32 results on '"Pewe LL"'
Search Results
2. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice.
- Author
-
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, and Impens F
- Subjects
- Animals, Mice, Proteomics methods, Antigens, Bacterial immunology, Mice, Inbred C57BL, Peptides immunology, Epitope Mapping methods, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class I metabolism, Bacterial Proteins immunology, Bacterial Proteins metabolism, Female, Spleen immunology, Spleen metabolism, Listeria monocytogenes immunology, Epitopes, T-Lymphocyte immunology, CD8-Positive T-Lymphocytes immunology, Listeriosis immunology, Listeriosis microbiology
- Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8
+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification., Competing Interests: Conflict of interest The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
3. Dynamic landscapes and protective immunity coordinated by influenza-specific lung-resident memory CD8 + T cells revealed by intravital imaging.
- Author
-
van de Wall S, Anthony SM, Hancox LS, Pewe LL, Langlois RA, Zehn D, Badovinac VP, and Harty JT
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Interferon-gamma metabolism, Interferon-gamma immunology, Intravital Microscopy, Monocytes immunology, Lung immunology, Lung virology, Orthomyxoviridae Infections immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Integrin alpha Chains metabolism, Influenza A virus immunology, Antigens, CD metabolism, Memory T Cells immunology
- Abstract
Lung-tissue-resident memory (T
RM ) CD8+ T cells are critical for heterosubtypic immunity against influenza virus (IAV) reinfection. How TRM cells surveil the lung, respond to infection, and interact with other cells remains unresolved. Here, we used IAV infection of mice in combination with intravital and static imaging to define the spatiotemporal dynamics of lung TRM cells before and after recall infection. CD69+ CD103+ TRM cells preferentially localized to lung sites of prior IAV infection, where they exhibited patrolling behavior. After rechallenge, lung TRM cells formed tight clusters in an antigen-dependent manner. Transcriptomic analysis of IAV-specific TRM cells revealed the expression of several factors that regulate myeloid cell biology. In vivo rechallenge experiments demonstrated that protection elicited by TRM cells is orchestrated in part by interferon (IFN)-γ-mediated recruitment of inflammatory monocytes into the lungs. Overall, these data illustrate the dynamic landscapes of CD103+ lung TRM cells that mediate early protective immunity against IAV infection., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
4. Regenerating murine CD8+ lung tissue resident memory T cells after targeted radiation exposure.
- Author
-
Hassert M, Pewe LL, He R, Heidarian M, Phruttiwanichakun P, van de Wall S, Mix MR, Salem AK, Badovinac VP, and Harty JT
- Subjects
- Mice, Animals, CD8-Positive T-Lymphocytes, Memory T Cells, Lung, Immunologic Memory, Orthomyxoviridae Infections, Influenza A virus, Radiation Exposure
- Abstract
Radiation exposure occurs during medical procedures, nuclear accidents, or spaceflight, making effective medical countermeasures a public health priority. Naïve T cells are highly sensitive to radiation-induced depletion, although their numbers recover with time. Circulating memory CD8+ T cells are also depleted by radiation; however, their numbers do not recover. Critically, the impact of radiation exposure on tissue-resident memory T cells (TRM) remains unknown. Here, we found that sublethal thorax-targeted radiation resulted in the rapid and prolonged numerical decline of influenza A virus (IAV)-specific lung TRM in mice, but no decline in antigen-matched circulating memory T cells. Prolonged loss of lung TRM was associated with decreased heterosubtypic immunity. Importantly, boosting with IAV-epitope expressing pathogens that replicate in the lungs or peripheral tissues or with a peripherally administered mRNA vaccine regenerated lung TRM that was derived largely from circulating memory CD8+ T cells. Designing effective vaccination strategies to regenerate TRM will be important in combating the immunological effects of radiation exposure., (© 2024 Hassert et al.)
- Published
- 2024
- Full Text
- View/download PDF
5. Discrete class I molecules on brain endothelium differentially regulate neuropathology in experimental cerebral malaria.
- Author
-
Fain CE, Zheng J, Jin F, Ayasoufi K, Wu Y, Lilley MT, Dropik AR, Wolf DM, Rodriguez RC, Aibaidula A, Tritz ZP, Bouchal SM, Pewe LL, Urban SL, Chen Y, Chang SY, Hansen MJ, Kachergus JM, Shi J, Thompson EA, Jensen HE, Harty JT, Parney IF, Sun J, Wu LJ, and Johnson AJ
- Subjects
- Mice, Humans, Animals, Endothelial Cells pathology, Brain pathology, Blood-Brain Barrier pathology, CD8-Positive T-Lymphocytes, Endothelium pathology, Mice, Inbred C57BL, Disease Models, Animal, Malaria, Cerebral pathology, Malaria, Cerebral prevention & control
- Abstract
Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
6. Sublethal whole-body irradiation induces permanent loss and dysfunction in pathogen-specific circulating memory CD8 T cell populations.
- Author
-
Heidarian M, Jensen IJ, Kannan SK, Pewe LL, Hassert M, Park S, Xue HH, Harty JT, and Badovinac VP
- Subjects
- Mice, Animals, Neoplasm Recurrence, Local, CD8-Positive T-Lymphocytes, Lymphocytic choriomeningitis virus, Immunologic Memory, Mice, Inbred C57BL, Whole-Body Irradiation, Lymphocytic Choriomeningitis
- Abstract
The increasing use of nuclear energy sources inevitably raises the risk of accidental or deliberate radiation exposure and associated immune dysfunction. However, the extent to which radiation exposure impacts memory CD8 T cells, potent mediators of immunity to recurring intracellular infections and malignancies, remains understudied. Using P14 CD8 T cell chimeric mice (P14 chimeras) with an lymphocytic choriomeningitis virus (LCMV) infection model, we observed that sublethal (5Gy) whole-body irradiation (WBI) induced a rapid decline in the number of naive (T
N ) and P14 circulating memory CD8 T cells (TCIRCM ), with the former being more susceptible to radiation-induced numeric loss. While TN cell numbers rapidly recovered, as previously described, the number of P14 TCIRCM cells remained low at least 9 mo after radiation exposure. Additionally, the remaining P14 TCIRCM in irradiated hosts exhibited an inefficient transition to a central memory (CD62L+ ) phenotype compared to nonirradiated P14 chimeras. WBI also resulted in long-lasting T cell intrinsic deficits in memory CD8 T cells, including diminished cytokine and chemokine production along with impaired secondary expansion upon cognate Ag reencounter. Irradiated P14 chimeras displayed significantly higher bacterial burden after challenge with Listeria monocytogenes expressing the LCMV GP33-41 epitope relative to nonirradiated controls, likely due to radiation-induced numerical and functional impairments. Taken together, our findings suggest that sublethal radiation exposure caused a long-term numerical, impaired differentiation, and functional dysregulation in preexisting TCIRCM , rendering previously protected hosts susceptible to reinfection.- Published
- 2023
- Full Text
- View/download PDF
7. CD4 T Cell-Dependent and -Independent Roles for IFN-γ in Blood-Stage Malaria.
- Author
-
Drewry LL, Pewe LL, Hancox LS, Van de Wall S, and Harty JT
- Subjects
- Mice, Animals, CD4-Positive T-Lymphocytes, Mice, Inbred C57BL, Interferon-gamma, Plasmodium chabaudi, Malaria
- Abstract
Production of IFN-γ by CD4 T cells is widely theorized to control Plasmodium parasite burden during blood-stage malaria. Surprisingly, the specific and crucial mechanisms through which this highly pleiotropic cytokine acts to confer protection against malarial disease remain largely untested in vivo. Here we used a CD4 T cell-restricted Cre-Lox IFN-γ excision mouse model to test whether and how CD4 T cell-derived IFN-γ controls blood-stage malaria. Although complete absence of IFN-γ compromised control of the acute and the chronic, recrudescent blood-stage infections with P. c. chabaudi, we identified a specific, albeit modest, role for CD4 T cell-derived IFN-γ in limiting parasite burden only during the chronic stages of P. c. chabaudi malaria. CD4 T cell IFN-γ promoted IgG Ab class switching to the IgG2c isotype during P. c. chabaudi malaria in C57BL/6 mice. Unexpectedly, our data do not support gross defects in phagocytic activity in IFN-γ-deficient hosts infected with blood-stage malaria. Together, our data confirm CD4 T cell-dependent roles for IFN-γ but suggest CD4 T cell-independent roles for IFN-γ in immune responses to blood-stage malaria., (Copyright © 2023 by The American Association of Immunologists, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
8. AIM2 sensors mediate immunity to Plasmodium infection in hepatocytes.
- Author
-
Marques-da-Silva C, Poudel B, Baptista RP, Peissig K, Hancox LS, Shiau JC, Pewe LL, Shears MJ, Kanneganti TD, Sinnis P, Kyle DE, Gurung P, Harty JT, and Kurup SP
- Subjects
- Animals, Humans, Hepatocytes metabolism, Liver, Caspases metabolism, DNA-Binding Proteins metabolism, Malaria parasitology, Plasmodium, Parasites
- Abstract
Malaria, caused by Plasmodium parasites is a severe disease affecting millions of people around the world. Plasmodium undergoes obligatory development and replication in the hepatocytes, before initiating the life-threatening blood-stage of malaria. Although the natural immune responses impeding Plasmodium infection and development in the liver are key to controlling clinical malaria and transmission, those remain relatively unknown. Here we demonstrate that the DNA of Plasmodium parasites is sensed by cytosolic AIM2 (absent in melanoma 2) receptors in the infected hepatocytes, resulting in Caspase-1 activation. Remarkably, Caspase-1 was observed to undergo unconventional proteolytic processing in hepatocytes, resulting in the activation of the membrane pore-forming protein, Gasdermin D, but not inflammasome-associated proinflammatory cytokines. Nevertheless, this resulted in the elimination of Plasmodium -infected hepatocytes and the control of malaria infection in the liver. Our study uncovers a pathway of natural immunity critical for the control of malaria in the liver.
- Published
- 2023
- Full Text
- View/download PDF
9. Cutting Edge: Subunit Booster Vaccination Confers Sterilizing Immunity against Liver-Stage Malaria in Mice Initially Primed with a Weight-Normalized Dose of Radiation-Attenuated Sporozoites.
- Author
-
Lefebvre MN, Drewry LL, Pewe LL, Hancox LS, Reyes-Sandoval A, and Harty JT
- Subjects
- Animals, Immunization, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Vaccination, Liver Diseases immunology, Malaria immunology, Malaria Vaccines immunology, Sporozoites immunology, Vaccines, Attenuated immunology, Vaccines, Subunit immunology
- Abstract
Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations., (Copyright © 2021 by The American Association of Immunologists, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
10. Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria.
- Author
-
Lefebvre MN, Surette FA, Anthony SM, Vijay R, Jensen IJ, Pewe LL, Hancox LS, Van Braeckel-Budimir N, van de Wall S, Urban SL, Mix MR, Kurup SP, Badovinac VP, Butler NS, and Harty JT
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes microbiology, CD8-Positive T-Lymphocytes parasitology, Disease Models, Animal, Female, Host-Parasite Interactions, Listeria monocytogenes immunology, Listeria monocytogenes pathogenicity, Listeriosis blood, Listeriosis immunology, Listeriosis microbiology, Liver metabolism, Liver microbiology, Liver parasitology, Lymphocyte Function-Associated Antigen-1 metabolism, Malaria blood, Malaria parasitology, Male, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Parasite Load, Phagocytes immunology, Phagocytes metabolism, Phagocytes microbiology, Phagocytes parasitology, Plasmodium berghei pathogenicity, Time Factors, Mice, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Liver immunology, Malaria immunology, Plasmodium berghei immunology
- Abstract
Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections., Competing Interests: Declarations of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. p53 Hinders CRISPR/Cas9-Mediated Targeted Gene Disruption in Memory CD8 T Cells In Vivo.
- Author
-
Kurup SP, Moioffer SJ, Pewe LL, and Harty JT
- Subjects
- Animals, Antigens immunology, DNA Damage genetics, DNA Damage immunology, Mice, Mice, Transgenic, CD8-Positive T-Lymphocytes immunology, CRISPR-Cas Systems, Cell Proliferation genetics, Immunologic Memory genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 immunology
- Abstract
CRISPR/Cas9 technology has revolutionized rapid and reliable gene editing in cells. Although many cell types have been subjected to CRISPR/Cas9-mediated gene editing, there is no evidence of success in genetic alteration of Ag-experienced memory CD8 T cells. In this study, we show that CRISPR/Cas9-mediated gene editing in memory CD8 T cells precludes their proliferation after Ag re-encounter in vivo. This defect is mediated by the proapoptotic transcription factor p53, a sensor of DNA damage. Temporarily inhibiting p53 function offers a window of opportunity for the memory CD8 T cells to repair the DNA damage, facilitating robust recall responses on Ag re-encounter. We demonstrate this by functionally altering memory CD8 T cells using CRISPR/Cas9-mediated targeted gene disruption under the aegis of p53siRNA in the mouse model. Our approach thus adapts the CRISPR/Cas9 technology for memory CD8 T cells to undertake gene editing in vivo, for the first time, to our knowledge., (Copyright © 2020 by The American Association of Immunologists, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
12. Peripherally induced brain tissue-resident memory CD8 + T cells mediate protection against CNS infection.
- Author
-
Urban SL, Jensen IJ, Shan Q, Pewe LL, Xue HH, Badovinac VP, and Harty JT
- Subjects
- Animals, Bacterial Infections immunology, Brain cytology, Lymphocyte Activation immunology, Mice, Virus Diseases immunology, Brain immunology, CD8-Positive T-Lymphocytes immunology, Central Nervous System Infections immunology, Immunologic Memory immunology
- Abstract
The central nervous system (CNS) is classically viewed as immune-privileged; however, recent advances highlight interactions between the peripheral immune system and CNS in controlling infections and tissue homeostasis. Tissue-resident memory (T
RM ) CD8+ T cells in the CNS are generated after brain infections, but it is unknown whether CNS infection is required to generate brain TRM cells. We show that peripheral infections generate antigen-specific CD8+ memory T cells in the brain that adopt a unique TRM signature. Upon depletion of circulating and perivascular memory T cells, this brain signature was enriched and the surveilling properties of brain TRM cells was revealed by intravital imaging. Notably, peripherally induced brain TRM cells showed evidence of rapid activation and enhanced cytokine production and mediated protection after brain infections. These data reveal that peripheral immunizations can generate brain TRM cells and will guide potential use of T cells as therapeutic strategies against CNS infections and neurological diseases.- Published
- 2020
- Full Text
- View/download PDF
13. Therapeutic intervention in relapsing autoimmune demyelinating disease through induction of myelin-specific regulatory CD8 T cell responses.
- Author
-
Brate AA, Boyden AW, Itani FR, Pewe LL, Harty JT, and Karandikar NJ
- Abstract
Multiple Sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). We have shown that CNS-specific CD8 T cells (CNS-CD8) possess a disease suppressive function in MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Previous studies have focused on the role of these cells predominantly in chronic models of disease, but the majority of MS patients present with a relapsing-remitting disease course. In this study, we evaluated the therapeutic role of CD8 T cells in the context of relapsing-remitting disease (RR-EAE), using SJL mice. We found that PLP
178-191 - and MBP84-104 -CD8 ameliorated disease severity in an antigen-specific manner. In contrast, PLP139-151 -CD8 did not suppress disease. PLP178-191 -CD8 were able to reduce the number of relapses even when transferred during ongoing disease. We further ascertained that the suppressive subset of CD8 T cells was contained within the CD25+ CD8 T cell compartment post- in vitro activation with PLP178-191 . Using Listeria monocytogenes (LM) encoding CNS antigens to preferentially prime suppressive CDS T cells in vivo , we show that LM infection induced disease suppressive CD8 T cells that protected and treated PLP178-191 disease. Importantly, a combination of PLP178-191 -CDs transfer boosted by LM-PLP175-194 infection effectively treated ongoing disease induced by a non-cognate peptide (PLP139-151 ), indicating that this approach could be effective even in the context of epitope spreading. These data support a potential immunotherapeutic strategy using CD8 transfer and/or LM vaccination to boost disease regulatory CD8 T cells., Competing Interests: Declarations of interest None.- Published
- 2019
- Full Text
- View/download PDF
14. Protective role for the N-terminal domain of α-dystroglycan in Influenza A virus proliferation.
- Author
-
de Greef JC, Slütter B, Anderson ME, Hamlyn R, O'Campo Landa R, McNutt EJ, Hara Y, Pewe LL, Venzke D, Matsumura K, Saito F, Harty JT, and Campbell KP
- Subjects
- Animals, Basement Membrane drug effects, Basement Membrane virology, Body Fluids drug effects, Body Fluids virology, Cell Line, Glycosylation drug effects, HEK293 Cells, Humans, Inflammation drug therapy, Inflammation virology, Influenza, Human drug therapy, Influenza, Human virology, Lung drug effects, Lung virology, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections drug therapy, Orthomyxoviridae Infections virology, Viral Load methods, Cell Proliferation drug effects, Dystroglycans pharmacology, Influenza A Virus, H1N1 Subtype drug effects, Protective Agents pharmacology
- Abstract
α-Dystroglycan (α-DG) is a highly glycosylated basement membrane receptor that is cleaved by the proprotein convertase furin, which releases its N-terminal domain (α-DGN). Before cleavage, α-DGN interacts with the glycosyltransferase LARGE1 and initiates functional O-glycosylation of the mucin-like domain of α-DG. Notably, α-DGN has been detected in a wide variety of human bodily fluids, but the physiological significance of secreted α-DGN remains unknown. Here, we show that mice lacking α-DGN exhibit significantly higher viral titers in the lungs after Influenza A virus (IAV) infection (strain A/Puerto Rico/8/1934 H1N1), suggesting an inability to control virus load. Consistent with this, overexpression of α-DGN before infection or intranasal treatment with recombinant α-DGN prior and during infection, significantly reduced IAV titers in the lungs of wild-type mice. Hemagglutination inhibition assays using recombinant α-DGN showed in vitro neutralization of IAV. Collectively, our results support a protective role for α-DGN in IAV proliferation., Competing Interests: The authors declare no conflict of interest., (Copyright © 2019 the Author(s). Published by PNAS.)
- Published
- 2019
- Full Text
- View/download PDF
15. Monocyte-Derived CD11c + Cells Acquire Plasmodium from Hepatocytes to Prime CD8 T Cell Immunity to Liver-Stage Malaria.
- Author
-
Kurup SP, Anthony SM, Hancox LS, Vijay R, Pewe LL, Moioffer SJ, Sompallae R, Janse CJ, Khan SM, and Harty JT
- Subjects
- CD11c Antigen analysis, Liver parasitology, Monocytes chemistry, Monocytes immunology, Plasmodium immunology, Antigen Presentation, CD8-Positive T-Lymphocytes immunology, Hepatocytes parasitology, Immunity, Cellular, Liver immunology, Malaria immunology, Monocytes parasitology
- Abstract
Plasmodium sporozoites inoculated by mosquitoes migrate to the liver and infect hepatocytes prior to release of merozoites that initiate symptomatic blood-stage malaria. Plasmodium parasites are thought to be restricted to hepatocytes throughout this obligate liver stage of development, and how liver-stage-expressed antigens prime productive CD8 T cell responses remains unknown. We found that a subset of liver-infiltrating monocyte-derived CD11c
+ cells co-expressing F4/80, CD103, CD207, and CSF1R acquired parasites during the liver stage of malaria, but only after initial hepatocyte infection. These CD11c+ cells found in the infected liver and liver-draining lymph nodes exhibited transcriptionally and phenotypically enhanced antigen-presentation functions and primed protective CD8 T cell responses against Plasmodium liver-stage-restricted antigens. Our findings highlight a previously unrecognized aspect of Plasmodium biology and uncover the fundamental mechanism by which CD8 T cell responses are primed against liver-stage malaria antigens., (Copyright © 2019 Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
16. Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection.
- Author
-
Schmidt ME, Knudson CJ, Hartwig SM, Pewe LL, Meyerholz DK, Langlois RA, Harty JT, and Varga SM
- Subjects
- Animals, Cells, Cultured, Female, Humans, Immune System Diseases pathology, Mice, Mice, Inbred BALB C, Respiratory Syncytial Virus Infections pathology, Respiratory Syncytial Viruses immunology, Severity of Illness Index, CD8-Positive T-Lymphocytes physiology, Immune System Diseases immunology, Immune System Diseases virology, Immunologic Memory, Respiratory Syncytial Virus Infections complications, Respiratory Syncytial Virus Infections immunology
- Abstract
Memory CD8 T cells can provide protection from re-infection by respiratory viruses such as influenza and SARS. However, the relative contribution of memory CD8 T cells in providing protection against respiratory syncytial virus (RSV) infection is currently unclear. To address this knowledge gap, we utilized a prime-boost immunization approach to induce robust memory CD8 T cell responses in the absence of RSV-specific CD4 T cells and antibodies. Unexpectedly, RSV infection of mice with pre-existing CD8 T cell memory led to exacerbated weight loss, pulmonary disease, and lethal immunopathology. The exacerbated disease in immunized mice was not epitope-dependent and occurred despite a significant reduction in RSV viral titers. In addition, the lethal immunopathology was unique to the context of an RSV infection as mice were protected from a normally lethal challenge with a recombinant influenza virus expressing an RSV epitope. Memory CD8 T cells rapidly produced IFN-γ following RSV infection resulting in elevated protein levels in the lung and periphery. Neutralization of IFN-γ in the respiratory tract reduced morbidity and prevented mortality. These results demonstrate that in contrast to other respiratory viruses, RSV-specific memory CD8 T cells can induce lethal immunopathology despite mediating enhanced viral clearance.
- Published
- 2018
- Full Text
- View/download PDF
17. Suppression of autoimmune demyelinating disease by preferential stimulation of CNS-specific CD8 T cells using Listeria-encoded neuroantigen.
- Author
-
Itani FR, Sinha S, Brate AA, Pewe LL, Gibson-Corley KN, Harty JT, and Karandikar NJ
- Subjects
- Animals, Encephalomyelitis, Autoimmune, Experimental pathology, Immunity, Cellular, Mice, Inbred C57BL, Mice, Knockout, Myelin Proteolipid Protein immunology, Organ Specificity, Antigens, Bacterial metabolism, CD8-Positive T-Lymphocytes immunology, Central Nervous System pathology, Encephalomyelitis, Autoimmune, Experimental immunology, Listeria metabolism, Lymphocyte Activation immunology
- Abstract
CD8 T-cells predominate in CNS lesions of MS patients and display oligoclonal expansion. However, the role of myelin-specific CD8 T-cells in disease remains unclear, with studies showing protective and pathogenic roles in EAE. We demonstrated a disease-suppressive function for CNS-specific CD8 T-cells in a model where the antigen is exogenously administered in vivo and used for in vitro activation. To probe the nature of the CD8 response elicited by endogenously presented myelin antigens in vivo, we developed a novel approach utilizing infection with Listeria monocytogenes (LM) encoding proteolipid protein peptide (PLP) amino acids 178-191 (LM-PLP). LM-PLP infection preferentially induced PLP-specific CD8 T-cell responses. Despite the induction of PLP-specific CD8 T-cells, LM-PLP infection did not result in disease. In fact, LM-PLP infection resulted in significant amelioration of PLP178-191-induced EAE. Disease suppression was not observed in mice deficient in CD8 T-cells, IFN-γ or perforin. DTH responses and CNS infiltration were reduced in protected mice, and their CD4 T-cells had reduced capacity to induce tissue inflammation. Importantly, infection with LM-PLP ameliorated established disease. Our studies indicate that CD8 T-cells induced by endogenous presentation of PLP178-191 attenuate CNS autoimmunity in models of EAE, implicating the potential of this approach as a novel immunotherapeutic strategy.
- Published
- 2017
- Full Text
- View/download PDF
18. Correction: Discriminating Protective from Nonprotective Plasmodium-Specific CD8+ T Cell Responses.
- Author
-
Doll KL, Pewe LL, Kurup SP, and Harty JT
- Published
- 2016
- Full Text
- View/download PDF
19. Context-dependent effects of SOCS3 in angiotensin II-induced vascular dysfunction and hypertension in mice: mechanisms and role of bone marrow-derived cells.
- Author
-
Li Y, Kinzenbaw DA, Modrick ML, Pewe LL, and Faraci FM
- Subjects
- Animals, Aorta drug effects, Aorta physiopathology, Basilar Artery drug effects, Basilar Artery physiopathology, Bone Marrow Transplantation, Carotid Arteries drug effects, Carotid Arteries physiopathology, Disease Models, Animal, Dose-Response Relationship, Drug, Female, Genotype, Hypertension chemically induced, Hypertension physiopathology, Hypertension prevention & control, Interleukin-6 metabolism, Male, Mice, Inbred C57BL, Mice, Knockout, NF-kappa B metabolism, Organ Culture Techniques, Phenotype, STAT3 Transcription Factor metabolism, Signal Transduction, Superoxides metabolism, Suppressor of Cytokine Signaling 3 Protein deficiency, Suppressor of Cytokine Signaling 3 Protein genetics, Vasodilator Agents pharmacology, Angiotensin II, Aorta metabolism, Basilar Artery metabolism, Bone Marrow Cells metabolism, Carotid Arteries metabolism, Hypertension metabolism, Suppressor of Cytokine Signaling 3 Protein metabolism, Vasodilation drug effects
- Abstract
Carotid artery disease is a major contributor to stroke and cognitive deficits. Angiotensin II (Ang II) promotes vascular dysfunction and disease through mechanisms that include the IL-6/STAT3 pathway. Here, we investigated the importance of suppressor of cytokine signaling 3 (SOCS3) in models of Ang II-induced vascular dysfunction. We examined direct effects of Ang II on carotid arteries from SOCS3-deficient (SOCS3(+/-)) mice and wild-type (WT) littermates using organ culture and then tested endothelial function with acetylcholine (ACh). A low concentration of Ang II (1 nmol/l) did not affect ACh-induced vasodilation in WT but reduced that of SOCS3(+/-) mice by ∼50% (P < 0.05). In relation to mechanisms, effects of Ang II in SOCS3(+/-) mice were prevented by inhibitors of STAT3, IL-6, NF-κB, or superoxide. Systemic Ang II (1.4 mg/kg per day for 14 days) also reduced vasodilation to ACh in WT. Surprisingly, SOCS3 deficiency prevented most of the endothelial dysfunction. To examine potential underlying mechanisms, we performed bone marrow transplantation. WT mice reconstituted with SOCS3(+/-) bone marrow were protected from Ang II-induced endothelial dysfunction, whereas reconstitution of SOCS3(+/-) mice with WT bone marrow exacerbated Ang II-induced effects. The SOCS3 genotype of bone marrow-derived cells did not influence direct effects of Ang II on vascular function. These data provide new mechanistic insight into the influence of SOCS3 on the vasculature, including divergent effects depending on the source of Ang II. Bone marrow-derived cells deficient in SOCS3 protect against systemic Ang II-induced vascular dysfunction., (Copyright © 2016 the American Physiological Society.)
- Published
- 2016
- Full Text
- View/download PDF
20. Discriminating Protective from Nonprotective Plasmodium-Specific CD8+ T Cell Responses.
- Author
-
Doll KL, Pewe LL, Kurup SP, and Harty JT
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Epitopes, T-Lymphocyte immunology, Female, Hepatocytes immunology, Hepatocytes parasitology, Immunization Schedule, Immunologic Memory, Liver parasitology, Malaria prevention & control, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Protozoan Proteins immunology, Sporozoites immunology, Vaccines, Subunit immunology, Antigens, Protozoan immunology, CD8-Positive T-Lymphocytes immunology, Malaria immunology, Malaria Vaccines immunology, Plasmodium berghei immunology
- Abstract
Despite decades of research, malaria remains a global health crisis. Current subunit vaccine approaches do not provide efficient long-term, sterilizing immunity against Plasmodium infections in humans. Conversely, whole parasite vaccinations with their larger array of target Ags have conferred long-lasting sterilizing protection to humans. Similar studies in rodent models of malaria reveal that CD8(+) T cells play a critical role in liver-stage immunity after whole parasite vaccination. However, it is unknown whether all CD8(+) T cell specificities elicited by whole parasite vaccination contribute to protection, an issue of great relevance for enhanced subunit vaccination. In this article, we show that robust CD8(+) T cell responses of similar phenotype are mounted after prime-boost immunization against Plasmodium berghei glideosome-associated protein 5041-48-, sporozoite-specific protein 20318-325-, thrombospondin-related adhesion protein (TRAP) 130-138-, or circumsporozoite protein (CSP) 252-260-derived epitopes in mice, but only CSP252-260- and TRAP130-138-specific CD8(+) T cells provide sterilizing immunity and reduce liver parasite burden after sporozoite challenge. Further, CD8(+) T cells specific to sporozoite surface-expressed CSP and TRAP proteins, but not intracellular glideosome-associated protein 50 and sporozoite-specific protein 20, efficiently recognize sporozoite-infected hepatocytes in vitro. These results suggest that: 1) protection-relevant antigenic targets, regardless of their immunogenic potential, must be efficiently presented by infected hepatocytes for CD8(+) T cells to eliminate liver-stage Plasmodium infection; and 2) proteins expressed on the surface of sporozoites may be good target Ags for protective CD8(+) T cells., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
21. Paradoxical Increase in Mortality and Rupture of Intracranial Aneurysms in Microsomal Prostaglandin E2 Synthase Type 1-Deficient Mice: Attenuation by Aspirin.
- Author
-
Peña Silva RA, Mitchell IJ, Kung DK, Pewe LL, Granja MF, Harty JT, Faraci FM, Heistad DD, and Hasan DM
- Subjects
- Aneurysm, Ruptured mortality, Animals, Aspirin pharmacology, Humans, Intracranial Aneurysm mortality, Mice, Mice, Inbred C57BL, Mice, Knockout, Microsomes drug effects, Mortality trends, Prostaglandin-E Synthases, Aneurysm, Ruptured drug therapy, Aneurysm, Ruptured enzymology, Aspirin therapeutic use, Intracranial Aneurysm drug therapy, Intracranial Aneurysm enzymology, Intramolecular Oxidoreductases deficiency, Microsomes enzymology
- Abstract
Background: Inflammation plays an important role in formation and rupture of intracranial aneurysms. Expression of microsomal prostaglandin E2 (PGE2) synthase type 1 (mPGES-1) is increased in the wall of intracranial aneurysms in humans. PGE2, a by-product of mPGES-1, is associated with inflammation and cerebrovascular dysfunction., Objective: To test the hypothesis that deletion of mPGES-1 decreases the formation and rupture of intracranial aneurysms in a murine model., Methods: Intracranial aneurysms were induced in wild-type and mPGES-1 knockout (mPGES-1 KO) mice by using a combination of deoxycorticosterone acetate-salt-induced hypertension and intracranial injection of elastase in the basal cistern. Prevalence of aneurysms, subarachnoid hemorrhage, and mortality were assessed. We also tested the effects of administration of aspirin (6 mg/kg/d) by gavage and PGE2 (1 mg/kg/d) by subcutaneous infusion., Results: Systolic blood pressure and prevalence of aneurysm were similar in wild-type and mPGES-1 KO mice. However, mortality and the prevalence of subarachnoid hemorrhage were markedly increased in mPGES-1 KO mice (P < .05). Bone marrow reconstitution studies suggest that mPGES-1 derived from leukocytes does not appear to increase rupture of intracranial aneurysms. Aspirin, but not PGE2, attenuated the increased mortality in mPGES-1 KO mice (P < .05)., Conclusion: Vascular mPGES-1 plays a protective role in blood vessels and attenuates rupture of cerebral aneurysms. In contrast to effects on abdominal aneurysms, mPGES-1 deficiency is associated with an increase in rupture of cerebral aneurysms and mortality, which are attenuated by low-dose aspirin.
- Published
- 2015
- Full Text
- View/download PDF
22. Inflammatory IL-15 is required for optimal memory T cell responses.
- Author
-
Richer MJ, Pewe LL, Hancox LS, Hartwig SM, Varga SM, and Harty JT
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, Cell Division genetics, Humans, Interferon Type I genetics, Interferon Type I immunology, Interleukin-15 genetics, Mechanistic Target of Rapamycin Complex 1, Mice, Mice, Knockout, Multiprotein Complexes genetics, Multiprotein Complexes immunology, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases immunology, CD8-Positive T-Lymphocytes immunology, Cell Division immunology, Immunologic Memory, Interleukin-15 immunology
- Abstract
Due to their ability to rapidly proliferate and produce effector cytokines, memory CD8+ T cells increase protection following reexposure to a pathogen. However, low inflammatory immunizations do not provide memory CD8+ T cells with a proliferation advantage over naive CD8+ T cells, suggesting that cell-extrinsic factors enhance memory CD8+ T cell proliferation in vivo. Herein, we demonstrate that inflammatory signals are critical for the rapid proliferation of memory CD8+ T cells following infection. Using murine models of viral infection and antigen exposure, we found that type I IFN-driven expression of IL-15 in response to viral infection prepares memory CD8+ T cells for rapid division independently of antigen reexposure by transiently inducing cell-cycle progression via a pathway dependent on mTOR complex-1 (mTORC1). Moreover, exposure to IL-15 allowed more rapid division of memory CD8+ T cells following antigen encounter and enhanced their protective capacity against viral infection. Together, these data reveal that inflammatory IL-15 promotes optimal responses by memory CD8+ T cells.
- Published
- 2015
- Full Text
- View/download PDF
23. Aged mice exhibit a severely diminished CD8 T cell response following respiratory syncytial virus infection.
- Author
-
Fulton RB, Weiss KA, Pewe LL, Harty JT, and Varga SM
- Subjects
- Animals, Down-Regulation, Female, Humans, Lung immunology, Lung virology, Lymphocyte Activation, Male, Mice, Mice, Inbred BALB C, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Viruses immunology, Th2 Cells immunology, Aging immunology, CD8-Positive T-Lymphocytes immunology, Respiratory Syncytial Virus Infections immunology, Respiratory Syncytial Viruses physiology
- Abstract
Respiratory virus infections in the elderly result in increased rates of hospitalization and death. Respiratory syncytial virus (RSV) is a leading cause of severe virus-induced respiratory disease in individuals over the age of 65. CD8 T cells play a critical role in mediating RSV clearance. While it is clear that T cell immunity declines with age, it is not clear to what extent the CD8 T cell response to RSV is altered. Using aged BALB/c mice, we demonstrated that RSV-specific CD8 T cell responses were significantly reduced in the lungs of aged mice at the peak of the T cell response and that this decrease correlated with delayed viral clearance. Despite a decrease in the overall numbers of RSV-specific CD8 T cells during acute infection, their capacity to produce effector cytokines was not impaired. Following viral clearance, the RSV-specific memory CD8 T cells were similar in total number and phenotype in young and aged mice. Furthermore, following infection with a heterologous pathogen expressing an RSV epitope, RSV-specific memory CD8 T cells exhibited similar activation and ability to provide early control of the infection in young and aged mice. These data demonstrate a decrease in the capacity of aged mice to induce a high-magnitude acute CD8 T cell response, leading to prolonged viral replication, which may contribute to the increased disease severity of RSV infection observed for aged individuals.
- Published
- 2013
- Full Text
- View/download PDF
24. Lung airway-surveilling CXCR3(hi) memory CD8(+) T cells are critical for protection against influenza A virus.
- Author
-
Slütter B, Pewe LL, Kaech SM, and Harty JT
- Subjects
- Adoptive Transfer, Animals, Bronchoalveolar Lavage Fluid immunology, CD8-Positive T-Lymphocytes chemistry, CD8-Positive T-Lymphocytes transplantation, Chemotaxis, Leukocyte, Dendritic Cells immunology, Immunization, Secondary, Influenza A virus immunology, Interleukin-12 antagonists & inhibitors, Interleukin-12 biosynthesis, Interleukin-12 immunology, Lymph Nodes immunology, Lymphocyte Depletion, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections virology, Receptor, Interferon alpha-beta deficiency, Receptors, CXCR3 biosynthesis, Receptors, CXCR3 deficiency, Receptors, CXCR3 genetics, Respiratory System immunology, STAT4 Transcription Factor physiology, T-Lymphocyte Subsets chemistry, T-Lymphocyte Subsets transplantation, Vaccination, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Influenza A virus pathogenicity, Influenza Vaccines immunology, Lung immunology, Orthomyxoviridae Infections prevention & control, Receptors, CXCR3 physiology, T-Lymphocyte Subsets immunology
- Abstract
Inducing memory CD8(+) T cells specific for conserved antigens from influenza A virus (IAV) is a potential strategy for broadly protective vaccines. Here we show that memory CD8(+) T cells in the airways played an important role in early control of IAV. Expression of the chemokine receptor CXCR3 was critical for memory CD8(+) T cells to populate the airways during the steady state and vaccination approaches were designed to favor the establishment of memory CD8(+) T cells in the airways. Specifically, we found that interleukin-12 (IL-12) signaling shortly after immunization limited CXCR3 expression on memory CD8(+) T cells. Neutralization of IL-12 or adjuvants that did not induce high amounts of IL-12 enhanced CXCR3 expression, sustained airway localization of memory CD8(+) T cells, and resulted in superior protection against IAV., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
25. In vivo CD8+ T cell dynamics in the liver of Plasmodium yoelii immunized and infected mice.
- Author
-
Cabrera M, Pewe LL, Harty JT, and Frevert U
- Subjects
- Adoptive Transfer, Animals, CD8-Positive T-Lymphocytes immunology, Hepatocytes immunology, Hepatocytes parasitology, Malaria prevention & control, Mice, Mice, Inbred BALB C, Sporozoites physiology, CD8-Positive T-Lymphocytes cytology, Immunization, Liver immunology, Liver parasitology, Malaria immunology, Plasmodium yoelii physiology
- Abstract
Plasmodium falciparum malaria remains one of the most serious health problems globally and a protective malaria vaccine is desperately needed. Vaccination with attenuated parasites elicits multiple cellular effector mechanisms that lead to Plasmodium liver stage elimination. While granule-mediated cytotoxicity requires contact between CD8+ effector T cells and infected hepatocytes, cytokine secretion should allow parasite killing over longer distances. To better understand the mechanism of parasite elimination in vivo, we monitored the dynamics of CD8+ T cells in the livers of naïve, immunized and sporozoite-infected mice by intravital microscopy. We found that immunization of BALB/c mice with attenuated P. yoelii 17XNL sporozoites significantly increases the velocity of CD8+ T cells patrolling the hepatic microvasculature from 2.69±0.34 μm/min in naïve mice to 5.74±0.66 μm/min, 9.26±0.92 μm/min, and 7.11±0.73 μm/min in mice immunized with irradiated, early genetically attenuated (Pyuis4-deficient), and late genetically attenuated (Pyfabb/f-deficient) parasites, respectively. Sporozoite infection of immunized mice revealed a 97% and 63% reduction in liver stage density and volume, respectively, compared to naïve controls. To examine cellular mechanisms of immunity in situ, naïve mice were passively immunized with hepatic or splenic CD8+ T cells. Unexpectedly, adoptive transfer rendered the motile CD8+ T cells from immunized mice immotile in the liver of P. yoelii infected mice. Similarly, when mice were simultaneously inoculated with viable sporozoites and CD8+ T cells, velocities 18 h later were also significantly reduced to 0.68±0.10 μm/min, 1.53±0.22 μm/min, and 1.06±0.26 μm/min for CD8+ T cells from mice immunized with irradiated wild type sporozoites, Pyfabb/f-deficient parasites, and P. yoelii CS280-288 peptide, respectively. Because immobilized CD8+ T cells are unable to make contact with infected hepatocytes, soluble mediators could potentially play a key role in parasite elimination under these experimental conditions.
- Published
- 2013
- Full Text
- View/download PDF
26. Cutting edge: rapid boosting of cross-reactive memory CD8 T cells broadens the protective capacity of the Flumist vaccine.
- Author
-
Slütter B, Pewe LL, Lauer P, and Harty JT
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Cross Reactions immunology, Female, Humans, Influenza A virus immunology, Influenza Vaccines immunology, Influenza Vaccines therapeutic use, Influenza, Human immunology, Influenza, Human virology, Mice, Mice, Inbred BALB C, Time Factors, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes virology, Immunologic Memory, Influenza Vaccines administration & dosage, Influenza, Human prevention & control, Lymphocyte Activation immunology
- Abstract
Memory CD8 T cells recognizing conserved proteins from influenza A virus (IAV), such as nucleoprotein, have the potential to provide protection in individuals who lack the proper neutralizing Abs. In this study, we show that the most potent CD8 T cell-inducing influenza vaccine on the market (Flumist) does not induce sufficient numbers of cross-reactive CD8 T cells to provide substantial protection against lethal nonhomologous IAV challenge. However, Flumist-primed CD8 T cells rapidly acquire memory characteristics and can respond to short-interval boosting to greatly enlarge the IAV-specific memory pool, which is sufficient to protect mice from nonhomologous IAV challenge. Thus, a current vaccine strategy, Flumist, may serve as a priming platform for the rapid induction of large numbers of memory CD8 T cells with the capacity for broad protection against influenza.
- Published
- 2013
- Full Text
- View/download PDF
27. Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection.
- Author
-
Butler NS, Moebius J, Pewe LL, Traore B, Doumbo OK, Tygrett LT, Waldschmidt TJ, Crompton PD, and Harty JT
- Subjects
- Acute Disease, Animals, Antigens, CD immunology, B-Lymphocytes drug effects, B-Lymphocytes immunology, B-Lymphocytes parasitology, B7-H1 Antigen immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes parasitology, Child, Child, Preschool, Chronic Disease, Erythrocytes immunology, Erythrocytes parasitology, Female, Germinal Center drug effects, Germinal Center immunology, Germinal Center parasitology, Humans, Malaria, Falciparum immunology, Mali, Mice, Mice, Inbred C57BL, Plasmodium falciparum immunology, United States, Up-Regulation drug effects, Lymphocyte Activation Gene 3 Protein, Antigens, CD drug effects, B7-H1 Antigen antagonists & inhibitors, CD4-Positive T-Lymphocytes drug effects, Malaria, Falciparum drug therapy, Plasmodium falciparum drug effects
- Abstract
Infection of erythrocytes with Plasmodium species induces clinical malaria. Parasite-specific CD4(+) T cells correlate with lower parasite burdens and severity of human malaria and are needed to control blood-stage infection in mice. However, the characteristics of CD4(+) T cells that determine protection or parasite persistence remain unknown. Here we show that infection of humans with Plasmodium falciparum resulted in higher expression of the inhibitory receptor PD-1 associated with T cell dysfunction. In vivo blockade of the PD-1 ligand PD-L1 and the inhibitory receptor LAG-3 restored CD4(+) T cell function, amplified the number of follicular helper T cells and germinal-center B cells and plasmablasts, enhanced protective antibodies and rapidly cleared blood-stage malaria in mice. Thus, chronic malaria drives specific T cell dysfunction, and proper function can be restored by inhibitory therapies to enhance parasite control.
- Published
- 2011
- Full Text
- View/download PDF
28. NFIL3/E4BP4 is a key transcription factor for CD8α⁺ dendritic cell development.
- Author
-
Kashiwada M, Pham NL, Pewe LL, Harty JT, and Rothman PB
- Subjects
- Animals, Basic-Leucine Zipper Transcription Factors biosynthesis, Basic-Leucine Zipper Transcription Factors genetics, Basic-Leucine Zipper Transcription Factors metabolism, CD8 Antigens genetics, CD8 Antigens metabolism, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Differentiation genetics, Dendritic Cells cytology, Dendritic Cells metabolism, Gene Expression Regulation genetics, Gene Expression Regulation immunology, Interleukin-12 genetics, Interleukin-12 immunology, Interleukin-12 metabolism, Membrane Proteins genetics, Membrane Proteins immunology, Membrane Proteins metabolism, Mice, Mice, Knockout, Plasma Cells cytology, Plasma Cells metabolism, Repressor Proteins biosynthesis, Repressor Proteins genetics, Repressor Proteins immunology, Toll-Like Receptor 3 genetics, Toll-Like Receptor 3 immunology, Toll-Like Receptor 3 metabolism, Basic-Leucine Zipper Transcription Factors immunology, CD8 Antigens immunology, Cell Differentiation immunology, Dendritic Cells immunology, Plasma Cells immunology
- Abstract
Antigen presentation by mature dendritic cells (DCs) is the first step for initiating adaptive immune responses. DCs are composed of heterogeneous functional subsets; however, the molecular mechanisms that regulate differentiation of specific DC subsets are not understood. Here, we report that the basic leucine zipper transcription factor NFIL3/E4BP4 is essential for the development of CD8α(+) conventional DCs (cDCs). Nfil3(-/-) mice specifically lack CD8α(+) cDCs but not CD8α(-) cDCs or plasmacytoid DCs in lymphoid tissues. Flt3 ligand-dependent generation of CD8α(+) cDCs in lymphoid tissues and CD8α(+)-equivalent cDCs from Nfil3(-/-) bone marrow cells was also impaired. NFIL3 regulates CD8α(+) cDC development in part through Batf3 expression. Importantly, Nfil3(-/-) mice exhibited impaired cross-priming of CD8(+) T cells against cell-associated antigen, a process normally performed by CD8α(+) cDCs, and failed to produce IL-12 after TLR3 stimulation. Thus, NFIL3 plays an essential role in the development of CD8α(+) cDCs.
- Published
- 2011
- Full Text
- View/download PDF
29. Exploiting cross-priming to generate protective CD8 T-cell immunity rapidly.
- Author
-
Pham NL, Pewe LL, Fleenor CJ, Langlois RA, Legge KL, Badovinac VP, and Harty JT
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes metabolism, Dendritic Cells immunology, Immunization methods, Immunization, Secondary, Leukocytes, Mononuclear immunology, Listeria monocytogenes immunology, Listeriosis immunology, Listeriosis microbiology, Malaria immunology, Malaria parasitology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Models, Immunological, Ovalbumin immunology, Plasmodium berghei immunology, Time Factors, CD8-Positive T-Lymphocytes immunology, Cross-Priming immunology, Immunity immunology, Immunologic Memory immunology
- Abstract
The number of memory CD8 T cells generated by infection or vaccination correlates strongly with the degree of protection observed in infection and tumor models. Therefore, rapid induction of protective numbers of effector and memory CD8 T cells may be crucial in the case of malignancy, pandemic infection, or bioterrorism. Many studies have shown that amplifying T-cell numbers by prime-boost vaccination is most effective with a substantial time interval between immunizations. In contrast, immunization with peptide-coated mature dendritic cells (DCs) results in a CD8 T-cell response exhibiting accelerated acquisition of memory characteristics, including the ability to respond to booster immunization within days of initial priming. However, personalized DC immunization is too costly, labor intensive, and time-consuming for large-scale vaccination. Here, we demonstrate that in vivo cross-priming with cell-associated antigens or antigen-coated, biodegradable microspheres in the absence of adjuvant quickly generates CD8 T cells that display the phenotype and function of long-term memory populations. Importantly, cross-primed CD8 T cells can respond to booster immunization within days of the initial immunization to generate rapidly large numbers of effector and memory T cells that can protect against bacterial, viral, and parasitic infections, including lethal influenza and malaria-causing Plasmodium infection. Thus, accelerated CD8 T-cell memory after in vivo cross-priming in the absence of adjuvant is generalizable and can be exploited to generate protective immunity rapidly.
- Published
- 2010
- Full Text
- View/download PDF
30. Very diverse CD8 T cell clonotypic responses after virus infections.
- Author
-
Pewe LL, Netland JM, Heard SB, and Perlman S
- Subjects
- Animals, CD8-Positive T-Lymphocytes virology, Clone Cells, Coronavirus Infections metabolism, Epitopes, T-Lymphocyte immunology, Gene Rearrangement, beta-Chain T-Cell Antigen Receptor, Lymphocyte Activation, Lymphocyte Count methods, Lymphocyte Count statistics & numerical data, Mice, Mice, Inbred C57BL, Peptide Fragments immunology, Poisson Distribution, Receptors, Antigen, T-Cell, alpha-beta genetics, Spleen cytology, Spleen immunology, Spleen metabolism, Viral Proteins immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Coronavirus Infections immunology, Murine hepatitis virus immunology, Receptors, Antigen, T-Cell, alpha-beta biosynthesis
- Abstract
We measured CD8 T cell clonotypic diversity to three epitopes recognized in C57BL/6 mice infected with mouse hepatitis virus, strain JHM, or lymphocytic choriomeningitis virus. We isolated epitope-specific T cells with an IFN-gamma capture assay or MHC class I/peptide tetramers and identified different clonotypes by Vbeta chain sequence analysis. In agreement with our previous results, the number of different clonotypes responding to all three epitopes fit a log-series distribution. From these distributions, we estimated that >1000 different clonotypes responded to each immunodominant CD8 T cell epitope; the response to a subdominant CD8 T cell epitope was modestly less diverse. These results suggest that T cell response diversity is greater by 1-2 orders of magnitude than predicted previously.
- Published
- 2004
- Full Text
- View/download PDF
31. Bystander CD8 T cell-mediated demyelination after viral infection of the central nervous system.
- Author
-
Haring JS, Pewe LL, and Perlman S
- Subjects
- Animals, Bystander Effect, Chemokines biosynthesis, Coronavirus Infections complications, Cytokines biosynthesis, Demyelinating Autoimmune Diseases, CNS immunology, Homeodomain Proteins physiology, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Multiple Sclerosis etiology, Murine hepatitis virus, CD8-Positive T-Lymphocytes immunology, Central Nervous System Viral Diseases complications, Demyelinating Autoimmune Diseases, CNS etiology
- Abstract
Multiple sclerosis, a chronic inflammatory disease of the CNS, is characterized by immune-mediated demyelination. Many patients have a remitting-relapsing course of disease with exacerbations often following unrelated microbial illnesses. The relationship between the two events remains obscure. One possibility is that T cells specific for the inciting microbial pathogen are able to effect demyelination at a site of ongoing inflammation within the CNS. This possibility was examined in mice infected with mouse hepatitis virus, a well-described model of virus-induced demyelination. Using transgenic TCR/recombination activation gene 2(-/-) mice with only non-mouse hepatitis virus-specific T cells, we show that CD8 T cells are able to cause demyelination in the absence of cognate Ag in the CNS, but only if specifically activated. These findings demonstrate a novel mechanism for immune-mediated neuropathology and show that activated CD8 T cells may serve as important mediators of bystander demyelination during times of infection, including in patients with multiple sclerosis.
- Published
- 2002
- Full Text
- View/download PDF
32. High-magnitude, virus-specific CD4 T-cell response in the central nervous system of coronavirus-infected mice.
- Author
-
Haring JS, Pewe LL, and Perlman S
- Subjects
- Animals, Central Nervous System Viral Diseases virology, Coronavirus Infections virology, Mice, Receptors, Antigen, T-Cell, alpha-beta metabolism, CD4-Positive T-Lymphocytes immunology, Central Nervous System Viral Diseases immunology, Coronavirus Infections immunology, Murine hepatitis virus immunology
- Abstract
The neurotropic JHM strain of mouse hepatitis virus (MHV) causes acute encephalitis and chronic demyelinating encephalomyelitis in rodents. Previous results indicated that CD8 T cells infiltrating the central nervous system (CNS) were largely antigen specific in both diseases. Herein we show that by 7 days postinoculation, nearly 30% of the CD4 T cells in the acutely infected CNS were MHV specific by using intracellular gamma interferon (IFN-gamma) staining assays. In mice with chronic demyelination, 10 to 15% of the CD4 T cells secreted IFN-gamma in response to MHV-specific peptides. Thus, these results show that infection of the CNS is characterized by a large influx of CD4 T cells specific for MHV and that these cells remain functional, as measured by cytokine secretion, in mice with chronic demyelination.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.