Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Antib Ther ; 7(1): 77-87, 2024 Jan.
Article En | MEDLINE | ID: mdl-38371956

Atopic dermatitis (AD) is a chronic inflammatory skin condition characterized by dysregulated immune responses. The key mediators of AD pathogenesis are T helper 2 (TH2) cells and TH2 cytokines. Targeting interleukin 4 (IL4), IL13 or IL31 has become a pivotal focus in both research and clinical treatments for AD. However, the need remains pressing for the development of a more effective and safer therapy, as the current approaches often yield low response rates and adverse effects. In response to this challenge, we have engineered a immunoglobulin G-single-chain fragment variable (scFv) format bispecific antibody (Ab) designed to concurrently target IL4R and IL31R. Our innovative design involved sequence optimization of VL-VH and the introduction of disulfide bond (VH44-VL100) within the IL31Rα Ab scFv region to stabilize the scFv structure. Our bispecific Ab efficiently inhibited the IL4/IL13/IL31 signaling pathways in vitro and reduced serum immunoglobulin E and IL31 levels in vivo. Consequently, this intervention led to improved inflammation profiles and notable amelioration of AD symptoms. This research highlighted a novel approach to AD therapy by employing bispecific Ab targeting IL4Rα and IL31Rα with potent efficacy.

3.
Adv Ther (Weinh) ; 5(1)2022 Jan.
Article En | MEDLINE | ID: mdl-35812344

Radiotherapy (RT) has demonstrated synergy with immune checkpoint blockade (ICB) in preclinical models. However, its potential as an immunoadjuvant is limited by low immunogenicity at low radiation doses and immunosuppression at high radiation doses. It is hypothesized that radiosensitizers can enhance both the anticancer and immunogenic effects of low-dose radiation. Herein the authors report the antitumor immunity of combined RT and immunotherapy with dimethylaminomicheliolide (DMAMCL), a prodrug of the anti-inflammatory sesquiterpene lactone micheliolide (MCL). DMAMCL sensitized cancer cells to a single fraction of RT in vitro by inducing apoptosis and DNA double-strand breaks. DMAMCL with 5 fractions of 2 Gy focal X-ray irradiation led to significant anticancer efficacy in subcutaneous and spontaneous models of murine cancer. DMAMCL-sensitized RT upregulated programmed death-ligand 1 (PD-L1) expression in the tumors. Combination of DMAMCL-sensitized RT with anti-PD-L1 ICB significantly enhanced antitumor efficacy by increasing tumor-infiltrating CD4+ and CD8+ T cells and establishing immune memory.

4.
Nat Biomed Eng ; 6(2): 144-156, 2022 02.
Article En | MEDLINE | ID: mdl-35190678

Checkpoint blockade elicits durable responses in immunogenic cancers, but it is largely ineffective in immunologically 'cold' tumours. Here we report the design, synthesis and performance of a bismuth-based nanoscale metal-organic framework that modulates the immunological and mechanical properties of the tumour microenvironment for enhanced radiotherapy-radiodynamic therapy. In mice with non-immunogenic prostate and pancreatic tumours irradiated with low X-ray doses, the intratumoural injection of the radiosensitizer mediated potent outcomes via the repolarization of immunosuppressive M2 macrophages into immunostimulatory M1 macrophages, the reduction of the concentration of intratumoural transforming growth factor beta (TGF-ß) and of collagen density, and the inactivation of cancer-associated fibroblasts. When intravenously injected in combination with checkpoint-blockade therapy, the radiosensitizer mediated the reversal of immunosuppression in primary and distant tumours via the systemic reduction of TGF-ß levels, which led to the downregulation of collagen expression, the stimulation of T-cell infiltration in the tumours and a robust abscopal effect. Nanoscale radiosensitizers that stimulate anti-tumour immunity and T-cell infiltration may enhance the therapeutic outcomes of checkpoint blockade in other tumour types.


Metal-Organic Frameworks , Neoplasms , Animals , Immunity , Immunotherapy , Male , Metal-Organic Frameworks/pharmacology , Mice , Tumor Microenvironment
5.
ACS Nano ; 15(11): 17515-17527, 2021 Nov 23.
Article En | MEDLINE | ID: mdl-34709030

Ineffective antigen cross-presentation in the tumor microenvironment compromises the generation of antitumor immune responses. Radiotherapy-radiodynamic therapy (RT-RDT) with nanoscale metal-organic frameworks (nMOFs) induces robust adaptive immune responses despite modest activation of canonical antigen presenting dendritic cells. Here, using transplantable and autochthonous murine tumor models, we demonstrate that RT-RDT induces antitumor immune responses via early neutrophil infiltration and reprogramming. Intravenous or intratumoral injection of nMOFs recruited peripheral CD11b+Ly6G+CD11c- neutrophils into tumors. The activation of nMOFs by low-dose X-rays significantly increased the population of CD11b+Ly6G+CD11c+ hybrid neutrophils with upregulated expression of the co-stimulatory molecules CD80 and CD86 as well as major histocompatibility complex class II molecules. Thus, nMOF-enabled RT-RDT reshapes a favorable tumor microenvironment for antitumor immune responses by reprogramming tumor-infiltrating neutrophils to function as non-canonical antigen presenting cells for effective cross-presentation of tumor antigens.


Metal-Organic Frameworks , Neutrophils , Mice , Animals , Antigen-Presenting Cells , Antigen Presentation , Metal-Organic Frameworks/pharmacology
6.
Sci Adv ; 6(40)2020 10.
Article En | MEDLINE | ID: mdl-33008911

Cancer vaccines have been actively pursued to bolster antitumor immunity. Here, we designed nanoscale metal-organic frameworks (nMOFs) as locally activable immunotherapeutics to release danger-associated molecular patterns (DAMPs) and tumor antigens and deliver pathogen-associated molecular patterns (PAMPs) for in situ personalized cancer vaccination. When activated by x-rays, nMOFs effectively generate reactive oxygen species to release DAMPs and tumor antigens while delivering CpG oligodeoxynucleotides as PAMPs to facilitate the maturation of antigen-presenting cells. Together, DAMPs, tumor antigens, and PAMPs expand cytotoxic T cells in tumor-draining lymph nodes to reinvigorate the adaptive immune system for local tumor regression. When treated in combination with an immune checkpoint inhibitor, the local therapeutic effects of nMOF-based vaccines were extended to distant tumors via attenuating T cell exhaustion. Our work demonstrates the potential of nMOFs as x-ray-activable in situ cancer vaccines to awaken the host's innate and adaptive immune systems for systemic antitumor immunity.


Cancer Vaccines , Metal-Organic Frameworks , Neoplasms , Antigens, Neoplasm , Humans , Metal-Organic Frameworks/chemistry , Neoplasms/therapy , Pathogen-Associated Molecular Pattern Molecules , Vaccination , X-Rays
7.
Biomaterials ; 210: 94-104, 2019 07.
Article En | MEDLINE | ID: mdl-31060867

Though early detection and treatment of primary tumors has significantly improved in recent years, metastatic disease remains among the most significant challenges in cancer therapy. Cancer cells can disseminate before the primary tumor is detected to form micro or gross metastases, requiring toxic systemic therapies. To prevent and suppress metastases, we have developed a nontoxic, long-circulating nanoscale coordination polymer (NCP) protecting microRNA (miRNA) in circulation and releasing it in tumors. PtIV(en)2 [en = ethylenediamine] containing NCPs (PtEN) can release a nontoxic, kinetically inert PtII(en)2 compound and carbon dioxide which aids the endosomal escape of its miRNA cargo, miR-655-3p. Without the presence of the PtEN core, the miRNA showed cellular uptake but no effect. When transfected into human colorectal HCT116 cells by NCPs, this oligometastatic miRNA limited proliferation and epithelial-to-mesenchymal transition by preventing ß-catenin nuclear translocation and tumor cell invasion. Systemic administrations of PtEN/miR-655-3p sustained effective transfection to reduce liver colonization and tumor burden in a xenogenic hepatic metastatic model of HCT116 without any observable toxicity.


Colorectal Neoplasms/secondary , Epithelial-Mesenchymal Transition , Gene Transfer Techniques , Liver Neoplasms/pathology , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Polymers/chemistry , Animals , Cell Adhesion , Cell Proliferation , Endosomes/metabolism , HCT116 Cells , Humans , Mice, Inbred BALB C , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , PTEN Phosphohydrolase/metabolism , beta Catenin/metabolism
8.
Nat Commun ; 10(1): 1899, 2019 04 23.
Article En | MEDLINE | ID: mdl-31015397

Nanoparticles can potentially stimulate tumour microenvironments to elicit antitumour immunity. Herein, we demonstrate effective immunotherapy of colorectal cancer via systemic delivery of an immunostimulatory chemotherapeutic combination in nanoscale coordination polymer (NCP) core-shell particles. Oxaliplatin and dihydroartemesinin have contrasting physicochemical properties but strong synergy in reactive oxygen species (ROS) generation and anticancer activity. The combined ROS generation is harnessed for immune activation to synergize with an anti-PD-L1 antibody for the treatment of murine colorectal cancer tumours. The favourable biodistribution and tumour uptake of NCPs and the absence of peripheral neuropathy allow for repeated dosing to afford 100% tumour eradication. The involvement of innate and adaptive immune systems elicit strong and long lasting antitumour immunity which prevents tumour formation when cured mice are challenged with cancer cells. The intrinsically biodegradable, well tolerated, and systemically available immunostimulatory NCP promises to enter clinical testing as an immunotherapy against colorectal cancer.


Adenocarcinoma/therapy , Cancer Vaccines/pharmacology , Colorectal Neoplasms/therapy , Immunologic Factors/pharmacology , Nanoparticles/administration & dosage , Polymers/administration & dosage , Adaptive Immunity/drug effects , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Artemisinins/pharmacokinetics , Artemisinins/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cancer Vaccines/chemical synthesis , Cancer Vaccines/pharmacokinetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Drug Compounding/methods , Humans , Immunity, Innate/drug effects , Immunologic Factors/chemical synthesis , Immunologic Factors/pharmacokinetics , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Neoplasm Transplantation , Oxaliplatin/pharmacokinetics , Oxaliplatin/pharmacology , Polymers/chemical synthesis , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/agonists , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Survival Analysis , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
9.
Matter ; 1(5): 1331-1353, 2019 Nov 06.
Article En | MEDLINE | ID: mdl-32832885

Checkpoint blockade immunotherapy (CBI) is effective in promoting a systemic immune response against some metastatic tumors. The reliance on the pre-existing immune environment of the tumor, however, limits the efficacy of CBI on a broad spectrum of cancers. Herein, we report the design of a novel nanoscale metal-organic layer (nMOL), Hf-MOL, for effective treatment of local tumors by enabling radiotherapy-radiodynamic therapy (RT-RDT) with low-dose X-rays and, when in combination with an immune checkpoint inhibitor, regression of metastatic tumors by re-activating anti-tumor immunity and inhibiting myeloid-derived suppressor cells. Owing to the reduced dimensionality, nMOLs allow facile diffusion of reactive oxygen species and exhibit superior RT-RDT effects. The synergy of Hf-MOL-enabled RT-RDT immune activation and anti-programmed death ligand 1 (anti-PD-L1) CBI led to robust abscopal effects on a series of bilateral models of colon, head and neck, and breast cancers and significant anti-metastatic effects on an orthotopic model of breast cancer.

10.
Adv Mater ; 30(37): e1707634, 2018 Sep.
Article En | MEDLINE | ID: mdl-29971835

Nanotechnology has played an important role in drug delivery and biomedical imaging over the past two decades. In particular, nanoscale metal-organic frameworks (nMOFs) are emerging as an important class of biomedically relevant nanomaterials due to their high porosity, multifunctionality, and biocompatibility. The high porosity of nMOFs allows for the encapsulation of exceptionally high payloads of therapeutic and/or imaging cargoes while the building blocks-both ligands and the secondary building units (SBUs)-can be utilized to load drugs and/or imaging agents via covalent attachment. The ligands and SBUs of nMOFs can also be functionalized for surface passivation or active targeting at overexpressed biomarkers. The metal ions or metal clusters on nMOFs also render them viable candidates as contrast agents for magnetic resonance imaging, computed tomography, or other imaging modalities. This review article summarizes recent progress on nMOF designs and their exploration in biomedical areas. First, the therapeutic applications of nMOFs, based on four distinct drug loading strategies, are discussed, followed by a summary of nMOF designs for imaging and biosensing. The review is concluded by exploring the fundamental challenges facing nMOF-based therapeutic, imaging, and biosensing agents. This review hopefully can stimulate interdisciplinary research at the intersection of MOFs and biomedicine.


Metal-Organic Frameworks/chemistry , Contrast Media , Drug Delivery Systems , Nanostructures
11.
Nat Commun ; 9(1): 2351, 2018 06 15.
Article En | MEDLINE | ID: mdl-29907739

Checkpoint blockade immunotherapy enhances systemic antitumor immune response by targeting T cell inhibitory pathways; however, inadequate T cell infiltration has limited its anticancer efficacy. Radiotherapy (RT) has local immunomodulatory effects that can alter the microenvironment of irradiated tumors to synergize with immune checkpoint blockade. However, even with high doses of radiation, RT has rarely elicited systemic immune responses. Herein, we report the design of two porous Hf-based nanoscale metal-organic frameworks (nMOFs) as highly effective radioenhancers that significantly outperform HfO2, a clinically investigated radioenhancer in vitro and in vivo. Importantly, the combination of nMOF-mediated low-dose RT with an anti-programmed death-ligand 1 antibody effectively extends the local therapeutic effects of RT to distant tumors via abscopal effects. Our work establishes the feasibility of combining nMOF-mediated RT with immune checkpoint blockade to elicit systemic antitumor immunity in non-T cell-inflamed tumor phenotypes without normal tissue toxicity, promising to broaden the application of checkpoint blockade immunotherapy.


B7-H1 Antigen/metabolism , Immunotherapy/methods , Metal-Organic Frameworks/pharmacology , Nanotechnology/methods , Radiotherapy/methods , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Combined Modality Therapy , Humans , Hydroxyl Radical , Immunosuppression Therapy , Lymphocytes/cytology , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Necrosis , Neoplasm Transplantation , Signal Transduction
12.
Nat Biomed Eng ; 2(8): 600-610, 2018 08.
Article En | MEDLINE | ID: mdl-31015630

Checkpoint blockade immunotherapy relies on energized cytotoxic T cells attacking tumour tissue systemically. However, for many cancers, the reliance on T cell infiltration leads to low response rates. Conversely, radiotherapy has served as a powerful therapy for local tumours over the past 100 years, yet is rarely sufficient to cause systemic tumour rejection. Here, we describe a treatment strategy that combines nanoscale metal-organic framework (nMOF)-enabled radiotherapy-radiodynamic therapy with checkpoint blockade immunotherapy for both local and systemic tumour elimination. In mouse models of breast and colorectal cancer, intratumorally injected nMOFs treated with low doses of X-ray irradiation led to the eradication of local tumours and, when loaded with an inhibitor of the immune checkpoint molecule indoleamine 2,3-dioxygenase, the irradiated nMOFs led to consistent abscopal responses that rejected distal tumours. By combining the advantages of local radiotherapy and systemic tumour rejection via synergistic X-ray-induced in situ vaccination and indoleamine 2,3-dioxygenase inhibition, nMOFs may overcome some of the limitations of checkpoint blockade in cancer treatment.


Immunotherapy/methods , Metal-Organic Frameworks/chemistry , Nanostructures/chemistry , X-Ray Therapy/methods , Animals , Antineoplastic Agents , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Combined Modality Therapy , Humans , Metal-Organic Frameworks/pharmacology , Mice , Nanomedicine
13.
Mol Ther ; 25(7): 1588-1595, 2017 07 05.
Article En | MEDLINE | ID: mdl-28457664

Multiple therapeutic agents are typically used in concert to effectively control metastatic tumors. Recently, we described microRNAs that are associated with the oligometastatic state, in which a limited number of metastatic tumors progress to more favorable outcomes. Here, we report the effective delivery of an oligometastatic microRNA (miR-655-3p) to colorectal liver metastases using nanoscale coordination polymers (NCPs). The NCPs demonstrated a targeted and prolonged distribution of microRNAs to metastatic liver tumors. Tumor-targeted microRNA miR-655-3p suppressed tumor growth when co-delivered with oxaliplatin, suggesting additive or synergistic interactions between microRNAs and platinum drugs. This is the first known example of systemically administered nanoparticles delivering an oligometastatic microRNA to advanced metastatic liver tumors and demonstrating tumor-suppressive effects. Our results suggest a potential therapeutic strategy for metastatic liver disease by the co-delivery of microRNAs and conventional cytotoxic agents using tumor-specific NCPs.


Antineoplastic Agents/pharmacology , Colorectal Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Liver Neoplasms/therapy , MicroRNAs/genetics , Nanostructures/administration & dosage , Organoplatinum Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Cholesterol/chemistry , Cholesterol/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Dihydroxyphenylalanine/chemistry , Dihydroxyphenylalanine/metabolism , Disease Models, Animal , Drug Carriers , Drug Synergism , Female , HCT116 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Mice , Mice, Nude , MicroRNAs/administration & dosage , MicroRNAs/metabolism , Nanostructures/chemistry , Organoplatinum Compounds/chemistry , Oxaliplatin , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
14.
J Am Chem Soc ; 138(51): 16686-16695, 2016 12 28.
Article En | MEDLINE | ID: mdl-27976881

An effective, nontoxic, tumor-specific immunotherapy is the ultimate goal in the battle against cancer, especially the metastatic disease. Checkpoint blockade-based immunotherapies have been shown to be extraordinarily effective but benefit only the minority of patients whose tumors have been pre-infiltrated by T cells. Here, we show that Zn-pyrophosphate (ZnP) nanoparticles loaded with the photosensitizer pyrolipid (ZnP@pyro) can kill tumor cells upon irradiation with light directly by inducing apoptosis and/or necrosis and indirectly by disrupting tumor vasculature and increasing tumor immunogenicity. Furthermore, immunogenic ZnP@pyro photodynamic therapy (PDT) treatment sensitizes tumors to checkpoint inhibition mediated by a PD-L1 antibody, not only eradicating the primary 4T1 breast tumor but also significantly preventing metastasis to the lung. The abscopal effects on both 4T1 and TUBO bilateral syngeneic mouse models further demonstrate that ZnP@pyro PDT treatment combined with anti-PD-L1 results in the eradication of light-irradiated primary tumors and the complete inhibition of untreated distant tumors by generating a systemic tumor-specific cytotoxic T cell response. These findings indicate that nanoparticle-mediated PDT can potentiate the systemic efficacy of checkpoint blockade immunotherapies by activating the innate and adaptive immune systems in tumor microenvironment.


Breast Neoplasms/immunology , Breast Neoplasms/therapy , Diphosphates/chemistry , Immunotherapy , Nanoparticles/chemistry , Photochemotherapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Breast Neoplasms/pathology , Cell Line, Tumor , Chlorophyll/analogs & derivatives , Chlorophyll/chemistry , Combined Modality Therapy , Humans , Light , Lipids/chemistry , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Necrosis , Neoplasm Metastasis , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Zinc/chemistry
15.
Nat Commun ; 7: 12499, 2016 08 17.
Article En | MEDLINE | ID: mdl-27530650

Advanced colorectal cancer is one of the deadliest cancers, with a 5-year survival rate of only 12% for patients with the metastatic disease. Checkpoint inhibitors, such as the antibodies inhibiting the PD-1/PD-L1 axis, are among the most promising immunotherapies for patients with advanced colon cancer, but their durable response rate remains low. We herein report the use of immunogenic nanoparticles to augment the antitumour efficacy of PD-L1 antibody-mediated cancer immunotherapy. Nanoscale coordination polymer (NCP) core-shell nanoparticles carry oxaliplatin in the core and the photosensitizer pyropheophorbide-lipid conjugate (pyrolipid) in the shell (NCP@pyrolipid) for effective chemotherapy and photodynamic therapy (PDT). Synergy between oxaliplatin and pyrolipid-induced PDT kills tumour cells and provokes an immune response, resulting in calreticulin exposure on the cell surface, antitumour vaccination and an abscopal effect. When combined with anti-PD-L1 therapy, NCP@pyrolipid mediates regression of both light-irradiated primary tumours and non-irradiated distant tumours by inducing a strong tumour-specific immune response.


Coordination Complexes/chemistry , Immunotherapy , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/immunology , Photochemotherapy , Polymers/chemistry , Animals , Apoptosis/drug effects , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Chlorophyll/analogs & derivatives , Cytotoxicity, Immunologic , Disease Models, Animal , Drug Compounding , Immunity , Inflammation/pathology , Lipids/chemistry , Male , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/ultrastructure , Neoplasms/pathology , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Tissue Distribution
16.
J Am Chem Soc ; 138(38): 12502-10, 2016 09 28.
Article En | MEDLINE | ID: mdl-27575718

Photodynamic therapy (PDT) can destroy local tumors and minimize normal tissue damage, but is ineffective at eliminating metastases. Checkpoint blockade immunotherapy has enjoyed recent success in the clinic, but only elicits limited rates of systemic antitumor response for most cancers due to insufficient activation of the host immune system. Here we describe a treatment strategy that combines PDT by a new chlorin-based nanoscale metal-organic framework (nMOF), TBC-Hf, and a small-molecule immunotherapy agent that inhibits indoleamine 2,3-dioxygenase (IDO), encapsulated in the nMOF channels to induce systemic antitumor immunity. The synergistic combination therapy achieved effective local and distant tumor rejection in colorectal cancer models. We detected increased T cell infiltration in the tumor microenvironment after activation of the immune system with the combination of IDO inhibition by the small-molecule immunotherapy agent and immunogenic cell death induced by PDT. We also elucidated the underlying immunological mechanisms and revealed compensatory roles of neutrophils and B cells in presenting tumor-associated antigens to T cells in this combination therapy. We believe that nMOF-enabled PDT has the potential to significantly enhance checkpoint blockade cancer immunotherapy, affording clinical benefits for the treatment of many difficult-to-treat cancers.


Colorectal Neoplasms/drug therapy , Immunotherapy/methods , Metal Nanoparticles/chemistry , Metal-Organic Frameworks/chemistry , Photochemotherapy , Porphyrins/chemistry , Animals , Colorectal Neoplasms/metabolism , Drug Delivery Systems , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplastic Cells, Circulating , Singlet Oxygen/metabolism
17.
Sci Rep ; 5: 12941, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-26251280

Transgenic expression of I-E molecules prevents diabetes in NOD mice. So far, the precise role of these non-classical MHC II molecules remains elusive. Here, we showed that transgenic expression of I-E(k) alpha 16 molecule in NOD mice selectively reduced Th17 cells in the thymus and pancreatic draining lymph nodes. The reduction in Th17 cells was associated with both attenuated IL-6 production and decreased activation of macrophages. Mechanistically, transgenic expression of the I-E molecule diminished expression of intracellular classical MHC II molecule and led to impaired TLR4-mediated signaling. In contrast to classical MHC II molecule, this non-classical MHC II molecule negatively regulates the inflammatory responses of macrophages. Altogether, our study reveals a novel regulatory role of I-E molecules in modulating inflammatory immune responses.


Macrophage Activation/immunology , Macrophages/immunology , Th17 Cells/immunology , Animals , Inflammation/immunology , Interleukin-6/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred NOD , Pancreas/immunology , Signal Transduction/immunology , Thymus Gland/immunology , Toll-Like Receptor 4/immunology , Trans-Activators
18.
J Virol ; 89(21): 10860-7, 2015 Nov.
Article En | MEDLINE | ID: mdl-26292317

UNLABELLED: Coxsackievirus A16 (CVA16) is one of the major etiological agents of hand, foot, and mouth disease (HFMD) in children. The host defense mechanisms against CVA16 infection remain almost entirely unknown. Unlike previous observations with enterovirus 71 (EV71) infection, here we show that gamma interferon (IFN-γ) or invariant NK T cell deficiency does not affect disease development or the survival of CVA16-infected mice. In contrast, type I interferon receptor deficiency resulted in the development of more severe disease in mice, and the mice had a lower survival rate than wild-type mice. Similarly, a deficiency of Toll-like receptor 3 (TLR3) and TRIF, but not other pattern recognition receptors, led to the decreased survival of CVA16-infected mice. TLR3-TRIF signaling was indispensable for the induction of type I interferons during CVA16 infection in mice and protected young mice from disease caused by the infection. In particular, TRIF-mediated immunity was critical for preventing CVA16 replication in the neuronal system before disease occurred. IFN-ß treatment was also found to compensate for TRIF deficiency in mice and decreased the disease severity in and mortality of CVA16-infected mice. Altogether, type I interferons induced by TLR3-TRIF signaling mediate protective immunity against CVA16 infection. These findings may shed light on therapeutic strategies to combat HFMD caused by CVA16 infection. IMPORTANCE: Hand, foot, and mouth disease (HFMD) is a major threat to public health in the Asia-Pacific region. Both CVA16 and EV71 are major pathogens that are responsible for HFMD. The majority of research efforts have focused on the more virulent EV71, but little has been done with CVA16. Thus far, host immune responses to CVA16 infection have not yet been elucidated. The present study discovered an initial molecular mechanism underlying host protective immunity against CVA16 infection, providing the first explanation for why CVA16 and EV71 cause different clinical outcomes upon infection of humans. Therefore, different therapeutic strategies should be developed to treat HFMD cases caused by these two viruses.


Adaptor Proteins, Vesicular Transport/metabolism , Coxsackievirus Infections/prevention & control , Interferon Type I/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Vesicular Transport/deficiency , Animals , Coxsackievirus Infections/drug therapy , DNA Primers/genetics , Dendritic Cells/immunology , Flow Cytometry , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 3/deficiency
19.
PLoS Pathog ; 11(1): e1004613, 2015 Jan.
Article En | MEDLINE | ID: mdl-25615690

Enterovirus 71 (EV71) is the most virulent pathogen among enteroviruses that cause hand, foot and mouth disease in children but rarely in adults. The mechanisms that determine the age-dependent susceptibility remain largely unclear. Here, we found that the paucity of invariant natural killer T (iNKT) cells together with immaturity of the immune system was related to the susceptibility of neonatal mice to EV71 infection. iNKT cells were crucial antiviral effector cells to protect young mice from EV71 infection before their adaptive immune systems were fully mature. EV71 infection led to activation of iNKT cells depending on signaling through TLR3 but not other TLRs. Surprisingly, iNKT cell activation during EV71 infection required TLR3 signaling in macrophages, but not in dendritic cells (DCs). Mechanistically, interleukin (IL)-12 and endogenous CD1d-restricted antigens were both required for full activation of iNKT cells. Furthermore, CD1d-deficiency led to dramatically increased viral loads in central nervous system and more severe disease in EV71-infected mice. Altogether, our results suggest that iNKT cells may be involved in controlling EV71 infection in children when their adaptive immune systems are not fully developed, and also imply that iNKT cells might be an intervention target for treating EV71-infected patients.


Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Immunity, Cellular , Macrophages/immunology , Natural Killer T-Cells/immunology , Toll-Like Receptor 3/physiology , Animals , Cells, Cultured , Enterovirus Infections/genetics , Humans , Immunity, Cellular/genetics , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Natural Killer T-Cells/metabolism , Signal Transduction/immunology
20.
Pharm Res ; 32(3): 741-8, 2015 Mar.
Article En | MEDLINE | ID: mdl-23619596

PURPOSE: How HCV virus affects the function of dendritic cells (DCs) and their ability to induce CD4+ T cell response remains not fully understood. This study was done to elucidate the impact of HCV on the function of DCs and on DC's capability to induce CD4+ T-cell response. METHODS: Monocyte-derived DCs (MoDCs) were treated with cell-culture HCV (HCVcc). The effects of HCVcc on DC maturation, CD40L-induced DC maturation, and cytokine production and the capacity of DCs to induce Th cytokine production of allogeneic CD4+ T cells were evaluated. RESULTS: HCVcc exposure increased expression of both IL-6 and IL-10 by MoDCs. HCV-exposed MoDCs also selectively facilitated allogeneic CD4+ T cells to further produce Th17-related cytokines interleukin 1 (IL-1), IL-6, and IL-17A. Pretreatment of IL-17A inhibited HCV production in Huh7.5 cells, suggesting that induction of Th17 cells may be beneficial to host anti-HCV immunity. Paradoxically, induction of IL-10 expression and the failure of HCV-exposed MoDCs to facilitate other Th cell development may hinder the anti-viral immunity. CONCLUSIONS: This study highlights both the therapeutic potential of IL-17A in treating HCV infection and the cautious consideration of HCV-induced immunosuppression in DC-based therapy.


Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Hepacivirus/immunology , Hepatitis C/immunology , Hepatitis C/virology , Immune Tolerance , Inflammation Mediators/immunology , Th17 Cells/immunology , Th17 Cells/virology , Antiviral Agents/pharmacology , Autocrine Communication , CD40 Ligand/immunology , CD40 Ligand/pharmacology , Cell Differentiation , Cell Line , Coculture Techniques , Cytokines/metabolism , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Hepacivirus/drug effects , Hepatitis C/drug therapy , Host-Pathogen Interactions , Humans , Immune Tolerance/drug effects , Immunologic Memory , Inflammation Mediators/metabolism , Inflammation Mediators/pharmacology , Interleukin-17/immunology , Interleukin-17/pharmacology , Lymphocyte Activation , Signal Transduction , Th17 Cells/drug effects , Th17 Cells/metabolism , Time Factors
...