Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 17(12): 1424-1435, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27695000

ABSTRACT

The final stages of restriction to the T cell lineage occur in the thymus after the entry of thymus-seeding progenitors (TSPs). The identity and lineage potential of TSPs remains unclear. Because the first embryonic TSPs enter a non-vascularized thymic rudiment, we were able to directly image and establish the functional and molecular properties of embryonic thymopoiesis-initiating progenitors (T-IPs) before their entry into the thymus and activation of Notch signaling. T-IPs did not include multipotent stem cells or molecular evidence of T cell-restricted progenitors. Instead, single-cell molecular and functional analysis demonstrated that most fetal T-IPs expressed genes of and had the potential to develop into lymphoid as well as myeloid components of the immune system. Moreover, studies of embryos deficient in the transcriptional regulator RBPJ demonstrated that canonical Notch signaling was not involved in pre-thymic restriction to the T cell lineage or the migration of T-IPs.


Subject(s)
Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Lymphoid Progenitor Cells/physiology , Myeloid Progenitor Cells/physiology , Receptors, Notch/metabolism , T-Lymphocytes/physiology , Thymus Gland/immunology , Animals , Cell Differentiation , Cell Lineage , Cell Movement , Cells, Cultured , Fetus , Gene Expression Regulation, Developmental , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
2.
Blood ; 142(19): 1622-1632, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37562000

ABSTRACT

A critical regulatory role of hematopoietic stem cell (HSC) vascular niches in the bone marrow has been implicated to occur through endothelial niche cell expression of KIT ligand. However, endothelial-derived KIT ligand is expressed in both a soluble and membrane-bound form and not unique to bone marrow niches, and it is also systemically distributed through the circulatory system. Here, we confirm that upon deletion of both the soluble and membrane-bound forms of endothelial-derived KIT ligand, HSCs are reduced in mouse bone marrow. However, the deletion of endothelial-derived KIT ligand was also accompanied by reduced soluble KIT ligand levels in the blood, precluding any conclusion as to whether the reduction in HSC numbers reflects reduced endothelial expression of KIT ligand within HSC niches, elsewhere in the bone marrow, and/or systemic soluble KIT ligand produced by endothelial cells outside of the bone marrow. Notably, endothelial deletion, specifically of the membrane-bound form of KIT ligand, also reduced systemic levels of soluble KIT ligand, although with no effect on stem cell numbers, implicating an HSC regulatory role primarily of soluble rather than membrane KIT ligand expression in endothelial cells. In support of a role of systemic rather than local niche expression of soluble KIT ligand, HSCs were unaffected in KIT ligand deleted bones implanted into mice with normal systemic levels of soluble KIT ligand. Our findings highlight the need for more specific tools to unravel niche-specific roles of regulatory cues expressed in hematopoietic niche cells in the bone marrow.


Subject(s)
Endothelial Cells , Stem Cell Factor , Mice , Animals , Stem Cell Factor/metabolism , Hematopoietic Stem Cells/metabolism , Bone Marrow/metabolism , Bone and Bones , Stem Cell Niche , Bone Marrow Cells/metabolism
3.
Nat Immunol ; 13(4): 412-9, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344248

ABSTRACT

The stepwise commitment from hematopoietic stem cells in the bone marrow to T lymphocyte-restricted progenitors in the thymus represents a paradigm for understanding the requirement for distinct extrinsic cues during different stages of lineage restriction from multipotent to lineage-restricted progenitors. However, the commitment stage at which progenitors migrate from the bone marrow to the thymus remains unclear. Here we provide functional and molecular evidence at the single-cell level that the earliest progenitors in the neonatal thymus had combined granulocyte-monocyte, T lymphocyte and B lymphocyte lineage potential but not megakaryocyte-erythroid lineage potential. These potentials were identical to those of candidate thymus-seeding progenitors in the bone marrow, which were closely related at the molecular level. Our findings establish the distinct lineage-restriction stage at which the T cell lineage-commitment process transits from the bone marrow to the remote thymus.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Lymphoid Progenitor Cells/cytology , Myeloid Cells/cytology , Precursor Cells, B-Lymphoid/cytology , T-Lymphocytes/cytology , Animals , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Thymus Gland/cytology
4.
Virol J ; 21(1): 166, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080643

ABSTRACT

BACKGROUND: Simian T-cell leukemia virus type 1 (STLV-1) is a retrovirus closely related to human T-cell leukemia virus type 1 (HTLV-1), the causative agent of adult T-cell leukemia (ATL). It has been shown that Japanese macaques (Macaca fuscata, JMs) are one of the main hosts of STLV-1 and that a high percentage of JMs (up to 60%) are infected with STLV-1; however, the molecular epidemiology of STLV-1 in JMs has not been examined. METHODS: In this study, we analyzed full-length STLV-1 genome sequences obtained from 5 independent troops including a total of 68 JMs. RESULTS: The overall nucleotide heterogeneity was 4.7%, and the heterogeneity among the troops was 2.1%, irrespective of the formation of distinct subclusters in each troop. Moreover, the heterogeneity within each troop was extremely low (>99% genome homology) compared with cases of STLV-1 in African non-human primates as well as humans. It was previously reported that frequent G-to-A single-nucleotide variants (SNVs) occur in HTLV-1 proviral genomes in both ATL patients and HTLV-1 carriers, and that a G-to-A hypermutation is associated with the cellular antiviral restriction factor, Apobec3G. Surprisingly, these SNVs were scarcely observed in the STLV-1 genomes in JMs. CONCLUSIONS: Taken together, these results indicate that STLV-1 genomes in JMs are highly homologous, at least in part due to the lack of Apobec3G-dependent G-to-A hypermutation.


Subject(s)
Genome, Viral , Macaca fuscata , Simian T-lymphotropic virus 1 , Animals , Simian T-lymphotropic virus 1/genetics , Simian T-lymphotropic virus 1/isolation & purification , Macaca fuscata/genetics , Phylogeny , Cohort Studies , Deltaretrovirus Infections/virology , Deltaretrovirus Infections/veterinary , Deltaretrovirus Infections/epidemiology , Japan , Humans , Sequence Analysis, DNA , Molecular Epidemiology , Genetic Variation
5.
Biologicals ; 85: 101754, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38428357

ABSTRACT

The Eighth Asian National Control Laboratory (NCL) Network meeting, entitled "Biological Products Quality Control and Self-Sufficiency Strategy focusing on plasma-derived medicinal products (PDMPs)" was held in Seoul on 31 August 2023. The participants were NCL experts from Indonesia, Japan, Malaysia, the Philippines, Vietnam, and the Republic of Korea. Special lectures included the PDMPs self-sufficiency strategies of the World Health Organization (WHO) and Indonesian Food and Drug Authority, and a case study on Global Benchmarking Tool (GBT) assessment for vaccines by the Korea Ministry of Food and Drug Safety. The NCL delegates shared their current experiences with national lot releases and biological standardisation. The meeting contributed to a mutual understanding of the progress of the PDMPs self-sufficiency among Asian countries, the WHO's support strategies, and the NCL's plan for the preparation of the WHO GBT assessment. In the panel discussion, all participants agreed that building capacity in blood safety in the Asian region and harmonisation of relevant international regulatory requirements will support appropriate emergency preparedness, particularly source materials in the region, and will build the foundation for resolving the PDMPs supply insecurity that has worsened after the COVID-19 pandemic in some countries.


Subject(s)
Pandemics , Humans , Pandemics/prevention & control , Asia , Indonesia , World Health Organization , Republic of Korea
6.
PLoS Pathog ; 17(8): e1009890, 2021 08.
Article in English | MEDLINE | ID: mdl-34460865

ABSTRACT

Aluminum hydroxide salts (alum) have been added to inactivated vaccines as safe and effective adjuvants to increase the effectiveness of vaccination. However, the exact cell types and immunological factors that initiate mucosal immune responses to alum adjuvants are unclear. In this study, the mechanism of action of alum adjuvant in nasal vaccination was investigated. Alum has been shown to act as a powerful and unique adjuvant when added to a nasal influenza split vaccine in mice. Alum is cytotoxic in the alveoli and stimulates the release of damage-associated molecular patterns, such as dsDNA, interleukin (IL)-1α, and IL-33. We found that Ag-specific IgA antibody (Ab) production was markedly reduced in IL-33-deficient mice. However, no decrease was observed in Ag-specific IgA Ab production with DNase I treatment, and no decrease was observed in IL-1α/ß or IL-6 production in IL-33-deficient mice. From the experimental results of primary cultured cells and immunofluorescence staining, although IL-1α was secreted by alveolar macrophage necroptosis, IL-33 release was observed in alveolar epithelial cell necroptosis but not in alveolar macrophages. Alum- or IL-33-dependent Ag uptake enhancement and elevation of OX40L expression were not observed. By stimulating the release of IL-33, alum induced Th2 immunity via IL-5 and IL-13 production in group 2 innate lymphoid cells (ILC2s) and increased MHC class II expression in antigen-presenting cells (APCs) in the lung. Our results suggest that IL-33 secretion by epithelial cell necroptosis initiates APC- and ILC2-mediated T cell activation, which is important for the enhancement of Ag-specific IgA Ab production by alum.


Subject(s)
Aluminum Hydroxide/chemistry , Alveolar Epithelial Cells/immunology , Immunoglobulin A/metabolism , Influenza Vaccines/administration & dosage , Interleukin-33/physiology , Orthomyxoviridae Infections/immunology , Th2 Cells/immunology , Adjuvants, Immunologic/administration & dosage , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Animals , Antibodies, Viral/immunology , Antibody Formation , Female , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoglobulin A/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/chemistry , Nasal Mucosa/metabolism , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Vaccination
7.
Biologicals ; 78: 17-26, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35840492

ABSTRACT

This online workshop Accelerating Global Deletion of the Abnormal Toxicity Test for vaccines and biologicals. Planning common next steps was organized on October 14th, 2021, by the Animal Free Safety Assessment Collaboration (AFSA), the Humane Society International (HSI), the European Federation of Pharmaceutical Industries and Associations (EFPIA), in collaboration with the International Alliance of Biological Standardization (IABS). The workshop saw a participation of over a hundred representatives from international organizations, pharmaceutical industries and associations, and regulatory authorities of 28 countries. Participants reported on country- and region-specific regulatory requirements and, where present, on the perspectives on the waiving and elimination of the Abnormal Toxicity Test. With AFSA, HSI, EFPIA and IABS representatives as facilitators, the participants also discussed specific country/global actions to further secure the deletion of ATT from all regulatory requirements worldwide.


Subject(s)
Toxicity Tests , Vaccines , Drug Industry , Humans , Reference Standards , Vaccines/adverse effects
8.
Retrovirology ; 17(1): 15, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576215

ABSTRACT

BACKGROUND: Simian T-cell leukemia virus type 1 (STLV-1) is disseminated among various non-human primate species and is closely related to human T-cell leukemia virus type 1 (HTLV-1), the causative agent of adult T-cell leukemia and HTLV-1-associated myelopathy/tropical spastic paraparesis. Notably, the prevalence of STLV-1 infection in Japanese macaques (JMs) is estimated to be > 60%, much greater than that in other non-human primates; however, the mechanism and mode of STLV-1 transmission remain unknown. The aim of this study is to examine the epidemiological background by which STLV-1 infection is highly prevalent in JMs. RESULTS: The prevalence of STLV-1 in the JMs rearing in our free-range facility reached up to 64% (180/280 JMs) with variation from 55 to 77% among five independent troops. Anti-STLV-1 antibody titers (ABTs) and STLV-1 proviral loads (PVLs) were normally distributed with mean values of 4076 and 0.62%, respectively, which were mostly comparable to those of HTLV-1-infected humans. Our initial hypothesis that some of the macaques might contribute to frequent horizontal STLV-1 transmission as viral super-spreaders was unlikely because of the absence of the macaques exhibiting abnormally high PVLs but poor ABTs. Rather, ABTs and PVLs were statistically correlated (p < 0.0001), indicating that the increasing PVLs led to the greater humoral immune response. Further analyses demonstrated that the STLV-1 prevalence as determined by detection of the proviral DNA was dramatically increased with age; 11%, 31%, and 58% at 0, 1, and 2 years of age, respectively, which was generally consistent with the result of seroprevalence and suggested the frequent incidence of mother-to-child transmission. Moreover, our longitudinal follow-up study indicated that 24 of 28 seronegative JMs during the periods from 2011 to 2012 converted to seropositive (86%) 4 years later; among them, the seroconversion rates of sexually matured (4 years of age and older) macaques and immature macaques (3 years of age and younger) at the beginning of study were comparably high (80% and 89%, respectively), suggesting the frequent incidence of horizontal transmission. CONCLUSIONS: Together with the fact that almost all of the full-adult JMs older than 9 years old were infected with STLV-1, our results of this study demonstrated for the first time that frequent horizontal and mother-to-child transmission may contribute to high prevalence of STLV-1 infection in JMs.


Subject(s)
Antibodies, Viral/blood , Deltaretrovirus Infections/transmission , Deltaretrovirus Infections/veterinary , Disease Transmission, Infectious , Infectious Disease Transmission, Vertical , Simian T-lymphotropic virus 1/physiology , Animals , Female , Follow-Up Studies , Japan , Macaca fuscata/virology , Male , Prevalence , Proviruses/genetics , Seroepidemiologic Studies , Simian T-lymphotropic virus 1/genetics
9.
Biologicals ; 55: 43-52, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30017557

ABSTRACT

Recently, many vaccine adjuvants have been developed; however, most of the newly developed adjuvants have been dropped out of preclinical and clinical trials owing to their unexpected toxicity. Thus, the development of highly quantitative and comparable screening methods for evaluating adjuvant safety is needed. In a previous study, we identified specific biomarkers for evaluating the safety of an intranasal influenza vaccine with CpG K3 adjuvant by comparing biomarker expression. We hypothesized that these biomarkers might be useful for screening newly developed adjuvant safety. We compared the expression of biomarkers in mouse lungs by the intranasal administration of 4 types of adjuvants: Alum, Pam3CSK4, NanoSiO2, and DMXAA with subvirion influenza vaccine. The control adjuvant alum did not show any significant increase in biomarker expression or preclinical parameters; however, NanoSiO2 and Pam3CSK4 increased the expression of biomarkers, such as Timp1 and Csf1. DMXAA at 300 µg induced the expression of over 80% of biomarkers. Hierarchical clustering analysis showed that 300 µg DMXAA was classified in the toxicity reference whole-particle influenza vaccine cluster. FACS analysis to confirm specific phenotypes that the number of T cells decreased in DMXAA-treated mouse lungs. Thus, our biomarkers are useful for initial adjuvant safety and toxicity screening.


Subject(s)
Adjuvants, Immunologic , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Drug Evaluation, Preclinical , Female , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C
10.
Biologicals ; 50: 100-108, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28838806

ABSTRACT

The leukopenic toxicity test (LTT) is used to evaluate the safety and lot-to-lot consistency of influenza hemagglutinin split vaccine (HAv) and is included in the Japanese Minimum Requirements for Biological Products. LTT assesses the reduced leukocyte levels in murine peripheral blood after HAv administration. However, they require large numbers of animals, and therefore it would be beneficial to develop a more accurate and sensitive alternative method. In this study, we selected biomarkers of leukocyte reduction from 18 previously identified marker genes that were associated with an abnormal toxicity test (ATT). Among these 18 genes, the expressions of 15 marker genes were strongly associated with leukocyte reduction levels. A stepwise single addition multiple regression analysis was used to further extract the genes responsible for leukocyte reduction, with significant (p < 0.25) regression coefficients. The expression of 7 genes significantly predicted the leukocyte reduction. The prediction accuracy of this approach was approximately >90% (mean) for the direct measurement of leukocyte numbers. These results indicate that the expression of these 18 previously identified genes can provide information for both ATT and LTT.


Subject(s)
Cytotoxicity Tests, Immunologic/methods , Influenza Vaccines/immunology , Leukocytes/immunology , Vaccines, Inactivated/immunology , Animals , Biomarkers/metabolism , Female , Gene Expression Profiling/methods , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/pharmacology , Influenza Vaccines/standards , Leukocyte Count , Leukocytes/drug effects , Leukocytes/metabolism , Lung/immunology , Lung/metabolism , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Vaccination , Vaccines, Inactivated/pharmacology , Vaccines, Inactivated/standards
11.
J Clin Microbiol ; 53(2): 587-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25502533

ABSTRACT

Quantitative PCR (qPCR) for human T-lymphotropic virus 1 (HTLV-1) is useful for measuring the amount of integrated HTLV-1 proviral DNA in peripheral blood mononuclear cells. Many laboratories in Japan have developed different HTLV-1 qPCR methods. However, when six independent laboratories analyzed the proviral load of the same samples, there was a 5-fold difference in their results. To standardize HTLV-1 qPCR, preparation of a well-defined reference material is needed. We analyzed the integrated HTLV-1 genome and the internal control (IC) genes of TL-Om1, a cell line derived from adult T-cell leukemia, to confirm its suitability as a reference material for HTLV-1 qPCR. Fluorescent in situ hybridization (FISH) showed that HTLV-1 provirus was monoclonally integrated in chromosome 1 at the site of 1p13 in the TL-Om1 genome. HTLV-1 proviral genome was not transferred from TL-Om1 to an uninfected T-cell line, suggesting that the HTLV-1 proviral copy number in TL-Om1 cells is stable. To determine the copy number of HTLV-1 provirus and IC genes in TL-Om1 cells, we used FISH, digital PCR, and qPCR. HTLV-1 copy numbers obtained by these three methods were similar, suggesting that their results were accurate. Also, the ratio of the copy number of HTLV-1 provirus to one of the IC genes, RNase P, was consistent for all three methods. These findings indicate that TL-Om1 cells are an appropriate reference material for HTLV-1 qPCR.


Subject(s)
Human T-lymphotropic virus 1/isolation & purification , Proviruses/isolation & purification , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Reference Standards , Viral Load/methods , Cell Line, Tumor , Human T-lymphotropic virus 1/genetics , Humans , Japan , Proviruses/genetics
12.
J Clin Microbiol ; 53(11): 3485-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26292315

ABSTRACT

Quantitative PCR (qPCR) analysis of human T-cell leukemia virus type 1 (HTLV-1) was used to assess the amount of HTLV-1 provirus DNA integrated into the genomic DNA of host blood cells. Accumulating evidence indicates that a high proviral load is one of the risk factors for the development of adult T-cell leukemia/lymphoma and HTLV-1-associated myelopathy/tropical spastic paraparesis. However, interlaboratory variability in qPCR results makes it difficult to assess the differences in reported proviral loads between laboratories. To remedy this situation, we attempted to minimize discrepancies between laboratories through standardization of HTLV-1 qPCR in a collaborative study. TL-Om1 cells that harbor the HTLV-1 provirus were serially diluted with peripheral blood mononuclear cells to prepare a candidate standard. By statistically evaluating the proviral loads of the standard and those determined using in-house qPCR methods at each laboratory, we determined the relative ratios of the measured values in the laboratories to the theoretical values of the TL-Om1 standard. The relative ratios of the laboratories ranged from 0.84 to 4.45. Next, we corrected the proviral loads of the clinical samples from HTLV-1 carriers using the relative ratio. As expected, the overall differences between the laboratories were reduced by half, from 7.4-fold to 3.8-fold on average, after applying the correction. HTLV-1 qPCR can be standardized using TL-Om1 cells as a standard and by determining the relative ratio of the measured to the theoretical standard values in each laboratory.


Subject(s)
DNA, Viral/analysis , Human T-lymphotropic virus 1/genetics , Real-Time Polymerase Chain Reaction/methods , Real-Time Polymerase Chain Reaction/standards , Viral Load/genetics , Cell Line, Tumor , DNA, Viral/genetics , HTLV-I Infections/genetics , HTLV-I Infections/virology , Humans , Japan , Jurkat Cells , Leukemia, T-Cell/genetics , Leukemia, T-Cell/virology , Leukocytes, Mononuclear/virology , Proviruses/genetics , Virus Integration/genetics
13.
Blood ; 119(10): 2376-84, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22262766

ABSTRACT

Fifty percent of Diamond-Blackfan anemia (DBA) patients possess mutations in genes coding for ribosomal proteins (RPs). To identify new mutations, we investigated large deletions in the RP genes RPL5, RPL11, RPL35A, RPS7, RPS10, RPS17, RPS19, RPS24, and RPS26. We developed an easy method based on quantitative-PCR in which the threshold cycle correlates to gene copy number. Using this approach, we were able to diagnose 7 of 27 Japanese patients (25.9%) possessing mutations that were not detected by sequencing. Among these large deletions, similar results were obtained with 6 of 7 patients screened with a single nucleotide polymorphism array. We found an extensive intragenic deletion in RPS19, including exons 1-3. We also found 1 proband with an RPL5 deletion, 1 patient with an RPL35A deletion, 3 with RPS17 deletions, and 1 with an RPS19 deletion. In particular, the large deletions in the RPL5 and RPS17 alleles are novel. All patients with a large deletion had a growth retardation phenotype. Our data suggest that large deletions in RP genes comprise a sizable fraction of DBA patients in Japan. In addition, our novel approach may become a useful tool for screening gene copy numbers of known DBA genes.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Gene Deletion , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/ethnology , Anemia, Diamond-Blackfan/pathology , Asian People/genetics , Child, Preschool , DNA Mutational Analysis/methods , Female , Genetic Association Studies , Humans , Infant , Infant, Newborn , Japan , Male , Microarray Analysis/methods , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Reproducibility of Results
14.
N Engl J Med ; 363(11): 1025-37, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20825315

ABSTRACT

BACKGROUND: The in vivo clinical significance of malignant stem cells remains unclear. METHODS: Patients who have the 5q deletion (del[5q]) myelodysplastic syndrome (interstitial deletions involving the long arm of chromosome 5) have complete clinical and cytogenetic remissions in response to lenalidomide treatment, but they often have relapse. To determine whether the persistence of rare but distinct malignant stem cells accounts for such relapses, we examined bone marrow specimens obtained from seven patients with the del(5q) myelodysplastic syndrome who became transfusion-independent while receiving lenalidomide treatment and entered cytogenetic remission. RESULTS: Virtually all CD34+, CD38+ progenitor cells and stem cells that were positive for CD34 and CD90, with undetectable or low CD38 (CD38−/low), had the 5q deletion before treatment. Although lenalidomide efficiently reduced these progenitors in patients in complete remission, a larger fraction of the minor, quiescent, CD34+,CD38-/low, CD90+ del(5q) stem cells as well as functionally defined del(5q) stem cells remained distinctly resistant to lenalidomide. Over time, lenalidomide resistance developed in most of the patients in partial and complete remission, with recurrence or expansion of the del(5q) clone and clinical and cytogenetic progression. CONCLUSIONS: In these patients with the del(5q) myelodysplastic syndrome, we identified rare and phenotypically distinct del(5q) myelodysplastic syndrome stem cells that were also selectively resistant to therapeutic targeting at the time of complete clinical and cytogenetic remission. (Funded by the EuroCancerStemCell Consortium and others.)


Subject(s)
Antineoplastic Agents/pharmacology , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Drug Resistance/genetics , Myelodysplastic Syndromes/pathology , Neoplastic Stem Cells/drug effects , Thalidomide/analogs & derivatives , ADP-ribosyl Cyclase 1/analysis , Aged , Aged, 80 and over , Antigens, CD34/analysis , Antineoplastic Agents/therapeutic use , Female , Gene Expression , Humans , Lenalidomide , Male , Middle Aged , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Neoplastic Stem Cells/immunology , Phenotype , Remission Induction , Thalidomide/pharmacology , Thalidomide/therapeutic use , Thy-1 Antigens/analysis
15.
Transfusion ; 53(10 Pt 2): 2545-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23590180

ABSTRACT

BACKGROUND: The risk of transferring blood-borne infections during transfusion is continually increasing because of newly emerging and reemerging viruses. Development of a rapid screening method for emerging viruses that might be transmitted by transfusion is required to eliminate such pathogens during blood donor screening. Owing to increased use of human materials in organ transplants and cell therapy, the risk of donor-transmitted viral infections is also increasing. Although nucleic acid amplification technology (NAT) is dedicated to blood screening, a small, convenient detection system is needed at the laboratory and hospital level. STUDY DESIGN AND METHODS: We developed a new pathogen detection system that can detect multiple viruses simultaneously, using originally designed degenerate polymerase chain reaction primers to amplify a wide range of viral genotypes. Amplified samples were identified using a DNA microarray of pathogen-specific probes. RESULTS: We detected very low copy numbers of multiple subtypes of viruses, such as human hepatitis C virus (HCV), human hepatitis B virus (HBV), human parvovirus B19 (PVB19), and West Nile virus (WNV), using a single plate. We also detected all genotypes of human immunodeficiency virus (HIV) but sensitivity was less than for the other viruses. CONCLUSION: We developed a microarray assay using novel primers for detection of a wide range of multiple pathogens and subtypes. Our NAT system was accurate and reliable for detection of HIV, HBV, HCV, PVB19, and WNV, with respect to specificity, sensitivity, and genotype inclusivity. Our system could be customized and extended for emerging pathogens and is suitable as a future NAT system.


Subject(s)
Blood Donors , Mass Screening/methods , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction/methods , Viruses/isolation & purification , Blood-Borne Pathogens/isolation & purification , DNA Primers/genetics , DNA, Viral/genetics , DNA, Viral/isolation & purification , Genotype , Humans , Oligonucleotide Array Sequence Analysis/instrumentation , Sensitivity and Specificity , Viruses/genetics , World Health Organization
16.
Mucosal Immunol ; 16(3): 275-286, 2023 06.
Article in English | MEDLINE | ID: mdl-36935091

ABSTRACT

The stimulation of local immunity by vaccination is desirable for controlling virus replication in the respiratory tract. However, the local immune stimulatory effects of adjuvanted vaccines administered through the non-mucosal route are poorly understood. Here, we clarify the mechanisms by which non-mucosal inoculation of adjuvants stimulates the plasmacytoid dendritic cell (pDC)-dependent immunoglobulin (Ig)A response in the lungs. After systemic inoculation with type 1 interferon (IFN)-inducing adjuvants, type 1 IFN promotes CXCL9/10/11 release from alveolar endothelial and epithelial cells and recruits CXCR3-expressing pDCs into the lungs. Because adjuvant-activated pulmonary pDCs highly express major histocompatibility complex II, cluster of differentiation 80, and cluster of differentiation 86, transplantation of such cells into the lungs successfully enhances antigen-specific IgA production by the intranasally sensitized vaccine. In contrast, pDC accumulation in the lungs and subsequent IgA production are impaired in pDC-depleted mice and Ifnar1-/- mice. Notably, the combination of systemic inoculation with type 1 IFN-inducing adjuvants and intranasal antigen sensitization protects mice against influenza virus infection due to the pDC-dependent IgA response and type I IFN response. Our results provide insights into the novel mucosal vaccine strategies using non-mucosal inoculated adjuvants.


Subject(s)
Influenza Vaccines , Interferon Type I , Animals , Mice , Immunoglobulin A , Immunoglobulin G , Immunity, Mucosal , Antibodies, Viral , Adjuvants, Immunologic , Administration, Intranasal , Dendritic Cells , Mice, Inbred BALB C
17.
J Med Chem ; 66(18): 13266-13279, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37676021

ABSTRACT

Protein or peptide cancer vaccines usually include immune potentiators, so-called adjuvants. However, it remains challenging to identify structurally simple, chemically accessible synthetic molecules that are effective and safe as vaccine adjuvant. Here, we present cholicamideß (6), a self-assembling small-molecule vaccine adjuvant with an improved toxicity profile and proven efficacy in vivo. We demonstrate that cholicamideß (6), which is less cytotoxic than its parent compound, forms virus-like particles to potently activate dendritic cells with the concomitant secretion of cytokines. When combined with a peptide antigen, cholicamideß (6) potentiated the antigen presentation on dendritic cells to induce antigen-specific T cells. As a therapeutic cancer vaccine adjuvant in mice, a mixture of cholicamideß (6) and a peptide antigen protected mice from the challenges of malignant cancer cells without overt toxicity. Cholicamideß (6) may offer a translational opportunity as an unprecedented class of small-molecule cancer vaccine adjuvants.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , Mice , Cancer Vaccines/therapeutic use , Adjuvants, Vaccine , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/chemistry , T-Lymphocytes , Adjuvants, Pharmaceutic , Vaccines, Subunit , Peptides , Dendritic Cells
18.
iScience ; 26(5): 106694, 2023 May 19.
Article in English | MEDLINE | ID: mdl-37124417

ABSTRACT

Evaluating the serum cross-neutralization responses after breakthrough infection with various SARS-CoV-2 variants provides valuable insight for developing variant-proof COVID-19 booster vaccines. However, fairly comparing the impact of breakthrough infections with distinct epidemic timing on cross-neutralization responses, influenced by the exposure interval between vaccination and infection, is challenging. To compare the impact of pre-Omicron to Omicron breakthrough infection, we estimated the effects on cross-neutralizing responses by the exposure interval using Bayesian hierarchical modeling. The saturation time required to generate saturated cross-neutralization responses differed by variant, with variants more antigenically distant from the ancestral strain requiring longer intervals of 2-4 months. The breadths of saturated cross-neutralization responses to Omicron lineages were comparable in pre-Omicron and Omicron breakthrough infections. Our results highlight the importance of vaccine dosage intervals of 4 months or longer, regardless of the antigenicity of the exposed antigen, to maximize the breadth of serum cross-neutralization covering SARS-CoV-2 Omicron lineages.

19.
J Toxicol Sci ; 47(11): 439-451, 2022.
Article in English | MEDLINE | ID: mdl-36328534

ABSTRACT

Aluminum salt adjuvants (Als) have been the most widely used adjuvants in vaccines and known to be effective in intramuscular inoculation. However, in rare cases, some Al containing vaccines caused serious adverse events such as chronic pain at the site of the injection. The Als cause mild tissue damage at the inoculation site, allowing the antigen to be locally retained at the inoculation site and thus potentiate innate immunity. This is required to elicit effectiveness of vaccination. However, there is concern that chronic muscle damage might potentially lead to serious adverse events, such as autoimmune disease and movement disorders. In this study, muscle damage caused by several Al containing vaccines were examined in guinea pigs. Mild and moderate inflammation were observed following Al containing split influenza virus vaccine, formalin-inactivated diphtheria-pertussis-tetanus and Salk polio vaccine. While massive inflammation and muscle damage were observed in Al-containing human papillomavirus vaccine-inoculated animals. However, the severities of damage were not associated with their Al contents. Masson's trichrome staining and immunostaining revealed that injured muscle at the inoculated site recovered within one month of vaccination, whereas inflammatory nodules remained. Flow cytometric analyses of the infiltrating cells revealed that the number of CD45+ lymphocytes and potential granulocytes were increased following vaccination. The number of infiltrated cells seemed to be associated with severity of muscle damages. These observations revealed that Al containing vaccine-induced muscle damage is reparable, and severity of transient muscle damages seemed to be determined by type of antigen or types of Al salts rather than Al content.


Subject(s)
Aluminum , Vaccines , Guinea Pigs , Humans , Animals , Aluminum/toxicity , Vaccines/adverse effects , Adjuvants, Immunologic/toxicity , Inflammation/chemically induced , Muscles
20.
Med ; 3(6): 406-421.e4, 2022 06 10.
Article in English | MEDLINE | ID: mdl-35815933

ABSTRACT

BACKGROUND: The Omicron variant of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) was identified in Japan in November 2021. This variant contains up to 36 mutations in the spike protein, the target of neutralizing antibodies, and can escape vaccine-induced immunity. A booster vaccination campaign began with healthcare workers and high-risk groups. The safety and immunogenicity of the three-dose vaccination against Omicron remain unknown. METHODS: A total of 272 healthcare workers were initially evaluated for long-term vaccine safety and immunogenicity. We further established a vaccinee panel to evaluate the safety and immunogenicity against variants of concern (VOCs), including the Omicron variants, using a live virus microneutralization assay. FINDINGS: Two-dose vaccination induced robust anti-spike antibodies and neutralization titers (NTs) against the ancestral strain WK-521, whereas NTs against VOCs were significantly lower. Within 93-247 days of the second vaccine dose, NTs against Omicron were completely abolished in up to 80% of individuals in the vaccinee panel. Booster dose induced a robust increase in anti-spike antibodies and NTs against the WK-521, Delta, and Omicron variants. There were no significant differences in the neutralization ability of sera from boosted individuals among the Omicron subvariants BA.1, BA.1.1, and BA.2. Boosting increased the breadth of humoral immunity and cross-reactivity with Omicron without changes in cytokine signatures and adverse event rate. CONCLUSIONS: The third vaccination dose is safe and increases neutralization against Omicron variants. FUNDING: This study was supported by grants from AMED (grants JP21fk0108104 and JP21mk0102146).


Subject(s)
Antibodies, Viral , BNT162 Vaccine , COVID-19 , Immunization, Secondary , Immunogenicity, Vaccine , Antibodies, Neutralizing , BNT162 Vaccine/immunology , COVID-19/prevention & control , Cross Reactions , Humans , Immunity, Humoral , Neutralization Tests , RNA, Messenger , SARS-CoV-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL