Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(52): e2301155120, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38109544

ABSTRACT

The protease MALT1 promotes lymphocyte activation and lymphomagenesis by cleaving a limited set of cellular substrates, most of which control gene expression. Here, we identified the integrin-binding scaffold protein Tensin-3 as a MALT1 substrate in activated human B cells. Activated B cells lacking Tensin-3 showed decreased integrin-dependent adhesion but exhibited comparable NF-κB1 and Jun N-terminal kinase transcriptional responses. Cells expressing a noncleavable form of Tensin-3, on the other hand, showed increased adhesion. To test the role of Tensin-3 cleavage in vivo, mice expressing a noncleavable version of Tensin-3 were generated, which showed a partial reduction in the T cell-dependent B cell response. Interestingly, human diffuse large B cell lymphomas and mantle cell lymphomas with constitutive MALT1 activity showed strong constitutive Tensin-3 cleavage and a decrease in uncleaved Tensin-3 levels. Moreover, silencing of Tensin-3 expression in MALT1-driven lymphoma promoted dissemination of xenografted lymphoma cells to the bone marrow and spleen. Thus, MALT1-dependent Tensin-3 cleavage reveals a unique aspect of the function of MALT1, which negatively regulates integrin-dependent B cell adhesion and facilitates metastatic spread of B cell lymphomas.


Subject(s)
Caspases , Lymphoma, Large B-Cell, Diffuse , Mice , Humans , Animals , Adult , Tensins/genetics , Caspases/metabolism , NF-kappa B/metabolism , Cell Adhesion/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Integrins
2.
Nat Immunol ; 24(5): 869-883, 2023 05.
Article in English | MEDLINE | ID: mdl-37081150

ABSTRACT

To date, no immunotherapy approaches have managed to fully overcome T-cell exhaustion, which remains a mandatory fate for chronically activated effector cells and a major therapeutic challenge. Understanding how to reprogram CD8+ tumor-infiltrating lymphocytes away from exhausted effector states remains an elusive goal. Our work provides evidence that orthogonal gene engineering of T cells to secrete an interleukin (IL)-2 variant binding the IL-2Rßγ receptor and the alarmin IL-33 reprogrammed adoptively transferred T cells to acquire a novel, synthetic effector state, which deviated from canonical exhaustion and displayed superior effector functions. These cells successfully overcame homeostatic barriers in the host and led-in the absence of lymphodepletion or exogenous cytokine support-to high levels of engraftment and tumor regression. Our work unlocks a new opportunity of rationally engineering synthetic CD8+ T-cell states endowed with the ability to avoid exhaustion and control advanced solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Immunotherapy, Adoptive , Interleukin-2 , Neoplasms, Experimental , CD8-Positive T-Lymphocytes/immunology , T-Cell Exhaustion , Lymphocytes, Tumor-Infiltrating/immunology , Interleukin-2/pharmacology , Interleukin-33 , Protein Engineering , Female , Animals , Mice , Mice, Inbred C57BL , Cell Line, Tumor , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/metabolism
3.
Immunity ; 56(4): 813-828.e10, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36809763

ABSTRACT

T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-33 , Lymphocytic Choriomeningitis , Animals , Mice , Alarmins/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Persistent Infection , T Cell Transcription Factor 1/metabolism
4.
Front Cell Infect Microbiol ; 12: 941860, 2022.
Article in English | MEDLINE | ID: mdl-36034709

ABSTRACT

The lymphatic system plays a crucial role in mounting immune response against intracellular pathogens, and recent studies have documented its role in facilitating tumor dissemination linked largely with cancer cells. However, in mucocutaneous leishmaniasis (MCL) caused by Leishmania Viannia subgenus showing infectious metastasis and resulting in severe distant secondary lesions, the route of escape of these parasites to secondary sites has not yet been investigated in detail. Our results demonstrated that when infection was associated with inflammation and additionally exacerbated by the presence of dsRNA viral endosymbiont (LRV1), lymphatic vessels could serve as efficient routes for infected cells to egress from the primary site and colonize distant organs. We challenged this hypothesis by using the intracellular Leishmania protozoan parasites Leishmania guyanensis (Lgy) associated with or without a dsRNA viral endosymbiont, exacerbating the infection and responsible for a strong inflammatory response, and favoring metastasis of the infection. We analyzed possible cargo cells and the routes of dissemination through flow cytometry, histological analysis, and in vivo imaging in our metastatic model to show that parasites disseminated not only intracellularly but also as free extracellular parasites using migrating immune cells, lymph nodes (LNs), and lymph vessels, and followed intricate connections of draining and non-draining lymph node to finally end up in the blood and in distant skin, causing new lesions.


Subject(s)
Leishmania braziliensis , Leishmania , Leishmaniasis, Mucocutaneous , Neoplasms , Humans , Lymphatic System
5.
Nat Immunol ; 23(8): 1246-1255, 2022 08.
Article in English | MEDLINE | ID: mdl-35817845

ABSTRACT

Lymph nodes (LNs) comprise two main structural elements: fibroblastic reticular cells that form dedicated niches for immune cell interaction and capsular fibroblasts that build a shell around the organ. Immunological challenge causes LNs to increase more than tenfold in size within a few days. Here, we characterized the biomechanics of LN swelling on the cellular and organ scale. We identified lymphocyte trapping by influx and proliferation as drivers of an outward pressure force, causing fibroblastic reticular cells of the T-zone (TRCs) and their associated conduits to stretch. After an initial phase of relaxation, TRCs sensed the resulting strain through cell matrix adhesions, which coordinated local growth and remodeling of the stromal network. While the expanded TRC network readopted its typical configuration, a massive fibrotic reaction of the organ capsule set in and countered further organ expansion. Thus, different fibroblast populations mechanically control LN swelling in a multitier fashion.


Subject(s)
Lymph Nodes , Stromal Cells , Animals , Fibroblasts , Lymphocytes , Mice , Mice, Inbred C57BL
6.
Nat Commun ; 13(1): 3983, 2022 07 09.
Article in English | MEDLINE | ID: mdl-35810168

ABSTRACT

The small intestinal villus tip is the first point of contact for lumen-derived substances including nutrients and microbial products. Electron microscopy studies from the early 1970s uncovered unusual spatial organization of small intestinal villus tip blood vessels: their exterior, epithelial-facing side is fenestrated, while the side facing the villus stroma is non-fenestrated, covered by pericytes and harbors endothelial nuclei. Such organization optimizes the absorption process, however the molecular mechanisms maintaining this highly specialized structure remain unclear. Here we report that perivascular LGR5+ villus tip telocytes (VTTs) are necessary for maintenance of villus tip endothelial cell polarization and fenestration by sequestering VEGFA signaling. Mechanistically, unique VTT expression of the protease ADAMTS18 is necessary for VEGFA signaling sequestration through limiting fibronectin accumulation. Therefore, we propose a model in which LGR5+ ADAMTS18+ telocytes are necessary to maintain a "just-right" level and location of VEGFA signaling in intestinal villus blood vasculature to ensure on one hand the presence of sufficient endothelial fenestrae, while avoiding excessive leakiness of the vessels and destabilization of villus tip epithelial structures.


Subject(s)
Intestines , Telocytes , Duodenum , Intestinal Mucosa/metabolism , Nutrients
7.
Nat Cardiovasc Res ; 1(5): 476-490, 2022 May.
Article in English | MEDLINE | ID: mdl-35602406

ABSTRACT

Stem and progenitor cells residing in the intestinal crypts drive the majority of colorectal cancers (CRCs), yet vascular contribution to this niche remains largely unexplored. VEGFA is a key driver of physiological and tumor angiogenesis. Accordingly, current anti-angiogenic cancer therapies target the VEGFA pathway. Here we report that in CRC expansion of the stem/progenitor pool in intestinal crypts requires VEGFA-independent growth and remodeling of blood vessels. Epithelial transformation induced expression of the endothelial peptide apelin, directs migration of distant venous endothelial cells towards progenitor niche vessels ensuring optimal perfusion. In the absence of apelin, loss of injury-inducible PROX1+ epithelial progenitors inhibited both incipient and advanced intestinal tumor growth. Our results establish fundamental principles for the reciprocal communication between vasculature and the intestinal progenitor niche and provide a mechanism for resistance to VEGFA-targeting drugs in CRCs.

8.
J Clin Invest ; 132(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35503257

ABSTRACT

Antigen-presenting cells (APCs) integrate signals emanating from local pathology and program appropriate T cell responses. In allogeneic hematopoietic stem cell transplantation (alloHCT), recipient conditioning releases damage-associated molecular patterns (DAMPs) that generate proinflammatory APCs that secrete IL-12, which is a driver of donor Th1 responses, causing graft-versus-host disease (GVHD). Nevertheless, other mechanisms exist to initiate alloreactive T cell responses, as recipients with disrupted DAMP signaling or lacking IL-12 develop GVHD. We established that tissue damage signals are perceived directly by donor CD4+ T cells and promoted T cell expansion and differentiation. Specifically, the fibroblastic reticular cell-derived DAMP IL-33 is increased by recipient conditioning and is critical for the initial activation, proliferation, and differentiation of alloreactive Th1 cells. IL-33 stimulation of CD4+ T cells was not required for lymphopenia-induced expansion, however. IL-33 promoted IL-12-independent expression of Tbet and generation of Th1 cells that infiltrated GVHD target tissues. Mechanistically, IL-33 augmented CD4+ T cell TCR-associated signaling pathways in response to alloantigen. This enhanced T cell expansion and Th1 polarization, but inhibited the expression of regulatory molecules such as IL-10 and Foxp3. These data establish an unappreciated role for IL-33 as a costimulatory signal for donor Th1 generation after alloHCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Interleukin-12 , Interleukin-33/genetics , Mice , Mice, Inbred BALB C , Th1 Cells/pathology
9.
Cancer Cell ; 40(3): 243-245, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35176235

ABSTRACT

Tumor-infiltrated T cells with stem-cell-like properties are important for determining the immunotherapy response. In this issue of Cancer Cell, Asrir and colleagues show that their entry requires specialized tumor-associated endothelial cells that resemble immature and inflamed lymph node vessels and that immunotherapy enhances the recruitment capacity of these endothelial cells.


Subject(s)
Neoplasms , T-Lymphocytes , CTLA-4 Antigen , Endothelial Cells , Humans , Immunotherapy , Lymph Nodes/pathology , Lymphocytes , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor , T-Lymphocytes/pathology , Venules/pathology
10.
Sci Adv ; 7(29)2021 07.
Article in English | MEDLINE | ID: mdl-34272244

ABSTRACT

The mechanisms maintaining adult lymphatic vascular specialization throughout life and their role in coordinating inter-organ communication to sustain homeostasis remain elusive. We report that inactivation of the mechanosensitive transcription factor Foxc2 in adult lymphatic endothelium leads to a stepwise intestine-to-lung systemic failure. Foxc2 loss compromised the gut epithelial barrier, promoted dysbiosis and bacterial translocation to peripheral lymph nodes, and increased circulating levels of purine metabolites and angiopoietin-2. Commensal microbiota depletion dampened systemic pro-inflammatory cytokine levels, corrected intestinal lymphatic dysfunction, and improved survival. Foxc2 loss skewed the specialization of lymphatic endothelial subsets, leading to populations with mixed, pro-fibrotic identities and to emergence of lymph node-like endothelial cells. Our study uncovers a cross-talk between lymphatic vascular function and commensal microbiota, provides single-cell atlas of lymphatic endothelial subtypes, and reveals organ-specific and systemic effects of dysfunctional lymphatics. These effects potentially contribute to the pathogenesis of diseases, such as inflammatory bowel disease, cancer, or lymphedema.


Subject(s)
Lymphatic Vessels , Lymphedema , Endothelial Cells/metabolism , Endothelium, Lymphatic/metabolism , Endothelium, Lymphatic/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Lymphatic Vessels/metabolism , Lymphedema/metabolism , Lymphedema/pathology
11.
Elife ; 102021 04 08.
Article in English | MEDLINE | ID: mdl-33830019

ABSTRACT

Innate immune cellular effectors are actively consumed during systemic inflammation, but the systemic traffic and the mechanisms that support their replenishment remain unknown. Here, we demonstrate that acute systemic inflammation induces the emergent activation of a previously unrecognized system of rapid migration of granulocyte-macrophage progenitors and committed macrophage-dendritic progenitors, but not other progenitors or stem cells, from bone marrow (BM) to regional lymphatic capillaries. The progenitor traffic to the systemic lymphatic circulation is mediated by Ccl19/Ccr7 and is NF-κB independent, Traf6/IκB-kinase/SNAP23 activation dependent, and is responsible for the secretion of pre-stored Ccl19 by a subpopulation of CD205+/CD172a+ conventional dendritic cells type 2 and upregulation of BM myeloid progenitor Ccr7 signaling. Mature myeloid Traf6 signaling is anti-inflammatory and necessary for lymph node myeloid cell development. This report unveils the existence and the mechanistic basis of a very early direct traffic of myeloid progenitors from BM to lymphatics during inflammation.


When the body becomes infected with disease-causing pathogens, such as bacteria, the immune system activates various mechanisms which help to fight off the infection. One of the immune system's first lines of defense is to launch an inflammatory response that helps remove the pathogen and recruit other immune cells. However, this response can become overactivated, leading to severe inflammatory conditions that damage healthy cells and tissues. A second group of cells counteract this over inflammation and are different to the ones involved in the early inflammatory response. Both types of cells ­ inflammatory and anti-inflammatory ­ develop from committed progenitors, which, unlike stem cells, are already destined to become a certain type of cell. These committed progenitors reside in the bone marrow and then rapidly travel to secondary lymphoid organs, such as the lymph nodes, where they mature into functioning immune cells. During this journey, committed progenitors pass from the bone marrow to the lymphatic vessels that connect up the different secondary lymphoid organs, and then spread to all tissues in the body. Yet, it is not fully understood what exact route these cells take and what guides them towards these lymphatic tissues during inflammation. To investigate this, Serrano-Lopez, Hegde et al. used a combination of techniques to examine the migration of progenitor cells in mice that had been treated with lethal doses of a bacterial product that triggers inflammation. This revealed that as early as one to three hours after the onset of infection, progenitor cells were already starting to travel from the bone marrow towards lymphatic vessels. Serrano-Lopez, Hegde et al. found that a chemical released by an "alarm" immune cell already residing in secondary lymphoid organs attracted these progenitor cells towards the lymphatic tissue. Further experiments showed that the progenitor cells travelling to secondary lymphoid organs were already activated by bacterial products. They then follow the chemical released by alarm immune cells ready to respond to the immune challenge and suppress inflammation. These committed progenitors were also found in the inflamed lymph nodes of patients. These findings suggest this rapid circulation of progenitors is a mechanism of defense that contributes to the fight against severe inflammation. Altering how these cells migrate from the bone marrow to secondary lymphoid organs could provide a more effective treatment for inflammatory conditions and severe infections. However, these approaches would need to be tested further in the laboratory and in clinical trials.


Subject(s)
Bone Marrow/metabolism , Cell Movement , Granulocyte-Macrophage Progenitor Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Lymphadenopathy/metabolism , Lymphatic System/metabolism , Myeloid Progenitor Cells/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow/immunology , Bone Marrow/pathology , Cell Lineage , Cells, Cultured , Child , Child, Preschool , Disease Models, Animal , Female , Granulocyte-Macrophage Progenitor Cells/immunology , Granulocyte-Macrophage Progenitor Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Lymphadenopathy/immunology , Lymphadenopathy/pathology , Lymphatic System/immunology , Lymphatic System/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Phenotype , Signal Transduction , Time Factors , Young Adult
12.
Eur J Immunol ; 51(1): 76-90, 2021 01.
Article in English | MEDLINE | ID: mdl-32700362

ABSTRACT

Upon viral infection, stressed or damaged cells can release alarmins like IL-33 that act as endogenous danger signals alerting innate and adaptive immune cells. IL-33 coming from nonhematopoietic cells has been identified as important factor triggering the expansion of antiviral CD8+ T cells. In LN the critical cellular source of IL-33 is unknown, as is its potential cell-intrinsic function as a chromatin-associated factor. Using IL-33-GFP reporter mice, we identify fibroblastic reticular cells (FRC) and lymphatic endothelial cells (LEC) as the main IL-33 source. In homeostasis, IL-33 is dispensable as a transcriptional regulator in FRC, indicating it functions mainly as released cytokine. Early during infection with lymphocytic choriomeningitis virus (LCMV) clone 13, both FRC and LEC lose IL-33 protein expression suggesting cytokine release, correlating timewise with IL-33 receptor expression by reactive CD8+ T cells and their greatly augmented expansion in WT versus ll33-/- mice. Using mice lacking IL-33 selectively in FRC versus LEC, we identify FRC as key IL-33 source driving acute and chronic antiviral T-cell responses. Collectively, these findings show that LN T-zone FRC not only regulate the homeostasis of naïve T cells but also their expansion and differentiation several days into an antiviral response.


Subject(s)
Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Acute Disease , Adaptive Immunity , Animals , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Endothelial Cells/immunology , Fibroblasts/immunology , Homeostasis , Humans , Immunity, Innate , Interleukin-33/deficiency , Interleukin-33/genetics , Lymph Nodes/immunology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological
13.
Immunity ; 53(5): 985-1000.e11, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33128876

ABSTRACT

Central memory CD8+ T cells (Tcm) control systemic secondary infections and can protect from chronic infection and cancer as a result of their stem-cell-like capacity to expand, differentiate, and self-renew. Central memory is generally thought to emerge following pathogen clearance and to form based on the de-differentiation of cytolytic effector cells. Here, we uncovered rare effector-phase CD8+ T cells expressing high amounts of the transcription factor Tcf7 (Tcf1) that showed no evidence of prior cytolytic differentiation and that displayed key hallmarks of Tcm cells. These effector-phase Tcf7hi cells quantitatively yielded Tcm cells based on lineage tracing. Mechanistically, Tcf1 counteracted the differentiation of Tcf7hi cells and sustained the expression of conserved adult stem-cell genes that were critical for CD8+ T cell stemness. The discovery of stem-cell-like CD8+ T cells during the effector response to acute infection provides an opportunity to optimize Tcm cell formation by vaccination.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cytotoxicity, Immunologic , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunologic Memory , T Cell Transcription Factor 1/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , Cytotoxicity, Immunologic/genetics , Fluorescent Antibody Technique , Gene Expression , Hepatocyte Nuclear Factor 1-alpha/chemistry , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Immunization , Immunologic Memory/genetics , Immunophenotyping , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Protein Conformation , Spleen/immunology , Spleen/metabolism , Structure-Activity Relationship , T Cell Transcription Factor 1/chemistry , T Cell Transcription Factor 1/genetics
14.
PLoS Biol ; 17(7): e3000072, 2019 07.
Article in English | MEDLINE | ID: mdl-31306410

ABSTRACT

Lymphoid T-zone fibroblastic reticular cells (FRCs) actively promote T-cell trafficking, homeostasis, and expansion but can also attenuate excessive T-cell responses via inducible nitric oxide (NO) and constitutive prostanoid release. It remains unclear how these FRC-derived mediators dampen T-cell responses and whether this occurs in vivo. Here, we confirm that murine lymph node (LN) FRCs produce prostaglandin E2 (PGE2) in a cyclooxygenase-2 (COX2)-dependent and inflammation-independent fashion. We show that this COX2/PGE2 pathway is active during both strong and weak T-cell responses, in contrast to NO, which only comes into play during strong T-cell responses. During chronic infections in vivo, PGE2-receptor signaling in virus-specific cluster of differentiation (CD)8 cytotoxic T cells was shown by others to suppress T-cell survival and function. Using COX2flox/flox mice crossed to mice expressing Cre recombinase expression under control of the CC chemokine ligand (CCL19) promoter (CCL19cre), we now identify CCL19+ FRC as the critical source of this COX2-dependent suppressive factor, suggesting PGE2-expressing FRCs within lymphoid tissues are an interesting therapeutic target to improve T-cell-mediated pathogen control during chronic infection.


Subject(s)
Cyclooxygenase 2/immunology , Fibroblasts/immunology , Lymph Nodes/immunology , Prostaglandins/immunology , T-Lymphocytes/immunology , Animals , Cell Line , Cell Movement/genetics , Cell Movement/immunology , Cell Proliferation/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Lymph Nodes/cytology , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prostaglandins/biosynthesis , T-Lymphocytes/virology
15.
Proc Natl Acad Sci U S A ; 116(27): 13490-13497, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31213547

ABSTRACT

Resident fibroblasts at sites of infection, chronic inflammation, or cancer undergo phenotypic and functional changes to support leukocyte migration and, in some cases, aggregation into tertiary lymphoid structures (TLS). The molecular programming that shapes these changes and the functional requirements of this population in TLS development are unclear. Here, we demonstrate that external triggers at mucosal sites are able to induce the progressive differentiation of a population of podoplanin (pdpn)-positive stromal cells into a network of immunofibroblasts that are able to support the earliest phases of TLS establishment. This program of events, that precedes lymphocyte infiltration in the tissue, is mediated by paracrine and autocrine signals mainly regulated by IL13. This initial fibroblast network is expanded and stabilized, once lymphocytes are recruited, by the local production of the cytokines IL22 and lymphotoxin. Interfering with this regulated program of events or depleting the immunofibroblasts in vivo results in abrogation of local pathology, demonstrating the functional role of immunofibroblasts in supporting TLS maintenance in the tissue and suggesting novel therapeutic targets in TLS-associated diseases.


Subject(s)
Fibroblasts/pathology , Tertiary Lymphoid Structures/pathology , Animals , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Interleukin-13/metabolism , Interleukins/metabolism , Lymphocytes/pathology , Mice , Salivary Glands/pathology , Interleukin-22
16.
Cell Rep ; 27(8): 2442-2458.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116987

ABSTRACT

Adaptive type 2 immune responses against the intestinal helminth Heligmosomoides polygyrus (Hp) require the interaction of follicle-associated CXCR5+ dendritic cells with naive T cells in the draining mesenteric lymph nodes (mLNs). However, the source of CXCL13 responsible for attracting CXCR5+ dendritic cells has remained unclear. Using multiplex imaging combined with deep tissue analysis, we observed new CXCL13+ fibroblastic reticular cells surrounding paracortical and cortical B cell follicles in the mLNs of infected mice. CXCL13+ fibroblasts expressed markers of marginal reticular cells (MRCs), and their expansion required lymphotoxin (LT)-dependent interactions between IL-4Rα-expressing B cells and CCL19+ fibroblasts. Infection-induced follicles did not necessarily contain follicular dendritic cells (FDCs), indicating that CXCL13+ fibroblasts may instead drive their formation. These data reveal a role for lymphotoxin signaling to CCL19+ fibroblasts in the development of CXCL13+ MRC-like cells and adaptive type 2 immunity in response to helminth infection.


Subject(s)
B-Lymphocytes/metabolism , Chemokine CXCL13/metabolism , Receptors, Cell Surface/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chemokine CCL19/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lymph Nodes/immunology , Lymph Nodes/parasitology , Lymph Nodes/pathology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism
17.
Immunity ; 50(1): 195-211.e10, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30635237

ABSTRACT

Checkpoint blockade mediates a proliferative response of tumor-infiltrating CD8+ T lymphocytes (TILs). The origin of this response has remained elusive because chronic activation promotes terminal differentiation or exhaustion of tumor-specific T cells. Here we identified a subset of tumor-reactive TILs bearing hallmarks of exhausted cells and central memory cells, including expression of the checkpoint protein PD-1 and the transcription factor Tcf1. Tcf1+PD-1+ TILs mediated the proliferative response to immunotherapy, generating both Tcf1+PD-1+ and differentiated Tcf1-PD-1+ cells. Ablation of Tcf1+PD-1+ TILs restricted responses to immunotherapy. Tcf1 was not required for the generation of Tcf1+PD-1+ TILs but was essential for the stem-like functions of these cells. Human TCF1+PD-1+ cells were detected among tumor-reactive CD8+ T cells in the blood of melanoma patients and among TILs of primary melanomas. Thus, immune checkpoint blockade relies not on reversal of T cell exhaustion programs, but on the proliferation of a stem-like TIL subset.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Stem Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation , Cell Proliferation , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Melanoma/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL
18.
J Invest Dermatol ; 139(6): 1318-1328, 2019 06.
Article in English | MEDLINE | ID: mdl-30594488

ABSTRACT

Neutrophils are rapidly recruited to the mammalian skin in response to infection with the cutaneous Leishmania pathogen. The parasites use neutrophils to establish the disease; however, the signals driving early neutrophil recruitment are poorly known. Here, we identified the functional importance of TLR2 signaling in this process. Using bone marrow chimeras and immunohistology, we identified the TLR2-expressing cells involved in this early neutrophil recruitment to be of nonhematopoietic origin. Keratinocytes are damaged and briefly in contact with the parasites during infection. We show that TLR2 triggering by Leishmania major is required for their secretion of neutrophil-attracting chemokines. Furthermore, TLR2 triggering by L. major phosphoglycans is critical for neutrophil recruitment to negatively affect disease development, as shown by better control of lesion size and parasite load in Tlr2-/- compared with wild-type infected mice. Conversely, restoring early neutrophil presence in Tlr2-/- mice through injection of wild-type neutrophils or CXCL1 at the onset of infection resulted in delayed disease resolution comparable to that observed in wild-type mice. Taken together, our data show a crucial role for TLR2-expressing nonhematopoietic skin cells in the recruitment of the first wave of neutrophils after L. major infection, a process that delays disease control.


Subject(s)
Keratinocytes/metabolism , Leishmania major/immunology , Leishmaniasis, Cutaneous/immunology , Neutrophils/immunology , Toll-Like Receptor 2/metabolism , Animals , Bone Marrow Transplantation , Cell Communication/immunology , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , Disease Models, Animal , Humans , Keratinocytes/immunology , Leishmania major/isolation & purification , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Mice , Mice, Knockout , Neutrophil Infiltration , Parasite Load , Signal Transduction/genetics , Signal Transduction/immunology , Skin/cytology , Skin/immunology , Skin/parasitology , Skin/pathology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Transplantation Chimera
19.
Front Immunol ; 9: 2573, 2018.
Article in English | MEDLINE | ID: mdl-30483254

ABSTRACT

Adult hematopoiesis takes place in the perivascular zone of the bone cavity, where endothelial cells, mesenchymal stromal/stem cells and their derivatives such as osteoblasts are key components of bone marrow (BM) niches. Defining the contribution of BM adipocytes to the hematopoietic stem cell niche remains controversial. While an excess of medullar adiposity is generally considered deleterious for hematopoiesis, an active role for adipocytes in shaping the niche has also been proposed. We thus investigated the consequences of total adipocyte deletion, including in the BM niche, on adult hematopoiesis using mice carrying a constitutive deletion of the gene coding for the nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ). We show that PpargΔ/Δ lipodystrophic mice exhibit severe extramedullary hematopoiesis (EMH), which we found to be non-cell autonomous, as it is reproduced when wild-type donor BM cells are transferred into PpargΔ/Δ recipients. This phenotype is not due to a specific alteration linked to Pparg deletion, such as chronic inflammation, since it is also found in AZIPtg/+ mice, another lipodystrophic mouse model with normal PPARγ expression, that display only very moderate levels of inflammation. In both models, the lack of adipocytes alters subpopulations of both myeloid and lymphoid cells. The CXCL12/CXCR4 axis in the BM is also dysregulated in an adipocyte deprived environment supporting the hypothesis that adipocytes are required for normal hematopoietic stem cell mobilization or retention. Altogether, these data suggest an important role for adipocytes, and possibly for the molecular interactions they provide within the BM, in maintaining the appropriate microenvironment for hematopoietic homeostasis.


Subject(s)
Adipocytes/physiology , Hematopoiesis/physiology , Adipocytes/metabolism , Adipogenesis/physiology , Animals , Bone Marrow/metabolism , Bone Marrow/physiology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone and Bones/metabolism , Bone and Bones/physiology , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Knockout , Mice, Transgenic , Osteoblasts/metabolism , Osteoblasts/physiology , PPAR gamma/metabolism , Receptors, CXCR4/metabolism , Stem Cell Niche/physiology
20.
J Exp Med ; 215(11): 2760-2777, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30355615

ABSTRACT

The mammalian lymphatic system consists of strategically located lymph nodes (LNs) embedded into a lymphatic vascular network. Mechanisms underlying development of this highly organized system are not fully understood. Using high-resolution imaging, we show that lymphoid tissue inducer (LTi) cells initially transmigrate from veins at LN development sites using gaps in venous mural coverage. This process is independent of lymphatic vasculature, but lymphatic vessels are indispensable for the transport of LTi cells that egress from blood capillaries elsewhere and serve as an essential LN expansion reservoir. At later stages, lymphatic collecting vessels ensure efficient LTi cell transport and formation of the LN capsule and subcapsular sinus. Perinodal lymphatics also promote local interstitial flow, which cooperates with lymphotoxin-ß signaling to amplify stromal CXCL13 production and thereby promote LTi cell retention. Our data unify previous models of LN development by showing that lymphatics intervene at multiple points to assist LN expansion and identify a new role for mechanical forces in LN development.


Subject(s)
Embryo, Mammalian/embryology , Lymph Nodes/embryology , Lymphangiogenesis/physiology , Lymphatic Vessels/embryology , Organogenesis/physiology , Signal Transduction/physiology , Animals , Embryo, Mammalian/immunology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...