219 results on '"Harty JT"'
Search Results
2. Type-I IFNs induce GBPs and lysosomal defense in hepatocytes to control malaria.
- Author
-
Marques-da-Silva C, Schmidt-Silva C, Bowers C, Charles-Chess E, Shiau JC, Park ES, Yuan Z, Kim BH, Kyle DE, Harty JT, MacMicking JD, and Kurup SP
- Abstract
Plasmodium parasites undergo development and replication within the hepatocytes before infecting the erythrocytes and initiating clinical malaria. Although type-I interferons (IFNs) are known to hinder Plasmodium infection within the liver, the underlying mechanisms remain unclear. Here, we describe two IFN-I-driven hepatocyte antimicrobial programs controlling liver-stage malaria. First, oxidative defense by NADPH oxidases 2 and 4 triggers a pathway of lysosomal fusion with the parasitophorous vacuole (PV) to help clear Plasmodium . Second, guanylate-binding protein (GBP) 1 disruption of the PV activates caspase-1 inflammasome, inducing pyroptosis to remove the infected host cells. Remarkably, both human and mouse hepatocytes enlist these cell-autonomous immune programs to eliminate Plasmodium ; their pharmacologic or genetic inhibition led to profound malarial susceptibility, and are essential in vivo . In addition to identifying the IFN-I-mediated cell-autonomous immune circuits controlling Plasmodium infection in the hepatocytes, this study extends our understanding of how non-immune cells are integral to protective immunity against malaria.
- Published
- 2024
- Full Text
- View/download PDF
3. 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
4. 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
5. 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
6. Defining Parameters That Modulate Susceptibility and Protection to Respiratory Murine Coronavirus MHV1 Infection.
- Author
-
Silva EE, Moioffer SJ, Hassert M, Berton RR, Smith MG, van de Wall S, Meyerholz DK, Griffith TS, Harty JT, and Badovinac VP
- Subjects
- Humans, Mice, Animals, Mice, Inbred C57BL, Mice, Inbred C3H, Mice, Inbred Strains, Genetic Predisposition to Disease, Murine hepatitis virus, Sepsis
- Abstract
Patients infected with SARS-CoV-2 experience variable disease susceptibility, and patients with comorbidities such as sepsis are often hospitalized for COVID-19 complications. However, the extent to which initial infectious inoculum dose determines disease outcomes and whether this can be used for immunological priming in a genetically susceptible host has not been completely defined. We used an established SARS-like murine model in which responses to primary and/or secondary challenges with murine hepatitis virus type 1 (MHV-1) were analyzed. We compared the response to infection in genetically susceptible C3H/HeJ mice, genetically resistant C57BL/6J mice, and genetically diverse, variably susceptible outbred Swiss Webster mice. Although defined as genetically susceptible to MHV-1, C3H/HeJ mice displayed decreasing dose-dependent pathological changes in disease severity and lung infiltrate/edema, as well as lymphopenia. Importantly, an asymptomatic dose (500 PFU) was identified that yielded no measurable morbidity/mortality postinfection in C3H/HeJ mice. Polymicrobial sepsis induced via cecal ligation and puncture converted asymptomatic infections in C3H/HeJ and C57BL/6J mice to more pronounced disease, modeling the impact of sepsis as a comorbidity to β-coronavirus infection. We then used low-dose infection as an immunological priming event in C3H/HeJ mice, which provided neutralizing Ab-dependent, but not circulating CD4/CD8 T cell-dependent, protection against a high-dose MHV-1 early rechallenge. Together, these data define how infection dose, immunological status, and comorbidities modulate outcomes of primary and secondary β-coronavirus infections in hosts with variable susceptibility., (Copyright © 2024 by The American Association of Immunologists, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
7. 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
8. Sepsis leads to lasting changes in phenotype and function of naïve CD8 T cells.
- Author
-
Berton RR, McGonagil PW, Jensen IJ, Ybarra TK, Bishop GA, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Humans, Mice, Animals, CD8-Positive T-Lymphocytes, Immunity, Innate, Phenotype, Mice, Inbred C57BL, Immunologic Memory, Cytokine Release Syndrome, Sepsis
- Abstract
Sepsis, an amplified immune response to systemic infection, is characterized by a transient cytokine storm followed by chronic immune dysfunction. Consequently, sepsis survivors are highly susceptible to newly introduced infections, suggesting sepsis can influence the function and composition of the naïve CD8 T cell pool and resulting pathogen-induced primary CD8 T cell responses. Here, we explored the extent to which sepsis induces phenotypic and functional changes within the naïve CD8 T cell pool. To interrogate this, the cecal ligation and puncture (CLP) mouse model of polymicrobial sepsis was used. In normal, non-septic mice, we show type-I interferon (IFN I)-mediated signaling plays an important role in driving the phenotypic and functional heterogeneity in the naïve CD8 T cell compartment leading to increased representation of Ly6C+ naïve CD8 T cells. In response to viral infection after sepsis resolution, naïve Ly6C+ CD8 T cells generated more primary effector and memory CD8 T cells with slower conversion to a central memory CD8 T cell phenotype (Tcm) than Ly6C- naïve CD8 T cells. Importantly, as a potent inducer of cytokine storm and IFN I production, sepsis leads to increased representation of Ly6C+ naïve CD8 T cells that maintained their heightened ability to respond (i.e., effector and memory CD8 T cell accumulation and cytokine production) to primary LCMV infection. Lastly, longitudinal analyses of peripheral blood samples obtained from septic patients revealed profound changes in CD8 T cell subset composition and frequency compared to healthy controls. Thus, sepsis has the capacity to alter the composition of naïve CD8 T cells, directly influencing primary CD8 T cell responses to newly introduced infections., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Berton et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
9. Alt-RNAtive vaccines elicit anti-malarial T RM cells.
- Author
-
Hassert M and Harty JT
- Subjects
- CD8-Positive T-Lymphocytes, Immunologic Memory, Antimalarials, Vaccines
- Published
- 2023
- Full Text
- View/download PDF
10. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury.
- Author
-
Todd BP, Luo Z, Gilkes N, Chimenti MS, Peterson Z, Mix MR, Harty JT, Nickl-Jockschat T, Ferguson PJ, Bassuk AG, and Newell EA
- Subjects
- Male, Animals, Mice, Neuropathology, Brain, Antibodies, Interferon Type I, Brain Injuries, Traumatic complications
- Abstract
Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/β receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Molecular alteration of reactive microglia also occurred with diminished expression of genes needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
11. 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
12. Memory CD8+ T cell-mediated protection against liver-stage malaria.
- Author
-
Hassert M, Arumugam S, and Harty JT
- Subjects
- Mice, Humans, Animals, Immunologic Memory, Liver, CD8-Positive T-Lymphocytes, Malaria Vaccines, Malaria, Plasmodium
- Abstract
Nearly half of the world's population is at risk of malaria, a disease caused by the protozoan parasite Plasmodium, which is estimated to cause more than 240,000,000 infections and kill more than 600,000 people annually. The emergence of Plasmodia resistant to chemoprophylactic treatment highlights the urgency to develop more effective vaccines. In this regard, whole sporozoite vaccination approaches in murine models and human challenge studies have provided substantial insight into the immune correlates of protection from malaria. From these studies, CD8+ T cells have come to the forefront, being identified as critical for vaccine-mediated liver-stage immunity that can prevent the establishment of the symptomatic blood stages and subsequent transmission of infection. However, the unique biological characteristics required for CD8+ T cell protection from liver-stage malaria dictate that more work must be done to design effective vaccines. In this review, we will highlight a subset of studies that reveal basic aspects of memory CD8+ T cell-mediated protection from liver-stage malaria infection., (© 2023 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
13. 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
14. Cutting Edge: Influenza-Induced CD11alo Airway CD103+ Tissue Resident Memory T Cells Exhibit Compromised IFN-γ Production after In Vivo TCR Stimulation.
- Author
-
van de Wall S, Crooks S, Varga SM, Badovinac VP, and Harty JT
- Subjects
- Humans, CD8-Positive T-Lymphocytes, Memory T Cells, Immunologic Memory, Lung, Interferon-gamma, Receptors, Antigen, T-Cell metabolism, Influenza, Human
- Abstract
Although tissue resident memory T cells (TRM) in the lung confer robust protection against secondary influenza infection, their in vivo production of IFN-γ is unknown. In this study, using a mouse model, we evaluated production of IFN-γ by influenza-induced TRM (defined as CD103+) that localize to the airways or lung parenchyma. Airway TRM consist of both CD11ahi and CD11alo populations, with low CD11a expression signifying prolonged airway residence. In vitro, high-dose peptide stimulation evoked IFN-γ from most CD11ahi airway and parenchymal TRM, whereas most CD11alo airway TRM did not produce IFN-γ. In vivo production of IFN-γ was clearly detectable in CD11ahi airway and parenchymal TRM but essentially absent in CD11alo airway TRM, irrespective of airway-instilled peptide concentration or influenza reinfection. The majority of IFN-γ-producing airway TRM in vivo were CD11ahi, suggesting recent airway entry. These results question the contribution of long-term CD11alo airway TRM to influenza immunity and reinforce the importance of defining TRM tissue compartment-specific contributions to protective immunity., (Copyright © 2023 by The American Association of Immunologists, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
15. 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
16. Cryopreservation of Plasmodium Sporozoites.
- Author
-
Bowers C, Hancox L, Peissig K, Shiau JC, Vantaux A, Witkowski B, Phal S, Maher SP, Harty JT, Kyle DE, and Kurup SP
- Abstract
Malaria is a deadly disease caused by the parasite, Plasmodium , and impacts the lives of millions of people around the world. Following inoculation into mammalian hosts by infected mosquitoes, the sporozoite stage of Plasmodium undergoes obligate development in the liver before infecting erythrocytes and causing clinical malaria. The most promising vaccine candidates for malaria rely on the use of attenuated live sporozoites to induce protective immune responses. The scope of widespread testing or clinical use of such vaccines is limited by the absence of efficient, reliable, or transparent strategies for the long-term preservation of live sporozoites. Here we outline a method to cryopreserve the sporozoites of various human and murine Plasmodium species. We found that the structural integrity, viability, and in vivo or in vitro infectiousness were conserved in the recovered cryopreserved sporozoites. Cryopreservation using our approach also retained the transgenic properties of sporozoites and immunization with cryopreserved radiation attenuated sporozoites (RAS) elicited strong immune responses. Our work offers a reliable protocol for the long-term storage and recovery of human and murine Plasmodium sporozoites and lays the groundwork for the widespread use of live sporozoites for research and clinical applications.
- Published
- 2022
- Full Text
- View/download PDF
17. Keeping T cell memories in mind.
- Author
-
Mix MR and Harty JT
- Subjects
- Animals, Antigens, Homeostasis, Mammals, Immunologic Memory, CD8-Positive T-Lymphocytes
- Abstract
The mammalian central nervous system (CNS) contains a vibrant community of resident adaptive immune cells at homeostasis. Among these are memory CD8
+ and CD4+ T cells, which reside in the CNS in the settings of health, aging, and neurological disease. These T cells commonly exhibit a tissue-resident memory (TRM ) phenotype, suggesting that they are antigen-experienced and remain separate from the circulation. Despite these characterizations, T cell surveillance of the CNS has only recently been studied through the lens of TRM immunology. In this Review, we outline emerging concepts of CNS TRM generation, localization, maintenance, function, and specificity. In this way, we hope to highlight roles of CNS TRM in health and disease to inform future studies of adaptive neuroimmunity., Competing Interests: Declaration of interests The authors declare no competing financial interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
18. Influenza-Induced CD103 + T Resident Memory Cells Exhibit Enhanced Functional Avidity over CD103 - Memory T Cells in the Mediastinal Lymph Node.
- Author
-
Crooks SD, Varga SM, and Harty JT
- Subjects
- Animals, Cytokines, Humans, Lymph Nodes, Memory T Cells, Mice, Peptides, Receptors, Antigen, T-Cell, Viral Proteins, Influenza, Human
- Abstract
Influenza virus-specific tissue-resident memory CD8 T cells (Trms) targeting conserved viral proteins provide strain-transcending heterosubtypic immunity to infection. Trms in the lung combat reinfection through rapid cytolytic function and production of inflammatory cytokines to recruit other immune cells. Influenza-specific Trms are also generated in the lung draining mediastinal lymph node (mLN) and can provide immunity to heterologous virus infection in this tissue, although their role in combating influenza infection is less well defined. Functional avidity, a measure of T cell sensitivity to Ag stimulation, correlates with control of viral infection and may be important for immune detection of recently infected cells, when low numbers of surface peptide-MHC complexes are displayed. However, the functional avidity of influenza-specific Trms has not been previously compared with that of other memory CD8 T cell subsets. In this article, a methodology is presented to compare the functional avidity of CD8 T cell subsets across murine tissues, with a focus on influenza-specific mLNs compared with splenic CD8 T cells, by stimulating both populations in the same well to account for CD8 T cell-extrinsic variables. The functional avidity of influenza-specific mLN effector CD8 T cells is slightly increased relative to splenic effector CD8 T cells. However, CD103
+ mLN Trms display increased functional avidity compared with splenic memory CD8 T cells and CD103- memory CD8 T cells within the mLN. In contrast, lung-derived CD103+ Trms did not exhibit enhanced functional avidity. mLN CD103+ Trms also exhibit increased TCR expression, providing a potential mechanism for their enhanced functional avidity., (Copyright © 2022 The Authors.)- Published
- 2022
- Full Text
- View/download PDF
19. Tissue resident memory T cells- A new benchmark for the induction of vaccine-induced mucosal immunity.
- Author
-
Hassert M and Harty JT
- Subjects
- Animals, Humans, Immunologic Memory, Benchmarking, Memory T Cells, Antibodies, Neutralizing, Immunity, Mucosal, Influenza Vaccines
- Abstract
Historically, the gold-standard benchmark for vaccine immunogenicity has been the induction of neutralizing antibodies detectable in the serum of peripheral blood. However, in recent years there has been a new appreciation for the mucosa as an important site for vaccine induced immunity. As a point of first contact, the mucosal tissue represents a major site of immune based detection and restriction of pathogen entry and dissemination. Tissue resident memory T cells (T
rm ) are one of the critical cell types involved in this early detection and restriction of mucosal pathogens. Following tissue-specific infection or vaccination, Trm lodge themselves within tissues and can perform rapid sensing and alarm functions to control local re-infections, in an effort that has been defined as important for restriction of a number of respiratory pathogens including influenza and respiratory syncytial virus. Despite this characterized importance, only minor attention has been paid to the importance of Trm as a benchmark for vaccine immunogenicity. The purpose of this review is to highlight the functions of Trm with particular emphasis on respiratory infections, and to suggest the inclusion of Trm elicitation as a benchmark for vaccine immunogenicity in animal models, and where possible, human samples., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Hassert and Harty.)- Published
- 2022
- Full Text
- View/download PDF
20. Inflammation Controls Susceptibility of Immune-Experienced Mice to Sepsis.
- Author
-
Berton RR, Jensen IJ, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Animals, Cecum surgery, Disease Models, Animal, Humans, Inflammation, Ligation, Mice, Sepsis
- Abstract
Sepsis, an amplified immune response to systemic infection that leads to life-threatening organ dysfunction, affects >125,000 people/day worldwide with 20% mortality. Modest therapeutic progress for sepsis has been made, in part because of the lack of therapeutic translatability between mouse-based experimental models and humans. One potential reason for this difference stems from the extensive use of immunologically naive specific pathogen-free mice in preclinical research. To address this issue, we used sequential infections with well-defined BSL-2 pathogens to establish a novel immune-experienced mouse model (specific pathogen experienced [SPexp]) to determine the extent to which immunological experience and/or inflammation influences the host capacity to respond to subsequent infections, including sepsis. Consistent with their immunological experience, SPexp inbred or outbred mice had significant changes in the composition and activation status of multiple leukocyte populations known to influence the severity of cecal ligation and puncture-induced sepsis. Importantly, by varying the timing of sepsis induction, we found the level of basal inflammation controls sepsis-induced morbidity and mortality in SPexp mice. In addition, although a beneficial role of NK cells in sepsis was recently demonstrated in specific pathogen-free mice, NK cell depletion before cecal ligation and puncture induction in SPexp mice lead to diminished mortality, suggesting NK cells may have beneficial or detrimental roles in the response to septic insult dependent on host immune status. Thus, data highlight the importance of utilizing immune-experienced models for preclinical studies to interrogate the cellular/molecular mechanism(s) that could be therapeutically exploited during severe and dysregulated infection-induced inflammatory responses, such as sepsis., (Copyright © 2022 The Authors.)
- Published
- 2022
- Full Text
- View/download PDF
21. 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
22. 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
23. Severity of Sepsis Determines the Degree of Impairment Observed in Circulatory and Tissue-Resident Memory CD8 T Cell Populations.
- Author
-
Moioffer SJ, Danahy DB, van de Wall S, Jensen IJ, Sjaastad FV, Anthony SM, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Animals, Blood Circulation, Cells, Cultured, Disease Progression, Humans, Immunologic Memory, Mice, Mice, Inbred C57BL, Mice, Transgenic, Organ Specificity, CD8-Positive T-Lymphocytes immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus physiology, Sepsis immunology, T-Lymphocyte Subsets immunology
- Abstract
Sepsis reduces the number and function of memory CD8 T cells within the host, contributing to the long-lasting state of immunoparalysis. Interestingly, the relative susceptibility of memory CD8 T cell subsets to quantitative/qualitative changes differ after cecal ligation and puncture (CLP)-induced sepsis. Compared with circulatory memory CD8 T cells (T
CIRCM ), moderate sepsis (0-10% mortality) does not result in numerical decline of CD8 tissue-resident memory T cells (TRM ), which retain their "sensing and alarm" IFN-γ-mediated effector function. To interrogate this biologically important dichotomy, vaccinia virus-immune C57BL/6 (B6) mice containing CD8 TCIRCM and skin TRM underwent moderate or severe (∼50% mortality) sepsis. Severe sepsis led to increased morbidity and mortality characterized by increased inflammation compared with moderate CLP or sham controls. Severe CLP mice also displayed increased vascular permeability in the ears. Interestingly, skin CD103+ CD8 TRM , detected by i.v. exclusion or two-photon microscopy, underwent apoptosis and subsequent numerical loss following severe sepsis, which was not observed in mice that experienced moderate CLP or sham surgeries. Consequently, severe septic mice showed diminished CD8 T cell-mediated protection to localized skin reinfection. Finally, the relationship between severity of sepsis and demise in circulatory versus tissue-embedded memory CD8 T cell populations was confirmed by examining tumor-infiltrating and nonspecific CD8 T cells in B16 melanoma tumors. Thus, sepsis can differentially affect the presence and function of Ag-specific CD8 T cells that reside inside tissues/tumors depending on the severity of the insult, a notion with direct relevance to sepsis survivors and their ability to mount protective memory CD8 T cell-dependent responses to localized Ag re-encounter., (Copyright © 2021 by The American Association of Immunologists, Inc.)- Published
- 2021
- Full Text
- View/download PDF
24. Protective function and durability of mouse lymph node-resident memory CD8 + T cells.
- Author
-
Anthony SM, Van Braeckel-Budimir N, Moioffer SJ, van de Wall S, Shan Q, Vijay R, Sompallae R, Hartwig SM, Jensen IJ, Varga SM, Butler NS, Xue HH, Badovinac VP, and Harty JT
- Subjects
- Animals, Antigens, CD genetics, Antigens, CD immunology, Antigens, CD metabolism, Cells, Cultured, Female, Influenza A virus immunology, Lung cytology, Lung immunology, Mice, Mice, Inbred C57BL, Orthomyxoviridae Infections immunology, Transcriptome genetics, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Lymph Nodes cytology, Lymph Nodes immunology
- Abstract
Protective lung tissue-resident memory CD8
+ T cells (Trm) form after influenza A virus (IAV) infection. We show that IAV infection of mice generates CD69+ CD103+ and other memory CD8+ T cell populations in lung-draining mediastinal lymph nodes (mLNs) from circulating naive or memory CD8+ T cells. Repeated antigen exposure, mimicking seasonal IAV infections, generates quaternary memory (4M) CD8+ T cells that protect mLN from viral infection better than 1M CD8+ T cells. Better protection by 4M CD8+ T cells associates with enhanced granzyme A/B expression and stable maintenance of mLN CD69+ CD103+ 4M CD8+ T cells, vs the steady decline of CD69+ CD103+ 1M CD8+ T cells, paralleling the durability of protective CD69+ CD103+ 4M vs 1M in the lung after IAV infection. Coordinated upregulation in canonical Trm-associated genes occurs in circulating 4M vs 1M populations without the enrichment of canonical downregulated Trm genes. Thus, repeated antigen exposure arms circulating memory CD8+ T cells with enhanced capacity to form long-lived populations of Trm that enhance control of viral infections of the mLN., Competing Interests: SA, NV, SM, Sv, QS, RV, RS, SH, IJ, SV, NB, HX, VB, JH No competing interests declared, (© 2021, Anthony et al.)- Published
- 2021
- Full Text
- View/download PDF
25. NK Cell-Derived IL-10 Supports Host Survival during Sepsis.
- Author
-
Jensen IJ, McGonagill PW, Butler NS, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Animals, Cytokine Release Syndrome blood, Cytokine Release Syndrome diagnosis, Humans, Interferon-gamma metabolism, Interleukin-10 genetics, Interleukin-15 metabolism, Killer Cells, Natural metabolism, Mice, Mice, Transgenic, Sepsis blood, Sepsis diagnosis, Sepsis immunology, Severity of Illness Index, Signal Transduction immunology, Cytokine Release Syndrome immunology, Interleukin-10 metabolism, Killer Cells, Natural immunology, Sepsis complications
- Abstract
The dysregulated sepsis-induced cytokine storm evoked during systemic infection consists of biphasic and interconnected pro- and anti-inflammatory responses. The contrasting inflammatory cytokine responses determine the severity of the septic event, lymphopenia, host survival, and the ensuing long-lasting immunoparalysis state. NK cells, because of their capacity to elaborate pro- (i.e., IFN-γ) and anti-inflammatory (i.e., IL-10) responses, exist at the inflection of sepsis-induced inflammatory responses. Thus, NK cell activity could be beneficial or detrimental during sepsis. In this study, we demonstrate that murine NK cells promote host survival during sepsis by limiting the scope and duration of the cytokine storm. Specifically, NK cell-derived IL-10, produced in response to IL-15, is relevant to clinical manifestations in septic patients and critical for survival during sepsis. This role of NK cells demonstrates that regulatory mechanisms of classical inflammatory cells are beneficial and critical for controlling systemic inflammation, a notion relevant for therapeutic interventions during dysregulated infection-induced inflammatory responses., (Copyright © 2021 by The American Association of Immunologists, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
26. γδ T cells burst malaria's bubble.
- Author
-
Lefebvre MN and Harty JT
- Subjects
- Homicide, Humans, Phagocytosis, T-Lymphocytes, Malaria, Falciparum, Plasmodium falciparum
- Published
- 2021
- Full Text
- View/download PDF
27. Influenza-Specific Lung-Resident Memory CD8 + T Cells.
- Author
-
van de Wall S, Badovinac VP, and Harty JT
- Subjects
- Animals, Humans, Influenza Vaccines, Influenza, Human prevention & control, CD8-Positive T-Lymphocytes, Influenza, Human immunology, Lung immunology, Memory T Cells
- Abstract
Despite the availability of seasonal vaccines, influenza A (IAV) prevails as a leading cause of respiratory infection worldwide. Current vaccination efforts aim at increasing protection against heterologous and potentially pandemic IAV strains. Lung-resident CD8
+ T cells (Trm) generated upon IAV infection are vital for heterosubtypic immunity to IAV reexposure and provide quick and robust responses upon reactivation. Yet, protection wanes with time as lung Trm cell numbers decline, a contrasting feature with Trm cells at other mucosal sites such as the skin. In this review, we discuss current data on lung Trm compared to Trm cells in other tissues. Furthermore, major knowledge gaps in the generation and maintenance of IAV-specific lung Trm are addressed and mechanisms that may contribute to their decline are discussed. Further understanding in the mechanisms that govern effector function versus immunopathology is paramount for future IAV vaccine design in enhancing durability of lung Trm cells., (Copyright © 2021 Cold Spring Harbor Laboratory Press; all rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
28. Balancing in a black box: Potential immunomodulatory roles for TGF-β signaling during blood-stage malaria.
- Author
-
Drewry LL and Harty JT
- Subjects
- Animals, Cytokines blood, Humans, Immunity, Cellular, Immunity, Humoral, Interferon-gamma, Interleukin-10, Malaria blood, Malaria parasitology, Mice, Th1 Cells immunology, Immunomodulation physiology, Malaria immunology, Transforming Growth Factor beta immunology
- Abstract
Malarial disease caused by Plasmodium parasites challenges the mammalian immune system with a delicate balancing act. Robust inflammatory responses are required to control parasite replication within red blood cells, which if unchecked, can lead to severe anemia and fatality. However, the same inflammatory response that controls parasite replication is also associated with immunopathology and severe disease, as is exemplified by cerebral malaria. A robust literature has identified critical roles for innate, cellular, and humoral immune responses orchestrated by IFN-γ and T
H 1 type responses in controlling blood stage malarial disease. In contrast, TGF-β and IL-10 have been identified as important anti-inflammatory immunomodulators that help to limit inflammation and pathology during malaria. TGF-β is a pleiotropic cytokine, with the ability to exert a wide variety of context-dependent immunomodulatory roles.The specific mechanisms that allow TGF-β to protect against malarial pathology remain essentially unexplored and offer a promising avenue to dissect the most critical elements of immunomodulation in avoiding severe malaria. Here we discuss potential immunomodulatory roles for TGF-β during malaria in light of recent advances in our understanding of the role of Tregs during blood-stage malaria.- Published
- 2020
- Full Text
- View/download PDF
29. 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
30. 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
31. RPL-6: An Achilles Needle in the Malaria Haystack?
- Author
-
Drewry LL and Harty JT
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Immunity, Cellular, Liver, Mice, Ribosomal Proteins, Malaria prevention & control, Malaria Vaccines, Plasmodium immunology
- Abstract
Malaria is a global health scourge for which a highly effective vaccine remains frustratingly elusive. Recent identification of an endogenous malaria antigen that stimulates robust T
RM -mediated immunity in mice by Valencia-Hernandez et al. strengthens the case for prime-and-trap malaria vaccines and will greatly aid further investigations of cellular antimalarial immunity., (Copyright © 2020. Published by Elsevier Ltd.)- Published
- 2020
- Full Text
- View/download PDF
32. Diverse CD8 T Cell Responses to Viral Infection Revealed by the Collaborative Cross.
- Author
-
Martin MD, Sompallae R, Winborn CS, Harty JT, and Badovinac VP
- Subjects
- Animals, Crosses, Genetic, Female, Genetic Variation genetics, Genotype, Haplotypes genetics, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred Strains, Models, Immunological, Phenotype, Quantitative Trait Loci genetics, T-Lymphocytes metabolism, Virus Diseases genetics, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, T-Lymphocytes immunology
- Abstract
Enhanced host protection against re-infection requires generation of memory T cells of sufficient quantity and functional quality. Unlike well-studied inbred mice, T cell responses of diverse size and quality are generated following infection of humans and outbred mice. Thus, additional models are needed that accurately reflect variation in immune outcomes in genetically diverse populations and to uncover underlying genetic causes. The Collaborative Cross (CC), a large recombinant inbred panel of mice, is an ideal model in this pursuit for the high degree of genetic variation present, because it allows for assessment of genetic factors underlying unique phenotypes. Here, we advance the utility of the CC as a tool to analyze the immune response to viral infection. We describe variability in resting immune cell composition and adaptive immune responses generated among CC strains following systemic virus infection and reveal quantitative trait loci responsible for generation of CD62L+ memory CD8 T cells., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Worry and FRET: ROS Production Leads to Fluorochrome Tandem Degradation and impairs Interpretation of Flow Cytometric Results.
- Author
-
Jensen IJ, McGonagill PW, Lefebvre MN, Griffith TS, Harty JT, and Badovinac VP
- Subjects
- Animals, Benzothiazoles chemistry, Benzothiazoles metabolism, Carbocyanines chemistry, Carbocyanines metabolism, Fluorescent Dyes chemistry, Granulocytes chemistry, Granulocytes metabolism, Humans, Mercaptoethanol chemistry, Mercaptoethanol metabolism, Phycocyanin chemistry, Phycocyanin metabolism, Reproducibility of Results, Flow Cytometry methods, Fluorescence Resonance Energy Transfer methods, Fluorescent Dyes metabolism, Reactive Oxygen Species metabolism
- Published
- 2020
- Full Text
- View/download PDF
34. You Shall Not Pass: Memory CD8 T Cells in Liver-Stage Malaria.
- Author
-
Lefebvre MN and Harty JT
- Subjects
- Antigens, Protozoan immunology, Immunologic Memory immunology, Liver parasitology, Malaria parasitology, Malaria Vaccines immunology, CD8-Positive T-Lymphocytes immunology, Malaria immunology
- Abstract
Each year over 200 million malaria infections occur, with over 400 000 associated deaths. Vaccines formed with attenuated whole parasites can induce protective memory CD8 T cell responses against liver-stage malaria; however, widespread administration of such vaccines is logistically challenging. Recent scientific findings are delineating how protective memory CD8 T cell populations are primed and maintained and how such cells mediate immunity to liver-stage malaria. Memory CD8 T cell anatomic localization and expression of transcription factors, homing receptors, and signaling molecules appear to play integral roles in protective immunity to liver-stage malaria. Further investigation of how such factors contribute to optimal protective memory CD8 T cell generation and maintenance in humans will inform efforts for improved vaccines., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
35. 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
36. Sepsis-Induced State of Immunoparalysis Is Defined by Diminished CD8 T Cell-Mediated Antitumor Immunity.
- Author
-
Danahy DB, Kurup SP, Winborn CS, Jensen IJ, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antigens, CD immunology, Cecum surgery, Cell Line, Tumor, Cell Proliferation drug effects, Disease Models, Animal, Female, Lymphocyte Count, Male, Melanoma, Experimental pathology, Mice, Mice, Inbred C57BL, Programmed Cell Death 1 Receptor immunology, Lymphocyte Activation Gene 3 Protein, Antigens, CD metabolism, CD8-Positive T-Lymphocytes immunology, Lymphocytes, Tumor-Infiltrating immunology, Melanoma, Experimental immunology, Programmed Cell Death 1 Receptor metabolism, Sepsis immunology
- Abstract
Patients who survive sepsis experience long-term immunoparalysis characterized by numerical and/or functional lesions in innate and adaptive immunity that increase the host's susceptibility to secondary complications. The extent to which tumor development/growth is affected in sepsis survivors remains unknown. In this study, we show cecal ligation and puncture (CLP) surgery renders mice permissive to increased B16 melanoma growth weeks/months after sepsis induction. CD8 T cells provide partial protection in this model, and tumors from sepsis survivors had a reduced frequency of CD8 tumor-infiltrating lymphocytes (TILs) concomitant with an increased tumor burden. Interestingly, the postseptic environment reduced the number of CD8 TILs with high expression of activating/inhibitory receptors PD-1 and LAG-3 (denoted PD-1
hi ) that define a tumor-specific CD8 T cell subset that retain some functional capacity. Direct ex vivo analysis of CD8 TILs from CLP hosts showed decreased proliferation, IFN-γ production, and survival compared with sham counterparts. To increase the frequency and/or functional capacity of PD-1hi CD8 TILs in tumor-bearing sepsis survivors, checkpoint blockade therapy using anti-PD-L1/anti-LAG-3 mAb was administered before or after the development of sepsis-induced lesions in CD8 TILs. Checkpoint blockade did not reduce tumor growth in CLP hosts when therapy was administered after PD-1hi CD8 TILs had become reduced in frequency and/or function. However, early therapeutic intervention before lesions were observed significantly reduced tumor growth to levels seen in nonseptic hosts receiving therapy. Thus, sepsis-induced immunoparalysis is defined by diminished CD8 T cell-mediated antitumor immunity that can respond to timely checkpoint blockade, further emphasizing the importance of early cancer detection in hosts that survive sepsis., (Copyright © 2019 by The American Association of Immunologists, Inc.)- Published
- 2019
- Full Text
- View/download PDF
37. T cell-mediated immunity to malaria.
- Author
-
Kurup SP, Butler NS, and Harty JT
- Subjects
- Animals, Cytokines physiology, Humans, Malaria Vaccines immunology, Malaria immunology, T-Lymphocytes immunology
- Abstract
Immunity to malaria has been linked to the availability and function of helper CD4
+ T cells, cytotoxic CD8+ T cells and γδ T cells that can respond to both the asymptomatic liver stage and the symptomatic blood stage of Plasmodium sp. infection. These T cell responses are also thought to be modulated by regulatory T cells. However, the precise mechanisms governing the development and function of Plasmodium-specific T cells and their capacity to form tissue-resident and long-lived memory populations are less well understood. The field has arrived at a point where the push for vaccines that exploit T cell-mediated immunity to malaria has made it imperative to define and reconcile the mechanisms that regulate the development and functions of Plasmodium-specific T cells. Here, we review our current understanding of the mechanisms by which T cell subsets orchestrate host resistance to Plasmodium infection on the basis of observational and mechanistic studies in humans, non-human primates and rodent models. We also examine the potential of new experimental strategies and human infection systems to inform a new generation of approaches to harness T cell responses against malaria.- Published
- 2019
- Full Text
- View/download PDF
38. Bystander responses impact accurate detection of murine and human antigen-specific CD8 T cells.
- Author
-
Martin MD, Jensen IJ, Ishizuka AS, Lefebvre M, Shan Q, Xue HH, Harty JT, Seder RA, and Badovinac VP
- Subjects
- Animals, Brefeldin A pharmacology, Cytokines metabolism, Female, Humans, Immunization, Immunologic Memory, Interferon-gamma metabolism, Leukocytes, Mononuclear cytology, Malaria prevention & control, Malaria Vaccines, Mice, Mice, Inbred C57BL, Phenotype, Signal Transduction, Spleen cytology, Vaccines, Attenuated immunology, Antigens immunology, Bystander Effect, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology
- Abstract
Induction of memory CD8 T cells is important for controlling infections such as malaria HIV/AIDS, and for cancer immunotherapy. Accurate assessment of antigen (Ag)-specific CD8 T-cells is critical for vaccine optimization and defining correlates of protection. However, conditions for determining Ag-specific CD8 T-cell responses ex-vivo using ICS may be variable, especially in humans with complex antigens. Here, we used an attenuated whole parasite malaria vaccine model in humans and various experimental infections in mice to show that the duration of antigenic stimulation and timing of brefeldin A (BFA) addition influences the magnitude of Ag-specific and bystander T cell responses. Indeed, following immunization with an attenuated whole sporozoite malaria vaccine in humans, significantly higher numbers of IFN-γ producing memory CD8 T-cells comprised of antigen specific and bystander responses were detected by increasing the duration of Ag-stimulation prior to addition of BFA. Mechanistic analyses of virus-specific CD8 T-cells in mice revealed that the increase in IFNg producing CD8 T-cells was due to bystander activation of Ag-experienced memory CD8 T-cells, and correlated with the proportion of Ag-experienced CD8 T-cells in the stimulated populations. Incubation with anti-cytokine antibodies (ex. IL-12) improved accuracy in detecting bona-fide memory CD8 T-cell responses suggesting this as the mechanism for the bystander activation. These data have important implications for accurate assessment of immune responses generated by vaccines intended to elicit protective memory CD8 T-cells.
- Published
- 2019
- Full Text
- View/download PDF
39. 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
40. 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
41. Repeated Antigen Exposure Extends the Durability of Influenza-Specific Lung-Resident Memory CD8 + T Cells and Heterosubtypic Immunity.
- Author
-
Van Braeckel-Budimir N, Varga SM, Badovinac VP, and Harty JT
- Subjects
- Animals, Female, Lung cytology, Mice, Mice, Inbred C57BL, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Lung immunology, Orthomyxoviridae Infections immunology
- Abstract
Lung-resident primary memory CD8
+ T cell populations (Trm ) induced by a single influenza infection decline within months, rendering the host susceptible to new heterosubtypic influenza infections. Here, we demonstrate that, relative to single virus exposure, repeated antigen exposure dramatically alters the dynamics of influenza-specific lung Trm populations. Lung Trm derived from repeatedly stimulated circulating memory CD8+ T cells exhibit extended durability and protective heterosubtypic immunity relative to primary lung Trm . Parabiosis studies reveal that the enhanced durability of lung Trm after multiple antigen encounters resulted from the generation of long-lasting circulating effector memory (Tem ) populations, which maintained the ability to be recruited to the lung parenchyma and converted to Trm , in combination with enhanced survival of these cells in the lung. Thus, generating a long-lasting Trm precursor pool through repeated intranasal immunizations might be a promising strategy to establish long-lasting lung Trm -mediated heterosubtypic immunity against influenza., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
42. 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
43. Revealing the Complexity in CD8 T Cell Responses to Infection in Inbred C57B/6 versus Outbred Swiss Mice.
- Author
-
Martin MD, Danahy DB, Hartwig SM, Harty JT, and Badovinac VP
- Abstract
Recent work has suggested that current mouse models may underrepresent the complexity of human immune responses. While most mouse immunology studies utilize inbred mouse strains, it is unclear if conclusions drawn from inbred mice can be extended to all mouse strains or generalized to humans. We recently described a "surrogate activation marker" approach that could be used to track polyclonal CD8 T cell responses in inbred and outbred mice and noted substantial discord in the magnitude and kinetics of CD8 T cell responses in individual outbred mice following infection. However, how the memory CD8 T cell response develops following infection and the correlates of memory CD8 T cell-mediated protection against re-infection in outbred mice remains unknown. In this study, we investigated development of pathogen-specific memory CD8 T cell responses in inbred C57B/6 and outbred National Institutes of Health Swiss mice following lymphocytic choriomeningitis virus or L. monocytogenes infection. Interestingly, the size of the memory CD8 T cell pool generated and rate of phenotypic progression was considerably more variable in individual outbred compared to inbred mice. Importantly, while prior infection provided both inbred and outbred cohorts of mice with protection against re-infection that was dependent on the dose of primary infection, levels of memory CD8 T cells generated and degree of protection against re-infection did not correlate with primary infection dose in all outbred mice. While variation in CD8 T cell responses to infection is not entirely surprising due to the genetic diversity present, analysis of infection-induced immunity in outbred hosts may reveal hidden complexity in CD8 T cell responses in genetically diverse populations and might help us further bridge the gap between mouse and human studies.
- Published
- 2017
- Full Text
- View/download PDF
44. A T Cell Receptor Locus Harbors a Malaria-Specific Immune Response Gene.
- Author
-
Van Braeckel-Budimir N, Gras S, Ladell K, Josephs TM, Pewe L, Urban SL, Miners KL, Farenc C, Price DA, Rossjohn J, and Harty JT
- Subjects
- 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, Histocompatibility Antigen H-2D genetics, Plasmodium berghei immunology, Protozoan Proteins immunology, Receptors, Antigen, T-Cell genetics
- 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 Vβ8.1+ precursors, which dictated susceptibility to cerebral malaria and conferred protection against recombinant Listeria monocytogenes infection. Structural analysis of a prototypical Vβ8.1+ TCR-H-2Db -GAP5040-48 ternary complex revealed that germline-encoded complementarity-determining region 1β residues present exclusively in the Vβ8.1 segment mediated essential interactions with the GAP5040-48 peptide. Collectively, these findings demonstrated that Vβ8.1 functioned as an Ir gene that was indispensable for immune reactivity against the malaria GAP5040-48 epitope., (Copyright © 2017 Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
45. Enzymatic synthesis of core 2 O-glycans governs the tissue-trafficking potential of memory CD8 + T cells.
- Author
-
Osborn JF, Mooster JL, Hobbs SJ, Munks MW, Barry C, Harty JT, Hill AB, and Nolz JC
- Subjects
- Animals, CD8-Positive T-Lymphocytes enzymology, Cell Movement, Cytoplasm immunology, Cytoplasm virology, Inflammation, Interleukin-15 genetics, Interleukin-15 immunology, Lymphocytic choriomeningitis virus immunology, Mice, Polysaccharides biosynthesis, CD8-Positive T-Lymphocytes immunology, Immunologic Memory, Polysaccharides immunology
- Abstract
Trafficking of memory CD8
+ T cells out of the circulation is essential to provide protective immunity against intracellular pathogens in nonlymphoid tissues. However, the molecular mechanisms that dictate the trafficking potential of diverse memory CD8+ T cell populations are not completely defined. We show that after infection or inflammatory challenge, central memory (TCM ) CD8+ T cells rapidly traffic into nonlymphoid tissues, whereas most effector memory cells remain in the circulation. Furthermore, we demonstrate that cellular migration of memory CD8+ T cells into nonlymphoid tissues is driven by interleukin-15 (IL-15)-stimulated enzymatic synthesis of core 2 O-glycans, which generates functional ligands for E- and P-selectins. Given that IL-15-stimulated expression of glycosyltransferase enzymes is largely a feature of TCM CD8+ T cells, this allows TCM to selectively migrate out of the circulation and into nonlymphoid tissues. Collectively, our data indicate that entry of memory CD8+ T cells into inflamed, nonlymphoid tissues is primarily restricted to TCM cells that have the capacity to synthesize core 2 O-glycans., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2017
- Full Text
- View/download PDF
46. Characterization of Inner and Outer Membrane Proteins from Francisella tularensis Strains LVS and Schu S4 and Identification of Potential Subunit Vaccine Candidates.
- Author
-
Post DMB, Slütter B, Schilling B, Chande AT, Rasmussen JA, Jones BD, D'Souza AK, Reinders LM, Harty JT, Gibson BW, and Apicella MA
- Subjects
- Adjuvants, Immunologic, Animals, Disease Models, Animal, Francisella tularensis chemistry, Francisella tularensis pathogenicity, Lactic Acid, Macrophages immunology, Macrophages microbiology, Mass Spectrometry, Membrane Proteins chemistry, Membrane Proteins genetics, Membrane Proteins isolation & purification, Mice, Mice, Inbred BALB C, Nanoparticles, Poly I-C immunology, Polyglycolic Acid, Polylactic Acid-Polyglycolic Acid Copolymer, Proteomics, Tularemia immunology, Vaccination, Vaccines, Attenuated immunology, Vaccines, Subunit genetics, Bacterial Vaccines immunology, Francisella tularensis immunology, Membrane Proteins immunology, Tularemia prevention & control, Vaccines, Subunit immunology
- Abstract
Francisella tularensis is the causative agent of tularemia and a potential bioterrorism agent. In the present study, we isolated, identified, and quantified the proteins present in the membranes of the virulent type A strain, Schu S4, and the attenuated type B strain, LVS (live vaccine strain). Spectral counting of mass spectrometric data showed enrichment for membrane proteins in both strains. Mice vaccinated with whole LVS membranes encapsulated in poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing the adjuvant polyinosinic-polycytidylic acid [poly(I·C)] showed significant protection against a challenge with LVS compared to the results seen with naive mice or mice vaccinated with either membranes or poly(I·C) alone. The PLGA-encapsulated Schu S4 membranes with poly(I·C) alone did not significantly protect mice from a lethal intraperitoneal challenge with Schu S4; however, this vaccination strategy provided protection from LVS challenge. Mice that received the encapsulated Schu S4 membranes followed by a booster of LVS bacteria showed significant protection with respect to a lethal Schu S4 challenge compared to control mice. Western blot analyses of the sera from the Schu S4-vaccinated mice that received an LVS booster showed four immunoreactive bands. One of these bands from the corresponding one-dimensional (1D) SDS-PAGE experiment represented capsule. The remaining bands were excised, digested with trypsin, and analyzed using mass spectrometry. The most abundant proteins present in these immunoreactive samples were an outer membrane OmpA-like protein, FopA; the type IV pilus fiber building block protein; a hypothetical membrane protein; and lipoproteins LpnA and Lpp3. These proteins should serve as potential targets for future recombinant protein vaccination studies. IMPORTANCE The low infectious dose, the high potential mortality/morbidity rates, and the ability to be disseminated as an aerosol make Francisella tularensis a potential agent for bioterrorism. These characteristics led the Centers for Disease Control (CDC) to classify F. tularensis as a Tier 1 pathogen. Currently, there is no vaccine approved for general use in the United States., (Copyright © 2017 Post et al.)
- Published
- 2017
- Full Text
- View/download PDF
47. Regulatory T cells impede acute and long-term immunity to blood-stage malaria through CTLA-4.
- Author
-
Kurup SP, Obeng-Adjei N, Anthony SM, Traore B, Doumbo OK, Butler NS, Crompton PD, and Harty JT
- Subjects
- Animals, Enzyme-Linked Immunosorbent Assay, Flow Cytometry, Germinal Center immunology, Humans, Immune Tolerance immunology, Lymph Nodes pathology, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Spleen pathology, CTLA-4 Antigen immunology, Malaria immunology, Parasitemia immunology, T-Lymphocytes, Helper-Inducer immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Malaria, caused by the protozoan Plasmodium, is a devastating mosquito-borne disease with the potential to affect nearly half the world's population. Despite mounting substantial T and B cell responses, humans fail to efficiently control blood-stage malaria or develop sterilizing immunity to reinfections. Although forkhead box P3 (FOXP3)
+ CD4+ regulatory T (Treg ) cells form a part of these responses, their influence remains disputed and their mode of action is unknown. Here we show that Treg cells expand in both humans and mice in blood-stage malaria and interfere with conventional T helper cell responses and follicular T helper (TFH )-B cell interactions in germinal centers. Mechanistically, Treg cells function in a critical temporal window to impede protective immunity through cytotoxic-T-lymphocyte-associated protein-4 (CTLA-4). Targeting Treg cells or CTLA-4 in this precise window accelerated parasite clearance and generated species-transcending immunity to blood-stage malaria in mice. Our study uncovers a critical mechanism of immunosuppression associated with blood-stage malaria that delays parasite clearance and prevents development of potent adaptive immunity to reinfection. These data also reveal a temporally discrete and potentially therapeutically amenable functional role for Treg cells and CTLA-4 in limiting antimalarial immunity.- Published
- 2017
- Full Text
- View/download PDF
48. Polymicrobial sepsis impairs bystander recruitment of effector cells to infected skin despite optimal sensing and alarming function of skin resident memory CD8 T cells.
- Author
-
Danahy DB, Anthony SM, Jensen IJ, Hartwig SM, Shan Q, Xue HH, Harty JT, Griffith TS, and Badovinac VP
- Subjects
- Animals, Antigens immunology, Cytokines metabolism, Interferon-gamma biosynthesis, Skin immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Sepsis immunology, Vaccinia virus immunology
- Abstract
Sepsis is a systemic infection that enhances host vulnerability to secondary infections normally controlled by T cells. Using CLP sepsis model, we observed that sepsis induces apoptosis of circulating memory CD8 T-cells (TCIRCM) and diminishes their effector functions, leading to impaired CD8 T-cell mediated protection to systemic pathogen re-infection. In the context of localized re-infections, tissue resident memory CD8 T-cells (TRM) provide robust protection in a variety of infectious models. TRM rapidly 'sense' infection in non-lymphoid tissues and 'alarm' the host by enhancing immune cell recruitment to the site of the infection to accelerate pathogen clearance. Here, we show that compared to pathogen-specific TCIRCM, sepsis does not invoke significant numerical decline of Vaccinia virus induced skin-TRM keeping their effector functions (e.g., Ag-dependent IFN-γ production) intact. IFN-γ-mediated recruitment of immune cells to the site of localized infection was, however, reduced in CLP hosts despite TRM maintaining their 'sensing and alarming' functions. The capacity of memory CD8 T-cells in the septic environment to respond to inflammatory cues and arrive to the site of secondary infection/antigen exposure remained normal suggesting T-cell-extrinsic factors contributed to the observed lesion. Mechanistically, we showed that IFN-γ produced rapidly during sepsis-induced cytokine storm leads to reduced IFN-γR1 expression on vascular endothelium. As a consequence, decreased expression of adhesion molecules and/or chemokines (VCAM1 and CXCL9) on skin endothelial cells in response to TRM-derived IFN-γ was observed, leading to sub-optimal bystander-recruitment of effector cells and increased susceptibility to pathogen re-encounter. Importantly, as visualized by intravital 2-photon microscopy, exogenous administration of CXCL9/10 was sufficient to correct sepsis-induced impairments in recruitment of effector cells at the localized site of TRM antigen recognition. Thus, sepsis has the capacity to alter skin TRM anamnestic responses without directly impacting TRM number and/or function, an observation that helps to further define the immunoparalysis phase in sepsis survivors.
- Published
- 2017
- Full Text
- View/download PDF
49. Influenza-induced lung T rm : not all memories last forever.
- Author
-
Van Braeckel-Budimir N and Harty JT
- Subjects
- Animals, Humans, Immunologic Memory, Lung virology, Mass Vaccination, CD8-Positive T-Lymphocytes immunology, Influenza Vaccines immunology, Influenza, Human immunology, Lung immunology, Orthomyxoviridae immunology, Orthomyxoviridae Infections immunology
- Abstract
In the light of new findings that lung tissue resident memory CD8
+ T cells (Trm ) represent major mediators of heterosubtypic immunity against influenza virus, it is of utmost importance to understand the basic biological mechanisms behind induction, formation and maintenance of this cell population. Addressing these important knowledge gaps will potentially inform development of superior, broadly protective influenza vaccines and new immunization strategies.- Published
- 2017
- Full Text
- View/download PDF
50. Differential Requirements for Tcf1 Long Isoforms in CD8 + and CD4 + T Cell Responses to Acute Viral Infection.
- Author
-
Gullicksrud JA, Li F, Xing S, Zeng Z, Peng W, Badovinac VP, Harty JT, and Xue HH
- Subjects
- Animals, Cell Differentiation, Cytotoxicity Tests, Immunologic, Germinal Center cytology, Germinal Center immunology, Germinal Center metabolism, Immunologic Memory, Inducible T-Cell Co-Stimulator Protein genetics, Inducible T-Cell Co-Stimulator Protein metabolism, Inhibitor of Differentiation Proteins genetics, Inhibitor of Differentiation Proteins metabolism, Lymphocytic choriomeningitis virus isolation & purification, Mice, Positive Regulatory Domain I-Binding Factor 1, Protein Isoforms, Proto-Oncogene Proteins c-bcl-6 genetics, Proto-Oncogene Proteins c-bcl-6 metabolism, T Cell Transcription Factor 1 deficiency, T Cell Transcription Factor 1 genetics, Transcription Factors genetics, Transcription Factors metabolism, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Lymphocytic Choriomeningitis immunology, Lymphocytic choriomeningitis virus immunology, T Cell Transcription Factor 1 chemistry, T Cell Transcription Factor 1 immunology
- Abstract
In response to acute viral infection, activated naive T cells give rise to effector T cells that clear the pathogen and memory T cells that persist long-term and provide heightened protection. T cell factor 1 (Tcf1) is essential for several of these differentiation processes. Tcf1 is expressed in multiple isoforms, with all isoforms sharing the same HDAC and DNA-binding domains and the long isoforms containing a unique N-terminal β-catenin-interacting domain. In this study, we specifically ablated Tcf1 long isoforms in mice, while retaining expression of Tcf1 short isoforms. During CD8
+ T cell responses, Tcf1 long isoforms were dispensable for generating cytotoxic CD8+ effector T cells and maintaining memory CD8+ T cell pool size, but they contributed to optimal maturation of central memory CD8+ T cells and their optimal secondary expansion in a recall response. In contrast, Tcf1 long isoforms were required for differentiation of T follicular helper (TFH ) cells, but not TH 1 effectors, elicited by viral infection. Although Tcf1 short isoforms adequately supported Bcl6 and ICOS expression in TFH cells, Tcf1 long isoforms remained important for suppressing the expression of Blimp1 and TH 1-associated genes and for positively regulating Id3 to restrain germinal center TFH cell differentiation. Furthermore, formation of memory TH 1 and memory TFH cells strongly depended on Tcf1 long isoforms. These data reveal that Tcf1 long and short isoforms have distinct, yet complementary, functions and may represent an evolutionarily conserved means to ensure proper programming of CD8+ and CD4+ T cell responses to viral infection., (Copyright © 2017 by The American Association of Immunologists, Inc.)- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.