scholarly journals Follicular B Helper T Cells Express Cxc Chemokine Receptor 5, Localize to B Cell Follicles, and Support Immunoglobulin Production

2000 ◽  
Vol 192 (11) ◽  
pp. 1545-1552 ◽  
Author(s):  
Dagmar Breitfeld ◽  
Lars Ohl ◽  
Elisabeth Kremmer ◽  
Joachim Ellwart ◽  
Federica Sallusto ◽  
...  

Chemokines and their receptors have been identified as major regulators controlling the functional organization of secondary lymphoid organs. Here we show that expression of CXC chemokine receptor 5 (CXCR5), a chemokine receptor required for B cell homing to B cell follicles, defines a novel subpopulation of B helper T cells localizing to follicles. In peripheral blood these cells coexpress CD45RO and the T cell homing CC chemokine receptor 7 (CCR7). In secondary lymphoid organs, CD4+CXCR5+ cells lose expression of CCR7, which allows them to localize to B cell follicles and germinal centers where they express high levels of CD40 ligand (CD40L), a costimulatory molecule required for B cell activation and inducible costimulator (ICOS), a recently identified costimulatory molecule of the CD28 family. Thus, when compared with CD4+CD45RO+CXCR5− cells, CD4+CD45RO+CXCR5+ tonsillar T cells efficiently support the production of immunoglobulin (Ig)A and IgG. In contrast, analysis of the memory response revealed that long-lasting memory cells are found within the CD4+CD45RO+CXCR5− population, suggesting that CXCR5+CD4 cells represent recently activated effector cells. Based on the characteristic localization within secondary lymphoid organs, we suggest to term these cells “follicular B helper T cells” (TFH).

1997 ◽  
Vol 185 (11) ◽  
pp. 1909-1918 ◽  
Author(s):  
Jérôme Fayette ◽  
Bertrand Dubois ◽  
Stéphane Vandenabeele ◽  
Jean-Michel Bridon ◽  
Béatrice Vanbervliet ◽  
...  

Within T cell–rich areas of secondary lymphoid organs, interdigitating dendritic cells recruit antigen-specific T cells that then induce B cells to secrete Igs. This study investigates the possible role(s) of dendritic cells in the regulation of human B cell responses. In the absence of exogenous cytokines, in vitro generated dendritic cells (referred to as Dendritic Langerhans cells, D-Lc) induced surface IgA expression on ∼10% of CD40-activated naive sIgD+ B cells. In the presence of IL-10 and TGF-β, a combination of cytokines previously identified for its capacity to induce IgA switch, D-Lc strongly potentiated the induction of sIgA on CD40-activated naive B cells from 5% to 40–50%. D-Lc alone did not induce the secretion of IgA by CD40-activated naive B cells, which required further addition of IL-10. Furthermore, D-Lc skewed towards the IgA isotype at the expense of IgG, the Ig production of CD40-activated naive B cells cultured in the presence of IL-10 and TGF-β. Importantly, under these culture conditions, both IgA1 and IgA2 were detected. In the presence of IL-10, secretion of IgA2 by CD40-activated naive B cells could be detected only in response to D-Lc and was further enhanced by TGF-β. Collectively, these results suggest that in addition to activating T cells in the extrafollicular areas of secondary lymphoid organs, human D-Lc also directly modulate T cell–dependent B cell growth and differentiation, by inducing the IgA isotype switch.


2011 ◽  
Vol 42 (3) ◽  
pp. 790-798 ◽  
Author(s):  
Raquel Planas ◽  
Ilijas Jelc̆ić ◽  
Sven Schippling ◽  
Roland Martin ◽  
Mireia Sospedra

1997 ◽  
Vol 185 (5) ◽  
pp. 941-952 ◽  
Author(s):  
Bertrand Dubois ◽  
Béatrice Vanbervliet ◽  
Jérome Fayette ◽  
Catherine Massacrier ◽  
Cees Van Kooten ◽  
...  

After antigen capture, dendritic cells (DC) migrate into T cell–rich areas of secondary lymphoid organs, where they induce T cell activation, that subsequently drives B cell activation. Here, we investigate whether DC, generated in vitro, can directly modulate B cell responses, using CD40L-transfected L cells as surrogate activated T cells. DC, through the production of soluble mediators, stimulated by 3- to 6-fold the proliferation and subsequent recovery of B cells. Furthermore, after CD40 ligation, DC enhanced by 30–300-fold the secretion of IgG and IgA by sIgD− B cells (essentially memory B cells). In the presence of DC, naive sIgD+ B cells produced, in response to interleukin-2, large amounts of IgM. Thus, in addition to activating naive T cells in the extrafollicular areas of secondary lymphoid organs, DC may directly modulate B cell growth and differentiation.


2000 ◽  
Vol 192 (11) ◽  
pp. 1553-1562 ◽  
Author(s):  
Patrick Schaerli ◽  
Katharina Willimann ◽  
Alois B. Lang ◽  
Martin Lipp ◽  
Pius Loetscher ◽  
...  

Leukocyte traffic through secondary lymphoid tissues is finely tuned by chemokines. We have studied the functional properties of a human T cell subset marked by the expression of CXC chemokine receptor 5 (CXCR5). Memory but not naive T cells from tonsils are CXCR5+ and migrate in response to the B cell–attracting chemokine 1 (BCA-1), which is selectively expressed by reticular cells and blood vessels within B cell follicles. Tonsillar CXCR5+ T cells do not respond to other chemokines present in secondary lymphoid tissues, including secondary lymphoid tissue chemokine (SLC), EBV-induced molecule 1 ligand chemokine (ELC), and stromal cell–derived factor 1 (SDF-1). The involvement of tonsillar CXCR5+ T cells in humoral immune responses is suggested by their localization in the mantle and light zone germinal centers of B cell follicles and by the concomitant expression of activation and costimulatory markers, including CD69, HLA-DR, and inducible costimulator (ICOS). Peripheral blood CXCR5+ T cells also belong to the CD4+ memory T cell subset but, in contrast to tonsillar cells, are in a resting state and migrate weakly to chemokines. CXCR5+ T cells are very inefficient in the production of cytokines but potently induce antibody production during coculture with B cells. These properties portray CXCR5+ T cells as a distinct memory T cell subset with B cell helper function, designated here as follicular B helper T cells (TFH).


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 212-212
Author(s):  
Heather E Stefanski ◽  
Emily Goren ◽  
Leslie Jonert ◽  
James J. Mule ◽  
Bruce R. Blazar

Abstract Abstract 212 Introduction: BMT conditioning causes substantial stromal cell injury, which may hinder lymphopoiesis and immune recovery post-transplant. The C-C chemokine CCL21, produced by LN fibroblastic reticular cells (FRCs), is critical for the trafficking of CCR7 expressing T cells into secondary lymphoid organs (i.e. spleen, LN). We first reported that radiation conditioning markedly reduces FRCs, CCL21 expression, and lymphoid tissue inducer cells that are critical for lymphoid organogenesis. As a consequence of these events, mice have substantial T cell lymphopenia and poor responses to pathogens and neoantigens (Kelly et al, Blood, 2010). We hypothesized that CCL21 deficiency would preclude the recruitment of recent thymic emigrants to secondary lymphoid organs where they would receive necessary signals for their survival. Moreover, within the secondary lymphoid organs, T cells would be unable to properly migrate to B cell zones. Thus, we tested whether restoring CCL21 protein could improve LN stromal cell architecture, T cell trafficking and consequently, T cell effector responses. Methods/Results: C57BL/6 mice were lethally irradiated, rescued with congenic T cell depleted bone marrow, and given CCL21 cDNA containing or null adenovirus vector transduced dendritic cells (DCs) that were pulsed with lysates from either vesicular stomatitis or attenuated Listeria monocytogenes expressing the nominal antigen ovalbumin (VSV-OVA; Listeria-OVA). The DC vaccines were given intramuscularly beginning 21 days post-transplant and then weekly for three total doses. On day 42, mice were challenged with either VSV-OVA or Listeria-OVA and euthanized on day 43 or 50, respectively. Compared to DC/null, DC/CCL21 vaccine substantially improved spleen and LN architecture and markedly increased ER-TR7+ LN stromal cell density. These architectural changes were associated with significantly increased CCL21 expression (p<0.05 for both CCL21 density per B cell area and normalization to non-BMT controls determined by ELISA), resulting in a significant increase in total LN cells, CD8+ T cells and lymphoid tissue inducer cells (all p<0.05). Expression of the B cell chemoattractant, CXCL13, and B cell follicle number were significantly increased (p<0.05) in the spleen and LN of DC/CCL21 vaccinated mice and T cells were found juxtaposed to B cells within B cell zones. Compared to DC/null, DC/CCL21 LN CD8+ T cell number normalized vs non-BMT controls, and clearance of Listeria-OVA by the liver was significantly (p<0.05) improved and similar to non-BMT controls (3.5 vs 1.8 vs 1.2 × 105 colony-forming units, respectively). Similarly, VSV-OVA clearance was significantly improved in DC/CCL21 vs DC/null vaccines, both less than non-BMT controls (13.5 vs 26 vs 8 splenic plaque-forming units, respectively; all group-wise comparisons, p<0.05). In order to determine whether improved immune function could enhance the endogenous CD8+ T cell response to acute myeloid leukemia (AML) challenge post-BMT, congeneic BMT recipients were vaccinated with DC/CCL21 or DC/null pre-loaded with AML lysates. All mice received a lethal dose (106) of C57BL/6 AML cells expressing firefly luciferase (C1498) on d.42 post-BMT (see Figure). Compared to the uniform lethality in both DC/null vaccines and non-vaccinated, non-BMT controls, mice receiving DC/CCL21 had a significant increase (p=.0241 compared to DC/null) in survival with 50% of mice surviving long-term. Conclusions: Taken together, these data indicate that DC/CCL21 vaccines can potently restore LN and splenic architecture as well as improve endogenous T cell responses to pathogens and AML challenge. Because DC/CCL21 vaccines are in clinical trial for patients with lung cancer or melanoma, our studies may provide the foundation for future clinical trials of DC/CCL21 vaccination in patients receiving pre-transplant conditioning regimens. If our preclinical data prove translatable, DC/CCL21 vaccination could increase immune reconstitution in BMT recipients. This could lead to decreased relapse and increased pathogenic responses to life-threatening infections and decreasing morbidity and mortality post transplant. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-47-SCI-47
Author(s):  
Ivan Maillard

Notch signaling is a highly conserved signaling pathway with multiple functions in health and disease. Notch ligands of the Delta-like and Jagged families interact with Notch1-4 on adjacent cells to trigger proteolytic activation of the Notch receptors, followed by context-dependent transcriptional activation of Notch target genes. In the hematopoietic system, Notch was first identified for its oncogenic role in T cell acute lymphoblastic leukemia (T-ALL). More recently, Notch has been recognized as a recurrent oncogenic pathway in multiple B cell lymphoproliferative disorders, including CLL/SLL, mantle cell lymphoma and marginal zone lymphoma. In parallel, essential functions of Notch signaling have been identified in early T cell development and B cell homeostasis, as well as in regulating T and B cell immune responses in secondary lymphoid organs. We have discovered a conserved pathogenic function of Notch signaling in the control of graft-versus-host disease (GVHD), both in mouse and non-human primate models of hematopoietic cell transplantation. Within days after allogeneic hematopoietic cell transplantation, donor-derived T cells interact with Delta-like Notch ligands expressed by specialized subsets of non-hematopoietic fibroblastic stromal cells in spleen and lymph nodes. In turn, Notch signaling programs alloreactive T cells to become pathogenic effector cells that mediate long-term damage in GVHD target tissues, while suppressing the expansion of regulatory T cells. Emerging evidence suggests the existence of an active crosstalk between T cells, B cells and well-defined specialized immunological stromal niches expressing Notch ligands in secondary lymphoid organs, both in GVHD and in other types of immune responses. Moreover, many B cell lymphomas demonstrate oncogenic Notch activation as a result of gain-of-function mutations that increase the persistence of intracellular Notch after proteolytic cleavage, thus requiring the presence of Notch ligands in the microenvironment to trigger oncogenic Notch signals. New evidence suggests that key Notch ligands in human lymphomas are present in the lymph node microenvironment, and that oncogenic Notch activation can happen even in the absence of mutations in a substantial fraction of lymphoma patients, including the majority of patients with CLL/SLL. In these cases, lymphoma cells appear to interact with microenvironmental Notch ligands in secondary lymphoid organs through unmutated Notch receptors, borrowing from the playbook of normal lymphocytes. Furthermore, the expression of Notch ligands by defined subsets of fibroblastic stromal cells illustrates a new paradigm through which microenvironmental signals can profoundly influence the differentiation and function of adjacent lymphoid cells. We speculate that better understanding the regulation of Notch signaling and fibroblastic stromal cells in secondary lymphoid organs will provide valuable new information with translational potential both in immunobiology and in malignant hematology. Disclosures Maillard: Genentech: Consultancy; Regeneron: Consultancy.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii96-ii96
Author(s):  
Catalina Lee Chang ◽  
Jason Miska ◽  
David Hou ◽  
Aida Rashidi ◽  
Peng Zhang ◽  
...  

Abstract Immunotherapy has revolutionized the treatment of many tumors. However, most glioblastoma (GBM) patients have not, so far, benefited from such successes. With the goal of exploring ways to boost anti-GBM immunity, we developed a B-cell-based vaccine (BVax) that consists of 4-1BBL+ B cells activated with CD40 agonism and IFNg stimulation. BVaxmigrate to key secondary lymphoid organs and are proficient at antigen cross-presentation, which promotes both the survival and functionality of CD8+ T cells. A combination of radiation, BVax, and PD-L1 blockade conferred tumor eradication in 80% of treated tumor-bearing animals. This treatment elicited immunologic memory that prevented the growth of new tumors upon subsequent re-injection in cured mice. GBM patient-derived BVax were successful in activating autologous CD8+ T cells; these T cells showed a strong ability to kill autologous glioma cells. In addition to the role in activating CD8+ T cells, BVax produce tumor-specific antibodies able to control tumor growth via antibody-mediated cell cytotoxicity. In conclusion, BVax tackles GBM immunosurveillance escape by using both cellular (CD8+ T-cell activation) and humoral (anti-tumor antibody production) immunity. Our study provides an efficient alternative to current immunotherapeutic approaches that can be readily translated to the clinic.


2004 ◽  
Vol 31 (8) ◽  
pp. 1021-1031 ◽  
Author(s):  
Ken Matsui ◽  
Zheng Wang ◽  
Timothy J. McCarthy ◽  
Paul M. Allen ◽  
David E. Reichert

2004 ◽  
Vol 64 (21) ◽  
pp. 7697-7701 ◽  
Author(s):  
Irene M. Mullins ◽  
Craig L. Slingluff ◽  
Jae K. Lee ◽  
Courtney F. Garbee ◽  
Jianfen Shu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document