Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
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
2.
PLoS Pathog ; 12(12): e1006075, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27973593

ABSTRACT

Many natural prion diseases of humans and animals are considered to be acquired through oral consumption of contaminated food or pasture. Determining the route by which prions establish host infection will identify the important factors that influence oral prion disease susceptibility and to which intervention strategies can be developed. After exposure, the early accumulation and replication of prions within small intestinal Peyer's patches is essential for the efficient spread of disease to the brain. To replicate within Peyer's patches, the prions must first cross the gut epithelium. M cells are specialised epithelial cells within the epithelia covering Peyer's patches that transcytose particulate antigens and microorganisms. M cell-development is dependent upon RANKL-RANK-signalling, and mice in which RANK is deleted only in the gut epithelium completely lack M cells. In the specific absence of M cells in these mice, the accumulation of prions within Peyer's patches and the spread of disease to the brain was blocked, demonstrating a critical role for M cells in the initial transfer of prions across the gut epithelium in order to establish host infection. Since pathogens, inflammatory stimuli and aging can modify M cell-density in the gut, these factors may also influence oral prion disease susceptibility. Mice were therefore treated with RANKL to enhance M cell density in the gut. We show that prion uptake from the gut lumen was enhanced in RANKL-treated mice, resulting in shortened survival times and increased disease susceptibility, equivalent to a 10-fold higher infectious titre of prions. Together these data demonstrate that M cells are the critical gatekeepers of oral prion infection, whose density in the gut epithelium directly limits or enhances disease susceptibility. Our data suggest that factors which alter M cell-density in the gut epithelium may be important risk factors which influence host susceptibility to orally acquired prion diseases.


Subject(s)
Disease Susceptibility , Epithelial Cells , Intestinal Mucosa , Prion Diseases/metabolism , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Transcytosis/physiology
3.
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
4.
J Gen Virol ; 97(8): 1725-1738, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27193137

ABSTRACT

Prion diseases are a unique group of transmissible, chronic, neurodegenerative disorders. Following peripheral exposure (e.g. oral), prions often accumulate first within the secondary lymphoid tissues before they infect the central nervous system (CNS). Prion replication within secondary lymphoid tissues is crucial for the efficient spread of disease to the CNS. Once within the CNS, the responses of innate immune cells within it can have a significant influence on neurodegeneration and disease progression. Recently, there have been substantial advances in our understanding of how cross-talk between the host and the vast community of commensal microorganisms present at barrier surfaces such as the gut influences the development and regulation of the host's immune system. These effects are evident not only in the mucosal immune system in the gut, but also in the CNS. The actions of this microbial community (the microbiota) have many important beneficial effects on host health, from metabolism of nutrients and regulation of host development to protection from pathogen infection. However, the microbiota can also have detrimental effects in some circumstances. In this review we discuss the many and varied interactions between prions, the host and the gut microbiota. Particular emphasis is given to the ways by which changes to the composition of the commensal gut microbiota or congruent pathogen infection may influence prion disease pathogenesis and/or disease susceptibility. Understanding how these factors influence prion pathogenesis and disease susceptibility is important for assessing the risk to infection and the design of novel opportunities for therapeutic intervention.


Subject(s)
Disease Susceptibility , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Prion Diseases/immunology , Prion Diseases/pathology , Animals , Humans
5.
J Virol ; 89(18): 9532-47, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26157121

ABSTRACT

UNLABELLED: Prion diseases are infectious neurodegenerative disorders characterized by accumulations of abnormally folded cellular prion protein in affected tissues. Many natural prion diseases are acquired orally, and following exposure, the early replication of some prion isolates upon follicular dendritic cells (FDC) within gut-associated lymphoid tissues (GALT) is important for the efficient spread of disease to the brain (neuroinvasion). Prion detection within large intestinal GALT biopsy specimens has been used to estimate human and animal disease prevalence. However, the relative contributions of the small and large intestinal GALT to oral prion pathogenesis were unknown. To address this issue, we created mice that specifically lacked FDC-containing GALT only in the small intestine. Our data show that oral prion disease susceptibility was dramatically reduced in mice lacking small intestinal GALT. Although these mice had FDC-containing GALT throughout their large intestines, these tissues were not early sites of prion accumulation or neuroinvasion. We also determined whether pathology specifically within the large intestine might influence prion pathogenesis. Congruent infection with the nematode parasite Trichuris muris in the large intestine around the time of oral prion exposure did not affect disease pathogenesis. Together, these data demonstrate that the small intestinal GALT are the major early sites of prion accumulation and neuroinvasion after oral exposure. This has important implications for our understanding of the factors that influence the risk of infection and the preclinical diagnosis of disease. IMPORTANCE: Many natural prion diseases are acquired orally. After exposure, the accumulation of some prion diseases in the gut-associated lymphoid tissues (GALT) is important for efficient spread of disease to the brain. However, the relative contributions of GALT in the small and large intestines to oral prion pathogenesis were unknown. We show that the small intestinal GALT are the essential early sites of prion accumulation. Furthermore, congruent infection with a large intestinal helminth (worm) around the time of oral prion exposure did not affect disease pathogenesis. This is important for our understanding of the factors that influence the risk of prion infection and the preclinical diagnosis of disease. The detection of prions within large intestinal GALT biopsy specimens has been used to estimate human and animal disease prevalence. However, our data suggest that using these biopsy specimens may miss individuals in the early stages of oral prion infection and significantly underestimate the disease prevalence.


Subject(s)
Dendritic Cells, Follicular/immunology , Intestine, Small/immunology , Lymphoid Tissue/immunology , Prion Diseases/immunology , Prion Diseases/transmission , Prions/immunology , Animals , Dendritic Cells, Follicular/pathology , Humans , Intestine, Large/immunology , Intestine, Large/parasitology , Intestine, Large/pathology , Intestine, Small/parasitology , Intestine, Small/pathology , Lymphoid Tissue/pathology , Mice , Prion Diseases/parasitology , Prions/pathogenicity , Trichuriasis/immunology , Trichuriasis/pathology , Trichuris/immunology
6.
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
7.
Immunology ; 133(4): 482-98, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21635249

ABSTRACT

Follicular dendritic cells (FDC) are an important subset of stromal cells within the germinal centres of lymphoid tissues. They are specialized to trap and retain antigen-containing immune complexes on their surfaces to promote B-cell maturation and immunoglobulin isotype class-switching. However, little is known of the cell types from which FDC originate. To address fundamental questions associated with the relationships between FDC and other cell populations, we took advantage of the growing body of publicly available data for transcriptome analysis. We obtained a large number of gene expression data files from a range of different primary mouse cells and cell lines and subjected these data to network-based cluster analysis using BiolayoutExpress(3D) . Genes with related function clustered together in distinct regions of the graph and enabled the identification of transcriptional networks that underpin the functional activity of distinct cell populations. Several gene clusters were identified that were selectively expressed by cells of mesenchymal lineage and contained classic mesenchymal cell markers and extracellular matrix genes including various collagens, Acta2, Bgn, Fbn1 and Twist1. Our analysis showed that FDC also express highly many of these mesenchyme-associated genes. Promoter analysis of the genes comprising the mesenchymal clusters identified several regulatory motifs that are binding sites for candidate transcription factors previously known to be candidate regulators of mesenchyme-specific genes. Together, these data suggest FDC are a specialized mesenchymal cell population within the germinal centres of lymphoid tissues.


Subject(s)
Dendritic Cells, Follicular/metabolism , Extracellular Matrix/genetics , Mesoderm/metabolism , Oligonucleotide Array Sequence Analysis , Animals , Biomarkers , Cell Line , Humans , Meta-Analysis as Topic , Mice
8.
Front Immunol ; 12: 761949, 2021.
Article in English | MEDLINE | ID: mdl-34938288

ABSTRACT

The decline in mucosal immunity during aging increases susceptibility, morbidity and mortality to infections acquired via the gastrointestinal and respiratory tracts in the elderly. We previously showed that this immunosenescence includes a reduction in the functional maturation of M cells in the follicle-associated epithelia (FAE) covering the Peyer's patches, diminishing the ability to sample of antigens and pathogens from the gut lumen. Here, co-expression analysis of mRNA-seq data sets revealed a general down-regulation of most FAE- and M cell-related genes in Peyer's patches from aged mice, including key transcription factors known to be essential for M cell differentiation. Conversely, expression of ACE2, the cellular receptor for SARS-Cov-2 virus, was increased in the aged FAE. This raises the possibility that the susceptibility of aged Peyer's patches to infection with the SARS-Cov-2 virus is increased. Expression of key Paneth cell-related genes was also reduced in the ileum of aged mice, consistent with the adverse effects of aging on their function. However, the increased expression of these genes in the villous epithelium of aged mice suggested a disturbed distribution of Paneth cells in the aged intestine. Aging effects on Paneth cells negatively impact on the regenerative ability of the gut epithelium and could indirectly impede M cell differentiation. Thus, restoring Paneth cell function may represent a novel means to improve M cell differentiation in the aging intestine and increase mucosal vaccination efficacy in the elderly.


Subject(s)
COVID-19 , Immunity, Mucosal/immunology , Immunosenescence/immunology , Paneth Cells/immunology , Peyer's Patches/immunology , Animals , Cell Differentiation/immunology , Mice , Mice, Inbred C57BL , SARS-CoV-2
9.
iScience ; 23(6): 101147, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32454449

ABSTRACT

Aging has a profound effect on the immune system, termed immunosenescence, resulting in increased incidence and severity of infections and decreased efficacy of vaccinations. We previously showed that immunosurveillance in the intestine, achieved primarily through antigen sampling M cells in the follicle associated epithelium (FAE) of Peyer's patches, was compromised during aging due to a decline in M cell functional maturation. The intestinal microbiota also changes significantly with age, but whether this affects M cell maturation was not known. We show that housing of aged mice on used bedding from young mice, or treatment with bacterial flagellin, were each sufficient to enhance the functional maturation of M cells in Peyer's patches. An understanding of the mechanisms underlying the influence of the intestinal microbiota on M cells has the potential to lead to new methods to enhance the efficacy of oral vaccination in aged individuals.

10.
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
11.
Adv Exp Med Biol ; 666: 1-18, 2009.
Article in English | MEDLINE | ID: mdl-20054971

ABSTRACT

Bacterial toxins are the causative agent at pathology in a variety of diseases. Although not always the primary target of these toxins, many have been shown to have potent immunomodulatory effects, for example, inducing immune responses to co-administered antigens and suppressing activation of immune cells. These abilities of bacterial toxins can be harnessed and used in a therapeutic manner, such as in vaccination or the treatment of autoimmune diseases. Furthermore, the ability of toxins to gain entry to cells can be used in novel bacterial toxin based immuno-therapies in order to deliver antigens into MHC Class I processing pathways. Whether the immunomodulatory properties of these toxins arose in order to enhance bacterial survival within hosts, to aid spread within the population or is pure serendipity, it is interesting to think that these same toxins potentially hold the key to preventing or treating human disease.


Subject(s)
Autoimmune Diseases/drug therapy , Bacterial Toxins/pharmacology , Immunologic Factors/pharmacology , Humans
12.
J Manipulative Physiol Ther ; 32(9): 781-91, 2009.
Article in English | MEDLINE | ID: mdl-20004807

ABSTRACT

OBJECTIVE: The purpose of this study is to describe the musculoskeletal rehabilitation model used to care for combat and severely wounded or ill US military service members at an integrated Comprehensive Combat and Complex Casualty Care center located at Naval Medical Center San Diego. METHODS: Through a collaborative and iterative process, providers from the various services included at the Comprehensive Combat and Complex Casualty Care program developed a description of the integration of services provided at this location. RESULTS: After construction of the facility in 2007, the program has provided services for approximately 2 years. Eighteen different health care providers from 10 different specialties provide integrated musculoskeletal services, which include primary care, physical therapy, occupational therapy, vestibular therapy, gait analysis, prosthetics, recreational therapy, and chiropractic care. At the time of this writing (early 2009), the program had provided musculoskeletal rehabilitation care to approximately 500 patients, 58 with amputations, from the operational theater, Veterans Affairs, other military treatment facilities, and local trauma centers. CONCLUSION: The complex nature of combat wounded and polytrauma patients requires an integrated and interdisciplinary team that is innovative, adaptable, and focused on the needs of the patient. This article presents a description of the model and the experiences of our musculoskeletal rehabilitation team; it is our hope that this article will assist other centers and add to the small but emerging literature on this topic.


Subject(s)
Comprehensive Health Care/organization & administration , Delivery of Health Care, Integrated/organization & administration , Health Services Administration , Military Medicine/organization & administration , Musculoskeletal Diseases/rehabilitation , Warfare , California , Humans , Interprofessional Relations , Medical Staff , Medicine/organization & administration , Occupational Therapy/organization & administration , Physical Therapy Specialty/organization & administration , Physical and Rehabilitation Medicine/organization & administration
13.
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
14.
Front Immunol ; 10: 2495, 2019.
Article in English | MEDLINE | ID: mdl-31695701

ABSTRACT

The follicle-associated epithelium (FAE) is a specialized structure that samples luminal antigens and transports them into mucosa-associated lymphoid tissues (MALT). In mammals, transcytosis of antigens across the gut epithelium is performed by a subset of FAE cells known as M cells. Here we show that colony-stimulating factor 1 receptor (CSF1R) is expressed by a subset of cells in the avian bursa of Fabricius FAE. Expression was initially detected using a CSF1R-reporter transgene that also label subsets of bursal macrophages. Immunohistochemical detection using a specific monoclonal antibody confirmed abundant expression of CSF1R on the basolateral membrane of FAE cells. CSF1R-transgene expressing bursal FAE cells were enriched for expression of markers previously reported as putative M cell markers, including annexin A10 and CD44. They were further distinguished from a population of CSF1R-transgene negative epithelial cells within FAE by high apical F-actin expression and differential staining with the lectins jacalin, PHA-L and SNA. Bursal FAE cells that express the CSF1R-reporter transgene were responsible for the bulk of FAE transcytosis of labeled microparticles in the size range 0.02-0.1 µm. Unlike mammalian M cells, they did not readily take up larger bacterial sized microparticles (0.5 µm). Their role in uptake of bacteria was tested using Salmonella, which can enter via M cells in mammals. Labeled Salmonella enterica serovar Typhimurium entered bursal tissue via the FAE. Entry was partially dependent upon Type III secretion system-1. However, the majority of invading bacteria were localized to CSF1R-negative FAE cells and in resident phagocytes that express the phosphatidylserine receptor TIM4. CSF1R-expressing FAE cells in infected follicles showed evidence of cell death and shedding into the bursal lumen. In mammals, CSF1R expression in the gut is restricted to macrophages which only indirectly control M cell differentiation. The novel expression of CSF1R in birds suggests that these functional equivalents to mammalian M cells may have different ontological origins and their development and function are likely to be regulated by different growth factors.


Subject(s)
Antigen Presentation/immunology , Avian Proteins/immunology , Bursa of Fabricius/immunology , Epithelial Cells/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Antigens, Bacterial , Antigens, Differentiation/immunology , Bursa of Fabricius/pathology , Chickens , Humans , Salmonella Infections/pathology
15.
Nat Commun ; 9(1): 1272, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29593242

ABSTRACT

Colony-stimulating factor 1 (CSF1) controls the growth and differentiation of macrophages.CSF1R signaling has been implicated in the maintenance of the intestinal stem cell niche and differentiation of Paneth cells, but evidence of expression of CSF1R within the crypt is equivocal. Here we show that CSF1R-dependent macrophages influence intestinal epithelial differentiation and homeostasis. In the intestinal lamina propria CSF1R mRNA expression is restricted to macrophages which are intimately associated with the crypt epithelium, and is undetectable in Paneth cells. Macrophage ablation following CSF1R blockade affects Paneth cell differentiation and leads to a reduction of Lgr5+ intestinal stem cells. The disturbances to the crypt caused by macrophage depletion adversely affect the subsequent differentiation of intestinal epithelial cell lineages. Goblet cell density is enhanced, whereas the development of M cells in Peyer's patches is impeded. We suggest that modification of the phenotype or abundance of macrophages in the gut wall alters the development of the intestinal epithelium and the ability to sample gut antigens.


Subject(s)
Intestinal Mucosa/metabolism , Macrophages/cytology , Mucous Membrane/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Differentiation , Cell Lineage , Female , Goblet Cells/metabolism , Green Fluorescent Proteins/metabolism , Homeostasis , Intestines , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Paneth Cells/cytology , Peyer's Patches/cytology , Phenotype , RNA, Messenger/metabolism , Signal Transduction , Stem Cell Niche , Stem Cells/cytology
16.
Immunobiology ; 222(2): 316-326, 2017 02.
Article in English | MEDLINE | ID: mdl-27663963

ABSTRACT

M cells reside within the follicle-associated epithelium (FAE) overlying the gut-associated lymphoid tissues. These unique phagocytic epithelial cells enable the mucosal immune system to sample antigens within the lumen of the intestine. The differentiation of M cells from uncommitted precursors in the FAE is dependent on the production of receptor activator of nuclear factor-κB ligand (RANKL) by subepithelial stromal cells. The ligation of a variety of cell surface receptors activates the nuclear factor-κB (NF-κB) family of transcription factors which in-turn induce the transcription of multiple target genes. RANKL-stimulation can stimulate the nuclear translocation of the NF-κB subunit c-Rel. We therefore used c-Rel-deficient mice to determine whether the differentiation and functional maturation of M cells in the Peyer's patches was dependent on c-Rel. Our data show that c-Rel-deficiency does not influence the expression of RANKL or RANK in Peyer's patches, or the induction of M-cell differentiation in the FAE. RANKL-stimulation in the differentiating M cells induces the expression of SpiB which is essential for their subsequent maturation. However, SpiB expression in the FAE was also unaffected in the absence of c-Rel. As a consequence, the functional maturation of M cells was not impaired in the Peyer's patches of c-Rel-deficient mice. Although our data showed that the specific expression of CCL20 and ubiquitin D in the FAE was not impeded in the absence of c-Rel, the expression of ubiquitin D was dramatically reduced in the B cell-follicles of c-Rel-deficient mice. Coincident with this, we also observed that the status of follicular dendritic cells in the B cell-follicles was dramatically reduced in Peyer's patches from c-Rel-deficient mice. Taken together, our data show that c-Rel is dispensable for the RANKL-mediated differentiation and functional maturation of M cells.


Subject(s)
Cell Differentiation/genetics , Peyer's Patches/cytology , Peyer's Patches/physiology , Proto-Oncogene Proteins c-rel/genetics , Proto-Oncogene Proteins c-rel/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Profiling , Mice , Mice, Knockout , Organ Specificity/genetics , Phenotype , RANK Ligand/genetics , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptor Activator of Nuclear Factor-kappa B/metabolism , Transcriptome , Transcytosis/genetics , Transcytosis/immunology
17.
Nat Nanotechnol ; 10(4): 361-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751305

ABSTRACT

In humans and other mammals it is known that calcium and phosphate ions are secreted from the distal small intestine into the lumen. However, why this secretion occurs is unclear. Here, we show that the process leads to the formation of amorphous magnesium-substituted calcium phosphate nanoparticles that trap soluble macromolecules, such as bacterial peptidoglycan and orally fed protein antigens, in the lumen and transport them to immune cells of the intestinal tissue. The macromolecule-containing nanoparticles utilize epithelial M cells to enter Peyer's patches, small areas of the intestine concentrated with particle-scavenging immune cells. In wild-type mice, intestinal immune cells containing these naturally formed nanoparticles expressed the immune tolerance-associated molecule 'programmed death-ligand 1', whereas in NOD1/2 double knockout mice, which cannot recognize peptidoglycan, programmed death-ligand 1 was undetected. Our results explain a role for constitutively formed calcium phosphate nanoparticles in the gut lumen and show how this helps to shape intestinal immune homeostasis.


Subject(s)
Antigens/immunology , Intestines/cytology , Intestines/immunology , Peptidoglycan/immunology , Peyer's Patches/immunology , Phosphates/immunology , Animals , Calcium/immunology , Calcium Phosphates/immunology , Cells, Cultured , Humans , Intestines/chemistry , Mice , Mice, Inbred BALB C , Minerals/immunology , Molecular Chaperones/immunology , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Particle Size , Peyer's Patches/cytology
18.
PLoS One ; 9(8): e105429, 2014.
Article in English | MEDLINE | ID: mdl-25137049

ABSTRACT

The MacBlue transgenic mouse uses the Csf1r promoter and first intron to drive expression of gal4-VP16, which in turn drives a cointegrated gal4-responsive UAS-ECFP cassette. The Csf1r promoter region used contains a deletion of a 150 bp conserved region covering trophoblast and osteoclast-specific transcription start sites. In this study, we examined expression of the transgene in embryos and adult mice. In embryos, ECFP was expressed in the large majority of macrophages derived from the yolk sac, and as the liver became a major site of monocytopoiesis. In adults, ECFP was detected at high levels in both Ly6C+ and Ly6C- monocytes and distinguished them from Ly6C+, F4/80+, CSF1R+ immature myeloid cells in peripheral blood. ECFP was also detected in the large majority of microglia and Langerhans cells. However, expression was lost from the majority of tissue macrophages, including Kupffer cells in the liver and F4/80+ macrophages of the lung, kidney, spleen and intestine. The small numbers of positive cells isolated from the liver resembled blood monocytes. In the gut, ECFP+ cells were identified primarily as classical dendritic cells or blood monocytes in disaggregated cell preparations. Immunohistochemistry showed large numbers of ECFP+ cells in the Peyer's patch and isolated lymphoid follicles. The MacBlue transgene was used to investigate the effect of treatment with CSF1-Fc, a form of the growth factor with longer half-life and efficacy. CSF1-Fc massively expanded both the immature myeloid cell (ECFP-) and Ly6C+ monocyte populations, but had a smaller effect on Ly6C- monocytes. There were proportional increases in ECFP+ cells detected in lung and liver, consistent with monocyte infiltration, but no generation of ECFP+ Kupffer cells. In the gut, there was selective infiltration of large numbers of cells into the lamina propria and Peyer's patches. We discuss the use of the MacBlue transgene as a marker of monocyte/macrophage/dendritic cell differentiation.


Subject(s)
Biomarkers/metabolism , Kupffer Cells/metabolism , Langerhans Cells/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/metabolism , Transgenes/genetics , Animals , Cell Differentiation/genetics , Female , Green Fluorescent Proteins/genetics , Intestinal Mucosa/metabolism , Kidney/metabolism , Langerhans Cells/drug effects , Liver/metabolism , Male , Mice , Mice, Transgenic/genetics , Microglia/metabolism , Peyer's Patches/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Spleen/metabolism , Trans-Activators/genetics , Yolk Sac/metabolism
19.
DNA Res ; 19(5): 407-22, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22991451

ABSTRACT

The follicle-associated epithelium (FAE) overlying the Peyer's patches and the microfold cells (M cells) within it are important sites of antigen transcytosis across the intestinal epithelium. Using a meta-analysis approach, we identified a transcriptional signature that distinguished the FAE from a large collection of mouse cells and tissues. A co-expressed cluster of 21 FAE-specific genes was identified, and the analysis of the transcription factor binding site motifs in their promoter regions indicated that these genes shared an underlying transcriptional programme. This cluster contained known FAE- (Anxa10, Ccl20, Psg18 and Ubd) and M-cell-specific (Gp2) genes, suggesting that the others were novel FAE-specific genes. Some of these novel candidate genes were expressed highly by the FAE and M cells (Calcb, Ces3b, Clca2 and Gjb2), and others only by the FAE (Ascl2, Cftr, Fgf15, Gpr133, Kcna1, Kcnj15, Mycl1, Pgap1 and Rps6kl). We also identified a subset of novel FAE-related genes that were induced in the intestinal epithelium after receptor activator of nuclear factor (NF)-κB ligand stimulation. These included Mfge8 which was specific to FAE enterocytes. This study provides new insight into the FAE transcriptome. Further characterization of the candidate genes identified here will aid the identification of novel regulators of cell function in the FAE.


Subject(s)
Intestinal Mucosa/metabolism , Transcriptome , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Connexin 26 , Connexins , Enterocytes/metabolism , Mice , Milk Proteins/genetics , Milk Proteins/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Organ Specificity , Peyer's Patches/metabolism
20.
Cell Host Microbe ; 12(5): 645-56, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23159054

ABSTRACT

Salmonella Typhimurium specifically targets antigen-sampling microfold (M) cells to translocate across the gut epithelium. Although M cells represent a small proportion of the specialized follicular-associated epithelium (FAE) overlying mucosa-associated lymphoid tissues, their density increases during Salmonella infection, but the underlying molecular mechanism remains unclear. Using in vitro and in vivo infection models, we demonstrate that the S. Typhimurium type III effector protein SopB induces an epithelial-mesenchymal transition (EMT) of FAE enterocytes into M cells. This cellular transdifferentiation is a result of SopB-dependent activation of Wnt/ß-catenin signaling leading to induction of both receptor activator of NF-κB ligand (RANKL) and its receptor RANK. The autocrine activation of RelB-expressing FAE enterocytes by RANKL/RANK induces the EMT-regulating transcription factor Slug that marks epithelial transdifferentiation into M cells. Thus, via the activity of a single secreted effector, S. Typhimurium transforms primed epithelial cells into M cells to promote host colonization and invasion.


Subject(s)
Enterocytes/cytology , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition , Intestinal Mucosa/microbiology , Salmonella typhimurium/pathogenicity , Aminophenols/pharmacology , Animals , Bacterial Proteins/metabolism , Benzylamines/pharmacology , Cell Differentiation , Cell Transdifferentiation , Cells, Cultured , Chromones/pharmacology , Enterocytes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Intestinal Mucosa/metabolism , Maleimides/pharmacology , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Peptides/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Quinoxalines/pharmacology , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Snail Family Transcription Factors , Transcription Factor RelB/biosynthesis , Transcription Factor RelB/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/biosynthesis , Transcription Factors/metabolism , Vimentin/antagonists & inhibitors , Vimentin/biosynthesis , Wnt Proteins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL