1
|
Wang C, Ge F, Ge F, Xu Z, Jiang J. Harnessing stem cell therapeutics in LPS-induced animal models: mechanisms, efficacies, and future directions. Stem Cell Res Ther 2025; 16:176. [PMID: 40221751 PMCID: PMC11993993 DOI: 10.1186/s13287-025-04290-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
The severity and threat posed by inflammation are well documented, and lipopolysaccharides (LPS), as important inducers of inflammatory responses, are widely recognized for studying host immunity and the resulting tissue and organ damage. The LPS-induced disease model, triggers a remarkable release of inflammatory factors, immune and coagulation dysfunction, and damage to vital organs such as the brain, lungs, heart, liver, and kidneys. Recently, the role of mesenchymal stem cells (MSCs) in various clinical diseases has garnered significant attention due to their immunomodulatory, anti-inflammatory, tissue healing, anti-apoptotic, and antibacterial properties. Despite the common use of LPS models to induce disease models and simulate acute inflammation, the integration of stem cell therapy within these models remains underexplored. This article integrates the LPS induced animal model and reviews the current evidence regarding the therapeutic mechanisms of stem cells in LPS-induced disease models across various human body systems. Furthermore, this review predicts and hypothesizes the feasibility and potential of using stem cells in disease models that have not yet been extensively studied, based on existing animal inflammation models.
Collapse
Affiliation(s)
- Chengran Wang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Fanghong Ge
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Fangjun Ge
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong Province, People's Republic of China
| | - Zhonghang Xu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
2
|
Sardar P, Beresford-Jones BS, Xia W, Shabana O, Suyama S, Ramos RJF, Soderholm AT, Tourlomousis P, Kuo P, Evans AC, Imianowski CJ, Conti AG, Wesolowski AJ, Baker NM, McCord EAL, Okkenhaug K, Whiteside SK, Roychoudhuri R, Bryant CE, Cross JR, Pedicord VA. Gut microbiota-derived hexa-acylated lipopolysaccharides enhance cancer immunotherapy responses. Nat Microbiol 2025; 10:795-807. [PMID: 39929976 PMCID: PMC11879847 DOI: 10.1038/s41564-025-01930-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/13/2025] [Indexed: 03/06/2025]
Abstract
The gut microbiome modulates immunotherapy treatment responses, and this may explain why immune checkpoint inhibitors, such as anti-PD-1, are only effective in some patients. Previous studies correlated lipopolysaccharide (LPS)-producing gut microbes with poorer prognosis; however, LPS from diverse bacterial species can range from immunostimulatory to inhibitory. Here, by functionally analysing faecal metagenomes from 112 patients with melanoma, we found that a subset of LPS-producing bacteria encoding immunostimulatory hexa-acylated LPS was enriched in microbiomes of clinical responders. In an implanted tumour mouse model of anti-PD-1 treatment, microbiota-derived hexa-acylated LPS was required for effective anti-tumour immune responses, and LPS-binding antibiotics and a small-molecule TLR4 antagonist abolished anti-PD-1 efficacy. Conversely, oral administration of hexa-acylated LPS to mice significantly augmented anti-PD-1-mediated anti-tumour immunity. Penta-acylated LPS did not improve anti-PD-1 efficacy in vivo and inhibited hexa-acylated LPS-induced immune activation in vitro. Microbiome hexa-acylated LPS therefore represents an accessible predictor and potential enhancer of immunotherapy responses.
Collapse
Affiliation(s)
- Puspendu Sardar
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Benjamin S Beresford-Jones
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Wangmingyu Xia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Omar Shabana
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Satoshi Suyama
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ruben J F Ramos
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amelia T Soderholm
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Panagiotis Tourlomousis
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Paula Kuo
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Alexander C Evans
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Charlotte J Imianowski
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Alberto G Conti
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Alexander J Wesolowski
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Natalie M Baker
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Emily A L McCord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Klaus Okkenhaug
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Sarah K Whiteside
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Rahul Roychoudhuri
- Department of Pathology, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Clare E Bryant
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, School of Biological Sciences, University of Cambridge, Cambridge, UK
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Virginia A Pedicord
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK.
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Roach KA, Anderson SE, Waggy C, Aldinger J, Stefaniak AB, Roberts JR. Assessment of dermal sensitization by nickel salts in a novel humanized TLR-4 mouse model. J Immunotoxicol 2024; 21:2414979. [PMID: 39632339 DOI: 10.1080/1547691x.2024.2414979] [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: 01/28/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 12/07/2024] Open
Abstract
The fundamental goal of this study was to determine the potential utility of a novel humanized Toll-like receptor-4 (hTLR-4) mouse model for future in vivo studies of nickel allergy. First, mice of both sexes and hTLR-4 expression profiles were incorporated into a Local Lymph Node Assay (LLNA) to assess skin sensitization. Next, a set of hTLR-4 hTLR-4-positive mice (female and male groups) was similarly exposed to vehicle control (VC) or 10% NiSO4 on Days 1, 2, and 3. Mice were euthanized on Day 10, lymph node (LN) cellularity was assessed, LN and spleen cells were phenotyped, and serum was collected to quantify circulating cytokine and IgE levels. In the LLNA, hTLR-4-positive mice of both sexes exhibited enhanced responsivity to nickel. NiSO4 (10%) had a stimulation index (SI) of 3.7 (females) and 3.8 (males) in hTLR-4-positive animals, and an SI of 0.5 (females) and 0.8 (males) in hTLR-4 hTLR-4-negative mice. In the 10d study, hTLR-4-positive mice exposed to 10% NiSO4 exhibited increased LN cellularity (6.0× increase in females, 3.2× in males) and significantly higher concentrations of circulating IgE (4.1× increase in females, 3.4× in males). Significant increases in serum interferon (IFN)-γ, interleukin (IL)-4, and IL-5 levels were seen in female mice, while altered concentrations of IL-4 and IL-10 were detected in male mice. The results of this study ultimately demonstrate that murine expression of hTLR-4 confers enhanced susceptibility to dermal sensitization by nickel, and consequently, the hTLR-4 mouse model represents a viable approach for future studies of nickel allergy in vivo.
Collapse
Affiliation(s)
- K A Roach
- Allergy and Clinical Immunology Branch (ACIB), National Institute of Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - S E Anderson
- Allergy and Clinical Immunology Branch (ACIB), National Institute of Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - C Waggy
- Office of the Director, NIOSH, Morgantown, WV, USA
| | - J Aldinger
- Allergy and Clinical Immunology Branch (ACIB), National Institute of Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| | - A B Stefaniak
- Respiratory Health Division, NIOSH, Morgantown, WV, USA
| | - J R Roberts
- Allergy and Clinical Immunology Branch (ACIB), National Institute of Occupational Safety and Health (NIOSH), Morgantown, WV, USA
| |
Collapse
|
4
|
Herrero-Fernández B, Ortega-Zapero M, Gómez-Bris R, Sáez A, Iborra S, Zorita V, Quintas A, Vázquez E, Dopazo A, Sánchez-Madrid F, Arribas SM, González-Granado JM. Role of lamin A/C on dendritic cell function in antiviral immunity. Cell Mol Life Sci 2024; 81:400. [PMID: 39264480 PMCID: PMC11393282 DOI: 10.1007/s00018-024-05423-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/29/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024]
Abstract
Dendritic cells (DCs) play a crucial role in orchestrating immune responses, particularly in promoting IFNγ-producing-CD8 cytotoxic T lymphocytes (CTLs) and IFNγ-producing-CD4 T helper 1 (Th1) cells, which are essential for defending against viral infections. Additionally, the nuclear envelope protein lamin A/C has been implicated in T cell immunity. Nevertheless, the intricate interplay between innate and adaptive immunity in response to viral infections, particularly the role of lamin A/C in DC functions within this context, remains poorly understood. In this study, we demonstrate that mice lacking lamin A/C in myeloid LysM promoter-expressing cells exhibit a reduced capacity to induce Th1 and CD8 CTL responses, leading to impaired clearance of acute primary Vaccinia virus (VACV) infection. Remarkably, in vitro-generated granulocyte macrophage colony-stimulating factor bone marrow-derived DCs (GM-CSF BMDCs) show high levels of lamin A/C. Lamin A/C absence on GM-CSF BMDCs does not affect the expression of costimulatory molecules on the cell membrane but it reduces the cellular ability to form immunological synapses with naïve CD4 T cells. Lamin A/C deletion induces alterations in NFκB nuclear localization, thereby influencing NF-κB-dependent transcription. Furthermore, lamin A/C ablation modifies the gene accessibility of BMDCs, predisposing these cells to mount a less effective antiviral response upon TLR stimulation. This study highlights the critical role of DCs in interacting with CD4 T cells during antiviral responses and proposes some mechanisms through which lamin A/C may modulate DC function via gene accessibility and transcriptional regulation.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernández
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Marina Ortega-Zapero
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Raquel Gómez-Bris
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain
| | - Angela Sáez
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), Pozuelo de Alarcón, 28223, Spain
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain
- Fundacion Inmunotek, Alcalá de Henares, 28805, Spain
| | - Virginia Zorita
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Enrique Vázquez
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Sánchez-Madrid
- Centro Nacional de Investigaciones, Cardiovasculares (CNIC), Madrid, 28029, Spain
- Immunology Unit, Medicine Department, Hospital Universitario La Princesa, Universidad Autónoma de Madrid, Instituto Investigacion Sanitaria-Princesa IIS-IP, Madrid, Spain, Madrid, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Silvia Magdalena Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, 28029, Spain.
| | - Jose Maria González-Granado
- LamImSys Lab, Instituto de Investigacion Sanitaria Hospital 12 de Octubre (imas12), Madrid, 28041, Spain.
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, 28040, Spain.
- Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
5
|
Moghaddam MM, Behzadi E, Sedighian H, Goleij Z, Kachuei R, Heiat M, Fooladi AAI. Regulation of immune responses to infection through interaction between stem cell-derived exosomes and toll-like receptors mediated by microRNA cargoes. Front Cell Infect Microbiol 2024; 14:1384420. [PMID: 38756232 PMCID: PMC11096519 DOI: 10.3389/fcimb.2024.1384420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Infectious diseases are among the factors that account for a significant proportion of disease-related deaths worldwide. The primary treatment approach to combat microbial infections is the use of antibiotics. However, the widespread use of these drugs over the past two decades has led to the emergence of resistant microbial species, making the control of microbial infections a serious challenge. One of the most important solutions in the field of combating infectious diseases is the regulation of the host's defense system. Toll-like receptors (TLRs) play a crucial role in the first primary defense against pathogens by identifying harmful endogenous molecules released from dying cells and damaged tissues as well as invading microbial agents. Therefore, they play an important role in communicating and regulating innate and adaptive immunity. Of course, excessive activation of TLRs can lead to disruption of immune homeostasis and increase the risk of inflammatory reactions. Targeting TLR signaling pathways has emerged as a new therapeutic approach for infectious diseases based on host-directed therapy (HDT). In recent years, stem cell-derived exosomes have received significant attention as factors regulating the immune system. The regulation effects of exosomes on the immune system are based on the HDT strategy, which is due to their cargoes. In general, the mechanism of action of stem cell-derived exosomes in HDT is by regulating and modulating immunity, promoting tissue regeneration, and reducing host toxicity. One of their most important cargoes is microRNAs, which have been shown to play a significant role in regulating immunity through TLRs. This review investigates the therapeutic properties of stem cell-derived exosomes in combating infections through the interaction between exosomal microRNAs and Toll-like receptors.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- The Academy of Medical Sciences of I.R. Iran, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zoleikha Goleij
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Dorando HK, Mutic EC, Li JY, Perrin EP, Wurtz MK, Quinn CC, Payton JE. LPS and type I and II interferons have opposing effects on epigenetic regulation of LAIR1 expression in mouse and human macrophages. J Leukoc Biol 2024; 115:547-564. [PMID: 38011310 DOI: 10.1093/jleuko/qiad148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Inhibitory immune receptors are important for maintaining immune homeostasis. We identified epigenetic alterations in 2 members of this group, LAIR1 and LAIR2, in lymphoma patients with inflammatory tissue damage and susceptibility to infection. We predicted that the expression of LAIR genes is controlled by immune mediators acting on transcriptional regulatory elements. Using flow cytometry, quantitative reverse-transcription polymerase chain reaction, and RNA sequencing, we measured LAIR1 and LAIR2 in human and murine immune cell subsets at baseline and posttreatment with immune mediators, including type I and II interferons, tumor necrosis factor α, and lipopolysaccharide (LPS). We identified candidate regulatory elements using epigenome profiling and measured their regulatory activity using luciferase reporters. LAIR1 expression substantially increases during monocyte differentiation to macrophages in both species. In contrast, murine and human macrophages exhibited opposite changes in LAIR1 in response to immune stimuli: human LAIR1 increased with LPS while mouse LAIR1 increased with interferon γ. LAIR genes had distinct patterns of enhancer activity with variable responses to immune stimuli. To identify relevant transcription factors (TFs), we developed integrative bioinformatic techniques applied to TF chromatin immunoprecipitation sequencing, RNA sequencing, and luciferase activity, revealing distinct sets of TFs for each LAIR gene. Most strikingly, LAIR1 TFs include nuclear factor kappa B factors RELA and RELB, while Lair1 and LAIR2 instead include STAT3 and/or STAT5. Regulation by nuclear factor kappa B factors may therefore explain the LPS-induced increase in LAIR1 expression, in contrast to Lair1 decrease. Our findings reveal new insights into transcriptional mechanisms that control distinct expression patterns of LAIR genes in response to inflammatory stimuli in human and murine myeloid and lymphoid cells.
Collapse
Affiliation(s)
- Hannah K Dorando
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Evan C Mutic
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Joanna Y Li
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Ezri P Perrin
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Mellisa K Wurtz
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Chaz C Quinn
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine in St.Louis, 660 S. Euclid Avenue, Box 8118, St. Louis, MO 63110, United States
| |
Collapse
|
7
|
Abstract
The immune system of multicellular organisms protects them from harmful microbes. To establish an infection in the face of host immune responses, pathogens must evolve specific strategies to target immune defense mechanisms. One such defense is the formation of intracellular protein complexes, termed inflammasomes, that are triggered by the detection of microbial components and the disruption of homeostatic processes that occur during bacterial infection. Formation of active inflammasomes initiates programmed cell death pathways via activation of inflammatory caspases and cleavage of target proteins. Inflammasome-activated cell death pathways such as pyroptosis lead to proinflammatory responses that protect the host. Bacterial infection has the capacity to influence inflammasomes in two distinct ways: activation and perturbation. In this review, we discuss how bacterial activities influence inflammasomes, and we discuss the consequences of inflammasome activation or evasion for both the host and pathogen.
Collapse
Affiliation(s)
- Beatrice I Herrmann
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James P Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
- Current affiliation: Oncology Discovery, Abbvie, Inc., Chicago, Illinois, USA;
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; ,
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Zsurka G, Appel MLT, Nastaly M, Hallmann K, Hansen N, Nass D, Baumgartner T, Surges R, Hartmann G, Bartok E, Kunz WS. Loss of the Immunomodulatory Transcription Factor BATF2 in Humans Is Associated with a Neurological Phenotype. Cells 2023; 12:227. [PMID: 36672163 PMCID: PMC9856319 DOI: 10.3390/cells12020227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Epilepsy and mental retardation are known to be associated with pathogenic mutations in a broad range of genes that are expressed in the brain and have a role in neurodevelopment. Here, we report on a family with three affected individuals whose clinical symptoms closely resemble a neurodevelopmental disorder. Whole-exome sequencing identified a homozygous stop-gain mutation, p.Gln19*, in the BATF2 gene in the patients. The BATF2 transcription factor is predominantly expressed in macrophages and monocytes and has been reported to modulate AP-1 transcription factor-mediated pro-inflammatory responses. Transcriptome analysis showed altered base-level expression of interferon-stimulated genes in the patients' blood, typical for type I interferonopathies. Peripheral blood mononuclear cells from all three patients demonstrated elevated responses to innate immune stimuli, which could be reproduced in CRISPR-Cas9-generated BATF2-/- human monocytic cell lines. BATF2 is, therefore, a novel disease-associated gene candidate for severe epilepsy and mental retardation related to dysregulation of immune responses, which underscores the relevance of neuroinflammation for epilepsy.
Collapse
Affiliation(s)
- Gábor Zsurka
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Maximilian L. T. Appel
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Institute of Experimental Haematology and Transfusion Medicine, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Maximilian Nastaly
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Kerstin Hallmann
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Niels Hansen
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Daniel Nass
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Tobias Baumgartner
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Rainer Surges
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Eva Bartok
- Institute of Experimental Haematology and Transfusion Medicine, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Wolfram S. Kunz
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
9
|
Harberts EM, Grubaugh D, Akuma DC, Shin S, Ernst RK, Brodsky IE. Position-Specific Secondary Acylation Determines Detection of Lipid A by Murine TLR4 and Caspase-11. Infect Immun 2022; 90:e0020122. [PMID: 35862717 PMCID: PMC9387250 DOI: 10.1128/iai.00201-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 01/16/2023] Open
Abstract
Immune sensing of the Gram-negative bacterial membrane glycolipid lipopolysaccharide (LPS) is both a critical component of host defense against bacterial infection and a contributor to the hyperinflammatory response, potentially leading to sepsis and death. Innate immune activation by LPS is due to the lipid A moiety, an acylated di-glucosamine molecule that can activate inflammatory responses via the extracellular sensor Toll-like receptor 4 (TLR4)/myeloid differentiation 2 (MD2) or the cytosolic sensor caspase-11 (Casp11). The number and length of acyl chains present on bacterial lipid A structures vary across bacterial species and strains, which affects the magnitude of TLR4 and Casp11 activation. TLR4 and Casp11 are thought to respond similarly to various lipid A structures, as tetra-acylated lipid A structures do not activate either sensor, whereas hexa-acylated structures activate both sensors. However, the precise features of lipid A that determine the differential activation of each receptor remain poorly defined, as direct analysis of extracellular and cytosolic responses to the same sources and preparations of LPS/lipid A structures have been limited. To address this question, we used rationally engineered lipid A isolated from a series of bacterial acyl-transferase mutants that produce novel, structurally defined molecules. Intriguingly, we found that the location of specific secondary acyl chains on lipid A resulted in differential recognition by TLR4 or Casp11, providing new insight into the structural features of lipid A required to activate either TLR4 or Casp11. Our findings indicate that TLR4 and Casp11 sense nonoverlapping areas of lipid A chemical space, thereby constraining the ability of Gram-negative pathogens to evade innate immunity.
Collapse
Affiliation(s)
- Erin M. Harberts
- Department of Microbial Pathogenesis, University of Maryland, School of Dentistry, Baltimore, Maryland, USA
| | - Daniel Grubaugh
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Daniel C. Akuma
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland, School of Dentistry, Baltimore, Maryland, USA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Sheppe AEF, Santelices J, Czyz DM, Edelmann MJ. Yersinia pseudotuberculosis YopJ Limits Macrophage Response by Downregulating COX-2-Mediated Biosynthesis of PGE2 in a MAPK/ERK-Dependent Manner. Microbiol Spectr 2021; 9:e0049621. [PMID: 34319170 PMCID: PMC8552654 DOI: 10.1128/spectrum.00496-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
Prostaglandin E2 (PGE2) is an essential immunomodulatory lipid released by cells in response to infection with many bacteria, yet its function in macrophage-mediated bacterial clearance is poorly understood. Yersinia overall inhibits the inflammatory circuit, but its effect on PGE2 production is unknown. We hypothesized that one of the Yersinia effector proteins is responsible for the inhibition of PGE2 biosynthesis. We identified that yopB-deficient Y. enterocolitica and Y. pseudotuberculosis deficient in the secretion of virulence proteins via a type 3 secretion system (T3SS) failed to inhibit PGE2 biosynthesis in macrophages. Consistently, COX-2-mediated PGE2 biosynthesis is upregulated in cells treated with heat-killed or T3SS-deficient Y. pseudotuberculosis but diminished in the presence of a MAPK/ERK inhibitor. Mutants expressing catalytically inactive YopJ induce similar levels of PGE2 as heat-killed or ΔyopB Y. pseudotuberculosis, reversed by YopJ complementation. Shotgun proteomics discovered host pathways regulated in a YopJ-mediated manner, including pathways regulating PGE2 synthesis and oxidative phosphorylation. Consequently, this study identified that YopJ-mediated inhibition of MAPK signal transduction serves as a mechanism targeting PGE2, an alternative means of inflammasome inhibition by Yersinia. Finally, we showed that EP4 signaling supports macrophage function in clearing intracellular bacteria. In summary, our unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription, thereby enhancing the intracellular survival of yersiniae. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic molecule to improve the outcomes of specific bacterial infections. Since other pathogens encode YopJ homologs, this mechanism is expected to be present in other infections. IMPORTANCE PGE2 is a critical immunomodulatory lipid, but its role in bacterial infection and pathogen clearance is poorly understood. We previously demonstrated that PGE2 leads to macrophage polarization toward the M1 phenotype and stimulates inflammasome activation in infected macrophages. Finally, we also discovered that PGE2 improved the clearance of Y. enterocolitica. The fact that Y. enterocolitica hampers PGE2 secretion in a type 3 secretion system (T3SS)-dependent manner and because PGE2 appears to assist macrophage in the clearance of this bacterium indicates that targeting of the eicosanoid pathway by Yersinia might be an adaption used to counteract host defenses. Our study identified a mechanism used by Yersinia that obstructs PGE2 biosynthesis in human macrophages. We showed that Y. pseudotuberculosis interferes with PGE2 biosynthesis by using one of its T3SS effectors, YopJ. Specifically, YopJ targets the host COX-2 enzyme responsible for PGE2 biosynthesis, which happens in a MAPK/ER-dependent manner. Moreover, in a shotgun proteomics study, we also discovered other pathways that catalytically active YopJ targets in the infected macrophages. YopJ was revealed to play a role in limiting host LPS responses, including repression of EGR1 and JUN proteins, which control transcriptional activation of proinflammatory cytokine production such as interleukin-1β. Since YopJ has homologs in other bacterial species, there are likely other pathogens that target and inhibit PGE2 biosynthesis. In summary, our study's unique contribution was to determine a bacterial virulence factor that targets COX-2 transcription. Future studies should investigate whether PGE2 or its stable synthetic derivatives could serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Austin E. F. Sheppe
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida, USA
| | - John Santelices
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida, USA
| | - Daniel M. Czyz
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida, USA
| | - Mariola J. Edelmann
- Department of Microbiology and Cell Science, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
11
|
Rosenzweig JA, Hendrix EK, Chopra AK. Plague vaccines: new developments in an ongoing search. Appl Microbiol Biotechnol 2021; 105:4931-4941. [PMID: 34142207 PMCID: PMC8211537 DOI: 10.1007/s00253-021-11389-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/25/2021] [Accepted: 06/02/2021] [Indexed: 11/17/2022]
Abstract
As the reality of pandemic threats challenges humanity, exemplified during the ongoing SARS-CoV-2 infections, the development of vaccines targeting these etiological agents of disease has become increasingly critical. Of paramount concern are novel and reemerging pathogens that could trigger such events, including the plague bacterium Yersinia pestis. Y. pestis is responsible for more human deaths than any other known pathogen and exists globally in endemic regions of the world, including the four corners region and Northern California in the USA. Recent cases have been scattered throughout the world, including China and the USA, with serious outbreaks in Madagascar during 2008, 2013-2014, and, most recently, 2017-2018. This review will focus on recent advances in plague vaccine development, a seemingly necessary endeavor, as there is no Food and Drug Administration-licensed vaccine available for human distribution in western nations, and that antibiotic-resistant strains are recovered clinically or intentionally developed. Progress and recent development involving subunit, live-attenuated, and nucleic acid-based plague vaccine candidates will be discussed in this review. KEY POINTS: • Plague vaccine development remains elusive yet critical. • DNA, animal, and live-attenuated vaccine candidates gain traction.
Collapse
Affiliation(s)
- Jason A Rosenzweig
- Department of Biology, Texas Southern University, Houston, TX, 77004, USA.
| | - Emily K Hendrix
- Departmnet of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Ashok K Chopra
- Departmnet of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
12
|
Anti-Leptospira immunoglobulin profiling in mice reveals strain specific IgG and persistent IgM responses associated with virulence and renal colonization. PLoS Negl Trop Dis 2021; 15:e0008970. [PMID: 33705392 PMCID: PMC8007020 DOI: 10.1371/journal.pntd.0008970] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/29/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Leptospira interrogans is a pathogenic spirochete responsible for leptospirosis, a neglected, zoonotic reemerging disease. Humans are sensitive hosts and may develop severe disease. Some animal species, such as rats and mice can become asymptomatic renal carriers. More than 350 leptospiral serovars have been identified, classified on the basis of the antibody response directed against the lipopolysaccharide (LPS). Similarly to whole inactivated bacteria used as human vaccines, this response is believed to confer only short-term, serogroup-specific protection. The immune response of hosts against leptospires has not been thoroughly studied, which complicates the testing of vaccine candidates. In this work, we studied the immunoglobulin (Ig) profiles in mice infected with L. interrogans over time to determine whether this humoral response confers long-term protection after homologous challenge six months post-infection. Groups of mice were injected intraperitoneally with 2×107 leptospires of one of three pathogenic serovars (Manilae, Copenhageni or Icterohaemorrhagiae), attenuated mutants or heat-killed bacteria. Leptospira-specific immunoglobulin (IgA, IgM, IgG and 4 subclasses) produced in the first weeks up to 6 months post-infection were measured by ELISA. Strikingly, we found sustained high levels of IgM in mice infected with the pathogenic Manilae and Copenhageni strains, both colonizing the kidney. In contrast, the Icterohaemorrhagiae strain did not lead to kidney colonization, even at high dose, and triggered a classical IgM response that peaked at day 8 post-infection and disappeared. The virulent Manilae and Copenhageni serovars elicited high levels and similar profiles of IgG subclasses in contrast to Icterohaemorrhagiae strains that stimulated weaker antibody responses. Inactivated heat-killed Manilae strains elicited very low responses. However, all mice pre-injected with leptospires challenged with high doses of homologous bacteria did not develop acute leptospirosis, and all antibody responses were boosted after challenge. Furthermore, we showed that 2 months post-challenge, mice pre-infected with the attenuated M895 Manilae LPS mutant or heat-killed bacterin were completely protected against renal colonization. In conclusion, we observed a sustained IgM response potentially associated with chronic leptospiral renal infection. We also demonstrated in mice different profiles of protective and cross-reactive antibodies after L. interrogans infection, depending on the serovar and virulence of strains. Leptospira interrogans is a pathogenic spirochete responsible for leptospirosis, a neglected zoonotic reemerging disease. The immune response of hosts against these bacteria has not been thoroughly studied. Here, we studied over 6 months the antibody profiles in mice infected with L. interrogans and determined whether this humoral response confers long-term protection after homologous challenge six months after primary infection. Groups of mice were infected intraperitoneally with 2×107 bacteria of one of three different pathogenic serovars (Manilae, Copenhageni and Icterohaemorrhagiae) and some corresponding attenuated avirulent mutants. We measured by ELISA each type of Leptospira-specific immunoglobulin (Ig) (IgA, IgM, IgG and 4 subclasses) produced in the first weeks up to 6 months post-infection and studied their cross-reactivities among serovars. We showed different profiles of antibody response after L. interrogans challenge in mice, depending on the serovar and virulence of strains. However, all infected mice, including the ones harboring low antibody levels, like mice vaccinated with an inactivated, heat-killed strain, were protected against leptospirosis after challenge. Notably, we also showed an unusual sustained IgM response associated with chronic leptospiral colonization. Altogether, this long-term immune protection is different from what is known in humans and warrants further investigation.
Collapse
|
13
|
Nair N, Guedes MS, Hajjar AM, Werts C, Gomes-Solecki M. Role of TLR4 in Persistent Leptospira interrogans Infection: A Comparative In Vivo Study in Mice. Front Immunol 2021; 11:572999. [PMID: 33519799 PMCID: PMC7843520 DOI: 10.3389/fimmu.2020.572999] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Toll-Like Receptor (TLR) 4, the LPS receptor, plays a central role in the control of leptospirosis and absence of TLR4 results in lethal infection in mice. Because human TLR4 does not sense the atypical leptospiral-LPS, we hypothesized that TLR4/MD-2 humanized transgenic mice (huTLR4) may be more susceptible to leptospirosis than wild-type mice, and thus may constitute a model of acute human leptospirosis. We infected huTLR4 mice, which express human TLR4 but not murine TLR4, with a high dose of L. interrogans serovar Copenhageni FioCruz (Leptospira) in comparison to C57BL/6J wild-type (WT) and, as a control, a congenic strain in which the tlr4 coding sequences are deleted (muTLR4Lps-del). We show that the huTLR4 gene is fully functional in the murine background. We found that dissemination of Leptospira in blood, shedding in urine, colonization of the kidney and overall kinetics of leptospirosis progression is equivalent between WT and huTLR4 C57BL/6J mice. Furthermore, inflammation of the kidney appeared to be subdued in huTLR4 compared to WT mice in that we observed less infiltrates of mononuclear lymphocytes, less innate immune markers and no relevant differences in fibrosis markers. Thus, huTLR4 mice showed less inflammation and kidney pathology, and are not more susceptible to leptospirosis than WT mice. This study is significant as it indicates that one intact TLR4 gene, be it mouse or human, is necessary to control acute leptospirosis.
Collapse
Affiliation(s)
- Nisha Nair
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mariana S. Guedes
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Catherine Werts
- Institut Pasteur, Biology and Genetics of the Bacterial Cell Wall Unit, Paris, France,CNRS, UMR 2001 Microbiologie intégrative et Moléculaire, Paris, France
| | - Maria Gomes-Solecki
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States,*Correspondence: Maria Gomes-Solecki,
| |
Collapse
|
14
|
Bollmann P, Werner F, Jaron M, Bruns TA, Wache H, Runte J, Boknik P, Kirchhefer U, Müller FU, Buchwalow IB, Rothemund S, Neumann J, Gergs U. Initial Characterization of Stressed Transgenic Mice With Cardiomyocyte-Specific Overexpression of Protein Phosphatase 2C. Front Pharmacol 2021; 11:591773. [PMID: 33597873 PMCID: PMC7883593 DOI: 10.3389/fphar.2020.591773] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/07/2020] [Indexed: 12/17/2022] Open
Abstract
As part of our ongoing studies on the potential pathophysiological role of serine/threonine phosphatases (PP) in the mammalian heart, we have generated mice with cardiac-specific overexpression of PP2Cβ (PP2C-TG) and compared them with littermate wild type mice (WT) serving as a control. Cardiac fibrosis was noted histologically in PP2C-TG. Collagen 1a, interleukin-6 and the natriuretic peptides ANP and BNP were augmented in PP2C-TG vs. WT (p < 0.05). Left atrial preparations from PP2C-TG were less resistant to hypoxia than atria from WT. PP2C-TG maintained cardiac function after the injection of lipopolysaccharide (LPS, a model of sepsis) and chronic isoproterenol treatment (a model of heart failure) better than WT. Crossbreeding of PP2C-TG mice with PP2A-TG mice (a genetic model of heart failure) resulted in double transgenic (DT) mice that exhibited a pronounced increase of heart weight in contrast to the mild hypertrophy noted in the mono-transgenic mice. The ejection fraction was reduced in PP2C-TG and in PP2A-TG mice compared with WT, but the reduction was the highest in DT compared with WT. PP2A enzyme activity was enhanced in PP2A-TG and DT mice compared with WT and PP2C-TG mice. In summary, cardiac overexpression of PP2Cβ and co-overexpression of both the catalytic subunit of PP2A and PP2Cβ were detrimental to cardiac function. PP2Cβ overexpression made cardiac preparations less resistant to hypoxia than WT, leading to fibrosis, but PP2Cβ overexpression led to better adaptation to some stressors, such as LPS or chronic β-adrenergic stimulation. Hence, the effect of PP2Cβ is context sensitive.
Collapse
Affiliation(s)
- Paula Bollmann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Franziska Werner
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Marko Jaron
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Tom A Bruns
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Hartmut Wache
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Jochen Runte
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Peter Boknik
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - Uwe Kirchhefer
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | - Frank U Müller
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Westfälische Wilhelms-Universität, Münster, Germany
| | | | | | - Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| | - Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle, Germany
| |
Collapse
|
15
|
Zamyatina A, Heine H. Lipopolysaccharide Recognition in the Crossroads of TLR4 and Caspase-4/11 Mediated Inflammatory Pathways. Front Immunol 2020; 11:585146. [PMID: 33329561 PMCID: PMC7732686 DOI: 10.3389/fimmu.2020.585146] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
The innate immune response to lipopolysaccharide is essential for host defense against Gram-negative bacteria. In response to bacterial infection, the TLR4/MD-2 complex that is expressed on the surface of macrophages, monocytes, dendritic, and epithelial cells senses picomolar concentrations of endotoxic LPS and triggers the production of various pro-inflammatory mediators. In addition, LPS from extracellular bacteria which is either endocytosed or transfected into the cytosol of host cells or cytosolic LPS produced by intracellular bacteria is recognized by cytosolic proteases caspase-4/11 and hosts guanylate binding proteins that are involved in the assembly and activation of the NLRP3 inflammasome. All these events result in the initiation of pro-inflammatory signaling cascades directed at bacterial eradication. However, TLR4-mediated signaling and caspase-4/11-induced pyroptosis are largely involved in the pathogenesis of chronic and acute inflammation. Both extra- and intracellular LPS receptors-TLR4/MD-2 complex and caspase-4/11, respectively-are able to directly bind the lipid A motif of LPS. Whereas the structural basis of lipid A recognition by the TLR4 complex is profoundly studied and well understood, the atomic mechanism of LPS/lipid A interaction with caspase-4/11 is largely unknown. Here we describe the LPS-induced TLR4 and caspase-4/11 mediated signaling pathways and their cross-talk and scrutinize specific structural features of the lipid A motif of diverse LPS variants that have been reported to activate caspase-4/11 or to induce caspase-4/11 mediated activation of NLRP3 inflammasome (either upon transfection of LPS in vitro or upon infection of cell cultures with intracellular bacteria or by LPS as a component of the outer membrane vesicles). Generally, inflammatory caspases show rather similar structural requirements as the TLR4/MD-2 complex, so that a "basic" hexaacylated bisphosphorylated lipid A architecture is sufficient for activation. However, caspase-4/11 can sense and respond to much broader variety of lipid A variants compared to the very "narrow" specificity of TLR4/MD-2 complex as far as the number and the length of lipid chains attached at the diglucosamine backbone of lipid A is concerned. Besides, modification of the lipid A phosphate groups with positively charged appendages such as phosphoethanolamine or aminoarabinose could be essential for the interaction of lipid A/LPS with inflammatory caspases and related proteins.
Collapse
Affiliation(s)
- Alla Zamyatina
- Institute of Organic Chemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Holger Heine
- Research Group Innate Immunity, Research Center Borstel—Leibniz Lung Center, Airway Research Center North (ARCN), German Center for Lung Disease (DZL), Borstel, Germany
| |
Collapse
|
16
|
Ciesielska A, Matyjek M, Kwiatkowska K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell Mol Life Sci 2020; 78:1233-1261. [PMID: 33057840 PMCID: PMC7904555 DOI: 10.1007/s00018-020-03656-y] [Citation(s) in RCA: 779] [Impact Index Per Article: 155.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
Toll-like receptor (TLR) 4 belongs to the TLR family of receptors inducing pro-inflammatory responses to invading pathogens. TLR4 is activated by lipopolysaccharide (LPS, endotoxin) of Gram-negative bacteria and sequentially triggers two signaling cascades: the first one involving TIRAP and MyD88 adaptor proteins is induced in the plasma membrane, whereas the second engaging adaptor proteins TRAM and TRIF begins in early endosomes after endocytosis of the receptor. The LPS-induced internalization of TLR4 and hence also the activation of the TRIF-dependent pathway is governed by a GPI-anchored protein, CD14. The endocytosis of TLR4 terminates the MyD88-dependent signaling, while the following endosome maturation and lysosomal degradation of TLR4 determine the duration and magnitude of the TRIF-dependent one. Alternatively, TLR4 may return to the plasma membrane, which process is still poorly understood. Therefore, the course of the LPS-induced pro-inflammatory responses depends strictly on the rates of TLR4 endocytosis and trafficking through the endo-lysosomal compartment. Notably, prolonged activation of TLR4 is linked with several hereditary human diseases, neurodegeneration and also with autoimmune diseases and cancer. Recent studies have provided ample data on the role of diverse proteins regulating the functions of early, late, and recycling endosomes in the TLR4-induced inflammation caused by LPS or phagocytosis of E. coli. In this review, we focus on the mechanisms of the internalization and intracellular trafficking of TLR4 and CD14, and also of LPS, in immune cells and discuss how dysregulation of the endo-lysosomal compartment contributes to the development of diverse human diseases.
Collapse
Affiliation(s)
- Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland.
| | - Marta Matyjek
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| |
Collapse
|
17
|
Early evolutionary loss of the lipid A modifying enzyme PagP resulting in innate immune evasion in Yersinia pestis. Proc Natl Acad Sci U S A 2020; 117:22984-22991. [PMID: 32868431 DOI: 10.1073/pnas.1917504117] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Immune evasion through membrane remodeling is a hallmark of Yersinia pestis pathogenesis. Yersinia remodels its membrane during its life cycle as it alternates between mammalian hosts (37 °C) and ambient (21 °C to 26 °C) temperatures of the arthropod transmission vector or external environment. This shift in growth temperature induces changes in number and length of acyl groups on the lipid A portion of lipopolysaccharide (LPS) for the enteric pathogens Yersinia pseudotuberculosis (Ypt) and Yersinia enterocolitica (Ye), as well as the causative agent of plague, Yersinia pestis (Yp). Addition of a C16 fatty acid (palmitate) to lipid A by the outer membrane acyltransferase enzyme PagP occurs in immunostimulatory Ypt and Ye strains, but not in immune-evasive Yp Analysis of Yp pagP gene sequences identified a single-nucleotide polymorphism that results in a premature stop in translation, yielding a truncated, nonfunctional enzyme. Upon repair of this polymorphism to the sequence present in Ypt and Ye, lipid A isolated from a Yp pagP+ strain synthesized two structures with the C16 fatty acids located in acyloxyacyl linkage at the 2' and 3' positions of the diglucosamine backbone. Structural modifications were confirmed by mass spectrometry and gas chromatography. With the genotypic restoration of PagP enzymatic activity in Yp, a significant increase in lipid A endotoxicity mediated through the MyD88 and TRIF/TRAM arms of the TLR4-signaling pathway was observed. Discovery and repair of an evolutionarily lost lipid A modifying enzyme provides evidence of lipid A as a crucial determinant in Yp infectivity, pathogenesis, and host innate immune evasion.
Collapse
|
18
|
Bruning EE, Coller JK, Wardill HR, Bowen JM. Site-specific contribution of Toll-like receptor 4 to intestinal homeostasis and inflammatory disease. J Cell Physiol 2020; 236:877-888. [PMID: 32730645 DOI: 10.1002/jcp.29976] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Toll-like receptor 4 (TLR4) is a highly conserved protein of innate immunity, responsible for the regulation and maintenance of homeostasis, as well as immune recognition of external and internal ligands. TLR4 is expressed on a variety of cell types throughout the gastrointestinal tract, including on epithelial and immune cell populations. In a healthy state, epithelial cell expression of TLR4 greatly assists in homeostasis by shaping the host microbiome, promoting immunoglobulin A production, and regulating follicle-associated epithelium permeability. In contrast, immune cell expression of TLR4 in healthy states is primarily centred on the maturation of dendritic cells in response to stimuli, as well as adequately priming the adaptive immune system to fight infection and promote immune memory. Hence, in a healthy state, there is a clear distinction in the site-specific roles of TLR4 expression. Similarly, recent research has indicated the importance of site-specific TLR4 expression in inflammation and disease, particularly the impact of epithelial-specific TLR4 on disease progression. However, the majority of evidence still remains ambiguous for cell-specific observations, with many studies failing to provide the distinction of epithelial versus immune cell expression of TLR4, preventing specific mechanistic insight and greatly impacting the translation of results. The following review provides a critical overview of the current understanding of site-specific TLR4 activity and its contribution to intestinal/immune homeostasis and inflammatory diseases.
Collapse
Affiliation(s)
- Elise E Bruning
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Janet K Coller
- Discipline of Pharmacology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| | - Hannah R Wardill
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia.,Department of Paediatric Oncology/Haematology, The University of Groningen (University Medical Centre Groningen), Groningen, The Netherlands
| | - Joanne M Bowen
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
19
|
Robison A, Snyder DT, Christensen K, Kimmel E, Hajjar AM, Jutila MA, Hedges JF. Expression of human TLR4/myeloid differentiation factor 2 directs an early innate immune response associated with modest increases in bacterial burden during Coxiella burnetii infection. Innate Immun 2019; 25:401-411. [PMID: 31180798 PMCID: PMC6900644 DOI: 10.1177/1753425919855420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/24/2019] [Accepted: 05/13/2019] [Indexed: 01/25/2023] Open
Abstract
Human TLR4 (hTLR4) and mouse TLR4 molecules respond differently to hypo-acylated LPS. The LPS of Coxiella burnetii is hypo-acylated and heavily glycosylated and causes a minimal response by human cells. Thus, we hypothesized that mice expressing hTLR4 molecules would be more susceptible to C. burnetii infection. Our results show that transgenic mice expressing hTLR4 and the human myeloid differentiation factor 2 (MD-2) adaptor protein (hTLR4/MD-2) respond similarly to wild type mice with respect to overall disease course. However, differences in bacterial burdens in tissues were noted, and lung pathology was increased in hTLR4/MD2 compared to wild type mice. Surprisingly, bone marrow chimera experiments indicated that hTLR4/MD-2 expression on non-hematopoietic cells, rather than the target cells for C. burnetii infection, accounted for increased bacterial burden. Early during infection, cytokines involved in myeloid cell recruitment were detected in the plasma of hTLR4/MD2 mice but not wild type mice. This restricted cytokine response was accompanied by neutrophil recruitment to the lung in hTLR4/MD2 mice. These data suggest that hTLR4/MD-2 alters early responses during C. burnetii infection. These early responses are precursors to later increased bacterial burdens and exacerbated pathology in the lung. Our data suggest an unexpected role for hTLR4/MD-2 in non-hematopoietic cells during C. burnetii infection.
Collapse
Affiliation(s)
- Amanda Robison
- Department of Microbiology and Immunology, Montana State University, USA
| | - Deann T Snyder
- Department of Microbiology and Immunology, Montana State University, USA
| | - Kelly Christensen
- Department of Microbiology and Immunology, Montana State University, USA
| | - Emily Kimmel
- Department of Microbiology and Immunology, Montana State University, USA
| | - Adeline M Hajjar
- Department of Comparative Medicine, School of Medicine, University of Washington, USA
| | - Mark A Jutila
- Department of Microbiology and Immunology, Montana State University, USA
| | - Jodi F Hedges
- Department of Microbiology and Immunology, Montana State University, USA
| |
Collapse
|
20
|
Gergs U, Jahn T, Werner F, Köhler C, Köpp F, Großmann C, Neumann J. Overexpression of protein phosphatase 5 in the mouse heart: Reduced contractility but increased stress tolerance - Two sides of the same coin? PLoS One 2019; 14:e0221289. [PMID: 31425567 PMCID: PMC6699691 DOI: 10.1371/journal.pone.0221289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/02/2019] [Indexed: 11/18/2022] Open
Abstract
The pathophysiological mechanisms of sepsis-induced cardiac dysfunction are largely unknown. The Toll-like receptor 4 (TLR4) is expressed in cardiac myocytes and is involved in bacterial endotoxin-mediated inflammatory disorders. TLR4 signaling leads to activation of the nuclear factor kappa B followed by increased expression of cytokines. Several protein phosphatases including PP2Cβ, PP2A or PP1 are known to act as regulators of this signaling pathway. Here, we examined the role of PP5 for the inflammatory response to the bacterial endotoxin lipopolysaccharide in the heart using a transgenic mouse model with cardiac myocyte directed overexpression of PP5. In these transgenic mice, basal cardiac contractility was reduced, in vivo as well as in vitro, but LPS-induced cardiac dysfunction was less pronounced compared to wild type mice. Quantitative RT-PCR suggested an attenuated NF-κB signaling in the heart and cardiac expression of heat shock protein 25 (HSP25) was increased in PP5 transgenic mice. From our data we assume that PP5 increases stress tolerance of cardiac myocytes by downregulation of NF-κB signaling and upregulation of HSP25 expression.
Collapse
Affiliation(s)
- Ulrich Gergs
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
- * E-mail:
| | - Tina Jahn
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Franziska Werner
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Carolin Köhler
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Friedrich Köpp
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Großmann
- Julius-Bernstein-Institut für Physiologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| | - Joachim Neumann
- Institut für Pharmakologie und Toxikologie, Medizinische Fakultät, Martin-Luther-Universität Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
21
|
Modeling Pneumonic Plague in Human Precision-Cut Lung Slices Highlights a Role for the Plasminogen Activator Protease in Facilitating Type 3 Secretion. Infect Immun 2019; 87:IAI.00175-19. [PMID: 31085709 PMCID: PMC6652753 DOI: 10.1128/iai.00175-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/06/2019] [Indexed: 12/30/2022] Open
Abstract
Pneumonic plague is the deadliest form of disease caused by Yersinia pestis Key to the progression of infection is the activity of the plasminogen activator protease Pla. Deletion of Pla results in a decreased Y. pestis bacterial burden in the lung and failure to progress into the lethal proinflammatory phase of disease. While a number of putative functions have been attributed to Pla, its precise role in the pathogenesis of pneumonic plague is yet to be defined. Here, we show that Pla facilitates type 3 secretion into primary alveolar macrophages but not into the commonly used THP-1 cell line. We also establish human precision-cut lung slices as a platform for modeling early host/pathogen interactions during pneumonic plague and solidify the role of Pla in promoting optimal type 3 secretion using primary human tissue with relevant host cell heterogeneity. These results position Pla as a key player in the early host/pathogen interactions that define pneumonic plague and showcase the utility of human precision-cut lung slices as a platform to evaluate pulmonary infection by bacterial pathogens.
Collapse
|
22
|
Chaudhary A, Kamischke C, Leite M, Altura MA, Kinman L, Kulasekara H, Blanc MP, Wang G, Terhorst C, Miller SI. β-Barrel outer membrane proteins suppress mTORC2 activation and induce autophagic responses. Sci Signal 2018; 11:11/558/eaat7493. [PMID: 30482849 DOI: 10.1126/scisignal.aat7493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The outer membranes of Gram-negative bacteria and mitochondria contain proteins with a distinct β-barrel tertiary structure that could function as a molecular pattern recognized by the innate immune system. Here, we report that purified outer membrane proteins (OMPs) from different bacterial and mitochondrial sources triggered the induction of autophagy-related endosomal acidification, LC3B lipidation, and p62 degradation. Furthermore, OMPs reduced the phosphorylation and therefore activation of the multiprotein complex mTORC2 and its substrate Akt in macrophages and epithelial cells. The cell surface receptor SlamF8 and the DNA-protein kinase subunit XRCC6 were required for these OMP-specific responses in macrophages and epithelial cells, respectively. The addition of OMPs to mouse bone marrow-derived macrophages infected with Salmonella Typhimurium facilitated bacterial clearance. These data identify a specific cellular response mediated by bacterial and mitochondrial OMPs that can alter inflammatory responses and influence the killing of pathogens.
Collapse
Affiliation(s)
- Anu Chaudhary
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA.,Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Cassandra Kamischke
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Mara Leite
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Melissa A Altura
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Loren Kinman
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Hemantha Kulasekara
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Marie-Pierre Blanc
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Guoxing Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Samuel I Miller
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA. .,Department of Immunology, University of Washington, Seattle, WA 98195, USA.,Department of Medicine, University of Washington, Seattle, WA 98195, USA.,Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
23
|
Kabanov DS, Rykov VA, Prokhorenko SV, Murashev AN, Prokhorenko IR. In vivo Proinflammatory Cytokine Production by CD-1 Mice in Response to Equipotential Doses of Rhodobacter capsulatus PG and Salmonella enterica Lipopolysaccharides. BIOCHEMISTRY (MOSCOW) 2018; 83:846-854. [PMID: 30200869 DOI: 10.1134/s0006297918070088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The capacities of relatively nontoxic lipopolysaccharide (LPS) from Rhodobacter capsulatus PG and highly potent LPS from Salmonella enterica serovar Typhimurium to evoke proinflammatory cytokine production have been compared in vivo. Intravenous administration of S. enterica LPS at a relatively low dose (1 mg/kg body weight) led to upregulation of TNF-α, IL-6, and IFN-γ production by non-sensitized CD-1 mice. LPS from R. capsulatus PG used at a four-times higher dose than that from S. enterica elicited production of almost the same amount of systemic TNF-α; therefore, the doses of 4 mg/kg LPS from R. capsulatus PG and 1 mg/kg LPS from S. enterica were considered to be approximately equipotential doses with respect to the LPS-dependent TNF-α production by CD-1 mice. Rhodobacter capsulatus PG LPS was a weaker inducer of the production of TNF-α, IL-6, and IFN-γ, as compared to the equipotential dose of S. enterica LPS. Administration of R. capsulatus PG LPS before S. enterica LPS decreased production of IFN-γ, but not of TNF-α and IL-6, induced by S. enterica LPS. Rhodobacter capsulatus PG LPS also suppressed IFN-γ production induced by S. enterica LPS when R. capsulatus PG LPS had been injected as little as 10 min after S. enterica LPS, but to a much lesser extent. Rhodobacter capsulatus PG LPS did not affect TNF-α and IL-6 production induced by the equipotential dose of S. enterica LPS. In order to draw conclusion on the endotoxic activity of particular LPSs, species-specific structure or arrangement of the animal or human immune systems should be considered.
Collapse
Affiliation(s)
- D S Kabanov
- Institute of Basic Biological Problems, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - V A Rykov
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - S V Prokhorenko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitation, Moscow, 107031, Russia
| | - A N Murashev
- Branch of Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - I R Prokhorenko
- Institute of Basic Biological Problems, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
24
|
Zha Z, Wang SY, Chu W, Lv Y, Kan H, Chen Q, Zhong L, Yue L, Xiao J, Wang Y, Yin H. Isolation, purification, structural characterization and immunostimulatory activity of water-soluble polysaccharides from Lepidium meyenii. PHYTOCHEMISTRY 2018; 147:184-193. [PMID: 29353155 DOI: 10.1016/j.phytochem.2018.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 06/07/2023]
Abstract
A water-soluble polysaccharide LMP-1 was isolated and purified by ion-exchange chromatography from maca (Lepidium meyenii Walp.). LMP-1 has a molecular weight of 1.01 × 104 Da, and is composed of glucose and arabinose with a molar ratio of 7.03:1.08. Methylation and the 1D and 2D NMR spectroscopy of LMP-1 revealed that it is mainly composed of →4)-α-D-Glcp-(1→, →6)-α-D-Glcp-(1→, →3)-α-D-Glcp-(1→, and β-D-Araf-(1→, with branching at O-6 of →4,6)-α-D-Glcp-(1 → . LMP-1 showed up-regulation of Toll-like receptor 4 (TLR4) and Toll-like receptor 2 (TLR2). The upstream proteins of Toll-like receptors (TLRs) (CD14 and MD2) and mRNA level of IL-1β also increased. Increased transcription factor nuclear factor-kappa B (NF-κB) p65 was found in the nuclei and cytoplasm in LMP-1-treated RAW264.7 macrophages. These results indicated that LMP-1 activated RAW264.7 macrophages and elicited immunostimulatory activities via the TLRs/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Zhengqi Zha
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Su-Yan Wang
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Weihua Chu
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yang Lv
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Hongjin Kan
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qiuli Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| | - Lili Zhong
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Long Yue
- School of Engineering, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Jinna Xiao
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ying Wang
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Hongping Yin
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
25
|
Martinez-Gil L, Goff PH, Tan GS. The Role of Self-Assembling Lipid Molecules in Vaccination. ADVANCES IN BIOMEMBRANES AND LIPID SELF-ASSEMBLY 2018. [PMCID: PMC7147077 DOI: 10.1016/bs.abl.2017.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The advent of vaccines represents one of the most significant advances in medical history. The protection provided by vaccines has greatly contributed in reducing the number of cases of infections and most notably to the eradication of small pox. A large number of new technologies and approaches in vaccine development are currently being investigated with the goal of providing the basis for the next generation of prophylactics against an ever-expanding list of emerging and reemerging pathogens. In this chapter, we will focus on the role of lipids and lipid self-assembling vesicles in new and promising vaccination approaches. We will start by describing how lipids can induce activation of the innate immune system and focus on some lipid-derived vaccine adjuvants. Next, we will review current lipid-based self-assembling particles used as vaccine platforms, specifically liposomes and virus-like particles, and how virus-like particles have facilitated research of highly pathogenic viruses such as Ebola.
Collapse
|
26
|
Expression level of human TLR4 rather than sequence is the key determinant of LPS responsiveness. PLoS One 2017; 12:e0186308. [PMID: 29020088 PMCID: PMC5636155 DOI: 10.1371/journal.pone.0186308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/28/2017] [Indexed: 12/14/2022] Open
Abstract
To address the role of Toll-like receptor 4 (TLR4) single nucleotide polymorphisms (SNP) in lipopolysaccharide (LPS) recognition, we generated mice that differed only in the sequence of TLR4. We used a bacterial artificial chromosome (BAC) transgenic approach and TLR4/MD-2 knockout mice to specifically examine the role of human TLR4 variants in recognition of LPS. Using in vitro and in vivo assays we found that the expression level rather than the sequence of TLR4 played a larger role in recognition of LPS, especially hypoacylated LPS.
Collapse
|
27
|
Harvey DJ. Analysis of carbohydrates and glycoconjugates by matrix-assisted laser desorption/ionization mass spectrometry: An update for 2011-2012. MASS SPECTROMETRY REVIEWS 2017; 36:255-422. [PMID: 26270629 DOI: 10.1002/mas.21471] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 01/15/2015] [Indexed: 06/04/2023]
Abstract
This review is the seventh update of the original article published in 1999 on the application of MALDI mass spectrometry to the analysis of carbohydrates and glycoconjugates and brings coverage of the literature to the end of 2012. General aspects such as theory of the MALDI process, matrices, derivatization, MALDI imaging, and fragmentation are covered in the first part of the review and applications to various structural types constitute the remainder. The main groups of compound are oligo- and poly-saccharides, glycoproteins, glycolipids, glycosides, and biopharmaceuticals. Much of this material is presented in tabular form. Also discussed are medical and industrial applications of the technique, studies of enzyme reactions, and applications to chemical synthesis. © 2015 Wiley Periodicals, Inc. Mass Spec Rev 36:255-422, 2017.
Collapse
Affiliation(s)
- David J Harvey
- Department of Biochemistry, Oxford Glycobiology Institute, University of Oxford, Oxford, OX1 3QU, UK
| |
Collapse
|
28
|
Watkins HC, Rappazzo CG, Higgins JS, Sun X, Brock N, Chau A, Misra A, Cannizzo JPB, King MR, Maines TR, Leifer CA, Whittaker GR, DeLisa MP, Putnam D. Safe Recombinant Outer Membrane Vesicles that Display M2e Elicit Heterologous Influenza Protection. Mol Ther 2017; 25:989-1002. [PMID: 28215994 DOI: 10.1016/j.ymthe.2017.01.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/02/2017] [Accepted: 01/06/2017] [Indexed: 01/10/2023] Open
Abstract
Recombinant, Escherichia coli-derived outer membrane vesicles (rOMVs), which display heterologous protein subunits, have potential as a vaccine adjuvant platform. One drawback to rOMVs is their lipopolysaccharide (LPS) content, limiting their translatability to the clinic due to potential adverse effects. Here, we explore a unique rOMV construct with structurally remodeled lipids containing only the lipid IVa portion of LPS, which does not stimulate human TLR4. The rOMVs are derived from a genetically engineered B strain of E. coli, ClearColi, which produces lipid IVa, and which was further engineered in our laboratory to hypervesiculate and make rOMVs. We report that rOMVs derived from this lipid IVa strain have substantially attenuated pyrogenicity yet retain high levels of immunogenicity, promote dendritic cell maturation, and generate a balanced Th1/Th2 humoral response. Additionally, an influenza A virus matrix 2 protein-based antigen displayed on these rOMVs resulted in 100% survival against a lethal challenge with two influenza A virus strains (H1N1 and H3N2) in mice with different genetic backgrounds (BALB/c, C57BL/6, and DBA/2J). Additionally, a two-log reduction of lung viral titer was achieved in a ferret model of influenza infection with human pandemic H1N1. The rOMVs reported herein represent a potentially safe and simple subunit vaccine delivery platform.
Collapse
Affiliation(s)
- Hannah C Watkins
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - C Garrett Rappazzo
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jaclyn S Higgins
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Xiangjie Sun
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Nicole Brock
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Annie Chau
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Aditya Misra
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Joseph P B Cannizzo
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Michael R King
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Taronna R Maines
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Cynthia A Leifer
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Gary R Whittaker
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14853, USA
| | - Matthew P DeLisa
- Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA
| | - David Putnam
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
29
|
Lipid A structural modifications in extreme conditions and identification of unique modifying enzymes to define the Toll-like receptor 4 structure-activity relationship. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1439-1450. [PMID: 28108356 DOI: 10.1016/j.bbalip.2017.01.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 01/23/2023]
Abstract
Strategies utilizing Toll-like receptor 4 (TLR4) agonists for treatment of cancer, infectious diseases, and other targets report promising results. Potent TLR4 antagonists are also gaining attention as therapeutic leads. Though some principles for TLR4 modulation by lipid A have been described, a thorough understanding of the structure-activity relationship (SAR) is lacking. Only through a complete definition of lipid A-TLR4 SAR is it possible to predict TLR4 signaling effects of discrete lipid A structures, rendering them more pharmacologically relevant. A limited 'toolbox' of lipid A-modifying enzymes has been defined and is largely composed of enzymes from mesophile human and zoonotic pathogens. Expansion of this 'toolbox' will result from extending the search into lipid A biosynthesis and modification by bacteria living at the extremes. Here, we review the fundamentals of lipid A structure, advances in lipid A uses in TLR4 modulation, and the search for novel lipid A-modifying systems in extremophile bacteria. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
Collapse
|
30
|
Marshall JD, Heeke DS, Rao E, Maynard SK, Hornigold D, McCrae C, Fraser N, Tovchigrechko A, Yu L, Williams N, King S, Cooper ME, Hajjar AM, Woo JC. A Novel Class of Small Molecule Agonists with Preference for Human over Mouse TLR4 Activation. PLoS One 2016; 11:e0164632. [PMID: 27736941 PMCID: PMC5063506 DOI: 10.1371/journal.pone.0164632] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/28/2016] [Indexed: 01/09/2023] Open
Abstract
The best-characterized Toll-like receptor 4 (TLR4) ligands are lipopolysaccharide (LPS) and its chemically modified and detoxified variant, monophosphoryl lipid A (MPL). Although both molecules are active for human TLR4, they demonstrate a potency preference for mouse TLR4 based on data from transfected cell lines and primary cells of both species. After a high throughput screening process of small molecule libraries, we have discovered a new class of TLR4 agonist with a species preference profile differing from MPL. Products of the 4-component Ugi synthesis reaction were demonstrated to potently trigger human TLR4-transfected HEK cells but not mouse TLR4, although inclusion of the human MD2 with mTLR4 was able to partially recover activity. Co-expression of CD14 was not required for optimal activity of Ugi compounds on transfected cells, as it is for LPS. The species preference profile for the panel of Ugi compounds was found to be strongly active for human and cynomolgus monkey primary cells, with reduced but still substantial activity for most Ugi compounds on guinea pig cells. Mouse, rat, rabbit, ferret, and cotton rat cells displayed little or no activity when exposed to Ugi compounds. However, engineering the human versions of TLR4 and MD2 to be expressed in mTLR4/MD2 deficient mice allowed for robust activity by Ugi compounds both in vitro and in vivo. These findings extend the range of compounds available for development as agonists of TLR4 and identify novel molecules which reverse the TLR4 triggering preference of MPL for mouse TLR4 over human TLR4. Such compounds may be amenable to formulation as more potent human-specific TLR4L-based adjuvants than typical MPL-based adjuvants.
Collapse
Affiliation(s)
- Jason D. Marshall
- Vaccine Platform Group, MedImmune, Gaithersburg, Maryland, United States of America
- * E-mail:
| | - Darren S. Heeke
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| | - Eileen Rao
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| | - Sean K. Maynard
- Vaccine Platform Group, MedImmune, Gaithersburg, Maryland, United States of America
| | - David Hornigold
- Department of Cardiovascular and Metabolic Diseases, MedImmune, Cambridge, United Kingdom
| | - Christopher McCrae
- Translational Science, Respiratory, Inflammation and Autoimmunity Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Neil Fraser
- Biology Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Andrey Tovchigrechko
- Research Bioinformatics, MedImmune, Gaithersburg, Maryland, United States of America
| | - Li Yu
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, United States of America
| | - Nicola Williams
- Biology Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Sarah King
- Medicinal Chemistry Department, AstraZeneca R&D, Charnwood, United Kingdom
| | - Martin E. Cooper
- Translational Science, Respiratory, Inflammation and Autoimmunity Innovative Medicines, AstraZeneca R&D, Mölndal, Sweden
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jennifer C. Woo
- Translational Biology Group, MedImmune, Mountain View, California, United States of America
| |
Collapse
|
31
|
A novel algorithm for detecting multiple covariance and clustering of biological sequences. Sci Rep 2016; 6:30425. [PMID: 27451921 PMCID: PMC4958985 DOI: 10.1038/srep30425] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/05/2016] [Indexed: 12/14/2022] Open
Abstract
Single genetic mutations are always followed by a set of compensatory mutations. Thus, multiple changes commonly occur in biological sequences and play crucial roles in maintaining conformational and functional stability. Although many methods are available to detect single mutations or covariant pairs, detecting non-synchronous multiple changes at different sites in sequences remains challenging. Here, we develop a novel algorithm, named Fastcov, to identify multiple correlated changes in biological sequences using an independent pair model followed by a tandem model of site-residue elements based on inter-restriction thinking. Fastcov performed exceptionally well at harvesting co-pairs and detecting multiple covariant patterns. By 10-fold cross-validation using datasets of different scales, the characteristic patterns successfully classified the sequences into target groups with an accuracy of greater than 98%. Moreover, we demonstrated that the multiple covariant patterns represent co-evolutionary modes corresponding to the phylogenetic tree, and provide a new understanding of protein structural stability. In contrast to other methods, Fastcov provides not only a reliable and effective approach to identify covariant pairs but also more powerful functions, including multiple covariance detection and sequence classification, that are most useful for studying the point and compensatory mutations caused by natural selection, drug induction, environmental pressure, etc.
Collapse
|
32
|
Zhang L, Mei M, Yu C, Shen W, Ma L, He J, Yi L. The Functions of Effector Proteins in Yersinia Virulence. Pol J Microbiol 2016; 65:5-12. [PMID: 27281989 DOI: 10.5604/17331331.1197324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Yersinia species are bacterial pathogens that can cause plague and intestinal diseases after invading into human cells through the Three Secretion System (TTSS). The effect of pathogenesis is mediated by Yersinia outer proteins (Yop) and manifested as down-regulation of the cytokine genes expression by inhibiting nuclear factor-κ-gene binding (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. In addition, its pathogenesis can also manipulate the disorder of host innate immune system and cell death such as apoptosis, pyroptosis, and autophagy. Among the Yersinia effector proteins, YopB and YopD assist the injection of other virulence effectors into the host cytoplasm, while YopE, YopH, YopJ, YopO, and YopT target on disrupting host cell signaling pathways in the host cytosols. Many efforts have been applied to reveal that intracellular proteins such as Rho-GTPase, and transmembrane receptors such as Toll-like receptors (TLRs) both play critical roles in Yersinia pathogenesis, establishing a connection between the pathogenic process and the signaling response. This review will mainly focus on how the effector proteins of Yersinia modulate the intrinsic signals in host cells and disturb the innate immunity of hosts through TTSS.
Collapse
|
33
|
Type I Interferon Counters or Promotes Coxiella burnetii Replication Dependent on Tissue. Infect Immun 2016; 84:1815-1825. [PMID: 27068091 DOI: 10.1128/iai.01540-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/01/2016] [Indexed: 01/09/2023] Open
Abstract
Coxiella burnetii is an intracellular pathogen and the cause of Q fever. Gamma interferon (IFN-γ) is critical for host protection from infection, but a role for type I IFN in C. burnetii infection has not been determined. Type I IFN supports host protection from a related pathogen, Legionella pneumophila, and we hypothesized that it would be similarly protective in C. burnetii infection. In contrast to our prediction, IFN-α receptor-deficient (IFNAR(-/-)) mice were protected from C. burnetii-induced infection. Therefore, the role of type I IFN in C. burnetii infection was distinct from that in L. pneumophila Mice treated with a double-stranded-RNA mimetic were protected from C. burnetii-induced weight loss through an IFNAR-independent pathway. We next treated mice with recombinant IFN-α (rIFN-α). When rIFN-α was injected by the intraperitoneal route during infection, disease-induced weight loss was exacerbated. Mice that received rIFN-α by this route had dampened interleukin 1β (IL-1β) expression in bronchoalveolar lavage fluids. However, when rIFN-α was delivered to the lung, bacterial replication was decreased in all tissues. Thus, the presence of type I IFN in the lung protected from infection, but when delivered to the periphery, type I IFN enhanced disease, potentially by dampening inflammatory cytokines. To better characterize the capacity for type I IFN induction by C. burnetii, we assessed expression of IFN-β transcripts by human macrophages following stimulation with lipopolysaccharide (LPS) from C. burnetii Understanding innate responses in C. burnetii infection will support the discovery of novel therapies that may be alternative or complementary to the current antibiotic treatment.
Collapse
|
34
|
Ma Y, Han F, Liang J, Yang J, Shi J, Xue J, Yang L, Li Y, Luo M, Wang Y, Wei J, Liu X. A species-specific activation of Toll-like receptor signaling in bovine and sheep bronchial epithelial cells triggered by Mycobacterial infections. Mol Immunol 2016; 71:23-33. [DOI: 10.1016/j.molimm.2016.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/04/2016] [Indexed: 01/29/2023]
|
35
|
TLR Stimulation Dynamically Regulates Heme and Iron Export Gene Expression in Macrophages. J Immunol Res 2016; 2016:4039038. [PMID: 27006955 PMCID: PMC4783552 DOI: 10.1155/2016/4039038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/22/2016] [Accepted: 01/31/2016] [Indexed: 01/01/2023] Open
Abstract
Pathogenic bacteria have evolved multiple mechanisms to capture iron or iron-containing heme from host tissues or blood. In response, organisms have developed defense mechanisms to keep iron from pathogens. Very little of the body's iron store is available as free heme; rather nearly all body iron is complexed with heme or other proteins. The feline leukemia virus, subgroup C (FeLV-C) receptor, FLVCR, exports heme from cells. It was unknown whether FLVCR regulates heme-iron availability after infection, but given that other heme regulatory proteins are upregulated in macrophages in response to bacterial infection, we hypothesized that macrophages dynamically regulate FLVCR. We stimulated murine primary macrophages or macrophage cell lines with LPS and found that Flvcr is rapidly downregulated in a TLR4/MD2-dependent manner; TLR1/2 and TLR3 stimulation also decreased Flvcr expression. We identified several candidate TLR-activated transcription factors that can bind to the Flvcr promoter. Macrophages must balance the need to sequester iron from systemic circulating or intracellular pathogens with the macrophage requirement for heme and iron to produce reactive oxygen species. Our findings underscore the complexity of this regulation and point to a new role for FLVCR and heme export in macrophages responses to infection and inflammation.
Collapse
|
36
|
Sironi M, Cagliani R, Forni D, Clerici M. Evolutionary insights into host-pathogen interactions from mammalian sequence data. Nat Rev Genet 2015; 16:224-36. [PMID: 25783448 PMCID: PMC7096838 DOI: 10.1038/nrg3905] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Infections are one of the major selective pressures acting on humans, and host-pathogen interactions contribute to shaping the genetic diversity of both organisms. Evolutionary genomic studies take advantage of experiments that natural selection has been performing over millennia. In particular, inter-species comparative genomic analyses can highlight the genetic determinants of infection susceptibility or severity. Recent examples show how evolution-guided approaches can provide new insights into host-pathogen interactions, ultimately clarifying the basis of host range and explaining the emergence of different diseases. We describe the latest developments in comparative immunology and evolutionary genetics, showing their relevance for understanding the molecular determinants of infection susceptibility in mammals.
Collapse
Affiliation(s)
- Manuela Sironi
- Bioinformatics, Scientific Institute IRCCS E. Medea, 23842 Bosisio Parini, Italy
| | - Rachele Cagliani
- Bioinformatics, Scientific Institute IRCCS E. Medea, 23842 Bosisio Parini, Italy
| | - Diego Forni
- Bioinformatics, Scientific Institute IRCCS E. Medea, 23842 Bosisio Parini, Italy
| | - Mario Clerici
- 1] Department of Physiopathology and Transplantation, University of Milan, 20090 Milan, Italy. [2] Don C. Gnocchi Foundation ONLUS, IRCCS, 20148 Milan, Italy
| |
Collapse
|
37
|
Abstract
During the past decade, the development of humanized mouse models and their general applications in biomedical research greatly accelerated the translation of outcomes obtained from basic research into potential diagnostic and therapeutic strategies in clinic. In this chapter, we firstly present an overview on the history and current progress of diverse humanized mouse models and then focus on those equipped with reconstituted human immune system. The update advancement in the establishment of humanized immune system mice and their applications in the studies of the development of human immune system and the pathogenesis of multiple human immune-related diseases are intensively reviewed here, while the shortcoming and perspective of these potent tools are discussed as well. As a valuable bridge across the gap between bench work and clinical trial, progressive humanized mouse models will undoubtedly continue to play an indispensable role in the wide area of biomedical research.
Collapse
|
38
|
Shin S, Brodsky IE. The inflammasome: Learning from bacterial evasion strategies. Semin Immunol 2015; 27:102-10. [PMID: 25914126 DOI: 10.1016/j.smim.2015.03.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 03/12/2015] [Indexed: 01/01/2023]
Abstract
The innate immune system plays a critical role in defense against microbial infection and employs germline-encoded pattern recognition receptors to detect broadly conserved microbial structures or activities. Pattern recognition receptors of the nucleotide binding domain/leucine rich repeat (NLR) family respond to particular microbial products or disruption of cellular physiology, and mediate the activation of an arm of the innate immune response termed the inflammasome. Inflammasomes are multiprotein complexes that are inducibly assembled in response to the contamination of the host cell cytosol by microbial products. Individual NLRs sense the presence of their cognate stimuli, and initiate assembly of inflammasomes via the adaptor protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and the effector pro-enzyme caspase-1. Inflammasome activation leads to rapid release of pro-inflammatory mediators of the IL-1 family as well as the release of intracellular alarmins due to a lytic form of programmed cell death termed pyroptosis. Over the past 15 years, a great deal has been learned about the mechanisms that drive inflammasome activation in response to infection by diverse pathogens. However, pathogens have also evolved mechanisms to evade or suppress host defenses, and the mechanisms by which pathogens evade inflammasome activation are not well-understood. Here, we will discuss emerging evidence on how diverse pathogens evade inflammasome activation, and what these studies have revealed about inflammasome biology. Deeper understanding of pathogen evasion of inflammasome activation has the potential to lead to development of novel classes of immunomodulatory factors that could be used in the context of human inflammatory diseases.
Collapse
Affiliation(s)
- Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Maeshima N, Evans-Atkinson T, Hajjar AM, Fernandez RC. Bordetella pertussis Lipid A Recognition by Toll-like Receptor 4 and MD-2 Is Dependent on Distinct Charged and Uncharged Interfaces. J Biol Chem 2015; 290:13440-53. [PMID: 25837248 DOI: 10.1074/jbc.m115.653881] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Indexed: 12/22/2022] Open
Abstract
Lipid A in LPS activates innate immunity through the Toll-like receptor 4 (TLR4)-MD-2 complex on host cells. Variation in lipid A has significant consequences for TLR4 activation and thus may be a means by which Gram-negative bacteria modulate host immunity. However, although even minor changes in lipid A structure have been shown to affect downstream immune responses, the mechanism by which the TLR4-MD-2 receptor complex recognizes these changes is not well understood. We previously showed that strain BP338 of the human pathogen Bordetella pertussis, the causative agent of whooping cough, modifies its lipid A by the addition of glucosamine moieties that promote TLR4 activation in human, but not mouse, macrophages. Using site-directed mutagenesis and an NFκB reporter assay screen, we have identified several charged amino acid residues in TLR4 and MD-2 that are important for these species-specific responses; some of these are novel for responses to penta-acyl B. pertussis LPS, and their mutation does not affect the response to hexa-acylated Escherichia coli LPS or tetra-acylated lipid IVA. We additionally show evidence that suggests that recognition of penta-acylated B. pertussis lipid A is dependent on uncharged amino acids in TLR4 and MD-2 and that this is true for both human and mouse TLR4-MD-2 receptors. Taken together, we have demonstrated that the TLR4-MD-2 receptor complex recognizes variation in lipid A molecules using multiple sites for receptor-ligand interaction and propose that host-specific immunity to a particular Gram-negative bacterium is, at least in part, mediated by very subtle tuning of one of the earliest interactions at the host-pathogen interface.
Collapse
Affiliation(s)
- Nina Maeshima
- From the Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3 and
| | - Tara Evans-Atkinson
- From the Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3 and
| | - Adeline M Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington 98195
| | - Rachel C Fernandez
- From the Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3 and
| |
Collapse
|
40
|
Zariri A, van der Ley P. Biosynthetically engineered lipopolysaccharide as vaccine adjuvant. Expert Rev Vaccines 2015; 14:861-76. [PMID: 25797360 DOI: 10.1586/14760584.2015.1026808] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lipopolysaccharide (LPS), a dominant component of the Gram-negative bacterial outer membrane, is a strong activator of the innate immune system, and thereby an important determinant in the adaptive immune response following bacterial infection. This adjuvant activity can be harnessed following immunization with bacteria-derived vaccines that naturally contain LPS, and when LPS or molecules derived from it are added to purified vaccine antigens. However, the downside of the strong biological activity of LPS is its ability to contribute to vaccine reactogenicity. Modification of the LPS structure allows triggering of a proper immune response needed in a vaccine against a particular pathogen while at the same time lowering its toxicity. Extensive modifications to the basic structure are possible by using our current knowledge of bacterial genes involved in LPS biosynthesis and modification. This review focuses on biosynthetic engineering of the structure of LPS and implications of these modifications for generation of safe adjuvants.
Collapse
Affiliation(s)
- Afshin Zariri
- Institute for Translational Vaccinology (InTraVacc), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | | |
Collapse
|
41
|
Manček-Keber M, Jerala R. Postulates for validating TLR4 agonists. Eur J Immunol 2015; 45:356-70. [DOI: 10.1002/eji.201444462] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 10/20/2014] [Accepted: 12/01/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Mateja Manček-Keber
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| | - Roman Jerala
- Department of Biotechnology; National Institute of Chemistry; Ljubljana Slovenia
- EN-FIST Centre of Excellence; Ljubljana Slovenia
| |
Collapse
|
42
|
Merritt PM, Nero T, Bohman L, Felek S, Krukonis ES, Marketon MM. Yersinia pestis targets neutrophils via complement receptor 3. Cell Microbiol 2014; 17:666-87. [PMID: 25359083 DOI: 10.1111/cmi.12391] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Revised: 10/18/2014] [Accepted: 10/28/2014] [Indexed: 01/20/2023]
Abstract
Yersinia species display a tropism for lymphoid tissues during infection, and the bacteria select innate immune cells for delivery of cytotoxic effectors by the type III secretion system. Yet, the mechanism for target cell selection remains a mystery. Here we investigate the interaction of Yersinia pestis with murine splenocytes to identify factors that participate in the targeting process. We find that interactions with primary immune cells rely on multiple factors. First, the bacterial adhesin Ail is required for efficient targeting of neutrophils in vivo. However, Ail does not appear to directly mediate binding to a specific cell type. Instead, we find that host serum factors direct Y. pestis to specific innate immune cells, particularly neutrophils. Importantly, specificity towards neutrophils was increased in the absence of bacterial adhesins because of reduced targeting of other cell types, but this phenotype was only visible in the presence of mouse serum. Addition of antibodies against complement receptor 3 and CD14 blocked target cell selection, suggesting that a combination of host factors participate in steering bacteria towards neutrophils during plague infection.
Collapse
Affiliation(s)
- Peter M Merritt
- Department of Biology, Indiana University, Bloomington, IN, USA
| | | | | | | | | | | |
Collapse
|
43
|
Tanner SM, Berryhill TF, Ellenburg JL, Jilling T, Cleveland DS, Lorenz RG, Martin CA. Pathogenesis of necrotizing enterocolitis: modeling the innate immune response. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 185:4-16. [PMID: 25447054 DOI: 10.1016/j.ajpath.2014.08.028] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 08/21/2014] [Accepted: 08/27/2014] [Indexed: 12/23/2022]
Abstract
Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. The pathophysiology is likely secondary to innate immune responses to intestinal microbiota by the premature infant's intestinal tract, leading to inflammation and injury. This review provides an updated summary of the components of the innate immune system involved in NEC pathogenesis. In addition, we evaluate the animal models that have been used to study NEC with regard to the involvement of innate immune factors and histopathological changes as compared to those seen in infants with NEC. Finally, we discuss new approaches to studying NEC, including mathematical models of intestinal injury and the use of humanized mice.
Collapse
Affiliation(s)
- Scott M Tanner
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama; Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Taylor F Berryhill
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - James L Ellenburg
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tamas Jilling
- Division of Neonatology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dava S Cleveland
- Department of Pediatric Pathology, Children's Hospital of Alabama, Birmingham, Alabama
| | - Robin G Lorenz
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.
| | - Colin A Martin
- Department of Pediatric Surgery, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
44
|
Loomis WP, Johnson ML, Brasfield A, Blanc MP, Yi J, Miller SI, Cookson BT, Hajjar AM. Temporal and anatomical host resistance to chronic Salmonella infection is quantitatively dictated by Nramp1 and influenced by host genetic background. PLoS One 2014; 9:e111763. [PMID: 25350459 PMCID: PMC4211889 DOI: 10.1371/journal.pone.0111763] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/30/2014] [Indexed: 01/09/2023] Open
Abstract
The lysosomal membrane transporter, Nramp1, plays a key role in innate immunity and resistance to infection with intracellular pathogens such as non-typhoidal Salmonella (NTS). NTS-susceptible C57BL/6 (B6) mice, which express the mutant Nramp1D169 allele, are unable to control acute infection with Salmonella enterica serovar Typhimurium following intraperitoneal or oral inoculation. Introducing functional Nramp1G169 into the B6 host background, either by constructing a congenic strain carrying Nramp1G169 from resistant A/J mice (Nramp-Cg) or overexpressing Nramp1G169 from a transgene (Nramp-Tg), conferred equivalent protection against acute Salmonella infection. In contrast, the contributions of Nramp1 for controlling chronic infection are more complex, involving temporal and anatomical differences in Nramp1-dependent host responses. Nramp-Cg, Nramp-Tg and NTS-resistant 129×1/SvJ mice survived oral Salmonella infection equally well for the first 2–3 weeks, providing evidence that Nramp1 contributes to the initial control of NTS bacteremia preceding establishment of chronic Salmonella infection. By day 30, increased host Nramp1 expression (Tg>Cg) provided greater protection as indicated by decreased splenic bacterial colonization (Tg<Cg). However, despite controlling bacterial growth within MLN as effectively as 129×1/SvJ mice, Nramp-Cg and Nramp-Tg mice eventually succumbed to infection. These data indicate: 1) discrete, anatomically localized host resistance is conferred by Nramp1 expression in NTS-susceptible mice, 2) restriction of systemic bacterial growth in the spleens of NTS-susceptible mice is enhanced by Nramp1 expression and dose-dependent, and 3) host genes other than Nramp1 also contribute to the ability of NTS-resistant 129×1/SvJ mice to control bacterial replication during chronic infection.
Collapse
Affiliation(s)
- Wendy P. Loomis
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Matthew L. Johnson
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Alicia Brasfield
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Marie-Pierre Blanc
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Jaehun Yi
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Samuel I. Miller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Departments of Medicine and Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | - Brad T. Cookson
- Department of Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
45
|
Nour A, Hayashi T, Chan M, Yao S, Tawatao RI, Crain B, Tsigelny IF, Kouznetsova VL, Ahmadiiveli A, Messer K, Pu M, Corr M, Carson DA, Cottam HB. Discovery of substituted 4-aminoquinazolines as selective Toll-like receptor 4 ligands. Bioorg Med Chem Lett 2014; 24:4931-8. [PMID: 25288184 DOI: 10.1016/j.bmcl.2014.09.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 09/10/2014] [Accepted: 09/12/2014] [Indexed: 01/23/2023]
Abstract
The Toll-like receptors (TLRs) are critical components of the innate immune system that regulate immune recognition in part through NF-κB activation. A human cell-based high throughput screen (HTS) revealed substituted 4-aminoquinazolines to be small molecular weight activators of NF-κB. The most potent hit compound predominantly stimulated through the human TLR4/MD2 complex, and had less activity with the mouse TLR4/MD2. There was no activity with other TLRs and the TLR4 activation was MD-2 dependent and CD14 independent. Synthetic modifications of the quinazoline scaffold at the 2 and 4 positions revealed trends in structure-activity relationships with respect to TLR dependent production of the NF-κB associated cytokine IL-8 in human peripheral blood mononuclear cells, as well as IL-6 in mouse antigen presenting cells. Furthermore, the hit compound in this series also activated the interferon signaling pathway resulting in type I interferon production. Substitution at the O-phenyl moiety with groups such as bromine, chlorine and methyl resulted in enhanced immunological activity. Computational studies indicated that the 4-aminoquinazoline compounds bind primarily to human MD-2 in the TLR4/MD-2 complex. These small molecules, which preferentially stimulate human rather than mouse innate immune cells, may be useful as adjuvants or immunotherapeutic agents.
Collapse
Affiliation(s)
- Afshin Nour
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Tomoko Hayashi
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Michael Chan
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Shiyin Yao
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Rommel I Tawatao
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Brian Crain
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Igor F Tsigelny
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States; San Diego Supercomputer Center, and University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States; Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - Valentina L Kouznetsova
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States; San Diego Supercomputer Center, and University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States
| | - Alast Ahmadiiveli
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Karen Messer
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Minya Pu
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Maripat Corr
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0663, United States
| | - Dennis A Carson
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| | - Howard B Cottam
- Moores Cancer Center, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, United States
| |
Collapse
|
46
|
Snyder JM, Treuting PM, Nagy L, Yam C, Yi J, Brasfield A, Nguyen LPA, Hajjar AM. Humanized TLR7/8 expression drives proliferative multisystemic histiocytosis in C57BL/6 mice. PLoS One 2014; 9:e107257. [PMID: 25229618 PMCID: PMC4168129 DOI: 10.1371/journal.pone.0107257] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/11/2014] [Indexed: 02/04/2023] Open
Abstract
A humanized TLR7/TLR8 transgenic mouse line was engineered for studies using TLR7/8 ligands as vaccine adjuvants. The mice developed a spontaneous immune-mediated phenotype prior to six months of age characterized by runting, lethargy, blepharitis, and corneal ulceration. Histological examination revealed a marked, multisystemic histiocytic infiltrate that effaced normal architecture. The histological changes were distinct from those previously reported in mouse models of systemic lupus erythematosus. When the mice were crossed with MyD88-/- mice, which prevented toll-like receptor signaling, the inflammatory phenotype resolved. Illness may be caused by constitutive activation of human TLR7 or TLR8 in the bacterial artificial chromosome positive mice as increased TLR7 and TLR8 expression or activation has previously been implicated in autoimmune disease.
Collapse
Affiliation(s)
- Jessica M. Snyder
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
- Comparative Pathology Program, University of Washington, Seattle, Washington, United States of America
| | - Piper M. Treuting
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
- Comparative Pathology Program, University of Washington, Seattle, Washington, United States of America
| | - Lee Nagy
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Cathy Yam
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Jaehun Yi
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Alicia Brasfield
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Lisa Phuong Anh Nguyen
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| | - Adeline M. Hajjar
- Department of Comparative Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
47
|
Pouliot K, Buglione-Corbett R, Marty-Roix R, Montminy-Paquette S, West K, Wang S, Lu S, Lien E. Contribution of TLR4 and MyD88 for adjuvant monophosphoryl lipid A (MPLA) activity in a DNA prime-protein boost HIV-1 vaccine. Vaccine 2014; 32:5049-56. [PMID: 25045815 PMCID: PMC10687719 DOI: 10.1016/j.vaccine.2014.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 06/17/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022]
Abstract
Recombinant protein vaccines are commonly formulated with an immune-stimulatory compound, or adjuvant, to boost immune responses to a particular antigen. Recent studies have shown that, through recognition of molecular motifs, receptors of the innate immune system are involved in the functions of adjuvants to generate and direct adaptive immune responses. However, it is not clear to which degree those receptors are also important when the adjuvant is used as part of a novel heterologous prime-boost immunization process in which the priming and boosting components are not the same type of vaccines. In the current study, we compared the immune responses elicited by a pentavalent HIV-1 DNA prime-protein boost vaccine in mice deficient in either Toll-like receptor 4 (TLR4) or myeloid differentiation primary response gene 88 (MyD88) to wildtype mice. HIV gp120 protein administered in the boost phase was formulated with either monophosphoryl lipid A (MPLA), QS-21, or Al(OH)3. Endpoint antibody titer, serum cytokine response and T-cell memory response were assessed. Neither TLR4 nor MyD88 deficiency had a significant effect on the immune response of mice given vaccine formulated with QS-21 or Al(OH)3. However, TLR4- and MyD88-deficiency decreased both the antibody and T-cell responses in mice administered HIV gp120 formulated with MPLA. These results further our understanding of the activation of TLR4 and MyD88 by MPLA in the context of a DNA prime/protein boost immunization strategy.
Collapse
Affiliation(s)
- Kimberly Pouliot
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Rachel Buglione-Corbett
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Robyn Marty-Roix
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Sara Montminy-Paquette
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Kim West
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Egil Lien
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States; Centre of Molecular Inflammation Research, Dept. of Cancer and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
48
|
Hayashi T, Crain B, Yao S, Caneda CD, Cottam HB, Chan M, Corr M, Carson DA. Novel synthetic toll-like receptor 4/MD2 ligands attenuate sterile inflammation. J Pharmacol Exp Ther 2014; 350:330-40. [PMID: 24893985 PMCID: PMC4109491 DOI: 10.1124/jpet.114.214312] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/02/2014] [Indexed: 01/19/2023] Open
Abstract
Toll-like receptor (TLR) stimulation has been implicated as a major contributor to chronic inflammation. Among these receptors, TLR4 has been described as a key regulator of endogenous inflammation and has been proposed as a therapeutic target. Previously, we discovered by high-throughput screening a group of substituted pyrimido[5,4-b]indoles that activated a nuclear factor-κB reporter in THP-1 human monocytic cells. A biologically active hit compound was resynthesized, and derivatives were prepared to assess structure-activity relationships. The derived compounds activated cells in a TLR4/myeloid differentiation protein 2 (MD2)-dependent and CD14-independent manner, using the myeloid differentiation primary response 88 and Toll/IL-1 receptor domain-containing adapter-inducing interferon-β pathways. Two lead compounds, 1Z105 and 1Z88, were selected for further analysis based on favorable biologic properties and lack of toxicity. In vivo pharmacokinetics indicated that 1Z105 was orally bioavailable, whereas 1Z88 was not. Oral or parenteral doses of 1Z105 and 1Z88 induced undetectable or negligible levels of circulating cytokines and did not induce hepatotoxicity when administered to galactosamine-conditioned mice, indicating good safety profiles. Both compounds were very effective in preventing lethal liver damage in lipopolysaccharide treated galatosamine-conditioned mice. Orally administered 1Z105 and parenteral 1Z88 prevented arthritis in an autoantibody-driven murine model. Hence, these low molecular weight molecules that target TLR4/MD2 were well tolerated and effective in reducing target organ damage in two different mouse models of sterile inflammation.
Collapse
Affiliation(s)
- Tomoko Hayashi
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Brian Crain
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Shiyin Yao
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Christa D Caneda
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Howard B Cottam
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Michael Chan
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Maripat Corr
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| | - Dennis A Carson
- Rebecca and John Moores UCSD Cancer Center (T.H., B.C., S.Y., H.B.C., M.Ch., D.A.C.) and Department of Medicine, University of California San Diego, La Jolla, California (C.D.C., M.Co.)
| |
Collapse
|
49
|
Wang X, Quinn PJ, Yan A. Kdo2 -lipid A: structural diversity and impact on immunopharmacology. Biol Rev Camb Philos Soc 2014; 90:408-27. [PMID: 24838025 PMCID: PMC4402001 DOI: 10.1111/brv.12114] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 04/10/2014] [Accepted: 04/17/2014] [Indexed: 12/11/2022]
Abstract
3-deoxy-d-manno-octulosonic acid-lipid A (Kdo2-lipid A) is the essential component of lipopolysaccharide in most Gram-negative bacteria and the minimal structural component to sustain bacterial viability. It serves as the active component of lipopolysaccharide to stimulate potent host immune responses through the complex of Toll-like-receptor 4 (TLR4) and myeloid differentiation protein 2. The entire biosynthetic pathway of Escherichia coli Kdo2-lipid A has been elucidated and the nine enzymes of the pathway are shared by most Gram-negative bacteria, indicating conserved Kdo2-lipid A structure across different species. Yet many bacteria can modify the structure of their Kdo2-lipid A which serves as a strategy to modulate bacterial virulence and adapt to different growth environments as well as to avoid recognition by the mammalian innate immune systems. Key enzymes and receptors involved in Kdo2-lipid A biosynthesis, structural modification and its interaction with the TLR4 pathway represent a clear opportunity for immunopharmacological exploitation. These include the development of novel antibiotics targeting key biosynthetic enzymes and utilization of structurally modified Kdo2-lipid A or correspondingly engineered live bacteria as vaccines and adjuvants. Kdo2-lipid A/TLR4 antagonists can also be applied in anti-inflammatory interventions. This review summarizes recent knowledge on both the fundamental processes of Kdo2-lipid A biosynthesis, structural modification and immune stimulation, and applied research on pharmacological exploitations of these processes for therapeutic development.
Collapse
Affiliation(s)
- Xiaoyuan Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China; Synergetic Innovation Center of Food Safety and Nutrition, Jiangnan University, Wuxi, China
| | | | | |
Collapse
|
50
|
Chang MY, Tanino Y, Vidova V, Kinsella MG, Chan CK, Johnson PY, Wight TN, Frevert CW. Reprint of: A rapid increase in macrophage-derived versican and hyaluronan in infectious lung disease. Matrix Biol 2014; 35:162-73. [PMID: 24727035 PMCID: PMC4096977 DOI: 10.1016/j.matbio.2014.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/21/2014] [Accepted: 01/21/2014] [Indexed: 01/08/2023]
Abstract
The goals of this study were to characterize the changes in chondroitin sulfate proteoglycans and hyaluronan in lungs in acute response to gram-negative bacterial infection and to identify cellular components responsible for these changes. Mice were treated with intratracheal (IT) live Escherichia coli, E. coli lipopolysaccharide (LPS), or PBS. Both E. coli and LPS caused rapid selective increases in mRNA expression of versican and hyaluronan synthase (Has) isoforms 1 and 2 associated with increased immunohistochemical and histochemical staining for versican and hyaluronan in the lungs. Versican was associated with a subset of alveolar macrophages. To examine whether macrophages contribute to versican and hyaluronan accumulation, in vitro studies with primary cultures of bone marrow-derived and alveolar macrophages were performed. Unstimulated macrophages expressed very low levels of versican and hyaluronan synthase mRNA, with no detectible versican protein or hyaluronan product. Stimulation with LPS caused rapid increases in versican mRNA and protein, a rapid increase in Has1 mRNA, and concomitant inhibition of hyaluronidases 1 and 2, the major hyaluronan degrading enzymes. Hyaluronan could be detected following chloroquine pre-treatment, indicating rapid turnover and degradation of hyaluronan by macrophages. In addition, the effects of LPS, the M1 macrophage classical activation agonist, were compared to those of IL-4/IL-13 or IL-10, the M2a and M2c alternative activation agonists, respectively. Versican and Has1 increased only in response to M1 activation. Finally, the up-regulation of versican and Has1 in the whole lungs of wild-type mice following IT LPS was completely abrogated in TLR-4(-/-) mice. These findings suggest that versican and hyaluronan synthesis may play an important role in the innate immune response to gram-negative lung infection.
Collapse
Affiliation(s)
- Mary Y Chang
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States.
| | - Yoshinori Tanino
- Fukushima Medical University School of Medicine, Department of Pulmonary Medicine, Fukushima, Japan
| | - Veronika Vidova
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Michael G Kinsella
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Christina K Chan
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Pamela Y Johnson
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Thomas N Wight
- Hope Heart Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Charles W Frevert
- Comparative Pathology Program, Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA, United States; Division of Pulmonary/Critical Care Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|