Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Immunol ; 210(11): 1687-1699, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37042681

ABSTRACT

Although CD4+CD25+FOXP3+ regulatory T (TREG) cells have been studied in patients with COVID-19, changes in the TREG cell population have not been longitudinally examined during the course of COVID-19. In this study, we longitudinally investigated the quantitative and qualitative changes in the TREG cell population in patients with COVID-19. We found that the frequencies of total TREG cells and CD45RA-FOXP3hi activated TREG cells were significantly increased 15-28 d postsymptom onset in severe patients, but not in mild patients. TREG cells from severe patients exhibited not only increased proliferation but also enhanced apoptosis, suggesting functional derangement of the TREG cell population during severe COVID-19. The suppressive functions of the TREG cell population did not differ between patients with severe versus mild COVID-19. The frequency of TREG cells inversely correlated with SARS-CoV-2-specific cytokine production by CD4+ T cells and their polyfunctionality in patients with mild disease, suggesting that TREG cells are major regulators of virus-specific CD4+ T cell responses during mild COVID-19. However, such correlations were not observed in patients with severe disease. Thus, in this study, we describe distinctive changes in the TREG cell population in patients with severe and mild COVID-19. Our study provides a deep understanding of host immune responses upon SARS-CoV-2 infection in regard to TREG cells.


Subject(s)
COVID-19 , T-Lymphocytes, Regulatory , Humans , SARS-CoV-2 , CD4-Positive T-Lymphocytes , Interleukin-2 Receptor alpha Subunit , Forkhead Transcription Factors
2.
Cancer Immunol Immunother ; 72(2): 371-384, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35902399

ABSTRACT

Immune checkpoint inhibitors are effective for advanced hepatocellular carcinoma (HCC), but there remains a need for peripheral blood biomarkers to predict the clinical response. Here, we analyzed the peripheral blood of 45 patients with advanced HCC who underwent nivolumab. During treatment, frequency of classical monocytes (CD14+CD16-) was increased on day 7, and the fold increase in the frequency on day 7 over day 0 (cMonocyteD7/D0) was significantly higher in patients with durable clinical benefit (DCB) than in patients with non-DCB (NDB). When we analyzed transcriptomes of classical monocytes, CD274, gene encoding PD-L1, was upregulated in NDB patients compared to DCB patients at day 7. Notably, gene signature of suppressive tumor-associated macrophages, or IL4l1+PD-L1+IDO1+ macrophages, was enriched after treatment in NDB patients, but not in DCB patients. Accordingly, the fold increase in the frequency of PD-L1+ classical monocytes at day 7 over day 0 (cMonocyte-PDL1D7/D0) was higher in NDB patients than DCB patients. The combined biomarker cMonocyteD7/D0/cMonocyte-PDL1D7/D0 was termed the "monocyte index", which was significantly higher in DCB patients than NDB patients. Moreover, the monocyte index was an independent prognostic factor for survival. Overall, our results suggest that early changes of circulating classical monocytes, represented as a monocyte index, could predict clinical outcomes of advanced HCC patients undergoing anti-PD-1 therapy.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Monocytes , Carcinoma, Hepatocellular/pathology , B7-H1 Antigen , Liver Neoplasms/pathology , Macrophages
3.
J Hepatol ; 77(4): 1059-1070, 2022 10.
Article in English | MEDLINE | ID: mdl-35644434

ABSTRACT

BACKGROUND & AIMS: The liver provides a unique niche of lymphocytes enriched with a large proportion of innate-like T cells. However, the heterogeneity and functional characteristics of the hepatic T-cell population remain to be fully elucidated. METHODS: We obtained liver sinusoidal mononuclear cells from the liver perfusate of healthy donors and recipients with HBV-associated chronic liver disease (CLD) during liver transplantation. We performed a CITE-seq analysis of liver sinusoidal CD45+ cells in combination with T cell receptor (TCR)-seq and flow cytometry to examine the phenotypes and functions of liver sinusoidal CD8+ T cells. RESULTS: We identified a distinct CD56hiCD161-CD8+ T-cell population characterized by natural killer (NK)-related gene expression and a uniquely restricted TCR repertoire. The frequency of these cells among the liver sinusoidal CD8+ T-cell population was significantly increased in patients with HBV-associated CLD. Although CD56hiCD161-CD8+ T cells exhibit weak responsiveness to TCR stimulation, CD56hiCD161-CD8+ T cells highly expressed various NK receptors, including CD94, killer immunoglobulin-like receptors, and NKG2C, and exerted NKG2C-mediated NK-like effector functions even in the absence of TCR stimulation. In addition, CD56hiCD161-CD8+ T cells highly respond to innate cytokines, such as IL-12/18 and IL-15, in the absence of TCR stimulation. We validated the results from liver sinusoidal CD8+ T cells using intrahepatic CD8+ T cells obtained from liver tissues. CONCLUSIONS: In summary, the current study found a distinct CD56hiCD161-CD8+ T-cell population characterized by NK-like activation via TCR-independent NKG2C ligation. Further studies are required to elucidate the roles of liver sinusoidal CD56hiCD161-CD8+ T cells in immune responses to microbial pathogens or liver immunopathology. LAY SUMMARY: The role of different immune cell populations in the liver is becoming an area of increasing interest. Herein, we identified a distinct T-cell population that had features similar to those of natural killer (NK) cells - a type of innate immune cell. This distinct population was expanded in the livers of patients with chronic liver disease and could thus have pathogenic relevance.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-15 , Immunoglobulins , Interleukin-12 , Liver , Receptors, Antigen, T-Cell
4.
Development ; 143(13): 2334-43, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27161150

ABSTRACT

During spinal cord development, motor neuron (MN) axons exit the spinal cord ventrally, although the molecular basis for this process remains poorly understood. STAM1 and HRS form a complex involved with endosomal targeting of cargo proteins, including the chemokine receptor CXCR4. Interestingly, the absence of CXCR4 signaling in spinal MNs is known to result in improper extension of the axons into the dorsal side of the spinal cord. Here, we report that the MN-specific ISL1-LHX3 complex directly transactivates the Stam1 gene and that STAM1 functions in determining the ventral spinal MN axonal projections. STAM1 is co-expressed with HRS in embryonic spinal MNs, and knockdown of STAM1 in the developing chick spinal cord results in downregulation of CXCR4 expression, accompanied by dorsally projecting motor axons. Interestingly, overexpression of STAM1 or CXCR4 also results in dorsal projection of motor axons, suggesting that proper CXCR4 protein level is necessary for the ventral motor axon trajectory. Our results reveal a crucial regulatory axis for the ventral axonal trajectory of developing spinal MNs, consisting of the ISL1-LHX3 complex, STAM1 and CXCR4.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Motor Neurons/metabolism , Phosphoproteins/metabolism , Spinal Cord/cytology , Adaptor Proteins, Signal Transducing/genetics , Animals , Axons/metabolism , Base Sequence , Binding Sites , Chickens , Chromatin Immunoprecipitation , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/metabolism , Female , HEK293 Cells , Humans , LIM-Homeodomain Proteins/metabolism , Mice, Knockout , Phosphoproteins/genetics , Protein Binding , Rats , Receptors, CXCR4/metabolism , Response Elements/genetics , Transcription Factors/metabolism
6.
PLoS Genet ; 10(4): e1004280, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24763339

ABSTRACT

The establishment of correct neurotransmitter characteristics is an essential step of neuronal fate specification in CNS development. However, very little is known about how a battery of genes involved in the determination of a specific type of chemical-driven neurotransmission is coordinately regulated during vertebrate development. Here, we investigated the gene regulatory networks that specify the cholinergic neuronal fates in the spinal cord and forebrain, specifically, spinal motor neurons (MNs) and forebrain cholinergic neurons (FCNs). Conditional inactivation of Isl1, a LIM homeodomain factor expressed in both differentiating MNs and FCNs, led to a drastic loss of cholinergic neurons in the developing spinal cord and forebrain. We found that Isl1 forms two related, but distinct types of complexes, the Isl1-Lhx3-hexamer in MNs and the Isl1-Lhx8-hexamer in FCNs. Interestingly, our genome-wide ChIP-seq analysis revealed that the Isl1-Lhx3-hexamer binds to a suite of cholinergic pathway genes encoding the core constituents of the cholinergic neurotransmission system, such as acetylcholine synthesizing enzymes and transporters. Consistently, the Isl1-Lhx3-hexamer directly coordinated upregulation of cholinergic pathways genes in embryonic spinal cord. Similarly, in the developing forebrain, the Isl1-Lhx8-hexamer was recruited to the cholinergic gene battery and promoted cholinergic gene expression. Furthermore, the expression of the Isl1-Lhx8-complex enabled the acquisition of cholinergic fate in embryonic stem cell-derived neurons. Together, our studies show a shared molecular mechanism that determines the cholinergic neuronal fate in the spinal cord and forebrain, and uncover an important gene regulatory mechanism that directs a specific neurotransmitter identity in vertebrate CNS development.


Subject(s)
Cholinergic Neurons/metabolism , LIM-Homeodomain Proteins/metabolism , Prosencephalon/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Line , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Gene Expression/genetics , Gene Regulatory Networks/genetics , HEK293 Cells , Humans , LIM-Homeodomain Proteins/genetics , Mice , Motor Neurons/metabolism , Rats , Transcription Factors/genetics
7.
Biochem Biophys Res Commun ; 472(3): 471-6, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26845353

ABSTRACT

Sclerostin, encoded by the Sost gene, is mainly produced by osteocytes in bone and antagonizes the Wnt/ß-catenin signaling pathway, which is a requisite for bone formation. Currently, human anti-sclerostin antibodies are being tested in phase III clinical trials. In addition, serum sclerostin levels are reported to be associated with bone mineral density and fracture risk in normal individuals; however, the correlation between serum sclerostin and bone mass remains controversial. To study the effects of the continuous exposure of exogenous sclerostin on bone, a ΦC31 integrase system, which has the characteristics of site-specificity and efficiency, was applied for the delivery of the Sost gene in this study. We injected Sost-attB plasmid with or without ΦC31 integrase plasmid into the mouse tail vein using a hydrodynamic-based method. The site-specific integration of the Sost gene into the mouse genome was confirmed by examining a pseudo-attP site on the hepatic genomic DNA. Sclerostin was expressed in the hepatocytes, secreted into the blood flow, and maintained at high concentrations in the mice with both Sost-attB plasmid and ΦC31 integrase plasmid injections, which was observed by serial measurement. Moreover, the mice with long-term high levels of serum sclerostin showed trabecular bone loss on micro-CT analysis. Peripheral B cell populations were not affected. Our results suggested that sclerostin could be expressed in the liver and sustained successfully at high levels in the blood by using the ΦC31 integrase system, leading to trabecular bone loss. These findings may help to further ascertain the effects of sclerostin introduced exogenously on the skeleton.


Subject(s)
Bacteriophages/enzymology , Bone and Bones/metabolism , Glycoproteins/blood , Glycoproteins/genetics , Integrases/genetics , Osteoporosis/classification , Adaptor Proteins, Signal Transducing , Animals , Bacteriophages/genetics , Bone and Bones/pathology , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred ICR , Osteoporosis/pathology , Transfection/methods , Up-Regulation/genetics
8.
J Infect Public Health ; 16(10): 1643-1649, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37597451

ABSTRACT

BACKGROUND: Despite the low measles antibody positivity rate among young healthcare workers (HCWs) who have previously received two doses of a measles-containing vaccine (MCV), whether an additional dose of MCV acts as a booster remains unknown. Thus, we aimed to evaluate the immune responses to a third dose of MCV in young HCWs. METHODS: Hospital-wide measles seroprevalence was assessed using enzyme-linked immunosorbent assay (ELISA). The immunogenicity of a third dose of MCV was determined in young seronegative HCWs (born between 1986 and 1997) who had previously received a two-dose measles vaccination. RESULTS: A total of 3033 (92.6%) HCWs had anti-measles immunoglobulin G. The lowest seropositivity rate was observed in HCWs aged 20-24 years (87.7%). In this group, HCWs who received a third dose of MCV had higher seropositivity than those who received a second dose (89.5% vs. 75.4%). A third dose of MCV was administered to 18 HCWs who did not have anti-measles IgG despite two doses. Neutralizing antibody titers increased significantly 4 weeks after the third vaccination. Although neutralizing antibody titers decreased 1 year post vaccination, 17 (94.4%) HCWs had medium (121-900 mIU/mL) or high (>900 mIU/mL) levels. Furthermore, the third dose of MCV increased the measles virus-specific T-cell effector function. CONCLUSIONS: The third dose of MCV induced a strong immune response against measles in young seronegative HCWs who had previously received a two-dose measles vaccination.


Subject(s)
Measles Vaccine , Measles , Humans , Seroepidemiologic Studies , Measles/prevention & control , Health Personnel , Vaccination , Antibodies, Neutralizing
9.
Clin Cancer Res ; 29(9): 1794-1806, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36716293

ABSTRACT

PURPOSE: Regulatory T cells (Tregs) exert immunosuppressive functions and hamper antitumor immune responses in the tumor microenvironment. Understanding the heterogeneity of intratumoral Tregs, and how it changes with tumor progression, will provide clues regarding novel target molecules of Treg-directed therapies. EXPERIMENTAL DESIGN: From 42 patients with renal cell carcinoma and 5 patients with ovarian cancer, immune cells from tumor and peripheral blood were isolated. We performed multicolor flow cytometry and RNA-sequencing to characterize the phenotypes and heterogeneity of intratumoral Tregs. In vitro functional assays were performed to evaluate suppressive capacity of Tregs and effect of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1)-mediated depletion. The CT26 tumor model was used to evaluate the association between intratumoral Tregs and tumor growth, and examine the in vivo role of CEACAM1+ intratumoral Tregs on antitumor immunity. RESULTS: We found that CEACAM1 was selectively expressed on intratumoral Tregs, whereas its expression on peripheral Tregs or other immune cells was low. The CEACAM1+ intratumoral Tregs accumulated with tumor progression, whereas the CEACAM1- subset did not. Notably, we found that CEACAM1 marked intratumoral Tregs that exhibited highly suppressive and activated phenotypes with substantial clonal expansion. Depletion of CEACAM1-expressing cells from tumor-infiltrating leukocytes led to increased effector functions of tumor-infiltrating T cells. Moreover, CEACAM1+ cell depletion further enhanced anti-PD-1-mediated reinvigoration of exhausted CD8+ T cells. CONCLUSIONS: CEACAM1 marks highly suppressive subset of intratumoral Tregs, and can be a target for selective depletion of intratumoral Tregs. These results may inform future studies on CEACAM1-mediated depletion in patients with cancer.


Subject(s)
Neoplasms , T-Lymphocytes, Regulatory , Humans , Cell Adhesion Molecule-1/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Neoplasms/genetics , Neoplasms/therapy , Tumor Microenvironment
10.
Sci Immunol ; 7(74): eabn8390, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36026440

ABSTRACT

Despite being a standard treatment option in breast cancer, immune checkpoint inhibitors (ICIs) are only efficacious for a subset of patients. To gain a better understanding of the antitumor immune response in breast cancer, we examined the heterogeneity of CD8+ T cells in tumors, metastatic lymph nodes (mLNs), and peripheral blood from patients with early breast cancer (n = 131). Among tissue-resident memory CD8+ T (TRM) cells, including virus- and tumor-specific CD8+ T cells, CD39 expression was observed in a tumor-specific and exhausted subpopulation in both tumors and mLNs. CD39+ TRM cells from tumors and mLNs exhibited a phenotypic similarity and clonally overlapped with each other. Moreover, tumor or mLN CD39+ TRM cells clonally overlapped with CD39- TRM and non-TRM cells in the same compartment, implying a tissue-specific differentiation process. These inter-subpopulationally overlapping CD39+ TRM clonotypes were frequently detected among effector memory CD8+ T cells in peripheral blood, suggesting a systemic clonal overlap. CD39+ TRM cell enrichment was heterogeneous among molecular subtypes of breast cancer, which is associated with the different role of antitumor immune responses in each subtype. In vitro blockade of PD-1 and/or CTLA-4 effectively restored proliferation of CD39+ TRM cells and enhanced cytokine production by CD8+ T cells from tumors or mLNs, particularly in the presence of CD39+ TRM enrichment. This suggests that CD39+ TRM cells have a capacity for functional restoration upon ICI treatment. Thus, our study indicates that CD39+ TRM cells with a clonal overlap across compartments are key players in antitumor immunity in breast cancer.


Subject(s)
Breast Neoplasms , CD8-Positive T-Lymphocytes , Female , Humans , Immunotherapy , Lymph Nodes
11.
FEBS Open Bio ; 10(4): 619-626, 2020 04.
Article in English | MEDLINE | ID: mdl-32129006

ABSTRACT

Tobacco etch virus (TEV) protease is a 27-kDa catalytic domain of the polyprotein nuclear inclusion a (NIa) in TEV, which recognizes the specific amino acid sequence ENLYFQG/S and cleaves between Q and G/S. Despite its substrate specificity, its use is limited by its autoinactivation through self-cleavage and poor solubility during purification. It was previously reported that T17S/N68D/I77V mutations improve the solubility and yield of TEV protease and S219 mutations provide protection against self-cleavage. In this study, we isolated TEV proteases with S219N and S219V mutations in the background of T17S, N68D, and I77V without the inclusion body, and measured their enzyme kinetics. The kcat of two isolated S219N and S219V mutants in the background of T17S, N68D, and I77V mutations was highly increased compared to that of the control, and S219N was twofold faster than S219V without Km change. This result indicates that combination of these mutations can further enhance TEV activity.


Subject(s)
Catalytic Domain/genetics , Endopeptidases/chemistry , Endopeptidases/genetics , Mutation , Potyvirus/enzymology , Viral Proteins/chemistry , Viral Proteins/genetics , Amino Acid Sequence , Endopeptidases/metabolism , Enzyme Activation , Escherichia coli/genetics , Escherichia coli/metabolism , Inclusion Bodies , Kinetics , Plasmids/genetics , Solubility , Substrate Specificity , Viral Proteins/metabolism
12.
Elife ; 82019 07 15.
Article in English | MEDLINE | ID: mdl-31305241

ABSTRACT

During spinal cord development, Sonic hedgehog (Shh), secreted from the floor plate, plays an important role in the production of motor neurons by patterning the ventral neural tube, which establishes MN progenitor identity. It remains unknown, however, if Shh signaling plays a role in generating columnar diversity of MNs that connect distinct target muscles. Here, we report that Shh, expressed in MNs, is essential for the formation of lateral motor column (LMC) neurons in vertebrate spinal cord. This novel activity of Shh is mediated by its downstream effector ARHGAP36, whose expression is directly induced by the MN-specific transcription factor complex Isl1-Lhx3. Furthermore, we found that AKT stimulates the Shh activity to induce LMC MNs through the stabilization of ARHGAP36 proteins. Taken together, our data reveal that Shh, secreted from MNs, plays a crucial role in generating MN diversity via a regulatory axis of Shh-AKT-ARHGAP36 in the developing mouse spinal cord.


Subject(s)
Body Patterning , GTPase-Activating Proteins/metabolism , Hedgehog Proteins/metabolism , Motor Neurons/metabolism , Signal Transduction , Animals , Cell Differentiation , Chickens , Cyclic AMP-Dependent Protein Kinases/metabolism , HEK293 Cells , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Protein Stability , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism
13.
Nat Commun ; 6: 7718, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26212498

ABSTRACT

While microRNAs have emerged as an important component of gene regulatory networks, it remains unclear how microRNAs collaborate with transcription factors in the gene networks that determines neuronal cell fate. Here we show that in the developing spinal cord, the expression of miR-218 is directly upregulated by the Isl1-Lhx3 complex, which drives motor neuron fate. Inhibition of miR-218 suppresses the generation of motor neurons in both chick neural tube and mouse embryonic stem cells, suggesting that miR-218 plays a crucial role in motor neuron differentiation. Results from unbiased RISC-trap screens, in vivo reporter assays and overexpression studies indicated that miR-218 directly represses transcripts that promote developmental programs for interneurons. In addition, we found that miR-218 activity is required for Isl1-Lhx3 to effectively induce motor neurons and suppress interneuron fates. Together our results reveal an essential role of miR-218 as a downstream effector of the Isl1-Lhx3 complex in establishing motor neuron identity.


Subject(s)
Gene Expression Regulation, Developmental , LIM-Homeodomain Proteins/genetics , MicroRNAs/genetics , Motor Neurons/cytology , Neural Tube/embryology , Neurogenesis/genetics , Spinal Cord/embryology , Transcription Factors/genetics , Animals , Chick Embryo , Electroporation , HEK293 Cells , Humans , LIM-Homeodomain Proteins/metabolism , Mice , Mouse Embryonic Stem Cells , Neural Tube/cytology , Real-Time Polymerase Chain Reaction , Spinal Cord/cytology , Transcription Factors/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL