Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nat Immunol ; 20(1): 86-96, 2019 01.
Article in English | MEDLINE | ID: mdl-30538335

ABSTRACT

Germinal center (GC) B cells feature repression of many gene enhancers to establish their characteristic transcriptome. Here we show that conditional deletion of Lsd1 in GCs significantly impaired GC formation, associated with failure to repress immune synapse genes linked to GC exit, which are also direct targets of the transcriptional repressor BCL6. We found that BCL6 directly binds LSD1 and recruits it primarily to intergenic and intronic enhancers. Conditional deletion of Lsd1 suppressed GC hyperplasia caused by constitutive expression of BCL6 and significantly delayed BCL6-driven lymphomagenesis. Administration of catalytic inhibitors of LSD1 had little effect on GC formation or GC-derived lymphoma cells. Using a CRISPR-Cas9 domain screen, we found instead that the LSD1 Tower domain was critical for dependence on LSD1 in GC-derived B cells. These results indicate an essential role for LSD1 in the humoral immune response, where it modulates enhancer function by forming repression complexes with BCL6.


Subject(s)
B-Lymphocytes/physiology , Germinal Center/pathology , Histone Demethylases/metabolism , Lymphoma/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , Animals , CRISPR-Cas Systems , Carcinogenesis , DNA, Intergenic/genetics , Germinal Center/immunology , Histone Demethylases/genetics , Hyperplasia , Immunological Synapses/genetics , Introns/genetics , Lymphoma/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics
2.
Cell Mol Life Sci ; 81(1): 29, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38212474

ABSTRACT

Involution of the mammary gland after lactation is a dramatic example of coordinated cell death. Weaning causes distension of the alveolar structures due to the accumulation of milk, which, in turn, activates STAT3 and initiates a caspase-independent but lysosome-dependent cell death (LDCD) pathway. Although the importance of STAT3 and LDCD in early mammary involution is well established, it has not been entirely clear how milk stasis activates STAT3. In this report, we demonstrate that protein levels of the PMCA2 calcium pump are significantly downregulated within 2-4 h of experimental milk stasis. Reductions in PMCA2 expression correlate with an increase in cytoplasmic calcium in vivo as measured by multiphoton intravital imaging of GCaMP6f fluorescence. These events occur concomitant with the appearance of nuclear pSTAT3 expression but prior to significant activation of LDCD or its previously implicated mediators such as LIF, IL6, and TGFß3, all of which appear to be upregulated by increased intracellular calcium. We further demonstrate that increased intracellular calcium activates STAT3 by inducing degradation of its negative regulator, SOCS3. We also observed that milk stasis, loss of PMCA2 expression and increased intracellular calcium levels activate TFEB, an important regulator of lysosome biogenesis through a process involving inhibition of CDK4/6 and cell cycle progression. In summary, these data suggest that intracellular calcium serves as an important proximal biochemical signal linking milk stasis to STAT3 activation, increased lysosomal biogenesis, and lysosome-mediated cell death.


Subject(s)
Calcium , Milk , Female , Animals , Milk/metabolism , Calcium/metabolism , Cell Death , Lactation , Lysosomes/metabolism , Mammary Glands, Animal/metabolism , STAT3 Transcription Factor/metabolism
3.
Nat Immunol ; 13(1): 44-50, 2011 Nov 27.
Article in English | MEDLINE | ID: mdl-22120118

ABSTRACT

Mouse invariant natural killer T cells (iNKT cells) provide cognate and noncognate help for lipid and protein-specific B cells, respectively. However, the long-term outcome for B cells after cognate help is provided by iNKT cells is unknown at present. Here we found that cognate iNKT cell help resulted in a B cell differentiation program characterized by extrafollicular plasmablasts, germinal-center formation, affinity maturation and a robust primary immunoglobulin G (IgG) antibody response that was uniquely dependent on iNKT cell-derived interleukin 21 (IL-21). However, cognate help from iNKT cells did not generate an enhanced humoral memory response. Thus, cognate iNKT cell help for lipid-specific B cells induces a unique signature that is a hybrid of classic T cell-dependent and T cell-independent type 2 B cell responses.


Subject(s)
Antigens/immunology , B-Lymphocytes/immunology , Interleukins/physiology , Lipids/immunology , Natural Killer T-Cells/immunology , Animals , Germinal Center/immunology , Immunity, Humoral , Immunologic Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Spleen/immunology
4.
Immunol Rev ; 288(1): 10-27, 2019 03.
Article in English | MEDLINE | ID: mdl-30874342

ABSTRACT

Throughout the developing GC response, B cell survival and fate choices made at the single cell level are dependent on signals received largely through interactions with other cells, often with cognate T cells. The type of signals that a given B cell can encounter is dictated by its location within tissue microarchitecture. The focus of this review is on the initiation and evolution of the GC response at the earliest time points. Here, we review the key factors influencing the progression of GC B cell differentiation that are both stage and context dependent. Finally, we describe the coevolution of niches within and surrounding the GC that influence the outcome of the GC response.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Stromal Cells/physiology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Humans , Lymphocyte Activation , Paracrine Communication , Signal Transduction
5.
Nature ; 522(7554): 94-7, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25849774

ABSTRACT

Tissue homeostasis is achieved through a balance of cell production (growth) and elimination (regression). In contrast to tissue growth, the cells and molecular signals required for tissue regression remain unknown. To investigate physiological tissue regression, we use the mouse hair follicle, which cycles stereotypically between phases of growth and regression while maintaining a pool of stem cells to perpetuate tissue regeneration. Here we show by intravital microscopy in live mice that the regression phase eliminates the majority of the epithelial cells by two distinct mechanisms: terminal differentiation of suprabasal cells and a spatial gradient of apoptosis of basal cells. Furthermore, we demonstrate that basal epithelial cells collectively act as phagocytes to clear dying epithelial neighbours. Through cellular and genetic ablation we show that epithelial cell death is extrinsically induced through transforming growth factor (TGF)-ß activation and mesenchymal crosstalk. Strikingly, our data show that regression acts to reduce the stem cell pool, as inhibition of regression results in excess basal epithelial cells with regenerative abilities. This study identifies the cellular behaviours and molecular mechanisms of regression that counterbalance growth to maintain tissue homeostasis.


Subject(s)
Cell Death , Epithelial Cells/cytology , Hair Follicle/cytology , Phagocytosis , Stem Cell Niche/physiology , Stem Cells/cytology , Animals , Apoptosis , Dermis/cytology , Dermis/metabolism , Epithelial Cells/metabolism , Hair Follicle/metabolism , Homeostasis , Mice , Phagocytes/cytology , Regeneration , Signal Transduction , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , beta Catenin/metabolism
6.
Immunity ; 34(6): 947-60, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21636295

ABSTRACT

We identify the interfollicular (IF) zone as the site where germinal center B cell and T follicular helper (Tfh) cell differentiation initiates. For the first 2 days postimmunization, antigen-specific T and B cells remained confined within the IF zone, formed long-lived interactions, and upregulated the transcriptional repressor Bcl6. T cells also acquired the Tfh cell markers CXCR5, PD-1, and GL7. Responding B and T cells migrated to the follicle interior directly from the IF zone, T cell immigration preceding B cells by 1 day. Notably, in the absence of cognate B cells, Tfh cells still formed and migrated to the follicle. However, without such B cells, PD-1, ICOS, and GL7 were no longer expressed on follicular Bcl6(hi) T cells that nevertheless persisted in the follicle. Thus, Ag-specific B cells are required for the maintenance of the PD-1(hi)ICOS(hi)GL7(hi) Tfh cell phenotype within the follicle, but not for their initial differentiation in the IF zone.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation , Germinal Center/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens/immunology , B-Lymphocytes/cytology , Cell Movement , Germinal Center/cytology , Mice , Phenotype , T-Lymphocytes, Helper-Inducer/cytology
7.
J Immunol ; 201(12): 3569-3579, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30446568

ABSTRACT

We examined the unique contributions of the cytokines IL-21 and IL-4 on germinal center (GC) B cell initiation and subsequent maturation in a murine model system. Similar to other reports, we found T follicular helper cell expression of IL-21 begins prior to T follicular helper cell migration into the B cell follicle and precedes that of IL-4. Consistent with this timing, IL-21 signaling has a greater influence on the perifollicular pre-GC B cell transition to the intrafollicular stage. Notably, Bcl6hi B cells can form in the combined absence of IL-21R- and STAT6-derived signals; however, these nascent GC B cells cease to proliferate and are more prone to apoptosis. When B cells lack either IL-21R or STAT6, aberrant GCs form atypical centroblasts and centrocytes that differ in their phenotypic maturation and costimulatory molecule expression. Thus, IL-4 and IL-21 play nonredundant roles in the phased progression of GC B cell development that can initiate in the combined absence of these cytokine signals.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/immunology , Interleukin-4/metabolism , Interleukins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Apoptosis , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Lymphocyte Activation , Mice , Mice, Knockout , Paracrine Communication , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, Interleukin-21/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction
8.
Nature ; 487(7408): 496-9, 2012 Jul 26.
Article in English | MEDLINE | ID: mdl-22763436

ABSTRACT

Tissue development and regeneration depend on cell-cell interactions and signals that target stem cells and their immediate progeny. However, the cellular behaviours that lead to a properly regenerated tissue are not well understood. Using a new, non-invasive, intravital two-photon imaging approach we study physiological hair-follicle regeneration over time in live mice. By these means we have monitored the behaviour of epithelial stem cells and their progeny during physiological hair regeneration and addressed how the mesenchyme influences their behaviour. Consistent with earlier studies, stem cells are quiescent during the initial stages of hair regeneration, whereas the progeny are more actively dividing. Moreover, stem cell progeny divisions are spatially organized within follicles. In addition to cell divisions, coordinated cell movements of the progeny allow the rapid expansion of the hair follicle. Finally, we show the requirement of the mesenchyme for hair regeneration through targeted cell ablation and long-term tracking of live hair follicles. Thus, we have established an in vivo approach that has led to the direct observation of cellular mechanisms of growth regulation within the hair follicle and that has enabled us to precisely investigate functional requirements of hair-follicle components during the process of physiological regeneration.


Subject(s)
Hair Follicle/cytology , Regeneration/physiology , Stem Cells/cytology , Animals , Cell Division , Cell Movement , Cell Survival , Cell Tracking , Dermis/cytology , Laser Therapy , Mesoderm/cytology , Mice , Mice, Transgenic , Microscopy, Fluorescence, Multiphoton
9.
Nature ; 484(7395): 510-3, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-22538615

ABSTRACT

NLRs (nucleotide-binding domain leucine-rich-repeat-containing receptors; NOD-like receptors) are a class of pattern recognition receptor (PRR) that respond to host perturbation from either infectious agents or cellular stress. The function of most NLR family members has not been characterized and their role in instructing adaptive immune responses remains unclear. NLRP10 (also known as PYNOD, NALP10, PAN5 and NOD8) is the only NLR lacking the putative ligand-binding leucine-rich-repeat domain, and has been postulated to be a negative regulator of other NLR members, including NLRP3 (refs 4-6). We did not find evidence that NLRP10 functions through an inflammasome to regulate caspase-1 activity nor that it regulates other inflammasomes. Instead, Nlrp10(-/-) mice had a profound defect in helper T-cell-driven immune responses to a diverse array of adjuvants, including lipopolysaccharide, aluminium hydroxide and complete Freund's adjuvant. Adaptive immunity was impaired in the absence of NLRP10 because of a dendritic cell (DC) intrinsic defect in emigration from inflamed tissues, whereas upregulation of DC costimulatory molecules and chemotaxis to CCR7-dependent and -independent ligands remained intact. The loss of antigen transport to the draining lymph nodes by a subset of migratory DCs resulted in an almost absolute loss in naive CD4(+) T-cell priming, highlighting the critical link between diverse innate immune stimulation, NLRP10 activity and the immune function of mature DCs.


Subject(s)
Adaptive Immunity/immunology , Apoptosis Regulatory Proteins/metabolism , Dendritic Cells/immunology , Adaptor Proteins, Signal Transducing , Adjuvants, Immunologic , Animals , Antigens/immunology , Apoptosis Regulatory Proteins/deficiency , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Caspase 1 , Cell Movement , Chemokines/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Deletion , Inflammasomes , Ligands , Lymph Nodes/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccines/immunology
10.
Nat Rev Immunol ; 7(7): 499-504, 2007 07.
Article in English | MEDLINE | ID: mdl-17589541

ABSTRACT

Affinity maturation of antibodies during the course of an adaptive immune response requires germinal centre (GC) formation within B-cell follicles. Much of the current understanding of GC function has been derived from histology, but these static views have left unresolved many questions about cell movement in GCs. In this Progress article, we describe how several recent studies using time-resolved multiphoton microscopy to track GC B-cell movement within lymph nodes have shed light on the processes that influence GC B-cell dynamics.


Subject(s)
Cell Differentiation , Cell Movement , Germinal Center/cytology , Microscopy/methods , Animals , B-Lymphocytes/cytology , Humans
12.
Nature ; 475(7357): 514-8, 2011 Jul 17.
Article in English | MEDLINE | ID: mdl-21765430

ABSTRACT

Interleukin (IL)-17-producing T helper cells (T(H)17) are a recently identified CD4(+) T cell subset distinct from T helper type 1 (T(H)1) and T helper type 2 (T(H)2) cells. T(H)17 cells can drive antigen-specific autoimmune diseases and are considered the main population of pathogenic T cells driving experimental autoimmune encephalomyelitis (EAE), the mouse model for multiple sclerosis. The factors that are needed for the generation of T(H)17 cells have been well characterized. However, where and how the immune system controls T(H)17 cells in vivo remains unclear. Here, by using a model of tolerance induced by CD3-specific antibody, a model of sepsis and influenza A viral infection (H1N1), we show that pro-inflammatory T(H)17 cells can be redirected to and controlled in the small intestine. T(H)17-specific IL-17A secretion induced expression of the chemokine CCL20 in the small intestine, facilitating the migration of these cells specifically to the small intestine via the CCR6/CCL20 axis. Moreover, we found that T(H)17 cells are controlled by two different mechanisms in the small intestine: first, they are eliminated via the intestinal lumen; second, pro-inflammatory T(H)17 cells simultaneously acquire a regulatory phenotype with in vitro and in vivo immune-suppressive properties (rT(H)17). These results identify mechanisms limiting T(H)17 cell pathogenicity and implicate the gastrointestinal tract as a site for control of T(H)17 cells.


Subject(s)
Intestine, Small/immunology , Th17 Cells/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Movement/drug effects , Chemokine CCL20/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Gene Expression Profiling , Gene Expression Regulation/immunology , Influenza A virus/immunology , Interleukin-17/immunology , Intestine, Small/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Receptors, CCR6/immunology , Sepsis/immunology , Staphylococcal Infections/immunology
13.
Nat Rev Immunol ; 3(9): 757-64, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949499

ABSTRACT

The close association of follicular dendritic cells (FDCs) and germinal-centre B cells has fostered the idea that B-cell recognition of retained antigen that is presented on the surface of FDCs is important for affinity maturation and memory B-cell development. We argue that the retention of immune complexes is not required for germinal-centre development, affinity maturation and memory B-cell maintenance. Instead, it is probable that FDCs support B-cell proliferation and differentiation in a non-specific manner. Other potential roles of immune complexes retained by FDCs are discussed.


Subject(s)
Antigen-Antibody Complex/immunology , B-Lymphocytes/immunology , Dendritic Cells, Follicular/immunology , Immunologic Memory/immunology , Animals , Antigen-Antibody Complex/metabolism , B-Lymphocytes/cytology , Dendritic Cells, Follicular/cytology , Dendritic Cells, Follicular/metabolism , Germinal Center/cytology , Germinal Center/immunology , Humans , Lymphocyte Activation/immunology , Peanut Agglutinin/immunology
14.
Viruses ; 16(2)2024 02 03.
Article in English | MEDLINE | ID: mdl-38400021

ABSTRACT

Seasonal infection rates of individual viruses are influenced by synergistic or inhibitory interactions between coincident viruses. Endemic patterns of SARS-CoV-2 and influenza infection overlap seasonally in the Northern hemisphere and may be similarly influenced. We explored the immunopathologic basis of SARS-CoV-2 and influenza A (H1N1pdm09) interactions in Syrian hamsters. H1N1 given 48 h prior to SARS-CoV-2 profoundly mitigated weight loss and lung pathology compared to SARS-CoV-2 infection alone. This was accompanied by the normalization of granulocyte dynamics and accelerated antigen-presenting populations in bronchoalveolar lavage and blood. Using nasal transcriptomics, we identified a rapid upregulation of innate and antiviral pathways induced by H1N1 by the time of SARS-CoV-2 inoculation in 48 h dual-infected animals. The animals that were infected with both viruses also showed a notable and temporary downregulation of mitochondrial and viral replication pathways. Quantitative RT-PCR confirmed a decrease in the SARS-CoV-2 viral load and lower cytokine levels in the lungs of animals infected with both viruses throughout the course of the disease. Our data confirm that H1N1 infection induces rapid and transient gene expression that is associated with the mitigation of SARS-CoV-2 pulmonary disease. These protective responses are likely to begin in the upper respiratory tract shortly after infection. On a population level, interaction between these two viruses may influence their relative seasonal infection rates.


Subject(s)
COVID-19 , Influenza A Virus, H1N1 Subtype , Influenza, Human , Cricetinae , Animals , Humans , COVID-19/pathology , Mesocricetus , SARS-CoV-2 , Influenza, Human/pathology , Lung , Disease Models, Animal
15.
Biomed Pharmacother ; 173: 116427, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38484558

ABSTRACT

Uncertainty exists regarding the mechanisms by which hypoxia-inducible factors (HIFs) control CD8+T-cell migration into tumor microenvironments. Here, we found that HIF-1α knockdown or overexpression resulted in increased or decreased CXCL9, -10, and -11 expression in vitro, respectively. Gene Set Variation Analysis revealed that elevated HIF-1α levels correlated with a poor prognosis, severe pathological stage, and an absence of CD8+ T cells in the tumor microenvironment in colorectal cancer (CRC) patients. HIF-1α was inversely associated with pathways beneficial to anti-tumor immunotherapy and cytokine/chemokine function. In vivo, inhibiting HIF-1α or its upstream regulator BIRC2 significantly suppressed tumor growth and promoted CD8+ T-cell infiltration. CXCR3 neutralizing antibodies reversed these effects, implicating the involvement of CXCL9, -10, and -11/CXCR3 axis. The presence of HIF-1α weakened the upregulation of CXCL9, -10, and -11 by bleomycin and doxorubicin. Combining HIF-1α inhibition with bleomycin promoted CD8+ T-cell infiltration and tumor suppression in vivo. Moreover, doxorubicin could upregulate CXCL9, -10 and -11 by suppressing HIF-1α. Our findings highlight the potential of HIF-1α inhibition to improve CRC microenvironments and increase chemotherapy sensitivity.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit , Humans , Bleomycin , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Cytokines , Doxorubicin/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Tumor Microenvironment
16.
J Exp Med ; 204(9): 2103-14, 2007 Sep 03.
Article in English | MEDLINE | ID: mdl-17698588

ABSTRACT

The study of murine memory B cells has been limited by small cell numbers and the lack of a definitive marker. We have addressed some of these difficulties with hapten-specific transgenic (Tg) mouse models that yield relatively large numbers of antigen-specific memory B cells upon immunization. Using these models, along with a 5-bromo-2'-deoxyuridine (BrdU) pulse-label strategy, we compared memory cells to their naive precursors in a comprehensive flow cytometric survey, thus revealing several new murine memory B cell markers. Most interestingly, memory cells were phenotypically heterogeneous. Particularly surprising was the finding of an unmutated memory B cell subset identified by the expression of CD80 and CD35. We confirmed these findings in an analogous V region knock-in mouse and/or in non-Tg mice. There also was anatomic heterogeneity, with BrdU(+) memory cells residing not just in the marginal zone, as had been thought, but also in splenic follicles. These studies impact the current understanding of murine memory B cells by identifying new phenotypes and by challenging assumptions about the location and V region mutation status of memory cells. The apparent heterogeneity in the memory compartment implies either different origins and/or different functions, which we discuss.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Immunologic Memory/immunology , Lymphocyte Subsets/cytology , Lymphocyte Subsets/immunology , Mutation/genetics , Animals , B7-1 Antigen/immunology , Base Sequence , Biomarkers/metabolism , Bromodeoxyuridine/metabolism , Cell Survival , DNA/biosynthesis , Immunity, Cellular , Immunization , Mice , Mice, Transgenic , Receptors, Complement 3b/immunology , Selection, Genetic , Spleen/cytology , Time Factors
17.
Am J Pathol ; 180(4): 1715-25, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22310467

ABSTRACT

Lymphatic vessels (LVs) are important structures for antigen presentation, for lipid metabolism, and as conduits for tumor metastases, but they have been difficult to visualize in vivo. Prox1 is a transcription factor that is necessary for lymphangiogenesis in ontogeny and the maintenance of LVs. To visualize LVs in the lymph node of a living mouse in real time, we made the ProxTom transgenic mouse in a C57BL/6 background using red fluorescent LVs that are suitable for in vivo imaging. The ProxTom transgene contained all Prox1 regulatory sequences and was faithfully expressed in LVs coincident with endogenous Prox1 expression. The progenies of a ProxTom × Hec6stGFP cross were imaged using two-photon laser scanning microscopy, allowing the simultaneous visualization of LVs and high endothelial venules in a lymph node of a living mouse for the first time. We confirmed the expression of Prox1 in the adult liver, lens, and dentate gyrus. These intensely fluorescent mice revealed the expression of Prox1 in three novel sites: the neuroendocrine cells of the adrenal medulla, megakaryocytes, and platelets. The novel sites identified herein suggest previously unknown roles for Prox1. The faithful expression of the fluorescent reporter in ProxTom LVs indicates that these mice have potential utility in the study of diseases as diverse as lymphedema, filariasis, transplant rejection, obesity, and tumor metastasis.


Subject(s)
Adrenal Medulla/metabolism , Blood Platelets/metabolism , Homeodomain Proteins/metabolism , Lymphatic Vessels/metabolism , Megakaryocytes/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cells, Cultured , Cytoplasm/metabolism , Endothelial Cells/metabolism , Gene Expression Regulation/physiology , Genotype , Glycoproteins/metabolism , Homeodomain Proteins/genetics , Luminescent Proteins/metabolism , Lymph Nodes/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Red Fluorescent Protein
18.
J Immunol ; 185(9): 5315-25, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20921525

ABSTRACT

It is unclear where within tissues subsets of effector and memory CD8 T cells persist during viral infection and whether their localization affects function and long-term survival. Following lymphocytic choriomeningitis virus infection, we found most killer cell lectin-like receptor G1 (KLRG1)(lo)IL-7R(hi) effector and memory cells, which are long-lived and high proliferative capacity, in the T cell zone of the spleen. In contrast, KLRG1(hi)IL-7R(lo) cells, which appear terminally differentiated and have shorter life spans, were exclusively localized to the red pulp. KLRG1(lo)IL-7R(hi) T cells homed to the T cell zone using pertussis toxin-sensitive chemokine receptors and appeared to contact gp38(+) stromal cells, which produce the chemokines CCL19 and CCL21 and the T cell survival cytokine IL-7. The transcription factors T-bet and B lymphocyte-induced maturation protein-1 controlled effector CD8 T cell splenic migration. Effector CD8 T cells overexpressing T-bet homed to the red pulp, whereas those lacking B lymphocyte-induced maturation protein-1 homed to the T cell zone. Upon memory formation, CD62L(+) memory T cells were predominantly found in the T cell zone, whereas CD62L(-) cells were found in the red pulp. Thus, effector and memory CD8 T cell subset localization within tissues is linked to their differentiation states, and this may identify anatomical niches that regulate their longevity and homeostasis.


Subject(s)
Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/cytology , Chemotaxis, Leukocyte/immunology , Immunologic Memory/immunology , Spleen/cytology , T-Lymphocyte Subsets/cytology , Animals , CD8-Positive T-Lymphocytes/immunology , Lymphocytic choriomeningitis virus , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , T-Lymphocyte Subsets/immunology
19.
J Immunol ; 183(11): 7314-25, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19917681

ABSTRACT

B lymphocytes producing high-affinity Abs are critical for protection from extracellular pathogens, such as bacteria and parasites. The process by which high-affinity B cells are selected during the immune response has never been elucidated. Although it has been shown that high-affinity cells directly outcompete low-affinity cells in the germinal center (GC), whether there are also intrinsic differences between these cells has not been addressed. It could be that higher affinity cells proliferate more rapidly or are more likely to enter cell cycle, thereby outgrowing lower affinity cells. Alternatively, higher affinity cells could be relatively more resistant to cell death in the GC. By comparing high- and low-affinity B cells for the same Ag, we show here that low-affinity cells have an intrinsically higher death rate than do cells of higher affinity, even in the absence of competition. This suggests that selection in the GC reaction is due at least in part to the control of survival of higher affinity B cells and not by a proliferative advantage conferred upon these cells compared with lower affinity B cells. Control over survival rather than proliferation of low- and high-affinity B cells in the GC allows greater diversity not only in the primary response but also in the memory response.


Subject(s)
B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Animals , Antibody Affinity , Apoptosis , Cell Division , Cell Lineage/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/cytology , Germinal Center/immunology , Mice , Models, Theoretical , Mutagenesis, Site-Directed
20.
J Exp Med ; 195(9): 1215-21, 2002 May 06.
Article in English | MEDLINE | ID: mdl-11994427

ABSTRACT

To understand the relationship between the affinity of the B cell antigen receptor (BCR) and the immune response to antigen, two lines of immunoglobulin H chain transgenic (Tg) mice were created. H50Gmu(a) and T1(V23)mu(a) mice express mu H chain transgenes that associate with the lambda1 L chains to bind the (4-hydroxy-3-nitrophenyl)acetyl hapten with association constants (K(a)s) of only 1.2 x 10(5) M(-1) and 3 x 10(4) M(-1), respectively. Both lines mounted substantial antibody-forming cell (AFC) and germinal center (GC) responses. H50Gmu(a) Tg mice also generated memory B cells. T1(V23)mu(a) B cells formed AFC and GCs, but were largely replaced in late GCs by antigen-specific cells that express endogenous BCRs. Thus, B lymphocytes carrying BCRs with affinities previously thought to be irrelevant in specific immune responses are in fact capable of complete T cell-dependent immune responses when relieved of substantial competition from other B cells. The failure to observe such B cells normally in late primary responses and in memory B cell populations is the result of competition, rather than an intrinsic inability of low affinity B cells.


Subject(s)
B-Lymphocytes/immunology , Immunologic Memory , Receptors, Antigen, B-Cell/immunology , Animals , Antibody Formation , Gene Rearrangement, B-Lymphocyte , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Mice , Mice, SCID , Mice, Transgenic , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL