Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Cell ; 148(4): 739-51, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22341446

ABSTRACT

B cells infected by Epstein-Barr virus (EBV), a transforming virus endemic in humans, are rapidly cleared by the immune system, but some cells harboring the virus persist for life. Under conditions of immunosuppression, EBV can spread from these cells and cause life-threatening pathologies. We have generated mice expressing the transforming EBV latent membrane protein 1 (LMP1), mimicking a constitutively active CD40 coreceptor, specifically in B cells. Like human EBV-infected cells, LMP1+ B cells were efficiently eliminated by T cells, and breaking immune surveillance resulted in rapid, fatal lymphoproliferation and lymphomagenesis. The lymphoma cells expressed ligands for a natural killer (NK) cell receptor, NKG2D, and could be targeted by an NKG2D-Fc fusion protein. These experiments indicate a central role for LMP1 in the surveillance and transformation of EBV-infected B cells in vivo, establish a preclinical model for B cell lymphomagenesis in immunosuppressed patients, and validate a new therapeutic approach.


Subject(s)
Disease Models, Animal , Herpesvirus 4, Human , Immunologic Surveillance , Lymphoma/immunology , Lymphoma/therapy , Viral Matrix Proteins/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Humans , Immunotherapy , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Viral Matrix Proteins/genetics
2.
Mol Cell ; 72(1): 84-98.e9, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30220558

ABSTRACT

Emerging evidence indicates that heterogeneity in ribosome composition can give rise to specialized functions. Until now, research mainly focused on differences in core ribosomal proteins and associated factors. The effect of posttranslational modifications has not been studied systematically. Analyzing ribosome heterogeneity is challenging because individual proteins can be part of different subcomplexes (40S, 60S, 80S, and polysomes). Here we develop polysome proteome profiling to obtain unbiased proteomic maps across ribosomal subcomplexes. Our method combines extensive fractionation by sucrose gradient centrifugation with quantitative mass spectrometry. The high resolution of the profiles allows us to assign proteins to specific subcomplexes. Phosphoproteomics on the fractions reveals that phosphorylation of serine 38 in RPL12/uL11, a known mitotic CDK1 substrate, is strongly depleted in polysomes. Follow-up experiments confirm that RPL12/uL11 phosphorylation regulates the translation of specific subsets of mRNAs during mitosis. Together, our results show that posttranslational modification of ribosomal proteins can regulate translation.


Subject(s)
Protein Biosynthesis , Protein Processing, Post-Translational/genetics , Proteomics , Ribosomal Proteins/genetics , Humans , Mass Spectrometry , Mitosis/genetics , Phosphorylation/genetics , Polyribosomes/genetics , Proteome/genetics
3.
Proc Natl Acad Sci U S A ; 120(10): e2219439120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36853944

ABSTRACT

Multiple myeloma (MM), a tumor of germinal center (GC)-experienced plasma cells, comprises distinct genetic subgroups, such as the t(11;14)/CCND1 and the t(4;14)/MMSET subtype. We have generated genetically defined, subgroup-specific MM models by the GC B cell-specific coactivation of mouse Ccnd1 or MMSET with a constitutively active Ikk2 mutant, mimicking the secondary NF-κB activation frequently seen in human MM. Ccnd1/Ikk2ca and MMSET/Ikk2ca mice developed a pronounced, clonally restricted plasma cell outgrowth with age, accompanied by serum M spikes, bone marrow insufficiency, and bone lesions. The transgenic plasma cells could be propagated in vivo and showed distinct transcriptional profiles, resembling their human MM counterparts. Thus, we show that targeting the expression of genes involved in MM subgroup-specific chromosomal translocations into mouse GC B cells translates into distinct MM-like diseases that recapitulate key features of the human tumors, opening the way to a better understanding of the pathogenesis and therapeutic vulnerabilities of different MM subgroups.


Subject(s)
Multiple Myeloma , Humans , Animals , Mice , Multiple Myeloma/genetics , Plasma Cells , B-Lymphocytes , Genes, cdc , Animals, Genetically Modified , Disease Models, Animal
4.
Genes Cells ; 28(6): 411-421, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36871192

ABSTRACT

The CARMA1-Bcl10-MALT1 (CBM) signalosome is a crucial module of NF-κB activation in B cell receptor (BCR) signaling. Biophysical studies have shown that the E3 ubiquitin ligase TRAF6 cooperatively modifies the CBM signalosome; however, the specific details regarding how TRAF6 is involved in BCR signal-induced CBM formation remain unclear. In this study, we aimed to reveal the influences of TRAF6 on CBM formation and TAK1 and IKK activities using DT40 B cells which lack all the exons of TRAF6. In TRAF6-null cells we found: (i) attenuation of TAK1 activity and abolishment of IKK activity and (ii) sustained binding of CARMA1 to Bcl10. To account for the molecular mechanism causing these dynamics, we performed a mathematical model analysis. The mathematical model analysis showed that the regulation of IKK activation by TRAF6 can reproduce TAK1 and IKK activities in TRAF6 null cells, and that the TRAF6 related signal-dependent inhibitor suppresses CARMA1 binding to Bcl10 in wild-type cells. These results suggest that TRAF6 contributes to the positive regulation of IKK activation via TAK1, alongside the negative signal-dependent regulation of CARMA1 binding to Bcl10.


Subject(s)
Adaptor Proteins, Signal Transducing , TNF Receptor-Associated Factor 6 , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , NF-kappa B/metabolism , Guanylate Cyclase/metabolism
5.
Int Immunol ; 35(4): 197-207, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36413150

ABSTRACT

The immune evasion of SARS-CoV-2 Omicron variants caused by multiple amino acid replacements in the receptor-binding domain (RBD) of the spike protein wanes the effectiveness of antibodies elicited by current SARS-CoV-2 booster vaccination. The vaccines that target Omicron strains have been recently developed, however, there has been a concern yet to be addressed regarding the negative aspect of the immune response known as original antigenic sin. Here, we demonstrate that the breadth of neutralizing antibodies against SARS-CoV-2 variants is barely elicited by immunizing monovalent viral antigens via vaccination or natural infection in mice and human subjects. However, vaccination of Omicron BA.1 RBD to pre-immunized mice with the original RBD conferred sustained neutralizing activity to BA.1 and BA.2 not only original pseudoviruses. The acquisition of neutralizing antibody breadth was further confirmed in vaccinated-then-Omicron convalescent human sera in which neutralizing activity against BA.1 and BA.2 pseudoviruses was highly induced. Thus, our data suggest that Omicron-specific vaccines or the infection with Omicron viruses can boost potent neutralizing antibodies to the Omicron variants even in the host pre-vaccinated with the original antigen.


Subject(s)
COVID-19 , Animals , Humans , Mice , Antibodies, Neutralizing , Antibodies, Viral , Broadly Neutralizing Antibodies , COVID-19/prevention & control , SARS-CoV-2 , Vaccination
6.
Immunity ; 43(6): 1075-86, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26620760

ABSTRACT

Phosphatidylinositol 3' OH kinase (PI3K) signaling and FOXO transcription factors play opposing roles at several B cell developmental stages. We show here abundant nuclear FOXO1 expression in the proliferative compartment of the germinal center (GC), its dark zone (DZ), and PI3K activity, downregulating FOXO1, in the light zone (LZ), where cells are selected for further differentiation. In the LZ, however, FOXO1 was expressed in a fraction of cells destined for DZ reentry. Upon FOXO1 ablation or induction of PI3K activity, GCs lost their DZ, owing at least partly to downregulation of the chemokine receptor CXCR4. Although this prevented proper cyclic selection of cells in GCs, somatic hypermutation and proliferation were maintained. Class switch recombination was partly lost due to a failure of switch region targeting by activation-induced deaminase (AID).


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Germinal Center/immunology , Phosphatidylinositol 3-Kinases/immunology , Animals , B-Lymphocytes/cytology , Cell Separation , Chromatography, Liquid , Cytidine Deaminase/immunology , Flow Cytometry , Fluorescent Antibody Technique , Forkhead Box Protein O1 , Gene Expression Regulation/immunology , Germinal Center/cytology , Immunoglobulin Class Switching/immunology , Lymphocyte Activation/immunology , Mice , Mice, Mutant Strains , Polymerase Chain Reaction , Somatic Hypermutation, Immunoglobulin/immunology , Tandem Mass Spectrometry
7.
PLoS Genet ; 17(8): e1009686, 2021 08.
Article in English | MEDLINE | ID: mdl-34351912

ABSTRACT

Although long noncoding RNAs (lncRNAs) are transcripts that do not encode proteins by definition, some lncRNAs actually contain small open reading frames that are translated. TINCR (terminal differentiation-induced ncRNA) has been recognized as a lncRNA that contributes to keratinocyte differentiation. However, we here show that TINCR encodes a ubiquitin-like protein that is well conserved among species and whose expression was confirmed by the generation of mice harboring a FLAG epitope tag sequence in the endogenous open reading frame as well as by targeted proteomics. Forced expression of this protein promoted cell cycle progression in normal human epidermal keratinocytes, and mice lacking this protein manifested a delay in skin wound healing associated with attenuated cell cycle progression in keratinocytes. We termed this protein TINCR-encoded ubiquitin-like protein (TUBL), and our results reveal a role for TINCR in the regulation of keratinocyte proliferation and skin regeneration that is dependent on TUBL.


Subject(s)
Keratinocytes/cytology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Cycle , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Gene Knock-In Techniques , Humans , Keratinocytes/metabolism , Mice , Open Reading Frames , Proteomics , Ubiquitins/genetics , Ubiquitins/metabolism , Wound Healing
8.
Proc Natl Acad Sci U S A ; 117(25): 14421-14432, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32522871

ABSTRACT

Epstein-Barr virus (EBV) is a B cell transforming virus that causes B cell malignancies under conditions of immune suppression. EBV orchestrates B cell transformation through its latent membrane proteins (LMPs) and Epstein-Barr nuclear antigens (EBNAs). We here identify secondary mutations in mouse B cell lymphomas induced by LMP1, to predict and identify key functions of other EBV genes during transformation. We find aberrant activation of early B cell factor 1 (EBF1) to promote transformation of LMP1-expressing B cells by inhibiting their differentiation to plasma cells. EBV EBNA3A phenocopies EBF1 activities in LMP1-expressing B cells, promoting transformation while inhibiting differentiation. In cells expressing LMP1 together with LMP2A, EBNA3A only promotes lymphomagenesis when the EBNA2 target Myc is also overexpressed. Collectively, our data support a model where proproliferative activities of LMP1, LMP2A, and EBNA2 in combination with EBNA3A-mediated inhibition of terminal plasma cell differentiation critically control EBV-mediated B cell lymphomagenesis.


Subject(s)
Cell Transformation, Viral , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , Lymphoma, B-Cell/pathology , Plasma Cells/pathology , Animals , Cell Differentiation , Cell Line, Tumor , DNA-Binding Proteins/genetics , Disease Models, Animal , Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/metabolism , Fibroblasts , Herpesvirus 4, Human/metabolism , Humans , Lymphoma, B-Cell/virology , Mice , Mice, Knockout , Plasma Cells/virology , Primary Cell Culture , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism
9.
J Pharmacol Sci ; 148(3): 307-314, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35177210

ABSTRACT

G protein-coupled receptor 3 (GPR3) constitutively activates Gαs proteins without any ligands and is predominantly expressed in neurons. Since the expression and physiological role of GPR3 in immune cells is still unknown, we examined the possible role of GPR3 in T lymphocytes. The expression of GPR3 was upregulated 2 h after phorbol 12-myristate 13-acetate (PMA)/ionomycin stimulation and was sustained in Jurkat cells, a human T lymphocyte cell line. In addition, the expression of nuclear receptor 4 group A member 2 (NR4A2) was highly modulated by GPR3 expression. Additionally, GPR3 expression was linked with the transcriptional promoter activity of NR4A in Jurkat cells. In mouse CD4+ T cells, transient GPR3 expression was induced immediately after the antigen receptor stimulation. However, the expression of NR4A2 was not modulated in CD4+ T cells from GPR3-knockout mice after stimulation, and the population of Treg cells in thymocytes and splenocytes was not affected by GPR3 knockout. By contrast, spontaneous effector activation in both CD4+ T cells and CD8+ T cells was observed in GPR3-knockout mice. In summary, GPR3 is immediately induced by T cell stimulation and play an important role in the suppression of effector T cell activation.


Subject(s)
Lymphocyte Activation/genetics , Receptors, G-Protein-Coupled/physiology , T-Lymphocytes/immunology , Animals , Cell Line , Chromogranins/metabolism , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Gene Expression , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , T-Lymphocytes/metabolism
10.
Immunity ; 32(6): 828-39, 2010 Jun 25.
Article in English | MEDLINE | ID: mdl-20605486

ABSTRACT

Although the cellular concentration of miRNAs is critical to their function, how miRNA expression and abundance are regulated during ontogeny is unclear. We applied miRNA-, mRNA-, and ChIP-Seq to characterize the microRNome during lymphopoiesis within the context of the transcriptome and epigenome. We show that lymphocyte-specific miRNAs are either tightly controlled by polycomb group-mediated H3K27me3 or maintained in a semi-activated epigenetic state prior to full expression. Because of miRNA biogenesis, the cellular concentration of mature miRNAs does not typically reflect transcriptional changes. However, we uncover a subset of miRNAs for which abundance is dictated by miRNA gene expression. We confirm that concentration of 5p and 3p miRNA strands depends largely on free energy properties of miRNA duplexes. Unexpectedly, we also find that miRNA strand accumulation can be developmentally regulated. Our data provide a comprehensive map of immunity's microRNome and reveal the underlying epigenetic and transcriptional forces that shape miRNA homeostasis.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation/genetics , Lymphocytes , Lymphopoiesis/genetics , MicroRNAs/genetics , Animals , Gene Expression , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction
11.
Proc Natl Acad Sci U S A ; 113(48): 13821-13826, 2016 11 29.
Article in English | MEDLINE | ID: mdl-27856754

ABSTRACT

Epstein-Barr Virus (EBV) infects human B cells and drives them into continuous proliferation. Two key viral factors in this process are the latent membrane proteins LMP1 and LMP2A, which mimic constitutively activated CD40 receptor and B-cell receptor signaling, respectively. EBV-infected B cells elicit a powerful T-cell response that clears the infected B cells and leads to life-long immunity. Insufficient immune surveillance of EBV-infected B cells causes life-threatening lymphoproliferative disorders, including mostly germinal center (GC)-derived B-cell lymphomas. We have modeled acute EBV infection of naive and GC B cells in mice through timed expression of LMP1 and LMP2A. Although lethal when induced in all B cells, induction of LMP1 and LMP2A in just a small fraction of naive B cells initiated a phase of rapid B-cell expansion followed by a proliferative T-cell response, clearing the LMP-expressing B cells. Interfering with T-cell activity prevented clearance of LMP-expressing B cells. This was also true for perforin deficiency, which in the human causes a life-threatening EBV-related immunoproliferative syndrome. LMP expression in GC B cells impeded the GC reaction but, upon loss of T-cell surveillance, led to fatal B-cell expansion. Thus, timed expression of LMP1 together with LMP2A in subsets of mouse B cells allows one to study major clinically relevant features of human EBV infection in vivo, opening the way to new therapeutic approaches.


Subject(s)
B-Lymphocytes/virology , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Viral Matrix Proteins/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD40 Antigens/genetics , Cell Proliferation/genetics , Disease Models, Animal , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral , Germinal Center/immunology , Germinal Center/metabolism , Herpesvirus 4, Human/pathogenicity , Humans , Mice , Perforin/deficiency , Perforin/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology , Viral Matrix Proteins/biosynthesis
12.
Proc Natl Acad Sci U S A ; 113(18): 5065-70, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27099294

ABSTRACT

Although canonical NF-κB signaling is crucial to generate a normal mature B-cell compartment, its role in the persistence of resting mature B cells is controversial. To resolve this conflict, we ablated NF-κB essential modulator (NEMO) and IκB kinase 2 (IKK2), two essential mediators of the canonical pathway, either early on in B-cell development or specifically in mature B cells. Early ablation severely inhibited the generation of all mature B-cell subsets, but follicular B-cell numbers could be largely rescued by ectopic expression of B-cell lymphoma 2 (Bcl2), despite a persisting block at the transitional stage. Marginal zone (MZ) B and B1 cells were not rescued, indicating a possible role of canonical NF-κB signals beyond the control of cell survival in these subsets. When canonical NF-κB signaling was ablated specifically in mature B cells, the differentiation and/or persistence of MZ B cells was still abrogated, but follicular B-cell numbers were only mildly affected. However, the mutant cells exhibited increased turnover as well as functional deficiencies upon activation, suggesting that canonical NF-κB signals contribute to their long-term persistence and functional fitness.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Survival/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Cells, Cultured , Mice , Mice, Inbred C57BL
13.
Curr Top Microbiol Immunol ; 393: 211-231, 2016.
Article in English | MEDLINE | ID: mdl-26275875

ABSTRACT

Mitogen-activated protein kinases (MAPKs) play roles in a cell type and context-dependent manner to convert extracellular stimuli to a variety of cellular responses, thereby directing cells to proliferation, differentiation, survival, apoptosis, and migration. Studies of genetically engineered mice or chemical inhibitors specific to each MAPK signaling pathway revealed that MAPKs have various, but non-redundant physiologically important roles among different families. MAPK cascades are obviously integrated in the B cell receptor signaling pathways as critical components to drive B cell-mediated immunity.


Subject(s)
B-Lymphocytes/enzymology , Extracellular Signal-Regulated MAP Kinases/immunology , MAP Kinase Signaling System , Animals , B-Lymphocytes/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Humans , Immunity, Cellular
14.
Immunity ; 28(4): 499-508, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18356083

ABSTRACT

The pre-B cell receptor (pre-BCR) plays a crucial role in the development of immature B cells. Although certain aspects of proximal pre-BCR signaling have been studied, the intermediate signal transducers and the distal transcription modulators are poorly characterized. Here, we demonstrate that deletion of both Erk1 and Erk2 kinases was associated with defective pre-BCR-mediated cell expansion as well as a block in the transition of pro-B to pre-B cells. Phosphorylation of transcription factors Elk1 and CREB was mediated by Erk, and a dominant-negative mutation in the Erk-mediated phosphorylation sites of Elk1 or CREB suppressed pre-BCR-mediated cell expansion as well as expression of genes including Myc, which is involved in the cell-cycle progression. Together, our results identify a crucial role for Erk kinases in regulating B cell development by initiating transcriptional regulatory network and thereby pre-BCR-mediated cell expansion.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/enzymology , Cell Differentiation/immunology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Pre-B Cell Receptors/physiology , Signal Transduction/immunology , Transcription, Genetic/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Gene Deletion , Mice , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/deficiency , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/deficiency , Mitogen-Activated Protein Kinase 3/genetics , Pre-B Cell Receptors/metabolism , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/enzymology , Stem Cells/immunology , Stem Cells/metabolism
15.
Sci Immunol ; 9(92): eadi0042, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38306418

ABSTRACT

Familial hemophagocytic lymphohistiocytosis (FHL) is an inherited, often fatal immune deficiency characterized by severe systemic hyperinflammation. Although allogeneic bone marrow transplantation can be curative, more effective therapies are urgently needed. FHL is caused by inactivating mutations in proteins that regulate cellular immunity. Here, we used an adeno-associated virus-based CRISPR-Cas9 system with an inhibitor of nonhomologous end joining to repair such mutations in potentially long-lived T cells ex vivo. Repaired CD8 memory T cells efficiently cured lethal hyperinflammation in a mouse model of Epstein-Barr virus-triggered FHL2, a subtype caused by perforin-1 (Prf1) deficiency. Furthermore, repair of PRF1 and Munc13-4 (UNC13D)-whose deficiency causes the FHL subtype FHL3-in mutant memory T cells from two critically ill patients with FHL restored T cell cytotoxicity. These results provide a starting point for the treatment of genetic T cell immune dysregulation syndromes with repaired autologous T cells.


Subject(s)
Epstein-Barr Virus Infections , Lymphohistiocytosis, Hemophagocytic , Animals , Mice , Humans , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphohistiocytosis, Hemophagocytic/therapy , CRISPR-Cas Systems , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/therapy , Memory T Cells , Herpesvirus 4, Human , Membrane Proteins/genetics
16.
Cancers (Basel) ; 15(11)2023 May 24.
Article in English | MEDLINE | ID: mdl-37296852

ABSTRACT

Epstein-Barr virus (EBV) is a ubiquitous herpesvirus, but also the first discovered human tumor virus [...].

17.
Exp Gerontol ; 174: 112130, 2023 04.
Article in English | MEDLINE | ID: mdl-36822486

ABSTRACT

Senescent cells were recently shown to play a role in aging-related malfunctions and pathologies. This consensus has been facilitated by evidence from senolytic model mice capable of eliminating senescent cells in tissues using well-characterized senescent markers, such as p16INK4a (hereafter p16). However, since the incomplete or artificial gene expression regulatory regions of manipulated marker genes affect their cognate expression, it currently remains unclear whether these models accurately reflect physiological senescence. We herein describe a novel approach to eliminate p16-expressing cells from mice at any given point in time, generating a new type of knock-in model, p16hCD2 mice and a toxin-conjugated anti-human CD2 antibody (hCD2-SAP) as an inducer. p16hCD2 mice possess an intact Cdkn2a locus that includes a p16 coding region and human CD2 (hCD2) expression unit. We confirmed cognate p16-associated hCD2 expression in mouse embryonic fibroblasts (MEFs) and in several tissues, such as the spleen, liver, and skin. We detected chronological increases in the hCD2-positive population in T lymphocytes that occurred in a p16-dependent manner, which reflected physiological aging. We then confirmed the high sensitivity of hCD2-SAP to hCD2 and validated its efficacy to remove p16-positive cells, particularly in T lymphocytes. The multiple administration of hCD2-SAP for a prolonged p16-positive cell deficiency partially restored aging-related phenotypes in T lymphocytes, such as the contraction of the CD4+ naïve population and expansion of senescence-associated T cells. Our novel approach of targeting p16-positive senescent cells will provide novel insights into the mechanisms underlying physiological aging in vivo.


Subject(s)
Immunotoxins , T-Lymphocytes , Mice , Animals , Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Immunotoxins/genetics , Immunotoxins/metabolism , Senotherapeutics , Fibroblasts/metabolism , Phenotype , CD4-Positive T-Lymphocytes
18.
Commun Biol ; 6(1): 395, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37041231

ABSTRACT

The decrease of antibody efficacy to mutated SARS-CoV-2 spike RBD explains the breakthrough infections and reinfections by Omicron variants. Here, we analyzed broadly neutralizing antibodies isolated from long-term hospitalized convalescent patients of early SARS-CoV-2 strains. One of the antibodies named NCV2SG48 is highly potent to broad SARS-CoV-2 variants including Omicron BA.1, BA.2, and BA.4/5. To reveal the mode of action, we determined the sequence and crystal structure of the Fab fragment of NCV2SG48 in a complex with spike RBD from the original, Delta, and Omicron BA.1. NCV2SG48 is from a minor VH but the multiple somatic hypermutations contribute to a markedly extended binding interface and hydrogen bonds to interact with conserved residues at the core receptor-binding motif of RBD, which efficiently neutralizes a broad spectrum of variants. Thus, eliciting the RBD-specific B cells to the longitudinal germinal center reaction confers potent immunity to broad SARS-CoV-2 variants emerging one after another.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antibodies , Immunoglobulin Fab Fragments
19.
Cells ; 11(11)2022 05 26.
Article in English | MEDLINE | ID: mdl-35681449

ABSTRACT

Isolation of both lymphocytes and myogenic cells from muscle tissue is required for elucidating the cellular and molecular mechanisms of muscle regeneration. Here, we aimed to establish an optimal method obtaining a high yield of lymphocytes during muscle regeneration. After the muscle injury, we observed higher infiltration of lymphocytic cells in the muscle on day 3 after injury. Then, we compared two different white blood cell isolation methods, the Percoll gradient and CD45-magnetic bead methods, to assess the percentage and number of T and B cells. Flow cytometry analysis showed that the CD45-magnetic bead method has a better efficiency in isolating CD4+, CD8+ T cells, and B cells from injured muscle tissues of wild-type and mdx mice than that by the Percoll gradient method. Moreover, we found that the CD45-negative fraction from wild-type and mdx mice includes myogenic cells. In conclusion, we report that the CD45-magnetic bead method is suitable to isolate T and B cells during muscle regeneration with higher purity and yield and can also isolate myogenic cells within the same sample. This method provides a technical basis for further studies on muscle regeneration, involving lymphocytes and muscle cells, with a wide range of clinical applications.


Subject(s)
CD8-Positive T-Lymphocytes , Muscle Development , Animals , Flow Cytometry/methods , Mice , Mice, Inbred mdx , Muscles
20.
Cancers (Basel) ; 14(17)2022 Aug 25.
Article in English | MEDLINE | ID: mdl-36077655

ABSTRACT

Most people infected by EBV acquire specific immunity, which then controls latent infection throughout their life. Immune surveillance of EBV-infected cells by cytotoxic CD4+ T cells has been recognized; however, the molecular mechanism of generating cytotoxic effector T cells of the CD4+ subset remains poorly understood. Here we compared phenotypic features and the transcriptome of EBV-specific effector-memory CD4+ T cells and CD8+ T cells in mice and found that both T cell types show cytotoxicity and, to our surprise, widely similar gene expression patterns relating to cytotoxicity. Similar to cytotoxic CD8+ T cells, EBV-specific cytotoxic CD4+ T cells from human peripheral blood expressed T-bet, Granzyme B, and Perforin and upregulated the degranulation marker, CD107a, immediately after restimulation. Furthermore, T-bet expression in cytotoxic CD4+ T cells was highly correlated with Granzyme B and Perforin expression at the protein level. Thus, differentiation of EBV-specific cytotoxic CD4+ T cells is possibly controlled by mechanisms shared by cytotoxic CD8+ T cells. T-bet-mediated transcriptional regulation may explain the similarity of cytotoxic effector differentiation between CD4+ T cells and CD8+ T cells, implicating that this differentiation pathway may be directed by environmental input rather than T cell subset.

SELECTION OF CITATIONS
SEARCH DETAIL