1
|
McErlean EM, McCarthy HO. Non-viral approaches in CAR-NK cell engineering: connecting natural killer cell biology and gene delivery. J Nanobiotechnology 2024; 22:552. [PMID: 39256765 PMCID: PMC11384716 DOI: 10.1186/s12951-024-02746-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 08/02/2024] [Indexed: 09/12/2024] Open
Abstract
Natural Killer (NK) cells are exciting candidates for cancer immunotherapy with potent innate cytotoxicity and distinct advantages over T cells for Chimeric Antigen Receptor (CAR) therapy. Concerns regarding the safety, cost, and scalability of viral vectors has ignited research into non-viral alternatives for gene delivery. This review comprehensively analyses recent advancements and challenges with non-viral genetic modification of NK cells for allogeneic CAR-NK therapies. Non-viral alternatives including electroporation and multifunctional nanoparticles are interrogated with respect to CAR expression and translational responses. Crucially, the link between NK cell biology and design of drug delivery technologies are made, which is essential for development of future non-viral approaches. This review provides valuable insights into the current state of non-viral CAR-NK cell engineering, aimed at realising the full potential of NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Emma M McErlean
- School of Pharmacy, Queen's University of Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK.
| | - Helen O McCarthy
- School of Pharmacy, Queen's University of Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
- School of Chemical Sciences, Dublin City University, Collins Avenue, Dublin 9, Ireland
- Biodesign Europe, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
2
|
Hu J, Xiang Y, Zhu X, Hu C, Xu X, Li D, Deng Z, Jiang Z. Grass carp (Ctenopharyngodon idella) Mex3B positively regulates innate immunity by promoting the K63-linked ubiquitination of TLR3. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109023. [PMID: 37625735 DOI: 10.1016/j.fsi.2023.109023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
As a member of Mex3 (muscle excess protein-3) family, Mex3B (Mex-3 RNA binding family member B) is crucial in cell proliferation and migration in mammals. In this study, an ortholog of mammalian Mex3B (denominated CiMex3B, MT276802.1) was cloned and identified in grass carp (Ctenopharyngodon idella). CiMex3B is 1578 bp in length and encodes a polypeptide of 525 amino acids. Consistent with its mammalian counterpart, CiMex3B also contains one C-terminal RING domain and two N-terminal conserved tandem KH domains. CiMex3B up-regulates the expressions of IFN1, ISG15, MX2, as well as the expressions of inflammatory cytokines such as IL6, IL8 and TNFα in response to poly(I:C). A screening test for identifying potential targets indicated that CiMex3B is associated with TLR3 and TRIF. CiMex3B co-localizes with TLR3 in the late endosome, mitochondria and endoplasmic reticulum after poly(I:C) stimulation, whereas they are rarely discovered in the lysosomes. CiMex3B serves as a positive regulator in the phosphorylation of IRF3 and induces IFN1 expression. In addition, two truncation mutants of CiMex3B (1-220 and 221-525) were constructed to better understand the molecular mechanism of CiMex3B-mediated ubiquitination of TLR3. In line with wild-type protein, CiMex3B mutant (1-220) was found mainly in the cytoplasm; however, CiMex3B mutant (221-525) resided in the cytoplasm and the nucleus as well, and it was further confirmed that CiMex3B mutant (221-525) still interacts with TLR3. We also observed that CiMex3B promotes the K63-linked ubiquitination of TLR3, while neither of the truncation mutants (1-220 or 221-525) retains this activity. To sum up, this study revealed that CiMex3B potentiates the K63-linked ubiquitination of TLR3, and then elicits the IRF3-mediated antiviral innate immune responses.
Collapse
Affiliation(s)
- Jihuan Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, China; Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Yang Xiang
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xuechun Zhu
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Chengyu Hu
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xiaowen Xu
- Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Dongming Li
- Fuzhou Medical College, Nanchang University, Fuzhou, 344000, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Zeyin Jiang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, 330047, Jiangxi, China; Department of Bioscience, College of Life Science, Nanchang University, Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
3
|
MicroRNA expression in immune tissues of adult chickens after embryo stimulation with bioactive substances. Sci Rep 2023; 13:3076. [PMID: 36813917 PMCID: PMC9946929 DOI: 10.1038/s41598-023-30299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
The microbiota has a profound impact on the host organisms. The interaction between the host and its microbiota has an epigenetic mode of action. In poultry species, gastrointestinal microbiota might be stimulated before hatching. This stimulation with bioactive substances has a broad spectrum and long-term effects. This study aimed to examine the role of miRNA expression stimulated by host-microbiota interaction via administering a bioactive substance at the stage of embryonic development. This paper is a continuation of earlier research in the field of molecular analyzes in immune tissues after in ovo administration of bioactive substances. Eggs of Ross 308 broiler chicken and Polish native breed chicken (Green-legged Partridgelike) were incubated in the commercial hatchery. On day 12 of incubation, eggs were injected: the control group with saline (0.2 mM physiological saline), probiotic-Lactococcus lactis subsp. cremoris, prebiotic-galactooligosaccharides, and synbiotic-mentioned above prebiotic with probiotic. The birds were intended for rearing. miRNA expression analysis was performed using the miRCURY LNA miRNA PCR Assay in the spleen and tonsils of adult chickens. Six miRNAs differed significantly, at least between one pair of treatment groups. The most miRNA changes were observed in the cecal tonsils of Green-legged Partridgelike chickens. At the same time, only miR-1598 and miR-1652 showed significant differences between the treatment groups in the cecal tonsils and spleen of Ross broiler chickens. Only two miRNAs showed significant GeneOntology (GO)enrichment with the ClueGo plug-in. gga-miR-1652 target genes showed only 2 GOs significantly enriched: chondrocyte differentiation and early endosome. gga-miR-1612 target genes, the most significant GO was regulating the RNA metabolic process. The enriched functions were associated with gene expression or protein regulation, the nervous system, and the immune system. Results suggest that early microbiome stimulation in chicken might regulate the miRNA expression in different immune tissues in a genotype-dependent manner.
Collapse
|
4
|
Hsu YW, Wong HSC, Huang WC, Yeh YH, Hsiao CD, Chang WC, Hsieh SL. Human rs75776403 polymorphism links differential phenotypic and clinical outcomes to a CLEC18A p.T151M-driven multiomics. J Biomed Sci 2022; 29:43. [PMID: 35717171 PMCID: PMC9206359 DOI: 10.1186/s12929-022-00822-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/03/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human traits, diseases susceptibility, and clinical outcomes vary hugely among individuals. Despite a fundamental understanding of genetic (or environmental) contributions, the detailed mechanisms of how genetic variation impacts molecular or cellular behaviours of a gene, and subsequently leads to such variability remain poorly understood. METHODS Here, in addition to phenome-wide correlations, we leveraged multiomics to exploit mechanistic links, from genetic polymorphism to protein structural or functional changes and a cross-omics perturbation landscape of a germline variant. RESULTS We identified a missense cis-acting expression quantitative trait locus in CLEC18A (rs75776403) in which the altered residue (T151→M151) disrupts the lipid-binding ability of the protein domain. The altered allele carriage led to a metabolic and proliferative shift, as well as immune deactivation, therefore determines human anthropometrics (body height), kidney, and hematological traits. CONCLUSIONS Collectively, we uncovered genetic pleiotropy in human complex traits and diseases via CLEC18A rs75776403-regulated pathways.
Collapse
Affiliation(s)
- Yu-Wen Hsu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Henry Sung-Ching Wong
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wan-Chen Huang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Institute of Medical Device and Imaging, National Taiwan University, Taipei, Taiwan
| | - Yi-Hung Yeh
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | | | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan. .,Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan. .,Integrative Research Center in Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Shie-Liang Hsieh
- Genomics Research Center, Academia Sinica, Taipei, Taiwan. .,Institute of Clinical Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan. .,Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan. .,Graduate of Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
5
|
Li H, Yang YG, Sun T. Nanoparticle-Based Drug Delivery Systems for Induction of Tolerance and Treatment of Autoimmune Diseases. Front Bioeng Biotechnol 2022; 10:889291. [PMID: 35464732 PMCID: PMC9019755 DOI: 10.3389/fbioe.2022.889291] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmune disease is a chronic inflammatory disease caused by disorders of immune regulation. Antigen-specific immunotherapy has the potential to inhibit the autoreactivity of inflammatory T cells and induce antigen-specific immune suppression without impairing normal immune function, offering an ideal strategy for autoimmune disease treatment. Tolerogenic dendritic cells (Tol DCs) with immunoregulatory functions play important roles in inducing immune tolerance. However, the effective generation of tolerogenic DCs in vivo remains a great challenge. The application of nanoparticle-based drug delivery systems in autoimmune disease treatment can increase the efficiency of inducing antigen-specific tolerance in vivo. In this review, we discuss multiple nanoparticles, with a focus on their potential in treatment of autoimmune diseases. We also discuss how the physical properties of nanoparticles influence their therapeutic efficacy.
Collapse
Affiliation(s)
- He Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- Department of Rehabilitation Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Institute of Immunology, The First Hospital, Jilin University, Changchun, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
| |
Collapse
|
6
|
Bodahl S, Cerps S, Uller L, Nilsson BO. LL-37 and Double-Stranded RNA Synergistically Upregulate Bronchial Epithelial TLR3 Involving Enhanced Import of Double-Stranded RNA and Downstream TLR3 Signaling. Biomedicines 2022; 10:biomedicines10020492. [PMID: 35203701 PMCID: PMC8962275 DOI: 10.3390/biomedicines10020492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/08/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
The human host defense peptide LL-37 influences double-stranded RNA signaling, but this process is not well understood. Here, we investigate synergistic actions of LL-37 and synthetic double-stranded RNA (poly I:C) on toll-like receptor 3 (TLR3) expression and signaling, and examine underlying mechanisms. In bronchial epithelial BEAS-2B cells, LL-37 potentiated poly I:C-induced TLR3 mRNA and protein expression demonstrated by qPCR and Western blot, respectively. Interestingly, these effects were associated with increased uptake of rhodamine-tagged poly I:C visualized by immunocytochemistry. The LL-37/poly I:C-induced upregulation of TLR3 mRNA expression was prevented by the endosomal acidification inhibitor chloroquine, indicating involvement of downstream TLR3 signaling. The glucocorticoid dexamethasone reduced LL-37/poly I:C-induced TLR3 expression on both mRNA and protein levels, and this effect was associated with increased IκBα protein expression, suggesting that dexamethasone acts via attenuation of NF-κB activity. We conclude that LL-37 potentiates poly I:C-induced upregulation of TLR3 through a mechanism that may involve enhanced import of poly I:C and that LL-37/poly I:C-induced TLR3 expression is associated with downstream TLR3 signaling and sensitive to inhibition of NF-κB activity.
Collapse
|
7
|
Harlan B, Park HG, Spektor R, Cummings B, Brenna JT, Soloway PD. Single-cell chromatin accessibility and lipid profiling reveals SCD1-dependent metabolic shift in adipocytes induced by bariatric surgery. PLoS One 2021; 16:e0261783. [PMID: 34972124 PMCID: PMC8719700 DOI: 10.1371/journal.pone.0261783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity promotes type 2 diabetes and cardiometabolic pathologies. Vertical sleeve gastrectomy (VSG) is used to treat obesity resulting in long-term weight loss and health improvements that precede weight loss; however, the mechanisms underlying the immediate benefits remain incompletely understood. Because adipose plays a crucial role in energy homeostasis and utilization, we hypothesized that VSG exerts its influences, in part, by modulating adipose functional states. We applied single-cell ATAC sequencing and lipid profiling to inguinal and epididymal adipose depots from mice that received sham surgery or VSG. We observed depot-specific cellular composition and chromatin accessibility patterns that were altered by VSG. Specifically, accessibility at Scd1, a fatty acid desaturase, was substantially reduced after VSG in mature adipocytes of inguinal but not epididymal depots. This was accompanied by reduced accumulation of SCD1-produced unsaturated fatty acids. Given these findings and reports that reductions in Scd1 attenuate obesity and insulin resistance our results suggest VSG exerts its beneficial effects through an inguinal depot-specific reduction of SCD1 activity.
Collapse
Affiliation(s)
- Blaine Harlan
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
| | - Roman Spektor
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Bethany Cummings
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| | - Paul D. Soloway
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
8
|
Roach TG, Lång HKM, Xiong W, Ryhänen SJ, Capelluto DGS. Protein Trafficking or Cell Signaling: A Dilemma for the Adaptor Protein TOM1. Front Cell Dev Biol 2021; 9:643769. [PMID: 33718385 PMCID: PMC7952518 DOI: 10.3389/fcell.2021.643769] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/09/2021] [Indexed: 12/29/2022] Open
Abstract
Lysosomal degradation of ubiquitinated transmembrane protein receptors (cargo) relies on the function of Endosomal Sorting Complex Required for Transport (ESCRT) protein complexes. The ESCRT machinery is comprised of five unique oligomeric complexes with distinct functions. Target of Myb1 (TOM1) is an ESCRT protein involved in the initial steps of endosomal cargo sorting. To exert its function, TOM1 associates with ubiquitin moieties on the cargo via its VHS and GAT domains. Several ESCRT proteins, including TOLLIP, Endofin, and Hrs, have been reported to form a complex with TOM1 at early endosomal membrane surfaces, which may potentiate the role of TOM1 in cargo sorting. More recently, it was found that TOM1 is involved in other physiological processes, including autophagy, immune responses, and neuroinflammation, which crosstalk with its endosomal cargo sorting function. Alteration of TOM1 function has emerged as a phosphoinositide-dependent survival mechanism for bacterial infections and cancer progression. Based on current knowledge of TOM1-dependent cellular processes, this review illustrates how TOM1 functions in coordination with an array of protein partners under physiological and pathological scenarios.
Collapse
Affiliation(s)
- Tiffany G. Roach
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| | - Heljä K. M. Lång
- Division of Hematology, Oncology, and Stem Cell Transplantation, Children’s Hospital, and Pediatric Research Center, The New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wen Xiong
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| | - Samppa J. Ryhänen
- Division of Hematology, Oncology, and Stem Cell Transplantation, Children’s Hospital, and Pediatric Research Center, The New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Daniel G. S. Capelluto
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
9
|
Dobson GP, Biros E, Letson HL, Morris JL. Living in a Hostile World: Inflammation, New Drug Development, and Coronavirus. Front Immunol 2021; 11:610131. [PMID: 33552070 PMCID: PMC7862725 DOI: 10.3389/fimmu.2020.610131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
We present a brief history of the immune response and show that Metchnikoff's theory of inflammation and phagocytotic defense was largely ignored in the 20th century. For decades, the immune response was believed to be triggered centrally, until Lafferty and Cunningham proposed the initiating signal came from the tissues. This shift opened the way for Janeway's pattern recognition receptor theory, and Matzinger's danger model. All models failed to appreciate that without inflammation, there can be no immune response. The situation changed in the 1990s when cytokine biology was rapidly advancing, and the immune system's role expanded from host defense, to the maintenance of host health. An inflammatory environment, produced by immune cells themselves, was now recognized as mandatory for their attack, removal and repair functions after an infection or injury. We explore the cellular programs of the immune response, and the role played by cytokines and other mediators to tailor the right response, at the right time. Normally, the immune response is robust, self-limiting and restorative. However, when the antigen load or trauma exceeds the body's internal tolerances, as witnessed in some COVID-19 patients, excessive inflammation can lead to increased sympathetic outflows, cardiac dysfunction, coagulopathy, endothelial and metabolic dysfunction, multiple organ failure and death. Currently, there are few drug therapies to reduce excessive inflammation and immune dysfunction. We have been developing an intravenous (IV) fluid therapy comprising adenosine, lidocaine and Mg2+ (ALM) that confers a survival advantage by preventing excessive inflammation initiated by sepsis, endotoxemia and sterile trauma. The multi-pronged protection appears to be unique and may provide a tool to examine the intersection points in the immune response to infection or injury, and possible ways to prevent secondary tissue damage, such as that reported in patients with COVID-19.
Collapse
Affiliation(s)
- Geoffrey P. Dobson
- Heart, Trauma and Sepsis Research Laboratory, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
| | | | | | | |
Collapse
|
10
|
Arnold NS, Noren Hooten N, Zhang Y, Lehrmann E, Wood W, Camejo Nunez W, Thorpe RJ, Evans MK, Dluzen DF. The association between poverty and gene expression within peripheral blood mononuclear cells in a diverse Baltimore City cohort. PLoS One 2020; 15:e0239654. [PMID: 32970748 PMCID: PMC7514036 DOI: 10.1371/journal.pone.0239654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/08/2020] [Indexed: 01/13/2023] Open
Abstract
Socioeconomic status (SES), living in poverty, and other social determinants of health contribute to health disparities in the United States. African American (AA) men living below poverty in Baltimore City have a higher incidence of mortality when compared to either white males or AA females living below poverty. Previous studies in our laboratory and elsewhere suggest that environmental conditions are associated with differential gene expression (DGE) patterns in peripheral blood mononuclear cells (PBMCs). DGE have also been associated with hypertension and cardiovascular disease (CVD) and correlate with race and sex. However, no studies have investigated how poverty status associates with DGE between male and female AAs and whites living in Baltimore City. We examined DGE in 52 AA and white participants of the Healthy Aging in Neighborhoods of Diversity across the Life Span (HANDLS) cohort, who were living above or below 125% of the 2004 federal poverty line at time of sample collection. We performed a microarray to assess DGE patterns in PBMCs from these participants. AA males and females living in poverty had the most genes differentially-expressed compared with above poverty controls. Gene ontology (GO) analysis identified unique and overlapping pathways related to the endosome, single-stranded RNA binding, long-chain fatty-acyl-CoA biosynthesis, toll-like receptor signaling, and others within AA males and females living in poverty and compared with their above poverty controls. We performed RT-qPCR to validate top differentially-expressed genes in AA males. We found that KLF6, DUSP2, RBM34, and CD19 are expressed at significantly lower levels in AA males in poverty and KCTD12 is higher compared to above poverty controls. This study serves as an additional link to better understand the gene expression response in peripheral blood mononuclear cells in those living in poverty.
Collapse
Affiliation(s)
- Nicole S. Arnold
- Department of Biology, Morgan State University, Baltimore, MD, United States of America
| | - Nicole Noren Hooten
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Elin Lehrmann
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - William Wood
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Wendy Camejo Nunez
- Department of Biology, Morgan State University, Baltimore, MD, United States of America
| | - Roland J. Thorpe
- Program for Research on Men’s Health, Hopkins Center for Health Disparities Solutions, Johns Hopkins University, Baltimore, MD, United States of America
| | - Michele K. Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States of America
| | - Douglas F. Dluzen
- Department of Biology, Morgan State University, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
11
|
Thorp EB, Boada C, Jarbath C, Luo X. Nanoparticle Platforms for Antigen-Specific Immune Tolerance. Front Immunol 2020; 11:945. [PMID: 32508829 PMCID: PMC7251028 DOI: 10.3389/fimmu.2020.00945] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Innovative approaches in nanoparticle design have facilitated the creation of new formulations of nanoparticles that are capable of selectively calibrating the immune response. These nanomaterials may be engineered to interact with specific cellular and molecular targets. Recent advancements in nanoparticle synthesis have enabled surface functionalization of particles that mimic the diversity of ligands on the cell surface. Platforms synthesized using these design principles, called "biomimetic" nanoparticles, have achieved increasingly sophisticated targeting specificity and cellular trafficking capabilities. This holds great promise for next generation therapies that seek to achieve immune tolerance. In this review, we discuss the importance of physical design parameters including size, shape, and biomimetic surface functionalization, on the biodistribution, safety and efficacy of biologic nanoparticles. We will also explore potential applications for immune tolerance for organ or stem cell transplantation.
Collapse
Affiliation(s)
- Edward B. Thorp
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Christian Boada
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Clarens Jarbath
- Departments of Pathology & Pediatrics at Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Xunrong Luo
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC, United States
- Duke Transplant Center, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
12
|
Zainol MIB, Kawasaki T, Monwan W, Murase M, Sueyoshi T, Kawai T. Innate immune responses through Toll-like receptor 3 require human-antigen-R-mediated Atp6v0d2 mRNA stabilization. Sci Rep 2019; 9:20406. [PMID: 31892731 PMCID: PMC6938500 DOI: 10.1038/s41598-019-56914-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/16/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 3 (TLR3) recognizes double-stranded RNA derived from virus and its synthetic analogue, polyinosinic–polycytidylic acid [poly(I:C)]. Upon poly(I:C) binding, TLR3 activates transcription factors to express inflammatory cytokines and type I interferon. TLR3 is located in the endosomes and its recognition of poly(I:C) and activation of downstream signaling is regulated by endosomal acidification. However, the mechanism of post-transcriptional regulation in TLR3-mediated innate responses remains unclear. Here, we focused on Human antigen R (HuR, also known as ELAVL1) that recognizes and binds to the 3′ untranslated regions (3′UTRs) of target mRNAs, thereby protecting them from mRNA degradation, and found that HuR-deficient murine macrophage cells showed significantly reduced Ifnb1 mRNA expression after poly(I:C) stimulation. HuR-deficient cells also showed a marked reduction in the expression of Atp6v0d2 mRNA, which encodes a subunit of vacuolar-type H+ ATPase (V-ATPase), and therefore reduced endosomal acidification. HuR associated with the 3′UTR of Atp6v0d2 mRNA and the stability of Atp6v0d2 mRNA was maintained by its association with HuR. Taken together, our results suggest that HuR stabilizes Atp6v0d2 mRNA, which is required for the TLR3-mediated innate immune responses.
Collapse
Affiliation(s)
- Mohd Izwan Bin Zainol
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan
| | - Takumi Kawasaki
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan.
| | - Warunthorn Monwan
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan
| | - Motoya Murase
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan
| | - Takuya Sueyoshi
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara, 630-0192, Japan.
| |
Collapse
|
13
|
Pais SV, Key CE, Borges V, Pereira IS, Gomes JP, Fisher DJ, Mota LJ. CteG is a Chlamydia trachomatis effector protein that associates with the Golgi complex of infected host cells. Sci Rep 2019; 9:6133. [PMID: 30992493 PMCID: PMC6468002 DOI: 10.1038/s41598-019-42647-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/22/2019] [Indexed: 12/17/2022] Open
Abstract
Chlamydia trachomatis is a bacterial pathogen causing ocular and genital infections in humans. C. trachomatis multiplies exclusively inside host cells within a characteristic vacuole, from where it manipulates host cells by injecting them with type III secretion effector proteins. Here, we identified CteG as the first C. trachomatiseffector associated with the Golgi. For this, C. trachomatis strains expressing candidate effectors fused to a double hemagglutinin (2HA) tag were constructed. Then, among these strains, immunofluorescence microscopy revealed that CteG-2HA was delivered into the cytoplasm of infected cells. Between 16–20 h post-infection, CteG-2HA mostly associated with the Golgi; however, CteG-2HA also appeared at the host cell plasma membrane, and at 30 or 40 h post-infection this was its predominant localization. This change in the main localization of CteG-2HA was independent of intact microfilaments or microtubules. Ectopic expression of different regions of CteG (656 amino acid residues) in uninfected cells revealed that its first 100 residues contain a Golgi targeting region. Although a C. trachomatis cteG mutant did not display a defect in intracellular multiplication, CteG induced a vacuolar protein sorting defect when expressed in Saccharomyces cerevisiae. This suggested that CteG might function by subverting host cell vesicular transport.
Collapse
Affiliation(s)
- Sara V Pais
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Charlotte E Key
- Department of Microbiology, Southern Illinois University, Carbondale, Illinois, USA
| | - Vítor Borges
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - Inês S Pereira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - João Paulo Gomes
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - Derek J Fisher
- Department of Microbiology, Southern Illinois University, Carbondale, Illinois, USA
| | - Luís Jaime Mota
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal.
| |
Collapse
|
14
|
Donahue ND, Acar H, Wilhelm S. Concepts of nanoparticle cellular uptake, intracellular trafficking, and kinetics in nanomedicine. Adv Drug Deliv Rev 2019; 143:68-96. [PMID: 31022434 DOI: 10.1016/j.addr.2019.04.008] [Citation(s) in RCA: 527] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticle-based therapeutics and diagnostics are commonly referred to as nanomedicine and may significantly impact the future of healthcare. However, the clinical translation of these technologies is challenging. One of these challenges is the efficient delivery of nanoparticles to specific cell populations and subcellular targets in the body to elicit desired biological and therapeutic responses. It is critical for researchers to understand the fundamental concepts of how nanoparticles interact with biological systems to predict and control in vivo nanoparticle transport for improved clinical benefit. In this overview article, we review and discuss cellular internalization pathways, summarize the field`s understanding of how nanoparticle physicochemical properties affect cellular interactions, and explore and discuss intracellular nanoparticle trafficking and kinetics. Our overview may provide a valuable resource for researchers and may inspire new studies to expand our current understanding of nanotechnology-biology interactions at cellular and subcellular levels with the goal to improve clinical translation of nanomedicines.
Collapse
Affiliation(s)
- Nathan D Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Handan Acar
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States; Stephenson Cancer Center, Oklahoma City, Oklahoma 73104, United States.
| |
Collapse
|
15
|
Martínez-García EA, Zavala-Cerna MG, Lujano-Benítez AV, Sánchez-Hernández PE, Martín-Márquez BT, Sandoval-García F, Vázquez-Del Mercado M. Potential Chronotherapeutic Optimization of Antimalarials in Systemic Lupus Erythematosus: Is Toll-Like Receptor 9 Expression Dependent on the Circadian Cycle in Humans? Front Immunol 2018; 9:1497. [PMID: 30034390 PMCID: PMC6043638 DOI: 10.3389/fimmu.2018.01497] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptor 9 (TLR9) belongs to the group of endosomal receptors of the innate immune system with the ability to recognize hypomethylated CpG sequences from DNA. There is scarce information about TLR9 expression and its association with the circadian cycle (CC). Different patterns of TLR9 expression are regulated by the CC in mice, with an elevated expression at Zeitgeber time 19 (1:00 a.m.); nevertheless, we still need to corroborate this in humans. In systemic lupus erythematosus (SLE), the inhibitory effect of chloroquine (CQ) on TLR9 is limited. TLR9 activation has been associated with the presence of some autoantibodies: anti-Sm/RNP, anti-histone, anti-Ro, anti-La, and anti-double-stranded DNA. Treatment with CQ for SLE has been proven to be useful, in part by interfering with HLA-antigen coupling and with TLR9 ligand recognition. Studies have shown that TLR9 inhibitors such as antimalarial drugs are able to mask TLR9-binding sites on nucleic acids. The data presented here provide the basic information that could be useful for other clinical researchers to design studies that will have an impact in achieving a chronotherapeutic effect by defining the ideal time for CQ administration in SLE patients, consequently reducing the pathological effects that follow the activation of TLR9.
Collapse
Affiliation(s)
- Erika Aurora Martínez-García
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- UDG-CA-703, Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Maria Guadalupe Zavala-Cerna
- Immunology Research Laboratory, Programa Internacional de Medicina, Universidad Autonoma de Guadalajara, Guadalajara, Mexico
| | - Andrea Verónica Lujano-Benítez
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Pedro Ernesto Sánchez-Hernández
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Laboratorio de Inmunología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Beatriz Teresita Martín-Márquez
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- UDG-CA-703, Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Flavio Sandoval-García
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Departamento de Clínicas Médicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- UDG CA-701, Inmunometabolismo en Enfermedades Emergentes (GIIEE), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Mónica Vázquez-Del Mercado
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- UDG-CA-703, Inmunología y Reumatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
- Hospital Civil de Guadalajara “Juan I. Menchaca”, Servicio de Reumatología, Programa Nacional de Posgrados de Calidad (PNPC), Consejo Nacional de Ciencia y Tecnología (CONACYT), Guadalajara, Mexico
| |
Collapse
|
16
|
Rider PJF, Musarrat F, Nabil R, Naidu S, Kousoulas KG. First Impressions-the Potential of Altering Initial Host-Virus Interactions for Rational Design of Herpesvirus Vaccine Vectors. CURRENT CLINICAL MICROBIOLOGY REPORTS 2018; 5:55-65. [PMID: 30560044 DOI: 10.1007/s40588-018-0082-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose The earliest host-virus interactions occur during virus attachment and entry into cells. These initial steps in the virus lifecycle influence the outcome of infection beyond delivery of the viral genome into the cell. Herpesviruses alter host signaling pathways and processes during attachment and entry to facilitate virus infection and modulate innate immune responses. We suggest in this review that understanding these early signaling events may inform the rational design of therapeutic and prevention strategies for herpesvirus infection, as well as the engineering of viral vectors for immunotherapy purposes. Recent Findings Recent reports demonstrate that modulation of Herpes Simplex Virus Type-1 (HSV-1) entry results in unexpected enhancement of antiviral immune responses. Summary A variety of evidence suggests that herpesviruses promote specific cellular signaling responses that facilitate viral replication after binding to cell surfaces, as well as during virus entry. Of particular interest is the ability of the virus to alter innate immune responses through these cellular signaling events. Uncovering the underlying immune evasion strategies may lead to the design of live-attenuated vaccines that can generate robust and protective anti-viral immune responses against herpesviruses. These adjuvant properties may be extended to a variety of heterologous antigens expressed by herpesviral vectors.
Collapse
Affiliation(s)
- Paul J F Rider
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA
| | - Farhana Musarrat
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA
| | - Rafiq Nabil
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA
| | - Shan Naidu
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA
| | - Konstantin G Kousoulas
- Division of Biotechnology and Molecular Medicine and Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge LA
| |
Collapse
|
17
|
Combes A, Camosseto V, N'Guessan P, Argüello RJ, Mussard J, Caux C, Bendriss-Vermare N, Pierre P, Gatti E. BAD-LAMP controls TLR9 trafficking and signalling in human plasmacytoid dendritic cells. Nat Commun 2017; 8:913. [PMID: 29030552 PMCID: PMC5640662 DOI: 10.1038/s41467-017-00695-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 07/20/2017] [Indexed: 12/28/2022] Open
Abstract
Toll-like receptors (TLR) are essential components of the innate immune system. Several accessory proteins, such as UNC93B1, are required for transport and activation of nucleic acid sensing Toll-like receptors in endosomes. Here, we show that BAD-LAMP (LAMP5) controls TLR9 trafficking to LAMP1+ late endosomes in human plasmacytoid dendritic cells (pDC), leading to NF-κB activation and TNF production upon DNA detection. An inducible VAMP3+/LAMP2+/LAMP1- endolysosome compartment exists in pDCs from which TLR9 activation triggers type I interferon expression. BAD-LAMP-silencing enhances TLR9 retention in this compartment and consequent downstream signalling events. Conversely, sustained BAD-LAMP expression in pDCs contributes to their lack of type I interferon production after exposure to a TGF-β-positive microenvironment or isolation from human breast tumours. Hence, BAD-LAMP limits interferon expression in pDCs indirectly, by promoting TLR9 sorting to late endosome compartments at steady state and in response to immunomodulatory cues.TLR9 is highly expressed by plasmacytoid dendritic cells and detects nucleic acids, but to discriminate between host and microbial nucleic acids TLR9 is sorted into different endosomal compartments. Here the authors show that BAD-LAMP limits type 1 interferon responses by sorting TLR9 to late endosomal compartments.
Collapse
Affiliation(s)
- Alexis Combes
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France
| | - Voahirana Camosseto
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France
- International associated laboratory (LIA) CNRS "Mistra", 13288, Marseille cedex 9, France
| | - Prudence N'Guessan
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France
| | - Julie Mussard
- Centre Léon Berard, 69373, LYON cedex 08, France
- Université de Lyon, 69373, LYON cedex 08, France
- INSERM U1052, 69373, LYON cedex 08, France
- CNRS UMR5286, 69373, LYON cedex 08, France
| | - Christophe Caux
- Centre Léon Berard, 69373, LYON cedex 08, France
- Université de Lyon, 69373, LYON cedex 08, France
- INSERM U1052, 69373, LYON cedex 08, France
- CNRS UMR5286, 69373, LYON cedex 08, France
| | - Nathalie Bendriss-Vermare
- Centre Léon Berard, 69373, LYON cedex 08, France
- Université de Lyon, 69373, LYON cedex 08, France
- INSERM U1052, 69373, LYON cedex 08, France
- CNRS UMR5286, 69373, LYON cedex 08, France
| | - Philippe Pierre
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France
- International associated laboratory (LIA) CNRS "Mistra", 13288, Marseille cedex 9, France
- Institute for Research in Biomedicine-iBiMED and Aveiro Health Sciences Program University of Aveiro, Aveiro, 3810-193, Portugal
| | - Evelina Gatti
- Aix Marseille Université, CNRS, INSERM, CIML, 13288, Marseille cedex 9, France.
- International associated laboratory (LIA) CNRS "Mistra", 13288, Marseille cedex 9, France.
- Institute for Research in Biomedicine-iBiMED and Aveiro Health Sciences Program University of Aveiro, Aveiro, 3810-193, Portugal.
| |
Collapse
|
18
|
HIV Fusion in Dendritic Cells Occurs Mainly at the Surface and Is Limited by Low CD4 Levels. J Virol 2017; 91:JVI.01248-17. [PMID: 28814521 DOI: 10.1128/jvi.01248-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/10/2017] [Indexed: 01/06/2023] Open
Abstract
HIV-1 poorly infects monocyte-derived dendritic cells (MDDCs). This is in large part due to SAMHD1, which restricts viral reverse transcription. Pseudotyping HIV-1 with vesicular stomatitis virus G protein (VSV-G) strongly enhances infection, suggesting that earlier steps of viral replication, including fusion, are also inefficient in MDDCs. The site of HIV-1 fusion remains controversial and may depend on the cell type, with reports indicating that it occurs at the plasma membrane or, conversely, in an endocytic compartment. Here, we examined the pathways of HIV-1 entry in MDDCs. Using a combination of temperature shift and fusion inhibitors, we show that HIV-1 fusion mainly occurs at the cell surface. We then asked whether surface levels or intracellular localization of CD4 modulates HIV-1 entry. Increasing CD4 levels strongly enhanced fusion and infection with various HIV-1 isolates, including reference and transmitted/founder strains, but not with BaL, which uses low CD4 levels for entry. Overexpressing coreceptors did not facilitate viral infection. To further study the localization of fusion events, we generated CD4 mutants carrying heterologous cytoplasmic tails (LAMP1 or Toll-like receptor 7 [TLR7]) to redirect the molecule to intracellular compartments. The intracellular CD4 mutants did not facilitate HIV-1 fusion and replication in MDDCs. Fusion of an HIV-2 isolate with MDDCs was also enhanced by increasing surface CD4 levels. Our results demonstrate that MDDCs are inefficiently infected by various HIV-1 and HIV-2 strains, in part because of low CD4 levels. In these cells, viral fusion occurs mainly at the surface, and probably not after internalization.IMPORTANCE Dendritic cells (DCs) are professional antigen-presenting cells inducing innate and adaptive immune responses. DCs express the HIV receptor CD4 and are potential target cells for HIV. There is debate about the sensitivity of DCs to productive HIV-1 and HIV-2 infection. The fusion step of the viral replication cycle is inefficient in DCs, and the underlying mechanisms are poorly characterized. We show that increasing the levels of CD4 at the plasma membrane allows more HIV fusion and productive infection in DCs. We further demonstrate that HIV fusion occurs mainly at the cell surface and not in an intracellular compartment. Our results help us understand why DCs are poorly sensitive to HIV infection.
Collapse
|
19
|
Regulation of sterile α- and armadillo motif (SARM) containing protein expression in Pam2CSK4- and Pam3CSK4-activated mouse macrophage cell line (RAW264.7) requires TLR9. Inflamm Res 2017; 66:1099-1105. [PMID: 28889202 DOI: 10.1007/s00011-017-1090-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/06/2017] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION We aimed to investigate the involvement of surface TLRs and endosomal TLRs in the regulation of SARM expression by TLR2 ligands (Pam2CSK4 and Pam3CSK4). MATERIALS AND METHODS Mouse macrophage cell line (RAW264.7) was treated with either Pam2CSK4 or Pam3CSK4 (TLR2 ligands) at a concentration of 100 ng/ml. At indicated time points, the treated cells were lysed. The gene and protein expression of SARM were determined by RT-PCR and immunoblotting, respectively. For silencing of TLR9 function, the cells were transfected with TLR9 siRNAs before stimulation by these two TLR2 ligands RESULTS: The SARM expression was upregulated at both transcriptional and translational levels in time-dependent manner during activation of Pam2CSK4 and Pam3CSK4 in mouse macrophages. Blocking of ligand internalization by cytochalasin D showed interference effect with SARM expression. Moreover, our results also demonstrated that endosomal acidification and TLR9 were required for SARM expression suggesting the essential role of endosomal compartment acidification and TLR9 in regulating SARM expression. CONCLUSION Our findings suggested the collaboration of TLR2-TLR9 at least in the regulation of SARM expression. However, the underlying mechanism that participated in these two TLRs cooperation is underinvestigated.
Collapse
|
20
|
Neefjes J, Jongsma MML, Berlin I. Stop or Go? Endosome Positioning in the Establishment of Compartment Architecture, Dynamics, and Function. Trends Cell Biol 2017; 27:580-594. [PMID: 28363667 DOI: 10.1016/j.tcb.2017.03.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 02/03/2023]
Abstract
The endosomal system constitutes a key negotiator between the environment of a cell and its internal affairs. Comprised of a complex membranous network, wherein each vesicle can in principle move autonomously throughout the cell, the endosomal system operates as a coherent unit to optimally face external challenges and maintain homeostasis. Our appreciation of how individual endosomes are controlled in time and space to best serve their collective purpose has evolved dramatically in recent years. In light of these efforts, the endoplasmic reticulum (ER) - with its expanse of membranes permeating the cytoplasmic space - has emerged as a potent spatiotemporal organizer of endosome biology. We review the latest advances in our understanding of the mechanisms underpinning endosomal transport and positioning, with emphasis on the contributions from the ER, and offer a perspective on how the interplay between these aspects shapes the architecture and dynamics of the endosomal system and drives its myriad cellular functions.
Collapse
Affiliation(s)
- Jacques Neefjes
- Department of Chemical Immunology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Marlieke M L Jongsma
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center (AMC)/Universiteit van Amsterdam (UvA), Plesmanlaan 125, 1066 CX Amsterdam, The Netherlands
| | - Ilana Berlin
- Department of Chemical Immunology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| |
Collapse
|
21
|
Bercusson A, de Boer L, Armstrong-James D. Endosomal sensing of fungi: current understanding and emerging concepts. Med Mycol 2017; 55:10-15. [PMID: 27596144 DOI: 10.1093/mmy/myw072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 05/01/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022] Open
Abstract
Endosomal sensing represents a key strategy by which mammalian cells detect parasitization by invading pathogens. This is critical for the control of fungal pathogens, which are for the most part phagocytosed by effector cells of the innate immune system. Despite rapid overall progress in our understanding of endosomal responses in recent times, relatively little is known about how the endosomal sensing system detects fungi and the ensuing immunological consequences. Considering that many fungal pathogens must overcome and evade endosomal killing in order to survive in the host, understanding this key area of the early innate response is crucial for our understanding of fungal infection. In this review we present a summary of our current knowledge of endosomal sensing within the context of fungal pathogens, with a focus on the myeloid compartment.
Collapse
Affiliation(s)
- Amelia Bercusson
- Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, London UK SW7 6NP
| | - Leon de Boer
- Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, London UK SW7 6NP
| | - Darius Armstrong-James
- Fungal Pathogens Laboratory, National Heart and Lung Institute, Imperial College London, London UK SW7 6NP
| |
Collapse
|
22
|
Quantitative Proteomic Analysis of Escherichia coli Heat-Labile Toxin B Subunit (LTB) with Enterovirus 71 (EV71) Subunit VP1. Int J Mol Sci 2016; 17:ijms17091419. [PMID: 27618897 PMCID: PMC5037698 DOI: 10.3390/ijms17091419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
The nontoxic heat-labile toxin (LT) B subunit (LTB) was used as mucosal adjuvant experimentally. However, the mechanism of LTB adjuvant was still unclear. The LTB and enterovirus 71 (EV71) VP1 subunit (EVP1) were constructed in pET32 and expressed in E. coli BL21, respectively. The immunogenicity of purified EVP1 and the adjuvanticity of LTB were evaluated via intranasal immunization EVP1 plus LTB in Balb/c mice. In order to elucidate the proteome change triggered by the adjuvant of LTB, the proteomic profiles of LTB, EVP1, and LTB plus EVP1 were quantitatively analyzed by iTRAQ-LC-MS/MS (isobaric tags for relative and absolute quantitation; liquid chromatography-tandem mass spectrometry) in murine macrophage RAW264.7. The proteomic data were analyzed by bioinformatics and validated by western blot analysis. The predicted protein interactions were confirmed using LTB pull-down and the LTB processing pathway was validated by confocal microscopy. The results showed that LTB significantly boosted EVP1 specific systematic and mucosal antibodies. A total of 3666 differential proteins were identified in the three groups. Pathway enrichment of proteomic data predicted that LTB upregulated the specific and dominant MAPK (mitogen-activated protein kinase) signaling pathway and the protein processing in endoplasmic reticulum (PPER) pathway, whereas LTB or EVP1 did not significantly upregulate these two signaling pathways. Confocal microscopy and LTB pull-down assays confirmed that the LTB adjuvant was endocytosed and processed through endocytosis (ENS)-lysosomal-endoplasmic reticulum (ER) system.
Collapse
|
23
|
Hill PB, Imai A. The immunopathogenesis of staphylococcal skin infections - A review. Comp Immunol Microbiol Infect Dis 2016; 49:8-28. [PMID: 27865269 DOI: 10.1016/j.cimid.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 07/22/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022]
Abstract
Staphylococcus aureus and S. pseudintermedius are the major causes of bacterial skin disease in humans and dogs. These organisms can exist as commensals on the skin, but they can also cause severe or even devastating infections. The immune system has evolved mechanisms to deal with pathogenic microorganisms and has strategies to combat bacteria of this type. What emerges is a delicate "peace" between the opposing sides, but this balance can be disrupted leading to a full blown "war". In the ferocious battle that ensues, both sides attempt to get the upper hand, using strategies that are comparable to those used by modern day armies. In this review article, the complex interactions between the immune system and the organisms are described using such military analogies. The process is described in a sequential manner, starting with the invasion itself, and progressing to the eventual battlezone in which there are heavy casualties on both sides. By the end, the appearance of a simple pustule on the skin surface will take on a whole new meaning.
Collapse
Affiliation(s)
- P B Hill
- Companion Animal Health Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, Roseworthy SA 5371, Australia.
| | - A Imai
- Dermatology resident, Synergy Animal General Hospital, 815 Kishigami Kawaguchi, Saitama, 333-0823, Japan
| |
Collapse
|
24
|
Hamzić E, Buitenhuis B, Hérault F, Hawken R, Abrahamsen MS, Servin B, Elsen JM, Pinard-van der Laan MH, Bed'Hom B. Genome-wide association study and biological pathway analysis of the Eimeria maxima response in broilers. Genet Sel Evol 2015; 47:91. [PMID: 26607727 PMCID: PMC4659166 DOI: 10.1186/s12711-015-0170-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 11/05/2015] [Indexed: 02/22/2023] Open
Abstract
Background Coccidiosis is the most common and costly disease in the poultry industry and is caused by protozoans of the Eimeria genus. The current control of coccidiosis, based on the use of anticoccidial drugs and vaccination, faces serious obstacles such as drug resistance and the high costs for the development of efficient vaccines, respectively. Therefore, the current control programs must be expanded with complementary approaches such as the use of genetics to improve the host response to Eimeria infections. Recently, we have performed a large-scale challenge study on Cobb500 broilers using E. maxima for which we investigated variability among animals in response to the challenge. As a follow-up to this challenge study, we performed a genome-wide association study (GWAS) to identify genomic regions underlying variability of the measured traits in the response to Eimeria maxima in broilers. Furthermore, we conducted a post-GWAS functional analysis to increase our biological understanding of the underlying response to Eimeria maxima challenge. Results In total, we identified 22 single nucleotide polymorphisms (SNPs) with q value <0.1 distributed across five chromosomes. The highly significant SNPs were associated with body weight gain (three SNPs on GGA5, one SNP on GGA1 and one SNP on GGA3), plasma coloration measured as optical density at wavelengths in the range 465–510 nm (10 SNPs and all on GGA10) and the percentage of β2-globulin in blood plasma (15 SNPs on GGA1 and one SNP on GGA2). Biological pathways related to metabolic processes, cell proliferation, and primary innate immune processes were among the most frequent significantly enriched biological pathways. Furthermore, the network-based analysis produced two networks of high confidence, with one centered on large tumor suppressor kinase 1 (LATS1) and 2 (LATS2) and the second involving the myosin heavy chain 6 (MYH6). Conclusions We identified several strong candidate genes and genomic regions associated with traits measured in response to Eimeria maxima in broilers. Furthermore, the post-GWAS functional analysis indicates that biological pathways and networks involved in tissue proliferation and repair along with the primary innate immune response may play the most important role during the early stage of Eimeria maxima infection in broilers. Electronic supplementary material The online version of this article (doi:10.1186/s12711-015-0170-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edin Hamzić
- UMR1313 Animal Genetics and Integrative Biology Unit, AgroParisTech, 16 rue Claude Bernard, 75005, Paris, France. .,UMR1313 Animal Genetics and Integrative Biology Unit, INRA, Domaine de Vilvert, 78350, Jouy-en-Josas, France. .,Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Blichers Allé 20, P.O. Box 50, 8830, Tjele, Denmark.
| | - Bart Buitenhuis
- Department of Molecular Biology and Genetics, Center for Quantitative Genetics and Genomics, Aarhus University, Blichers Allé 20, P.O. Box 50, 8830, Tjele, Denmark.
| | - Frédéric Hérault
- UMR1348 Physiology, Environment and Genetics for the Animal and Livestock Systems Unit, INRA, Domaine de la Prise, 35590, Saint Gilles, France.
| | | | | | - Bertrand Servin
- UMR1388 Genetics, Physiology and Breeding Systems, INRA, 24 chemin de Borde-Rouge, 31326, Castanet-Tolosan, France.
| | - Jean-Michel Elsen
- UMR1388 Genetics, Physiology and Breeding Systems, INRA, 24 chemin de Borde-Rouge, 31326, Castanet-Tolosan, France.
| | - Marie-Hélène Pinard-van der Laan
- UMR1313 Animal Genetics and Integrative Biology Unit, AgroParisTech, 16 rue Claude Bernard, 75005, Paris, France. .,UMR1313 Animal Genetics and Integrative Biology Unit, INRA, Domaine de Vilvert, 78350, Jouy-en-Josas, France.
| | - Bertrand Bed'Hom
- UMR1313 Animal Genetics and Integrative Biology Unit, AgroParisTech, 16 rue Claude Bernard, 75005, Paris, France. .,UMR1313 Animal Genetics and Integrative Biology Unit, INRA, Domaine de Vilvert, 78350, Jouy-en-Josas, France.
| |
Collapse
|
25
|
Posch B, Irsara C, Gamper FS, Herrmann M, Bindreither D, Fuchs D, Reider N, Redl B, Heufler C. Allergenic Can f 1 and its human homologue Lcn-1 direct dendritic cells to induce divergent immune responses. J Cell Mol Med 2015. [PMID: 26218644 PMCID: PMC4594679 DOI: 10.1111/jcmm.12616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Why and when the immune system skews to Th2 mediated allergic immune responses is still poorly characterized. With two homologous lipocalins, the major respiratory dog allergen Can f 1 and the human endogenous, non-allergenic Lipocalin-1, we investigated their impact on human monocyte-derived dendritic cells (DC). The two lipocalins had differential effects on DC according to their allergenic potential. Compared to Lipocalin-1, Can f 1 persistently induced lower levels of the Th1 skewing maturation marker expression, tryptophan breakdown and interleukin (IL)-12 production in DC. As a consequence, T cells stimulated by DC treated with Can f 1 produced more of the Th2 signature cytokine IL-13 and lower levels of the Th1 signature cytokine interferon-γ than T cells stimulated by Lipocalin-1 treated DC. These data were partially verified by a second pair of homologous lipocalins, the cat allergen Fel d 4 and its putative human homologue major urinary protein. Our data indicate that the crosstalk of DC with lipocalins alone has the potential to direct the type of immune response to these particular antigens. A global gene expression analysis further supported these results and indicated significant differences in intracellular trafficking, sorting and antigen presentation pathways when comparing Can f 1 and Lipocalin-1 stimulated DC. With this study we contribute to a better understanding of the induction phase of a Th2 immune response.
Collapse
Affiliation(s)
- Beate Posch
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Irsara
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Fabian S Gamper
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Martin Herrmann
- Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Bindreither
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Dietmar Fuchs
- Division of Medical Biochemistry, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Norbert Reider
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard Redl
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Christine Heufler
- Department of Dermatology, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
26
|
Zhou MT, Qin Y, Li M, Chen C, Chen X, Shu HB, Guo L. Quantitative Proteomics Reveals the Roles of Peroxisome-associated Proteins in Antiviral Innate Immune Responses. Mol Cell Proteomics 2015; 14:2535-49. [PMID: 26124285 DOI: 10.1074/mcp.m115.048413] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Indexed: 11/06/2022] Open
Abstract
Compared with whole-cell proteomic analysis, subcellular proteomic analysis is advantageous not only for the increased coverage of low abundance proteins but also for generating organelle-specific data containing information regarding dynamic protein movement. In the present study, peroxisome-enriched fractions from Sendai virus (SeV)-infected or uninfected HepG2 cells were obtained and subjected to quantitative proteomics analysis. We identified 311 proteins that were significantly changed by SeV infection. Among these altered proteins, 25 are immune response-related proteins. Further bioinformatic analysis indicated that SeV infection inhibits cell cycle-related proteins and membrane attack complex-related proteins, all of which are beneficial for the survival and replication of SeV within host cells. Using Luciferase reporter assays on several innate immune-related reporters, we performed functional analysis on 11 candidate proteins. We identified LGALS3BP and CALU as potential negative regulators of the virus-induced activation of the type I interferons.
Collapse
Affiliation(s)
- Mao-Tian Zhou
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Yue Qin
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Mi Li
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University
| | - Chen Chen
- From the ‡State Key Laboratory of Virology, College of Life Sciences
| | - Xi Chen
- ¶Wuhan Institute of Biotechnology, Wuhan, China
| | - Hong-Bing Shu
- From the ‡State Key Laboratory of Virology, College of Life Sciences; §Medical Research Institute, Wuhan University;
| | - Lin Guo
- From the ‡State Key Laboratory of Virology, College of Life Sciences;
| |
Collapse
|
27
|
Dunston CR, Herbert R, Griffiths HR. Improving T cell-induced response to subunit vaccines: opportunities for a proteomic systems approach. ACTA ACUST UNITED AC 2015; 67:290-9. [PMID: 25708693 DOI: 10.1111/jphp.12383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 11/23/2014] [Indexed: 11/30/2022]
Abstract
UNLABELLED Prophylactic vaccines are an effective strategy to prevent development of many infectious diseases. With new and re-emerging infections posing increasing risks to food stocks and the health of the population in general, there is a need to improve the rationale of vaccine development. One key challenge lies in development of an effective T cell-induced response to subunit vaccines at specific sites and in different populations. OBJECTIVES In this review, we consider how a proteomic systems-based approach can be used to identify putative novel vaccine targets, may be adopted to characterise subunit vaccines and adjuvants fully. KEY FINDINGS Despite the extensive potential for proteomics to aid our understanding of subunit vaccine nature, little work has been reported on identifying MHC 1-binding peptides for subunit vaccines generating T cell responses in the literature to date. SUMMARY In combination with predictive and structural biology approaches to mapping antigen presentation, proteomics offers a powerful and as yet un-tapped addition to the armoury of vaccine discovery to predict T-cell subset responses and improve vaccine design strategies.
Collapse
Affiliation(s)
- Christopher R Dunston
- Life & Health Sciences, Aston University, Birmingham, West Midlands, UK; Mologic, Bedford Technology Park, Thurleigh, Bedfordshire, MK44 2YP
| | | | | |
Collapse
|