Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 288, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436577

ABSTRACT

Vaccines and therapeutics are urgently needed for the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here, we screen human monoclonal antibodies (mAb) targeting the receptor binding domain (RBD) of the viral spike protein via antibody library constructed from peripheral blood mononuclear cells of a convalescent patient. The CT-P59 mAb potently neutralizes SARS-CoV-2 isolates including the D614G variant without antibody-dependent enhancement effect. Complex crystal structure of CT-P59 Fab/RBD shows that CT-P59 blocks interaction regions of RBD for angiotensin converting enzyme 2 (ACE2) receptor with an orientation that is notably different from previously reported RBD-targeting mAbs. Furthermore, therapeutic effects of CT-P59 are evaluated in three animal models (ferret, hamster, and rhesus monkey), demonstrating a substantial reduction in viral titer along with alleviation of clinical symptoms. Therefore, CT-P59 may be a promising therapeutic candidate for COVID-19.


Subject(s)
Antibodies, Neutralizing/pharmacology , COVID-19 Drug Treatment , Protein Binding/drug effects , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/drug effects , Angiotensin-Converting Enzyme 2/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chlorocebus aethiops , Disease Models, Animal , Female , Ferrets , Humans , Leukocytes, Mononuclear , Macaca mulatta , Male , Mesocricetus , Models, Molecular , Protein Conformation , Spike Glycoprotein, Coronavirus/chemistry , Vero Cells
2.
PLoS One ; 15(7): e0236172, 2020.
Article in English | MEDLINE | ID: mdl-32726321

ABSTRACT

There are several broadly neutralizing monoclonal antibodies that neutralize influenza viruses with different mechanisms from traditional polyclonal antibodies induced by vaccination. CT149, which is one of the broadly neutralizing antibodies, was also previously reported to neutralize group 2 and some of group 1 influenza viruses (13 out of 13 tested group 2 viruses and 5 out of 11 group 1 viruses). In this study, we developed another antibody with the aim of compensating partial coverage of CT149 against group 1 influenza viruses. CT120 was screened among different antibody candidates and mixed with CT149. Importantly, although the binding sites of CT120 and CT149 are close to each other, the two antibodies do not interfere. The mixture of CT120 and CT149, which we named as CT-P27, showed broad efficacy by neutralizing 37 viruses from 11 different subtypes, of both group 1 and 2 influenza A viruses. Moreover, CT-P27 showed in vivo therapeutic efficacy, long prophylactic potency, and synergistic effect with oseltamivir in influenza virus-challenged mouse models. Our findings provide a novel therapeutic opportunity for more efficient treatment of influenza.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibody Specificity/immunology , Antigens, Viral/immunology , Hemagglutination/immunology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A virus , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Neutralization Tests , Vaccination
3.
Mol Cell Proteomics ; 14(10): 2722-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26229149

ABSTRACT

Macrophages are crucial in controlling infectious agents and tissue homeostasis. Macrophages require a wide range of functional capabilities in order to fulfill distinct roles in our body, one being rapid and robust immune responses. To gain insight into macrophage plasticity and the key regulatory protein networks governing their specific functions, we performed quantitative analyses of the proteome and phosphoproteome of murine primary GM-CSF and M-CSF grown bone marrow derived macrophages (GM-BMMs and M-BMMs, respectively) using the latest isobaric tag based tandem mass tag (TMT) labeling and liquid chromatography-tandem mass spectrometry (LC-MS/MS). Strikingly, metabolic processes emerged as a major difference between these macrophages. Specifically, GM-BMMs show significant enrichment of proteins involving glycolysis, the mevalonate pathway, and nitrogen compound biosynthesis. This evidence of enhanced glycolytic capability in GM-BMMs is particularly significant regarding their pro-inflammatory responses, because increased production of cytokines upon LPS stimulation in GM-BMMs depends on their acute glycolytic capacity. In contrast, M-BMMs up-regulate proteins involved in endocytosis, which correlates with a tendency toward homeostatic functions such as scavenging cellular debris. Together, our data describes a proteomic network that underlies the pro-inflammatory actions of GM-BMMs as well as the homeostatic functions of M-BMMs.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/metabolism , Animals , Bone Marrow Cells/cytology , Cell Line, Tumor , Cytokines/metabolism , Glycolysis , Male , Mice, Inbred C57BL , Microspheres , Phagocytosis , Proteome/metabolism , Proteomics
4.
PLoS One ; 10(2): e0118203, 2015.
Article in English | MEDLINE | ID: mdl-25680189

ABSTRACT

Macrophages play important roles in defense against infection, as well as in homeostasis maintenance. Thus alterations of macrophage function can have unexpected pathological results. Cyclooxygenase (COX) inhibitors are widely used to relieve pain, but the effects of long-term usage on macrophage function remain to be elucidated. Using bone marrow-derived macrophage culture and long-term COX inhibitor treatments in BALB/c mice and zebrafish, we showed that chronic COX inhibition drives macrophages into an inflammatory state. Macrophages differentiated in the presence of SC-560 (COX-1 inhibitor), NS-398 (COX-2 inhibitor) or indomethacin (COX-1/2 inhibitor) for 7 days produced more TNFα or IL-12p70 with enhanced p65/IκB phosphoylation. YmI and IRF4 expression was reduced significantly, indicative of a more inflammatory phenotype. We further observed that indomethacin or NS-398 delivery accelerated zebrafish death rates during LPS induced sepsis. When COX inhibitors were released over 30 days from an osmotic pump implant in mice, macrophages from peritoneal cavities and adipose tissue produced more TNFα in both the basal state and under LPS stimulation. Consequently, indomethacin-exposed mice showed accelerated systemic inflammation after LPS injection. Our findings suggest that macrophages exhibit a more inflammatory phenotype when COX activities are chronically inhibited.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Inflammation/metabolism , Macrophages/drug effects , Macrophages/enzymology , Phenotype , Prostaglandin-Endoperoxide Synthases/metabolism , Animals , Biomarkers , Cell Differentiation/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Female , Inflammation/immunology , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Zebrafish
5.
J Biol Chem ; 289(9): 6225-35, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24421315

ABSTRACT

PDGF-C, which is abundant in the malignant breast tumor microenvironment, plays an important role in cell growth and survival. Because tumor-associated macrophages (TAMs) contribute to cancer malignancy, macrophage survival mechanisms are an attractive area of research into controlling tumor progression. In this study, we investigated PDGF-C-mediated signaling pathways involved in anti-apoptotic effects in macrophages. We found that the human malignant breast cancer cell line MDA-MB-231 produced high quantities of PDGF-C, whereas benign MCF-7 cells did not. Recombinant PDGF-C induced PDGF receptor α chain phosphorylation, followed by Akt and Bad phosphorylation in THP-1-derived macrophages. MDA-MB-231 culture supernatants also activated macrophage PDGF-Rα. PDGF-C prevented staurosporine-induced macrophage apoptosis by inhibiting the activation of caspase-3, -7, -8, and -9 and cleavage of poly(ADP-ribose) polymerase. Finally, TAMs isolated from the PDGF-C knockdown murine breast cancer cell line 4T1 and PDGF-C knockdown MDA-MB-231-derived tumor mass showed higher rates of apoptosis than the respective WT controls. Collectively, our results suggest that tumor cell-derived PDGF-C enhances TAM survival, promoting tumor malignancy.


Subject(s)
Apoptosis , Lymphokines/metabolism , Macrophages/metabolism , Neoplasms/metabolism , Platelet-Derived Growth Factor/metabolism , Signal Transduction , bcl-Associated Death Protein/metabolism , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Humans , Lymphokines/genetics , Macrophages/pathology , Male , Neoplasms/genetics , Neoplasms/physiopathology , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins c-akt , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Staurosporine/pharmacology , bcl-Associated Death Protein/genetics
6.
Mol Cancer ; 12: 71, 2013 Jul 09.
Article in English | MEDLINE | ID: mdl-23835085

ABSTRACT

BACKGROUND: With high throughput screening, novel therapeutic agents can be efficiently identified. Unfortunately, researchers only resort to in vitro cell viability assays for screening of anticancer drugs for retinoblastoma, the most common intraocular cancer in the childhood. Current available animal models of retinoblastoma require more than 2 weeks for tumour formation and the investigation of the efficacy of therapeutic agents. In this study, we established a novel orthotopic transplantation model of retinoblastoma in zebrafish as an in vivo animal model for screening of anticancer drugs. METHODS: We injected retinoblastoma cells into the vitreous cavity of zebrafish at 48 hours after fertilization. Eyeballs of zebrafish were scanned daily under the confocal laser microscope, and the tumor population was quantitatively analyzed by measuring the mean intensity of green fluorescent protein (GFP). Transplanted retinoblastoma cells were isolated to perform further analyses including Western blotting and reverse transcriptase-polymerase chain reaction to confirm that retinoblastoma cells maintained their characteristics as tumor cells even after transplantation and further isolation. To figure out the potential of this model for screening of anticancer drugs, zebrafish were cultured in Ringer's solution containing carboplatin and melphalan after the injection of retinoblastoma cells. RESULTS: The degree of the tumor population was dependent on the number of retinoblastoma cells injected and maintained stably for at least 4 days. Transplanted retinoblastoma cells maintain their proliferative potential and characteristics as retinoblastoma cells after isolation. Interestingly, systemic application of carboplatin and melphalan demonstrated significant reduction in the tumor population, which could be quantitatively analyzed by the estimation of the mean intensity of GFP. CONCLUSIONS: This orthotopic retinoblastoma model in zebrafish is expected to be utilized for the screening of anticancer drugs for the treatment of retinoblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays , Humans , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Zebrafish
7.
PLoS One ; 8(5): e63451, 2013.
Article in English | MEDLINE | ID: mdl-23667623

ABSTRACT

Tumor cells are often associated with abundant macrophages that resemble the alternatively activated M2 subset. Tumor-associated macrophages (TAMs) inhibit anti-tumor immune responses and promote metastasis. Cyclooxygenase-2 (COX-2) inhibition is known to prevent breast cancer metastasis. This study hypothesized that COX-2 inhibition affects TAM characteristics potentially relevant to tumor cell metastasis. We found that the specific COX-2 inhibitor, etodolac, inhibited human M2 macrophage differentiation, as determined by decreased CD14 and CD163 expressions and increased TNFα production. Several key metastasis-related mediators, such as vascular endothelial growth factor-A, vascular endothelial growth factor-C, and matrix metalloproteinase-9, were inhibited in the presence of etodolac as compared to untreated M2 macrophages. Murine bone marrow derived M2 macrophages also showed enhanced surface MHCII IA/IE and CD80, CD86 expressions together with enhanced TNFα expressions with etodolac treatment during differentiation. Using a BALB/c breast cancer model, we found that etodolac significantly reduced lung metastasis, possibly due to macrophages expressing increased IA/IE and TNFα, but decreased M2 macrophage-related genes expressions (Ym1, TGFß). In conclusion, COX-2 inhibition caused loss of the M2 macrophage characteristics of TAMs and may assist prevention of breast cancer metastasis.


Subject(s)
Cell Differentiation/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Lung Neoplasms/secondary , Macrophages/pathology , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Etodolac/pharmacology , Female , Humans , Inflammation Mediators/metabolism , Lung Neoplasms/pathology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Mammary Neoplasms, Animal/immunology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Phenotype
8.
Toxicol Lett ; 216(1): 65-71, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23178550

ABSTRACT

The development of novel alternative testing methods is required to identify the sensitizing capacity of chemicals as a replacement for animal experimentation. We aimed to evaluate in vitro assays as screening tools for detecting skin sensitizers. The murine epidermal keratinocyte cell line HEL-30 was exposed to 16 relevant skin sensitizers and 6 skin irritants. The dose causing 75% cell viability (CV(75)) measured by an MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was chosen as a highest dose and three more doses (0.5×, 0.1×, and 0.01× of CV(75)) were tested. As parameters, levels of interleukin 1α (IL-1α), macrophage inflammatory protein 2 (MIP-2), IL-6, and IL-18 production were measured using 4 different doses. The accuracy of detecting sensitizers or irritants by IL-1α or MIP-2 alone was exactly same: 75% (12 out of 16) for sensitizers, 83% (5 out of 6) for irritants, and overall 77% (17 out of 22). However, combination of IL-1α and MIP-2 showed better accuracy: 94% (15 out of 16), 67% (4 out of 6), and overall 86% (19 out of 22). IL-6 and IL-18 could not differentiate sensitizers from irritants. This study suggests that the combination of pro-inflammatory cytokines IL-1α and MIP-2 in murine HEL-30 cells can be a reliable in vitro method for identifying chemicals that may act as skin sensitizers.


Subject(s)
Animal Testing Alternatives/methods , Chemokine CXCL2/metabolism , Interleukin-1alpha/metabolism , Irritants/toxicity , Keratinocytes/drug effects , Skin/drug effects , Animals , Cell Adhesion/drug effects , Cell Survival/drug effects , Chemokine CXCL2/genetics , Gene Expression Regulation/drug effects , Interleukin-1alpha/genetics , Keratinocytes/metabolism , Lymph Nodes/drug effects , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...