Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
ACS Biomater Sci Eng ; 10(6): 3792-3805, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38814749

ABSTRACT

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.


Subject(s)
Copper , Metal-Organic Frameworks , Nanoparticles , Reactive Oxygen Species , Wound Healing , Zinc , Reactive Oxygen Species/metabolism , Wound Healing/drug effects , Animals , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Copper/chemistry , Copper/pharmacology , Zinc/chemistry , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Rats , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Infection/pathology , Wound Infection/metabolism , Rats, Sprague-Dawley , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Male , Staphylococcus aureus/drug effects , Oxidative Stress/drug effects , Antioxidants/pharmacology , Antioxidants/chemistry
2.
Cancer Immunol Res ; 11(4): 501-514, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36758174

ABSTRACT

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.


Subject(s)
Cell-Derived Microparticles , Lung Neoplasms , Animals , Mice , T-Lymphocytes/metabolism , Methotrexate/pharmacology , Methotrexate/metabolism , Neutrophils/metabolism , Cell-Derived Microparticles/metabolism , Programmed Cell Death 1 Receptor/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , B7-H1 Antigen/metabolism , Tumor Microenvironment , Cell Line, Tumor
3.
Signal Transduct Target Ther ; 8(1): 22, 2023 01 20.
Article in English | MEDLINE | ID: mdl-36658134

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Cell-Derived Microparticles , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Macrophages , Humans , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Lysosomes , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism
4.
Br J Cancer ; 127(12): 2108-2117, 2022 12.
Article in English | MEDLINE | ID: mdl-36229578

ABSTRACT

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.


Subject(s)
Cytostatic Agents , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy
6.
Nat Biomed Eng ; 4(7): 743-753, 2020 07.
Article in English | MEDLINE | ID: mdl-32632227

ABSTRACT

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.


Subject(s)
Antitussive Agents/therapeutic use , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Cholestasis/drug therapy , Methotrexate/therapeutic use , Animals , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Bone Marrow , Cell Death , Cholangiocarcinoma/diagnostic imaging , Cholangiocarcinoma/pathology , Cholestasis/diagnostic imaging , Cholestasis/pathology , Disease Models, Animal , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neutrophils/drug effects , Perfusion
7.
Cancer Immunol Res ; 8(9): 1193-1205, 2020 09.
Article in English | MEDLINE | ID: mdl-32661094

ABSTRACT

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.


Subject(s)
Cell-Derived Microparticles/metabolism , Neutrophils/metabolism , Pleural Effusion, Malignant/drug therapy , Animals , Female , Humans , Mice
8.
Nat Commun ; 11(1): 1769, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32286295

ABSTRACT

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.


Subject(s)
Glycogen/metabolism , Inflammation/metabolism , Macrophages/metabolism , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Gene Silencing/physiology , Humans , Interleukin-4/metabolism , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , THP-1 Cells
9.
Nat Cell Biol ; 22(1): 18-25, 2020 01.
Article in English | MEDLINE | ID: mdl-31871320

ABSTRACT

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.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Pentose Phosphate Pathway/drug effects , Phosphoenolpyruvate Carboxykinase (GTP)/drug effects , Animals , CD8-Positive T-Lymphocytes/metabolism , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Glycogen/metabolism , Homeostasis/drug effects , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Transcription Factors/metabolism , Transcriptional Activation/drug effects
10.
Cell Mol Immunol ; 17(12): 1233-1244, 2020 12.
Article in English | MEDLINE | ID: mdl-31649305

ABSTRACT

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.


Subject(s)
Carcinogenesis/pathology , Cell-Derived Microparticles/metabolism , Cellular Reprogramming , Interleukin-1beta/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Animals , Calcium/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Ligands , Lymphocyte Activation/immunology , Lysosomes/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Reactive Oxygen Species/metabolism , Toll-Like Receptor 3/metabolism , Up-Regulation
11.
Oncogene ; 38(44): 6970-6984, 2019 10.
Article in English | MEDLINE | ID: mdl-31409901

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Cell Hypoxia/physiology , Citric Acid Cycle/physiology , Breast Neoplasms/metabolism , Down-Regulation , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplastic Stem Cells/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Tumor Microenvironment
12.
Sci Transl Med ; 11(474)2019 01 09.
Article in English | MEDLINE | ID: mdl-30626714

ABSTRACT

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.


Subject(s)
Cell-Derived Microparticles/metabolism , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Pleural Effusion, Malignant/complications , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell-Derived Microparticles/ultrastructure , Disease Models, Animal , Endocytosis , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice, Inbred C57BL , Neoplasm Staging , Pleural Effusion, Malignant/immunology , Pleural Effusion, Malignant/pathology , Tissue Distribution/drug effects , Transplantation, Autologous , Tumor Microenvironment/drug effects
13.
Cancer Immunol Res ; 6(9): 1057-1068, 2018 09.
Article in English | MEDLINE | ID: mdl-30018046

ABSTRACT

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.


Subject(s)
Antigen Presentation , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell-Derived Microparticles/immunology , Dendritic Cells/immunology , Animals , B7-1 Antigen/genetics , B7-2 Antigen/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cancer Vaccines/immunology , Cell Differentiation , Cells, Cultured , Endocytosis/immunology , Female , Histocompatibility Antigens Class I/immunology , Lysosomes/physiology , Melanoma, Experimental , Mice , Mice, Inbred C57BL
14.
Cancer Immunol Res ; 6(9): 1046-1056, 2018 09.
Article in English | MEDLINE | ID: mdl-30002156

ABSTRACT

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.


Subject(s)
Cell-Derived Microparticles/immunology , Inflammation , Lung Neoplasms/pathology , Macrophages/immunology , Neoplasm Metastasis/immunology , Animals , Cell-Derived Microparticles/pathology , Female , Lung/cytology , Lung/immunology , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Microenvironment/immunology
15.
Nat Cell Biol ; 20(1): 21-27, 2018 01.
Article in English | MEDLINE | ID: mdl-29230018

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glycogen/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Melanoma, Experimental/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Skin Neoplasms/genetics , 3-Mercaptopropionic Acid/pharmacology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Enzyme Inhibitors/pharmacology , Female , Gluconeogenesis/drug effects , Gluconeogenesis/genetics , Gluconeogenesis/immunology , Glucose/immunology , Glycogen/immunology , Glycolysis/drug effects , Glycolysis/genetics , Glycolysis/immunology , Homeostasis/immunology , Immunologic Memory , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , NADP/immunology , NADP/metabolism , Pentose Phosphate Pathway/drug effects , Pentose Phosphate Pathway/genetics , Pentose Phosphate Pathway/immunology , Phosphoenolpyruvate Carboxykinase (GTP)/antagonists & inhibitors , Phosphoenolpyruvate Carboxykinase (GTP)/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
16.
Oncoimmunology ; 6(6): e1309487, 2017.
Article in English | MEDLINE | ID: mdl-28680743

ABSTRACT

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.

17.
Biomaterials ; 113: 93-104, 2017 01.
Article in English | MEDLINE | ID: mdl-27810645

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/administration & dosage , Cell-Derived Microparticles/metabolism , Drug Carriers/metabolism , Lysosomes/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/metabolism , Urinary Bladder/drug effects , Animals , Antineoplastic Agents/therapeutic use , Biocompatible Materials/metabolism , Cell Line, Tumor , Cell-Derived Microparticles/pathology , Female , Humans , Lysosomes/drug effects , Lysosomes/pathology , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology
18.
Cell Res ; 26(6): 713-27, 2016 06.
Article in English | MEDLINE | ID: mdl-27167569

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell-Derived Microparticles/metabolism , Drug Resistance, Neoplasm/drug effects , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Doxorubicin/pharmacology , Humans , Mice, Inbred BALB C , Microtubules/drug effects , Microtubules/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pleural Effusion/pathology , Survival Analysis
19.
Cancer Res ; 75(7): 1191-6, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25712344

ABSTRACT

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.


Subject(s)
Melanoma, Experimental/enzymology , Neoplastic Stem Cells/enzymology , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cytosol/enzymology , Female , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoplasm Transplantation , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Tumor Burden , Up-Regulation
20.
Clin Immunol ; 149(3): 411-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24211715

ABSTRACT

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.


Subject(s)
Hashimoto Disease/pathology , Interleukin-17/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Thyroid Gland/pathology , Adult , Aged , Animals , Case-Control Studies , Cell Differentiation/drug effects , Cell Movement/drug effects , Female , Fibrosis , Hashimoto Disease/blood , Hashimoto Disease/immunology , Humans , Interleukin-17/biosynthesis , Interleukin-17/blood , Iodine/pharmacology , Male , Mice , Middle Aged , Organ Specificity , Spleen/immunology , Spleen/metabolism , Spleen/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Thyroid Gland/immunology , Thyroid Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...