Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 21(6): 649-659, 2020 06.
Article in English | MEDLINE | ID: mdl-32424359

ABSTRACT

Efficient generation of germinal center (GC) responses requires directed movement of B cells between distinct microenvironments underpinned by specialized B cell-interacting reticular cells (BRCs). How BRCs are reprogrammed to cater to the developing GC remains unclear, and studying this process is largely hindered by incomplete resolution of the cellular composition of the B cell follicle. Here we used genetic targeting of Cxcl13-expressing cells to define the molecular identity of the BRC landscape. Single-cell transcriptomic analysis revealed that BRC subset specification was predetermined in the primary B cell follicle. Further topological remodeling of light and dark zone follicular dendritic cells required CXCL12-dependent crosstalk with B cells and dictated GC output by retaining B cells in the follicle and steering their interaction with follicular helper T cells. Together, our results reveal that poised BRC-defined microenvironments establish a feed-forward system that determines the efficacy of the GC reaction.


Subject(s)
Darkness , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Immunomodulation/radiation effects , Light , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Cell Communication , Chemokine CXCL12/metabolism , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Immunity ; 52(3): 542-556.e13, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187520

ABSTRACT

Fibrosis is an incurable disorder of unknown etiology. Segregated-nucleus-containing atypical monocytes (SatMs) are critical for the development of fibrosis. Here we examined the mechanisms that recruit SatMs to pre-fibrotic areas. A screen based on cytokine expression in the fibrotic lung revealed that the chemokine Cxcl12, which is produced by apoptotic nonhematopoietic cells, was essential for SatM recruitment. Analyses of lung tissues at fibrosis onset showed increased expression of Rbm7, a component of the nuclear exosome targeting complex. Rbm7 deletion suppressed bleomycin-induced fibrosis and at a cellular level, suppressed apoptosis of nonhematopoietic cells. Mechanistically, Rbm7 bound to noncoding (nc)RNAs that form subnuclear bodies, including Neat1 speckles. Dysregulated expression of Rbm7 resulted in the nuclear degradation of Neat1 speckles, the dispersion of the DNA repair protein BRCA1, and the triggering of apoptosis. Thus, Rbm7 in epithelial cells plays a critical role in the development of fibrosis by regulating ncRNA decay and thereby the production of chemokines that recruit SatMs.


Subject(s)
Apoptosis/immunology , Cell Nucleus/immunology , Exosomes/immunology , Pulmonary Fibrosis/immunology , RNA-Binding Proteins/immunology , Animals , Apoptosis/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Exosomes/genetics , Exosomes/metabolism , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , NIH 3T3 Cells , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
Cell ; 153(5): 1025-35, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23706740

ABSTRACT

Unique among leukocytes, neutrophils follow daily cycles of release from and migration back into the bone marrow, where they are eliminated. Because removal of dying cells generates homeostatic signals, we explored whether neutrophil elimination triggers circadian events in the steady state. Here, we report that the homeostatic clearance of neutrophils provides cues that modulate the physiology of the bone marrow. We identify a population of CD62L(LO) CXCR4(HI) neutrophils that have "aged" in the circulation and are eliminated at the end of the resting period in mice. Aged neutrophils infiltrate the bone marrow and promote reductions in the size and function of the hematopoietic niche. Modulation of the niche depends on macrophages and activation of cholesterol-sensing nuclear receptors and is essential for the rhythmic egress of hematopoietic progenitors into the circulation. Our results unveil a process that synchronizes immune and hematopoietic rhythms and expand the ascribed functions of neutrophils beyond inflammation. PAPERFLICK:


Subject(s)
Bone Marrow/physiology , Circadian Rhythm , Neutrophils/cytology , Neutrophils/physiology , Animals , Cell Movement , Cellular Senescence , Female , Hematopoietic Stem Cells/metabolism , Homeostasis , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Orphan Nuclear Receptors/metabolism
4.
Immunity ; 48(2): 286-298.e6, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29396162

ABSTRACT

Glucocorticoids are steroid hormones with strong anti-inflammatory and immunosuppressive effects that are produced in a diurnal fashion. Although glucocorticoids have the potential to induce interleukin-7 receptor (IL-7R) expression in T cells, whether they control T cell homeostasis and responses at physiological concentrations remains unclear. We found that glucocorticoid receptor signaling induces IL-7R expression in mouse T cells by binding to an enhancer of the IL-7Rα locus, with a peak at midnight and a trough at midday. This diurnal induction of IL-7R supported the survival of T cells and their redistribution between lymph nodes, spleen, and blood by controlling expression of the chemokine receptor CXCR4. In mice, T cell accumulation in the spleen at night enhanced immune responses against soluble antigens and systemic bacterial infection. Our results reveal the immunoenhancing role of glucocorticoids in adaptive immunity and provide insight into how immune function is regulated by the diurnal rhythm.


Subject(s)
Circadian Rhythm/physiology , Glucocorticoids/pharmacology , Receptors, CXCR4/physiology , Receptors, Interleukin-7/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Chemokine CXCL12/biosynthesis , Female , Immunologic Memory , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/physiology
5.
Genes Dev ; 32(5-6): 359-372, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29563184

ABSTRACT

Bone marrow is the tissue filling the space between bone surfaces. Hematopoietic stem cells (HSCs) are maintained by special microenvironments known as niches within bone marrow cavities. Mesenchymal cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells or leptin receptor-positive (LepR+) cells, are a major cellular component of HSC niches that gives rise to osteoblasts in bone marrow. However, it remains unclear how osteogenesis is prevented in most CAR/LepR+ cells to maintain HSC niches and marrow cavities. Here, using lineage tracing, we found that the transcription factor early B-cell factor 3 (Ebf3) is preferentially expressed in CAR/LepR+ cells and that Ebf3-expressing cells are self-renewing mesenchymal stem cells in adult marrow. When Ebf3 is deleted in CAR/LepR+ cells, HSC niche function is severely impaired, and bone marrow is osteosclerotic with increased bone in aged mice. In mice lacking Ebf1 and Ebf3, CAR/LepR+ cells exhibiting a normal morphology are abundantly present, but their niche function is markedly impaired with depleted HSCs in infant marrow. Subsequently, the mutants become progressively more osteosclerotic, leading to the complete occlusion of marrow cavities in early adulthood. CAR/LepR+ cells differentiate into bone-producing cells with reduced HSC niche factor expression in the absence of Ebf1/Ebf3 Thus, HSC cellular niches express Ebf3 that is required to create HSC niches, to inhibit their osteoblast differentiation, and to maintain spaces for HSCs.


Subject(s)
Bone Marrow/metabolism , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Age Factors , Animals , Bone Marrow/pathology , Cell Differentiation , Cell Lineage , Gene Expression Regulation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred C57BL , Osteogenesis/genetics , Stem Cell Niche , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
6.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38430523

ABSTRACT

Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche.


Subject(s)
B-Lymphocytes , Bone Marrow , Chemokine CXCL12 , Malaria , Mice, Inbred C57BL , Animals , Chemokine CXCL12/metabolism , Chemokine CXCL12/immunology , Mice , Malaria/immunology , Malaria/parasitology , B-Lymphocytes/immunology , Bone Marrow/immunology , Bone Marrow/parasitology , Stem Cell Niche/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism
7.
Immunity ; 45(6): 1219-1231, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27913094

ABSTRACT

Hematopoietic stem cells (HSCs) self-renew in bone marrow niches formed by mesenchymal progenitors and endothelial cells expressing the chemokine CXCL12, but whether a separate niche instructs multipotent progenitor (MPP) differentiation remains unclear. We show that MPPs resided in HSC niches, where they encountered lineage-instructive differentiation signals. Conditional deletion of the chemokine receptor CXCR4 in MPPs reduced differentiation into common lymphoid progenitors (CLPs), which decreased lymphopoiesis. CXCR4 was required for CLP positioning near Interleukin-7+ (IL-7) cells and for optimal IL-7 receptor signaling. IL-7+ cells expressed CXCL12 and the cytokine SCF, were mesenchymal progenitors capable of differentiation into osteoblasts and adipocytes, and comprised a minor subset of sinusoidal endothelial cells. Conditional Il7 deletion in mesenchymal progenitors reduced B-lineage committed CLPs, while conditional Cxcl12 or Scf deletion from IL-7+ cells reduced HSC and MPP numbers. Thus, HSC maintenance and multilineage differentiation are distinct cell lineage decisions that are both controlled by HSC niches.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/cytology , Multipotent Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Cell Lineage/physiology , Cell Separation , Chemokine CXCL2/metabolism , Flow Cytometry , Interleukin-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic
8.
Immunity ; 42(1): 13-4, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25607454

ABSTRACT

The nature and functions of cells creating hematopoietic niches during inflammation remain incompletely understood. In this issue of Immunity, Kwak et al. (2015) reveal that myeloid cell-produced reactive oxygen species stimulate proliferation of myeloid progenitors establishing an additional mechanism to regulate hematopoiesis.


Subject(s)
Escherichia coli Infections/immunology , Escherichia coli/immunology , Granulocytes/physiology , Hematopoiesis , Myeloid Cells/physiology , Myeloid Progenitor Cells/physiology , Animals
9.
Nature ; 563(7730): 254-258, 2018 11.
Article in English | MEDLINE | ID: mdl-30401834

ABSTRACT

Skeletal stem cells regulate bone growth and homeostasis by generating diverse cell types, including chondrocytes, osteoblasts and marrow stromal cells. The emerging concept postulates that there exists a distinct type of skeletal stem cell that is closely associated with the growth plate1-4, which is a type of cartilaginous tissue that has critical roles in bone elongation5. The resting zone maintains the growth plate by expressing parathyroid hormone-related protein (PTHrP), which interacts with Indian hedgehog (Ihh) that is released from the hypertrophic zone6-10, and provides a source of other chondrocytes11. However, the identity of skeletal stem cells and how they are maintained in the growth plate are unknown. Here we show, in a mouse model, that skeletal stem cells are formed among PTHrP-positive chondrocytes within the resting zone of the postnatal growth plate. PTHrP-positive chondrocytes expressed a panel of markers for skeletal stem and progenitor cells, and uniquely possessed the properties of skeletal stem cells in cultured conditions. Cell-lineage analysis revealed that PTHrP-positive chondrocytes in the resting zone continued to form columnar chondrocytes in the long term; these chondrocytes underwent hypertrophy, and became osteoblasts and marrow stromal cells beneath the growth plate. Transit-amplifying chondrocytes in the proliferating zone-which was concertedly maintained by a forward signal from undifferentiated cells (PTHrP) and a reverse signal from hypertrophic cells (Ihh)-provided instructive cues to maintain the cell fates of PTHrP-positive chondrocytes in the resting zone. Our findings unravel a type of somatic stem cell that is initially unipotent and acquires multipotency at the post-mitotic stage, underscoring the malleable nature of the skeletal cell lineage. This system provides a model in which functionally dedicated stem cells and their niches are specified postnatally, and maintained throughout tissue growth by a tight feedback regulation system.


Subject(s)
Growth Plate/cytology , Stem Cells/cytology , Animals , Cell Lineage , Chondrocytes/cytology , Chondrocytes/metabolism , Growth Plate/metabolism , In Vitro Techniques , Mice , Osteoblasts/cytology , Parathyroid Hormone-Related Protein/metabolism , Stem Cell Niche , Stem Cells/metabolism , Stromal Cells/cytology
10.
J Med Internet Res ; 26: e51749, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38373022

ABSTRACT

BACKGROUND: Given the global shortage of child psychiatrists and barriers to specialized care, remote assessment is a promising alternative for diagnosing and managing attention-deficit/hyperactivity disorder (ADHD). However, only a few studies have validated the accuracy and acceptability of these remote methods. OBJECTIVE: This study aimed to test the agreement between remote and face-to-face assessments. METHODS: Patients aged between 6 and 17 years with confirmed Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition diagnoses of ADHD or autism spectrum disorder (ASD) were recruited from multiple institutions. In a randomized order, participants underwent 2 evaluations, face-to-face and remotely, with distinct evaluators administering the ADHD Rating Scale-IV (ADHD-RS-IV). Intraclass correlation coefficient (ICC) was used to assess the reliability of face-to-face and remote assessments. RESULTS: The participants included 74 Japanese children aged between 6 and 16 years who were primarily diagnosed with ADHD (43/74, 58%) or ASD (31/74, 42%). A total of 22 (30%) children were diagnosed with both conditions. The ADHD-RS-IV ICCs between face-to-face and remote assessments showed "substantial" agreement in the total ADHD-RS-IV score (ICC=0.769, 95% CI 0.654-0.849; P<.001) according to the Landis and Koch criteria. The ICC in patients with ADHD showed "almost perfect" agreement (ICC=0.816, 95% CI 0.683-0.897; P<.001), whereas in patients with ASD, it showed "substantial" agreement (ICC=0.674, 95% CI 0.420-0.831; P<.001), indicating the high reliability of both methods across both conditions. CONCLUSIONS: Our study validated the feasibility and reliability of remote ADHD testing, which has potential benefits such as reduced hospital visits and time-saving effects. Our results highlight the potential of telemedicine in resource-limited areas, clinical trials, and treatment evaluations, necessitating further studies to explore its broader application. TRIAL REGISTRATION: UMIN Clinical Trials Registry UMIN000039860; http://tinyurl.com/yp34x6kh.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Autism Spectrum Disorder , Neurodevelopmental Disorders , Psychiatry , Telemedicine , Adolescent , Child , Humans , Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/therapy , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/therapy , Caregivers , Feasibility Studies , Reproducibility of Results
11.
Dev Biol ; 477: 70-84, 2021 09.
Article in English | MEDLINE | ID: mdl-34015362

ABSTRACT

The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.


Subject(s)
Blood Vessels/embryology , Endothelial Cells/physiology , Neovascularization, Physiologic/physiology , Receptors, CXCR4/physiology , Animals , Blood Vessels/anatomy & histology , Endothelial Cells/metabolism , Gain of Function Mutation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, CXCR4/biosynthesis , Vascular Remodeling/physiology
12.
Blood ; 135(23): 2071-2084, 2020 06 04.
Article in English | MEDLINE | ID: mdl-31990287

ABSTRACT

Sickle cell disease (SCD) is a monogenic red blood cell (RBC) disorder with high morbidity and mortality. Here, we report, for the first time, the impact of SCD on the bone marrow (BM) vascular niche, which is critical for hematopoiesis. In SCD mice, we find a disorganized and structurally abnormal BM vascular network of increased numbers of highly tortuous arterioles occupying the majority of the BM cavity, as well as fragmented sinusoidal vessels filled with aggregates of erythroid and myeloid cells. By in vivo imaging, sickle and control RBCs have significantly slow intravascular flow speeds in sickle cell BM but not in control BM. In sickle cell BM, we find increased reactive oxygen species production in expanded erythroblast populations and elevated levels of HIF-1α. The SCD BM exudate exhibits increased levels of proangiogenic growth factors and soluble vascular cell adhesion molecule-1. Transplantation of SCD mouse BM cells into wild-type mice recapitulates the SCD vascular phenotype. Our data provide a model of SCD BM, in which slow RBC flow and vaso-occlusions further diminish local oxygen availability in the physiologic hypoxic BM cavity. These events trigger a milieu that is conducive to aberrant vessel growth. The distorted neovascular network is completely reversed by a 6-week blood transfusion regimen targeting hemoglobin S to <30%, highlighting the plasticity of the vascular niche. A better insight into the BM microenvironments in SCD might provide opportunities to optimize approaches toward efficient and long-term hematopoietic engraftment in the context of curative therapies.


Subject(s)
Anemia, Sickle Cell/complications , Blood Transfusion/methods , Bone Marrow/pathology , Erythrocytes, Abnormal/pathology , Hematopoiesis , Neovascularization, Pathologic/prevention & control , Splenomegaly/prevention & control , Animals , Bone Marrow/metabolism , Erythrocytes, Abnormal/metabolism , Female , Humans , Male , Mice , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/pathology , Splenomegaly/etiology , Splenomegaly/pathology
13.
Curr Top Microbiol Immunol ; 434: 33-54, 2021.
Article in English | MEDLINE | ID: mdl-34850281

ABSTRACT

Most types of blood cells, including immune cells are generated from hematopoietic stem cells (HSCs) within bone marrow in the adult. Most HSCs are in contact with and require the special microenvironment known as a niche for their maintenance. It has been thought that HSC niches comprise various types of support cells that provide critical signals, including cytokines and extracellular matrix for HSC regulation. However, among these cells, several lines of evidence have demonstrated that the population of bone marrow-specific mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor-expressing (LepR+) cells, is the major cellular component of HSC niches. CAR/LepR+ cells give rise to most adipocytes and osteoblasts in adult bone marrow and express much higher levels of HSC niche factors, including cytokines CXCL12 and stem cell factor (SCF), which are essential for HSC maintenance, and transcription factors Foxc1 and Ebf3, which are essential for the formation and maintenance of HSC niches than other types of cells. CAR/LepR+ cells are present in human bone marrow, undergo fibrotic expansion, and have reduced expression of HSC niche factors in hematopoietic malignancies.


Subject(s)
Hematologic Neoplasms , Neoplasms , Bone Marrow , Hematologic Neoplasms/genetics , Hematopoiesis , Hematopoietic Stem Cells , Homeostasis , Humans , Stem Cell Niche , Tumor Microenvironment
14.
Int Immunol ; 33(12): 821-826, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34668936

ABSTRACT

Most lineages of blood cells, including immune cells, are generated from hematopoietic stem cells (HSCs) in bone marrow throughout adult life. Since HSCs cannot expand on their own, they require and contact the special microenvironments, termed niches for their maintenance. HSC niches comprise supportive cells that provide adjacent HSCs with critical signals, including cytokines. Although bone marrow microenvironments have been thought to be complex, recent studies have demonstrated that the bone marrow-specific population of fibroblastic reticular cells with long processes, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor (LepR)-expressing (LepR+) cells, is the major cellular component of niches for HSCs and lymphoid progenitors. CAR cells have salient features, expressing much higher levels of critical HSC niche factors than any other cell populations and function as self-renewing mesenchymal stem cells. Human counterpart of CAR cells is present and affected in diseases, including leukemia. Foxl1+ telocytes recently identified as the niche for intestinal stem cells share some features with CAR cells, suggesting that CAR cells might serve as a prototype for fibroblastic reticular cells creating niche for long-lived cells, including tissue stem cells and memory lymphocytes. These findings provided the basis for future mechanistic studies on the cross-talk between hematopoietic cells and microenvironments in both health and disease.


Subject(s)
Bone Marrow/immunology , Fibroblasts/immunology , Hematopoietic Stem Cells/immunology , Stem Cell Niche/immunology , Animals , Humans
15.
Immunity ; 39(5): 912-24, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24184055

ABSTRACT

Germinal center (GC) B cells cycle between the dark zone (DZ) and light zone (LZ) during antibody affinity maturation. Whether this movement is necessary for GC function has not been tested. Here we show that CXCR4-deficient GC B cells, which are restricted to the LZ, are gradually outcompeted by WT cells indicating an essential role for DZ access. Remarkably, the transition between DZ centroblast and LZ centrocyte phenotypes occurred independently of positioning. However, CXCR4-deficient cells carried fewer mutations and were overrepresented in the CD73(+) memory compartment. These findings are consistent with a model where GC B cells change from DZ to LZ phenotype according to a timed cellular program but suggest that spatial separation of DZ cells facilitates more effective rounds of mutation and selection. Finally, we identify a network of DZ CXCL12-expressing reticular cells that likely support DZ functions.


Subject(s)
B-Lymphocytes/cytology , Germinal Center/cytology , Lymphopoiesis/physiology , Animals , Antibody Affinity , Antigens, Differentiation, B-Lymphocyte/metabolism , Cell Cycle , Cell Movement , Chemokine CXCL12/analysis , Clonal Selection, Antigen-Mediated , Germinal Center/ultrastructure , Immunologic Memory , Lymph Nodes/ultrastructure , Mediastinum , Mice , Orthomyxoviridae Infections/immunology , Peyer's Patches/cytology , Phenotype , Plasma Cells/cytology , Radiation Chimera , Receptors, CXCR4/analysis , Receptors, CXCR4/deficiency , Specific Pathogen-Free Organisms , Time Factors
16.
J Allergy Clin Immunol ; 148(6): 1575-1588.e7, 2021 12.
Article in English | MEDLINE | ID: mdl-33965431

ABSTRACT

BACKGROUND: Epidemiologic studies have yielded conflicting results regarding the influence of a single bout of prolonged high-intensity exercise on viral infection. OBJECTIVE: We sought to learn whether prolonged high-intensity exercise either exacerbates or ameliorates herpes simplex virus type 2 (HSV-2) infection according to the interval between virus exposure and exercise. METHODS: Mice were intravaginally infected with HSV-2 and exposed to run on the treadmill. RESULTS: Prolonged high-intensity exercise 17 hours after infection impaired the clearance of HSV-2, while exercise 8 hours after infection enhanced the clearance of HSV-2. These impaired or enhanced immune responses were related to a transient decrease or increase in the number of blood-circulating plasmacytoid dendritic cells. Exercise-induced glucocorticoids transiently decreased the number of circulating plasmacytoid dendritic cells by facilitating their homing to the bone marrow via the CXCL12-CXCR4 axis, which led to their subsequent increase in the blood. CONCLUSION: A single bout of prolonged high-intensity exercise can be either deleterious or beneficial to antiviral immunity.


Subject(s)
Dendritic Cells/immunology , Glucocorticoids/metabolism , Herpes Simplex/immunology , Herpesvirus 2, Human/physiology , Animals , Chemokine CXCL12/metabolism , Exercise , Humans , Immunity , Mice , Mice, Inbred C57BL , Models, Animal , Physical Conditioning, Animal , Receptors, CXCR4/metabolism
17.
Br J Haematol ; 193(3): 659-668, 2021 05.
Article in English | MEDLINE | ID: mdl-33837967

ABSTRACT

A population of mesenchymal stem cells, termed CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells or leptin receptor-expressing cells, are the major cellular component of niches for haematopoietic stem cells (HSCs) in murine bone marrow. CAR cells are characterized by several salient features, including much higher expression of CXCL12, stem cell factor (SCF), forkhead box C1 (FOXC1) and early B-cell factor 3 (EBF3), which are essential for HSC maintenance, than other cells. However, the human counterpart of CAR cells has not been fully described. Here, we show the presence of cells expressing much higher CXCL12 than other cells in human adult bone marrow using a flow cytometry-based in situ technique that enables high-throughput detection of mRNA at single-cell resolution. Most CXCL12hi cells expressed high levels of SCF, FOXC1 and EBF3 and had the potential to differentiate into adipocytes and osteoblasts. Histologically, the nuclei of CXCL12hi cells were identified and quantified by EBF3 expression in fixed marrow sections. CXCL12hi cells sorted from residual bone marrow aspirates of chronic myeloid leukaemia patients expressed reduced levels of CXCL12, SCF, FOXC1 and EBF3 in correlation with increased leukaemic burden. Together, we identified the human counterpart of CAR cells, enabling the evaluation of their alterations in various haematological disorders by flow cytometric and histological analyses.


Subject(s)
Chemokine CXCL12/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mesenchymal Stem Cells/metabolism , Neoplasm Proteins/metabolism , Stem Cell Niche , Adult , Female , Forkhead Transcription Factors/metabolism , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mesenchymal Stem Cells/pathology , Stem Cell Factor/metabolism , Transcription Factors/metabolism
18.
Invest New Drugs ; 39(6): 1568-1576, 2021 12.
Article in English | MEDLINE | ID: mdl-34160752

ABSTRACT

BACKGROUND: Talazoparib is a poly(ADP-ribose) polymerase enzyme inhibitor. This open-label, non-randomized, phase 1 study of talazoparib investigated the safety, pharmacokinetics, and preliminary antitumor activity in Japanese patients with locally advanced or metastatic solid tumors, regardless of mutations in DNA damage repair-related genes, who are resistant to/ineligible for standard therapies. METHODS: Patients received talazoparib dosed orally at 0.75 or 1 mg once daily using a modified 3 + 3 dose-escalation scheme. Primary endpoint was dose-limiting toxicities during the first cycle of talazoparib. RESULTS: Nine patients (median age 62.0 years) were included: 3 and 6 patients at the 0.75 and 1.0 mg once-daily dose levels, respectively. No dose-limiting toxicities were reported. The most commonly reported treatment-emergent adverse events (≥2 patients) were anemia, stomatitis, maculopapular rash, platelet count decreased, neutrophil count decreased, and alanine aminotransferase increased. Three patients had grade ≥ 3 treatment-emergent adverse events (anemia, brain metastases [1 patient each], and neutrophil and white blood cell count decreased [same patient]). Two patients temporarily discontinued treatment due to a treatment-emergent adverse event, and 1 patient required a dose reduction for neutrophil count decreased (all at 1 mg once daily). Talazoparib exposure (Cmax and AUC) after single and multiple dosing was slightly higher proportionally with talazoparib 1 mg than talazoparib 0.75 mg. The overall disease control rate was 44.4%, including 2 patients with stable disease. The recommended phase 2 dose of talazoparib was established as 1 mg once daily. CONCLUSIONS: Single-agent talazoparib was well tolerated and had preliminary antitumor activity in Japanese patients with advanced solid tumors. ClinicalTrials.gov identifier: NCT03343054 (November 17, 2017).


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Phthalazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Asian People , Dose-Response Relationship, Drug , Humans , Japan , Maximum Tolerated Dose , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasms/pathology , Phthalazines/administration & dosage , Phthalazines/adverse effects , Phthalazines/pharmacokinetics , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/adverse effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics
19.
Cancer Sci ; 111(10): 3726-3738, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32681682

ABSTRACT

Lorlatinib is a potent, brain-penetrant, third-generation anaplastic lymphoma kinase (ALK)/ROS proto-oncogene 1 (ROS1) tyrosine kinase inhibitor (TKI) that is active against most known resistance mutations. This is an ongoing phase 1/2, multinational study (NCT01970865) investigating the efficacy, safety and pharmacokinetics of lorlatinib in ALK-rearranged/ROS1-rearranged advanced non-small cell lung cancer (NSCLC) with or without intracranial (IC) metastases. Because patterns of ALK TKI use in Japan differ from other regions, we present a subgroup analysis of Japanese patients. Patients were enrolled into six expansion (EXP) cohorts based on ALK/ROS1 mutation status and treatment history. The primary endpoint was the objective response rate (ORR) and the IC-ORR based on independent central review. Secondary endpoints included pharmacokinetic evaluations. At data cutoff, 39 ALK-rearranged/ROS1-rearranged Japanese patients were enrolled across the six expansion cohorts; all received lorlatinib 100 mg once daily. Thirty-one ALK-rearranged patients previously treated with ≥1 ALK TKI (EXP2 to EXP5) were evaluable for ORR and 15 were evaluable for IC-ORR. The ORR and the IC-ORR for Japanese patients in EXP2-5 were 54.8% (95% confidence interval [CI]: 36.0-72.7) and 46.7% (95% CI: 21.3-73.4), respectively. Among patients who had received prior alectinib only (EXP3B), the ORR was 42.9%; 95% CI: 9.9-81.6). The most common treatment-related adverse event (TRAE) was hypercholesterolemia (79.5%). Hypertriglyceridemia was the most common grade 3/4 TRAE (25.6%). Single-dose and multiple-dose pharmacokinetic profiles among Japanese patients were similar to those in non-Japanese patients. Lorlatinib showed clinically meaningful responses and IC responses among ALK-rearranged Japanese patients with NSCLC who received ≥1 prior ALK TKI, including meaningful responses among those receiving prior alectinib only. Lorlatinib was generally well tolerated.


Subject(s)
Brain Neoplasms/drug therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Lactams, Macrocyclic/administration & dosage , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Aged , Aged, 80 and over , Aminopyridines , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/drug effects , Drug-Related Side Effects and Adverse Reactions/epidemiology , Humans , Hypercholesterolemia/chemically induced , Hypercholesterolemia/epidemiology , Lactams , Lactams, Macrocyclic/adverse effects , Middle Aged , Mutation/drug effects , Neoplasm Metastasis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles
20.
Int Immunol ; 31(1): 5-11, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30169696

ABSTRACT

The special microenvironments, termed niches, with which hematopoietic stem cells (HSCs) are in contact, have been thought to be required for the maintenance of HSCs and the generation of immune cells in bone marrow. Although the identity of the HSC niche has been a subject of long-standing debate, recent findings demonstrate that a population of mesenchymal stem cells, termed CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells or leptin receptor-expressing (LepR+) cells, are the major cellular components of niches for HSCs and lymphoid progenitors, which express specific transcription factors, including Foxc1 and Ebf3, and cytokines, including CXCL12 and stem cell factor (SCF), essential for their niche functions. The identity and functions of other types of cells, including osteoblasts, sinusoidal endothelial cells, periarteriolar cells, megakaryocytes and a population of macrophages in HSC maintenance, have also been shown.


Subject(s)
Cell Differentiation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Immune System/cytology , Immune System/immunology , Stem Cell Niche , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Immune System/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Schwann Cells/immunology , Schwann Cells/metabolism , Stem Cell Niche/genetics , Stem Cell Niche/immunology
SELECTION OF CITATIONS
SEARCH DETAIL