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Neutrófilos/metabolismo , Derrame Pleural Maligno/tratamiento farmacológico , Animales , Femenino , Humanos , Ratones
8.
Nat Biomed Eng ; 4(7): 743-753, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32632227

RESUMEN

Most patients with cholangiocarcinoma (CCA) develop extrahepatic malignant biliary obstructions, which require palliative drainage to normalize bilirubin levels and to improve the patients' overall survival. Here, we report that the infusion of methotrexate-containing plasma-membrane microvesicles derived from apoptotic human tumour cells into the bile-duct lumen of patients with extrahepatic CCA mobilized and activated neutrophils and relieved biliary obstruction in 25% of the patients. Neutrophil recruitment by the microvesicles was associated with an increase in uridine diphosphate glucose and complement C5, and led to the degradation of the stromal barrier of CCA. The microvesicles induced pyroptosis of CCA cells through a gasdermin E-dependent pathway, and their intracellular contents released upon CCA-cell death activated patient-derived macrophages into producing proinflammatory cytokines, which attracted a secondary wave of neutrophils to the tumour site. Our findings suggest a possible treatment for the alleviation of obstructive extrahepatic CCA with few adverse effects, and highlight the potential of tumour-cell-derived microvesicles as drug carriers for antitumour therapies.


Asunto(s)
Antitusígenos/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Colestasis/tratamiento farmacológico , Metotrexato/uso terapéutico , Animales , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Médula Ósea , Muerte Celular , Colangiocarcinoma/diagnóstico por imagen , Colangiocarcinoma/patología , Colestasis/diagnóstico por imagen , Colestasis/patología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Perfusión
9.
Nat Commun ; 11(1): 1769, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286295

RESUMEN

Our current understanding of how sugar metabolism affects inflammatory pathways in macrophages is incomplete. Here, we show that glycogen metabolism is an important event that controls macrophage-mediated inflammatory responses. IFN-γ/LPS treatment stimulates macrophages to synthesize glycogen, which is then channeled through glycogenolysis to generate G6P and further through the pentose phosphate pathway to yield abundant NADPH, ensuring high levels of reduced glutathione for inflammatory macrophage survival. Meanwhile, glycogen metabolism also increases UDPG levels and the receptor P2Y14 in macrophages. The UDPG/P2Y14 signaling pathway not only upregulates the expression of STAT1 via activating RARß but also promotes STAT1 phosphorylation by downregulating phosphatase TC45. Blockade of this glycogen metabolic pathway disrupts acute inflammatory responses in multiple mouse models. Glycogen metabolism also regulates inflammatory responses in patients with sepsis. These findings show that glycogen metabolism in macrophages is an important regulator and indicate strategies that might be used to treat acute inflammatory diseases.


Asunto(s)
Glucógeno/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Animales , Western Blotting , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Silenciador del Gen/fisiología , Humanos , Interleucina-4/metabolismo , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Células THP-1
10.
Nat Cell Biol ; 22(1): 18-25, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31871320

RESUMEN

Glycogen has long been considered to have a function in energy metabolism. However, our recent study indicated that glycogen metabolism, directed by cytosolic phosphoenolpyruvate carboxykinase Pck1, controls the formation and maintenance of CD8+ memory T (Tmem) cells by regulating redox homeostasis1. This unusual metabolic program raises the question of how Pck1 is upregulated in CD8+ Tmem cells. Here, we show that mitochondrial acetyl coenzyme A is diverted to the ketogenesis pathway, which indirectly regulates Pck1 expression. Mechanistically, ketogenesis-derived ß-hydroxybutyrate is present in CD8+ Tmem cells; ß-hydroxybutyrate epigenetically modifies Lys 9 of histone H3 (H3K9) of Foxo1 and Ppargc1a (which encodes PGC-1α) with ß-hydroxybutyrylation, upregulating the expression of these genes. As a result, FoxO1 and PGC-1α cooperatively upregulate Pck1 expression, therefore directing the carbon flow along the gluconeogenic pathway to glycogen and the pentose phosphate pathway. These results reveal that ketogenesis acts as an unusual metabolic pathway in CD8+ Tmem cells, linking epigenetic modification required for memory development.


Asunto(s)
Ácido 3-Hidroxibutírico/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Vía de Pentosa Fosfato/efectos de los fármacos , Fosfoenolpiruvato Carboxiquinasa (GTP)/efectos de los fármacos , Animales , Linfocitos T CD8-positivos/metabolismo , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Glucógeno/metabolismo , Homeostasis/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/efectos de los fármacos
11.
Cell Mol Immunol ; 17(12): 1233-1244, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-31649305

RESUMEN

Despite their mutual antagonism, inflammation and immunosuppression coexist in tumor microenvironments due to tumor and immune cell interactions, but the underlying mechanism remains unclear. Previously, we showed that tumor cell-derived microparticles induce an M2 phenotype characterized by immunosuppression in tumor-infiltrating macrophages. Here, we further showed that lung cancer microparticles (L-MPs) induce macrophages to release a key proinflammatory cytokine, IL-1ß, thus promoting lung cancer development. The underlying mechanism involves the activation of TLR3 and the NLRP3 inflammasome by L-MPs. More importantly, tyrosine kinase inhibitor treatment-induced L-MPs also induce human macrophages to release IL-1ß, leading to a tumor-promoting effect in a humanized mouse model. These findings demonstrated that in addition to their anti-inflammatory effect, L-MPs induce a proinflammatory phenotype in tumor-infiltrating macrophages, promoting the development of inflammatory and immunosuppressive tumor microenvironments.


Asunto(s)
Carcinogénesis/patología , Micropartículas Derivadas de Células/metabolismo , Reprogramación Celular , Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Animales , Calcio/metabolismo , Carcinogénesis/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ligandos , Activación de Linfocitos/inmunología , Lisosomas/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 3/metabolismo , Regulación hacia Arriba
12.
Oncogene ; 38(44): 6970-6984, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31409901

RESUMEN

Clinical applications of antiangiogenic agents profoundly affect tumor cell behaviors via the resultant hypoxia. To date, how the hypoxia regulates tumor cells remains unclear. Here, we show that hypoxia promotes the growth of human breast tumorigenic cells that repopulate tumors [tumor-repopulating cells (TRCs)] in vitro and in vivo. This stimulating effect is ascribed to hypoxia-induced reactive oxygen species (ROS) that activates Akt and NF-κB, dependent on the attenuated tricarboxylic acid (TCA) cycle. We find that fumarate is accumulated in the TCA cycle of hypoxic TRCs, leading to glutathione succination, NADPH/NADP+ decrease, and an increase in ROS levels. Mechanistically, hypoxia-increased HIF-1α transcriptionally downregulates the expression of mitochondrial phosphoenolpyruvate carboxykinase (PCK2), leading to TCA cycle attenuation and fumarate accumulation. These findings reveal that hypoxia-reprogrammed TCA cycle promotes human breast TRCs growth via a HIF-1α-downregulated PCK2 pathway, implying a need for a combination of an antiangiogenic therapy with an antioxidant modulator.


Asunto(s)
Neoplasias de la Mama/patología , Hipoxia de la Célula/fisiología , Ciclo del Ácido Cítrico/fisiología , Neoplasias de la Mama/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Neoplásicas/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Microambiente Tumoral
13.
Sci Transl Med ; 11(474)2019 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-30626714

RESUMEN

Cell membrane-derived microparticles (MPs), the critical mediators of intercellular communication, have gained much interest for use as natural drug delivery systems. Here, we examined the therapeutic potential of tumor cell-derived MPs (TMPs) in the context of malignant pleural effusion (MPE). TMPs packaging the chemotherapeutic drug methotrexate (TMPs-MTX) markedly restricted MPE growth and provided a survival benefit in MPE models induced by murine Lewis lung carcinoma and colon adenocarcinoma cells. On the basis of the potential benefit and minimal toxicity of TMPs-MTX, we conducted a human study of intrapleural delivery of a single dose of autologous TMPs packaging methotrexate (ATMPs-MTX) to assess their safety, immunogenicity, and clinical activity. We report our findings on 11 advanced lung cancer patients with MPE. We found that manufacturing and infusing ATMPs-MTX were feasible and safe, without evidence of toxic effects of grade 3 or higher. Evaluation of the tumor microenvironment in MPE demonstrated notable reductions in tumor cells and CD163+ macrophages in MPE after ATMP-MTX infusion, which then translated into objective clinical responses. Moreover, ATMP-MTX treatment stimulated CD4+ T cells to release IL-2 and CD8+ cells to release IFN-γ. Our initial experience with ATMPs-MTX in advanced lung cancer with MPE suggests that ATMPs targeting malignant cells and the immunosuppressive microenvironment may be a promising therapeutic platform for treating malignancies.


Asunto(s)
Micropartículas Derivadas de Células/metabolismo , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/tratamiento farmacológico , Derrame Pleural Maligno/complicaciones , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Micropartículas Derivadas de Células/ultraestructura , Modelos Animales de Enfermedad , Endocitosis , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Metotrexato/farmacología , Metotrexato/uso terapéutico , Ratones Endogámicos C57BL , Estadificación de Neoplasias , Derrame Pleural Maligno/inmunología , Derrame Pleural Maligno/patología , Distribución Tisular/efectos de los fármacos , Trasplante Autólogo , Microambiente Tumoral/efectos de los fármacos
14.
Cancer Immunol Res ; 6(9): 1057-1068, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30018046

RESUMEN

Tumor cell-derived microparticles (T-MP) contain tumor antigen profiles as well as innate signals, endowing them with vaccine potential; however, the precise mechanism by which DCs present T-MP antigens to T cells remains unclear. Here, we show that T-MPs activate a lysosomal pathway that is required for DCs presenting tumor antigens of T-MPs. DCs endocytose T-MPs to lysosomes, where T-MPs increase lysosomal pH from 5.0 to a peak of 8.5 via NOX2-catalyzed reactive oxygen species (ROS) production. This increased pH, coupled with T-MP-driven lysosomal centripetal migration, promotes the formation of MHC class I-tumor antigen peptide complexes. Concurrently, endocytosis of T-MPs results in the upregulation of CD80 and CD86. T-MP-increased ROS activate lysosomal Ca2+ channel Mcoln2, leading to Ca2+ release. Released Ca2+ activates transcription factor EB (TFEB), a lysosomal master regulator that directly binds to CD80 and CD86 promoters, promoting gene expression. These findings elucidate a pathway through which DCs efficiently present tumor antigen from T-MPs to CD8+ T cells, potentiating T-MPs as a novel tumor cell-free vaccine with clinical applications. Cancer Immunol Res; 6(9); 1057-68. ©2018 AACR.


Asunto(s)
Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Micropartículas Derivadas de Células/inmunología , Células Dendríticas/inmunología , Animales , Antígeno B7-1/genética , Antígeno B7-2/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Células Cultivadas , Endocitosis/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Lisosomas/fisiología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL
15.
Cancer Immunol Res ; 6(9): 1046-1056, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30002156

RESUMEN

Despite the frequency of lung metastasis and its associated mortality, the mechanisms behind metastatic tumor cell survival and colonization in the lungs remain elusive. Here, we show that tumor cell-released microparticles (T-MPs) from the primary tumor site play a critical role in the metastatic process. The T-MPs remodeled the lung parenchyma via a macrophage-dependent pathway to create an altered inflammatory and mechanical response to tumor cell invasion. Mechanistically, we show that circulating T-MPs readily enter the lung parenchyma where they are taken up by local macrophages and induce CCL2 production. CCL2 recruits CD11b+Ly6Chigh inflammatory monocytes to the lungs where they mature into F4/80+CD11b+Ly6C- macrophages that not only produce IL6 but also trigger fibrin deposition. IL6 and the deposited fibrin facilitate the survival and growth of tumor-repopulating cells in the lungs by providing chemical and mechanical signals, respectively, thus setting the stage for lung metastasis. These data illustrate that T-MPs reprogram the lung microenvironment promoting metastasis. Cancer Immunol Res; 6(9); 1046-56. ©2018 AACR.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Inflamación , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Metástasis de la Neoplasia/inmunología , Animales , Micropartículas Derivadas de Células/patología , Femenino , Pulmón/citología , Pulmón/inmunología , Neoplasias Pulmonares/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microambiente Tumoral/inmunología
16.
Nat Cell Biol ; 20(1): 21-27, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29230018

RESUMEN

CD8+ memory T (Tm) cells are fundamental for protective immunity against infections and cancers 1-5 . Metabolic activities are crucial in controlling memory T-cell homeostasis, but mechanisms linking metabolic signals to memory formation and survival remain elusive. Here we show that CD8+ Tm cells markedly upregulate cytosolic phosphoenolpyruvate carboxykinase (Pck1), the hub molecule regulating glycolysis, tricarboxylic acid cycle and gluconeogenesis, to increase glycogenesis via gluconeogenesis. The resultant glycogen is then channelled to glycogenolysis to generate glucose-6-phosphate and the subsequent pentose phosphate pathway (PPP) that generates abundant NADPH, ensuring high levels of reduced glutathione in Tm cells. Abrogation of Pck1-glycogen-PPP decreases GSH/GSSG ratios and increases levels of reactive oxygen species (ROS), leading to impairment of CD8+ Tm formation and maintenance. Importantly, this metabolic regulatory mechanism could be readily translated into more efficient T-cell immunotherapy in mouse tumour models.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucógeno/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Melanoma Experimental/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Neoplasias Cutáneas/genética , Ácido 3-Mercaptopropiónico/farmacología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Ciclo del Ácido Cítrico/efectos de los fármacos , Ciclo del Ácido Cítrico/genética , Ciclo del Ácido Cítrico/inmunología , Inhibidores Enzimáticos/farmacología , Femenino , Gluconeogénesis/efectos de los fármacos , Gluconeogénesis/genética , Gluconeogénesis/inmunología , Glucosa/inmunología , Glucógeno/inmunología , Glucólisis/efectos de los fármacos , Glucólisis/genética , Glucólisis/inmunología , Homeostasis/inmunología , Memoria Inmunológica , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/inmunología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADP/inmunología , NADP/metabolismo , Vía de Pentosa Fosfato/efectos de los fármacos , Vía de Pentosa Fosfato/genética , Vía de Pentosa Fosfato/inmunología , Fosfoenolpiruvato Carboxiquinasa (GTP)/antagonistas & inhibidores , Fosfoenolpiruvato Carboxiquinasa (GTP)/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo
17.
Oncoimmunology ; 6(6): e1309487, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28680743

RESUMEN

Stem cell-like tumor-repopulating cells (TRCs) have a critical role in establishing a tumor immunosuppressive microenvironment. However, means to enhance antitumor immunity by disrupting TRCs are absent. Our previous studies have shown that tumor cell-derived microparticles (T-MPs) preferentially abrogate TRCs by delivering antitumor drugs into nuclei of TRCs. Here, we show that low dose irradiation (LDI) enhances the effect of cisplatin-packaging T-MPs (Cis-MPs) on TRCs, leading to inhibiting tumor growth in different tumor models. This antitumor effect is not due to the direct killing of tumor cells but is T cell-dependent and relies on macrophages for their efficacy. The underlying mechanism is involved in therapeutic reprograming macrophages from tumor-promotion to tumor-inhibition by disrupting TRCs and curtailing their vicious education on macrophages. These findings provide a novel strategy to reset macrophage polarization and confer their function more like M1 than M2 types with highly promising potential clinical applications.

18.
Biomaterials ; 113: 93-104, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27810645

RESUMEN

Nonmuscle-invasive bladder cancer (NMIBC) is treated with transurethral resection followed by intravesical chemotherapy. However, drug-resistant tumorigenic cells cannot be eliminated, leading to half of the treated cancers recur with increased stage and grade. Innovative approaches to enhance drug sensitivity and eradicate tumorigenic cells in NMIBC treatment are urgently needed. Here, we show that pre-instillation of tumor cell-derived microparticles (T-MP) as natural biomaterials markedly enhance the inhibitory effects of intravesical chemotherapy on growth and hematuria occurrence of orthotropic bladder cancer in mice. We provide evidence that T-MPs enter and increase the pH value of lysosomes from 4.6 to 5.6, leading to the migration of drug-loaded lysosomes along microtubule tracks toward the nucleus and discharging the drugs whereby for the entry of the nucleus. We propose that T-MPs may function as a potent sensitizer for augmenting NMIBC chemotherapy with unprecedented clinical benefits.


Asunto(s)
Antineoplásicos/administración & dosificación , Micropartículas Derivadas de Células/metabolismo , Portadores de Fármacos/metabolismo , Lisosomas/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/metabolismo , Vejiga Urinaria/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Materiales Biocompatibles/metabolismo , Línea Celular Tumoral , Micropartículas Derivadas de Células/patología , Femenino , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Vejiga Urinaria/metabolismo , Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/patología
19.
Cell Res ; 26(6): 713-27, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27167569

RESUMEN

Developing novel approaches to reverse the drug resistance of tumor-repopulating cells (TRCs) or stem cell-like cancer cells is an urgent clinical need to improve outcomes of cancer patients. Here we show an innovative approach that reverses drug resistance of TRCs using tumor cell-derived microparticles (T-MPs) containing anti-tumor drugs. TRCs, by virtue of being more deformable than differentiated cancer cells, preferentially take up T-MPs that release anti-tumor drugs after entering cells, which in turn lead to death of TRCs. The underlying mechanisms include interfering with drug efflux and promoting nuclear entry of the drugs. Our findings demonstrate the importance of tumor cell softness in uptake of T-MPs and effectiveness of a novel approach in reversing drug resistance of TRCs with promising clinical applications.


Asunto(s)
Antineoplásicos/uso terapéutico , Micropartículas Derivadas de Células/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/patología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Doxorrubicina/farmacología , Humanos , Ratones Endogámicos BALB C , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Derrame Pleural/patología , Análisis de Supervivencia
20.
Cancer Res ; 75(7): 1191-6, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25712344

RESUMEN

Although metabolic defects have been investigated extensively in differentiated tumor cells, much less attention has been directed to the metabolic properties of stem-like cells that repopulate tumors [tumor-repopulating cells (TRC)]. Here, we show that melanoma TRCs cultured in three-dimensional soft fibrin gels reprogram glucose metabolism by hijacking the cytosolic enzyme phosphoenolpyruvate carboxykinase (PCK1), a key player in gluconeogenesis. Surprisingly, upregulated PCK1 in TRCs did not mediate gluconeogenesis but promoted glucose side-branch metabolism, including in the serine and glycerol-3-phosphate pathways. Moreover, this retrograde glucose carbon flow strengthened rather than antagonized glycolysis and glucose consumption. Silencing PCK1 or inhibiting its enzymatic activity slowed the growth of TRCs in vitro and impeded tumorigenesis in vivo. Overall, our work unveiled metabolic features of TRCs in melanoma that have implications for targeting a unique aspect of this disease.


Asunto(s)
Melanoma Experimental/enzimología , Células Madre Neoplásicas/enzimología , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Citosol/enzimología , Femenino , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Carga Tumoral , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA