Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 9(1): 66, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38514656

ABSTRACT

Antigens from viruses or immunizations can persist or are archived in lymph node stromal cells such as lymphatic endothelial cells (LEC) and fibroblastic reticular cells (FRC). Here, we find that, during the time frame of antigen archiving, LEC apoptosis caused by a second, but unrelated, innate immune stimulus such as vaccina viral infection or CpG DNA administration resulted in cross-presentation of archived antigens and boosted memory CD8 + T cells specific to the archived antigen. In contrast to "bystander" activation associated with unrelated infections, the memory CD8 + T cells specific to the archived antigen from the immunization were significantly higher than memory CD8 + T cells of a different antigen specificity. Finally, the boosted memory CD8 + T cells resulted in increased protection against Listeria monocytogenes expressing the antigen from the immunization, but only for the duration that the antigen was archived. These findings outline an important mechanism by which lymph node stromal cell archived antigens, in addition to bystander activation, can augment memory CD8 + T cell responses during repeated inflammatory insults.

2.
J Immunol ; 209(11): 2149-2159, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36426978

ABSTRACT

Successful vaccination strategies offer the potential for lifelong immunity against infectious diseases and cancer. There has been increased attention regarding the limited translation of some preclinical findings generated using specific pathogen-free (SPF) laboratory mice to humans. One potential reason for the difference between preclinical and clinical findings lies in maturation status of the immune system at the time of challenge. In this study, we used a "dirty" mouse model, where SPF laboratory mice were cohoused (CoH) with pet store mice to permit microbe transfer and immune system maturation, to investigate the priming of a naive T cell response after vaccination with a peptide subunit mixed with polyinosinic-polycytidylic acid and agonistic anti-CD40 mAb. Although this vaccination platform induced robust antitumor immunity in SPF mice, it failed to do so in microbially experienced CoH mice. Subsequent investigation revealed that despite similar numbers of Ag-specific naive CD4 and CD8 T cell precursors, the expansion, differentiation, and recall responses of these CD4 and CD8 T cell populations in CoH mice were significantly reduced compared with SPF mice after vaccination. Evaluation of the dendritic cell compartment revealed reduced IL-27p28 expression by XCR1+ dendritic cells from CoH mice after vaccination, correlating with reduced T cell expansion. Importantly, administration of recombinant IL-27:EBI3 complex to CoH mice shortly after vaccination significantly boosted Ag-specific CD8 and CD4 T cell expansion, further implicating the defect to be T cell extrinsic. Collectively, our data show the potential limitation of exclusive use of SPF mice when testing vaccine efficacy.


Subject(s)
Interleukin-27 , Humans , Mice , Animals , Interleukin-27/metabolism , CD8-Positive T-Lymphocytes , CD40 Antigens , Cell Differentiation , Dendritic Cells
3.
J Biol Chem ; 298(12): 102694, 2022 12.
Article in English | MEDLINE | ID: mdl-36375639

ABSTRACT

Lymphatic endothelial cells (LECs) comprise lymphatic capillaries and vessels that guide immune cells to lymph nodes (LNs) and form the subcapsular sinus and cortical and medullary lymphatic structures of the LN. During an active immune response, the lymphatics remodel to accommodate the influx of immune cells from the tissue, but factors involved in remodeling are unclear. Here, we determined that a TSS motif within the cytoplasmic domain of programmed death ligand 1 (PD-L1), expressed by LECs in the LN, participates in lymphatic remodeling. Mutation of the TSS motif to AAA does not affect surface expression of PD-L1, but instead causes defects in LN cortical and medullary lymphatic organization following immunostimulant, Poly I:C, administration in vivo. Supporting this observation, in vitro treatment of the LEC cell line, SVEC4-10, with cytokines TNFα and IFNα significantly impeded SVEC4-10 movement in the presence of the TSS-AAA cytoplasmic mutation. The cellular movement defects coincided with reduced F-actin polymerization, consistent with differences previously found in dendritic cells. Here, in addition to loss of actin polymerization, we define STAT3 and Paxillin as important PD-L1 binding partners. STAT3 and Paxillin were previously demonstrated to be important at focal adhesions for cellular motility. We further demonstrate the PD-L1 TSS-AAA motif mutation reduced the amount of pSTAT3 and Paxillin bound to PD-L1 both before and after exposure to TNFα and IFNα. Together, these findings highlight PD-L1 as an important component of a membrane complex that is involved in cellular motility, which leads to defects in lymphatic organization.


Subject(s)
B7-H1 Antigen , Paxillin , Tumor Necrosis Factor-alpha , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Endothelial Cells , Paxillin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Lymphoid Tissue/metabolism , Animals , Mice , Mutation
4.
Front Immunol ; 13: 814203, 2022.
Article in English | MEDLINE | ID: mdl-35145521

ABSTRACT

T cells and B cells have been identified in human and murine islets, but the phenotype and role of islet lymphocytes is unknown. Resident immune populations set the stage for responses to inflammation in the islets during homeostasis and diabetes. Thus, we sought to identify the phenotype and effector function of islet lymphocytes to better understand their role in normal islets and in islets under metabolic stress. Lymphocytes were located in the islet parenchyma, and were comprised of a mix of naïve, activated, and memory T cell and B cell subsets, with an enrichment for regulatory B cell subsets. Use of a Nur77 reporter indicated that CD8 T cells and B cells both received local antigen stimulus, indicating that they responded to antigens present in the islets. Analysis of effector function showed that islet T cells and B cells produced the regulatory cytokine IL-10. The regulatory phenotype of islet T cells and B cells and their response to local antigenic stimuli remained stable under conditions of metabolic stress in the diet induced obesity (DIO) model. T cells present in human islets retained a similar activated and memory phenotype in non-diabetic and T2D donors. Under steady-state conditions, islet T cells and B cells have a regulatory phenotype, and thus may play a protective role in maintaining tissue homeostasis.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Homeostasis/physiology , Islets of Langerhans/immunology , Stress, Physiological/physiology , T-Lymphocytes/immunology , Animals , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 2/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Obesity/immunology , Phenotype
5.
EMBO J ; 40(22): e108966, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34618370

ABSTRACT

Viremia in the vertebrate host is a major determinant of arboviral reservoir competency, transmission efficiency, and disease severity. However, immune mechanisms that control arboviral viremia are poorly defined. Here, we identify critical roles for the scavenger receptor MARCO in controlling viremia during arthritogenic alphavirus infections in mice. Following subcutaneous inoculation, arthritogenic alphavirus particles drain via the lymph and are rapidly captured by MARCO+ lymphatic endothelial cells (LECs) in the draining lymph node (dLN), limiting viral spread to the bloodstream. Upon reaching the bloodstream, alphavirus particles are cleared from the circulation by MARCO-expressing Kupffer cells in the liver, limiting viremia and further viral dissemination. MARCO-mediated accumulation of alphavirus particles in the draining lymph node and liver is an important host defense mechanism as viremia and viral tissue burdens are elevated in MARCO-/- mice and disease is more severe. In contrast to prior studies implicating a key role for lymph node macrophages in limiting viral dissemination, these findings exemplify a previously unrecognized arbovirus-scavenging role for lymphatic endothelial cells and improve our mechanistic understanding of viremia control during arthritogenic alphavirus infection.


Subject(s)
Alphavirus Infections/virology , Lymph Nodes/cytology , Receptors, Immunologic/metabolism , Viremia/pathology , Alphavirus/pathogenicity , Animals , Chikungunya Fever/genetics , Chikungunya Fever/virology , Endothelial Cells/virology , Host-Pathogen Interactions , Kupffer Cells/virology , Lymph Nodes/virology , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , RNA, Viral/metabolism , Receptors, Immunologic/genetics , Single-Cell Analysis , Viremia/virology
6.
Elife ; 102021 04 12.
Article in English | MEDLINE | ID: mdl-33843587

ABSTRACT

The detection of foreign antigens in vivo has relied on fluorescent conjugation or indirect read-outs such as antigen presentation. In our studies, we found that these widely used techniques had several technical limitations that have precluded a complete picture of antigen trafficking or retention across lymph node cell types. To address these limitations, we developed a 'molecular tracking device' to follow the distribution, acquisition, and retention of antigen in the lymph node. Utilizing an antigen conjugated to a nuclease-resistant DNA tag, acting as a combined antigen-adjuvant conjugate, and single-cell mRNA sequencing, we quantified antigen abundance in the lymph node. Variable antigen levels enabled the identification of caveolar endocytosis as a mechanism of antigen acquisition or retention in lymphatic endothelial cells. Thus, these molecular tracking devices enable new approaches to study dynamic tissue dissemination of antigen-adjuvant conjugates and identify new mechanisms of antigen acquisition and retention at cellular resolution in vivo.


The lymphatic system is a network of ducts that transports fluid, proteins, and immune cells from different organs around the body. Lymph nodes provide pit stops at hundreds of points along this network where immune cells reside, and lymph fluid can be filtered and cleaned. When pathogens, such as viruses or bacteria, enter the body during an infection, fragments of their proteins can get swept into the lymph nodes. These pathogenic proteins or protein fragments activate resident immune cells and kickstart the immune response. Vaccines are designed to mimic this process by introducing isolated pathogenic proteins in a controlled way to stimulate similar immune reactions in lymph nodes. Once an infection has been cleared by the immune system, or a vaccination has triggered the immune system, most pathogenic proteins get cleared away. However, a small number of pathogenic proteins remain in the lymph nodes to enable immune cells to respond more strongly and quickly the next time they see the same pathogen. Yet it is largely unclear how much protein remains for training and how or where it is all stored. Current techniques are not sensitive or long-lived enough to accurately detect and track these small protein deposits over time. Walsh, Sheridan, Lucas, et al. have addressed this problem by developing biological tags that can be attached to the pathogenic proteins so they can be traced. These tags were designed so the body cannot easily break them down, helping them last as long as the proteins they are attached to. Walsh, Sheridan, Lucas et al. tested whether vaccinating mice with the tagged proteins allowed the proteins to be tracked. The method they used was designed to identify individual cell types based on their genetic information along with the tag. This allowed them to accurately map the complex network of cells involved in storing and retrieving archived protein fragments, as well as those involved in training new immune cells to recognize them. These results provide important insights into the protein archiving system that is involved in enhancing immune memory. This may help guide the development of new vaccination strategies that can manipulate how proteins are archived to establish more durable immune protection. The biological tags developed could also be used to track therapeutic proteins, allowing scientists to determine how long cancer drugs, antibody therapies or COVID19 anti-viral agents remain in the body. This information could then be used by doctors to plan specific and personalized treatment timetables for patients.


Subject(s)
Antigens/metabolism , Lymph Nodes/metabolism , Single-Cell Analysis , Animals , Antigen Presentation , Antigens/genetics , Antigens/immunology , Caveolae/immunology , Caveolae/metabolism , Cells, Cultured , DNA/genetics , DNA/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis , Endothelial Cells/immunology , Endothelial Cells/metabolism , Lymph Nodes/immunology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Peptide Fragments/genetics , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phosphorothioate Oligonucleotides/genetics , Phosphorothioate Oligonucleotides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Time Factors , Tissue Distribution , Transcriptome
7.
Cell Rep ; 33(2): 108258, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053342

ABSTRACT

Although the function of the extracellular region of programmed death ligand 1 (PD-L1) through its interactions with PD-1 on T cells is well studied, little is understood regarding the intracellular domain of PD-L1. Here, we outline a major role for PD-L1 intracellular signaling in the control of dendritic cell (DC) migration from the skin to the draining lymph node (dLN). Using a mutant mouse model, we identify a TSS signaling motif within the intracellular domain of PD-L1. The TSS motif proves critical for chemokine-mediated DC migration to the dLN during inflammation. This loss of DC migration, in the PD-L1 TSS mutant, leads to a significant decline in T cell priming when DC trafficking is required for antigen delivery to the dLN. Finally, the TSS motif is required for chemokine receptor signaling downstream of the Gα subunit of the heterotrimeric G protein complex, ERK phosphorylation, and actin polymerization in DCs.


Subject(s)
B7-H1 Antigen/metabolism , Cell Movement , Dendritic Cells/metabolism , Dermis/cytology , Immunity , Signal Transduction , Actins/metabolism , Amino Acids/genetics , Animals , B7-H1 Antigen/chemistry , B7-H1 Antigen/deficiency , Base Sequence , CD8-Positive T-Lymphocytes/immunology , Cell Count , Cell Movement/drug effects , Chemokine CCL21/pharmacology , Chemotaxis/drug effects , Dendritic Cells/drug effects , Exons/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Proteins/metabolism , Immunity/drug effects , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mutation/genetics , Phosphorylation/drug effects , Poly I-C/pharmacology , Polymerization , Protein Domains , Receptors, CCR7/metabolism , Signal Transduction/drug effects
9.
Front Immunol ; 10: 36, 2019.
Article in English | MEDLINE | ID: mdl-30740101

ABSTRACT

Lymphatic endothelial cells (LECs) form the structure of the lymphatic vessels and the sinuses of the lymph nodes, positioning them to be key players in many different aspects of the immune response. Following an inflammatory stimulus, LECs produce chemokines that recruit immune cells to the lymph nodes. The recruitment of immune cells aids in the coordination of both LEC and lymph node expansion and contraction. More recent data has demonstrated that to coordinate LEC division and death, cell surface molecules, such as PD-L1 and interferon receptors, are required. During homeostasis, LECs use PD-L1 to maintain peripheral tolerance by presenting specific peripheral tissue antigens in order to eliminate tissue specific responses. LECs also have the capacity to acquire, present, and exchange foreign antigens following viral infection or immunization. Here we will review how lymph node LECs require immune cells to expand and contract in response to an immune stimulus, the factors involved and how direct LEC-immune cell interactions are important for programming immunity.


Subject(s)
Endothelial Cells/immunology , Immunity, Cellular/immunology , Lymph Nodes/immunology , Antigen Presentation/immunology , Apoptosis/immunology , B7-H1 Antigen/metabolism , Cell Communication/immunology , Cell Division/immunology , Cell Proliferation , Endothelial Cells/cytology , Humans , Inflammation/immunology , Lymphatic Vessels/cytology , Platelet Factor 4/metabolism , Receptors, Interferon/metabolism
10.
J Immunol ; 201(6): 1735-1747, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30045970

ABSTRACT

Lymph node (LN) expansion during an immune response is a complex process that involves the relaxation of the fibroblastic network, germinal center formation, and lymphatic vessel growth. These processes require the stromal cell network of the LN to act deliberately to accommodate the influx of immune cells to the LN. The molecular drivers of these processes are not well understood. Therefore, we asked whether the immediate cytokines type 1 IFN produced during viral infection influence the lymphatic network of the LN in mice. We found that following an IFN-inducing stimulus such as viral infection or polyI:C, programmed cell death ligand 1 (PD-L1) expression is dynamically upregulated on lymphatic endothelial cells (LECs). We found that reception of type 1 IFN by LECs is important for the upregulation of PD-L1 of mouse and human LECs and the inhibition of LEC expansion in the LN. Expression of PD-L1 by LECs is also important for the regulation of LN expansion and contraction after an IFN-inducing stimulus. We demonstrate a direct role for both type 1 IFN and PD-L1 in inhibiting LEC division and in promoting LEC survival. Together, these data reveal a novel mechanism for the coordination of type 1 IFN and PD-L1 in manipulating LEC expansion and survival during an inflammatory immune response.


Subject(s)
B7-H1 Antigen/immunology , Cell Proliferation , Endothelial Cells/immunology , Endothelium, Lymphatic/immunology , Interferon Type I/immunology , Animals , B7-H1 Antigen/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/immunology , Endothelial Cells/pathology , Endothelium, Lymphatic/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Type I/genetics , Mice , Mice, Knockout , Poly I-C/pharmacology
11.
JCI Insight ; 3(13)2018 07 12.
Article in English | MEDLINE | ID: mdl-29997290

ABSTRACT

Chikungunya virus (CHIKV) causes acute and chronic rheumatologic disease. Pathogenic CHIKV strains persist in joints of immunocompetent mice, while the attenuated CHIKV strain 181/25 is cleared by adaptive immunity. We analyzed the draining lymph node (dLN) to define events in lymphoid tissue that may contribute to CHIKV persistence or clearance. Acute 181/25 infection resulted in dLN enlargement and germinal center (GC) formation, while the dLN of mice infected with pathogenic CHIKV became highly disorganized and depleted of lymphocytes. Using CHIKV strains encoding ovalbumin-specific TCR epitopes, we found that lymphocyte depletion was not due to impaired lymphocyte proliferation. Instead, the accumulation of naive lymphocytes transferred from the vasculature to the dLN was reduced, which was associated with fewer high endothelial venule cells and decreased CCL21 production. Following NP-OVA immunization, NP-specific GC B cells in the dLN were decreased during pathogenic, but not attenuated, CHIKV infection. Our data suggest that pathogenic, persistent strains of CHIKV disable the development of adaptive immune responses within the dLN.


Subject(s)
Adaptive Immunity , Chikungunya Fever/immunology , Chikungunya virus/pathogenicity , Lymph Nodes/immunology , Animals , B-Lymphocytes , Cell Proliferation , Chemokine CCL21 , Chikungunya Fever/virology , Disease Models, Animal , Epitopes , Immunization , Lymph Nodes/pathology , Lymph Nodes/virology , Lymphocyte Activation , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Ovalbumin , Stromal Cells/immunology , Venules
12.
Nat Commun ; 8(1): 2034, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229919

ABSTRACT

Antigens derived from viral infection or vaccination can persist within a host for many weeks after resolution of the infection or vaccine responses. We previously identified lymphatic endothelial cells (LEC) as the repository for this antigen archival, yet LECs are unable to present their archived antigens to CD8+ T cells, and instead transfer their antigens to CD11c+ antigen-presenting cells (APC). Here we show that the exchange of archived antigens between LECs and APCs is mediated by migratory dendritic cells (DC). After vaccination, both migratory basic leucine zipper ATF-like transcription factor 3 (BatF3)-dependent and BatF3-independent DCs are responsible for antigen exchange and cross-presentation. However, exchange of archived viral antigens is mediated only by BatF3-dependent migratory DCs potentially acquiring apoptotic LECs. In conclusion, LEC-archived antigens are exchanged with migratory DCs, both directly and through LEC apoptosis, to cross-present archived antigens to circulating T cells.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Movement/immunology , Female , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Repressor Proteins/immunology , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...