1
|
Sainz-Mejías M, Ma C, Hou Y, Jurado-Martin I, Romerio A, Franco AR, Shaik MM, Tomás-Cortázar J, Peri F, McClean S. Monosaccharide-Based Synthetic TLR4 Agonist Enhances Vaccine Efficacy against Pseudomonas aeruginosa Challenge. ACS Infect Dis 2025; 11:894-904. [PMID: 40129118 PMCID: PMC11998000 DOI: 10.1021/acsinfecdis.4c00932] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/26/2025]
Abstract
Vaccine adjuvants are critical to improve the immunogenicity, efficacy, and durability of vaccines; however, their development has lagged behind that of vaccine antigens. Monophosphoryl lipid A (MPLA), a clinically approved adjuvant that stimulates Toll-like receptor 4 (TLR4), faces manufacturing challenges due to its complex and long synthesis. With the aim of simplifying the structure of MPLA while retaining its biological activity, we developed monosaccharide-based molecules FP18 and FP20Rha that activate TLR4 signaling. Both TLR4 agonists induced robust antibody activity against the model antigen, ovalbumin. Here, we report the potential of these TLR4 agonists to enhance the protective efficacy of the well-characterized OprF antigen against P. aeruginosa infection. OprF adjuvanted with FP18 showed reduced bacterial loads in lungs and spleens, relative to antigen alone in an acute P. aeruginosa pneumonia model. FP18-adjuvanted OprF also enhanced the production of anti-OprF antibodies and stimulated IFNγ and TNF in CD4+ T cells, suggesting a Th1-skewed cellular immune response. These adjuvants have promise for accelerating the development of effective vaccines against P. aeruginosa and other infectious diseases.
Collapse
Affiliation(s)
- Maite Sainz-Mejías
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Chaoying Ma
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Yueran Hou
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Irene Jurado-Martin
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Ana Rita Franco
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Mohammed Monsoor Shaik
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Julen Tomás-Cortázar
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, Milano 20126, Italy
| | - Siobhán McClean
- School
of Biomolecular and Biomedical Sciences and Conway Institute of Biomolecular
and Biomedical Research, University College
Dublin, Belfield, Dublin 4 D04 V1W8, Ireland
| |
Collapse
|
2
|
Solleti SK, Matthews BE, Wu J, Rowe RK. SHIP-1 Differentially Regulates IgE-Induced IL-10 and Antiviral Responses in Human Monocytes. Eur J Immunol 2025; 55:e202451065. [PMID: 39668409 DOI: 10.1002/eji.202451065] [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: 02/12/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
IgE-mediated stimulation of monocytes regulates multiple cellular functions including cellular maturation, cytokine release, antiviral responses, and T-cell differentiation. Expression of the high-affinity IgE receptor, FcεRI, is closely linked to serum IgE levels and atopic disease. The signaling molecules regulating FcεRI effector functions have been well studied in mast cells and basophils; however, less is known about the signaling and regulatory mechanisms in monocytes. This study sought to identify regulators of IgE-mediated cytokine release in human monocytes. SHIP-1 was identified as a negative regulator of IgE-induced IL-10 production. It was also determined that IgE-mediated stimulation and SHIP-1 inhibition decreased antiviral IP-10 production after liposomal poly(I:C) stimulation, indicating differential regulation by SHIP-1 in IgE-driven and antiviral response pathways. SHIP-1 and NF-κB were activated following IgE-mediated stimulation of monocytes, and NF-κB activation was related to both SHIP-1 and FcεRIα cellular expression levels. To our knowledge, this is the first study to identify a role for SHIP-1 in regulating IgE-mediated and antiviral responses in human monocytes. Given the importance of monocytes in inflammation and immune responses, a better understanding of the signaling and regulatory mechanisms downstream of the FcεRI receptor could lead to new therapeutic targets in allergic disease.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Bailey E Matthews
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Jingyi Wu
- Biomedical Genetics and Genomics Graduate Program, University of Rochester Medical Center, Rochester, New York, USA
| | - Regina K Rowe
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
3
|
Matera A, Compagnion AC, Pedicone C, Kotah JM, Ivanov A, Monsorno K, Labouèbe G, Leggio L, Pereira-Iglesias M, Beule D, Mansuy-Aubert V, Williams TL, Iraci N, Sierra A, Marro SG, Goate AM, Eggen BJL, Kerr WG, Paolicelli RC. Microglial lipid phosphatase SHIP1 limits complement-mediated synaptic pruning in the healthy developing hippocampus. Immunity 2025; 58:197-217.e13. [PMID: 39657671 DOI: 10.1016/j.immuni.2024.11.003] [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: 04/25/2023] [Revised: 10/13/2024] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
The gene inositol polyphosphate-5-phosphatase D (INPP5D), which encodes the lipid phosphatase SH2-containing inositol polyphosphate 5-phosphatase 1 (SHIP1), is associated with the risk of Alzheimer's disease (AD). How it influences microglial function and brain physiology is unclear. Here, we showed that SHIP1 was enriched in early stages of healthy brain development. By combining in vivo loss-of-function approaches and proteomics, we discovered that mice conditionally lacking microglial SHIP1 displayed increased complement and synapse loss in the early postnatal brain. SHIP1-deficient microglia showed altered transcriptional signatures and abnormal synaptic pruning that was dependent on the complement system. Mice exhibited cognitive defects in adulthood only when microglial SHIP1 was depleted early postnatally but not at later stages. Induced pluripotent stem cell (iPSC)-derived microglia lacking SHIP1 also showed increased engulfment of synaptic structures. These findings suggest that SHIP1 is essential for proper microglia-mediated synapse remodeling in the healthy developing brain. Disrupting this process has lasting behavioral effects and may be linked to vulnerability to neurodegeneration.
Collapse
Affiliation(s)
- Alessandro Matera
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Chiara Pedicone
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Janssen M Kotah
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Andranik Ivanov
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katia Monsorno
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gwenaël Labouèbe
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Loredana Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Marta Pereira-Iglesias
- Achucarro Basque Center for Neuroscience, Barrio Sarriena s/n, Leioa, Spain; Department of Neuroscience, University of the Basque Country EHU/UPV, Barrio Sarriena s/n, Leioa, Spain
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Tim L Williams
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Nunzio Iraci
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, Barrio Sarriena s/n, Leioa, Spain; Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, Barrio Sarriena, Leioa, Spain; Ikerbasque Foundation, Bilbao, Spain
| | - Samuele G Marro
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosciences, Black Family Stem Cell Institute, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Alison M Goate
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - William G Kerr
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA; Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
4
|
Yeoh WJ, Krebs P. SHIP1 and its role for innate immune regulation-Novel targets for immunotherapy. Eur J Immunol 2023; 53:e2350446. [PMID: 37742135 DOI: 10.1002/eji.202350446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/03/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023]
Abstract
Phosphoinositide-3-kinase/AKT (PI3K/AKT) signaling plays key roles in the regulation of cellular activity in both health and disease. In immune cells, this PI3K/AKT pathway is critically regulated by the phosphoinositide phosphatase SHIP1, which has been reported to modulate the function of most immune subsets. In this review, we summarize our current knowledge of SHIP1 with a focus on innate immune cells, where we reflect on the most pertinent aspects described in the current literature. We also present several small-molecule agonists and antagonists of SHIP1 developed over the last two decades, which have led to improved outcomes in several preclinical models of disease. We outline these promising findings and put them in relation to human diseases with unmet medical needs, where we discuss the most attractive targets for immune therapies based on SHIP1 modulation.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Astragalus Saponins, Astragaloside VII and Newly Synthesized Derivatives, Induce Dendritic Cell Maturation and T Cell Activation. Vaccines (Basel) 2023; 11:vaccines11030495. [PMID: 36992079 DOI: 10.3390/vaccines11030495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
Astragaloside VII (AST VII), a triterpenic saponin isolated from Astragalus species, shows promise as a vaccine adjuvant, as it supported a balanced Th1/Th2 immune response in previous in vivo studies. However, the underlying mechanisms of its adjuvant activity have not been defined. Here, we investigated the impact of AST VII and its newly synthesized semi-synthetic analogs on human whole blood cells, as well as on mouse bone marrow-derived dendritic cells (BMDCs). Cells were stimulated with AST VII and its derivatives in the presence or absence of LPS or PMA/ionomycin and the secretion of cytokines and the expression of activation markers were analyzed using ELISA and flow cytometry, respectively. AST VII and its analogs increased the production of IL-1β in PMA/ionomycin-stimulated human whole blood cells. In LPS-treated mouse BMDCs, AST VII increased the production of IL-1β and IL-12, and the expression of MHC II, CD86, and CD80. In mixed leukocyte reaction, AST VII and derivatives increased the expression of the activation marker CD44 on mouse CD4+ and CD8+ T cells. In conclusion, AST VII and its derivatives strengthen pro-inflammatory responses and support dendritic cell maturation and T cell activation in vitro. Our results provide insights into the mechanisms of the adjuvant activities of AST VII and its analogs, which will be instrumental to improve their utility as a vaccine adjuvant.
Collapse
|
6
|
Thomas JHL, Lui L, Abell A, Tieu W, Somogyi AA, Bajic JE, Hutchinson MR. Toll-like receptors change morphine-induced antinociception, tolerance and dependence: Studies using male and female TLR and signalling gene KO mice. Brain Behav Immun 2022; 102:71-85. [PMID: 35131445 DOI: 10.1016/j.bbi.2022.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/22/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Toll-like receptors (TLR) have been proposed as a site of action that alters opioid pharmacodynamics. However, a comprehensive assessment of acute opioid antinociception, tolerance and withdrawal behaviours in genetic null mutant strains with altered innate immune signalling has not been performed. Nor has the impact of genetic deletion of TLR2/4 on high-affinity opioid receptor binding. Here we show that diminished TLR signalling potentiates acute morphine antinociception equally in male and female mice. However, only male TIR8 null mutant mice showed reduced morphine analgesia. Analgesic tolerance was prevented in TLR2 and TLR4 null mutants, but not MyD88 animals. Withdrawal behaviours were only protected in TLR2-/- mice. In silico docking simulations revealed opioid ligands bound preferentially to the LPS binding pocket of MD-2 rather than TLR4. There was no binding of [3H](-)-naloxone or [3H]diprenorphine to TLR4 in the concentrations explored. These data confirm that opioids have high efficacy activity at innate immune pattern recognition binding sites but do not bind to TLR4 and identify critical pathway and sex-specific effects of the complex innate immune signalling contributions to opioid pharmacodynamics. These data further support the behavioural importance of the TLR-opioid interaction but fail to demonstrate direct evidence for high-affinity binding of the TLR4 signalling complex to ligands.
Collapse
Affiliation(s)
- Jacob H L Thomas
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Liang Lui
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew Abell
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia
| | - William Tieu
- Discipline of Chemistry, University of Adelaide, Adelaide, SA 5005, Australia
| | - Andrew A Somogyi
- Discipline of Pharmacology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Juliana E Bajic
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia
| | - Mark R Hutchinson
- Discipline of Physiology, Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia; ARC Centre for Nanoscale BioPhotonics, University of Adelaide, SA 5005, Australia.
| |
Collapse
|
7
|
Zhang K, Huang Q, Deng S, Yang Y, Li J, Wang S. Mechanisms of TLR4-Mediated Autophagy and Nitroxidative Stress. Front Cell Infect Microbiol 2021; 11:766590. [PMID: 34746034 PMCID: PMC8570305 DOI: 10.3389/fcimb.2021.766590] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/04/2021] [Indexed: 01/23/2023] Open
Abstract
Pathogenic infections have badly affected public health and the development of the breeding industry. Billions of dollars are spent every year fighting against these pathogens. The immune cells of a host produce reactive oxygen species and reactive nitrogen species which promote the clearance of these microbes. In addition, autophagy, which is considered an effective method to promote the destruction of pathogens, is involved in pathological processes. As research continues, the interplay between autophagy and nitroxidative stress has become apparent. Autophagy is always intertwined with nitroxidative stress. Autophagy regulates nitroxidative stress to maintain homeostasis within an appropriate range. Intracellular oxidation, in turn, is a strong inducer of autophagy. Toll-like receptor 4 (TLR4) is a pattern recognition receptor mainly involved in the regulation of inflammation during infectious diseases. Several studies have suggested that TLR4 is also a key regulator of autophagy and nitroxidative stress. In this review, we describe the role of TLR4 in autophagy and oxidation, and focus on its function in influencing autophagy-nitroxidative stress interactions.
Collapse
Affiliation(s)
- Kunli Zhang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Qiuyan Huang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Shoulong Deng
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Yecheng Yang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding/Guangdong Provincial Research Center of Gene Editing Engineering Technology, Foshan University, Foshan, China
| | - Jianhao Li
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Sutian Wang
- State Key Laboratory of Livestock and Poultry Breeding, Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
8
|
Cho S, Dong J, Lu LF. Cell-intrinsic and -extrinsic roles of miRNAs in regulating T cell immunity. Immunol Rev 2021; 304:126-140. [PMID: 34549446 DOI: 10.1111/imr.13029] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/10/2021] [Accepted: 09/11/2021] [Indexed: 12/14/2022]
Abstract
T cells are crucial to generate an effective response against numerous invading microbial pathogens and play a pivotal role in tumor surveillance and elimination. However, unwanted T cell activation can also lead to deleterious immune-mediated inflammation and tissue damage. To ensure that an optimal T cell response can be established, each step, beginning from T cell development in the thymus to their activation and function in the periphery, is tightly regulated by many transcription factors and epigenetic regulators including microRNAs (miRNAs). Here, we first summarize recent progress in identifying major immune regulatory miRNAs in controlling the differentiation and function of distinct T cell subsets. Moreover, as emerging evidence has demonstrated that miRNAs can impact T cell immunity through targeting both immune- and non-immune cell populations that T cells closely interact with, the T cell-extrinsic role of miRNAs in regulating different aspects of T cell biology is also addressed. Finally, we discuss the complex nature of miRNA-mediated control of T cell immunity and highlight important questions that remain to be further investigated.
Collapse
Affiliation(s)
- Sunglim Cho
- Division of Biological Sciences, University of California, La Jolla, California, USA
| | - Jiayi Dong
- Division of Biological Sciences, University of California, La Jolla, California, USA
| | - Li-Fan Lu
- Division of Biological Sciences, University of California, La Jolla, California, USA.,Moores Cancer Center, University of California, La Jolla, California, USA.,Center for Microbiome Innovation, University of California, La Jolla, California, USA
| |
Collapse
|
9
|
MicroRNA-155: Regulation of Immune Cells in Sepsis. Mediators Inflamm 2021; 2021:8874854. [PMID: 33505221 PMCID: PMC7810547 DOI: 10.1155/2021/8874854] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/15/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs are small noncoding RNAs which regulate gene expression at the posttranscriptional level. miR-155 is encoded by the miR-155 host gene (miR155HG), also known as the noncoding B cell integration cluster (BIC). MicroRNAs are widely expressed in various hematopoietic cells and are involved in regulating the immune system. In this review, we summarized how miR-155 modulates specific immune cells and the regulatory role of miR-155 in sepsis. miR-155 is expressed by different populations of innate and adaptive immune cells and is involved in the regulation of development, proliferation, and function in these cells. Sepsis is associated with uncontrollable inflammatory responses, accompanied by unacceptably high mortality. Due to the inadequacy of diagnostic markers as well as treatment strategies, treating sepsis can be a huge challenge. So far, a large number of experiments have shown that the expression of miR-155 is increased at an early stage of sepsis and that this increase is positively correlated with disease progression and severity. In addition, by blocking the proinflammatory effects of miR-155, it can effectively improve sepsis-related organ injury, providing novel insights to identify potential biomarkers and therapeutic targets for sepsis. However, since most of the current research is limited to animal experiments, further clinical research is required to determine the function of miR-155 and its mechanism related to sepsis.
Collapse
|
10
|
Kayhan M, Koyas A, Akdemir I, Savas AC, Cekic C. Adenosine Receptor Signaling Targets Both PKA and Epac Pathways to Polarize Dendritic Cells to a Suppressive Phenotype. THE JOURNAL OF IMMUNOLOGY 2019; 203:3247-3255. [PMID: 31722989 DOI: 10.4049/jimmunol.1900765] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 02/05/2023]
Abstract
Extracellular adenosine accumulates in tumors and causes suppression of immune cells. Suppressive adenosine signaling is achieved through adenosine A2A and A2B receptors, which are Gs coupled, and their activation elevates cAMP levels. Gs-coupled GPCR signaling causes cAMP accumulation, which plays an anti-inflammatory role in immune cells. Protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac) are two intracellular receptors of cAMP. In this study we showed that adenosine receptor signaling polarizes activated murine dendritic cells (DCs) into a tumor-promoting suppressive phenotype. Adenosine receptor signaling activates cAMP pathway and upregulates the negative regulators of NF-κB but does not influence phosphorylation of immediate inflammatory signaling molecules downstream of TLR signaling. Pharmacologic activation of both PKA and Epac pathways by specific cAMP analogues phenocopied the effects of adenosine signaling on murine DCs, such as suppression of proinflammatory cytokines, elevation of anti-inflammatory IL-10, increased expression of regulators of NF-κB pathway, and finally suppression of T cell activation. Inhibition of effector cytokine, IL-12p40 production, and increased immunosuppressive IL-10 production by adenosine signaling is significantly reversed only when both PKA and Epac pathways were inhibited together. Adenosine signaling increased IL-10 secretion while decreasing IL-12p40 secretion in human monocyte-derived DCs. Stimulation of both PKA and Epac pathways also caused combinatorial effects in regulation of IL-12p40 secretion in human monocyte-derived DCs. Interestingly, PKA signaling alone caused similar increase in IL-10 secretion to that of adenosine signaling in human monocyte-derived DCs. Our data suggest adenosine/cAMP signaling targets both PKA/Epac pathways to fully differentiate DCs into a suppressive phenotype.
Collapse
Affiliation(s)
- Merve Kayhan
- Department of Molecular Biology and Genetics, Bilkent University, 06800 Ankara, Turkey
| | - Altay Koyas
- Department of Molecular Biology and Genetics, Bilkent University, 06800 Ankara, Turkey
| | - Imran Akdemir
- Department of Molecular Biology and Genetics, Bilkent University, 06800 Ankara, Turkey
| | - Ali Can Savas
- Department of Molecular Biology and Genetics, Bilkent University, 06800 Ankara, Turkey
| | - Caglar Cekic
- Department of Molecular Biology and Genetics, Bilkent University, 06800 Ankara, Turkey
| |
Collapse
|
11
|
Mortazavi‐Jahromi SS, Aslani M, Omidian S, Ahmadzadeh A, Rezaieyazdi Z, Mirshafiey A. Immunopharmacological effect of β‐
d
‐mannuronic acid (M2000), as a new immunosuppressive drug, on gene expression of miR‐155 and its target molecules (SOCS1, SHIP1) in a clinical trial on rheumatoid arthritis patients. Drug Dev Res 2019; 81:295-304. [DOI: 10.1002/ddr.21619] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 10/02/2019] [Accepted: 10/11/2019] [Indexed: 12/26/2022]
Affiliation(s)
| | - Mona Aslani
- Department of Immunology, School of Public HealthTehran University of Medical Sciences Tehran Iran
| | - Saiedeh Omidian
- Department of Immunology, School of Public HealthTehran University of Medical Sciences Tehran Iran
| | - Arman Ahmadzadeh
- Department of Rheumatology, Loghman Hakim HospitalShahid Beheshti University of Medical Sciences Tehran Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research CenterMashhad University of Medical Sciences Mashhad Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public HealthTehran University of Medical Sciences Tehran Iran
| |
Collapse
|
12
|
Crowley T, Buckley CD, Clark AR. Stroma: the forgotten cells of innate immune memory. Clin Exp Immunol 2018; 193:24-36. [PMID: 29729109 PMCID: PMC6038004 DOI: 10.1111/cei.13149] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/01/2018] [Accepted: 05/01/2018] [Indexed: 12/20/2022] Open
Abstract
All organisms are exposed constantly to a variety of infectious and injurious stimuli. These induce inflammatory responses tailored to the threat posed. While the innate immune system is the front line of response to each stimulant, it has been considered traditionally to lack memory, acting in a generic fashion until the adaptive immune arm can take over. This outmoded simplification of the roles of innate and acquired arms of the immune system has been challenged by evidence of myeloid cells altering their response to subsequent encounters based on earlier exposure. This concept of 'innate immune memory' has been known for nearly a century, and is accepted among myeloid biologists. In recent years other innate immune cells, such as natural killer cells, have been shown to display memory, suggesting that innate immune memory is a trait common to several cell types. During the last 30 years, evidence has slowly accumulated in favour of not only haematopoietic cells, but also stromal cells, being imbued with memory following inflammatory episodes. A recent publication showing this also to be true in epithelial cells suggests innate immune memory to be widespread, if under-appreciated, in non-haematopoietic cells. In this review, we will examine the evidence supporting the existence of innate immune memory in stromal cells. We will also discuss the ramifications of memory in long-lived tissue-resident cells. Finally, we will pose questions we feel to be important in the understanding of these forgotten cells in the field of innate memory.
Collapse
Affiliation(s)
- T. Crowley
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirmingham, UK
| | - C. D. Buckley
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirmingham, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UKUniversity of OxfordOxfordUK
| | - A. R. Clark
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirmingham, UK
| |
Collapse
|
13
|
Chentouh R, Fitting C, Cavaillon JM. Specific features of human monocytes activation by monophosphoryl lipid A. Sci Rep 2018; 8:7096. [PMID: 29728623 PMCID: PMC5935727 DOI: 10.1038/s41598-018-25367-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/09/2018] [Indexed: 01/01/2023] Open
Abstract
We deciphered the mechanisms of production of pro- and anti-inflammatory cytokines by adherent human blood mononuclear cells (PBMC) activated by lipopolysaccharide (LPS) or monophosphoryl lipid A (MPLA). Both LPS and MPLA induced tumor necrosis factor (TNF) production proved to be dependent on the production of interleukin-1β (IL-1β). Of note, MPLA induced IL-1β release in human adherent PBMCs whereas MPLA was previously reported to not induce this cytokine in murine cells. Both LPS and MPLA stimulatory effects were inhibited by Toll-like receptor-4 (TLR4) antagonists. Only monocytes activation by LPS was dependent on CD14. Other differences were noticed between LPS and MPLA. Among the different donors, a strong correlation existed in terms of the levels of TNF induced by different LPSs. In contrast, there was no correlation between the TNF productions induced by LPS and those induced by MPLA. However, there was a strong correlation when IL-6 production was analyzed. Blocking actin polymerization and internalization of the agonists inhibited MPLA induced TNF production while the effect on LPS induced TNF production depended on the donors (i.e. high TNF producers versus low TNF producers). Finally, conventional LPS, tolerized adherent PBMCs to TLR2 agonists, while MPLA primed cells to further challenge with TLR2 agonists.
Collapse
Affiliation(s)
- Ryme Chentouh
- Unit "Cytokines & Inflammation", Institut Pasteur, Paris, France
| | | | | |
Collapse
|
14
|
Mann M, Mehta A, Zhao JL, Lee K, Marinov GK, Garcia-Flores Y, Lu LF, Rudensky AY, Baltimore D. An NF-κB-microRNA regulatory network tunes macrophage inflammatory responses. Nat Commun 2017; 8:851. [PMID: 29021573 PMCID: PMC5636846 DOI: 10.1038/s41467-017-00972-z] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 08/10/2017] [Indexed: 01/02/2023] Open
Abstract
The innate inflammatory response must be tightly regulated to ensure effective immune protection. NF-κB is a key mediator of the inflammatory response, and its dysregulation has been associated with immune-related malignancies. Here, we describe a miRNA-based regulatory network that enables precise NF-κB activity in mouse macrophages. Elevated miR-155 expression potentiates NF-κB activity in miR-146a-deficient mice, leading to both an overactive acute inflammatory response and chronic inflammation. Enforced miR-155 expression overrides miR-146a-mediated repression of NF-κB activation, thus emphasizing the dominant function of miR-155 in promoting inflammation. Moreover, miR-155-deficient macrophages exhibit a suboptimal inflammatory response when exposed to low levels of inflammatory stimuli. Importantly, we demonstrate a temporal asymmetry between miR-155 and miR-146a expression during macrophage activation, which creates a combined positive and negative feedback network controlling NF-κB activity. This miRNA-based regulatory network enables a robust yet time-limited inflammatory response essential for functional immunity. MicroRNAs (miR) are important regulators of gene transcription, with miR-155 and miR-146a both implicated in macrophage activation. Here the authors show that NF-κB signalling, miR-155 and miR-146a form a complex network of cross-regulations to control gene transcription in macrophages for modulating inflammatory responses.
Collapse
Affiliation(s)
- Mati Mann
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Arnav Mehta
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.,David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jimmy L Zhao
- Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medical College, 525 E 68th Street, New York, NY, 10065, USA.,Division of Hematology Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Kevin Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Georgi K Marinov
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yvette Garcia-Flores
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Li-Fan Lu
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, 92093, USA.,Moores Cancer Center, University of California, La Jolla, San Diego, CA, 92093, USA.,Center for Microbiome Innovation, University of California, La Jolla, San Diego, CA, 92093, USA
| | - Alexander Y Rudensky
- Howard Hughes Medical Institute and Immunology Program, Ludwig Center at Memorial Sloan-Kettering Cancer Center, Memorial Sloan-Kettering Cancer Center, New York, NY, 10065, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
15
|
Adjei-Fremah S, Ekwemalor K, Asiamah EK, Ismail H, Ibrahim S, Worku M. Effect of probiotic supplementation on growth and global gene expression in dairy cows. JOURNAL OF APPLIED ANIMAL RESEARCH 2017. [DOI: 10.1080/09712119.2017.1292913] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Sarah Adjei-Fremah
- Department of Energy and Environmental Systems, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Kingsley Ekwemalor
- Department of Energy and Environmental Systems, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Emmanuel K. Asiamah
- Department of Energy and Environmental Systems, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Hamid Ismail
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Salam Ibrahim
- Department of Family and Consumer Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Mulumebet Worku
- Department of Animal Sciences, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| |
Collapse
|
16
|
Yan S, Yim LY, Tam RCY, Chan A, Lu L, Lau CS, Chan VSF. MicroRNA-155 Mediates Augmented CD40 Expression in Bone Marrow Derived Plasmacytoid Dendritic Cells in Symptomatic Lupus-Prone NZB/W F1 Mice. Int J Mol Sci 2016; 17:ijms17081282. [PMID: 27509492 PMCID: PMC5000679 DOI: 10.3390/ijms17081282] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 07/23/2016] [Accepted: 08/02/2016] [Indexed: 12/18/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic multi-organ autoimmune disease characterized by hyperactivated immune responses to self-antigens and persistent systemic inflammation. Previously, we reported abnormalities in circulating and bone marrow (BM)-derived plasmacytoid dendritic cells (pDCs) from SLE patients. Here, we aim to seek for potential regulators that mediate functional aberrations of pDCs in SLE. BM-derived pDCs from NZB/W F1 mice before and after the disease onset were compared for toll-like receptor (TLR) induced responses and microRNA profile changes. While pDCs derived from symptomatic mice were phenotypically comparable to pre-symptomatic ones, functionally they exhibited hypersensitivity to TLR7 but not TLR9 stimulation, as represented by the elevated upregulation of CD40, CD86 and MHC class II molecules upon R837 stimulation. Upregulated induction of miR-155 in symptomatic pDCs following TLR7 stimulation was observed. Transfection of miR-155 mimics in pre-symptomatic pDCs induced an augmented expression of Cd40, which is consistent with the increased CD40 expression in symptomatic pDCs. Overall, our results provide evidence for miR-155-mediated regulation in pDC functional abnormalities in SLE. Findings from this study contribute to a better understanding of SLE pathogenesis and ignite future interests in evaluating the molecular regulation in autoimmunity.
Collapse
Affiliation(s)
- Sheng Yan
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Lok Yan Yim
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Rachel Chun Yee Tam
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Albert Chan
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Liwei Lu
- Departments of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Chak Sing Lau
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Vera Sau-Fong Chan
- Departments of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
17
|
Sag D, Cekic C, Wu R, Linden J, Hedrick CC. The cholesterol transporter ABCG1 links cholesterol homeostasis and tumour immunity. Nat Commun 2015; 6:6354. [PMID: 25724068 PMCID: PMC4347884 DOI: 10.1038/ncomms7354] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 01/22/2015] [Indexed: 02/07/2023] Open
Abstract
ATP-binding Cassette Transporter G1 (ABCG1) promotes cholesterol efflux from cells and regulates intracellular cholesterol homeostasis. Here, we demonstrate a role of ABCG1 as a mediator of tumor immunity. Abcg1−/− mice have dramatically suppressed subcutaneous MB49-bladder carcinoma and B16-melanoma growth and prolonged survival. We show that reduced tumor growth in Abcg1−/− mice is myeloid cell-intrinsic and is associated with a phenotypic shift of the macrophages from a tumor-promoting M2 to a tumor-fighting M1 within the tumor. Abcg1−/− macrophages exhibit an intrinsic bias toward M1 polarization with increased NF-κB activation and direct cytotoxicity for tumor cells in vitro. Overall, our study demonstrates that absence of ABCG1 inhibits tumor growth through modulation of macrophage function within the tumor and illustrates a link between cholesterol homeostasis and cancer.
Collapse
Affiliation(s)
- Duygu Sag
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Caglar Cekic
- Department of Molecular Biology and Genetics, Bilkent University, Ankara 06800, Turkey
| | - Runpei Wu
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| | - Catherine C Hedrick
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA
| |
Collapse
|
18
|
Jacquier V, Estellé J, Schmaltz-Panneau B, Lecardonnel J, Moroldo M, Lemonnier G, Turner-Maier J, Duranthon V, Oswald IP, Gidenne T, Rogel-Gaillard C. Genome-wide immunity studies in the rabbit: transcriptome variations in peripheral blood mononuclear cells after in vitro stimulation by LPS or PMA-Ionomycin. BMC Genomics 2015; 16:26. [PMID: 25613284 PMCID: PMC4326531 DOI: 10.1186/s12864-015-1218-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 01/02/2015] [Indexed: 12/13/2022] Open
Abstract
Background Our purpose was to obtain genome-wide expression data for the rabbit species on the responses of peripheral blood mononuclear cells (PBMCs) after in vitro stimulation by lipopolysaccharide (LPS) or phorbol myristate acetate (PMA) and ionomycin. This transcriptome profiling was carried out using microarrays enriched with immunity-related genes, and annotated with the most recent data available for the rabbit genome. Results The LPS affected 15 to 20 times fewer genes than PMA-Ionomycin after both 4 hours (T4) and 24 hours (T24), of in vitro stimulation, in comparison with mock-stimulated PBMCs. LPS induced an inflammatory response as shown by a significant up-regulation of IL12A and CXCL11 at T4, followed by an increased transcription of IL6, IL1B, IL1A, IL36, IL37, TNF, and CCL4 at T24. Surprisingly, we could not find an up-regulation of IL8 either at T4 or at T24, and detected a down-regulation of DEFB1 and BPI at T24. A concerted up-regulation of SAA1, S100A12 and F3 was found upon stimulation by LPS. PMA-Ionomycin induced a very early expression of Th1, Th2, Treg, and Th17 responses by PBMCs at T4. The Th1 response increased at T24 as shown by the increase of the transcription of IFNG and by contrast to other cytokines which significantly decreased from T4 to T24 (IL2, IL4, IL10, IL13, IL17A, CD69) by comparison to mock-stimulation. The granulocyte-macrophage colony-stimulating factor (CSF2) was by far the most over-expressed gene at both T4 and T24 by comparison to mock-stimulated cells, confirming a major impact of PMA-Ionomycin on cell growth and proliferation. A significant down-regulation of IL16 was observed at T4 and T24, in agreement with a role of IL16 in PBMC apoptosis. Conclusions We report new data on the responses of PBMCs to LPS and PMA-Ionomycin in the rabbit species, thus enlarging the set of mammalian species for which such reports exist. The availability of the rabbit genome assembly together with high throughput genomic tools should pave the way for more intense genomic studies for this species, which is known to be a very relevant biomedical model in immunology and physiology. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1218-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vincent Jacquier
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,INRA, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31326, Castanet-Tolosan, France. .,Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31326, Castanet-Tolosan, France. .,Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31076, Toulouse, France.
| | - Jordi Estellé
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | - Barbara Schmaltz-Panneau
- INRA, UMR1198 Biologie du Développement et Reproduction, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | - Jérôme Lecardonnel
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | - Marco Moroldo
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | - Gaëtan Lemonnier
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | | | - Véronique Duranthon
- INRA, UMR1198 Biologie du Développement et Reproduction, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| | - Isabelle P Oswald
- INRA, UMR1331, Toxalim, Research Center in Food Toxicology, 180 chemin de Tournefeuille, BP 93173, F-31027, Toulouse, France. .,Université de Toulouse, INP, UMR1331, Toxalim, Research Center in Food Toxicology, F-31000, Toulouse, France.
| | - Thierry Gidenne
- INRA, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31326, Castanet-Tolosan, France. .,Université de Toulouse, INP, ENSAT, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31326, Castanet-Tolosan, France. .,Université de Toulouse, INP, ENVT, GenPhySE (Génétique, Physiologie et Systèmes d'Elevage), F-31076, Toulouse, France.
| | - Claire Rogel-Gaillard
- INRA, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France. .,AgroParisTech, UMR 1313 Génétique Animale et Biologie Intégrative, Domaine de Vilvert, F-78350, Jouy-en-Josas, France.
| |
Collapse
|
19
|
Stark R, Choi H, Koch S, Lamb F, Sherwood E. Monophosphoryl lipid A inhibits the cytokine response of endothelial cells challenged with LPS. Innate Immun 2014; 21:565-74. [PMID: 25540284 DOI: 10.1177/1753425914564172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/15/2014] [Indexed: 01/06/2023] Open
Abstract
Monophosphoryl lipid A (MPLA) is a TLR4 agonist that is used as an immunomodulator in human vaccines; additionally, it has been shown to be protective in models of sepsis. As endothelial cells regulate inflammation, we hypothesized that MPLA would decrease activation of human umbilical vein endothelial cells (HUVECs) to LPS. We studied HUVECs challenged with LPS (100 ng/ml), MPLA (0.001-100 µg/ml) or a combination. Secretion of IL-6, RANTES (CCL5) and IP-10 (CXCL10) were assessed by ELISA. Activation of MAPK phosphorylation and cytokine transcription were assessed by Western blot analysis and PCR, respectively. MPLA alone was a weak stimulator of myeloid differentiation primary response protein 88-dependent IL-6 and did not induce TIR-domain-containing adapter-inducing IFN-β (TRIF)-dependent chemokine responses. MPLA significantly reduced LPS-mediated IL-6 production. This inhibitory effect was also conferred for the TRIF-dependent chemokines RANTES and IP-10. Inhibition of LPS-mediated activation by MPLA was associated with reduced p38 phosphorylation and mRNAs encoding inflammatory cytokines. MPLA inhibition of LPS signaling appeared to be at the level of the TLR4 receptor, acting as a receptor antagonist with weak agonistic properties. This study provides evidence of a novel mechanism for the inhibitory effect of MPLA on LPS-induced endothelial activation.
Collapse
Affiliation(s)
- Ryan Stark
- Department of Pediatric Critical Care, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hyehun Choi
- Department of Pediatric Critical Care, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Stephen Koch
- Department of Pediatric Critical Care, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Fred Lamb
- Department of Pediatric Critical Care, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Edward Sherwood
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
20
|
Cekic C, Day YJ, Sag D, Linden J. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res 2014; 74:7250-9. [PMID: 25377469 DOI: 10.1158/0008-5472.can-13-3583] [Citation(s) in RCA: 227] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
High concentrations of adenosine in tumor microenvironments inhibit antitumor cytotoxic lymphocyte responses. Although T cells express inhibitory adenosine A2A receptors (A2AR) that suppress their activation and inhibit immune killing of tumors, a role for myeloid cell A2ARs in suppressing the immune response to tumors has yet to be investigated. In this study, we show that the growth of transplanted syngeneic B16F10 melanoma or Lewis lung carcinoma cells is slowed in Adora2a(f/f)-LysMCre(+/-) mice, which selectively lack myeloid A2ARs. Reduced melanoma growth is associated with significant increases in MHCII and IL12 expression in tumor-associated macrophages and with >90% reductions in IL10 expression in tumor-associated macrophages, dendritic cells (DC), and Ly6C(+) or Ly6G(+) myeloid-derived suppressor cells (MDSC). Myeloid deletion of A2ARs significantly increases CD44 expression on tumor-associated T cells and natural killer (NK) cells. Depletion of CD8(+) T cells or NK cells in tumor-bearing mice indicates that both cell types initially contribute to slowing melanoma growth in mice lacking myeloid A2A receptors, but tumor suppression mediated by CD8(+) T cells is more persistent. Myeloid-selective A2AR deletion significantly reduces lung metastasis of melanomas that express luciferase (for in vivo tracking) and ovalbumin (as a model antigen). Reduced metastasis is associated with increased numbers and activation of NK cells and antigen-specific CD8(+) T cells in lung infiltrates. Overall, the findings indicate that myeloid cell A2ARs have direct myelosuppressive effects that indirectly contribute to the suppression of T cells and NK cells in primary and metastatic tumor microenvironments. The results indicate that tumor-associated myeloid cells, including macrophages, DCs, and MDSCs all express immunosuppressive A2ARs that are potential targets of adenosine receptor blockers to enhance immune killing of tumors.
Collapse
Affiliation(s)
- Caglar Cekic
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California. Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Yuan-Ji Day
- Department of Anesthesiology, Chang Gung Memorial Hospital, Institute of Clinical Medical Science, Chang Gung University, Tauyuan, Taiwan
| | - Duygu Sag
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California
| | - Joel Linden
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, California.
| |
Collapse
|
21
|
Lee SJ, Choi EK, Seo KW, Bae JU, Park SY, Kim CD. TLR4-mediated expression of Mac-1 in monocytes plays a pivotal role in monocyte adhesion to vascular endothelium. PLoS One 2014; 9:e104588. [PMID: 25116953 PMCID: PMC4130585 DOI: 10.1371/journal.pone.0104588] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/10/2014] [Indexed: 11/18/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is known to mediate monocyte adhesion to endothelial cells, however, its role on the expression of monocyte adhesion molecules is unclear. In the present study, we investigated the role of TLR4 on the expression of monocyte adhesion molecules, and determined the functional role of TLR4-induced adhesion molecules on monocyte adhesion to endothelial cells. When THP-1 monocytes were stimulated with Kdo2-Lipid A (KLA), a specific TLR4 agonist, Mac-1 expression was markedly increased in association with an increased adhesion of monocytes to endothelial cells. These were attenuated by anti-Mac-1 antibody, suggesting a functional role of TLR4-induced Mac-1 on monocyte adhesion to endothelial cells. In monocytes treated with MK886, a 5-lipoxygenase (LO) inhibitor, both Mac-1 expression and monocyte adhesion to endothelial cells induced by KLA were markedly attenuated. Moreover, KLA increased the expression of mRNA and protein of 5-LO, suggesting a pivotal role of 5-LO on these processes. In in vivo studies, KLA increased monocyte adhesion to aortic endothelium of wild-type (WT) mice, which was attenuated in WT mice treated with anti-Mac-1 antibody as well as in TLR4-deficient mice. Taken together, TLR4-mediated expression of Mac-1 in monocytes plays a pivotal role on monocyte adhesion to vascular endothelium, leading to increased foam cell formation in the development of atherosclerosis.
Collapse
Affiliation(s)
- Seung Jin Lee
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Eun Kyoung Choi
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Kyo Won Seo
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Ung Bae
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - So Youn Park
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, and MRC for Ischemic Tissue Regeneration, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
22
|
Ireton GC, Reed SG. Adjuvants containing natural and synthetic Toll-like receptor 4 ligands. Expert Rev Vaccines 2014; 12:793-807. [PMID: 23885824 DOI: 10.1586/14760584.2013.811204] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The last decade has seen an increased focus on the development of adjuvants for vaccines, and several novel adjuvants are now in licensed products or in late-stage clinical development. These advancements have been aided by the discovery of receptors and signaling pathways of the innate immune system and an increased understanding of how these innate responses influence the adaptive immune response. Successful vaccine development relies on knowledge of which adjuvants to use and the proper formulation of adjuvants and antigens to achieve safe, stable and immunogenic vaccines. In this review, the authors focus on the current use of natural and synthetic lipopolysaccharide analogues that retain their adjuvant properties with reduced toxicity compared with the parent compound for use in emerging vaccines. The authors review how these compounds initiate signal transduction through Toll-like receptor 4, insights from structure-function studies and how formulation parameters can influence their effectiveness as vaccine adjuvants.
Collapse
Affiliation(s)
- Gregory C Ireton
- Infectious Disease Research Institute, 1124 Columbia St., Ste 400, Seattle, WA 98104, USA
| | | |
Collapse
|
23
|
Darveau RP, Chilton PM. Naturally occurring low biological reactivity lipopolysaccharides as vaccine adjuvants. Expert Rev Vaccines 2014; 12:707-9. [DOI: 10.1586/14760584.2013.811181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
24
|
In vivo and in vitro characterization of the immune stimulating activity of the Neisserial porin PorB. PLoS One 2013; 8:e82171. [PMID: 24349212 PMCID: PMC3859593 DOI: 10.1371/journal.pone.0082171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/30/2013] [Indexed: 12/22/2022] Open
Abstract
Vaccines play a vital role in modern medicine. The development of novel vaccines for emerging and resistant pathogens has been aided in recent years by the use of novel adjuvants in subunit vaccines. A deeper understanding of the molecular pathways behind adjuvanticity is required to better select immunostimulatory molecules for use in individual vaccines. To this end, we have undertaken a study of the essential signaling pathways involved in the innate and adaptive immune responses to the Neisseria meningitidis outer membrane protein Porin B (PorB). We have previously demonstrated that PorB is an agonist of Toll-Like Receptor 2 (TLR2) and acts as an adjuvant in vaccines for protein, carbohydrate and lipopolysaccharide antigens using murine models. Here we demonstrate NFκB translocation following stimulation with PorB only occurs in the presence of TLR2. IL-6 and TNF-α secretion was shown to be MAPK dependent. Surface expression of activation markers on macrophages, including CD40, CD69, and CD86, was increased following PorB stimulation in vitro. Interestingly, some upregulation of CD54 and CD69 was still observed in macrophages obtained from TLR2 KO mice, indicating a possible non-TLR2 mediated activation pathway induced by PorB. In a murine vaccination model, using ovalbumin as the antigen and PorB as the adjuvant, a decreased antigen-specific IgG production was observed in TLR2 KO mice; adjuvant-dependent increased IgG production was entirely ablated in MyD88 KO mice. These observations demonstrate the importance of the above pathways to the adjuvant activity of PorB. The potential TLR2 independent effect is currently being explored.
Collapse
|
25
|
Cekic C, Sag D, Day YJ, Linden J. Extracellular adenosine regulates naive T cell development and peripheral maintenance. ACTA ACUST UNITED AC 2013; 210:2693-706. [PMID: 24145516 PMCID: PMC3832923 DOI: 10.1084/jem.20130249] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Adenosine receptor signaling inhibits TCR-induced activation of PI3K–Akt to reduce IL-7Rα expression on T cells, thereby regulating development and maintenance of naive T cells in the periphery. Adenosine produced as a byproduct of metabolic activity is present in all tissues and produces dose-dependent suppression of TCR signaling. Naive T cell maintenance depends on inhibition of TCR signals by environmental sensors, which are yet to be fully defined. We produced mice with a floxed adenosine A2A receptor (A2AR) gene, Adora2a, and show that either global A2AR deletion or cre-mediated T cell deletion elicits a decline in the number of naive but not memory T cells. A2AR signaling maintains naive T cells in a quiescent state by inhibiting TCR-induced activation of the phosphatidylinositide 3-kinase (PI3K)–AKT pathway, thereby reducing IL-7Rα down-regulation and naive T cell apoptosis. Patterns of IL-7Rα expression on T cells in chimeric mice reconstituted with Adora2a+/+ and Adora2a−/− bone marrow cells suggest that decreased IL-7Rα in naive T cells is a cell-intrinsic consequence of Adora2a deletion. In addition, A2AR expression increases in early thymic T cell development and contributes to progression of double-negative thymic precursors to single-positive thymocytes with increased IL-7Rα expression. Therefore, A2AR signaling regulates T cell development and maintenance to sustain normal numbers of naive T cells in the periphery.
Collapse
Affiliation(s)
- Caglar Cekic
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | | | | | | |
Collapse
|
26
|
Stenton GR, Mackenzie LF, Tam P, Cross JL, Harwig C, Raymond J, Toews J, Chernoff D, MacRury T, Szabo C. Characterization of AQX-1125, a small-molecule SHIP1 activator: Part 2. Efficacy studies in allergic and pulmonary inflammation models in vivo. Br J Pharmacol 2013; 168:1519-29. [PMID: 23121409 DOI: 10.1111/bph.12038] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 09/14/2012] [Accepted: 10/16/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The efficacy of AQX-1125, a small-molecule SH2-containing inositol-5'-phosphatase 1 (SHIP1) activator and clinical development candidate, is investigated in rodent models of inflammation. EXPERIMENTAL APPROACH AQX-1125 was administered orally in a mouse model of passive cutaneous anaphylaxis (PCA) and a number of rodent models of respiratory inflammation including: cigarette smoke, LPS and ovalbumin (OVA)-mediated airway inflammation. SHIP1 dependency of the AQX-1125 mechanism of action was investigated by comparing the efficacy in wild-type and SHIP1-deficient mice subjected to an intrapulmonary LPS challenge. RESULTS AQX-1125 exerted anti-inflammatory effects in all of the models studied. AQX-1125 decreased the PCA response at all doses tested. Using bronchoalveolar lavage (BAL) cell counts as an end point, oral or aerosolized AQX-1125 dose dependently decreased the LPS-mediated pulmonary neutrophilic infiltration at 3-30 mg kg⁻¹ and 0.15-15 μg kg⁻¹ respectively. AQX-1125 suppressed the OVA-mediated airway inflammation at 0.1-10 mg kg⁻¹. In the smoke-induced airway inflammation model, AQX-1125 was tested at 30 mg kg⁻¹ and significantly reduced the neutrophil infiltration of the BAL fluid. AQX-1125 (10 mg kg⁻¹) decreased LPS-induced pulmonary neutrophilia in wild-type mice but not in SHIP1-deficient mice. CONCLUSIONS The SHIP1 activator, AQX-1125, suppresses leukocyte accumulation and inflammatory mediator release in rodent models of pulmonary inflammation and allergy. As shown in the mouse model of LPS-induced lung inflammation, the efficacy of the compound is dependent on the presence of SHIP1. Pharmacological SHIP1 activation may have clinical potential for the treatment of pulmonary inflammatory diseases.
Collapse
|
27
|
Casella CR, Mitchell TC. Inefficient TLR4/MD-2 heterotetramerization by monophosphoryl lipid A. PLoS One 2013; 8:e62622. [PMID: 23638128 PMCID: PMC3637451 DOI: 10.1371/journal.pone.0062622] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 03/22/2013] [Indexed: 11/18/2022] Open
Abstract
Synthetic forms of E. coli monophosphoryl lipid A (sMLA) weakly activate the MyD88 (myeloid differentiation primary response protein) branch of the bifurcated TLR4 (Toll-like receptor 4) signaling pathway, in contrast to diphosphoryl lipid A (sDLA), which is a strong activator of both branches of TLR4. sMLA's weak MyD88 signaling activity is apparent downstream of TLR4/MyD88 signaling as we show that sMLA, unlike sDLA, is unable to efficiently recruit the TNF receptor-associated factor 6 (TRAF6) to the Interleukin-1 receptor-associated kinase 1 (IRAK1). This reduced recruitment of TRAF6 explains MLA's lower MAPK (Mitogen Activated Protein Kinase) and NF-κB activity. As further tests of sMLA's ability to activate TLR4/Myeloid differentiation factor 2 (MD-2), we used the antibody MTS510 as an indicator for TLR4/MD-2 heterotetramer formation. Staining patterns with this antibody indicated that sMLA does not effectively drive heterotetramerization of TLR4/MD-2 when compared to sDLA. However, a F126A mutant of MD-2, which allows lipid A binding but interferes with TLR4/MD-2 heterotetramerization, revealed that while sMLA is unable to efficiently form TLR4/MD-2 heterotetramers, it still needs heterotetramer formation for the full extent of signaling it is able to achieve. Monophosphoryl lipid A's weak ability to form TLR4/MD-2 heterotetramers was not restricted to synthetic E. coli type because cells exposed to a biological preparation of S. minnesota monophosphoryl lipid A (MPLA) also showed reduced TLR4/MD-2 heterotetramer formation. The low potency with which sMLA and MPLA drive heterotetramerization of TLR4/MD-2 contributes to their weak MyD88 signaling activities.
Collapse
Affiliation(s)
- Carolyn R. Casella
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
| | - Thomas C. Mitchell
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
28
|
Abstract
Phosphoinositide signalling molecules interact with a plethora of effector proteins to regulate cell proliferation and survival, vesicular trafficking, metabolism, actin dynamics and many other cellular functions. The generation of specific phosphoinositide species is achieved by the activity of phosphoinositide kinases and phosphatases, which phosphorylate and dephosphorylate, respectively, the inositol headgroup of phosphoinositide molecules. The phosphoinositide phosphatases can be classified as 3-, 4- and 5-phosphatases based on their specificity for dephosphorylating phosphates from specific positions on the inositol head group. The SAC phosphatases show less specificity for the position of the phosphate on the inositol ring. The phosphoinositide phosphatases regulate PI3K/Akt signalling, insulin signalling, endocytosis, vesicle trafficking, cell migration, proliferation and apoptosis. Mouse knockout models of several of the phosphoinositide phosphatases have revealed significant physiological roles for these enzymes, including the regulation of embryonic development, fertility, neurological function, the immune system and insulin sensitivity. Importantly, several phosphoinositide phosphatases have been directly associated with a range of human diseases. Genetic mutations in the 5-phosphatase INPP5E are causative of the ciliopathy syndromes Joubert and MORM, and mutations in the 5-phosphatase OCRL result in Lowe's syndrome and Dent 2 disease. Additionally, polymorphisms in the 5-phosphatase SHIP2 confer diabetes susceptibility in specific populations, whereas reduced protein expression of SHIP1 is reported in several human leukaemias. The 4-phosphatase, INPP4B, has recently been identified as a tumour suppressor in human breast and prostate cancer. Mutations in one SAC phosphatase, SAC3/FIG4, results in the degenerative neuropathy, Charcot-Marie-Tooth disease. Indeed, an understanding of the precise functions of phosphoinositide phosphatases is not only important in the context of normal human physiology, but to reveal the mechanisms by which these enzyme families are implicated in an increasing repertoire of human diseases.
Collapse
|
29
|
Sharmin T, Satho T, Irie K, Watanabe M, Hosokawa M, Hiramatsu Y, Talukder P, Okuno T, Tsuruda S, Uyeda S, Fukmits Y, Tamura Y, Nakashima Y, Imoto M, Toda A, Kashige N, Miake F. Pyriproxyfen enhances the immunoglobulin G immune response in mice. Microbiol Immunol 2013; 57:316-22. [DOI: 10.1111/1348-0421.12035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 11/22/2012] [Accepted: 01/09/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Tanjina Sharmin
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Tomomitsu Satho
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Keiichi Irie
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Mineo Watanabe
- Graduate School of Infection Control Sciences, Kitasato University; 5-9-1 Shirokane, Minato-ku; Tokyo; 108-8641; Japan
| | - Masato Hosokawa
- Department of Dementia and Higher Brain Function; Tokyo Metropolitan Institute of Medical Science; 2-1-6, Kamikitazawa, Setagaya-ku; Tokyo; 156-8506; Japan
| | - Yukihiro Hiramatsu
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Parimal Talukder
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Takahiro Okuno
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Shodai Tsuruda
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Saori Uyeda
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Yuki Fukmits
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Yukie Tamura
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Yukihiko Nakashima
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Masumi Imoto
- Daiichi College of Pharmaceutical Sciences; 22-1 Tamagawa-cho, Minami-Ku; Fukuoka; 815-8511; Japan
| | - Akihisa Toda
- Daiichi College of Pharmaceutical Sciences; 22-1 Tamagawa-cho, Minami-Ku; Fukuoka; 815-8511; Japan
| | - Nobuhiro Kashige
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| | - Fumio Miake
- Faculty of Pharmaceutical Sciences; Fukuoka University; 8-19-1, Nanakuma, Jonan-ku; Fukuoka; 814-0180; Japan
| |
Collapse
|
30
|
Bowen WS, Gandhapudi SK, Kolb JP, Mitchell TC. Immunopharmacology of Lipid A Mimetics. ADVANCES IN PHARMACOLOGY 2013; 66:81-128. [DOI: 10.1016/b978-0-12-404717-4.00003-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Tu J, Xing Y, Guo Y, Tang F, Guo L, Xi T. TanshinoneIIA ameliorates inflammatory microenvironment of colon cancer cells via repression of microRNA-155. Int Immunopharmacol 2012; 14:353-61. [DOI: 10.1016/j.intimp.2012.08.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 07/25/2012] [Accepted: 08/14/2012] [Indexed: 11/25/2022]
|
32
|
Chilton PM, Embry CA, Mitchell TC. Effects of Differences in Lipid A Structure on TLR4 Pro-Inflammatory Signaling and Inflammasome Activation. Front Immunol 2012; 3:154. [PMID: 22707952 PMCID: PMC3374416 DOI: 10.3389/fimmu.2012.00154] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 05/25/2012] [Indexed: 12/22/2022] Open
Abstract
The vertebrate immune system exists in equilibrium with the microbial world. The innate immune system recognizes pathogen-associated molecular patterns via a family of Toll-like receptors (TLR) that activate cells upon detection of potential pathogens. Because some microbes benefit their hosts, mobilizing the appropriate response, and then controlling that response is critical in the maintenance of health. TLR4 recognizes the various forms of lipid A produced by Gram-negative bacteria. Depending on the structural form of the eliciting lipid A molecule, TLR4 responses range from a highly inflammatory endotoxic response involving inflammasome and other pro-inflammatory mediators, to an inhibitory, protective response. Mounting the correct response against an offending microbe is key to maintaining health when exposed to various bacterial species. Further study of lipid A variants may pave the way to understanding how TLR4 responses are generally able to avoid chronic inflammatory damage.
Collapse
Affiliation(s)
- Paula M Chilton
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, School of Medicine, University of Louisville Louisville, KY, USA
| | | | | |
Collapse
|
33
|
Busch M, Zernecke A. microRNAs in the regulation of dendritic cell functions in inflammation and atherosclerosis. J Mol Med (Berl) 2012; 90:877-85. [PMID: 22307520 DOI: 10.1007/s00109-012-0864-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/14/2012] [Accepted: 01/16/2012] [Indexed: 12/18/2022]
Abstract
Atherosclerosis has been established as a chronic inflammatory disease of the vessel wall. Among the mononuclear cell types recruited to the lesions, specialized dendritic cells (DCs) have gained increasing attention, and their secretory products and interactions shape the progression of atherosclerotic plaques. The regulation of DC functions by microRNAs (miRNAs) may thus be of primary importance in disease. We here systematically summarize the biogenesis and functions of miRNAs and provide an overview of miRNAs in DCs, their targets, and potential implications for atherosclerosis, with a particular focus on the best characterized miRNAs in DCs, namely, miR-155 and miR-146. MiRNA functions in DCs range from regulation of lipid uptake to cytokine production and T cell responses with a complex picture emerging, in which miRNAs cooperate or antagonize DC behavior, thereby promoting or counterbalancing inflammatory responses. As miRNAs regulate key functions of DCs known to control atherosclerotic vascular disease, their potential as a therapeutic target holds promise and should be attended to in future research.
Collapse
Affiliation(s)
- Martin Busch
- Rudolf-Virchow-Center/DFG Research Center for Experimental Biomedicine, University of Würzburg, Josef-Schneider Str. 2, Haus D15, 97080 Würzburg, Germany
| | | |
Collapse
|