Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 13749, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877012

ABSTRACT

Prion diseases are fatal, infectious, neurodegenerative disorders resulting from accumulation of misfolded cellular prion protein in the brain. Early pathological changes during CNS prion disease also include reactive astrocyte activation with increased CD44 expression, microgliosis, as well as loss of dendritic spines and synapses. CD44 is a multifunctional cell surface adhesion and signalling molecule which is considered to play roles in astrocyte morphology and the maintenance of dendritic spine integrity and synaptic plasticity. However, the role of CD44 in prion disease was unknown. Here we used mice deficient in CD44 to determine the role of CD44 during prion disease. We show that CD44-deficient mice displayed no difference in their response to CNS prion infection when compared to wild type mice. Furthermore, the reactive astrocyte activation and microgliosis that accompanies CNS prion infection was unimpaired in the absence of CD44. Together, our data show that although CD44 expression is upregulated in reactive astrocytes during CNS prion disease, it is dispensable for astrocyte and microglial activation and the development of prion neuropathogenesis.


Subject(s)
Astrocytes , Hyaluronan Receptors , Prion Diseases , Animals , Astrocytes/metabolism , Astrocytes/pathology , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Prion Diseases/metabolism , Prion Diseases/pathology , Prion Diseases/genetics , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Brain/metabolism , Brain/pathology , Mice, Inbred C57BL
2.
Front Cell Neurosci ; 16: 918883, 2022.
Article in English | MEDLINE | ID: mdl-35875357

ABSTRACT

Prion diseases such as Creutzfeldt-Jakob disease in humans, bovine spongiform encephalopathy in cattle, and scrapie in sheep, are infectious and chronic neurodegenerative diseases to which there are no cures. Infection with prions in the central nervous system (CNS) ultimately causes extensive neurodegeneration, and this is accompanied by prominent microglial and astrocytic activation in affected regions. The microglia are the CNS macrophages and help maintain neuronal homeostasis, clear dead or dying cells and provide defense against pathogens. The microglia also provide neuroprotection during CNS prion disease, but their pro-inflammatory activation may exacerbate the development of the neuropathology. Innate immune tolerance induced by consecutive systemic bacterial lipopolysaccharide (LPS) treatment can induce long-term epigenetic changes in the microglia in the brain that several months later can dampen their responsiveness to subsequent LPS treatment and impede the development of neuritic damage in a transgenic mouse model of Alzheimer's disease-like pathology. We therefore reasoned that innate immune tolerance in microglia might similarly impede the subsequent development of CNS prion disease. To test this hypothesis groups of mice were first infected with prions by intracerebral injection, and 35 days later given four consecutive systemic injections with LPS to induce innate immune tolerance. Our data show that consecutive systemic LPS treatment did not affect the subsequent development of CNS prion disease. Our data suggests innate immune tolerance in microglia does not influence the subsequent onset of prion disease-induced neuropathology in mice, despite previously published evidence of this effect in an Alzheimer's disease mouse model.

3.
Glia ; 70(11): 2169-2187, 2022 11.
Article in English | MEDLINE | ID: mdl-35852018

ABSTRACT

Prion diseases are transmissible, neurodegenerative disorders associated with misfolding of the prion protein. Previous studies show that reduction of microglia accelerates central nervous system (CNS) prion disease and increases the accumulation of prions in the brain, suggesting that microglia provide neuroprotection by phagocytosing and destroying prions. In Csf1rΔFIRE mice, the deletion of an enhancer within Csf1r specifically blocks microglia development, however, their brains develop normally and show none of the deficits reported in other microglia-deficient models. Csf1rΔFIRE mice were used as a refined model in which to study the impact of microglia-deficiency on CNS prion disease. Although Csf1rΔFIRE mice succumbed to CNS prion disease much earlier than wild-type mice, the accumulation of prions in their brains was reduced. Instead, astrocytes displayed earlier, non-polarized reactive activation with enhanced phagocytosis of neuronal contents and unfolded protein responses. Our data suggest that rather than simply phagocytosing and destroying prions, the microglia instead provide host-protection during CNS prion disease and restrict the harmful activities of reactive astrocytes.


Subject(s)
Prion Diseases , Prions , Animals , Astrocytes/metabolism , Brain/metabolism , Mice , Microglia/metabolism , Prion Diseases/metabolism , Prions/genetics , Prions/metabolism
4.
Development ; 147(23)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323375

ABSTRACT

The central nervous system hosts parenchymal macrophages, known as microglia, and non-parenchymal macrophages, collectively termed border-associated macrophages (BAMs). Microglia, but not BAMs, were reported to be absent in mice lacking a conserved Csf1r enhancer: the fms-intronic regulatory element (FIRE). However, it is unknown whether FIRE deficiency also impacts BAM arrival and/or maintenance. Here, we show that macrophages in the ventricular system of the brain, including Kolmer's epiplexus macrophages, are absent in Csf1rΔFIRE/ΔFIRE mice. Stromal choroid plexus BAMs are also considerably reduced. During normal development, we demonstrate that intracerebroventricular macrophages arrive from embryonic day 10.5, and can traverse ventricular walls in embryonic slice cultures. In Csf1rΔFIRE/ΔFIRE embryos, the arrival of both primitive microglia and intracerebroventricular macrophages was eliminated, whereas the arrival of cephalic mesenchyme and stromal choroid plexus BAMs was only partially restricted. Our results provide new insights into the development and regulation of different CNS macrophage populations.


Subject(s)
Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Macrophages/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Brain/growth & development , Brain/metabolism , Central Nervous System/growth & development , Embryo, Mammalian , Introns/genetics , Mice , Microglia/metabolism , Parenchymal Tissue/growth & development , Parenchymal Tissue/metabolism , Regulatory Sequences, Nucleic Acid
5.
Cell Rep ; 33(1): 108232, 2020 10 06.
Article in English | MEDLINE | ID: mdl-33027650

ABSTRACT

T follicular helper (Tfh) cells are crucial for the establishment of germinal centers (GCs) and potent antibody responses. Nevertheless, the T cell-intrinsic factors that are required for the maintenance of already-established Tfh cells and GCs remain largely unknown. Here, we use temporally guided gene ablation in CD4+ T cells to dissect the contributions of the Tfh-associated chemokine receptor CXCR5 and the transcription factor Bcl6. Induced ablation of Cxcr5 has minor effects on the function of established Tfh cells, and Cxcr5-ablated cells still exhibit most of the features of CXCR5+ Tfh cells. In contrast, continued Bcl6 expression is critical to maintain the GC Tfh cell phenotype and also the GC reaction. Importantly, Bcl6 ablation during acute viral infection results in the transdifferentiation of established Tfh into Th1 cells, thus highlighting the plasticity of Tfh cells. These findings have implications for strategies that boost or restrain Tfh cells and GCs in health and disease.


Subject(s)
Proto-Oncogene Proteins c-bcl-6/metabolism , T Follicular Helper Cells/metabolism , Th1 Cells/immunology , Virus Diseases/immunology , Acute Disease , Cell Differentiation , Humans
6.
Int J Mol Sci ; 21(19)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023255

ABSTRACT

Prion diseases are a unique group of infectious chronic neurodegenerative disorders to which there are no cures. Although prion infections do not stimulate adaptive immune responses in infected individuals, the actions of certain immune cell populations can have a significant impact on disease pathogenesis. After infection, the targeting of peripherally-acquired prions to specific immune cells in the secondary lymphoid organs (SLO), such as the lymph nodes and spleen, is essential for the efficient transmission of disease to the brain. Once the prions reach the brain, interactions with other immune cell populations can provide either host protection or accelerate the neurodegeneration. In this review, we provide a detailed account of how factors such as inflammation, ageing and pathogen co-infection can affect prion disease pathogenesis and susceptibility. For example, we discuss how changes to the abundance, function and activation status of specific immune cell populations can affect the transmission of prion diseases by peripheral routes. We also describe how the effects of systemic inflammation on certain glial cell subsets in the brains of infected individuals can accelerate the neurodegeneration. A detailed understanding of the factors that affect prion disease transmission and pathogenesis is essential for the development of novel intervention strategies.


Subject(s)
Brain/immunology , Immune System/immunology , Prion Diseases/immunology , Prions/immunology , Aging/immunology , Aging/pathology , Brain/metabolism , Disease Susceptibility , Humans , Immune System/metabolism , Immunomodulation/genetics , Prion Diseases/genetics , Prion Diseases/pathology , Prions/genetics
7.
Front Immunol ; 11: 1118, 2020.
Article in English | MEDLINE | ID: mdl-32582198

ABSTRACT

Infection of the mammalian host with African trypanosomes begins when the tsetse fly vector injects the parasites into the skin dermis during blood feeding. After injection into the skin, trypanosomes first accumulate in the draining lymph node before disseminating systemically. Whether this early accumulation within the draining lymph node is important for the trypanosomes to establish infection was not known. Lymphotoxin-ß-deficient mice (LTß-/- mice) lack most secondary lymphoid tissues, but retain the spleen and mesenteric lymph nodes. These mice were used to test the hypothesis that the establishment of infection after intradermal (ID) T. brucei infection would be impeded in the absence of the skin draining lymph nodes. However, LTß-/- mice revealed greater susceptibility to ID T. brucei infection than wild-type mice, indicating that the early accumulation of the trypanosomes in the draining lymph nodes was not essential to establish systemic infection. Although LTß-/- mice were able to control the first parasitemia wave as effectively as wild-type mice, they were unable to control subsequent parasitemia waves. LTß-/- mice also lack organized B cell follicles and germinal centers within their remaining secondary lymphoid tissues. As a consequence, LTß-/- mice have impaired immunoglobulin (Ig) isotype class-switching responses. When the disturbed microarchitecture of the B cell follicles in the spleens of LTß-/- mice was restored by reconstitution with wild-type bone marrow, their susceptibility to ID T. brucei infection was similar to that of wild-type control mice. This effect coincided with the ability to produce significant serum levels of Ig isotype class-switched parasite-specific antibodies. Thus, our data suggest that organized splenic microarchitecture and the production of parasite-specific Ig isotype class-switched antibodies are essential for the control of ID African trypanosome infections.


Subject(s)
Lymph Nodes/immunology , Skin/parasitology , Spleen/immunology , Trypanosomiasis, African/immunology , Animals , Antibodies, Protozoan , Female , Lymphotoxin-beta/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/immunology , Spleen/parasitology , Trypanosoma brucei brucei
8.
Sci Rep ; 10(1): 4554, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165661

ABSTRACT

Prion infections in the central nervous system (CNS) can cause extensive neurodegeneration. Systemic inflammation can affect the progression of some neurodegenerative disorders. Therefore, we used the gastrointestinal helminth pathogen Trichuris muris to test the hypothesis that a chronic systemic inflammatory response to a gastrointestinal infection would similarly affect CNS prion disease pathogenesis. Mice were injected with prions directly into the CNS and subsequently orally co-infected with T. muris before the onset of clinical signs. We show that co-infection with a low dose of T. muris that leads to the development of a chronic T helper cell type 1-polarized systemic immune response accelerated the onset of clinical prion disease. In contrast, co-infection with a high dose of T. muris that induces a T helper cell type 2-polarized immune response did not affect prion disease pathogenesis. The reduced survival times in mice co-infected with a low dose of T. muris on d 105 after CNS prion infection coincided with enhanced astrocyte activation in the brain during the preclinical phase. These data aid our understanding of how systemic inflammation may augment the progression of neurodegeneration in the CNS.


Subject(s)
Gastrointestinal Diseases/parasitology , Prion Diseases/pathology , Th1 Cells/metabolism , Th2 Cells/metabolism , Trichuriasis/immunology , Animals , Cell Polarity , Central Nervous System/immunology , Central Nervous System/pathology , Coinfection , Disease Models, Animal , Disease Progression , Female , Gastrointestinal Diseases/immunology , Mice , Prion Diseases/immunology , Prion Diseases/parasitology , Trichuriasis/parasitology
9.
Sci Rep ; 9(1): 19119, 2019 12 13.
Article in English | MEDLINE | ID: mdl-31836813

ABSTRACT

Prion diseases are a unique, infectious, neurodegenerative disorders that can affect animals and humans. Data from mouse transmissions show that efficient infection of the host after intravenous (IV) prion exposure is dependent upon the early accumulation and amplification of the prions on stromal follicular dendritic cells (FDC) in the B cell follicles. How infectious prions are initially conveyed from the blood-stream to the FDC in the spleen is uncertain. Addressing this issue is important as susceptibility to peripheral prion infections can be reduced by treatments that prevent the early accumulation of prions upon FDC. The marginal zone (MZ) in the spleen contains specialized subsets of B cells and macrophages that are positioned to continuously monitor the blood-stream and remove pathogens, toxins and apoptotic cells. The continual shuttling of MZ B cells between the MZ and the B-cell follicle enables them to efficiently capture and deliver blood-borne antigens and antigen-containing immune complexes to splenic FDC. We tested the hypothesis that MZ B cells also play a role in the initial shuttling of prions from the blood-stream to FDC. MZ B cells were temporarily depleted from the MZ by antibody-mediated blocking of integrin function. We show that depletion of MZ B cells around the time of IV prion exposure did not affect the early accumulation of blood-borne prions upon splenic FDC or reduce susceptibility to IV prion infection. In conclusion, our data suggest that the initial delivery of blood-borne prions to FDC in the spleen occurs independently of MZ B cells.


Subject(s)
B-Lymphocytes/cytology , Lymphoma, B-Cell, Marginal Zone/metabolism , Prion Diseases/physiopathology , Prion Proteins/metabolism , Animals , Antigen-Antibody Complex/immunology , Apoptosis , Dendritic Cells, Follicular/immunology , Lymphoid Tissue/immunology , Lymphoma, B-Cell, Marginal Zone/immunology , Macrophages/cytology , Mice , Mice, Inbred C57BL , Prion Diseases/immunology , Prions/immunology , Spleen/immunology
10.
Front Cell Neurosci ; 13: 411, 2019.
Article in English | MEDLINE | ID: mdl-31551718

ABSTRACT

Prion diseases or transmissible spongiform encephalopathies are fatal, progressive, neurodegenerative, protein-misfolding disorders. Prion diseases may arise spontaneously, be inherited genetically or be acquired by infection and affect a variety of mammalian species including humans. Prion infections in the central nervous system (CNS) cause extensive neuropathology, including abnormal accumulations of misfolded host prion protein, vacuolar change resulting in sponge-like (spongiform) appearance of CNS tissue, neurodegeneration and reactive glial responses. Many different prion agent strains exist and these can differ based on disease duration, clinical signs and the targeting and distribution of the neuropathology in distinct brain areas. Reactive astrocytes are a prominent feature in the prion disease affected CNS as revealed by distinct morphological changes and upregulation of glial fibrillary acidic protein (GFAP). The CD44 antigen is a transmembrane glycoprotein involved in cell-cell interactions, cell adhesion and migration. Here we show that CD44 is also highly expressed in a subset of reactive astrocytes in regions of the CNS targeted by prions. Astrocyte heterogeneity revealed by differential CD44 upregulation occurs coincident with the earliest neuropathological changes during the pre-clinical phase of disease, and is not affected by the route of infection. The expression and distribution of CD44 was compared in brains from a large collection of 15 distinct prion agent strains transmitted to mice of different prion protein (Prnp) genotype backgrounds. Our data show that the pattern of CD44 upregulation observed in the hippocampus in each prion agent strain and host Prnp genotype combination was unique. Many mouse-adapted prion strains and hosts have previously been characterized based on the pattern of the distribution of the spongiform pathology or the misfolded PrP deposition within the brain. Our data show that CD44 expression also provides a reliable discriminatory marker of prion infection with a greater dynamic range than misfolded prion protein deposition, aiding strain identification. Together, our data reveal CD44 as a novel marker to detect reactive astrocyte heterogeneity during CNS prion disease and for enhanced identification of distinct prion agent strains.

11.
Sci Rep ; 9(1): 6674, 2019 04 30.
Article in English | MEDLINE | ID: mdl-31040320

ABSTRACT

The early replication of some orally-acquired prion strains upon stromal-derived follicular dendritic cells (FDC) within the small intestinal Peyer's patches is essential to establish host infection, and for the disease to efficiently spread to the brain. Factors that influence the early accumulation of prions in Peyer's patches can directly influence disease pathogenesis. The host's immune response to a gastrointestinal helminth infection can alter susceptibility to co-infection with certain pathogenic bacteria and viruses. Here we used the natural mouse small intestine-restricted helminth pathogen Heligmosomoides polygyrus to test the hypothesis that pathology specifically within the small intestine caused by a helminth co-infection would influence oral prion disease pathogenesis. When mice were co-infected with prions on d 8 after H. polygyrus infection the early accumulation of prions within Peyer's patches was reduced and survival times significantly extended. Natural prion susceptible hosts such as sheep, deer and cattle are regularly exposed to gastrointestinal helminth parasites. Our data suggest that co-infections with small intestine-restricted helminth pathogens may be important factors that influence oral prion disease pathogenesis.


Subject(s)
Coinfection , Disease Susceptibility , Helminthiasis, Animal/complications , Helminthiasis, Animal/parasitology , Helminths , Intestinal Diseases, Parasitic/complications , Intestinal Diseases, Parasitic/parasitology , Prion Diseases/etiology , Animals , Disease Models, Animal , Disease Susceptibility/immunology , Helminthiasis, Animal/pathology , Immunity, Mucosal , Immunohistochemistry , Intestinal Diseases, Parasitic/pathology , Mice , Peyer's Patches/immunology , Peyer's Patches/pathology
12.
J Exp Med ; 216(4): 728-742, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30814299

ABSTRACT

Intestinal immune homeostasis is dependent upon tightly regulated and dynamic host interactions with the commensal microbiota. Immunoglobulin A (IgA) produced by mucosal B cells dictates the composition of commensal bacteria residing within the intestine. While emerging evidence suggests the majority of IgA is produced innately and may be polyreactive, mucosal-dwelling species can also elicit IgA via T cell-dependent mechanisms. However, the mechanisms that modulate the magnitude and quality of T cell-dependent IgA responses remain incompletely understood. Here we demonstrate that group 3 innate lymphoid cells (ILC3) regulate steady state interactions between T follicular helper cells (TfH) and B cells to limit mucosal IgA responses. ILC3 used conserved migratory cues to establish residence within the interfollicular regions of the intestinal draining lymph nodes, where they act to limit TfH responses and B cell class switching through antigen presentation. The absence of ILC3-intrinsic antigen presentation resulted in increased and selective IgA coating of bacteria residing within the colonic mucosa. Together these findings implicate lymph node resident, antigen-presenting ILC3 as a critical regulatory checkpoint in the generation of T cell-dependent colonic IgA and suggest ILC3 act to maintain tissue homeostasis and mutualism with the mucosal-dwelling commensal microbiota.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , Colon/microbiology , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Bacteria/immunology , Colon/immunology , Female , Gastrointestinal Microbiome/immunology , Helicobacter/immunology , Histocompatibility Antigens Class II/immunology , Homeostasis/immunology , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
J Exp Med ; 216(3): 621-637, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30723095

ABSTRACT

Ectopic lymphoid structures form in a wide range of inflammatory conditions, including infection, autoimmune disease, and cancer. In the context of infection, this response can be beneficial for the host: influenza A virus infection-induced pulmonary ectopic germinal centers give rise to more broadly cross-reactive antibody responses, thereby generating cross-strain protection. However, despite the ubiquity of ectopic lymphoid structures and their role in both health and disease, little is known about the mechanisms by which inflammation is able to convert a peripheral tissue into one that resembles a secondary lymphoid organ. Here, we show that type I IFN produced after viral infection can induce CXCL13 expression in a phenotypically distinct population of lung fibroblasts, driving CXCR5-dependent recruitment of B cells and initiating ectopic germinal center formation. This identifies type I IFN as a novel inducer of CXCL13, which, in combination with other stimuli, can promote lung remodeling, converting a nonlymphoid tissue into one permissive to functional tertiary lymphoid structure formation.


Subject(s)
Chemokine CXCL13/metabolism , Germinal Center/pathology , Interferon Type I/metabolism , Orthomyxoviridae Infections/pathology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Chemokine CXCL13/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/virology , Germinal Center/drug effects , Germinal Center/metabolism , Interferon-beta/pharmacology , Lung/metabolism , Lung/pathology , Lung/virology , Male , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/metabolism , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
14.
J Virol ; 92(19)2018 10 01.
Article in English | MEDLINE | ID: mdl-30021891

ABSTRACT

The early replication of certain prion strains within Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain after oral exposure. Our data show that orally acquired prions utilize specialized gut epithelial cells known as M cells to enter Peyer's patches. M cells express the cellular isoform of the prion protein, PrPC, and this may be exploited by some pathogens as an uptake receptor to enter Peyer's patches. This suggested that PrPC might also mediate the uptake and transfer of prions across the gut epithelium into Peyer's patches in order to establish infection. Furthermore, the expression level of PrPC in the gut epithelium could influence the uptake of prions from the lumen of the small intestine. To test this hypothesis, transgenic mice were created in which deficiency in PrPC was specifically restricted to epithelial cells throughout the lining of the small intestine. Our data clearly show that efficient prion neuroinvasion after oral exposure occurred independently of PrPC expression in small intestinal epithelial cells. The specific absence of PrPC in the gut epithelium did not influence the early replication of prions in Peyer's patches or disease susceptibility. Acute mucosal inflammation can enhance PrPC expression in the intestine, implying the potential to enhance oral prion disease pathogenesis and susceptibility. However, our data suggest that the magnitude of PrPC expression in the epithelium lining the small intestine is unlikely to be an important factor which influences the risk of oral prion disease susceptibility.IMPORTANCE The accumulation of orally acquired prions within Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain. Little is known of how the prions initially establish infection within Peyer's patches. Some gastrointestinal pathogens utilize molecules, such as the cellular prion protein PrPC, expressed on gut epithelial cells to enter Peyer's patches. Acute mucosal inflammation can enhance PrPC expression in the intestine, implying the potential to enhance oral prion disease susceptibility. We used transgenic mice to determine whether the uptake of prions into Peyer's patches was dependent upon PrPC expression in the gut epithelium. We show that orally acquired prions can establish infection in Peyer's patches independently of PrPC expression in gut epithelial cells. Our data suggest that the magnitude of PrPC expression in the epithelium lining the small intestine is unlikely to be an important factor which influences oral prion disease susceptibility.


Subject(s)
Brain/metabolism , Intestine, Small/metabolism , Peyer's Patches/metabolism , PrPC Proteins/genetics , Prion Diseases/metabolism , Administration, Oral , Animals , Brain/pathology , Brain Mapping , Dendritic Cells, Follicular/metabolism , Dendritic Cells, Follicular/pathology , Disease Susceptibility , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peyer's Patches/pathology , PrPC Proteins/metabolism , Prion Diseases/mortality , Prion Diseases/pathology , Survival Analysis
15.
Parasite Immunol ; 40(8): e12566, 2018 08.
Article in English | MEDLINE | ID: mdl-29920694

ABSTRACT

Trichuris muris is a natural mouse helminth pathogen which establishes infection specifically in the caecum and proximal colon. The rapid expulsion of T. muris in resistant mouse strains is associated with the induction of a protective T helper cell type 2 (Th2)-polarized immune response. Susceptible mouse strains, in contrast, mount an inappropriate Th1 response to T. muris infection. Expression of the chemokine CXCL13 by stromal follicular dendritic cells attracts CXCR5-expressing cells towards the B-cell follicles. Previous studies using a complex in vivo depletion model have suggested that CXCR5-expressing conventional dendritic cells (cDC) help regulate the induction of Th2-polarized responses. Here, transgenic mice with CXCR5 deficiency specifically restricted to CD11c+ cells were used to determine whether the specific absence CXCR5 on CD11c+ cells such as cDC would influence susceptibility to oral T. muris infection by affecting the Th1/Th2 balance. We show that in contrast to control mice, those which lacked CXCR5 expression on CD11c+ cells failed to clear T. muris infection and developed cytokine and antibody responses that suggested a disturbed Th1/Th2 balance with enhanced IFN-γ expression. These data suggest an important role of CXCR5-expressing CD11c+ cells such as cDC in immunity to oral T. muris infection.


Subject(s)
CD11c Antigen/analysis , Receptors, CXCR5/analysis , Trichuriasis/immunology , Trichuris/immunology , Administration, Oral , Animals , Antibody Formation , B-Lymphocytes , Cytokines/analysis , Dendritic Cells/immunology , Disease Models, Animal , Disease Susceptibility , Mice , Mice, Inbred C57BL , Mice, Transgenic , Specific Pathogen-Free Organisms , Th2 Cells/immunology , Trichuriasis/parasitology
16.
J Gen Virol ; 98(7): 1943-1952, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28708055

ABSTRACT

Prion diseases are a unique group of transmissible, typically sub-acute, neurodegenerative disorders. During central nervous system (CNS) prion disease, the microglia become activated and are thought to provide a protective response by scavenging and clearing prions. The mammalian intestine is host to a large burden of commensal micro-organisms, especially bacteria, termed the microbiota. The commensal microbiota has beneficial effects on host health, including through the metabolism of essential nutrients, regulation of host development and protection against pathogens. The commensal gut microbiota also constitutively regulates the functional maturation of microglia in the CNS, and microglial function is impaired when it is absent in germ-free mice. In the current study, we determined whether the absence of the commensal gut microbiota might also affect prion disease pathogenesis. Our data clearly show that the absence of the commensal microbiota in germ-free mice did not affect prion disease duration or susceptibility after exposure to prions by intraperitoneal or intracerebral injection. Furthermore, the magnitude and distribution of the characteristic neuropathological hallmarks of terminal prion disease in the CNS, including the development of spongiform pathology, accumulation of prion disease-specific protein (PrP), astrogliosis and microglial activation, were similar in conventionally housed and germ-free mice. Thus, although the commensal gut microbiota constitutively promotes the maintenance of the microglia in the CNS under steady-state conditions in naïve mice, our data suggest that dramatic changes to the abundance or complexity of the commensal gut microbiota are unlikely to influence CNS prion disease pathogenesis.


Subject(s)
Gastrointestinal Microbiome/physiology , Germ-Free Life/physiology , Microglia/pathology , Prion Diseases/pathology , Prions/pathogenicity , Animals , Central Nervous System/pathology , Mice , Mice, Inbred C3H
17.
J Virol ; 91(10)2017 05 15.
Article in English | MEDLINE | ID: mdl-28275192

ABSTRACT

After oral exposure, the early replication of certain prion strains upon stromal cell-derived follicular dendritic cells (FDC) in the Peyer's patches in the small intestine is essential for the efficient spread of disease to the brain. However, little is known of how prions are initially conveyed from the gut lumen to establish infection on FDC. Our previous data suggest that mononuclear phagocytes such as CD11c+ conventional dendritic cells play an important role in the initial propagation of prions from the gut lumen into Peyer's patches. However, whether these cells conveyed orally acquired prions toward FDC within Peyer's patches was not known. The chemokine CXCL13 is expressed by FDC and follicular stromal cells and modulates the homing of CXCR5-expressing cells toward the FDC-containing B cell follicles. Here, novel compound transgenic mice were created in which a CXCR5 deficiency was specifically restricted to CD11c+ cells. These mice were used to determine whether CXCR5-expressing conventional dendritic cells propagate prions toward FDC after oral exposure. Our data show that in the specific absence of CXCR5-expressing conventional dendritic cells the early accumulation of prions upon FDC in Peyer's patches and the spleen was impaired, and disease susceptibility significantly reduced. These data suggest that CXCR5-expressing conventional dendritic cells play an important role in the efficient propagation of orally administered prions toward FDC within Peyer's patches in order to establish host infection.IMPORTANCE Many natural prion diseases are acquired by oral consumption of contaminated food or pasture. Once the prions reach the brain they cause extensive neurodegeneration, which ultimately leads to death. In order for the prions to efficiently spread from the gut to the brain, they first replicate upon follicular dendritic cells within intestinal Peyer's patches. How the prions are first delivered to follicular dendritic cells to establish infection was unknown. Understanding this process is important since treatments which prevent prions from infecting follicular dendritic cells can block their spread to the brain. We created mice in which mobile conventional dendritic cells were unable to migrate toward follicular dendritic cells. In these mice the early accumulation of prions on follicular dendritic cells was impaired and oral prion disease susceptibility was reduced. This suggests that prions exploit conventional dendritic cells to facilitate their initial delivery toward follicular dendritic cells to establish host infection.


Subject(s)
Dendritic Cells, Follicular/immunology , Encephalopathy, Bovine Spongiform/immunology , Encephalopathy, Bovine Spongiform/physiopathology , Gene Expression , Prions/pathogenicity , Receptors, CXCR5/genetics , Animals , Brain/pathology , Cattle , Chemokine CXCL13/genetics , Dendritic Cells/pathology , Dendritic Cells/physiology , Dendritic Cells, Follicular/pathology , Disease Susceptibility , Intestine, Small/immunology , Intestine, Small/pathology , Mice , Mice, Transgenic , Peyer's Patches/immunology , Peyer's Patches/pathology , Prions/physiology , Scrapie/physiopathology , Spleen/immunology , Spleen/pathology
18.
Virology ; 497: 337-345, 2016 10.
Article in English | MEDLINE | ID: mdl-27522473

ABSTRACT

Prion diseases are infectious neurodegenerative disorders characterised by accumulations of abnormal prion glycoprotein in affected tissues. Following peripheral exposure, many prion strains replicate upon follicular dendritic cells (FDC) in lymphoid tissues before infecting the brain. An intact splenic marginal zone is important for the efficient delivery of prions to FDC. The marginal zone contains a ring of specific intercellular adhesion molecule-3-grabbing non-integrin related 1 (SIGN-R1)-expressing macrophages. This lectin binds dextran and capsular pneumococcal polysaccharides, and also enhances the clearance of apoptotic cells via interactions with complement components. Since prions are acquired as complement-opsonized complexes we determined the role of SIGN-R1 in disease pathogenesis. We show that transient down-regulation of SIGN-R1 prior to intravenous prion exposure had no effect on the early accumulation of prions upon splenic FDC or their subsequent spread to the brain. Thus, SIGN-R1 expression by marginal zone macrophages is not rate-limiting for peripheral prion disease pathogenesis.


Subject(s)
Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Prion Diseases/etiology , Prion Diseases/metabolism , Receptors, Cell Surface/metabolism , Animals , Cell Adhesion Molecules/genetics , Dendritic Cells, Follicular/metabolism , Disease Models, Animal , Disease Susceptibility , Down-Regulation , Gene Expression Regulation , Lectins, C-Type/genetics , Macrophages/metabolism , Mice , Prion Proteins/metabolism , Receptors, Cell Surface/genetics , Spleen/metabolism
19.
Biogerontology ; 16(2): 133-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24705962

ABSTRACT

Bacterial and viral infections of the gastrointestinal tract are more common in the elderly and represent a major cause of morbidity and mortality. The mucosal immune system provides the first line of defence against pathogens acquired by ingestion and inhalation, but its function is adversely affected in the elderly. This aging-related decline in the immune function is termed immunosenescence and is associated with diminished abilities to generate protective immunity, reduced vaccine efficacy, increased incidence of cancer, inflammation and autoimmunity, and the impaired ability to generate tolerance to harmless antigens. In this review we describe our current understanding of the effects immunosenescence has on the innate and adaptive arms of the mucosal immune system in the intestine. Current estimates suggest that by the year 2050 up to 40% of the UK population will be over 65 years old, bringing with it important health challenges. A thorough understanding of the mechanisms that contribute to the development of immunosenescence is therefore crucial to help identify novel approaches to improve mucosal immunity in the elderly.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Immunosenescence/immunology , Intestinal Mucosa/immunology , Models, Immunological , Animals , Humans , Intestinal Mucosa/pathology
20.
Immunology ; 143(1): 120-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24684244

ABSTRACT

Prions are a unique group of pathogens, which are considered to comprise solely of an abnormally folded isoform of the cellular prion protein. The accumulation and replication of prions within secondary lymphoid organs is important for their efficient spread from the periphery to the brain where they ultimately cause neurodegeneration and death. Mononuclear phagocytes (MNP) play key roles in prion disease pathogenesis. Some MNP appear to facilitate the propagation of prions to and within lymphoid tissues, whereas others may aid their clearance by phagocytosis and by destroying them. Our recent data show that an intact splenic marginal zone is important for the efficient delivery of prions into the B-cell follicles where they subsequently replicate upon follicular dendritic cells before infecting the nervous system. Sialoadhesin is an MNP-restricted cell adhesion molecule that binds sialylated glycoproteins. Sialoadhesin is constitutively expressed upon splenic marginal zone metallophilic and lymph node sub-capsular sinus macrophage populations, where it may function to bind sialylated glycoproteins, pathogens and exosomes in the blood and lymph via recognition of terminal sialic acid residues. As the prion glycoprotein is highly sialylated, we tested the hypothesis that sialoadhesin may influence prion disease pathogenesis. We show that after peripheral exposure, prion pathogenesis was unaltered in sialoadhesin-deficient mice; revealing that lymphoid sequestration of prions is not mediated via sialoadhesin. Hence, although an intact marginal zone is important for the efficient uptake and delivery of prions into the B-cell follicles of the spleen, this is not influenced by sialoadhesin expression by the MNP within it.


Subject(s)
Mononuclear Phagocyte System/metabolism , Prion Diseases/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , Animals , Immunohistochemistry , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mononuclear Phagocyte System/immunology , Prion Diseases/genetics , Prion Diseases/immunology , Sialic Acid Binding Ig-like Lectin 1/genetics , Spleen/immunology , Spleen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...