Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nat Immunol ; 23(8): 1236-1245, 2022 08.
Article in English | MEDLINE | ID: mdl-35882933

ABSTRACT

Tissue-resident memory T cells (TRM cells) provide rapid and superior control of localized infections. While the transcription factor Runx3 is a critical regulator of CD8+ T cell tissue residency, its expression is repressed in CD4+ T cells. Here, we show that, as a direct consequence of this Runx3-deficiency, CD4+ TRM cells lacked the transforming growth factor (TGF)-ß-responsive transcriptional network that underpins the tissue residency of epithelial CD8+ TRM cells. While CD4+ TRM cell formation required Runx1, this, along with the modest expression of Runx3 in CD4+ TRM cells, was insufficient to engage the TGF-ß-driven residency program. Ectopic expression of Runx3 in CD4+ T cells incited this TGF-ß-transcriptional network to promote prolonged survival, decreased tissue egress, a microanatomical redistribution towards epithelial layers and enhanced effector functionality. Thus, our results reveal distinct programming of tissue residency in CD8+ and CD4+ TRM cell subsets that is attributable to divergent Runx3 activity.


Subject(s)
Immunologic Memory , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Transforming Growth Factor beta/metabolism
2.
Nat Immunol ; 22(9): 1140-1151, 2021 09.
Article in English | MEDLINE | ID: mdl-34426691

ABSTRACT

Tissue-resident memory T (TRM) cells are non-recirculating cells that exist throughout the body. Although TRM cells in various organs rely on common transcriptional networks to establish tissue residency, location-specific factors adapt these cells to their tissue of lodgment. Here we analyze TRM cell heterogeneity between organs and find that the different environments in which these cells differentiate dictate TRM cell function, durability and malleability. We find that unequal responsiveness to TGFß is a major driver of this diversity. Notably, dampened TGFß signaling results in CD103- TRM cells with increased proliferative potential, enhanced function and reduced longevity compared with their TGFß-responsive CD103+ TRM counterparts. Furthermore, whereas CD103- TRM cells readily modified their phenotype upon relocation, CD103+ TRM cells were comparatively resistant to transdifferentiation. Thus, despite common requirements for TRM cell development, tissue adaptation of these cells confers discrete functional properties such that TRM cells exist along a spectrum of differentiation potential that is governed by their local tissue microenvironment.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Plasticity/immunology , Cellular Microenvironment/immunology , Immunologic Memory/immunology , Animals , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/cytology , Female , Integrin alpha Chains/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , Transforming Growth Factor beta1/metabolism
3.
Nat Immunol ; 21(8): 914-926, 2020 08.
Article in English | MEDLINE | ID: mdl-32424363

ABSTRACT

Adoptive cell therapies using genetically engineered T cell receptor or chimeric antigen receptor T cells are emerging forms of immunotherapy that redirect T cells to specifically target cancer. However, tumor antigen heterogeneity remains a key challenge limiting their efficacy against solid cancers. Here, we engineered T cells to secrete the dendritic cell (DC) growth factor Fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L-secreting T cells expanded intratumoral conventional type 1 DCs and substantially increased host DC and T cell activation when combined with immune agonists poly (I:C) and anti-4-1BB. Importantly, combination therapy led to enhanced inhibition of tumor growth and the induction of epitope spreading towards antigens beyond those recognized by adoptively transferred T cells in solid tumor models of T cell receptor and chimeric antigen receptor T cell therapy. Our data suggest that augmenting endogenous DCs is a promising strategy to overcome the clinical problem of antigen-negative tumor escape following adoptive cell therapy.


Subject(s)
Dendritic Cells/immunology , Immunotherapy, Adoptive , Membrane Proteins/immunology , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Humans , Immunologic Factors , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology
4.
Immunity ; 56(7): 1664-1680.e9, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37392736

ABSTRACT

Memory CD8+ T cells can be broadly divided into circulating (TCIRCM) and tissue-resident memory T (TRM) populations. Despite well-defined migratory and transcriptional differences, the phenotypic and functional delineation of TCIRCM and TRM cells, particularly across tissues, remains elusive. Here, we utilized an antibody screening platform and machine learning prediction pipeline (InfinityFlow) to profile >200 proteins in TCIRCM and TRM cells in solid organs and barrier locations. High-dimensional analyses revealed unappreciated heterogeneity within TCIRCM and TRM cell lineages across nine different organs after either local or systemic murine infection models. Additionally, we demonstrated the relative effectiveness of strategies allowing for the selective ablation of TCIRCM or TRM populations across organs and identified CD55, KLRG1, CXCR6, and CD38 as stable markers for characterizing memory T cell function during inflammation. Together, these data and analytical framework provide an in-depth resource for memory T cell classification in both steady-state and inflammatory conditions.


Subject(s)
CD8-Positive T-Lymphocytes , Memory T Cells , Mice , Animals , Cell Lineage , Immunologic Memory
5.
Immunity ; 53(2): 303-318.e5, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32579887

ABSTRACT

Granulocyte-monocyte progenitors (GMPs) have been previously defined for their potential to generate various myeloid progenies such as neutrophils and monocytes. Although studies have proposed lineage heterogeneity within GMPs, it is unclear if committed progenitors already exist among these progenitors and how they may behave differently during inflammation. By combining single-cell transcriptomic and proteomic analyses, we identified the early committed progenitor within the GMPs responsible for the strict production of neutrophils, which we designate as proNeu1. Our dissection of the GMP hierarchy led us to further identify a previously unknown intermediate proNeu2 population. Similar populations could be detected in human samples. proNeu1s, but not proNeu2s, selectively expanded during the early phase of sepsis at the expense of monocytes. Collectively, our findings help shape the neutrophil maturation trajectory roadmap and challenge the current definition of GMPs.


Subject(s)
Granulocyte Precursor Cells/cytology , Monocytes/cytology , Myelopoiesis/physiology , Neutrophils/cytology , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Single-Cell Analysis
6.
Immunity ; 50(2): 390-402.e10, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30709741

ABSTRACT

Neutrophils eliminate pathogens efficiently but can inflict severe damage to the host if they over-activate within blood vessels. It is unclear how immunity solves the dilemma of mounting an efficient anti-microbial defense while preserving vascular health. Here, we identify a neutrophil-intrinsic program that enabled both. The gene Bmal1 regulated expression of the chemokine CXCL2 to induce chemokine receptor CXCR2-dependent diurnal changes in the transcriptional and migratory properties of circulating neutrophils. These diurnal alterations, referred to as neutrophil aging, were antagonized by CXCR4 (C-X-C chemokine receptor type 4) and regulated the outer topology of neutrophils to favor homeostatic egress from blood vessels at night, resulting in boosted anti-microbial activity in tissues. Mice engineered for constitutive neutrophil aging became resistant to infection, but the persistence of intravascular aged neutrophils predisposed them to thrombo-inflammation and death. Thus, diurnal compartmentalization of neutrophils, driven by an internal timer, coordinates immune defense and vascular protection.


Subject(s)
Blood Vessels/immunology , Circadian Rhythm/immunology , Neutrophils/immunology , Phagocytosis/immunology , Animals , Blood Vessels/metabolism , Candida albicans/immunology , Candida albicans/physiology , Cells, Cultured , Cellular Senescence/immunology , Chemokine CXCL2/immunology , Chemokine CXCL2/metabolism , Host-Pathogen Interactions/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Time Factors
7.
Immunity ; 48(2): 364-379.e8, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29466759

ABSTRACT

Neutrophils are specialized innate cells that require constant replenishment from proliferative bone marrow (BM) precursors as a result of their short half-life. Although it is established that neutrophils are derived from the granulocyte-macrophage progenitor (GMP), the differentiation pathways from GMP to functional mature neutrophils are poorly defined. Using mass cytometry (CyTOF) and cell-cycle-based analysis, we identified three neutrophil subsets within the BM: a committed proliferative neutrophil precursor (preNeu) which differentiates into non-proliferating immature neutrophils and mature neutrophils. Transcriptomic profiling and functional analysis revealed that preNeu require the C/EBPε transcription factor for their generation from the GMP, and their proliferative program is substituted by a gain of migratory and effector function as they mature. preNeus expand under microbial and tumoral stress, and immature neutrophils are recruited to the periphery of tumor-bearing mice. In summary, our study identifies specialized BM granulocytic populations that ensure supply under homeostasis and stress responses.


Subject(s)
Bone Marrow Cells/physiology , Neutrophils/physiology , Animals , Bone Marrow Cells/immunology , CCAAT-Enhancer-Binding Proteins/physiology , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Gene Expression Profiling , Humans , Mice , Neoplasms, Experimental/immunology , Neutrophils/immunology
8.
Immunity ; 49(2): 326-341.e7, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30054204

ABSTRACT

The maintenance of appropriate arterial tone is critically important for normal physiological arterial function. However, the cellular and molecular mechanisms remain poorly defined. Here, we have shown that in the mouse aorta, resident macrophages prevented arterial stiffness and collagen deposition in the steady state. Using phenotyping, transcriptional profiling, and targeted deletion of Csf1r, we have demonstrated that these macrophages-which are a feature of blood vessels invested with smooth muscle cells (SMCs) in both mouse and human tissues-expressed the hyaluronan (HA) receptor LYVE-l. Furthermore, we have shown they possessed the unique ability to modulate collagen expression in SMCs by matrix metalloproteinase MMP-9-dependent proteolysis through engagement of LYVE-1 with the HA pericellular matrix of SMCs. Our study has unveiled a hitherto unknown homeostatic contribution of arterial LYVE-1+ macrophages through the control of collagen production by SMCs and has identified a function of LYVE-1 in leukocytes.


Subject(s)
Collagen/metabolism , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Vascular Stiffness/physiology , Animals , Aorta/physiology , Female , Glycoproteins/genetics , Humans , Hyaluronic Acid/metabolism , Male , Matrix Metalloproteinase 9/metabolism , Membrane Transport Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics
10.
Immunity ; 47(2): 374-388.e6, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813662

ABSTRACT

The liver is positioned at the interface between two routes traversed by pathogens in disseminating infection. Whereas blood-borne pathogens are efficiently cleared in hepatic sinusoids by Kupffer cells (KCs), it is unknown how the liver prevents dissemination of peritoneal pathogens accessing its outer membrane. We report here that the hepatic capsule harbors a contiguous cellular network of liver-resident macrophages phenotypically distinct from KCs. These liver capsular macrophages (LCMs) were replenished in the steady state from blood monocytes, unlike KCs that are embryonically derived and self-renewing. LCM numbers increased after weaning in a microbiota-dependent process. LCMs sensed peritoneal bacteria and promoted neutrophil recruitment to the capsule, and their specific ablation resulted in decreased neutrophil recruitment and increased intrahepatic bacterial burden. Thus, the liver contains two separate and non-overlapping niches occupied by distinct resident macrophage populations mediating immunosurveillance at these two pathogen entry points to the liver.


Subject(s)
Kupffer Cells/physiology , Listeria monocytogenes/immunology , Listeriosis/immunology , Liver/immunology , Macrophages/immunology , Neutrophils/immunology , Peritoneum/microbiology , Animals , Cell Communication , Cell Self Renewal , Host-Pathogen Interactions , Humans , Immunity, Innate , Kupffer Cells/microbiology , Liver/microbiology , Liver/pathology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Neutrophil Infiltration , Peritoneum/pathology
11.
Blood ; 137(20): 2770-2784, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33512478

ABSTRACT

Dendritic cells (DCs) encompass several cell subsets that collaborate to initiate and regulate immune responses. Proper DC localization determines their function and requires the tightly controlled action of chemokine receptors. All DC subsets express CXCR4, but the genuine contribution of this receptor to their biology has been overlooked. We addressed this question using natural CXCR4 mutants resistant to CXCL12-induced desensitization and harboring a gain of function that cause the warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome (WS), a rare immunodeficiency associated with high susceptibility to the pathogenesis of human papillomavirus (HPV). We report a reduction in the number of circulating plasmacytoid DCs (pDCs) in WHIM patients, whereas that of conventional DCs is preserved. This pattern was reproduced in an original mouse model of WS, enabling us to show that the circulating pDC defect can be corrected upon CXCR4 blockade and that pDC differentiation and function are preserved, despite CXCR4 dysfunction. We further identified proper CXCR4 signaling as a critical checkpoint for Langerhans cell and DC migration from the skin to lymph nodes, with corollary alterations of their activation state and tissue inflammation in a model of HPV-induced dysplasia. Beyond providing new hypotheses to explain the susceptibility of WHIM patients to HPV pathogenesis, this study shows that proper CXCR4 signaling establishes a migration threshold that controls DC egress from CXCL12-containing environments and highlights the critical and subset-specific contribution of CXCR4 signal termination to DC biology.


Subject(s)
Dendritic Cells/physiology , Inflammation/pathology , Primary Immunodeficiency Diseases/physiopathology , Receptors, CXCR4/physiology , Warts/physiopathology , Alphapapillomavirus/genetics , Animals , Benzylamines/pharmacology , Cell Count , Cell Differentiation , Chemokine CXCL12/physiology , Chemotaxis , Cyclams/pharmacology , Dendritic Cells/classification , Epidermis/pathology , Female , Gene Knock-In Techniques , Genes, Viral , Humans , Inflammation/metabolism , Langerhans Cells/physiology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Organ Specificity , Parabiosis , Primary Immunodeficiency Diseases/blood , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/pathology , Recombinant Proteins/metabolism , Warts/blood , Warts/genetics , Warts/pathology
13.
Eur J Immunol ; 51(12): 2708-3145, 2021 12.
Article in English | MEDLINE | ID: mdl-34910301

ABSTRACT

The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers.


Subject(s)
Autoimmune Diseases/immunology , Flow Cytometry , Infections/immunology , Neoplasms/immunology , Animals , Chronic Disease , Humans , Mice , Practice Guidelines as Topic
14.
Cytometry A ; 101(11): 922-941, 2022 11.
Article in English | MEDLINE | ID: mdl-35349225

ABSTRACT

Understanding the complex elements affecting signal resolution in cytometry is key for quality experimental design and data. In this study, we incorporate autofluorescence as a contributing factor to our understanding of resolution in cytometry and corroborate its impact in fluorescence signal detection through mathematical predictions supported by empirical evidence. Our findings illustrate the critical importance of autofluorescence extraction via full spectrum unmixing in unmasking dim signals and delineating the expression and subset distribution of low abundance markers in discovery projects. We apply our findings to the precise definition of the tissue and cellular distribution of a weakly expressed fluorescent protein that reports on a low-abundance immunological gene. Exploiting the full spectrum coverage enabled by Aurora 5L, we describe a novel approach to the isolation of pure cell subset-specific autofluorescence profiles based on high dimensionality reduction algorithms. This method can also be used to unveil differences in the autofluorescent fingerprints of tissues in homeostasis and after immunological challenges.


Subject(s)
Algorithms , Coloring Agents , Immunophenotyping
16.
Bioinformatics ; 35(2): 301-308, 2019 01 15.
Article in English | MEDLINE | ID: mdl-29931307

ABSTRACT

Motivation: Recent flow and mass cytometers generate datasets of dimensions 20 to 40 and a million single cells. From these, many tools facilitate the discovery of new cell populations associated with diseases or physiology. These new cell populations require the identification of new gating strategies, but gating strategies become exponentially more difficult to optimize when dimensionality increases. To facilitate this step, we developed Hypergate, an algorithm which given a cell population of interest identifies a gating strategy optimized for high yield and purity. Results: Hypergate achieves higher yield and purity than human experts, Support Vector Machines and Random-Forests on public datasets. We use it to revisit some established gating strategies for the identification of innate lymphoid cells, which identifies concise and efficient strategies that allow gating these cells with fewer parameters but higher yield and purity than the current standards. For phenotypic description, Hypergate's outputs are consistent with fields' knowledge and sparser than those from a competing method. Availability and implementation: Hypergate is implemented in R and available on CRAN. The source code is published at http://github.com/ebecht/hypergate under an Open Source Initiative-compliant licence. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Cell Separation/methods , Computational Biology , Flow Cytometry , Lymphocytes/cytology , Humans , Immunity, Innate
17.
Eur J Immunol ; 48(6): 1014-1019, 2018 06.
Article in English | MEDLINE | ID: mdl-29510451

ABSTRACT

Pressure ulcers are a chronic problem for patients or the elderly who require extended periods of bed rest. The formation of ulcers is due to repeated cycles of ischemia-reperfusion (IR), which initiates an inflammatory response. Advanced ulcers disrupt the skin barrier, resulting in further complications. To date, the immunological aspect of skin IR has been understudied, partly due to the complexity of the skin immune cells. Through a combination of mass cytometry, confocal imaging and intravital multiphoton imaging, this study establishes a workflow for multidimensionality single cell analysis of skin myeloid cell responses in the context of IR injury with high spatiotemporal resolution. The data generated has provided us with previously uncharacterized insights into the distinct cellular behavior of resident dendritic cells (DCs) and recruited neutrophils post IR. Of interest, we observed a drop in DDC numbers in the IR region, which was subsequently replenished 48h post IR. More importantly, in these cells, we observe an attenuated response to repeated injuries, which may have implications in the subsequent wound healing process.


Subject(s)
Dendritic Cells/immunology , Neutrophils/immunology , Pressure Ulcer/immunology , Reperfusion Injury/immunology , Skin/pathology , Aged , Animals , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cell Movement , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Single-Cell Analysis
18.
Blood ; 122(22): 3666-77, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24113869

ABSTRACT

Lymphangiogenesis is an important physiological response to inflammatory insult, acting to limit inflammation. Macrophages, dendritic cells, and lymphocytes are known to drive lymphangiogenesis. In this study, we show that neutrophils recruited to sites of inflammation can also coordinate lymphangiogenesis. In the absence of B cells, intranodal lymphangiogenesis induced during prolonged inflammation as a consequence of immunization is dependent on the accumulation of neutrophils. When neutrophils are depleted in wild-type mice developing skin inflammation in response to immunization or contact hypersensitization, lymphangiogenesis is decreased and local inflammation is increased. We demonstrate that neutrophils contribute to lymphangiogenesis primarily by modulating vascular endothelial growth factor (VEGF)-A bioavailability and bioactivity and, to a lesser extent, secreting VEGF-D. We further show that neutrophils increased VEGF-A bioavailability and bioactivity via the secretion of matrix metalloproteinases 9 and heparanase. Together, these findings uncover a novel function for neutrophils as organizers of lymphangiogenesis during inflammation.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Lymphangiogenesis/physiology , Neutrophils/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor D/metabolism , Animals , B-Lymphocytes/immunology , Dermatitis/etiology , Dermatitis/metabolism , Dermatitis/pathology , Female , Glucuronidase/metabolism , Inflammation/pathology , Lymphangiogenesis/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neutrophils/immunology , Neutrophils/pathology , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
J Immunol ; 191(3): 1006-10, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23817414

ABSTRACT

The complement system is a potent component of the innate immune response, promoting inflammation and orchestrating defense against pathogens. However, dysregulation of complement is critical to several autoimmune and inflammatory syndromes. Elevated expression of the proinflammatory cytokine IL-1ß is often linked to such diseases. In this study, we reveal the mechanistic link between complement and IL-1ß secretion using murine dendritic cells. IL-1ß secretion occurs following intracellular caspase-1 activation by inflammasomes. We show that complement elicits secretion of both IL-1ß and IL-18 in vitro and in vivo via the NLRP3 inflammasome. This effect depends on the inflammasome components NLRP3 and ASC, as well as caspase-1 activity. Interestingly, sublethal complement membrane attack complex formation, but not the anaphylatoxins C3a and C5a, activated the NLRP3 inflammasome in vivo. These findings provide insight into the molecular processes underlying complement-mediated inflammation and highlight the possibility of targeting IL-1ß to control complement-induced disease and pathological inflammation.


Subject(s)
Carrier Proteins/metabolism , Caspase 1/metabolism , Dendritic Cells/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Animals , Bone Marrow Cells , Carrier Proteins/genetics , Cells, Cultured , Complement C6/deficiency , Complement C6/genetics , Complement System Proteins/immunology , Dendritic Cells/metabolism , Enzyme Activation , Inflammation/immunology , Interleukin-18/biosynthesis , Interleukin-18/immunology , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptors, Complement/deficiency , Receptors, Complement/genetics , Signal Transduction
20.
Nat Commun ; 15(1): 2619, 2024 Mar 23.
Article in English | MEDLINE | ID: mdl-38521764

ABSTRACT

Immunity to infectious diseases is predominantly studied by measuring immune responses towards a single pathogen, although co-infections are common. In-depth mechanisms on how co-infections impact anti-viral immunity are lacking, but are highly relevant to treatment and prevention. We established a mouse model of co-infection with unrelated viruses, influenza A (IAV) and Semliki Forest virus (SFV), causing disease in different organ systems. SFV infection eight days before IAV infection results in prolonged IAV replication, elevated cytokine/chemokine levels and exacerbated lung pathology. This is associated with impaired lung IAV-specific CD8+ T cell responses, stemming from suboptimal CD8+ T cell activation and proliferation in draining lymph nodes, and dendritic cell paralysis. Prior SFV infection leads to increased blood brain barrier permeability and presence of IAV RNA in brain, associated with increased trafficking of IAV-specific CD8+ T cells and establishment of long-term tissue-resident memory. Relative to lung IAV-specific CD8+ T cells, brain memory IAV-specific CD8+ T cells have increased TCR repertoire diversity within immunodominant DbNP366+CD8+ and DbPA224+CD8+ responses, featuring suboptimal TCR clonotypes. Overall, our study demonstrates that infection with an unrelated neurotropic virus perturbs IAV-specific immune responses and exacerbates IAV disease. Our work provides key insights into therapy and vaccine regimens directed against unrelated pathogens.


Subject(s)
Coinfection , Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Viruses , Mice , Animals , Humans , Influenza, Human/pathology , CD8-Positive T-Lymphocytes , Coinfection/pathology , Receptors, Antigen, T-Cell , Lung/pathology
SELECTION OF CITATIONS
SEARCH DETAIL