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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Br J Cancer ; 127(12): 2108-2117, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36229578

RESUMO

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.


Assuntos
Citostáticos , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/tratamento farmacológico
3.
J Immunol ; 190(12): 6051-8, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23677469

RESUMO

Despite various suspected causes, ranging from endocrine and genetic to infectious and immunological aspects, the molecular mechanisms of miscarriage still remain enigmatic. This work provides evidence that downregulation of 11ß-hydroxysteroid dehydrogenase (HSD) type 2, the key enzyme inactivating glucocorticoid activities, insults the pregnant inflammatory milieu by inhibiting the biosynthesis of lipoxin A4 (LXA4), a metabolite of arachidonic acid, leading to an early loss of the pregnancy. Both LXA4 and its biosynthetic enzymes were found to be decreased in women with spontaneous miscarriages and in the murine miscarriage model. Replenishing LXA4 reversed LPS-induced miscarriages in mouse models, whereas blocking LXA4 signaling resulted in miscarriages in the pregnant mice. The protective effect of LXA4 might be explained by LXA4's role in regulating uterine and placental inflammatory factors and mast cells. The underlying molecular mechanism involved miscarriage-inducing infections or stresses that downregulate the expression of 11ß-HSD2, but not 11ß-HSD1, resulting in increases in glucocorticoid activity and decreases in LXA4. Together, these findings suggest that the stress/glucocorticoid/LXA4 axis might be a common pathway through which miscarriages occur.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/metabolismo , Aborto Espontâneo/fisiopatologia , Glucocorticoides/metabolismo , Lipoxinas/metabolismo , Estresse Fisiológico/fisiologia , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
4.
J Immunol ; 191(6): 3453-61, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23956429

RESUMO

Mechanisms by which tumor cells metastasize to distant organs still remain enigmatic. Immune cells have been assumed to be the root of metastasis by their fusing with tumor cells. This fusion theory, although interpreting tumor metastasis analogically and intriguingly, is arguable to date. We show in this study an alternative explanation by immune cell-derived microparticles (MPs). Upon stimulation by PMA or tumor cell-derived supernatants, immune cells released membrane-based MPs, which were taken up by H22 tumor cells, leading to tumor cell migration in vitro and metastasis in vivo. The underlying molecular basis was involved in integrin α(M)ß2 (CD11b/CD18), which could be effectively relayed from stimulated innate immune cells to MPs, then to tumor cells. Blocking either CD11b or CD18 led to significant decreases in MP-mediated tumor cell metastasis. This MP-mediated transfer of immune phenotype to tumor cells might also occur in vivo. These findings suggest that tumor cells may usurp innate immune cell phenotypes via MP pathway for their metastasis, providing new insight into tumor metastatic mechanism.


Assuntos
Carcinoma Hepatocelular/patologia , Micropartículas Derivadas de Células/metabolismo , Imunidade Inata , Neoplasias Hepáticas/patologia , Antígeno de Macrófago 1/metabolismo , Invasividade Neoplásica/patologia , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Imunidade Inata/imunologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Confocal , Microscopia Eletrônica de Transmissão
5.
Lab Chip ; 24(14): 3470-3479, 2024 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-38896021

RESUMO

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.


Assuntos
Técnicas de Cocultura , Neoplasias Hepáticas , Microambiente Tumoral , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Técnicas Analíticas Microfluídicas/instrumentação , Dispositivos Lab-On-A-Chip , Quimiocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Linhagem Celular Tumoral , Alicerces Teciduais/química , Células Hep G2 , Esferoides Celulares
6.
ACS Biomater Sci Eng ; 10(6): 3792-3805, 2024 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-38814749

RESUMO

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.


Assuntos
Cobre , Estruturas Metalorgânicas , Nanopartículas , Espécies Reativas de Oxigênio , Cicatrização , Zinco , Espécies Reativas de Oxigênio/metabolismo , Cicatrização/efeitos dos fármacos , Animais , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Cobre/química , Cobre/farmacologia , Zinco/química , Nanopartículas/química , Nanopartículas/uso terapêutico , Ratos , Infecção dos Ferimentos/tratamento farmacológico , Infecção dos Ferimentos/microbiologia , Infecção dos Ferimentos/patologia , Infecção dos Ferimentos/metabolismo , Ratos Sprague-Dawley , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/uso terapêutico , Masculino , Staphylococcus aureus/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Antioxidantes/farmacologia , Antioxidantes/química
7.
Clin Immunol ; 149(3): 411-20, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24211715

RESUMO

Hashimoto's thyroiditis (HT) has long been epidemiologically associated with excess iodine levels. However, the underlying immunological mechanisms still remain largely unexplored. Th17 cells are commonly recognized as playing vital roles in various autoimmune diseases. Here we show that intra-thyroid infiltrating Th17 cells and serum IL-17 levels were significantly increased in HT patients. However, the concentration of serum IL-17 was inversely correlated with patients' residual thyroid function while the heterogeneously expressed thyroid IL-17 was directly correlated with local fibrosis. Administration of moderate high levels of iodine was found to facilitate the polarization of murine splenic naïve T cells into Th17 cells, whereas extreme high levels of iodine favored Th1 polarization and inhibited Treg development. These findings suggest that both Th1 and Th17 cells may be involved in the pathogenesis of HT and high levels of iodine may play a critical role in this process by modulating T cell differentiation.


Assuntos
Doença de Hashimoto/patologia , Interleucina-17/imunologia , Células Th1/patologia , Células Th17/patologia , Glândula Tireoide/patologia , Adulto , Idoso , Animais , Estudos de Casos e Controles , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Feminino , Fibrose , Doença de Hashimoto/sangue , Doença de Hashimoto/imunologia , Humanos , Interleucina-17/biossíntese , Interleucina-17/sangue , Iodo/farmacologia , Masculino , Camundongos , Pessoa de Meia-Idade , Especificidade de Órgãos , Baço/imunologia , Baço/metabolismo , Baço/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Glândula Tireoide/imunologia , Glândula Tireoide/metabolismo
8.
Nat Mater ; 11(8): 734-41, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22751180

RESUMO

The identification of stem-cell-like cancer cells through conventional methods that depend on stem cell markers is often unreliable. We developed a mechanical method for selecting tumorigenic cells by culturing single cancer cells in fibrin matrices of ~100 Pa in stiffness. When cultured within these gels, primary human cancer cells or single cancer cells from mouse or human cancer cell lines grew within a few days into individual round colonies that resembled embryonic stem cell colonies. Subcutaneous or intravenous injection of 10 or 100 fibrin-cultured cells in syngeneic or severe combined immunodeficiency mice led to the formation of solid tumours at the site of injection or at the distant lung organ much more efficiently than control cancer cells selected using conventional surface marker methods or cultured on conventional rigid dishes or on soft gels. Remarkably, as few as ten such cells were able to survive and form tumours in the lungs of wild-type non-syngeneic mice.


Assuntos
Separação Celular/métodos , Fibrina , Géis , Melanoma Experimental/patologia , Células-Tronco Neoplásicas , Esferoides Celulares , Animais , Sobrevivência Celular , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Células Tumorais Cultivadas
9.
Cancer Immunol Res ; 11(4): 501-514, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36758174

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células , Neoplasias Pulmonares , Animais , Camundongos , Linfócitos T/metabolismo , Metotrexato/farmacologia , Metotrexato/metabolismo , Neutrófilos/metabolismo , Micropartículas Derivadas de Células/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Antígeno B7-H1/metabolismo , Microambiente Tumoral , Linhagem Celular Tumoral
10.
Signal Transduct Target Ther ; 8(1): 22, 2023 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-36658134

RESUMO

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.


Assuntos
Antineoplásicos , Micropartículas Derivadas de Células , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B , Macrófagos , Humanos , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Lisossomos , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo
11.
Biomed Chromatogr ; 25(9): 1038-44, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21154890

RESUMO

Coenzyme Q(10) (CoQ(10) ) is a naturally occurring compound located in all membranes throughout the cell. A rapid and sensitive HPLC method was developed to determine the concentration of CoQ(10) in dog plasma using a surrogate matrix. Chromatographic separation was carried out on a Diamonsil C(18) column with the UV detector set at 275 nm. Methanol-2-propanol (40:60, v/v) was used as a mobile phase delivered at a flow rate of 1.0 mL/min. Calibrators were prepared using blank plasma-K(2) HPO(4) buffer (50 mm, pH 8.0)-saline (1:3:6, v/v/v) as surrogate matrix. It was shown that the surrogate matrix had similar properties to dog plasma for CoQ(10) in extraction, freeze-thaw and stability. The assay was linear over the concentration range of 0.10-100 µg/mL. The intra- and inter-day precisions were within 13.3% in terms of relative standard deviation (RSD%) and the accuracy was within ±7.5% in terms of relative error. This simple and reproducible HPLC method with less plasma volume (0.4 mL) and adequate sensitivity was successfully applied to pharmacokinetic studies of CoQ(10) in dogs and an investigation of the effect of CoQ(10) formulation on CoQ(10) baseline levels.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Ubiquinona/análogos & derivados , Animais , Cães , Sensibilidade e Especificidade , Ubiquinona/sangue , Ubiquinona/farmacocinética
12.
Cell Mol Immunol ; 17(12): 1233-1244, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31649305

RESUMO

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.


Assuntos
Carcinogênese/patologia , Micropartículas Derivadas de Células/metabolismo , Reprogramação Celular , Interleucina-1beta/metabolismo , Neoplasias Pulmonares/metabolismo , Macrófagos/metabolismo , Animais , Cálcio/metabolismo , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Ligantes , Ativação Linfocitária/imunologia , Lisossomos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptor 3 Toll-Like/metabolismo , Regulação para Cima
13.
Nat Cell Biol ; 22(1): 18-25, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31871320

RESUMO

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.


Assuntos
Ácido 3-Hidroxibutírico/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Via de Pentose Fosfato/efeitos dos fármacos , Fosfoenolpiruvato Carboxiquinase (GTP)/efeitos dos fármacos , Animais , Linfócitos T CD8-Positivos/metabolismo , Gluconeogênese/efeitos dos fármacos , Gluconeogênese/genética , Glicogênio/metabolismo , Homeostase/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoenolpiruvato Carboxiquinase (GTP)/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional/efeitos dos fármacos
14.
Nat Commun ; 11(1): 1769, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286295

RESUMO

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.


Assuntos
Glicogênio/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Animais , Western Blotting , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Inativação Gênica/fisiologia , Humanos , Interleucina-4/metabolismo , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Células THP-1
15.
Cancer Immunol Res ; 8(9): 1193-1205, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32661094

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Neutrófilos/metabolismo , Derrame Pleural Maligno/tratamento farmacológico , Animais , Feminino , Humanos , Camundongos
16.
Nat Biomed Eng ; 4(7): 743-753, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32632227

RESUMO

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.


Assuntos
Antitussígenos/uso terapêutico , Neoplasias dos Ductos Biliares/tratamento farmacológico , Colangiocarcinoma/tratamento farmacológico , Colestase/tratamento farmacológico , Metotrexato/uso terapêutico , Animais , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos/patologia , Medula Óssea , Morte Celular , Colangiocarcinoma/diagnóstico por imagem , Colangiocarcinoma/patologia , Colestase/diagnóstico por imagem , Colestase/patologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Perfusão
17.
Oncogene ; 38(44): 6970-6984, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31409901

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Hipóxia Celular/fisiologia , Ciclo do Ácido Cítrico/fisiologia , Neoplasias da Mama/metabolismo , Regulação para Baixo , Feminino , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Microambiente Tumoral
18.
Sci Transl Med ; 11(474)2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30626714

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/tratamento farmacológico , Derrame Pleural Maligno/complicações , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Micropartículas Derivadas de Células/ultraestrutura , Modelos Animais de Doenças , Endocitose , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Metotrexato/farmacologia , Metotrexato/uso terapêutico , Camundongos Endogâmicos C57BL , Estadiamento de Neoplasias , Derrame Pleural Maligno/imunologia , Derrame Pleural Maligno/patologia , Distribuição Tecidual/efeitos dos fármacos , Transplante Autólogo , Microambiente Tumoral/efeitos dos fármacos
19.
Cancer Immunol Res ; 6(9): 1046-1056, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30002156

RESUMO

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.


Assuntos
Micropartículas Derivadas de Células/imunologia , Inflamação , Neoplasias Pulmonares/patologia , Macrófagos/imunologia , Metástase Neoplásica/imunologia , Animais , Micropartículas Derivadas de Células/patologia , Feminino , Pulmão/citologia , Pulmão/imunologia , Neoplasias Pulmonares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microambiente Tumoral/imunologia
20.
Cancer Immunol Res ; 6(9): 1057-1068, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30018046

RESUMO

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.


Assuntos
Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Micropartículas Derivadas de Células/imunologia , Células Dendríticas/imunologia , Animais , Antígeno B7-1/genética , Antígeno B7-2/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Vacinas Anticâncer/imunologia , Diferenciação Celular , Células Cultivadas , Endocitose/imunologia , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Lisossomos/fisiologia , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA