Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Exp Med ; 221(3)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38363548

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.


Subject(s)
Influenza A virus , Orthomyxoviridae Infections , Radiation Exposure , Mice , Animals , CD8-Positive T-Lymphocytes , Memory T Cells , Lung , Immunologic Memory
2.
Brain ; 147(2): 566-589, 2024 02 01.
Article in English | MEDLINE | ID: mdl-37776513

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.


Subject(s)
Malaria, Cerebral , Mice , Humans , Animals , Malaria, Cerebral/pathology , Malaria, Cerebral/prevention & control , Endothelial Cells/pathology , Brain/pathology , Blood-Brain Barrier/pathology , CD8-Positive T-Lymphocytes , Endothelium/pathology , Mice, Inbred C57BL , Disease Models, Animal
3.
Proc Natl Acad Sci U S A ; 120(27): e2302785120, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37364124

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 (TN) 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.


Subject(s)
Lymphocytic Choriomeningitis , Whole-Body Irradiation , Mice , Animals , Neoplasm Recurrence, Local , CD8-Positive T-Lymphocytes , Lymphocytic choriomeningitis virus , Immunologic Memory , Mice, Inbred C57BL
4.
J Immunol ; 210(9): 1305-1313, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36939394

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.


Subject(s)
Malaria , Plasmodium chabaudi , Mice , Animals , CD4-Positive T-Lymphocytes , Mice, Inbred C57BL , Interferon-gamma
5.
Proc Natl Acad Sci U S A ; 120(2): e2210181120, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36595704

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.


Subject(s)
Malaria , Parasites , Plasmodium , Animals , Humans , Hepatocytes/metabolism , Liver , Malaria/parasitology , Caspases/metabolism , DNA-Binding Proteins/metabolism
6.
Cell Rep ; 37(5): 109956, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34731605

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Liver/immunology , Malaria/immunology , Plasmodium berghei/immunology , 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
7.
J Immunol ; 207(11): 2631-2635, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34716185

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.


Subject(s)
Liver Diseases/immunology , Malaria Vaccines/immunology , Malaria/immunology , Sporozoites/immunology , Vaccines, Attenuated/immunology , Vaccines, Subunit/immunology , Animals , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Vaccination
8.
J Immunol ; 205(8): 2222-2230, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32887747

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems , Cell Proliferation/genetics , Immunologic Memory/genetics , Tumor Suppressor Protein p53 , Animals , Antigens/immunology , DNA Damage/genetics , DNA Damage/immunology , Mice , Mice, Transgenic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
9.
Nat Immunol ; 21(8): 938-949, 2020 08.
Article in English | MEDLINE | ID: mdl-32572242

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 (TRM) 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.


Subject(s)
Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System Infections/immunology , Immunologic Memory/immunology , Animals , Bacterial Infections/immunology , Brain/cytology , Lymphocyte Activation/immunology , Mice , Virus Diseases/immunology
10.
Proc Natl Acad Sci U S A ; 116(23): 11396-11401, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31097590

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.


Subject(s)
Cell Proliferation/drug effects , Dystroglycans/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Protective Agents/pharmacology , 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
11.
Cell Host Microbe ; 25(4): 565-577.e6, 2019 Apr 10.
Article in English | MEDLINE | ID: mdl-30905437

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.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Hepatocytes/parasitology , Immunity, Cellular , Liver/immunology , Malaria/immunology , Monocytes/parasitology , CD11c Antigen/analysis , Liver/parasitology , Monocytes/chemistry , Monocytes/immunology , Plasmodium/immunology
12.
Article in English | MEDLINE | ID: mdl-32161909

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 PLP178-191- 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.

13.
PLoS Pathog ; 14(1): e1006810, 2018 01.
Article in English | MEDLINE | ID: mdl-29293660

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.


Subject(s)
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 , 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
14.
Immunity ; 47(5): 835-847.e4, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29150238

ABSTRACT

Immune response (Ir) genes, originally proposed by Baruj Benacerraf to explain differential antigen-specific responses in animal models, have become synonymous with the major histocompatibility complex (MHC). We discovered a non-MHC-linked Ir gene in a T cell receptor (TCR) locus that was required for CD8+ T cell responses to the Plasmodium berghei GAP5040-48 epitope in mice expressing the MHC class I allele H-2Db. GAP5040-48-specific CD8+ T cell responses emerged from a very large pool of naive Vß8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vß8.1+ TCR-H-2Db-GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1ß residues present exclusively in the Vß8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vß8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope.


Subject(s)
Histocompatibility Antigen H-2D/genetics , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Receptors, Antigen, T-Cell/genetics , Animals , CD8-Positive T-Lymphocytes/immunology , Complementarity Determining Regions , Epitopes , Genes, T-Cell Receptor beta , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Peptide Fragments/immunology
15.
Sci Rep ; 7(1): 1519, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28484224

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.


Subject(s)
Antigens, Bacterial/metabolism , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Listeria/metabolism , Lymphocyte Activation/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunity, Cellular , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteolipid Protein/immunology , Organ Specificity
17.
Am J Physiol Heart Circ Physiol ; 311(1): H146-56, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27106041

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.


Subject(s)
Angiotensin II , Aorta/metabolism , Basilar Artery/metabolism , Bone Marrow Cells/metabolism , Carotid Arteries/metabolism , Hypertension/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vasodilation , 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 , Vasodilation/drug effects , Vasodilator Agents/pharmacology
18.
J Immunol ; 196(10): 4253-62, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27084099

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.


Subject(s)
Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium berghei/immunology , 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
19.
J Clin Invest ; 125(9): 3477-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26241055

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Division/immunology , Immunologic Memory , Interleukin-15/immunology , 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
20.
Neurosurgery ; 77(4): 613-20, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26134597

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.


Subject(s)
Aneurysm, Ruptured/drug therapy , Aneurysm, Ruptured/enzymology , Aspirin/therapeutic use , Intracranial Aneurysm/drug therapy , Intracranial Aneurysm/enzymology , Intramolecular Oxidoreductases/deficiency , Microsomes/enzymology , Aneurysm, Ruptured/mortality , Animals , Aspirin/pharmacology , Humans , Intracranial Aneurysm/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Microsomes/drug effects , Mortality/trends , Prostaglandin-E Synthases
SELECTION OF CITATIONS
SEARCH DETAIL
...