71 results on '"Lynette, Beattie"'
Search Results
2. A CAF01-adjuvanted whole asexual blood-stage liposomal malaria vaccine induces a CD4+ T-cell-dependent strain-transcending protective immunity in rodent models
- Author
-
Winter A. Okoth, Mei-Fong Ho, Mehfuz Zaman, Emily Cooper, Priyanka Som, Mark Burgess, Maddison Walton, Reshma J. Nevagi, Lynette Beattie, Declan Murphy, Danielle I. Stanisic, and Michael F. Good
- Subjects
CAF01 adjuvanted ,whole blood stage ,malaria ,vaccine ,liposomes ,Microbiology ,QR1-502 - Abstract
ABSTRACTMalaria is a leading cause of illness and death in children under 5 years of age in Sub-Saharan Africa. Currently, there is no highly efficacious malaria vaccine capable of inducing long-lasting immunity. This has increased interest in exploring different vaccine development strategies, including whole-parasite vaccines, and utilizing more effective adjuvant delivery systems. Here, we evaluate the immunogenicity and protective efficacy of a whole-parasite Plasmodium yoelii 17X blood-stage vaccine formulated with the clinically tested cationic adjuvant formulation, CAF01. The vaccine protected both inbred and outbred mice, and protected mice from homologous and heterologous challenge infections. Tracking of RBC subsets in vaccinated mice demonstrated that vaccine-induced immunity cleared parasitized normocytes as well as reticulocytes. Infection prior to vaccination led to an augmented level of protection and boosted immunity post vaccination. The vaccine induced parasite-specific IgG, mainly of the IgG1 subclass, and cellular responses, with a mixed Th1/Th2/Th17 cytokine profile. Mechanistic studies demonstrated that CD4+ T-cells (but not CD8+ T-cells), and Th1 and Th2 cytokines (interferon gamma and tumor necrosis factor, IL-10) were critical in controlling parasitemia and survival following challenge. Vaccinated µMT mice were not protected, suggesting that B-cells also play a role in protective immunity. Depletion of splenic macrophages with clodronate did not affect vaccine efficacy. These pre-clinical findings will inform the transition of this vaccine candidate into clinical trials.IMPORTANCEMalaria is a devastating disease that has claimed many lives, especially children
- Published
- 2023
- Full Text
- View/download PDF
3. Vaccine-induced inflammation and inflammatory monocytes promote CD4+ T cell-dependent immunity against murine salmonellosis.
- Author
-
Nancy Wang, Timothy A Scott, Andreas Kupz, Meghanashree M Shreenivas, Newton G Peres, Dianna M Hocking, Chenying Yang, Leila Jebeli, Lynette Beattie, Joanna R Groom, Thomas P Pierce, Linda M Wakim, Sammy Bedoui, and Richard A Strugnell
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Prior infection can generate protective immunity against subsequent infection, although the efficacy of such immunity can vary considerably. Live-attenuated vaccines (LAVs) are one of the most effective methods for mimicking this natural process, and analysis of their efficacy has proven instrumental in the identification of protective immune mechanisms. Here, we address the question of what makes a LAV efficacious by characterising immune responses to a LAV, termed TAS2010, which is highly protective (80-90%) against lethal murine salmonellosis, in comparison with a moderately protective (40-50%) LAV, BRD509. Mice vaccinated with TAS2010 developed immunity systemically and were protected against gut-associated virulent infection in a CD4+ T cell-dependent manner. TAS2010-vaccinated mice showed increased activation of Th1 responses compared with their BRD509-vaccinated counterparts, leading to increased Th1 memory populations in both lymphoid and non-lymphoid organs. The optimal development of Th1-driven immunity was closely correlated with the activation of CD11b+Ly6GnegLy6Chi inflammatory monocytes (IMs), the activation of which can be modulated proportionally by bacterial load in vivo. Upon vaccination with the LAV, IMs expressed T cell chemoattractant CXCL9 that attracted CD4+ T cells to the foci of infection, where IMs also served as a potent source of antigen presentation and Th1-promoting cytokine IL-12. The expression of MHC-II in IMs was rapidly upregulated following vaccination and then maintained at an elevated level in immune mice, suggesting IMs may have a role in sustained antigen stimulation. Our findings present a longitudinal analysis of CD4+ T cell development post-vaccination with an intracellular bacterial LAV, and highlight the benefit of inflammation in the development of Th1 immunity. Future studies focusing on the induction of IMs may reveal key strategies for improving vaccine-induced T cell immunity.
- Published
- 2023
- Full Text
- View/download PDF
4. Plasmodium berghei Hsp90 contains a natural immunogenic I-Ab-restricted antigen common to rodent and human Plasmodium species
- Author
-
Matthias H. Enders, Ganchimeg Bayarsaikhan, Sonia Ghilas, Yu Cheng Chua, Rose May, Maria N. de Menezes, Zhengyu Ge, Peck Szee Tan, Anton Cozijnsen, Vanessa Mollard, Katsuyuki Yui, Geoffrey I. McFadden, Mireille H. Lahoud, Irina Caminschi, Anthony W. Purcell, Ralf B. Schittenhelm, Lynette Beattie, William R. Heath, and Daniel Fernandez-Ruiz
- Subjects
CD4 T cell subsets ,Plasmodium epitope ,Vaccination ,T cell memory ,Experimental cerebral malaria ,Malaria immunity ,Specialties of internal medicine ,RC581-951 - Abstract
Thorough understanding of the role of CD4 T cells in immunity can be greatly assisted by the study of responses to defined specificities. This requires knowledge of Plasmodium-derived immunogenic epitopes, of which only a few have been identified, especially for the mouse C57BL/6 background. We recently developed a TCR transgenic mouse line, termed PbT-II, that produces CD4+ T cells specific for an MHC class II (I-Ab)-restricted Plasmodium epitope and is responsive to both sporozoites and blood-stage P. berghei. Here, we identify a peptide within the P. berghei heat shock protein 90 as the cognate epitope recognised by PbT-II cells. We show that C57BL/6 mice infected with P. berghei blood-stage induce an endogenous CD4 T cell response specific for this epitope, indicating cells of similar specificity to PbT-II cells are present in the naïve repertoire. Adoptive transfer of in vitro activated TH1-, or particularly TH2-polarised PbT-II cells improved control of P. berghei parasitemia in C57BL/6 mice and drastically reduced the onset of experimental cerebral malaria. Our results identify a versatile, potentially protective MHC-II restricted epitope useful for exploration of CD4 T cell-mediated immunity and vaccination strategies against malaria.
- Published
- 2021
- Full Text
- View/download PDF
5. Type I Interferons Regulate Immune Responses in Humans with Blood-Stage Plasmodium falciparum Infection
- Author
-
Marcela Montes de Oca, Rajiv Kumar, Fabian de Labastida Rivera, Fiona H. Amante, Meru Sheel, Rebecca J. Faleiro, Patrick T. Bunn, Shannon E. Best, Lynette Beattie, Susanna S. Ng, Chelsea L. Edwards, Glen M. Boyle, Ric N. Price, Nicholas M. Anstey, Jessica R. Loughland, Julie Burel, Denise L. Doolan, Ashraful Haque, James S. McCarthy, and Christian R. Engwerda
- Subjects
malaria ,type I interferons ,CD4+ T cells ,immune regulation ,Tr1 cells ,Th1 cells ,Biology (General) ,QH301-705.5 - Abstract
The development of immunoregulatory networks is important to prevent disease. However, these same networks allow pathogens to persist and reduce vaccine efficacy. Here, we identify type I interferons (IFNs) as important regulators in developing anti-parasitic immunity in healthy volunteers infected for the first time with Plasmodium falciparum. Type I IFNs suppressed innate immune cell function and parasitic-specific CD4+ T cell IFNγ production, and they promoted the development of parasitic-specific IL-10-producing Th1 (Tr1) cells. Type I IFN-dependent, parasite-specific IL-10 production was also observed in P. falciparum malaria patients in the field following chemoprophylaxis. Parasite-induced IL-10 suppressed inflammatory cytokine production, and IL-10 levels after drug treatment were positively associated with parasite burdens before anti-parasitic drug administration. These findings have important implications for understanding the development of host immune responses following blood-stage P. falciparum infection, and they identify type I IFNs and related signaling pathways as potential targets for therapies or vaccine efficacy improvement.
- Published
- 2016
- Full Text
- View/download PDF
6. Macrophage Transactivation for Chemokine Production Identified as a Negative Regulator of Granulomatous Inflammation Using Agent-Based Modeling
- Author
-
Daniel Moyo, Lynette Beattie, Paul S. Andrews, John W. J. Moore, Jon Timmis, Amy Sawtell, Stefan Hoehme, Adam T. Sampson, and Paul M. Kaye
- Subjects
kupffer cells ,granulomas ,inflammation ,Leishmania ,natural killer T cells ,agent-based modeling ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Cellular activation in trans by interferons, cytokines, and chemokines is a commonly recognized mechanism to amplify immune effector function and limit pathogen spread. However, an optimal host response also requires that collateral damage associated with inflammation is limited. This may be particularly so in the case of granulomatous inflammation, where an excessive number and/or excessively florid granulomas can have significant pathological consequences. Here, we have combined transcriptomics, agent-based modeling, and in vivo experimental approaches to study constraints on hepatic granuloma formation in a murine model of experimental leishmaniasis. We demonstrate that chemokine production by non-infected Kupffer cells in the Leishmania donovani-infected liver promotes competition with infected KCs for available iNKT cells, ultimately inhibiting the extent of granulomatous inflammation. We propose trans-activation for chemokine production as a novel broadly applicable mechanism that may operate early in infection to limit excessive focal inflammation.
- Published
- 2018
- Full Text
- View/download PDF
7. CD8+ and CD4+ T Cells Infiltrate into the Brain during Plasmodium berghei ANKA Infection and Form Long-Term Resident Memory
- Author
-
William R. Heath, Scott N. Mueller, Julia L. Gregory, Sapna Devi, Nazanin Ghazanfari, Daniel Fernandez-Ruiz, and Lynette Beattie
- Subjects
Pathology ,medicine.medical_specialty ,Adoptive cell transfer ,biology ,Secondary infection ,T cell ,Immunology ,medicine.disease ,biology.organism_classification ,medicine.anatomical_structure ,parasitic diseases ,Parenchyma ,medicine ,Immunology and Allergy ,Plasmodium berghei ,Memory T cell ,Infiltration (medical) ,CD8 - Abstract
In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection.
- Published
- 2021
- Full Text
- View/download PDF
8. Development of Plasmodium ‐specific liver‐resident memory CD8 + T cells after heat‐killed sporozoite immunization in mice
- Author
-
Daniel Fernandez-Ruiz, Ian A. Cockburn, Anton Cozijnsen, Rose May, Geoffrey I. McFadden, Lynette Beattie, Vanessa Mollard, Matthias H. Enders, Lauren E. Holz, Sonia Ghilas, and William R. Heath
- Subjects
0301 basic medicine ,Transgene ,medicine.medical_treatment ,Immunology ,Spleen ,Biology ,3. Good health ,Vaccination ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immunization ,Immunity ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Adjuvant ,CD8 ,030215 immunology - Abstract
Malaria remains a major cause of mortality in the world and an efficient vaccine is the best chance of reducing the disease burden. Vaccination strategies for the liver stage of disease that utilise injection of live radiation-attenuated sporozoites (RAS) confer sterile immunity, which is mediated by CD8+ memory T cells, with liver-resident memory T cells (TRM ) being particularly important. We have previously described a TCR transgenic mouse, termed PbT-I, where all CD8+ T cells recognize a specific peptide from Plasmodium. PbT-I form liver TRM cells upon RAS injection and are capable of protecting mice against challenge infection. Here, we utilize this transgenic system to examine whether nonliving sporozoites, killed by heat treatment (HKS), could trigger the development of Plasmodium-specific liver TRM cells. We found that HKS vaccination induced the formation of memory CD8+ T cells in the spleen and liver, and importantly, liver TRM cells were fewer in number than that induced by RAS. Crucially, we showed the number of TRM cells was significantly higher when HKS were combined with the glycolipid α-galactosylceramide as an adjuvant. In the future, this work could lead to development of an antimalaria vaccination strategy that does not require live sporozoites, providing greater utility.
- Published
- 2021
- Full Text
- View/download PDF
9. Harnessing liver-resident memory T cells for protection against malaria
- Author
-
Sonia Ghilas, Maria N. de Menezes, Daniel Fernandez-Ruiz, Lauren E. Holz, Lynette Beattie, and William R. Heath
- Subjects
0301 basic medicine ,Plasmodium ,T-Lymphocytes ,Immunology ,03 medical and health sciences ,0302 clinical medicine ,Malaria Vaccines ,Drug Discovery ,Animals ,Humans ,Medicine ,Cytotoxic T cell ,030212 general & internal medicine ,Liver immunology ,Pharmacology ,business.industry ,Vaccination ,medicine.disease ,Acquired immune system ,Malaria ,3. Good health ,030104 developmental biology ,Liver ,Molecular Medicine ,business ,Immunologic Memory - Abstract
Tissue-resident memory T cells (TRM cells) are powerful mediators of protracted adaptive immunity to infection in peripheral organs. Harnessing TRM cells through vaccination hence promises unpreced...
- Published
- 2021
- Full Text
- View/download PDF
10. Plasmodium berghei Hsp90 contains a natural immunogenic I-Ab-restricted antigen common to rodent and human Plasmodium species
- Author
-
Ganchimeg Bayarsaikhan, William R. Heath, Lynette Beattie, Rose May, Zhengyu Ge, Vanessa Mollard, Peck Szee Tan, Daniel Fernandez-Ruiz, Anton Cozijnsen, Mireille H. Lahoud, Yu Cheng Chua, Ralf B. Schittenhelm, Matthias H. Enders, Anthony W. Purcell, Katsuyuki Yui, Maria N. de Menezes, Irina Caminschi, Sonia Ghilas, and Geoffrey I. McFadden
- Subjects
MHC class II ,Adoptive cell transfer ,biology ,Vaccination ,Immunology ,Specialties of internal medicine ,Plasmodium epitope ,T cell memory ,Parasitemia ,CD4 T cell subsets ,medicine.disease ,biology.organism_classification ,Virology ,Epitope ,RC581-951 ,Antigen ,Immunity ,Malaria immunity ,Heat shock protein ,Experimental cerebral malaria ,parasitic diseases ,medicine ,biology.protein ,Plasmodium berghei - Abstract
Thorough understanding of the role of CD4 T cells in immunity can be greatly assisted by the study of responses to defined specificities. This requires knowledge of Plasmodium-derived immunogenic epitopes, of which only a few have been identified, especially for the mouse C57BL/6 background. We recently developed a TCR transgenic mouse line, termed PbT-II, that produces CD4+ T cells specific for an MHC class II (I-Ab)-restricted Plasmodium epitope and is responsive to both sporozoites and blood-stage P. berghei. Here, we identify a peptide within the P. berghei heat shock protein 90 as the cognate epitope recognised by PbT-II cells. We show that C57BL/6 mice infected with P. berghei blood-stage induce an endogenous CD4 T cell response specific for this epitope, indicating cells of similar specificity to PbT-II cells are present in the naive repertoire. Adoptive transfer of in vitro activated TH1-, or particularly TH2-polarised PbT-II cells improved control of P. berghei parasitemia in C57BL/6 mice and drastically reduced the onset of experimental cerebral malaria. Our results identify a versatile, potentially protective MHC-II restricted epitope useful for exploration of CD4 T cell-mediated immunity and vaccination strategies against malaria.
- Published
- 2021
- Full Text
- View/download PDF
11. A Petri Net Model of Granulomatous Inflammation.
- Author
-
Luca Albergante, Jon Timmis, Paul S. Andrews, Lynette Beattie, and Paul M. Kaye
- Published
- 2010
- Full Text
- View/download PDF
12. IFNAR1-Signalling Obstructs ICOS-mediated Humoral Immunity during Non-lethal Blood-Stage Plasmodium Infection.
- Author
-
Ismail Sebina, Kylie R James, Megan S F Soon, Lily G Fogg, Shannon E Best, Fabian de Labastida Rivera, Marcela Montes de Oca, Fiona H Amante, Bryce S Thomas, Lynette Beattie, Fernando Souza-Fonseca-Guimaraes, Mark J Smyth, Paul J Hertzog, Geoffrey R Hill, Andreas Hutloff, Christian R Engwerda, and Ashraful Haque
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Parasite-specific antibodies protect against blood-stage Plasmodium infection. However, in malaria-endemic regions, it takes many months for naturally-exposed individuals to develop robust humoral immunity. Explanations for this have focused on antigenic variation by Plasmodium, but have considered less whether host production of parasite-specific antibody is sub-optimal. In particular, it is unclear whether host immune factors might limit antibody responses. Here, we explored the effect of Type I Interferon signalling via IFNAR1 on CD4+ T-cell and B-cell responses in two non-lethal murine models of malaria, P. chabaudi chabaudi AS (PcAS) and P. yoelii 17XNL (Py17XNL) infection. Firstly, we demonstrated that CD4+ T-cells and ICOS-signalling were crucial for generating germinal centre (GC) B-cells, plasmablasts and parasite-specific antibodies, and likewise that T follicular helper (Tfh) cell responses relied on B cells. Next, we found that IFNAR1-signalling impeded the resolution of non-lethal blood-stage infection, which was associated with impaired production of parasite-specific IgM and several IgG sub-classes. Consistent with this, GC B-cell formation, Ig-class switching, plasmablast and Tfh differentiation were all impaired by IFNAR1-signalling. IFNAR1-signalling proceeded via conventional dendritic cells, and acted early by limiting activation, proliferation and ICOS expression by CD4+ T-cells, by restricting the localization of activated CD4+ T-cells adjacent to and within B-cell areas of the spleen, and by simultaneously suppressing Th1 and Tfh responses. Finally, IFNAR1-deficiency accelerated humoral immune responses and parasite control by boosting ICOS-signalling. Thus, we provide evidence of a host innate cytokine response that impedes the onset of humoral immunity during experimental malaria.
- Published
- 2016
- Full Text
- View/download PDF
13. Correction: Blimp-1-Dependent IL-10 Production by Tr1 Cells Regulates TNF-Mediated Tissue Pathology.
- Author
-
Marcela Montes de Oca, Rajiv Kumar, Fabian de Labastida Rivera, Fiona H Amante, Meru Sheel, Rebecca J Faleiro, Patrick T Bunn, Shannon E Best, Lynette Beattie, Susanna S Ng, Chelsea L Edwards, Werner Muller, Erika Cretney, Stephen L Nutt, Mark J Smyth, Ashraful Haque, Geoffrey R Hill, Shyam Sundar, Axel Kallies, and Christian R Engwerda
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Published
- 2016
- Full Text
- View/download PDF
14. Combined Immune Therapy for the Treatment of Visceral Leishmaniasis.
- Author
-
Rebecca J Faleiro, Rajiv Kumar, Patrick T Bunn, Neetu Singh, Shashi Bhushan Chauhan, Meru Sheel, Fiona H Amante, Marcela Montes de Oca, Chelsea L Edwards, Susanna S Ng, Shannon E Best, Ashraful Haque, Lynette Beattie, Louise M Hafner, David Sacks, Susanne Nylen, Shyam Sundar, and Christian R Engwerda
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Chronic disease caused by infections, cancer or autoimmunity can result in profound immune suppression. Immunoregulatory networks are established to prevent tissue damage caused by inflammation. Although these immune checkpoints preserve tissue function, they allow pathogens and tumors to persist, and even expand. Immune checkpoint blockade has recently been successfully employed to treat cancer. This strategy modulates immunoregulatory mechanisms to allow host immune cells to kill or control tumors. However, the utility of this approach for controlling established infections has not been extensively investigated. Here, we examined the potential of modulating glucocorticoid-induced TNF receptor-related protein (GITR) on T cells to improve anti-parasitic immunity in blood and spleen tissue from visceral leishmaniasis (VL) patients infected with Leishmania donovani. We found little effect on parasite growth or parasite-specific IFNγ production. However, this treatment reversed the improved anti-parasitic immunity achieved by IL-10 signaling blockade. Further investigations using an experimental VL model caused by infection of C57BL/6 mice with L. donovani revealed that this negative effect was prominent in the liver, dependent on parasite burden and associated with an accumulation of Th1 cells expressing high levels of KLRG-1. Nevertheless, combined anti-IL-10 and anti-GITR mAb treatment could improve anti-parasitic immunity when used with sub-optimal doses of anti-parasitic drug. However, additional studies with VL patient samples indicated that targeting GITR had no overall benefit over IL-10 signaling blockade alone at improving anti-parasitic immune responses, even with drug treatment cover. These findings identify several important factors that influence the effectiveness of immune modulation, including parasite burden, target tissue and the use of anti-parasitic drug. Critically, these results also highlight potential negative effects of combining different immune modulation strategies.
- Published
- 2016
- Full Text
- View/download PDF
15. CD4+ Recent Thymic Emigrants Are Recruited into Granulomas during Leishmania donovani Infection but Have Limited Capacity for Cytokine Production.
- Author
-
John W J Moore, Lynette Beattie, Mohamed Osman, Benjamin M J Owens, Najmeeyah Brown, Jane E Dalton, Asher Maroof, and Paul M Kaye
- Subjects
Medicine ,Science - Abstract
Recent thymic emigrants (RTEs) represent a source of antigen-naïve T cells that enter the periphery throughout life. However, whether RTEs contribute to the control of chronic parasitic infection and how their potential might be harnessed by therapeutic intervention is currently unclear. Here, we show that CD4+ recent thymic emigrants emerging into the periphery of mice with ongoing Leishmania donovani infection undergo partial activation and are recruited to sites of granulomatous inflammation. However, CD4+ RTEs displayed severely restricted differentiation either into IFNγ+ or IFNγ+TNFα+ effectors, or into IL-10-producing regulatory T cells. Effector cell differentiation in the chronically infected host was not promoted by adoptive transfer of activated dendritic cells or by allowing extended periods of post-thymic differentiation in the periphery. Nevertheless, CD4+ RTEs from infected mice retained the capacity to transfer protection into lymphopenic RAG2-/- mice. Taken together, our data indicate that RTEs emerging into a chronically inflamed environment are not recruited into the effector pool, but retain the capacity for subsequent differentiation into host protective T cells when placed in a disease-free environment.
- Published
- 2016
- Full Text
- View/download PDF
16. Blimp-1-Dependent IL-10 Production by Tr1 Cells Regulates TNF-Mediated Tissue Pathology.
- Author
-
Marcela Montes de Oca, Rajiv Kumar, Fabian de Labastida Rivera, Fiona H Amante, Meru Sheel, Rebecca J Faleiro, Patrick T Bunn, Shannon E Best, Lynette Beattie, Susanna S Ng, Chelsea L Edwards, Werner Muller, Erika Cretney, Stephen L Nutt, Mark J Smyth, Ashraful Haque, Geoffrey R Hill, Shyam Sundar, Axel Kallies, and Christian R Engwerda
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation.
- Published
- 2016
- Full Text
- View/download PDF
17. Marginal zone B cells acquire dendritic cell functions by trogocytosis
- Author
-
Patrick Schriek, Alan C. Ching, Nagaraj S. Moily, Jessica Moffat, Lynette Beattie, Thiago M. Steiner, Laine M. Hosking, Joshua M. Thurman, V. Michael Holers, Satoshi Ishido, Mireille H. Lahoud, Irina Caminschi, William R. Heath, Justine D. Mintern, and Jose A. Villadangos
- Subjects
Adult ,Male ,Antigen Presentation ,B-Lymphocytes ,HLA-D Antigens ,Mice, Inbred BALB C ,Mice, Inbred C3H ,Multidisciplinary ,Lymphoid Tissue ,T-Lymphocytes ,Ubiquitin-Protein Ligases ,Cell Membrane ,Histocompatibility Antigens Class II ,Ubiquitination ,Complement C3 ,Dendritic Cells ,Middle Aged ,Trogocytosis ,Mice, Inbred C57BL ,Mice ,Animals ,Humans ,Female ,Receptors, Complement 3d ,Complement Activation - Abstract
Marginal zone (MZ) B cells produce broad-spectrum antibodies that protect against infection early in life. In some instances, antibody production requires MZ B cells to display pathogen antigens bound to major histocompatibility complex class II (MHC II) molecules to T cells. We describe the trogocytic acquisition of these molecules from conventional dendritic cells (cDCs). Complement component 3 (C3) binds to murine and human MHC II on cDCs. MZ B cells recognize C3 with complement receptor 2 (CR2) and trogocytose the MHC II–C3 complexes, which become exposed on their cell surface. The ubiquitin ligase MARCH1 limits the number of MHC II–C3 complexes displayed on cDCs to prevent their elimination through excessive trogocytosis. Capture of C3 by MHC II thus enables the transfer of cDC-like properties to MZ B cells.
- Published
- 2022
- Full Text
- View/download PDF
18. The neurotrophic receptor Ntrk2 directs lymphoid tissue neovascularization during Leishmania donovani infection.
- Author
-
Jane E Dalton, Amy C Glover, Laura Hoodless, Eng-Kiat Lim, Lynette Beattie, Alun Kirby, and Paul M Kaye
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The neurotrophic tyrosine kinase receptor type 2 (Ntrk2, also known as TrkB) and its ligands brain derived neurotrophic factor (Bdnf), neurotrophin-4 (NT-4/5), and neurotrophin-3 (NT-3) are known primarily for their multiple effects on neuronal differentiation and survival. Here, we provide evidence that Ntrk2 plays a role in the pathologic remodeling of the spleen that accompanies chronic infection. We show that in Leishmania donovani-infected mice, Ntrk2 is aberrantly expressed on splenic endothelial cells and that new maturing blood vessels within the white pulp are intimately associated with F4/80(hi)CD11b(lo)CD11c(+) macrophages that express Bdnf and NT-4/5 and have pro-angiogenic potential in vitro. Furthermore, administration of the small molecule Ntrk2 antagonist ANA-12 to infected mice significantly inhibited white pulp neovascularization but had no effect on red pulp vascular remodeling. We believe this to be the first evidence of the Ntrk2/neurotrophin pathway driving pathogen-induced vascular remodeling in lymphoid tissue. These studies highlight the therapeutic potential of modulating this pathway to inhibit pathological angiogenesis.
- Published
- 2015
- Full Text
- View/download PDF
19. A Petri Net Model of Granulomatous Inflammation: Implications for IL-10 Mediated Control of Leishmania donovani Infection.
- Author
-
Luca Albergante, Jon Timmis, Lynette Beattie, and Paul M. Kaye
- Published
- 2013
- Full Text
- View/download PDF
20. Development of Plasmodium-specific liver-resident memory CD8
- Author
-
Sonia, Ghilas, Matthias H, Enders, Rose, May, Lauren E, Holz, Daniel, Fernandez-Ruiz, Anton, Cozijnsen, Vanessa, Mollard, Ian A, Cockburn, Geoffrey I, McFadden, William R, Heath, and Lynette, Beattie
- Subjects
Plasmodium ,Hot Temperature ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Host-Parasite Interactions ,Malaria ,Disease Models, Animal ,Mice ,Liver ,Vaccines, Inactivated ,Malaria Vaccines ,Animals ,Immunization ,Immunologic Memory - Abstract
Malaria remains a major cause of mortality in the world and an efficient vaccine is the best chance of reducing the disease burden. Vaccination strategies for the liver stage of disease that utilise injection of live radiation-attenuated sporozoites (RAS) confer sterile immunity, which is mediated by CD8
- Published
- 2020
21. Author response for 'Development of Plasmodium ‐specific liver resident‐memory CD8 + T cells after heat‐killed sporozoite immunization in mice'
- Author
-
Geoffrey I. McFadden, Matthias H. Enders, Lynette Beattie, Sonia Ghilas, William R. Heath, Rose May, Ian A. Cockburn, Lauren E. Holz, Anton Cozijnsen, Vanessa Mollard, and Daniel Fernandez-Ruiz
- Subjects
Immunization ,biology ,Cytotoxic T cell ,biology.organism_classification ,Plasmodium ,Virology - Published
- 2020
- Full Text
- View/download PDF
22. CD8
- Author
-
Nazanin, Ghazanfari, Julia L, Gregory, Sapna, Devi, Daniel, Fernandez-Ruiz, Lynette, Beattie, Scott N, Mueller, and William R, Heath
- Subjects
CD4-Positive T-Lymphocytes ,Plasmodium berghei ,Malaria, Cerebral ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Parasitemia ,Adoptive Transfer ,Mice, Inbred C57BL ,Disease Models, Animal ,Mice ,Blood-Brain Barrier ,Animals ,Female ,Immunologic Memory ,Spleen - Abstract
In the
- Published
- 2020
23. Glycolipid-peptide vaccination induces liver-resident memory CD8 + T cells that protect against rodent malaria
- Author
-
Patrick Bertolino, Anton Cozijnsen, Stephen J. Turner, Mireille H. Lahoud, Daniel Fernandez-Ruiz, Benjamin J. Compton, Matthias H. Enders, Lauren E. Holz, Geoffrey I. McFadden, Kathryn J. Farrand, Kirsteen M. Tullett, Ana Maria Valencia-Hernandez, Juby Mathew, Ian F. Hermans, Taryn L. Osmond, Dale I. Godfrey, William R. Heath, David G. Bowen, Vanessa Mollard, Rose May, Thiago M. Steiner, Zhongfang Wang, Gavin F. Painter, Joshua Lange, Lynette Beattie, Catarina F. Almeida, Lukasz Kedzierski, Sarah L. Draper, Jasmine Li, Susanna T. S. Chan, Maria N. de Menezes, Yu Cheng Chua, Katherine Kedzierska, Irina Caminschi, Sonia Ghilas, Regan J. Anderson, and Rebecca Seneviratna
- Subjects
0301 basic medicine ,Synthetic vaccine ,biology ,medicine.medical_treatment ,Immunology ,General Medicine ,Natural killer T cell ,Major histocompatibility complex ,Virology ,Epitope ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Cytotoxic T cell ,Adjuvant ,CD8 - Abstract
Liver resident-memory CD8+ T cells (TRM cells) can kill liver-stage Plasmodium-infected cells and prevent malaria, but simple vaccines for generating this important immune population are lacking. Here, we report the development of a fully synthetic self-adjuvanting glycolipid-peptide conjugate vaccine designed to efficiently induce liver TRM cells. Upon cleavage in vivo, the glycolipid-peptide conjugate vaccine releases an MHC I-restricted peptide epitope (to stimulate Plasmodium-specific CD8+ T cells) and an adjuvant component, the NKT cell agonist α-galactosylceramide (α-GalCer). A single dose of this vaccine in mice induced substantial numbers of intrahepatic malaria-specific CD8+ T cells expressing canonical markers of liver TRM cells (CD69, CXCR6, and CD101), and these cells could be further increased in number upon vaccine boosting. We show that modifications to the peptide, such as addition of proteasomal-cleavage sequences or epitope-flanking sequences, or the use of alternative conjugation methods to link the peptide to the glycolipid improved liver TRM cell generation and led to the development of a vaccine able to induce sterile protection in C57BL/6 mice against Plasmodium berghei sporozoite challenge after a single dose. Furthermore, this vaccine induced endogenous liver TRM cells that were long-lived (half-life of ~425 days) and were able to maintain >90% sterile protection to day 200. Our findings describe an ideal synthetic vaccine platform for generating large numbers of liver TRM cells for effective control of liver-stage malaria and, potentially, a variety of other hepatotropic infections.
- Published
- 2020
- Full Text
- View/download PDF
24. IL-10-producing Th1 cells and disease progression are regulated by distinct CD11c⁺ cell populations during visceral leishmaniasis.
- Author
-
Benjamin M J Owens, Lynette Beattie, John W J Moore, Najmeeyah Brown, Jason L Mann, Jane E Dalton, Asher Maroof, and Paul M Kaye
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
IL-10 is a critical regulatory cytokine involved in the pathogenesis of visceral leishmaniasis caused by Leishmania donovani and clinical and experimental data indicate that disease progression is associated with expanded numbers of CD4⁺ IFNγ⁺ T cells committed to IL-10 production. Here, combining conditional cell-specific depletion with adoptive transfer, we demonstrate that only conventional CD11c(hi) DCs that produce both IL-10 and IL-27 are capable of inducing IL-10-producing Th1 cells in vivo. In contrast, CD11c(hi) as well as CD11c(int/lo) cells isolated from infected mice were capable of reversing the host protective effect of diphtheria toxin-mediated CD11c⁺ cell depletion. This was reflected by increased splenomegaly, inhibition of NO production and increased parasite burden. Thus during chronic infection, multiple CD11c⁺ cell populations can actively suppress host resistance and enhance immunopathology, through mechanisms that do not necessarily involve IL-10-producing Th1 cells.
- Published
- 2012
- Full Text
- View/download PDF
25. B cell: T cell interactions occur within hepatic granulomas during experimental visceral leishmaniasis.
- Author
-
John W J Moore, Lynette Beattie, Jane E Dalton, Benjamin M J Owens, Asher Maroof, Mark C Coles, and Paul M Kaye
- Subjects
Medicine ,Science - Abstract
Hepatic resistance to Leishmania donovani infection in mice is associated with the development of granulomas, in which a variety of lymphoid and non-lymphoid populations accumulate. Although previous studies have identified B cells in hepatic granulomas and functional studies in B cell-deficient mice have suggested a role for B cells in the control of experimental visceral leishmaniasis, little is known about the behaviour of B cells in the granuloma microenvironment. Here, we first compared the hepatic B cell population in infected mice, where ≈60% of B cells are located within granulomas, with that of naïve mice. In infected mice, there was a small increase in mIgM(lo)mIgD(+) mature B2 cells, but no enrichment of B cells with regulatory phenotype or function compared to the naïve hepatic B cell population, as assessed by CD1d and CD5 expression and by IL-10 production. Using 2-photon microscopy to quantify the entire intra-granuloma B cell population, in conjunction with the adoptive transfer of polyclonal and HEL-specific BCR-transgenic B cells isolated from L. donovani-infected mice, we demonstrated that B cells accumulate in granulomas over time in an antigen-independent manner. Intra-vital dynamic imaging was used to demonstrate that within the polyclonal B cell population obtained from L. donovani-infected mice, the frequency of B cells that made multiple long contacts with endogenous T cells was greater than that observed using HEL-specific B cells obtained from the same inflammatory environment. These data indicate, therefore, that a subset of this polyclonal B cell population is capable of making cognate interactions with T cells within this unique environment, and provide the first insights into the dynamics of B cells within an inflammatory site.
- Published
- 2012
- Full Text
- View/download PDF
26. Dynamic imaging of experimental Leishmania donovani-induced hepatic granulomas detects Kupffer cell-restricted antigen presentation to antigen-specific CD8 T cells.
- Author
-
Lynette Beattie, Adam Peltan, Asher Maroof, Alun Kirby, Najmeeyah Brown, Mark Coles, Deborah F Smith, and Paul M Kaye
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Kupffer cells (KCs) represent the major phagocytic population within the liver and provide an intracellular niche for the survival of a number of important human pathogens. Although KCs have been extensively studied in vitro, little is known of their in vivo response to infection and their capacity to directly interact with antigen-specific CD8(+) T cells. Here, using a combination of approaches including whole mount and thin section confocal microscopy, adoptive cell transfer and intra-vital 2-photon microscopy, we demonstrate that KCs represent the only detectable population of mononuclear phagocytes within granulomas induced by Leishmania donovani infection that are capable of presenting parasite-derived peptide to effector CD8(+) T cells. This restriction of antigen presentation to KCs within the Leishmania granuloma has important implications for the identification of new candidate vaccine antigens and for the design of novel immuno-therapeutic interventions.
- Published
- 2010
- Full Text
- View/download PDF
27. Innate killing of Leishmania donovani by macrophages of the splenic marginal zone requires IRF-7.
- Author
-
Rebecca Phillips, Mattias Svensson, Naveed Aziz, Asher Maroof, Najmeeyah Brown, Lynette Beattie, Nathalie Signoret, and Paul M Kaye
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Highly phagocytic macrophages line the marginal zone (MZ) of the spleen and the lymph node subcapsular sinus. Although these macrophages have been attributed with a variety of functions, including the uptake and clearance of blood and lymph-borne pathogens, little is known about the effector mechanisms they employ after pathogen uptake. Here, we have combined gene expression profiling and RNAi using a stromal macrophage cell line with in situ analysis of the leishmanicidal activity of marginal zone macrophages (MZM) and marginal metallophilic macrophages (MMM) in wild type and gene targeted mice. Our data demonstrate a critical role for interferon regulatory factor-7 (IRF-7) in regulating the killing of intracellular Leishmania donovani by these specialised splenic macrophage sub-populations. This study, therefore, identifies a new role for IRF-7 as a regulator of innate microbicidal activity against this, and perhaps other, non-viral intracellular pathogens. This study also highlights the importance of selecting appropriate macrophage populations when studying pathogen interactions with this functionally diverse lineage of cells.
- Published
- 2010
- Full Text
- View/download PDF
28. Plasmodium strain determines dendritic cell function essential for survival from malaria.
- Author
-
Michelle N Wykes, Xue Q Liu, Lynette Beattie, Danielle I Stanisic, Katryn J Stacey, Mark J Smyth, Ranjeny Thomas, and Michael F Good
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The severity of malaria can range from asymptomatic to lethal infections involving severe anaemia and cerebral disease. However, the molecular and cellular factors responsible for these differences in disease severity are poorly understood. Identifying the factors that mediate virulence will contribute to developing antiparasitic immune responses. Since immunity is initiated by dendritic cells (DCs), we compared their phenotype and function following infection with either a nonlethal or lethal strain of the rodent parasite, Plasmodium yoelii, to identify their contribution to disease severity. DCs from nonlethal infections were fully functional and capable of secreting cytokines and stimulating T cells. In contrast, DCs from lethal infections were not functional. We then transferred DCs from mice with nonlethal infections to mice given lethal infections and showed that these DCs mediated control of parasitemia and survival. IL-12 was necessary for survival. To our knowledge, our studies have shown for the first time that during a malaria infection, DC function is essential for survival. More importantly, the functions of these DCs are determined by the strain of parasite. Our studies may explain, in part, why natural malaria infections may have different outcomes.
- Published
- 2007
- Full Text
- View/download PDF
29. Glycolipid-peptide vaccination induces liver-resident memory CD8
- Author
-
Lauren E, Holz, Yu Cheng, Chua, Maria N, de Menezes, Regan J, Anderson, Sarah L, Draper, Benjamin J, Compton, Susanna T S, Chan, Juby, Mathew, Jasmine, Li, Lukasz, Kedzierski, Zhongfang, Wang, Lynette, Beattie, Matthias H, Enders, Sonia, Ghilas, Rose, May, Thiago M, Steiner, Joshua, Lange, Daniel, Fernandez-Ruiz, Ana Maria, Valencia-Hernandez, Taryn L, Osmond, Kathryn J, Farrand, Rebecca, Seneviratna, Catarina F, Almeida, Kirsteen M, Tullett, Patrick, Bertolino, David G, Bowen, Anton, Cozijnsen, Vanessa, Mollard, Geoffrey I, McFadden, Irina, Caminschi, Mireille H, Lahoud, Katherine, Kedzierska, Stephen J, Turner, Dale I, Godfrey, Ian F, Hermans, Gavin F, Painter, and William R, Heath
- Subjects
Male ,Mice, Inbred C57BL ,Mice, Knockout ,Mice ,Liver ,Malaria Vaccines ,Vaccination ,Animals ,CD8-Positive T-Lymphocytes ,Glycolipids ,Peptides ,Malaria - Abstract
Liver resident-memory CD8
- Published
- 2019
30. Raster adaptive optics for video rate aberration correction and large FOV multiphoton imaging
- Author
-
Woei Ming Lee, Yean J. Lim, William R. Heath, Elizabeth E. Gardiner, Yongxiao Li, Qiongkai Xu, Lynette Beattie, and Katharina Gaus
- Subjects
Wavefront ,Physics ,0303 health sciences ,Microscope ,Laser scanning ,Image quality ,business.industry ,Image processing ,01 natural sciences ,Atomic and Molecular Physics, and Optics ,Deformable mirror ,Article ,law.invention ,010309 optics ,03 medical and health sciences ,Optics ,law ,0103 physical sciences ,Microscopy ,Adaptive optics ,business ,030304 developmental biology ,Biotechnology - Abstract
Removal of complex aberrations at millisecond time scales over millimeters in distance in multiphoton laser scanning microscopy limits the total spatiotemporal imaging throughput for deep tissue imaging. Using a single low resolution deformable mirror and time multiplexing (TM) adaptive optics, we demonstrate video rate aberration correction (5 ms update rate for a single wavefront mask) for a complex heterogeneous distribution of refractive index differences through a depth of up to 1.1 mm and an extended imaging FOV of up to 0.8 mm, with up to 167% recovery of fluorescence intensity 335 µm from the center of the FOV. The proposed approach, termed raster adaptive optics (RAO), integrates image-based aberration retrieval and video rate removal of arbitrarily defined regions of dominant, spatially varied wavefronts. The extended FOV was achieved by demonstrating rapid recovery of up to 50 distinct wavefront masks at 500 ms update rates that increased imaging throughput by 2.3-fold. Because RAO only requires a single deformable mirror with image-based aberration retrieval, it can be directly implemented on a standard laser scanning multiphoton microscope.
- Published
- 2019
31. A Natural Peptide Antigen within the Plasmodium Ribosomal Protein RPL6 Confers Liver T
- Author
-
Ana Maria, Valencia-Hernandez, Wei Yi, Ng, Nazanin, Ghazanfari, Sonia, Ghilas, Maria N, de Menezes, Lauren E, Holz, Cheng, Huang, Kieran, English, Myo, Naung, Peck Szee, Tan, Kirsteen M, Tullett, Thiago M, Steiner, Matthias H, Enders, Lynette, Beattie, Yu Cheng, Chua, Claerwen M, Jones, Anton, Cozijnsen, Vanessa, Mollard, Yeping, Cai, David G, Bowen, Anthony W, Purcell, Nicole L, La Gruta, Jose A, Villadangos, Tania, de Koning-Ward, Alyssa E, Barry, Winfried, Barchet, Ian A, Cockburn, Geoffrey I, McFadden, Stephanie, Gras, Mireille H, Lahoud, Patrick, Bertolino, Ralf B, Schittenhelm, Irina, Caminschi, William R, Heath, and Daniel, Fernandez-Ruiz
- Subjects
Male ,Ribosomal Proteins ,Immunity, Cellular ,Plasmodium berghei ,Antigens, Protozoan ,Dendritic Cells ,CD8-Positive T-Lymphocytes ,Cell Line ,Malaria ,Mice, Inbred C57BL ,Mice ,Liver ,Sporozoites ,Anopheles ,Malaria Vaccines ,Animals ,Female ,Immunization ,Malaria, Falciparum ,Peptides ,Immunologic Memory - Abstract
Liver-resident memory CD8
- Published
- 2019
32. Type I Interferons Regulate Immune Responses in Humans with Blood-Stage Plasmodium falciparum Infection
- Author
-
Rebecca J. Faleiro, Ric N. Price, Nicholas M. Anstey, Christian R. Engwerda, Marcela Montes de Oca, Susanna S. Ng, Meru Sheel, Julie G. Burel, Rajiv Kumar, Chelsea L. Edwards, Shannon E. Best, Patrick T. Bunn, Glen M. Boyle, Jessica R. Loughland, Ashraful Haque, Fabian de Labastida Rivera, James S. McCarthy, Fiona H. Amante, Lynette Beattie, and Denise L. Doolan
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,medicine.medical_treatment ,T cell ,Plasmodium falciparum ,malaria ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Host-Parasite Interactions ,03 medical and health sciences ,Interferon-gamma ,0302 clinical medicine ,Immune system ,CD4+ T cells ,Immunity ,Th1 cells ,medicine ,Humans ,Malaria, Falciparum ,lcsh:QH301-705.5 ,Innate immune system ,Antiparasitic Agents ,immune regulation ,Tr1 cells ,Vaccine efficacy ,biology.organism_classification ,Virology ,Healthy Volunteers ,Immunity, Innate ,3. Good health ,Interleukin-10 ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,type I interferons ,lcsh:Biology (General) ,Chemoprophylaxis ,Immunology ,Interferon Type I ,030215 immunology - Abstract
Summary The development of immunoregulatory networks is important to prevent disease. However, these same networks allow pathogens to persist and reduce vaccine efficacy. Here, we identify type I interferons (IFNs) as important regulators in developing anti-parasitic immunity in healthy volunteers infected for the first time with Plasmodium falciparum. Type I IFNs suppressed innate immune cell function and parasitic-specific CD4+ T cell IFNγ production, and they promoted the development of parasitic-specific IL-10-producing Th1 (Tr1) cells. Type I IFN-dependent, parasite-specific IL-10 production was also observed in P. falciparum malaria patients in the field following chemoprophylaxis. Parasite-induced IL-10 suppressed inflammatory cytokine production, and IL-10 levels after drug treatment were positively associated with parasite burdens before anti-parasitic drug administration. These findings have important implications for understanding the development of host immune responses following blood-stage P. falciparum infection, and they identify type I IFNs and related signaling pathways as potential targets for therapies or vaccine efficacy improvement.
- Published
- 2016
33. Spatiotemporal Characterization of the Cellular and Molecular Contributors to Liver Fibrosis in a Murine Hepatotoxic-Injury Model
- Author
-
Gregory Miller, Katie E. Lineburg, Andrew D. Clouston, Laetitia Le Texier, Glen M. Boyle, Victoria L. Gadd, Bianca E. Teal, Michelle Melino, Kylie A. Alexander, Kelli P. A. MacDonald, Michelle Martinez, Lynette Beattie, Katharine M. Irvine, Geoffrey R. Hill, and Elizabeth E. Powell
- Subjects
0301 basic medicine ,Macrophage colony-stimulating factor ,CCR2 ,Chemokine ,Pathology ,medicine.medical_specialty ,Inflammation ,Hepatitis, Animal ,Thioacetamide ,CCL2 ,Liver Cirrhosis, Experimental ,Collagen Type I ,Monocytes ,Pathology and Forensic Medicine ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Fibrosis ,medicine ,Animals ,Humans ,Oligonucleotide Array Sequence Analysis ,Liver injury ,biology ,Gene Expression Profiling ,Macrophage Colony-Stimulating Factor ,Macrophages ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Gene Expression Regulation ,Liver ,chemistry ,Disease Progression ,biology.protein ,Female ,Chemokines ,medicine.symptom - Abstract
The interplay between the inflammatory infiltrate and tissue resident cell populations invokes fibrogenesis. However, the temporal and mechanistic contributions of these cells to fibrosis are obscure. To address this issue, liver inflammation, ductular reaction (DR), and fibrosis were induced in C57BL/6 mice by thioacetamide administration for up to 12 weeks. Thioacetamide treatment induced two phases of liver fibrosis. A rapid pericentral inflammatory infiltrate enriched in F4/80(+) monocytes co-localized with SMA(+) myofibroblasts resulted in early collagen deposition, marking the start of an initial fibrotic phase (1 to 6 weeks). An expansion of bone marrow-derived macrophages preceded a second phase, characterized by accelerated progression of fibrosis (>6 weeks) after DR migration from the portal tracts to the centrilobular site of injury, in association with an increase in DR/macrophage interactions. Although chemokine (C-C motif) ligand 2 (CCL2) mRNA was induced rapidly in response to thioacetamide, CCL2 deficiency only partially abrogated fibrosis. In contrast, colony-stimulating factor 1 receptor blockade diminished C-C chemokine receptor type 2 [CCR2(neg) (Ly6C(lo))] monocytes, attenuated the DR, and significantly reduced fibrosis, illustrating the critical role of colony-stimulating factor 1-dependent monocyte/macrophage differentiation and linking the two phases of injury. In response to liver injury, colony-stimulating factor 1 drives early monocyte-mediated myofibroblast activation and collagen deposition, subsequent macrophage differentiation, and their association with the advancing DR, the formation of fibrotic septa, and the progression of liver fibrosis to cirrhosis.
- Published
- 2016
- Full Text
- View/download PDF
34. A Natural Peptide Antigen within the Plasmodium Ribosomal Protein RPL6 Confers Liver TRM Cell-Mediated Immunity against Malaria in Mice
- Author
-
Geoffrey I. McFadden, Mireille H. Lahoud, Kieran English, Irina Caminschi, Sonia Ghilas, Maria N. de Menezes, Wei Yi Ng, Thiago M. Steiner, Matthias H. Enders, Patrick Bertolino, Peck Szee Tan, Ana Maria Valencia-Hernandez, Vanessa Mollard, Lauren E. Holz, Nicole L. La Gruta, Anton Cozijnsen, Jose A Villadangos, Yeping Cai, Daniel Fernandez-Ruiz, David G. Bowen, Kirsteen M. Tullett, Ralf B. Schittenhelm, William R. Heath, Lynette Beattie, Tania F. de Koning-Ward, Claerwen M. Jones, Stephanie Gras, Yu Cheng Chua, Alyssa E. Barry, Anthony W. Purcell, Ian A. Cockburn, Winfried Barchet, Cheng Huang, Myo T. Naung, and Nazanin Ghazanfari
- Subjects
0303 health sciences ,biology ,biology.organism_classification ,Microbiology ,Virology ,Epitope ,Vaccination ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Immunity ,parasitic diseases ,biology.protein ,Cytotoxic T cell ,Parasitology ,Plasmodium berghei ,Antibody ,030217 neurology & neurosurgery ,CD8 ,030304 developmental biology - Abstract
Liver-resident memory CD8+ T (TRM) cells remain in and constantly patrol the liver to elicit rapid immunity upon antigen encounter and can mediate efficient protection against liver-stage Plasmodium infection. This finding has prompted the development of immunization strategies where T cells are activated in the spleen and then trapped in the liver to form TRM cells. Here, we identify PbRPL6120-127, a H2-Kb-restricted epitope from the putative 60S ribosomal protein L6 (RPL6) of Plasmodium berghei ANKA, as an optimal antigen for endogenous liver TRM cell generation and protection against malaria. A single dose vaccination targeting RPL6 provided effective and prolonged sterilizing immunity against high dose sporozoite challenges. Expressed throughout the parasite life cycle, across Plasmodium species, and highly conserved, RPL6 exhibits strong translation potential as a vaccine candidate. This is further advocated by the identification of a broadly conserved, immunogenic HLA-A∗02:01-restricted epitope in P. falciparum RPL6.
- Published
- 2020
- Full Text
- View/download PDF
35. Rapid loss of group 1 innate lymphoid cells during blood stage Plasmodium infection
- Author
-
Meru Sheel, Mark J. Smyth, Fabian de Labastida Rivera, Lynette Beattie, James S. McCarthy, Katherine T. Andrews, Fiona H. Amante, Fernando Souza-Fonseca-Guimaraes, J. Alejandro Lopez, Teija C. M. Frame, Eric Vivier, Rajiv Kumar, Christian R. Engwerda, Marcela Montes de Oca, Yulong Gao, Dale I. Godfrey, Daniel G. Pellicci, Camille Guillerey, Chelsea L. Edwards, Nicholas D. Huntington, Rebecca J. Faleiro, Patrick T. Bunn, Susanna S. Ng, QIMR Berghofer Medical Research Institute, University of Melbourne, Immunology-Hematology, Central Queensland University (CQU), National Centre for Immunisation Research and Surveillance, Centre d'Immunologie de Marseille - Luminy (CIML), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Innate Pharma, Griffith University [Brisbane], and Queensland State GovernmentNational Health and Medical Research Council of Australia (NHMRC)Australia Research Council (ARC)Griffith University, School of Natural SciencesUniversity of Queensland, School of MedicineIndian government Department of Science and TechnologyMedicines for Malaria Venture (MMV)Wellcome TrustBill and Melinda Gates Foundation
- Subjects
0301 basic medicine ,Immunology ,Population ,Spleen ,Inflammation ,Plasmodium chabaudi ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immunity ,parasitic diseases ,medicine ,Immunology and Allergy ,education ,General Nursing ,education.field_of_study ,natural killer cells ,biology ,Innate lymphoid cell ,Plasmodium falciparum ,biology.organism_classification ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,inflammation ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,medicine.symptom ,030215 immunology ,parasitic–protozoan - Abstract
International audience; Objectives. Innate lymphoid cells (ILCs) share many characteristics with CD4(+) T cells, and group 1 ILCs share a requirement for T-bet and the ability to produce IFNc with T helper 1 (Th1) cells. Given this similarity, and the importance of Th1 cells for protection against intracellular protozoan parasites, we aimed to characterise the role of group 1 ILCs during Plasmodium infection. Methods. We quantified group 1 ILCs in peripheral blood collected from subjects infected with with Plasmodium falciparum 3D7 as part of a controlled human malaria infection study, and in the liver and spleens of PcAS-infected mice. We used genetically-modified mouse models, as well as cell-depletion methods in mice to characterise the role of group 1 ILCs during PcAS infection. Results. In a controlled human malaria infection study, we found that the frequencies of circulating ILC1s and NK cells decreased as infection progressed but recovered after volunteers were treated with antiparasitic drug. A similar observation was made for liver and splenic ILC1s in P. chabaudi chabaudi AS (PcAS)-infected mice. The decrease in mouse liver ILC1 frequencies was associated with increased apoptosis. We also identified a population of cells within the liver and spleen that expressed both ILC1 and NK cell markers, indicative of plasticity between these two cell lineages. Studies using genetic and cell-depletion approaches indicated that group 1 ILCs have a limited role in antiparasitic immunity during PcAS infection in mice. Discussion. Our results are consistent with a previous study indicating a limited role for natural killer (NK) cells during Plasmodium chabaudi infection in mice. Additionally, a recent study reported the redundancy of ILCs in humans with competent B and T cells. Nonetheless, our results do not rule out a role for group 1 ILCs in human malaria in endemic settings given that blood stage infection was initiated intravenously in our experimental models, and thus bypassed the liver stage of infection, which may influence the immune response during the blood stage. Conclusion. Our results show that ILC1s are lost early during mouse and human malaria, and this observation may help to explain the limited role for these cells in controlling blood stage infection.
- Published
- 2018
- Full Text
- View/download PDF
36. IL-17A–Producing γδ T Cells Suppress Early Control of Parasite Growth by Monocytes in the Liver
- Author
-
Christian R. Engwerda, Shaun R. McColl, Ashraful Haque, Rachel D. Kuns, Patrick T. Bunn, Marcela Montes de Oca, Antiopi Varelias, Fabian de Labastida Rivera, Meru Sheel, Fiona H. Amante, Kelli P. A. MacDonald, Rebecca J. Faleiro, Geoff R. Hill, Mark J. Smyth, Lynette Beattie, Shannon E. Best, Rajiv Kumar, Chelsea L. Edwards, Teija C. M. Frame, and Susanna S. Ng
- Subjects
Receptors, CCR2 ,T-Lymphocytes ,T cell ,Immunology ,Leishmania donovani ,Monocytes ,Proinflammatory cytokine ,Mice ,Immune system ,Antigen ,medicine ,Animals ,Humans ,Immunology and Allergy ,Immunosuppression Therapy ,biology ,Superoxide Dismutase ,Intracellular parasite ,Interleukin-17 ,Receptors, Antigen, T-Cell, gamma-delta ,medicine.disease ,biology.organism_classification ,Mice, Inbred C57BL ,Disease Models, Animal ,Visceral leishmaniasis ,medicine.anatomical_structure ,Liver ,Leishmaniasis, Visceral ,Interleukin 17 - Abstract
Intracellular infections, such as those caused by the protozoan parasite Leishmania donovani, a causative agent of visceral leishmaniasis (VL), require a potent host proinflammatory response for control. IL-17 has emerged as an important proinflammatory cytokine required for limiting growth of both extracellular and intracellular pathogens. However, there are conflicting reports on the exact roles for IL-17 during parasitic infections and limited knowledge about cellular sources and the immune pathways it modulates. We examined the role of IL-17 in an experimental model of VL caused by infection of C57BL/6 mice with L. donovani and identified an early suppressive role for IL-17 in the liver that limited control of parasite growth. IL-17–producing γδ T cells recruited to the liver in the first week of infection were the critical source of IL-17 in this model, and CCR2+ inflammatory monocytes were an important target for the suppressive effects of IL-17. Improved parasite control was independent of NO generation, but associated with maintenance of superoxide dismutase mRNA expression in the absence of IL-17 in the liver. Thus, we have identified a novel inhibitory function for IL-17 in parasitic infection, and our results demonstrate important interactions among γδ T cells, monocytes, and infected macrophages in the liver that can determine the outcome of parasitic infection.
- Published
- 2015
- Full Text
- View/download PDF
37. Bile canalicular dynamics in hepatocyte sandwich cultures
- Author
-
Raymond Reif, Amruta Vartak, Paul M. Kaye, Lynette Beattie, Jan G. Hengstler, Georgia Günther, Johan Karlsson, Simone Melega, Brigitte Begher-Tibbe, Mats Jirstrand, Seddik Hammad, and David Wrangborg
- Subjects
Male ,medicine.medical_specialty ,Health, Toxicology and Mutagenesis ,Cell Culture Techniques ,Cholestasis, Intrahepatic ,Biology ,Toxicology ,Bone canaliculus ,digestive system ,Dexamethasone ,Excretion ,Mice ,chemistry.chemical_compound ,Cholestasis ,In vivo ,Internal medicine ,medicine ,Animals ,Insulin ,Fluorescein ,Cells, Cultured ,Bile Canaliculi ,General Medicine ,Fluoresceins ,medicine.disease ,In vitro ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Cell culture ,Hepatocyte ,Hepatocytes ,Collagen ,Chemical and Drug Induced Liver Injury - Abstract
Many substances are hepatotoxic due to their ability to cause intrahepatic cholestasis. Therefore, there is a high demand for in vitro systems for the identification of cholestatic properties of new compounds. Primary hepatocytes cultivated in collagen sandwich cultures are known to establish bile canaliculi which enclose secreted biliary components. Cholestatic compounds are mainly known to inhibit bile excretion dynamics, but may also alter canalicular volume, or hepatocellular morphology. So far, techniques to assess time-resolved morphological changes of bile canaliculi in sandwich cultures are not available. In this study, we developed an automated system that quantifies dynamics of bile canaliculi recorded in conventional time-lapse image sequences. We validated the hepatocyte sandwich culture system as an appropriate model to study bile canaliculi in vitro by showing structural similarity measured as bile canaliculi length per hepatocyte to that observed in vivo. Moreover, bile canalicular excretion kinetics of CMFDA (5-chloromethylfluorescein diacetate) in sandwich cultures resembled closely the kinetics observed in vivo. The developed quantification technique enabled the quantification of dynamic changes in individual bile canaliculi. With this technique, we were able to clearly distinguish between sandwich cultures supplemented with dexamethasone and insulin from control cultures. In conclusion, the automated quantification system offers the possibility to systematically study the causal relationship between disturbed bile canalicular dynamics and cholestasis.
- Published
- 2015
- Full Text
- View/download PDF
38. Rapid loss of group 1 innate lymphoid cells during blood stage
- Author
-
Susanna S, Ng, Fernando, Souza-Fonseca-Guimaraes, Fabian de Labastida, Rivera, Fiona H, Amante, Rajiv, Kumar, Yulong, Gao, Meru, Sheel, Lynette, Beattie, Marcela, Montes de Oca, Camille, Guillerey, Chelsea L, Edwards, Rebecca J, Faleiro, Teija, Frame, Patrick T, Bunn, Eric, Vivier, Dale I, Godfrey, Daniel G, Pellicci, J Alejandro, Lopez, Katherine T, Andrews, Nicholas D, Huntington, Mark J, Smyth, James, McCarthy, and Christian R, Engwerda
- Subjects
natural killer cells ,inflammation ,parasitic diseases ,Original Article ,Original Articles ,parasitic–protozoan - Abstract
Objectives Innate lymphoid cells (ILCs) share many characteristics with CD4+ T cells, and group 1 ILCs share a requirement for T‐bet and the ability to produce IFNγ with T helper 1 (Th1) cells. Given this similarity, and the importance of Th1 cells for protection against intracellular protozoan parasites, we aimed to characterise the role of group 1 ILCs during Plasmodium infection. Methods We quantified group 1 ILCs in peripheral blood collected from subjects infected with with Plasmodium falciparum 3D7 as part of a controlled human malaria infection study, and in the liver and spleens of Pc AS‐infected mice. We used genetically‐modified mouse models, as well as cell‐depletion methods in mice to characterise the role of group 1 ILCs during Pc AS infection. Results In a controlled human malaria infection study, we found that the frequencies of circulating ILC1s and NK cells decreased as infection progressed but recovered after volunteers were treated with antiparasitic drug. A similar observation was made for liver and splenic ILC1s in P. chabaudi chabaudi AS (Pc AS)‐infected mice. The decrease in mouse liver ILC1 frequencies was associated with increased apoptosis. We also identified a population of cells within the liver and spleen that expressed both ILC1 and NK cell markers, indicative of plasticity between these two cell lineages. Studies using genetic and cell‐depletion approaches indicated that group 1 ILCs have a limited role in antiparasitic immunity during Pc AS infection in mice. Discussion Our results are consistent with a previous study indicating a limited role for natural killer (NK) cells during Plasmodium chabaudi infection in mice. Additionally, a recent study reported the redundancy of ILCs in humans with competent B and T cells. Nonetheless, our results do not rule out a role for group 1 ILCs in human malaria in endemic settings given that blood stage infection was initiated intravenously in our experimental models, and thus bypassed the liver stage of infection, which may influence the immune response during the blood stage. Conclusion Our results show that ILC1s are lost early during mouse and human malaria, and this observation may help to explain the limited role for these cells in controlling blood stage infection.
- Published
- 2017
39. IFNAR1-Signalling Obstructs ICOS-mediated Humoral Immunity during Non-lethal Blood-Stage Plasmodium Infection
- Author
-
Paul J. Hertzog, Fabian de Labastida Rivera, Ashraful Haque, Andreas Hutloff, Megan S. F. Soon, Lynette Beattie, Marcela Montes de Oca, Fiona H. Amante, Bryce S. Thomas, Ismail Sebina, Fernando Souza-Fonseca-Guimaraes, Christian R. Engwerda, Geoffrey R. Hill, Lily Fogg, Shannon E. Best, Mark J. Smyth, and Kylie R. James
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,B Cells ,Humoral Immune Response ,Antibodies, Protozoan ,Receptor, Interferon alpha-beta ,Lymphocyte Activation ,Plasmodium chabaudi ,White Blood Cells ,Mice ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Immune Response ,lcsh:QH301-705.5 ,Protozoans ,Mice, Knockout ,B-Lymphocytes ,Microscopy, Confocal ,biology ,T Cells ,Malarial Parasites ,Acquired immune system ,Flow Cytometry ,Female ,Antibody ,Cellular Types ,Plasmodium yoelii ,Research Article ,Signal Transduction ,lcsh:Immunologic diseases. Allergy ,Immune Cells ,Immunology ,Enzyme-Linked Immunosorbent Assay ,Microbiology ,Inducible T-Cell Co-Stimulator Protein ,03 medical and health sciences ,Immune system ,Immunity ,Virology ,Genetics ,Antigenic variation ,Parasitic Diseases ,Animals ,Antibody-Producing Cells ,Molecular Biology ,Blood Cells ,Organisms ,Biology and Life Sciences ,Cell Biology ,biology.organism_classification ,Tropical Diseases ,Parasitic Protozoans ,Malaria ,Immunity, Humoral ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,lcsh:Biology (General) ,Humoral immunity ,Humoral Immunity ,biology.protein ,Parasitology ,lcsh:RC581-607 ,030215 immunology - Abstract
Parasite-specific antibodies protect against blood-stage Plasmodium infection. However, in malaria-endemic regions, it takes many months for naturally-exposed individuals to develop robust humoral immunity. Explanations for this have focused on antigenic variation by Plasmodium, but have considered less whether host production of parasite-specific antibody is sub-optimal. In particular, it is unclear whether host immune factors might limit antibody responses. Here, we explored the effect of Type I Interferon signalling via IFNAR1 on CD4+ T-cell and B-cell responses in two non-lethal murine models of malaria, P. chabaudi chabaudi AS (PcAS) and P. yoelii 17XNL (Py17XNL) infection. Firstly, we demonstrated that CD4+ T-cells and ICOS-signalling were crucial for generating germinal centre (GC) B-cells, plasmablasts and parasite-specific antibodies, and likewise that T follicular helper (Tfh) cell responses relied on B cells. Next, we found that IFNAR1-signalling impeded the resolution of non-lethal blood-stage infection, which was associated with impaired production of parasite-specific IgM and several IgG sub-classes. Consistent with this, GC B-cell formation, Ig-class switching, plasmablast and Tfh differentiation were all impaired by IFNAR1-signalling. IFNAR1-signalling proceeded via conventional dendritic cells, and acted early by limiting activation, proliferation and ICOS expression by CD4+ T-cells, by restricting the localization of activated CD4+ T-cells adjacent to and within B-cell areas of the spleen, and by simultaneously suppressing Th1 and Tfh responses. Finally, IFNAR1-deficiency accelerated humoral immune responses and parasite control by boosting ICOS-signalling. Thus, we provide evidence of a host innate cytokine response that impedes the onset of humoral immunity during experimental malaria., Author Summary Plasmodium parasites cause malaria by invading, replicating within, and rupturing out of red blood cells. Natural immunity to malaria, which depends on generating Plasmodium-specific antibodies, often takes years to develop. Explanations for this focus on antigenic variation by the parasite, but consider less whether antibody responses themselves may be sub-optimal. Surprisingly little is known about how Plasmodium-specific antibody responses are generated in the host, and whether these can be enhanced. Using mouse models, we found that cytokine-signalling via the receptor IFNAR1 delayed the production of Plasmodium-specific antibody responses. IFNAR1-signalling hindered the resolution of infection, and acted early via conventional dendritic cells to restrict CD4+ T-cell activation and their interactions with B-cells. Thus, we reveal that an innate cytokine response, which occurs during blood-stage Plasmodium infection in humans, obstructs the onset of antibody–mediated immunity during experimental malaria.
- Published
- 2016
40. Correction: Blimp-1-Dependent IL-10 Production by Tr1 Cells Regulates TNF-Mediated Tissue Pathology
- Author
-
Fabian de Labastida Rivera, Shannon E. Best, Meru Sheel, Axel Kallies, Fiona H. Amante, Rajesh Kumar, Marcela Montes de Oca, Erika Cretney, Mark J. Smyth, Werner Müller, Lynette Beattie, Geoffrey R. Hill, Shyam Sundar, Christian R. Engwerda, Chelsea L. Edwards, Rebecca J. Faleiro, Ashraful Haque, Stephen L. Nutt, Patrick T. Bunn, and Susanna S. Ng
- Subjects
Male ,0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,medicine.medical_specialty ,Immunology ,T-Lymphocytes, Regulatory ,Microbiology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,lcsh:QH301-705.5 ,health care economics and organizations ,Inflammation ,Tumor Necrosis Factor-alpha ,business.industry ,Correction ,Th1 Cells ,Flow Cytometry ,Mice, Mutant Strains ,Interleukin-10 ,Malaria ,Mice, Inbred C57BL ,Repressor Proteins ,Disease Models, Animal ,Interleukin 10 ,030104 developmental biology ,Microscopy, Fluorescence ,lcsh:Biology (General) ,Research centre ,Tropical medicine ,Leishmaniasis, Visceral ,Female ,Parasitology ,Tumor necrosis factor alpha ,Positive Regulatory Domain I-Binding Factor 1 ,business ,lcsh:RC581-607 ,030215 immunology - Abstract
Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation.
- Published
- 2016
41. Combined Immune Therapy for the Treatment of Visceral Leishmaniasis
- Author
-
Shannon E. Best, Ashraful Haque, Susanna S. Ng, Shyam Sundar, Patrick T. Bunn, Louise M. Hafner, Fiona H. Amante, Rebecca J. Faleiro, Shashi Bhushan Chauhan, Meru Sheel, Susanne Nylén, Christian R. Engwerda, David B. Sacks, Chelsea L. Edwards, Lynette Beattie, Neetu Singh, Marcela Montes de Oca, and Rajiv Kumar
- Subjects
0301 basic medicine ,Physiology ,medicine.medical_treatment ,White Blood Cells ,Mice ,Animal Cells ,Antiparasitic Therapy ,Immune Physiology ,Zoonoses ,Medicine and Health Sciences ,Public and Occupational Health ,Immune Response ,Leishmaniasis ,biology ,Pharmaceutics ,T Cells ,lcsh:Public aspects of medicine ,Vaccination and Immunization ,Interleukin-10 ,3. Good health ,Infectious Diseases ,Cytokines ,Leishmaniasis, Visceral ,Female ,Tumor necrosis factor alpha ,Immunotherapy ,Cellular Types ,medicine.symptom ,Research Article ,Neglected Tropical Diseases ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Immune Cells ,Immunology ,Leishmania donovani ,Inflammation ,03 medical and health sciences ,Immune system ,Drug Therapy ,Immunity ,Parasitic Diseases ,medicine ,Animals ,Humans ,T Helper Cells ,Blood Cells ,Protozoan Infections ,Public Health, Environmental and Occupational Health ,Biology and Life Sciences ,lcsh:RA1-1270 ,Cell Biology ,Th1 Cells ,Tropical Diseases ,biology.organism_classification ,medicine.disease ,Immune checkpoint ,Mice, Inbred C57BL ,030104 developmental biology ,Visceral leishmaniasis ,Preventive Medicine ,Spleen - Abstract
Chronic disease caused by infections, cancer or autoimmunity can result in profound immune suppression. Immunoregulatory networks are established to prevent tissue damage caused by inflammation. Although these immune checkpoints preserve tissue function, they allow pathogens and tumors to persist, and even expand. Immune checkpoint blockade has recently been successfully employed to treat cancer. This strategy modulates immunoregulatory mechanisms to allow host immune cells to kill or control tumors. However, the utility of this approach for controlling established infections has not been extensively investigated. Here, we examined the potential of modulating glucocorticoid-induced TNF receptor-related protein (GITR) on T cells to improve anti-parasitic immunity in blood and spleen tissue from visceral leishmaniasis (VL) patients infected with Leishmania donovani. We found little effect on parasite growth or parasite-specific IFNγ production. However, this treatment reversed the improved anti-parasitic immunity achieved by IL-10 signaling blockade. Further investigations using an experimental VL model caused by infection of C57BL/6 mice with L. donovani revealed that this negative effect was prominent in the liver, dependent on parasite burden and associated with an accumulation of Th1 cells expressing high levels of KLRG-1. Nevertheless, combined anti-IL-10 and anti-GITR mAb treatment could improve anti-parasitic immunity when used with sub-optimal doses of anti-parasitic drug. However, additional studies with VL patient samples indicated that targeting GITR had no overall benefit over IL-10 signaling blockade alone at improving anti-parasitic immune responses, even with drug treatment cover. These findings identify several important factors that influence the effectiveness of immune modulation, including parasite burden, target tissue and the use of anti-parasitic drug. Critically, these results also highlight potential negative effects of combining different immune modulation strategies., Author Summary Despite a decline in the numbers of visceral leishmaniasis (VL) cases and the availability of drugs to treat patients, there is still a need for more efficient treatment options. This is especially important as many VL endemic areas enter phases of disease elimination where parasite burdens need to be reduced as far as possible to minimize the risk of future parasite transmission. Immunotherapy involving the activation of anti-parasitic immunity in infected individuals is a promising approach to help achieve these goals. We tested whether immune modulation could improve anti-parasitic immunity using VL patient samples and an experimental mouse model of infection. Our results provide guidance on the utility of the experimental model, as well as highlighting potential problems associated with immune modulation to improve disease outcomes. These findings have implications for understanding how immunotherapy should be tested and implemented.
- Published
- 2016
42. Blimp-1-Dependent IL-10 Production by Tr1 Cells Regulates TNF-Mediated Tissue Pathology
- Author
-
Ashraful Haque, Fiona H. Amante, Stephen L. Nutt, Werner Müller, Chelsea L. Edwards, Susanna S. Ng, Christian R. Engwerda, Marcela Montes de Oca, Rebecca J. Faleiro, Erika Cretney, Patrick T. Bunn, Meru Sheel, Lynette Beattie, Shannon E. Best, Fabian de Labastida Rivera, Geoffrey R. Hill, Shyam Sundar, Axel Kallies, Rajiv Kumar, and Mark J. Smyth
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Cell ,Inflammation ,Biology ,Microbiology ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virology ,Genetics ,medicine ,Interferon gamma ,lcsh:QH301-705.5 ,Molecular Biology ,medicine.diagnostic_test ,Interleukin 10 ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,lcsh:Biology (General) ,Parasitology ,Tumor necrosis factor alpha ,medicine.symptom ,lcsh:RC581-607 ,Research Article ,030215 immunology ,medicine.drug - Abstract
Tumor necrosis factor (TNF) is critical for controlling many intracellular infections, but can also contribute to inflammation. It can promote the destruction of important cell populations and trigger dramatic tissue remodeling following establishment of chronic disease. Therefore, a better understanding of TNF regulation is needed to allow pathogen control without causing or exacerbating disease. IL-10 is an important regulatory cytokine with broad activities, including the suppression of inflammation. IL-10 is produced by different immune cells; however, its regulation and function appears to be cell-specific and context-dependent. Recently, IL-10 produced by Th1 (Tr1) cells was shown to protect host tissues from inflammation induced following infection. Here, we identify a novel pathway of TNF regulation by IL-10 from Tr1 cells during parasitic infection. We report elevated Blimp-1 mRNA levels in CD4+ T cells from visceral leishmaniasis (VL) patients, and demonstrate IL-12 was essential for Blimp-1 expression and Tr1 cell development in experimental VL. Critically, we show Blimp-1-dependent IL-10 production by Tr1 cells prevents tissue damage caused by IFNγ-dependent TNF production. Therefore, we identify Blimp-1-dependent IL-10 produced by Tr1 cells as a key regulator of TNF-mediated pathology and identify Tr1 cells as potential therapeutic tools to control inflammation., Author Summary Many parasitic diseases are associated with the generation of potent inflammatory responses. These are often needed to control infection, but can also cause tissue damage if not appropriately regulated. IL-10 has emerged as an important immune regulator that protects tissues by dampening inflammation. Recently, some T cells that initially produce inflammatory cytokines have been found to start producing IL-10 as a mechanism of auto-regulation. We identified an important transcriptional regulator called B lymphocyte-induced maturation protein 1 (Blimp-1), which promotes IL-10 production by IFNγ-producing CD4+ T (Tr1) cells during malaria and visceral leishmaniasis, two important diseases caused by protozoan parasites. We found that Tr1 cell-derived IL-10 suppressed anti-parasitic immunity, but played a critical role in preventing tissue damage caused by the potent pro-inflammatory cytokine TNF. Specifically, IL-10 protected macrophages from TNF-mediated destruction, and this enabled lymphocytes to continue to migrate to regions in the spleen where T and B cell responses are generated. These findings allow us to better understand how parasites persist in a host, but also identify new opportunities to control inflammation to prevent disease.
- Published
- 2016
- Full Text
- View/download PDF
43. In vivo imaging of systemic transport and elimination of xenobiotics and endogenous molecules in mice
- Author
-
Raymond Reif, Lars Kuepfer, Ahmed Ghallab, Georgia Günther, Jan G. Hengstler, Paul M. Kaye, and Lynette Beattie
- Subjects
0301 basic medicine ,Male ,Kupffer Cells ,Health, Toxicology and Mutagenesis ,Video Recording ,Mice, Transgenic ,Biology ,Bone canaliculus ,Toxicology ,Kidney ,Xenobiotics ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,In vivo ,Molecular Toxicology ,Bile canaliculi ,Intravital imaging ,medicine ,Animals ,Hepatocyte ,Renal tubules ,Acetaminophen ,Microscopy ,Cholic acid ,Drug transport ,General Medicine ,Small intestine ,3. Good health ,Cell biology ,Intestines ,030104 developmental biology ,medicine.anatomical_structure ,Biochemistry ,chemistry ,Liver ,Hepatocytes ,Glomerulus ,Lacteal ,030211 gastroenterology & hepatology ,Lymph ,Chemical and Drug Induced Liver Injury ,Preclinical imaging - Abstract
We describe a two-photon microscopy-based method to evaluate the in vivo systemic transport of compounds. This method comprises imaging of the intact liver, kidney and intestine, the main organs responsible for uptake and elimination of xenobiotics and endogenous molecules. The image quality of the acquired movies was sufficient to distinguish subcellular structures like organelles and vesicles. Quantification of the movement of fluorescent dextran and fluorescent cholic acid derivatives in different organs and their sub-compartments over time revealed significant dynamic differences. Calculated half-lives were similar in the capillaries of all investigated organs but differed in the specific sub-compartments, such as parenchymal cells and bile canaliculi of the liver, glomeruli, proximal and distal tubules of the kidney and lymph vessels (lacteals) of the small intestine. Moreover, tools to image immune cells, which can influence transport processes in inflamed tissues, are described. This powerful approach provides new possibilities for the analysis of compound transport in multiple organs and can support physiologically based pharmacokinetic modeling, in order to obtain more precise predictions at the whole body scale. Electronic supplementary material The online version of this article (doi:10.1007/s00204-016-1906-5) contains supplementary material, which is available to authorized users.
- Published
- 2016
- Full Text
- View/download PDF
44. Leishmania-host interactions: what has imaging taught us?
- Author
-
Paul M. Kaye and Lynette Beattie
- Subjects
Innate immune system ,Live cell imaging ,Virology ,Immunology ,Computational biology ,WHOLE ANIMAL ,Life history ,Biology ,Leishmania ,biology.organism_classification ,Microbiology - Abstract
Summary Leishmania parasites are well adapted to initiate infection, resist the onslaught of innate immunity and achieve a state of long-lived persistence. In recent years, the tools available to study these interactions have developed enormously and have become much more widely available. Confocal microscopy, live cell imaging, whole animal imaging and intra-vital 2-photon now complement and extend the classical light and electron microscopical techniques. Coupled with approaches to generate transgenic parasites that express imaging friendly reporter proteins, these tools are making the full breadth of the life cycle accessible to imaging studies. New insights into the life history of these highly successful parasites are emerging with increasing frequency, and often with startling clarity and visual drama. In this short review, we focus on how this new generation of imaging-based research tools has augmented our understanding of the complex interplay that occurs between Leishmania and the cells that it infects in mammalian hosts.
- Published
- 2011
- Full Text
- View/download PDF
45. Leishmania donovani-induced expression of signal regulatory protein α on Kupffer cells enhances hepatic invariant NKT-cell activation
- Author
-
Paul M. Kaye, Lynette Beattie, Alison Bune, Mattias Svensson, Soombul Zubairi, Katharine R. Smith, Najmeeyah Brown, and Asher Maroof
- Subjects
Kupffer Cells ,medicine.medical_treatment ,Immunology ,Leishmania donovani ,CD47 Antigen ,Biology ,Lymphocyte Activation ,Signal regulatory protein α ,Immune tolerance ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Interferon ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,Receptors, Immunologic ,CD47 ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Immunity to Infection ,Natural killer T cell ,biology.organism_classification ,3. Good health ,Cell biology ,Cytokine ,Liver ,Invariant NKT cells ,Leishmaniasis, Visceral ,Natural Killer T-Cells ,Signal transduction ,030215 immunology ,medicine.drug - Abstract
Signal regulatory protein alpha (SIRPalpha) and its cognate ligand CD47 have been documented to have a broad range of cellular functions in development and immunity. Here, we investigated the role of SIRPalpha-CD47 signalling in invariant NKT (iNKT) cell responses. We found that CD47 was required for the optimal production of IFN-gamma from splenic iNKT cells following exposure to the alphaGalCer analogue PBS-57 and in vivo infection of mice with Leishmania donovani. Surprisingly, although SIRPalpha was undetectable in the liver of uninfected mice, the hepatic iNKT-cell response to infection was also impaired in CD47-/- mice. However, we found that SIRPalpha was rapidly induced on Kupffer cells following L. donovani infection, via a mechanism involving G-protein-coupled receptors. Thus, we describe a novel amplification pathway affecting cytokine production by hepatic iNKT cells, which may facilitate the breakdown of hepatic tolerance after infection.
- Published
- 2009
- Full Text
- View/download PDF
46. Posttranscriptional Regulation of Il10 Gene Expression Allows Natural Killer Cells to Express Immunoregulatory Function
- Author
-
Simona Stager, Paul M. Kaye, Lynette Beattie, Soombul Zubairi, Mattias Svensson, and Asher Maroof
- Subjects
CD4-Positive T-Lymphocytes ,Immunology ,Gene Expression ,Biology ,Article ,CD49b ,Mice ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,Immunity ,Animals ,Immunology and Allergy ,RNA, Messenger ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,Granuloma ,Lymphokine-activated killer cell ,Macrophages ,Janus kinase 3 ,Natural killer T cell ,Interleukin-10 ,3. Good health ,Killer Cells, Natural ,Mice, Inbred C57BL ,Interleukin 10 ,Infectious Diseases ,Liver ,CELLIMMUNO ,Interleukin 12 ,Leishmaniasis, Visceral ,Leishmania donovani ,030215 immunology - Abstract
Natural killer (NK) cells play a well-recognized role in early pathogen containment and in shaping acquired cell-mediated immunity. However, indirect evidence in humans and experimental models has suggested that NK cells also play negative regulatory roles during chronic disease. To formally test this hypothesis, we employed a well-defined experimental model of visceral leishmaniasis. Our data demonstrated that NKp46(+)CD49b(+)CD3(-) NK cells were recruited to the spleen and into hepatic granulomas, where they inhibited host protective immunity in an interleukin-10 (IL-10)-dependent manner. Although IL-10 mRNA could be detected in activated NK cells 24 hr after infection, the inhibitory function of NK cells was only acquired later during infection, coincident with increased IL-10 mRNA stability and an enhanced capacity to secrete IL-10 protein. Our data support a growing body of literature that implicates NK cells as negative regulators of cell-mediated immunity and suggest that NK cells, like CD4(+) T helper 1 cells, may acquire immunoregulatory functions as a consequence of extensive activation.
- Published
- 2008
- Full Text
- View/download PDF
47. Transgenic Leishmania and the immune response to infection
- Author
-
Paul M. Kaye, Lynette Beattie, Krystal J. Evans, and Deborah F. Smith
- Subjects
Leishmania ,Intracellular parasite ,Transgene ,Immunology ,Virulence ,Biology ,biology.organism_classification ,Article ,Host-Parasite Interactions ,Animals, Genetically Modified ,Immune system ,RNA interference ,Animals ,Humans ,Parasite hosting ,Parasitology ,Leishmaniasis ,Gene - Abstract
SUMMARY Genetic manipulation of single-celled organisms such as the Leishmania parasite enables in depth analysis of the consequences of genotypic change on biological function. In probing the immune responses to infection, use of transgenic Leishmania has the potential to unravel both the contribution of the parasite to the infection process and the cellular interactions and mechanisms that characterize the innate and adaptive immune responses of the host. Here, we briefly review recent technical advances in parasite genetics and explore how these methods are being used to investigate parasite virulence factors, elucidate immune regulatory mechanisms and contribute to the development of novel therapeutics for the leishmaniases. Recent developments in imaging technology, such as bioluminescence and intravital imaging, combined with parasite transfection with fluorescent or enzyme-encoding marker genes, provides a rich opportunity for novel assessment of intimate, real-time host–parasite interactions at a previously unexplored level. Further advances in transgenic technology, such as the introduction of robust inducible gene cassettes for expression in intracellular parasite stages or the development of RNA interference methods for down-regulation of parasite gene expression in the host, will further advance our ability to probe host–parasite interactions and unravel disease-promoting mechanisms in the leishmaniases.
- Published
- 2008
- Full Text
- View/download PDF
48. Lessons from other diseases: granulomatous inflammation in leishmaniasis
- Author
-
Paul M. Kaye and Lynette Beattie
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Chemokine ,Lymphoid Tissue ,Immunology ,Antiprotozoal Agents ,Review ,Mouse models ,03 medical and health sciences ,Mice ,Immune system ,hemic and lymphatic diseases ,Intravital imaging ,medicine ,Immunology and Allergy ,Animals ,Humans ,Computational models ,Leishmaniasis ,Mononuclear Phagocyte System ,Leishmania ,Granuloma ,biology ,business.industry ,Mononuclear phagocyte system ,medicine.disease ,biology.organism_classification ,3. Good health ,030104 developmental biology ,Lymphatic system ,Visceral leishmaniasis ,Liver ,Immune System ,biology.protein ,Cytokines ,Chemokines ,business - Abstract
The Leishmania granuloma shares some, though not all, properties with that formed following mycobacterial infection. As a simplified, noncaseating granuloma composed of relatively few and largely mononuclear cell populations, it provides a tractable model system to investigate intra-granuloma cellular dynamics, immune regulation, and antimicrobial resistance. Here, the occurrence of granulomatous pathology across the spectrum of leishmaniasis, in humans and animal reservoir hosts, is first described. However, this review focuses on the process of hepatic granuloma formation as studied in rodent models of visceral leishmaniasis, starting from the initial infection of Kupffer cells to the involution of the granuloma after pathogen clearance. It describes how the application of intravital imaging and the use of computational modeling have changed some of our thoughts on granuloma function, and illustrates how host-directed therapies have been used to manipulate granuloma form and function for therapeutic benefit. Where appropriate, lessons that may be equally applicable across the spectrum of granulomatous diseases are highlighted. Electronic supplementary material The online version of this article (doi:10.1007/s00281-015-0548-7) contains supplementary material, which is available to authorized users.
- Published
- 2015
49. Potencies of Human Immunodeficiency Virus Protease Inhibitors In Vitro against Plasmodium falciparum and In Vivo against Murine Malaria
- Author
-
James S. McCarthy, Colin Berry, Robert Reid, Martin J. Stoermer, Lynette Beattie, Donald L. Gardiner, Petrina M. Hilton, Tina S. Skinner-Adams, David P. Fairlie, John E. Ray, Lewis A. Melville, Praveen K. Madala, David Wyatt, and Katherine T. Andrews
- Subjects
Protein Conformation ,Plasmodium falciparum ,Protozoan Proteins ,Plasmepsin ,Plasmodium chabaudi ,Hemoglobins ,Mice ,HIV-1 protease ,parasitic diseases ,medicine ,Animals ,Aspartic Acid Endopeptidases ,HIV Protease Inhibitor ,Pharmacology (medical) ,Pharmacology ,Binding Sites ,biology ,virus diseases ,Lopinavir ,HIV Protease Inhibitors ,biology.organism_classification ,Virology ,Malaria ,Mice, Inbred C57BL ,Infectious Diseases ,Susceptibility ,biology.protein ,Female ,Ritonavir ,Crystallization ,Saquinavir ,medicine.drug - Abstract
Parasite resistance to antimalarial drugs is a serious threat to human health, and novel agents that act on enzymes essential for parasite metabolism, such as proteases, are attractive targets for drug development. Recent studies have shown that clinically utilized human immunodeficiency virus (HIV) protease inhibitors can inhibit the in vitro growth of Plasmodium falciparum at or below concentrations found in human plasma after oral drug administration. The most potent in vitro antimalarial effects have been obtained for parasites treated with saquinavir, ritonavir, or lopinavir, findings confirmed in this study for a genetically distinct P. falciparum line (3D7). To investigate the potential in vivo activity of antiretroviral protease inhibitors (ARPIs) against malaria, we examined the effect of ARPI combinations in a murine model of malaria. In mice infected with Plasmodium chabaudi AS and treated orally with ritonavir-saquinavir or ritonavir-lopinavir, a delay in patency and a significant attenuation of parasitemia were observed. Using modeling and ligand docking studies we examined putative ligand binding sites of ARPIs in aspartyl proteases of P. falciparum (plasmepsins II and IV) and P. chabaudi (plasmepsin) and found that these in silico analyses support the antimalarial activity hypothesized to be mediated through inhibition of these enzymes. In addition, in vitro enzyme assays demonstrated that P. falciparum plasmepsins II and IV are both inhibited by the ARPIs saquinavir, ritonavir, and lopinavir. The combined results suggest that ARPIs have useful antimalarial activity that may be especially relevant in geographical regions where HIV and P. falciparum infections are both endemic.
- Published
- 2006
- Full Text
- View/download PDF
50. Dendritic cells and follicular dendritic cells express a novel ligand for CD38 which influences their maturation and antibody responses
- Author
-
Lynette Beattie, G. Gordon MacPherson, Michelle N. Wykes, and Derek N.J. Hart
- Subjects
Immunology ,CD11c ,CD38 ,Ligands ,Major histocompatibility complex ,Mice ,Antigen ,Antigens, CD ,immune system diseases ,hemic and lymphatic diseases ,Animals ,Humans ,Immunology and Allergy ,ADP-ribosyl Cyclase ,Cells, Cultured ,Membrane Glycoproteins ,biology ,Follicular dendritic cells ,Cell Differentiation ,hemic and immune systems ,Dendritic Cells ,Original Articles ,Flow Cytometry ,Germinal Center ,ADP-ribosyl Cyclase 1 ,Molecular biology ,Cell biology ,Mice, Inbred C57BL ,Immunoglobulin G ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Antibody ,Cyclase activity ,Dendritic Cells, Follicular ,Spleen ,Ex vivo - Abstract
CD38 is a cell surface molecule with ADP-ribosyl cyclase activity, which is predominantly expressed on lymphoid and myeloid cells. CD38 has a significant role in B-cell function as some anti-CD38 antibodies can deliver potent growth and differentiation signals, but the ligand that delivers this signal in mice is unknown. We used a chimeric protein of mouse CD38 and human immunogobulin G (IgG) (CD38-Ig) to identify a novel ligand for murine CD38 (CD38L) on networks of follicular dendritic cells (FDCs) as well as dendritic cells (DCs) in the spleen. Flow-cytometry found that all DC subsets expressed cytoplasmic CD38L but only fresh ex vivo CD11c+ CD11b− DCs had cell surface CD38L. Anti-CD38 antibody blocked the binding of CD38-Ig to CD38L, confirming the specificity of detection. CD38-Ig immuno-precipitated ligands of 66 and 130 kDa. Functional studies found that CD38-Ig along with anti-CD40 and anti-major histocompatibility complex (MHC) class II antibody provided maturation signals to DCs in vitro. When CD38-Ig was administered in vivo with antigen, IgG2a responses were significantly reduced, suggesting that B and T cells expressing CD38 may modulate the isotype of antibodies produced through interaction with CD38L on DCs. CD38-Ig also expanded FDC networks when administered in vivo. In conclusion, this study has identified a novel ligand for CD38 which has a role in functional interactions between lymphocytes and DCs or FDCs.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.