Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Int J Infect Dis ; 143: 107014, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38499058

ABSTRACT

Tropical infectious diseases inflict an unacceptable burden of disease on humans living in developing countries. Although anti-pathogenic drugs have been widely used, they carry a constant threat of selecting for resistance. Vaccines offer a promising means by which to enhance the global control of tropical infectious diseases; however, these have been difficult to develop, mostly because of the complex nature of the pathogen lifecycles. Here, we present recently developed vaccine candidates for five tropical infectious diseases in the form of a catalog that have either entered clinical trials or have been licensed for use. We deliberate on recently licensed dengue vaccines, provide evidence why combination vaccination could have a synergistic impact on schistosomiasis, critically appraise the value of typhoid conjugate vaccines, and discuss the potential of vaccines in the efforts to eliminate vivax malaria and hookworms.


Subject(s)
Dengue , Humans , Dengue/prevention & control , Dengue Vaccines/immunology , Dengue Vaccines/administration & dosage , Schistosomiasis/prevention & control , Communicable Diseases , Tropical Medicine , Vaccines/immunology , Typhoid Fever/prevention & control , Malaria, Vivax/prevention & control , Vaccine Development
2.
Trends Parasitol ; 40(1): 28-44, 2024 01.
Article in English | MEDLINE | ID: mdl-38065791

ABSTRACT

Cerebral malaria (CM) is a severe neurological complication caused by Plasmodium falciparum parasites; it is characterized by the sequestration of infected red blood cells within the cerebral microvasculature. New findings, combined with a better understanding of the central nervous system (CNS) barriers, have provided greater insight into the players and events involved in CM, including site-specific T cell responses in the human brain. Here, we review the updated roles of innate and adaptive immune responses in CM, with a focus on the role of the perivascular macrophage-endothelium unit in antigen presentation, in the vascular and perivascular compartments. We suggest that these events may be pivotal in the development of CM.


Subject(s)
Malaria, Cerebral , Humans , Brain , Plasmodium falciparum/physiology , Host-Parasite Interactions , Erythrocytes/parasitology
3.
Clin Microbiol Rev ; 34(4): e0034820, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34494873

ABSTRACT

About half of the world's population and 80% of the world's biodiversity can be found in the tropics. Many diseases are specific to the tropics, with at least 41 diseases caused by endemic bacteria, viruses, parasites, and fungi. Such diseases are of increasing concern, as the geographic range of tropical diseases is expanding due to climate change, urbanization, change in agricultural practices, deforestation, and loss of biodiversity. While traditional medicines have been used for centuries in the treatment of tropical diseases, the active natural compounds within these medicines remain largely unknown. In this review, we describe infectious diseases specific to the tropics, including their causative pathogens, modes of transmission, recent major outbreaks, and geographic locations. We further review current treatments for these tropical diseases, carefully consider the biodiscovery potential of the tropical biome, and discuss a range of technologies being used for drug development from natural resources. We provide a list of natural products with antimicrobial activity, detailing the source organisms and their effectiveness as treatment. We discuss how technological advancements, such as next-generation sequencing, are driving high-throughput natural product screening pipelines to identify compounds with therapeutic properties. This review demonstrates the impact natural products from the vast tropical biome have in the treatment of tropical infectious diseases and how high-throughput technical capacity will accelerate this discovery process.


Subject(s)
Biological Products , Communicable Diseases , Biodiversity , Biological Products/therapeutic use , Communicable Diseases/drug therapy , Communicable Diseases/epidemiology , Humans , Tropical Climate
4.
Clin Transl Immunology ; 10(4): e1273, 2021.
Article in English | MEDLINE | ID: mdl-33854773

ABSTRACT

OBJECTIVES: The immunologic events that build up to the fatal neurological stage of experimental cerebral malaria (ECM) are incompletely understood. Here, we dissect immune cell behaviour occurring in the central nervous system (CNS) when Plasmodium berghei ANKA (PbA)-infected mice show only minor clinical signs. METHODS: A 2-photon intravital microscopy (2P-IVM) brain imaging model was used to study the spatiotemporal context of early immunological events in situ during ECM. RESULTS: Early in the disease course, antigen-specific CD8+ T cells came in contact and arrested on the endothelium of post-capillary venules. CD8+ T cells typically adhered adjacent to, or were in the near vicinity of, perivascular macrophages (PVMs) that line post-capillary venules. Closer examination revealed that CD8+ T cells crawled along the inner vessel wall towards PVMs that lay on the abluminal side of large post-capillary venules. 'Activity hotspots' in large post-capillary venules were characterised by T-cell localisation, activated morphology and clustering of PVM, increased abutting of post-capillary venules by PVM and augmented monocyte accumulation. In the later stages of infection, when mice exhibited neurological signs, intravascular CD8+ T cells increased in number and changed their behaviour, actively crawling along the endothelium and displaying frequent, short-term interactions with the inner vessel wall at hotspots. CONCLUSION: Our study suggests an active interaction between PVM and CD8+ T cells occurs across the blood-brain barrier (BBB) in early ECM, which may be the initiating event in the inflammatory cascade leading to BBB alteration and neuropathology.

5.
Sci Rep ; 10(1): 13115, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753607

ABSTRACT

Toxoplasmic encephalitis is an AIDS-defining condition. The decline of IFN-γ-producing CD4+ T cells in AIDS is a major contributing factor in reactivation of quiescent Toxoplasma gondii to an actively replicating stage of infection. Hence, it is important to characterize CD4-independent mechanisms that constrain acute T. gondii infection. We investigated the in vivo regulation of IFN-γ production by CD8+ T cells, DN T cells and NK cells in response to acute T. gondii infection. Our data show that processing of IFN-γ by these non-CD4 cells is dependent on both IL-12 and IL-18 and the secretion of bioactive IL-18 in response to T. gondii requires the sensing of viable parasites by multiple redundant inflammasome sensors in multiple hematopoietic cell types. Importantly, our results show that expansion of CD8+ T cells, DN T cells and NK cell by S4B6 IL-2 complex pre-treatment increases survival rates of mice infected with T. gondii and this is dependent on IL-12, IL-18 and IFN-γ. Increased survival is accompanied by reduced pathology but is independent of expansion of TReg cells or parasite burden. This provides evidence for a protective role of IL2C-mediated expansion of non-CD4 cells and may represent a promising lead to adjunct therapy for acute toxoplasmosis.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-12/metabolism , Interleukin-18/metabolism , Interleukin-2/pharmacology , Toxoplasmosis/immunology , Toxoplasmosis/prevention & control , Animals , Brain/parasitology , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-2/chemistry , Mice , Toxoplasma/drug effects , Toxoplasma/physiology , Toxoplasmosis/metabolism
6.
Clin Transl Immunology ; 9(7): e1158, 2020.
Article in English | MEDLINE | ID: mdl-32714552

ABSTRACT

The global prevalence of respiratory infectious and inflammatory diseases remains a major public health concern. Prevention and management strategies have not kept pace with the increasing incidence of these diseases. The airway mucosa is the most common portal of entry for infectious and inflammatory agents. Therefore, significant benefits would be derived from a detailed understanding of how immune responses regulate the filigree of the airways. Here, the role of different antigen-presenting cells (APC) in the lower airways and the mechanisms used by pathogens to modulate APC function during infectious disease is reviewed. Features of APC that are unique to the airways and the influence they have on uptake and presentation of antigen to T cells directly in the airways are discussed. Current information on the crucial role that airway APC play in regulating respiratory infection is summarised. We examine the clinical implications of APC dysregulation in the airways on asthma and tuberculosis, two chronic diseases that are the major cause of illness and death in the developed and developing world. A brief overview of emerging therapies that specifically target APC function in the airways is provided.

7.
Sci Adv ; 6(10): eaaz1767, 2020 03.
Article in English | MEDLINE | ID: mdl-32181361

ABSTRACT

Tuberculosis (TB) is the deadliest infectious disease worldwide. Bacille-Calmette-Guérin (BCG), the only licensed TB vaccine, affords variable protection against TB but remains the gold standard. BCG improvement is focused around three strategies: recombinant BCG strains, heterologous routes of administration, and booster vaccination. It is currently unknown whether combining these strategies is beneficial. The preclinical evaluation for new TB vaccines is heavily skewed toward immunogenicity and efficacy; however, safety and efficacy are the dominant considerations in human use. To facilitate stage gating of TB vaccines, we developed a simple empirical model to systematically rank vaccination strategies by integrating multiple measurements of safety, immunogenicity, and efficacy. We assessed 24 vaccination regimens, composed of three BCG strains and eight combinations of delivery. The model presented here highlights that mucosal booster vaccination may cause adverse outcomes and provides a much needed strategy to evaluate and rank data obtained from TB vaccine studies using different routes, strains, or animal models.


Subject(s)
BCG Vaccine/administration & dosage , Immunization, Secondary/methods , Immunogenicity, Vaccine , Mycobacterium tuberculosis/drug effects , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Animals , Female , Humans , Immunization Schedule , Injections, Spinal , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Patient Safety , Research Design , Treatment Outcome , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology , Vaccines, Synthetic
8.
Front Immunol ; 10: 532, 2019.
Article in English | MEDLINE | ID: mdl-30949177

ABSTRACT

Tuberculosis (TB) is a major global public health problem causing significant mortality and morbidity. In addition to ~10.4 million cases of active TB annually, it is estimated that about two billion people are latently infected with Mycobacterium tuberculosis (Mtb), the causative agent of TB. Reactivation of latent Mtb infection is the leading cause of death in patients with immunodeficiency virus (HIV) infection. The low efficiency of the only licensed anti-TB vaccine, Bacille Calmette-Guérin (BCG) to reduce pulmonary TB in adults contributes to this problem. Here we investigated if vaccination with conventional BCG or the genetically modified experimental BCGΔBCG1419c strain can prevent reactivation of latent lymphatic TB in a mouse model of induced reactivation, following the depletion of CD4+ T cells, as it occurs in HIV+ individuals. Vaccination with conventional BCG or BCGΔBCG1419c prevented reactivation of Mtb from the infected lymph node and the systemic spread of Mtb to spleen and lung. Prevention of reactivation was independent of vaccination route and was accompanied by reduced levels of circulating inflammatory cytokines and the absence of lung pathology. Our results demonstrate that vaccine-induced CD4+ T cells are not essential to prevent reactivation of latent lymphatic murine TB, and highlight the need to better understand how non-CD4+ immune cell populations participate in protective immune responses to control latent TB.


Subject(s)
BCG Vaccine/therapeutic use , Latent Tuberculosis/therapy , Animals , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Recurrence , Vaccination
10.
Front Immunol ; 7: 246, 2016.
Article in English | MEDLINE | ID: mdl-27540379

ABSTRACT

The kynurenine pathway (KP) is the major metabolic pathway of the essential amino acid tryptophan (TRP). Stimulation by inflammatory molecules, such as interferon-γ (IFN-γ), is the trigger for induction of the KP, driving a complex cascade of production of both neuroprotective and neurotoxic metabolites, and in turn, regulation of the immune response and responses of brain cells to the KP metabolites. Consequently, substantial evidence has accumulated over the past couple of decades that dysregulation of the KP and the production of neurotoxic metabolites are associated with many neuroinflammatory and neurodegenerative diseases, including Parkinson's disease, AIDS-related dementia, motor neurone disease, schizophrenia, Huntington's disease, and brain cancers. In the past decade, evidence of the link between the KP and multiple sclerosis (MS) has rapidly grown and has implicated the KP in MS pathogenesis. KP enzymes, indoleamine 2,3-dioxygenase (IDO-1) and tryptophan dioxygenase (highest expression in hepatic cells), are the principal enzymes triggering activation of the KP to produce kynurenine from TRP. This is in preference to other routes such as serotonin and melatonin production. In neurological disease, degradation of the blood-brain barrier, even if transient, allows the entry of blood monocytes into the brain parenchyma. Similar to microglia and macrophages, these cells are highly responsive to IFN-γ, which upregulates the expression of enzymes, including IDO-1, producing neurotoxic KP metabolites such as quinolinic acid. These metabolites circulate systemically or are released locally in the brain and can contribute to the excitotoxic death of oligodendrocytes and neurons in neurological disease principally by virtue of their agonist activity at N-methyl-d-aspartic acid receptors. The latest evidence is presented and discussed. The enzymes that control the checkpoints in the KP represent an attractive therapeutic target, and consequently several KP inhibitors are currently in clinical trials for other neurological diseases, and hence may make suitable candidates for MS patients. Underpinning these drug discovery endeavors, in recent years, several advances have been made in how KP metabolites are assayed in various biological fluids, and tremendous advancements have been made in how specimens are imaged to determine disease progression and involvement of various cell types and molecules in MS.

11.
PLoS Pathog ; 10(7): e1004236, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25033406

ABSTRACT

During experimental cerebral malaria (ECM) mice develop a lethal neuropathological syndrome associated with microcirculatory dysfunction and intravascular leukocyte sequestration. The precise spatio-temporal context in which the intravascular immune response unfolds is incompletely understood. We developed a 2-photon intravital microscopy (2P-IVM)-based brain-imaging model to monitor the real-time behaviour of leukocytes directly within the brain vasculature during ECM. Ly6C(hi) monocytes, but not neutrophils, started to accumulate in the blood vessels of Plasmodium berghei ANKA (PbA)-infected MacGreen mice, in which myeloid cells express GFP, one to two days prior to the onset of the neurological signs (NS). A decrease in the rolling speed of monocytes, a measure of endothelial cell activation, was associated with progressive worsening of clinical symptoms. Adoptive transfer experiments with defined immune cell subsets in recombinase activating gene (RAG)-1-deficient mice showed that these changes were mediated by Plasmodium-specific CD8(+) T lymphocytes. A critical number of CD8(+) T effectors was required to induce disease and monocyte adherence to the vasculature. Depletion of monocytes at the onset of disease symptoms resulted in decreased lymphocyte accumulation, suggesting reciprocal effects of monocytes and T cells on their recruitment within the brain. Together, our studies define the real-time kinetics of leukocyte behaviour in the central nervous system during ECM, and reveal a significant role for Plasmodium-specific CD8(+) T lymphocytes in regulating vascular pathology in this disease.


Subject(s)
CD8-Positive T-Lymphocytes , Endothelial Cells , Malaria, Cerebral , Monocytes , Plasmodium berghei/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Malaria, Cerebral/physiopathology , Mice , Mice, Knockout , Microscopy, Fluorescence , Monocytes/metabolism , Monocytes/pathology
12.
Front Cell Neurosci ; 6: 67, 2012.
Article in English | MEDLINE | ID: mdl-23316136

ABSTRACT

Intravital imaging of the superficial brain tissue in mice represents a powerful tool for the dissection of the cellular and molecular cues underlying inflammatory and infectious central nervous system (CNS) diseases. We present here a step-by-step protocol that will enable a non-specialist to set up a two-photon brain-imaging model. The protocol offers a two-part approach that is specifically optimized for imaging leukocytes but can be easily adapted to answer varied CNS-related biological questions. The protocol enables simultaneous visualization of fluorescently labeled immune cells, the pial microvasculature and extracellular structures such as collagen fibers at high spatial and temporal resolution. Intracranial structures are exposed through a cranial window, and physiologic conditions are maintained during extended imaging sessions via continuous superfusion of the brain surface with artificial cerebrospinal fluid (aCSF). Experiments typically require 1-2 h of preparation, which is followed by variable periods of immune cell tracking. Our methodology converges the experience of two laboratories over the past 10 years in diseased animal models such as cerebral ischemia, lupus, cerebral malaria, and toxoplasmosis. We exemplify the utility of this protocol by tracking leukocytes in transgenic mice in the pial vessels under steady-state conditions.

13.
J Immunol ; 187(8): 4018-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21900177

ABSTRACT

Costimulation-deficient dendritic cells (DCs) prevent autoimmune disease in mouse models. However, autoimmune-prone mice and humans fail to control expansion of peripheral autoreactive effector memory T cells (T(EMs)), which resist immunoregulation by costimulation-deficient DCs. In contrast, activation of DC costimulation may be coupled with regulatory capacity. To test whether costimulatory DCs control T(EMs) and attenuate established autoimmune disease, we used RelB-deficient mice, which have multiorgan inflammation, expanded peripheral autoreactive T(EMs), and dysfunctional Foxp3(+) regulatory T cells (Tregs) cells and conventional DCs. T(EMs) were regulated by Foxp3(+) Tregs when costimulated by CD3/CD28-coated beads or wild-type DCs but not DCs deficient in RelB or CD80/CD86. After transfer, RelB and CD80/CD86-sufficient DCs restored tolerance and achieved a long-term cure of autoimmune disease through costimulation of T(EM) and Foxp3(+) Treg IFN-γ production, as well as induction of IDO by host APCs. IDO was required for regulation of T(EMs) and suppression of organ inflammation. Our data challenge the paradigm that costimulation-deficient DCs are required to regulate established autoimmune disease to avoid T(EM) activation and demonstrate cooperative cross-talk between costimulatory DCs, IFN-γ, and IDO-dependent immune regulation. IFN-γ and IDO activity may be good surrogate biomarkers measured against clinical efficacy in trials of autoimmune disease immunoregulation.


Subject(s)
Autoimmune Diseases/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/therapy , Dendritic Cells/transplantation , Flow Cytometry , Immune Tolerance/immunology , Immunomagnetic Separation , Inflammation/immunology , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Signal Transduction/immunology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Transcription Factor RelB/immunology
14.
J Autoimmun ; 31(3): 245-51, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18539434

ABSTRACT

Nuclear factor (NF)-kappaB is a transcription factor family which transmits signals from the cell surface to the nucleus, resulting in transcriptional effects on genes involved in inflammation, cell differentiation and survival. The signaling of NF-kappaB and mitogen-activated protein (MAP) kinases through adapter molecules is of critical importance to survival and activation of all cells in the body, including those regulating innate and adaptive immunity. Here we review the individual and intersecting roles played by the alternate and classical NF-kappaB pathways in the pathogenesis of autoimmune disease. Understanding the differences in classical and alternate NF-kappaB function has greatly assisted the development of models of their contribution to different autoimmune diseases. To exemplify these concepts, we consider the contribution of NF-kappaB to rheumatoid arthritis and type 1 diabetes pathogenesis and approaches to immunotherapy.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/immunology , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Animals , Apoptosis/immunology , Autoimmune Diseases/genetics , Autoimmunity/genetics , Humans , Immune Tolerance/genetics , Immunity, Active/genetics , Immunity, Innate/genetics , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics
15.
J Immunol ; 180(5): 3166-75, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18292540

ABSTRACT

Dendritic cell (DC) differentiation is abnormal in type 1 diabetes mellitus (T1DM). However, the nature of the relationship between this abnormality and disease pathogenesis is unknown. We studied the LPS response in monocytes and monocyte-derived DCs isolated from T1DM patients and from non-T1DM controls. In T1DM patients, late LPS-mediated nuclear DNA binding by RelA, p50, c-Rel, and RelB was impaired as compared with type 2 DM, rheumatoid arthritis, and healthy subjects, associated with impaired DC CD40 and MHC class I induction but normal cytokine production. In TIDM monocytes, RelA and RelB were constitutively activated, and the src homology 2 domain-containing protein tyrosine phosphatase (SHP-1), a negative regulator of NF-kappaB, was overexpressed. Addition of sodium stibogluconate, a SHP-1 inhibitor, to DCs differentiating from monocyte precursors restored their capacity to respond to LPS in approximately 60% of patients. The monocyte and DC NF-kappaB response to LPS is thus a novel phenotypic and likely pathogenetic marker for human T1DM. SHP-1 is at least one NF-kappaB regulatory mechanism which might be induced as a result of abnormal inflammatory signaling responses in T1DM monocytes.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/pathology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Monocytes/immunology , Monocytes/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Adolescent , Adult , Aged , Antigen Presentation/immunology , Cell Differentiation/immunology , Cells, Cultured , Child , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Humans , Lymphocyte Activation/immunology , MAP Kinase Signaling System/immunology , Male , Middle Aged , Monocytes/metabolism , NF-kappa B/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
16.
Immunol Cell Biol ; 85(5): 370-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17372611

ABSTRACT

Sequence variation in the Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) oncogene structure may affect antigen-presenting cell (APC) function of infected B cells and immune escape by EBV-specific T cells and thus contribute to the development of malignancy. Normal B cell-associated LMP1 (B-LMP1) upregulates B cell APC function through activation of the necrosis factor (NF)-kappaB subunit, RelB. We examined the ability of B-LMP1 and a nasopharyngeal carcinoma-associated LMP1 (NPC-LMP1) to modulate B cell APC function and T-cell responses. B lymphoma cells transfected with NPC-LMP1 stimulated resting T cells in mixed lymphocyte reaction less efficiently than B-LMP1 transfectants. Unexpectedly, antigen presentation to CD4(+) T helper cells was reduced owing to potentiation of regulatory T-cell function by NPC-LMP1 transfectants, which produce increased levels of interleukin-10, rendering CD4(+) T cells hyporesponsive. Thus, after primary EBV infection, T cells may escape activation by NPC-LMP1. These observations have important implications for the establishment of EBV-associated malignancy in the context of infection with tumour-associated EBV LMP1 variants.


Subject(s)
Nasopharyngeal Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Viral Matrix Proteins/immunology , Antigen-Presenting Cells/drug effects , B-Lymphocytes/drug effects , CD4 Antigens/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Humans , Interleukin-10/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Mutation/genetics , NF-kappa B/metabolism , Protein Transport/drug effects , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , Transfection
17.
J Immunother ; 29(6): 606-15, 2006.
Article in English | MEDLINE | ID: mdl-17063123

ABSTRACT

Fever is an evolutionarily conserved mechanism to improve survival during infection. Previous studies have shown that feverlike temperatures directly enhance the function of murine bone marrow-derived dendritic cells (DCs). In the present study, we examined the response of human monocyte-derived DC to 39.5 degrees C hyperthermia. When primed with toll-like receptor agonists or bacterial extract but not proinflammatory cytokines, hyperthermia specifically enhanced secretion of interleukin (IL)-12p70 by DC, without altering the secretion of IL-10, tumor necrosis factor alpha or IL-1beta. These DC induced significantly higher levels of T-cell proliferation and interferon gamma production in assays of antigen presentation and MLR. Endogenous heat-sock protein 70 colocalized with CD40 in DC exposed to hyperthermic conditions. Recombinant CD40-Fc fusion protein blocked the increase in IL-12p70 secretion by DC primed with bacterial extract and hyperthermia. Thus, DC primed with toll-like receptor-agonists respond to hyperthermia with increased IL-12p70 secretion, mediated by heat-shock protein binding and activation of CD40. The data have important applications for clinical immunotherapy and the mechanism of fever.


Subject(s)
CD40 Antigens/metabolism , Dendritic Cells/metabolism , Hot Temperature , Interleukin-12/metabolism , Toll-Like Receptors/agonists , Antigen Presentation/immunology , Antigens, Bacterial/pharmacology , Antigens, CD/metabolism , B7-2 Antigen/metabolism , CD40 Antigens/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Cytokines/metabolism , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-DR Antigens/metabolism , HSP70 Heat-Shock Proteins/metabolism , Humans , Immunoglobulins/metabolism , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Interleukin-4/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Membrane Glycoproteins/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Peptidoglycan/pharmacology , Poly I-C/pharmacology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tetanus Toxoid/immunology , CD83 Antigen
18.
J Immunol ; 176(12): 7278-87, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16751371

ABSTRACT

IL-1 is a key proinflammatory driver of several autoimmune diseases including juvenile inflammatory arthritis, diseases with mutations in the NALP/cryopyrin complex and Crohn's disease, and is genetically or clinically associated with many others. IL-1 is a pleiotropic proinflammatory cytokine; however the mechanisms by which increased IL-1 signaling promotes autoreactive T cell activity are not clear. Here we show that autoimmune-prone NOD and IL-1 receptor antagonist-deficient C57BL/6 mice both produce high levels of IL-1, which drives autoreactive effector cell expansion. IL-1beta drives proliferation and cytokine production by CD4(+)CD25(+)FoxP3(-) effector/memory T cells, attenuates CD4(+)CD25(+)FoxP3(+) regulatory T cell function, and allows escape of CD4(+)CD25(-) autoreactive effectors from suppression. Thus, inflammation or constitutive overexpression of IL-1beta in a genetically predisposed host can promote autoreactive effector T cell expansion and function, which attenuates the ability of regulatory T cells to maintain tolerance to self.


Subject(s)
Cell Proliferation , Interleukin-1/physiology , Receptors, Interleukin-2/biosynthesis , Self Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Cell Division/genetics , Cell Division/immunology , Cells, Cultured , Clonal Anergy/genetics , Clonal Anergy/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Forkhead Transcription Factors/biosynthesis , Genetic Predisposition to Disease , Immunologic Memory/genetics , Interferon-gamma/biosynthesis , Interleukin-1/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Interleukin-2/deficiency , Receptors, Interleukin-2/genetics , Self Tolerance/genetics , T-Lymphocytes, Regulatory/cytology
19.
J Virol ; 76(4): 1914-21, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11799186

ABSTRACT

Previous studies have shown that Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) is uniquely able to up-regulate the expression of the peptide transporters (referred to as TAP-1 and TAP-2) and major histocompatibility complex (MHC) class I in Burkitt's lymphoma (BL) cell lines. This up-regulation is often accompanied by a restoration of antigen-presenting function as measured by the ability of these cells to present endogenously expressed viral antigen to cytotoxic T lymphocytes. Here we show that the expression of LMP1 resulted in up-regulation and nuclear translocation of RelB that were coincident with increased expression of MHC class I in BL cells. Deletion of the C-terminal activator regions (CTARs) of LMP1 significantly impaired the abilities of LMP1 to translocate RelB into the nucleus and to up-regulate the expression of antigen-processing genes. Further analysis with single-point mutations within the CTARs confirmed that the residues critical for NF-kappa B activation directly contribute to antigen-processing function regulation in BL cells. This LMP1-mediated effect was blocked following expression of either dominant negative I kappa B alpha S32/36A, an NF-kappa B inhibitor, or antisense RelB. These observations indicate that upregulation of antigen-presenting function in B cells mediated by LMP1 is signaled through the NF-kappa B subunit RelB. The data provide a mechanism by which LMP1 modulates immunogenicity of Epstein-Barr virus-infected normal and malignant cells.


Subject(s)
Antigen Presentation/genetics , B-Lymphocytes/virology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/physiology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Viral Matrix Proteins/metabolism , Active Transport, Cell Nucleus , Antigen Presentation/physiology , B-Lymphocytes/immunology , Burkitt Lymphoma/virology , Cell Line, Transformed , Cell Nucleus/metabolism , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/genetics , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factor RelB , Transcription Factors/genetics , Tumor Cells, Cultured , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...