Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
1.
Cell ; 162(3): 493-504, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26189681

ABSTRACT

Dengue is the most common vector-borne viral disease, causing nearly 400 million infections yearly. Currently there are no approved therapies. Antibody epitopes that elicit weak humoral responses may not be accessible by conventional B cell panning methods. To demonstrate an alternative strategy to generating a therapeutic antibody, we employed a non-immunodominant, but functionally relevant, epitope in domain III of the E protein, and engineered by structure-guided methods an antibody directed to it. The resulting antibody, Ab513, exhibits high-affinity binding to, and broadly neutralizes, multiple genotypes within all four serotypes. To assess therapeutic relevance of Ab513, activity against important human clinical features of dengue was investigated. Ab513 mitigates thrombocytopenia in a humanized mouse model, resolves vascular leakage, reduces viremia to nearly undetectable levels, and protects mice in a maternal transfer model of lethal antibody-mediated enhancement. The results demonstrate that Ab513 may reduce the public health burden from dengue.


Subject(s)
Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/chemistry , Dengue Virus/physiology , Dengue/therapy , Immunodominant Epitopes/chemistry , Amino Acid Sequence , Animals , Dengue/immunology , Dengue/virology , Dengue Virus/immunology , Disease Models, Animal , Mice , Models, Molecular , Molecular Sequence Data , Phagocytosis , Protein Engineering , Receptors, Fc/immunology , Sequence Alignment
2.
Nature ; 630(8016): 360-367, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38778109

ABSTRACT

Implanted biomaterials and devices face compromised functionality and efficacy in the long term owing to foreign body reactions and subsequent formation of fibrous capsules at the implant-tissue interfaces1-4. Here we demonstrate that an adhesive implant-tissue interface can mitigate fibrous capsule formation in diverse animal models, including rats, mice, humanized mice and pigs, by reducing the level of infiltration of inflammatory cells into the adhesive implant-tissue interface compared to the non-adhesive implant-tissue interface. Histological analysis shows that the adhesive implant-tissue interface does not form observable fibrous capsules on diverse organs, including the abdominal wall, colon, stomach, lung and heart, over 12 weeks in vivo. In vitro protein adsorption, multiplex Luminex assays, quantitative PCR, immunofluorescence analysis and RNA sequencing are additionally carried out to validate the hypothesis. We further demonstrate long-term bidirectional electrical communication enabled by implantable electrodes with an adhesive interface over 12 weeks in a rat model in vivo. These findings may offer a promising strategy for long-term anti-fibrotic implant-tissue interfaces.


Subject(s)
Biocompatible Materials , Fibrosis , Foreign-Body Reaction , Prostheses and Implants , Tissue Adhesives , Animals , Female , Humans , Male , Mice , Rats , Abdominal Wall , Adsorption , Biocompatible Materials/chemistry , Colon , Electrodes, Implanted , Fibrosis/pathology , Fibrosis/prevention & control , Foreign-Body Reaction/prevention & control , Foreign-Body Reaction/pathology , Heart , Lung , Mice, Inbred C57BL , Organ Specificity , Polymerase Chain Reaction , Rats, Sprague-Dawley , Stomach , Swine , Time Factors , Tissue Adhesives/chemistry , Fluorescent Antibody Technique , Reproducibility of Results , Sequence Analysis, RNA
3.
Cell ; 156(3): 590-602, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485462

ABSTRACT

Therapy-resistant microenvironments represent a major barrier toward effective elimination of disseminated malignancies. Here, we show that select microenvironments can underlie resistance to antibody-based therapy. Using a humanized model of treatment refractory B cell leukemia, we find that infiltration of leukemia cells into the bone marrow rewires the tumor microenvironment to inhibit engulfment of antibody-targeted tumor cells. Resistance to macrophage-mediated killing can be overcome by combination regimens involving therapeutic antibodies and chemotherapy. Specifically, the nitrogen mustard cyclophosphamide induces an acute secretory activating phenotype (ASAP), releasing CCL4, IL8, VEGF, and TNFα from treated tumor cells. These factors induce macrophage infiltration and phagocytic activity in the bone marrow. Thus, the acute induction of stress-related cytokines can effectively target cancer cells for removal by the innate immune system. This synergistic chemoimmunotherapeutic regimen represents a potent strategy for using conventional anticancer agents to alter the tumor microenvironment and promote the efficacy of targeted therapeutics.


Subject(s)
Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Tumor Microenvironment , Animals , Cyclophosphamide/therapeutic use , Cytokines/immunology , Drug Resistance, Neoplasm , Heterografts , Humans , Immunity, Innate , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrophages/immunology , Mice , Neoplasm Transplantation
4.
Cell ; 144(2): 296-309, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241896

ABSTRACT

Though many individual transcription factors are known to regulate hematopoietic differentiation, major aspects of the global architecture of hematopoiesis remain unknown. Here, we profiled gene expression in 38 distinct purified populations of human hematopoietic cells and used probabilistic models of gene expression and analysis of cis-elements in gene promoters to decipher the general organization of their regulatory circuitry. We identified modules of highly coexpressed genes, some of which are restricted to a single lineage but most of which are expressed at variable levels across multiple lineages. We found densely interconnected cis-regulatory circuits and a large number of transcription factors that are differentially expressed across hematopoietic states. These findings suggest a more complex regulatory system for hematopoiesis than previously assumed.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Hematopoiesis , Transcription Factors/metabolism , Gene Expression Profiling , Humans
5.
Proc Natl Acad Sci U S A ; 120(52): e2311752120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38134199

ABSTRACT

The emergence of highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that are resistant to the current COVID-19 vaccines highlights the need for continued development of broadly protective vaccines for the future. Here, we developed two messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines, TU88mCSA and ALCmCSA, using the ancestral SARS-CoV-2 spike sequence, optimized 5' and 3' untranslated regions (UTRs), and LNP combinations. Our data showed that these nanocomplexes effectively activate CD4+ and CD8+ T cell responses and humoral immune response and provide complete protection against WA1/2020, Omicron BA.1 and BQ.1 infection in hamsters. Critically, in Omicron BQ.1 challenge hamster models, TU88mCSA and ALCmCSA not only induced robust control of virus load in the lungs but also enhanced protective efficacy in the upper respiratory airways. Antigen-specific immune analysis in mice revealed that the observed cross-protection is associated with superior UTRs [Carboxylesterase 1d (Ces1d)/adaptor protein-3ß (AP3B1)] and LNP formulations that elicit robust lung tissue-resident memory T cells. Strong protective effects of TU88mCSA or ALCmCSA against both WA1/2020 and VOCs suggest that this mRNA-LNP combination can be a broadly protective vaccine platform in which mRNA cargo uses the ancestral antigen sequence regardless of the antigenic drift. This approach could be rapidly adapted for clinical use and timely deployment of vaccines against emerging and reemerging VOCs.


Subject(s)
COVID-19 Vaccines , COVID-19 , Cricetinae , Animals , Humans , Mice , RNA, Messenger/genetics , COVID-19 Vaccines/genetics , mRNA Vaccines , SARS-CoV-2/genetics , COVID-19/prevention & control , 3' Untranslated Regions , Antibodies, Neutralizing , Antibodies, Viral
6.
Proc Natl Acad Sci U S A ; 119(25): e2122379119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696582

ABSTRACT

Acute myeloid leukemia (AML) remains a therapeutic challenge, and a paucity of tumor-specific targets has significantly hampered the development of effective immune-based therapies. Recent paradigm-changing studies have shown that natural killer (NK) cells exhibit innate memory upon brief activation with IL-12 and IL-18, leading to cytokine-induced memory-like (CIML) NK cell differentiation. CIML NK cells have enhanced antitumor activity and have shown promising results in early phase clinical trials in patients with relapsed/refractory AML. Here, we show that arming CIML NK cells with a neoepitope-specific chimeric antigen receptor (CAR) significantly enhances their antitumor responses to nucleophosphmin-1 (NPM1)-mutated AML while avoiding off-target toxicity. CIML NK cells differentiated from peripheral blood NK cells were efficiently transduced to express a TCR-like CAR that specifically recognizes a neoepitope derived from the cytosolic oncogenic NPM1-mutated protein presented by HLA-A2. These CAR CIML NK cells displayed enhanced activity against NPM1-mutated AML cell lines and patient-derived leukemic blast cells. CAR CIML NK cells persisted in vivo and significantly improved AML outcomes in xenograft models. Single-cell RNA sequencing and mass cytometry analyses identified up-regulation of cell proliferation, protein folding, immune responses, and major metabolic pathways in CAR-transduced CIML NK cells, resulting in tumor-specific, CAR-dependent activation and function in response to AML target cells. Thus, efficient arming of CIML NK cells with an NPM1-mutation-specific TCR-like CAR substantially improves their innate antitumor responses against an otherwise intracellular mutant protein. These preclinical findings justify evaluating this approach in clinical trials in HLA-A2+ AML patients with NPM1c mutations.


Subject(s)
Immunologic Memory , Immunological Memory Cells , Immunotherapy, Adoptive , Killer Cells, Natural , Leukemia, Myeloid, Acute , Nucleophosmin , Receptors, Chimeric Antigen , HLA-A2 Antigen/immunology , Humans , Immunological Memory Cells/immunology , Immunological Memory Cells/transplantation , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mutation , Nucleophosmin/genetics , Nucleophosmin/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology
7.
J Immunol ; 208(9): 2227-2238, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35396222

ABSTRACT

Malignant ascites is a common clinical problem in ovarian cancer. NK cells are present in the ascites, but their antitumor activity is inhibited. The underlying mechanisms of the inhibition have yet to be fully elucidated. Using an Fcγ receptor-mediated NK cell activation assay, we show that ascites from ovarian cancer patients potently inhibits NK cell activation. Part of the inhibitory activity is mediated by CA125, a mucin 16 fragment shed from ovarian cancer tumors. Moreover, transcriptional analyses by RNA sequencing reveal upregulation of genes involved in multiple metabolic pathways but downregulation of genes involved in cytotoxicity and signaling pathways in NK cells purified from ovarian cancer patient ascites. Transcription of genes involved in cytotoxicity pathways are also downregulated in NK cells from healthy donors after in vitro treatment with ascites or with a CA125-enriched protein fraction. These results show that ascites and CA125 inhibit antitumor activity of NK cells at transcriptional levels by suppressing expression of genes involved in NK cell activation and cytotoxicity. Our findings shed light on the molecular mechanisms by which ascites inhibits the activity of NK cells and suggest possible approaches to reactivate NK cells for ovarian cancer immunotherapy.


Subject(s)
Ascites , CA-125 Antigen , Killer Cells, Natural , Ovarian Neoplasms , Ascites/metabolism , CA-125 Antigen/genetics , CA-125 Antigen/metabolism , Female , Humans , Killer Cells, Natural/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Transcriptional Activation
8.
Annu Rev Biomed Eng ; 24: 85-109, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35231177

ABSTRACT

mRNA vaccines have brought about a great revolution in the vaccine fields owing to their simplicity and adaptability in antigen design, potential to induce both humoral and cell-mediated immune responses and demonstrated high efficacy, and rapid and low-cost production by using the same manufacturing platform for different mRNA vaccines. Multiple mRNA vaccines have been investigated for both infectious diseases and cancers, showing significant superiority to other types of vaccines. Although great success of mRNA vaccines has been achieved in the control of the coronavirus disease 2019 pandemic, there are still multiple challenges for the future development of mRNA vaccines. In this review, the most recent developments of mRNA vaccines against both infectious diseases and cancers are summarized for an overview of this field. Moreover, the challenges are also discussed on the basis of these developments.


Subject(s)
COVID-19 , Communicable Diseases , Neoplasms , COVID-19/prevention & control , Humans , RNA, Messenger/genetics , Vaccines, Synthetic , mRNA Vaccines
9.
J Immunol ; 202(6): 1885-1894, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30710044

ABSTRACT

Development of targeted cancer therapy requires a thorough understanding of mechanisms of tumorigenesis as well as mechanisms of action of therapeutics. This is challenging because by the time patients are diagnosed with cancer, early events of tumorigenesis have already taken place. Similarly, development of cancer immunotherapies is hampered by a lack of appropriate small animal models with autologous human tumor and immune system. In this article, we report the development of a mouse model of human acute myeloid leukemia (AML) with autologous immune system for studying early events of human leukemogenesis and testing the efficacy of immunotherapeutics. To develop such a model, human hematopoietic stem/progenitor cells (HSPC) are transduced with lentiviruses expressing a mutated form of nucleophosmin (NPM1), referred to as NPM1c. Following engraftment into immunodeficient mice, transduced HSPCs give rise to human myeloid leukemia, whereas untransduced HSPCs give rise to human immune cells in the same mice. The de novo AML, with CD123+ leukemic stem or initiating cells (LSC), resembles NPM1c+ AML from patients. Transcriptional analysis of LSC and leukemic cells confirms similarity of the de novo leukemia generated in mice with patient leukemia and suggests Myc as a co-operating factor in NPM1c-driven leukemogenesis. We show that a bispecific conjugate that binds both CD3 and CD123 eliminates CD123+ LSCs in a T cell-dependent manner both in vivo and in vitro. These results demonstrate the utility of the NPM1c+ AML model with an autologous immune system for studying early events of human leukemogenesis and for evaluating efficacy and mechanism of immunotherapeutics.


Subject(s)
Carcinogenesis , Leukemia, Myeloid , Nuclear Proteins , Xenograft Model Antitumor Assays , Animals , Hematopoietic Stem Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Nucleophosmin
10.
PLoS Pathog ; 14(10): e1007298, 2018 10.
Article in English | MEDLINE | ID: mdl-30286211

ABSTRACT

Natural killer (NK) cells provide the first line of defense against malaria parasite infection. However, the molecular mechanisms through which NK cells are activated by parasites are largely unknown, so is the molecular basis underlying the variation in NK cell responses to malaria infection in the human population. Here, we compared transcriptional profiles of responding and non-responding NK cells following exposure to Plasmodium-infected red blood cells (iRBCs) and identified MDA5, a RIG-I-like receptor involved in sensing cytosolic RNAs, to be differentially expressed. Knockout of MDA5 in responding human NK cells by CRISPR/cas9 abolished NK cell activation, IFN-γ secretion, lysis of iRBCs. Similarly, inhibition of TBK1/IKKε, an effector molecule downstream of MDA5, also inhibited activation of responding NK cells. Conversely, activation of MDA5 by liposome-packaged poly I:C restored non-responding NK cells to lyse iRBCs. We further show that microvesicles containing large parasite RNAs from iRBCs activated NK cells by fusing with NK cells. These findings suggest that NK cells are activated through the MDA5 pathway by parasite RNAs that are delivered to the cytoplasm of NK cells by microvesicles from iRBCs. The difference in MDA5 expression between responding and non-responding NK cells following exposure to iRBCs likely contributes to the variation in NK cell responses to malaria infection in the human population.


Subject(s)
Cell-Derived Microparticles/immunology , Erythrocytes/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Killer Cells, Natural/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , CRISPR-Cas Systems , Cells, Cultured , Cytoplasm/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Interferon-Induced Helicase, IFIH1/antagonists & inhibitors , Interferon-Induced Helicase, IFIH1/genetics , Killer Cells, Natural/metabolism , Killer Cells, Natural/parasitology , Lymphocyte Activation , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/isolation & purification
11.
J Virol ; 91(14)2017 07 15.
Article in English | MEDLINE | ID: mdl-28468882

ABSTRACT

Dengue is an acute febrile illness caused by dengue virus (DENV) and a major cause of morbidity and mortality in tropical and subtropical regions of the world. The lack of an appropriate small-animal model of dengue infection has greatly hindered the study of dengue pathogenesis and the development of therapeutics. In this study, we conducted mass spectrometry-based serum metabolic profiling from a model using humanized mice (humice) with DENV serotype 2 infection at 0, 3, 7, 14, and 28 days postinfection (dpi). Forty-eight differential metabolites were identified, including fatty acids, purines and pyrimidines, acylcarnitines, acylglycines, phospholipids, sphingolipids, amino acids and derivatives, free fatty acids, and bile acid. These metabolites showed a reversible-change trend-most were significantly perturbed at 3 or 7 dpi and returned to control levels at 14 or 28 dpi, indicating that the metabolites might serve as prognostic markers of the disease in humice. The major perturbed metabolic pathways included purine and pyrimidine metabolism, fatty acid ß-oxidation, phospholipid catabolism, arachidonic acid and linoleic acid metabolism, sphingolipid metabolism, tryptophan metabolism, phenylalanine metabolism, lysine biosynthesis and degradation, and bile acid biosynthesis. Most of these disturbed pathways are similar to our previous metabolomics findings in a longitudinal cohort of adult human dengue patients across different infection stages. Our analyses revealed the commonalities of host responses to DENV infection between humice and humans and suggested that humice could be a useful small-animal model for the study of dengue pathogenesis and the development of dengue therapeutics.IMPORTANCE Dengue virus is the most widespread arbovirus, causing an estimated 390 million dengue infections worldwide every year. There is currently no effective treatment for the disease, and the lack of an appropriate small-animal model of dengue infection has greatly increased the challenges in the study of dengue pathogenesis and the development of therapeutics. Metabolomics provides global views of small-molecule metabolites and is a useful tool for finding metabolic pathways related to disease processes. Here, we conducted a serum metabolomics study on a model using humanized mice with dengue infection that had significant levels of human platelets, monocytes/macrophages, and hepatocytes. Forty-eight differential metabolites were identified, and the underlying perturbed metabolic pathways are quite similar to the pathways found to be altered in dengue patients in previous metabolomics studies, indicating that humanized mice could be a highly relevant small-animal model for the study of dengue pathogenesis and the development of dengue therapeutics.


Subject(s)
Dengue/pathology , Metabolome , Serum/chemistry , Animals , Disease Models, Animal , Mass Spectrometry , Metabolomics , Mice , Mice, SCID , Time Factors
12.
Blood ; 128(10): 1396-407, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27338099

ABSTRACT

Epstein-Barr virus (EBV) is an oncovirus associated with several human malignancies including posttransplant lymphoproliferative disease in immunosuppressed patients. We show here that anti-EBV T-cell receptor-like monoclonal antibodies (TCR-like mAbs) E1, L1, and L2 bound to their respective HLA-A*0201-restricted EBV peptides EBNA1562-570, LMP1125-133, and LMP2A426-434 with high affinities and specificities. These mAbs recognized endogenously presented targets on EBV B lymphoblastoid cell lines (BLCLs), but not peripheral blood mononuclear cells, from which they were derived. Furthermore, these mAbs displayed similar binding activities on several BLCLs, despite inherent heterogeneity between different donor samples. A single weekly administration of the naked mAbs reduced splenomegaly, liver tumor spots, and tumor burden in BLCL-engrafted immunodeficient NOD-SCID/Il2rg(-/-) mice. In particular, mice that were treated with the E1 mAb displayed a delayed weight loss and significantly prolonged survival. In vitro, these TCR-like mAbs induced early apoptosis of BLCLs, thereby enhancing their Fc-dependent phagocytic uptake by macrophages. These data provide evidence for TCR-like mAbs as potential therapeutic modalities to target EBV-associated diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B-Lymphocytes/immunology , HLA-A2 Antigen/immunology , Herpesvirus 4, Human/immunology , Liver Neoplasms/prevention & control , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Flow Cytometry , Humans , Leukocytes, Mononuclear/immunology , Liver Neoplasms/immunology , Liver Neoplasms/virology , Mice , Mice, Inbred NOD , Mice, SCID , Phagocytosis/immunology
13.
J Immunol ; 195(4): 1591-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26163589

ABSTRACT

Recall responses by memory CD8 T cells are impaired in the absence of CD4 T cells. Although several mechanisms have been proposed, the molecular basis is still largely unknown. Using a local influenza virus infection in the respiratory tract and the lung of CD4(-/-) mice, we show that memory CD8 T cell impairment is limited to the lungs and the lung-draining lymph nodes, where viral Ags are unusually persistent and abundant in these mice. Persistent Ag exposure results in prolonged activation of the AKT-mTORC1 pathway in Ag-specific CD8 T cells, favoring their development into effector memory T cells at the expense of central memory T cells, and inhibition of mTORC1 by rapamycin largely corrects the impairment by promoting central memory T cell development. The findings suggest that the prolonged AKT-mTORC1 activation driven by persistent Ag is a critical mechanism underlying the impaired memory CD8 T cell development and responses in the absence of CD4 T cells.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Immunophenotyping , Lymphocyte Activation/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Phenotype , Sirolimus/pharmacology
14.
J Immunol ; 194(10): 4737-49, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25862818

ABSTRACT

Scavenger receptor class B, member 2 (SCARB2) is essential for endosome biogenesis and reorganization and serves as a receptor for both ß-glucocerebrosidase and enterovirus 71. However, little is known about its function in innate immune cells. In this study, we show that, among human peripheral blood cells, SCARB2 is most highly expressed in plasmacytoid dendritic cells (pDCs), and its expression is further upregulated by CpG oligodeoxynucleotide stimulation. Knockdown of SCARB2 in pDC cell line GEN2.2 dramatically reduces CpG-induced type I IFN production. Detailed studies reveal that SCARB2 localizes in late endosome/lysosome of pDCs, and knockdown of SCARB2 does not affect CpG oligodeoxynucleotide uptake but results in the retention of TLR9 in the endoplasmic reticulum and an impaired nuclear translocation of IFN regulatory factor 7. The IFN-I production by TLR7 ligand stimulation is also impaired by SCARB2 knockdown. However, SCARB2 is not essential for influenza virus or HSV-induced IFN-I production. These findings suggest that SCARB2 regulates TLR9-dependent IFN-I production of pDCs by mediating endosomal translocation of TLR9 and nuclear translocation of IFN regulatory factor 7.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-7/metabolism , Interferon Type I/biosynthesis , Lysosomal Membrane Proteins/immunology , Receptors, Scavenger/immunology , Toll-Like Receptor 9/metabolism , Blotting, Western , Cells, Cultured , Dendritic Cells/metabolism , Endosomes/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Humans , Lysosomal Membrane Proteins/metabolism , Protein Transport/physiology , Real-Time Polymerase Chain Reaction , Receptors, Scavenger/metabolism
15.
Proc Natl Acad Sci U S A ; 111(4): 1479-84, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474774

ABSTRACT

Immunodeficient mouse-human chimeras provide a powerful approach to study host-specific pathogens, such as Plasmodium falciparum that causes human malaria. Supplementation of immunodeficient mice with human RBCs supports infection by human Plasmodium parasites, but these mice lack the human immune system. By combining human RBC supplementation and humanized mice that are optimized for human immune cell reconstitution, we have developed RBC-supplemented, immune cell-optimized humanized (RICH) mice that support multiple cycles of P. falciparum infection. Depletion of human natural killer (NK) cells, but not macrophages, in RICH mice results in a significant increase in parasitemia. Further studies in vitro show that NK cells preferentially interact with infected RBCs (iRBCs), resulting in the activation of NK cells and the elimination of iRBCs in a contact-dependent manner. We show that the adhesion molecule lymphocyte-associated antigen 1 is required for NK cell interaction with and elimination of iRBCs. Development of RICH mice and validation of P. falciparum infection should facilitate the dissection of human immune responses to malaria parasite infection and the evaluation of therapeutics and vaccines.


Subject(s)
Erythrocytes/parasitology , Killer Cells, Natural/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Animals , Cell Adhesion , Humans , Malaria, Falciparum/blood , Mice , Parasitemia/immunology
16.
Gut ; 65(10): 1744-53, 2016 10.
Article in English | MEDLINE | ID: mdl-26149491

ABSTRACT

OBJECTIVE: HCV infection affects millions of people worldwide, and many patients develop chronic infection leading to liver cancers. For decades, the lack of a small animal model that can recapitulate HCV infection, its immunopathogenesis and disease progression has impeded the development of an effective vaccine and therapeutics. We aim to provide a humanised mouse model for the understanding of HCV-specific human immune responses and HCV-associated disease pathologies. DESIGN: Recently, we have established human liver cells with a matched human immune system in NOD-scid Il2rg(-/-) (NSG) mice (HIL mice). These mice are infected with HCV by intravenous injection, and the pathologies are investigated. RESULTS: In this study, we demonstrate that HIL mouse is capable of supporting HCV infection and can present some of the clinical symptoms found in HCV-infected patients including hepatitis, robust virus-specific human immune cell and cytokine responses as well as liver fibrosis and cirrhosis. Similar to results obtained from the analysis of patient samples, the human immune cells, particularly T cells and macrophages, play critical roles during the HCV-associated liver disease development in the HIL mice. Furthermore, our model is demonstrated to be able to reproduce the therapeutic effects of human interferon alpha 2a antiviral treatment. CONCLUSIONS: The HIL mouse provides a model for the understanding of HCV-specific human immune responses and HCV-associated disease pathologies. It could also serve as a platform for antifibrosis and immune-modulatory drug testing.


Subject(s)
Disease Models, Animal , Hepatitis C, Chronic , Interferon-alpha/therapeutic use , Mice, Inbred NOD , Animals , Antiviral Agents/therapeutic use , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/physiopathology , Humans , Immunity, Cellular/immunology , Interferon alpha-2 , Mice , Recombinant Proteins/therapeutic use , Reproducibility of Results
17.
Biomacromolecules ; 16(1): 351-6, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25486335

ABSTRACT

A number of N-alkylated polyethylenimines (PEIs) were covalently attached to glass-slide surfaces, and their virucidal efficacies against three different strains of influenza viruses were examined quantitatively. The anti-influenza activities of the modified surfaces varied widely, with the most potent, immobilized N,N-hexyl,methyl-PEI and N,N-dodecyl,methyl-PEI, reducing the viral titer by over three logs (i.e., >99.9%). While the virucidal activities of the glass surfaces derivatized with N-alkylated PEIs displayed no discernible correlation with such surface properties as hydrophobicity, charge, protein affinity, roughness, adhesive interactions, and polymer-chain extension lengths, they exhibited a marginal correlation with the surface density of the quaternary ammonium group, as titrated by means of fluorescein binding. However, this correlation markedly improved (to the correlation coefficient of 0.97 with a two-tailed p value of 0.044) when the titration was instead carried out using a macromolecular conjugate, the dye coupled to the protein lysozyme, suggesting that the critical determinant of the virucidal activity is the density of the immobilized quaternary ammonium groups accessible to influenza virions.


Subject(s)
Antiviral Agents/pharmacology , Influenza A virus/drug effects , Polyethyleneimine/pharmacology , Virus Inactivation/drug effects , Antiviral Agents/chemistry , Glass , Humans , Hydrophobic and Hydrophilic Interactions , Influenza A virus/growth & development , Materials Testing , Polyethyleneimine/chemistry , Surface Properties , Viral Load/drug effects
18.
J Immunol ; 191(6): 3192-9, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23935193

ABSTRACT

Engraftment of human CD34⁺ hematopoietic stem/progenitor cells into immunodeficient mice leads to robust reconstitution of human T and B cells but not monocytes and macrophages. To identify the cause underlying the poor monocyte and macrophage reconstitution, we analyzed human myeloid cell development in humanized mice and found that it was blocked at the promonocyte stage in the bone marrow. Expression of human M-CSF or GM-CSF by hydrodynamic injection of cytokine-encoding plasmid completely abolished the accumulation of promonocytes in the bone marrow. M-CSF promoted the development of mature monocytes and tissue-resident macrophages whereas GM-CSF did not. Moreover, correlating with an increased human macrophages at the sites of infection, M-CSF-treated humanized mice exhibited an enhanced protection against influenza virus and Mycobacterium infection. Our study identifies the precise stage at which human monocyte/macrophage development is blocked in humanized mice and reveals overlapping and distinct functions of M-CSF and GM-CSF in human monocyte and macrophage development. The improved reconstitution and functionality of monocytes/macrophages in the humanized mice following M-CSF expression provide a superior in vivo system to investigate the role of macrophages in physiological and pathological processes.


Subject(s)
Cell Differentiation/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Monocyte-Macrophage Precursor Cells/cytology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Cell Differentiation/drug effects , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Humans , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Monocyte-Macrophage Precursor Cells/immunology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
19.
Proc Natl Acad Sci U S A ; 109(50): 20385-90, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23185023

ABSTRACT

Covalently conjugating multiple copies of the drug zanamivir (ZA; the active ingredient in Relenza) via a flexible linker to poly-l-glutamine (PGN) enhances the anti-influenza virus activity by orders of magnitude. In this study, we investigated the mechanisms of this phenomenon. Like ZA itself, the PGN-attached drug (PGN-ZA) binds specifically to viral neuraminidase and inhibits both its enzymatic activity and the release of newly synthesized virions from infected cells. Unlike monomeric ZA, however, PGN-ZA also synergistically inhibits early stages of influenza virus infection, thus contributing to the markedly increased antiviral potency. This inhibition is not caused by a direct virucidal effect, aggregation of viruses, or inhibition of viral attachment to target cells and the subsequent endocytosis; rather, it is a result of interference with intracellular trafficking of the endocytosed viruses and the subsequent virus-endosome fusion. These findings both rationalize the great anti-influenza potency of PGN-ZA and reveal that attaching ZA to a polymeric chain confers a unique mechanism of antiviral action potentially useful for minimizing drug resistance.


Subject(s)
Antiviral Agents/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Influenza, Human/drug therapy , Influenza, Human/virology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Zanamivir/analogs & derivatives , Animals , Antiviral Agents/chemistry , Dogs , Drug Synergism , Endocytosis/drug effects , Hemagglutinin Glycoproteins, Influenza Virus/drug effects , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N1 Subtype/ultrastructure , Madin Darby Canine Kidney Cells , Microscopy, Electron, Transmission , Neuraminidase/antagonists & inhibitors , Peptides/chemistry , Zanamivir/administration & dosage , Zanamivir/chemistry
20.
Proc Natl Acad Sci U S A ; 109(12): 4580-5, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22403068

ABSTRACT

Algorithms derived from measurements of short-peptide (8-10 mers) binding to class I MHC proteins suggest that the binding groove of a class I MHC protein, such as K(b), can bind well over 1 million different peptides with significant affinity (<500 nM), a level of ligand-binding promiscuity approaching the level of heat shock protein binding of unfolded proteins. MHC proteins can, nevertheless, discriminate between similar peptides and bind many of them with high (nanomolar) affinity. Some insights into this high-promiscuity/high-affinity behavior and its impact on immunodominant peptides in T-cell responses to some infections and vaccination are suggested by results obtained here from testing a model developed to predict the number of cell surface peptide-MHC complexes that form on cells exposed to extracellular (exogenous) peptides.


Subject(s)
Histocompatibility Antigens Class I/genetics , Animals , Binding Sites , Cell Membrane/metabolism , Endocytosis , Genes, MHC Class I , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Mice , Models, Biological , Ovalbumin/chemistry , Peptides/chemistry , Protein Binding , Protein Denaturation , Protein Folding , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL