Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Front Immunol ; 14: 1123342, 2023.
Article in English | MEDLINE | ID: mdl-37056754

ABSTRACT

Introduction: In people with HIV (PWH) both off and on antiretroviral therapy (ART), the expression of immune checkpoint (IC) proteins is elevated on the surface of total and HIV-specific T-cells, indicating T-cell exhaustion. Soluble IC proteins and their ligands can also be detected in plasma, but have not been systematically examined in PWH. Since T-cell exhaustion is associated with HIV persistence on ART, we aimed to determine if soluble IC proteins and their ligands also correlated with the size of the HIV reservoir and HIV-specific T-cell function. Methods: We used multiplex bead-based immunoassay to quantify soluble programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin domain and mucin domain 3 (TIM-3), PD-1 Ligand 1 (PD-L1) and PD-1 Ligand 2 (PD-L2) in plasma from PWH off ART (n=20), on suppressive ART (n=75) and uninfected controls (n=20). We also quantified expression of membrane-bound IC and frequencies of functional T-cells to Gag and Nef peptide stimulation on CD4+ and CD8+ T-cells using flow cytometry. The HIV reservoir was quantified in circulating CD4+ T-cells using qPCR for total and integrated HIV DNA, cell-associated unspliced HIV RNA and 2LTR circles. Results: Soluble (s) PD-L2 level was higher in PWH off and on ART compared to uninfected controls. Higher levels of sPD-L2 correlated with lower levels of HIV total DNA and higher frequencies of gag-specific CD8+ T-cells expressing CD107a, IFNγ or TNFα. In contrast, the concentration of sLAG-3 was similar in uninfected individuals and PWH on ART, but was significantly elevated in PWH off ART. Higher levels of sLAG-3 correlated with higher levels of HIV total and integrated DNA, and lower frequency of gag-specific CD4+ T cells expressing CD107a. Similar to sLAG-3, levels of sPD-1 were elevated in PWH off ART and normalized in PWH on ART. sPD-1 was positively correlated with the frequency of gag-specific CD4+ T cells expressing TNF-a and the expression of membrane-bound PD-1 on total CD8+ T-cells in PWH on ART. Discussion: Plasma soluble IC proteins and their ligands correlate with markers of the HIV reservoir and HIV-specific T-cell function and should be investigated further in in large population-based studies of the HIV reservoir or cure interventions in PWH on ART.


Subject(s)
HIV Infections , Programmed Cell Death 1 Receptor , Humans , Programmed Cell Death 1 Receptor/metabolism , Ligands , Lymphocyte Activation , CD4-Positive T-Lymphocytes
2.
Pathog Glob Health ; 117(2): 134-151, 2023 03.
Article in English | MEDLINE | ID: mdl-35550001

ABSTRACT

The ongoing pandemic of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has globally devastated public health, the economies of many countries and quality of life universally. The recent emergence of immune-escaped variants and scenario of vaccinated individuals being infected has raised the global concerns about the effectiveness of the current available vaccines in transmission control and disease prevention. Given the high rate mutation of SARS-CoV-2, an efficacious vaccine targeting against multiple variants that contains virus-specific epitopes is desperately needed. An immunoinformatics approach is gaining traction in vaccine design and development due to the significant reduction in time and cost of immunogenicity studies and increasing reliability of the generated results. It can underpin the development of novel therapeutic methods and accelerate the design and production of peptide vaccines for infectious diseases. Structural proteins, particularly spike protein (S), along with other proteins have been studied intensively as promising coronavirus vaccine targets. Numbers of promising online immunological databases, tools and web servers have widely been employed for the design and development of next generation COVID-19 vaccines. This review highlights the role of immunoinformatics in identifying immunogenic peptides as potential vaccine targets, involving databases, and prediction and characterization of epitopes which can be harnessed for designing future coronavirus vaccines.


Subject(s)
COVID-19 , Viral Vaccines , Humans , COVID-19 Vaccines , SARS-CoV-2 , Viral Vaccines/chemistry , Viral Vaccines/genetics , Quality of Life , Reproducibility of Results , Epitopes, B-Lymphocyte , Epitopes, T-Lymphocyte , Molecular Docking Simulation
3.
Front Immunol ; 11: 1377, 2020.
Article in English | MEDLINE | ID: mdl-32733457

ABSTRACT

The blockade of programmed cell death-1 (PD1) and its ligand PDL1 has been proven to be a successful immunotherapy against several cancers. Similar to cancer, PD1 contributes to the establishment of several chronic infectious diseases, including malaria. While monoclonal antibodies (mAbs) targeting checkpoint receptors are revolutionary in cancer treatment, the immune-related adverse events (irAEs) may prevent their utilization in prophylactic and therapeutic treatments of infectious diseases. The irAEs are, in part, due to the prolonged half-life of mAbs resulting in prolonged activation of the immune system. As an alternative modality to mAbs, peptides represent a viable option because they possess a shorter pharmacokinetic half-life and offer more formulation and delivery options. Here, we report on a 22-amino acid immunomodulatory peptide, LD01, derived from a Bacillus bacteria. When combined prophylactically with an adenovirus-based or irradiated sporozoite-based malaria vaccine, LD01 significantly enhanced antigen-specific CD8+ T cell expansion. Therapeutically, LD01 treatment of mice infected with a lethal malaria strain resulted in survival that was associated with lower numbers of FOXP3+Tbet+CD4+ regulatory T cells. Taken together, our results demonstrate that LD01 is a potent immunomodulator that acts upon the adaptive immune system to stimulate T cell responses both prophylactically and therapeutically.


Subject(s)
Lymphocyte Activation/drug effects , Malaria Vaccines/immunology , Malaria Vaccines/pharmacology , Malaria/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Lymphocyte Activation/immunology , Mice , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
4.
J Clin Oncol ; 37(34): 3300-3309, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31461379

ABSTRACT

PURPOSE: Understanding the immunobiology of the 15% to 30% of patients with follicular lymphoma (FL) who experience progression of disease within 24 months (POD24) remains a priority. Solid tumors with low levels of intratumoral immune infiltration have inferior outcomes. It is unknown whether a similar relationship exists between POD24 in FL. PATIENTS AND METHODS: Digital gene expression using a custom code set-five immune effector, six immune checkpoint, one macrophage molecules-was applied to a discovery cohort of patients with early- and advanced-stage FL (n = 132). T-cell receptor repertoire analysis, flow cytometry, multispectral immunofluorescence, and next-generation sequencing were performed. The immune infiltration profile was validated in two independent cohorts of patients with advanced-stage FL requiring systemic treatment (n = 138, rituximab plus cyclophosphamide, vincristine, prednisone; n = 45, rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone), with the latter selected to permit comparison of patients experiencing a POD24 event with those having no progression at 5 years or more. RESULTS: Immune molecules showed distinct clustering, characterized by either high or low expression regardless of categorization as an immune effector, immune checkpoint, or macrophage molecule. Low programmed death-ligand 2 (PD-L2) was the most sensitive/specific marker to segregate patients with adverse outcomes; therefore, PD-L2 expression was chosen to distinguish immune infiltrationHI (ie, high PD-L2) FL biopsies from immune infiltrationLO (ie, low PD-L2) tumors. Immune infiltrationHI tissues were highly infiltrated with macrophages and expanded populations of T-cell clones. Of note, the immune infiltrationLO subset of patients with FL was enriched for POD24 events (odds ratio [OR], 4.32; c-statistic, 0.81; P = .001), validated in the independent cohorts (rituximab plus cyclophosphamide, vincristine, prednisone: OR, 2.95; c-statistic, 0.75; P = .011; and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone: OR, 7.09; c-statistic, 0.88; P = .011). Mutations were equally proportioned across tissues, which indicated that degree of immune infiltration is capturing aspects of FL biology distinct from its mutational profile. CONCLUSION: Assessment of immune-infiltration by PD-L2 expression is a promising tool with which to help identify patients who are at risk for POD24.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Biomarkers, Tumor/analysis , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphoma, Follicular/drug therapy , Programmed Cell Death 1 Ligand 2 Protein/analysis , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/genetics , Databases, Factual , Disease Progression , Germany , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphoma, Follicular/genetics , Lymphoma, Follicular/immunology , Lymphoma, Follicular/mortality , North America , Programmed Cell Death 1 Ligand 2 Protein/genetics , Progression-Free Survival , Queensland , Risk Factors , Time Factors , Transcriptome
5.
Clin Transl Immunology ; 8(7): e01071, 2019.
Article in English | MEDLINE | ID: mdl-31367378

ABSTRACT

OBJECTIVE: Crohn's disease (CD) is characterised by inflammation, predominantly associated with ilea. To investigate the basis for this inflammation in patients with CD, we examined dendritic cells (DC) which are pivotal for maintenance of immunological tolerance in the gut. METHODS: Ileal biopsies and blood DCs from CD patients and controls were examined by microscopy and flow cytometry for PD-L1 and PD-L2 expression, as PD-L1 has been implicated in colitis but the contribution of PD-L2 is less clear. In vitro studies, of blood samples from CD patients, were used to demonstrate a functional role for PD-L2 in disease pathogenesis. RESULTS: Quantitative microscopy of CD11c+ DCs in inflamed and noninflamed ilea from CD patient showed > 75% loss of these cells from the villi, lamina propria and Peyer's patches compared with non-CD controls. Given this loss of DCs from ilia of CD patients, we hypothesised DCs may have migrated to the blood as these patients can have extra-intestinal symptoms. We thus examined blood DCs from CD patients by flow cytometry and found significant increases in PD-L1 and PD-L2 expression compared with control samples. Microscopy revealed an aggregated form of PD-L2 expression, known to drive Th1 immunity, in CD patients but not in controls. In vitro functional studies with PD-L2 blockade confirmed PD-L2 contributes significantly to the secretion of pro-inflammatory cytokines known to cause disease pathogenesis. CONCLUSION: Taken together, this study shows that PD-L2 can influence the progression of CD and blockade of PD-L2 may have therapeutic potential.

6.
Nat Rev Immunol ; 18(2): 91-104, 2018 02.
Article in English | MEDLINE | ID: mdl-28990586

ABSTRACT

The upregulation of immune checkpoint molecules, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte antigen 4 (CTLA4), on immune cells occurs during acute infections, such as malaria, as well as during chronic persistent viral infections, including HIV and hepatitis B virus. These pathways are important for preventing immune-driven pathology but can also limit immune-mediated clearance of the infection. The recent success of immune checkpoint blockade in cancer therapy suggests that targeting these pathways would also be effective for preventing and treating a range of infectious diseases. Here, we review our current understanding of immune checkpoint pathways in the pathogenesis of infectious diseases and discuss the potential for therapeutically targeting these pathways in this setting.


Subject(s)
CTLA-4 Antigen/antagonists & inhibitors , Communicable Diseases/immunology , Communicable Diseases/therapy , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CTLA-4 Antigen/immunology , HIV Infections/immunology , HIV Infections/therapy , Hepatitis B/immunology , Hepatitis B/therapy , Humans , Immune Tolerance , Malaria/immunology , Malaria/therapy , Models, Immunological , Neoplasms/immunology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Signal Transduction/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/therapy , T-Lymphocytes/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/therapy
7.
Front Immunol ; 9: 2926, 2018.
Article in English | MEDLINE | ID: mdl-30631323

ABSTRACT

Plasmodium spp., the causative agent of malaria, caused 212 million infections in 2016 with 445,000 deaths, mostly in children. Adults acquire enough immunity to prevent clinical symptoms but never develop sterile immunity. The only vaccine for malaria, RTS,S, shows promising protection of a limited duration against clinical malaria in infants but no significant protection against severe disease. There is now abundant evidence that T cell functions are inhibited during malaria, which may explain why vaccine are not efficacious. Studies have now clearly shown that T cell immunity against malaria is subdued by multiple the immune regulatory receptors, in particular, by programmed cell-death-1 (PD-1). Given there is an urgent need for an efficacious malarial treatment, compounded with growing drug resistance, a better understanding of malarial immunity is essential. This review will examine molecular signals that affect T cell-mediated immunity against malaria.


Subject(s)
Antimalarials/immunology , Immunity, Cellular/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Adult , Child , Humans , Infant , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Plasmodium/physiology , T-Lymphocytes/parasitology
8.
Bio Protoc ; 7(11): e2302, 2017 Jun 05.
Article in English | MEDLINE | ID: mdl-34541071

ABSTRACT

Interferon-gamma (IFN-γ) is crucial for immunity against intracellular pathogens and for tumor control. It is produced predominantly by natural killer (NK) and natural killer T cells (NKT) as well as by antigen-specific Th1 CD4+ and CD8+ effector T cells. When investigating immune responses against pathogens and cancer cells, measuring antigen-specific cytokine-responses by cells of adaptive immunity offers an advantage over total non-specific cytokine responses. Significantly, the measurement of antigen-specific IFN-γ responses against pathogens or cancer cells, when compared to a treatment group, provides a quantitative measure of how well the treatment works. Measuring antigen-specific IFN-γ responses involves culture of the cells being considered (CD4+ or CD8+ T cells) with antigen presenting cells (APC) and a specific peptide from the target pathogen or cancer cell compared to control cultures without a peptide. After a suitable timeframe, the cytokine released is measured by an ELISPOT assay. The difference in the number of cells secreting IFN-γ, with and without peptide, is a measure of antigen-specific IFN-γ responses. This assay can be applied to other cytokines such as IL-10.

9.
Immunity ; 45(2): 333-45, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533014

ABSTRACT

Many pathogens, including Plasmodium spp., exploit the interaction of programmed death-1 (PD-1) with PD-1-ligand-1 (PD-L1) to "deactivate" T cell functions, but the role of PD-L2 remains unclear. We studied malarial infections to understand the contribution of PD-L2 to immunity. Here we have shown that higher PD-L2 expression on blood dendritic cells, from Plasmodium falciparum-infected individuals, correlated with lower parasitemia. Mechanistic studies in mice showed that PD-L2 was indispensable for establishing effective CD4(+) T cell immunity against malaria, because it not only inhibited PD-L1 to PD-1 activity but also increased CD3 and inducible co-stimulator (ICOS) expression on T cells. Importantly, administration of soluble multimeric PD-L2 to mice with lethal malaria was sufficient to dramatically improve immunity and survival. These studies show immuno-regulation by PD-L2, which has the potential to be translated into an effective treatment for malaria and other diseases where T cell immunity is ineffective or short-lived due to PD-1-mediated signaling.


Subject(s)
B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adamantane/analogs & derivatives , Adamantane/therapeutic use , Adult , Animals , Antimalarials/therapeutic use , B7-H1 Antigen/genetics , Cells, Cultured , Clinical Trials as Topic , Dendritic Cells/parasitology , Female , Humans , Immunity, Cellular , Lymphocyte Activation , Malaria, Falciparum/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Parasitemia/immunology , Peroxides/therapeutic use , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Receptor/genetics , Pyrimidines/therapeutic use , Triazoles/therapeutic use , Young Adult
10.
Sci Rep ; 6: 26210, 2016 05 24.
Article in English | MEDLINE | ID: mdl-27217330

ABSTRACT

Even after years of experiencing malaria, caused by infection with Plasmodium species, individuals still have incomplete immunity and develop low-density parasitemia on re-infection. Previous studies using the P. chabaudi (Pch) mouse model to understand the reason for chronic malaria, found that mice with a deletion of programmed cell death-1 (PD-1KO) generate sterile immunity unlike wild type (WT) mice. Here we investigated if the mechanism underlying this defect during acute immunity also impacts on long-term immunity. We infected WT and PD-1KO mice with Pch-malaria and measured protection as well as immune responses against re-infections, 15 or 20 weeks after the original infection had cleared. WT mice showed approximately 1% parasitemia compared to sterile immunity in PD-1KO mice on re-infection. An examination of the mechanisms of immunity behind this long-term protection in PD-1KO mice showed a key role for parasite-specific CD8(+) T cells even when CD4(+) T cells and B cells responded to re-infection. These studies indicate that long-term CD8(+) T cell-meditated protection requires consideration for future malaria vaccine design, as part of a multi-cell type response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria/immunology , Plasmodium chabaudi/immunology , Programmed Cell Death 1 Receptor/metabolism , Animals , Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency
11.
J Virol ; 89(17): 9137-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26109734

ABSTRACT

Here we present evidence for previously unappreciated B-cell immune dysregulation during acute Epstein-Barr virus (EBV)-associated infectious mononucleosis (IM). Longitudinal analyses revealed that patients with acute IM have undetectable EBV-specific neutralizing antibodies and gp350-specific B-cell responses, which were associated with a significant reduction in memory B cells and no evidence of circulating antibody-secreting cells. These observations correlate with dysregulation of tumor necrosis factor family members BAFF and APRIL and increased expression of FAS on circulating B cells.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Herpesvirus 4, Human/immunology , Infectious Mononucleosis/immunology , B-Cell Activating Factor/immunology , B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Humans , Immunologic Memory/immunology , Infectious Mononucleosis/virology , Lymphocyte Activation/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Viral Matrix Proteins/immunology , fas Receptor/metabolism
12.
J Infect Dis ; 212(12): 1957-61, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26080368

ABSTRACT

Acute infectious mononucleosis (IM) is associated with altered expression of inflammatory cytokines and disturbed T-cell homeostasis, however, the precise mechanism of this immune dysregulation remains unresolved. In the current study we demonstrated a significant loss of circulating myeloid and plasmacytoid dendritic cells (DCs) during acute IM, a loss correlated with the severity of clinical symptoms. In vitro exposure of blood DCs to acute IM plasma resulted in loss of plasmacytoid DCs, and further studies with individual cytokines showed that exposure to interleukin 10 could replicate this effect. Our data provide important mechanistic insight into dysregulated immune homeostasis during acute IM.


Subject(s)
Blood Cells/drug effects , Dendritic Cells/immunology , Immune Tolerance , Infectious Mononucleosis/pathology , Interleukin-10/blood , Adolescent , Adult , Cell Survival/drug effects , Humans , Infectious Mononucleosis/immunology , Interleukin-10/metabolism , Young Adult
13.
Front Microbiol ; 5: 249, 2014.
Article in English | MEDLINE | ID: mdl-24904561

ABSTRACT

Malaria is a significant global burden but after >30 years of effort there is no vaccine on the market. While the complex life cycle of the parasite presents several challenges, many years of research have also identified several mechanisms of immune evasion by Plasmodium spp. Recent research on malaria, has investigated the programmed cell death-1 (PD-1) pathway which mediates exhaustion of T cells, characterized by poor effector functions and recall responses and in some cases loss of the cells by apoptosis. Such studies have shown exhaustion of CD4(+) T cells and an unappreciated role for CD8(+) T cells in promoting sterile immunity against blood stage malaria. This is because PD-1 mediates up to a 95% reduction in numbers and functional capacity of parasite-specific CD8(+) T cells, thus masking their role in protection. The role of T cell exhaustion during malaria provides an explanation for the absence of sterile immunity following the clearance of acute disease which will be relevant to future malaria-vaccine design and suggests the need for novel therapeutic solutions. This review will thus examine the role of PD-1-mediated T cell exhaustion in preventing lasting immunity against malaria.

14.
Cell Rep ; 5(5): 1204-13, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24316071

ABSTRACT

Malaria is a highly prevalent disease caused by infection by Plasmodium spp., which infect hepatocytes and erythrocytes. Blood-stage infections cause devastating symptoms and can persist for years. Antibodies and CD4(+) T cells are thought to protect against blood-stage infections. However, there has been considerable difficulty in developing an efficacious malaria vaccine, highlighting our incomplete understanding of immunity against this disease. Here, we used an experimental rodent malaria model to show that PD-1 mediates up to a 95% reduction in numbers and functional capacity of parasite-specific CD8(+) T cells. Furthermore, in contrast to widely held views, parasite-specific CD8(+) T cells are required to control both acute and chronic blood-stage disease even when parasite-specific antibodies and CD4(+) T cells are present. Our findings provide a molecular explanation for chronic malaria that will be relevant to future malaria-vaccine design and may need consideration when vaccine development for other infections is problematic.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Malaria/blood , Programmed Cell Death 1 Receptor/metabolism , Animals , CD8-Positive T-Lymphocytes/pathology , Malaria/immunology , Malaria/metabolism , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics
16.
J Immunol ; 190(6): 2692-701, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23401589

ABSTRACT

Streptococcus pyogenes (group A Streptococcus [GAS]) is a leading human pathogen associated with a diverse array of mucosal and systemic infections. Vaccination with J8, a conserved region synthetic peptide derived from the M-protein of GAS and containing only 12 aa from GAS, when conjugated to diphtheria toxoid, has been shown to protect mice against a lethal GAS challenge. Protection has been previously shown to be Ab-mediated. J8 does not contain a dominant GAS-specific T cell epitope. The current study examined long-term Ab memory and dissected the role of B and T cells. Our results demonstrated that vaccination generates specific memory B cells (MBC) and long-lasting Ab responses. The MBC response can be activated following boost with Ag or limiting numbers of whole bacteria. We further show that these memory responses protect against systemic infection with GAS. T cell help is required for activation of MBC but can be provided by naive T cells responding directly to GAS at the time of infection. Thus, individuals whose T cells do not recognize the short synthetic peptide in the vaccine will be able to generate a protective and rapid memory Ab response at the time of infection. These studies significantly strengthen previous findings, which showed that protection by the J8-diphtheria toxoid vaccine is Ab-mediated and suggest that in vaccine design for other organisms the source of T cell help for Ab responses need not be limited to sequences from the organism itself.


Subject(s)
Antibodies, Bacterial/biosynthesis , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/microbiology , Immunologic Memory/immunology , Streptococcal Infections/prevention & control , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Vaccines, Subunit/administration & dosage , Animals , Antibodies, Bacterial/administration & dosage , B-Lymphocyte Subsets/metabolism , Immunization, Secondary/methods , Mice , Mice, Inbred BALB C , Mice, SCID , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Time Factors , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
17.
Eur J Immunol ; 42(12): 3291-301, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22936176

ABSTRACT

Malaria is a major cause of morbidity worldwide with reports of over 200-500 million infected individuals and nearly 1 million deaths each year. Antibodies have been shown to play a critical role in controlling the blood stage of this disease; however, in malaria-endemic areas antibody immunity is slow to develop despite years of exposure to Plasmodium spp. the causative parasite. Using rodent Plasmodium yoelii YM, we provide evidence that malarial infections result in a decrease in the proportion of DCs that express the B-cell survival factor, BAFF, resulting in a decreased ability of these DCs to support memory B-cell differentiation into antibody secreting cells (ASCs) and/or the survival of ASCs. Further, compared with infected WT mice, ASC numbers were significantly increased in malaria-infected transgenic mice that either overexpressed BAFF or mice with BAFF-independent B-cell survival (B-cell-restricted TRAF3 deletion). Remarkably, BAFF-overexpressing mice were protected from lethal malaria infections, indicating the significance of the role BAFF plays in determining the outcome of malaria infections. These findings describe a previously unappreciated mechanism by which Plasmodium spp. can depress the generation of protective antibody responses.


Subject(s)
Antibodies, Protozoan/immunology , B-Cell Activating Factor/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunologic Memory , Malaria/immunology , Plasma Cells/immunology , Plasmodium yoelii/immunology , Animals , Antibodies, Protozoan/genetics , Antibodies, Protozoan/metabolism , Antibody Formation/genetics , B-Cell Activating Factor/biosynthesis , B-Cell Activating Factor/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation/genetics , Humans , Malaria/genetics , Malaria/metabolism , Mice , Mice, Transgenic , Plasma Cells/metabolism
18.
BMC Evol Biol ; 12: 140, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22871040

ABSTRACT

BACKGROUND: Proteins of the mammalian PYHIN (IFI200/HIN-200) family are involved in defence against infection through recognition of foreign DNA. The family member absent in melanoma 2 (AIM2) binds cytosolic DNA via its HIN domain and initiates inflammasome formation via its pyrin domain. AIM2 lies within a cluster of related genes, many of which are uncharacterised in mouse. To better understand the evolution, orthology and function of these genes, we have documented the range of PYHIN genes present in representative mammalian species, and undertaken phylogenetic and expression analyses. RESULTS: No PYHIN genes are evident in non-mammals or monotremes, with a single member found in each of three marsupial genomes. Placental mammals show variable family expansions, from one gene in cow to four in human and 14 in mouse. A single HIN domain appears to have evolved in the common ancestor of marsupials and placental mammals, and duplicated to give rise to three distinct forms (HIN-A, -B and -C) in the placental mammal ancestor. Phylogenetic analyses showed that AIM2 HIN-C and pyrin domains clearly diverge from the rest of the family, and it is the only PYHIN protein with orthology across many species. Interestingly, although AIM2 is important in defence against some bacteria and viruses in mice, AIM2 is a pseudogene in cow, sheep, llama, dolphin, dog and elephant. The other 13 mouse genes have arisen by duplication and rearrangement within the lineage, which has allowed some diversification in expression patterns. CONCLUSIONS: The role of AIM2 in forming the inflammasome is relatively well understood, but molecular interactions of other PYHIN proteins involved in defence against foreign DNA remain to be defined. The non-AIM2 PYHIN protein sequences are very distinct from AIM2, suggesting they vary in effector mechanism in response to foreign DNA, and may bind different DNA structures. The PYHIN family has highly varied gene composition between mammalian species due to lineage-specific duplication and loss, which probably indicates different adaptations for fighting infectious disease. Non-genomic DNA can indicate infection, or a mutagenic threat. We hypothesise that defence of the genome against endogenous retroelements has been an additional evolutionary driver for PYHIN proteins.


Subject(s)
Evolution, Molecular , Mammals/genetics , Nuclear Proteins/genetics , Animals , Bayes Theorem , DNA-Binding Proteins , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/chemistry , Nuclear Proteins/immunology , Phylogeny , Rats , Transcriptome
19.
Int J Parasitol ; 42(6): 583-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22726753

ABSTRACT

Malaria, caused by Plasmodium spp., is responsible for over 200 million infections worldwide and 650,000 deaths annually. Until recently, it was thought that blood-stage parasites survived and replicated in hepatocytes and red blood cells exclusively. We recently showed that blood-stage parasites could infect, survive and replicate within plasmacytoid dendritic cells of the spleen and that these cells could release infective parasites. Here we discuss the implications of this novel niche in the spleen.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/parasitology , Host-Pathogen Interactions , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Plasmodium/pathogenicity , Humans , Spleen/immunology , Spleen/parasitology
20.
Trends Parasitol ; 28(5): 182-6, 2012 May.
Article in English | MEDLINE | ID: mdl-22365902

ABSTRACT

Dendritic cells (DCs), the sentinels of immunity, reside in almost every organ of the body. These cells are responsible for initiating immune responses against infectious agents. DCs are divided into different subsets based on their biological functions, with plasmacytoid DCs (pDCs) and conventional DCs (cDCs) being two major populations. The ability of DCs to protect against malaria infection was recently questioned when pDCs were reported to be a reservoir for rodent Plasmodium spp. in the spleen. This opinion article explores how the occupation of pDCs by the parasite may corrupt immunity against malaria.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/parasitology , Malaria/immunology , Malaria/parasitology , Animals , Humans , Mice , Plasmodium/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...