51
|
Ghaffari S, Roshanravan N, Tutunchi H, Ostadrahimi A, Pouraghaei M, Kafil B. Oleoylethanolamide, A Bioactive Lipid Amide, as A Promising Treatment Strategy for Coronavirus/COVID-19. Arch Med Res 2020; 51:464-467. [PMID: 32327293 PMCID: PMC7158763 DOI: 10.1016/j.arcmed.2020.04.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 12/12/2022]
Abstract
The current outbreak of COVID-19 (coronavirus) has been identified by World Health Organization (WHO) as a global pandemic. With the emergence of the COVID-19 virus and considering the lack of effective pharmaceutical treatment for it, there is an urgent need to identify safe and effective drugs or potential adjuvant therapy in this regard. Bioactive lipids with an array of known health-promoting properties can be suggested as effective agents in alleviating acute respiratory stress induced by virus. The bioactive lipid amide, oleoylethanolamide (OEA), due to several distinctive homeostatic properties, including anti-inflammatory activities, modulation of immune response, and anti-oxidant effects can be considered as a novel potential pharmacological alternative for the management of COVID-19.
Collapse
Affiliation(s)
- Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Helda Tutunchi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboub Pouraghaei
- Emergency Medicine Research Team, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnam Kafil
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
52
|
Ge L, Xu L, Lu S, Yan H. Expression and Function of Toll-Like Receptor 10 (TLR10) in Diffuse Large B Cell Lymphoma, Acute Myeloid Leukemia, and Glioma. Med Sci Monit 2020; 26:e921500. [PMID: 32287174 PMCID: PMC7174897 DOI: 10.12659/msm.921500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Toll-like receptor (TLR) family members are part of the major pathogen-recognition system for innate immunity. TLR10, the only remaining orphan receptor with an unknown ligand, has been poorly studied in tumors, and its functional and clinical relevance are unclear. Material/Methods We analyzed TLR10 expression data in The Cancer Genome Atlas (TCGA) by established computational approaches (UALCAN, GEPIA, CGGA, and TIMER) and confirmed them by immunohistochemistry analysis. Results Bioinformatics analysis showed that TLR10 was most highly expressed in diffuse large B cell lymphoma (DLBC), acute myeloid leukemia (LAML), and glioblastoma multiforme (GBM) patients. A data-mining study also revealed that TLR10 levels were positively correlated with WHO grade in glioma, and patients with high TLR10 levels showed shorter overall survival (OS) and disease-free survival (DFS) times than patients with low TLR10 levels. TISIDB and TIMER data showed that TLR10 expression was significantly positively correlated with immune infiltrates, especially infiltrating levels of B cells. Importantly, immunohistochemistry analysis revealed that TLR10 expression was a potential biomarker for distinguishing CNS-DLBC (also known as primary central nervous system lymphoma, PCNSL) from GBM. Conclusions Taken together, these results suggest that TLR10 could serve as a promising theranostic target for patients with glioma and is a potential biomarker for distinguishing PCNSL from GBM.
Collapse
Affiliation(s)
- Le Ge
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Lixia Xu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Shan Lu
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| | - Hua Yan
- Tianjin Neurosurgical Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China (mainland)
| |
Collapse
|
53
|
Xiang Q, Zhu L, Zheng K, Ding Y, Chen N, Liu G, He Q. Association of toll-like receptor 10 polymorphisms with pediatric pneumococcal meningitis. APMIS 2020; 128:335-342. [PMID: 31976578 DOI: 10.1111/apm.13028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/16/2020] [Indexed: 01/26/2023]
Abstract
We aimed to investigate whether the gene polymorphisms of TLR10 were associated with risk and severity of pneumococcal meningitis (PM) and serum cytokine levels in children. Three single nucleotide polymorphisms (SNPs) of TLR10 rs4129009 (2676A > G), rs10004195 (1018T > A) and rs11466617 (40735A > G) were studied in 95 laboratory-confirmed PM pediatric patients and 330 healthy controls by PCR-based sequencing. Ten serum cytokines were determined by multiplex immunoassay. The frequency of variant haplotype GAG of TLR10 was significantly lower in patients than controls (11.3% vs 33.3%, p < 0.001), although frequencies of the genotypes and alleles of the three SNPs did not differ between patients and controls. Frequency of variant haplotype GAG was significantly lower in patients who had CSF protein >1000 mg/L than those who had CSF protein ≤1000 mg/L (3.50% vs 32.4%, p < 0.001). Moreover, higher frequency of the haplotype GAG was found in patients who had higher levels of inflammatory cytokines such as IFN-γ, TNF-α and IL-1β. Our finding suggested that the variant haplotype GAG in TLR10 is associated with decreased risk of PM in Chinese children.
Collapse
Affiliation(s)
- Qiaoyan Xiang
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Liang Zhu
- Department of Infectious Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Kai Zheng
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Yiwei Ding
- Department of Medical Microbiology, Capital Medical University, Beijing, China.,Department of Laboratory Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ning Chen
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Gang Liu
- Department of Infectious Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Qiushui He
- Department of Medical Microbiology, Capital Medical University, Beijing, China.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| |
Collapse
|
54
|
Single Nucleotide Polymorphisms in TLR4 Affect Susceptibility to Tuberculosis in Mexican Population from the State of Veracruz. J Immunol Res 2020; 2020:2965697. [PMID: 32411792 PMCID: PMC7204096 DOI: 10.1155/2020/2965697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/12/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
Tuberculosis is still a global public health problem, with an estimated 10 million new cases and 1.6 million deaths in 2017. Of all humans infected with M. tuberculosis, only 10-15% will develop active tuberculosis disease during their lifetime, and data suggest that along with environmental factors, genetic factors influence susceptibility to develop active disease. Toll-like receptors (TLRs) are pattern recognition receptors that play a central role in the initiation and shaping of adaptive immune responses, and several TLRs have been shown to recognize mycobacterial components. In this work, we performed a case-control study to determine if common single nucleotide polymorphisms (SNPs) in genes encoding TLRs 1, 2, 4, 6, and 10 are associated with susceptibility to develop active tuberculosis in population from the state of Veracruz, Mexico. The study included 279 cases and 569 controls. The results show that the frequency of two SNPs in TLR4 was significantly higher in controls than in tuberculosis patients. The minor allele (G) of rs4986790 in TLR4 (D299G) decreased the risk of active tuberculosis in the allelic (A vs. G, OR = 0.31, 95%CI = 0.09‐0.81, p = 0.01) and in the dominant genetic model (AA vs. GG+AG, OR = 0.26, 95%CI = 0.09‐0.77, p = 0.02). Similarly, the minor allele (T) of rs4986791 in TLR4 (T399I) decreased the risk of active disease in the allelic model (C vs. T, OR = 0.29, 95%CI = 0.10‐0.90, p = 0.03). We did not find an association of SNPs in TLR1 (N248S), TLR2 (R753Q), TLR6 (S249P), and TLR10 (A153S and V298I) with tuberculosis disease. These results suggest that in this population, genetic variants of TLR4 affect the susceptibility for suffering active tuberculosis disease.
Collapse
|
55
|
Mourits VP, Arts RJW, Novakovic B, Matzaraki V, de Bree LCJ, Koeken VACM, Moorlag SJCFM, van Puffelen JH, Groh L, van der Heijden CDCC, Keating ST, Netea MG, Oosting M, Joosten LAB. The role of Toll-like receptor 10 in modulation of trained immunity. Immunology 2019; 159:289-297. [PMID: 31671203 PMCID: PMC7011636 DOI: 10.1111/imm.13145] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/30/2019] [Accepted: 10/28/2019] [Indexed: 02/06/2023] Open
Abstract
Toll‐like receptor 10 (TLR10) is the only member of the human Toll‐like receptor family with an inhibitory function on the induction of innate immune responses and inflammation. However, its role in the modulation of trained immunity (innate immune memory) is unknown. In the present study, we assessed whether TLR10 modulates the induction of trained immunity induced by β‐glucan or bacillus Calmette–Guérin (BCG). Interleukin 10 receptor antagonist production was increased upon activation of TLR10 ex vivo after BCG vaccination, and TLR10 protein expression on monocytes was increased after BCG vaccination, whereas anti‐TLR10 antibodies did not significantly modulate β‐glucan or BCG‐induced trained immunity in vitro. A known immunomodulatory TLR10 missense single‐nucleotide polymorphism (rs11096957) influenced trained immunity responses by β‐glucan or BCG in vitro. However, the in vivo induction of trained immunity by BCG vaccination was not influenced by TLR10 polymorphisms. In conclusion, TLR10 has a limited, non‐essential impact on the induction of trained immunity in humans.
Collapse
Affiliation(s)
- Vera P Mourits
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rob J W Arts
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Boris Novakovic
- Epigenetics Research, Murdoch Children's Research Institute, Parkville, Vic., Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - Vasiliki Matzaraki
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - L Charlotte J de Bree
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark.,Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Valerie A C M Koeken
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Simone J C F M Moorlag
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jelmer H van Puffelen
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Health Evidence, Radboud University Medical Center, Radboud Institute for Health Sciences (RIHS), Nijmegen, The Netherlands
| | - Laszlo Groh
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charlotte D C C van der Heijden
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sam T Keating
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Marije Oosting
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
56
|
Bou Karroum N, Moarbess G, Guichou JF, Bonnet PA, Patinote C, Bouharoun-Tayoun H, Chamat S, Cuq P, Diab-Assaf M, Kassab I, Deleuze-Masquefa C. Novel and Selective TLR7 Antagonists among the Imidazo[1,2- a]pyrazines, Imidazo[1,5- a]quinoxalines, and Pyrazolo[1,5- a]quinoxalines Series. J Med Chem 2019; 62:7015-7031. [PMID: 31283223 DOI: 10.1021/acs.jmedchem.9b00411] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The Toll-like receptors (TLRs) 7 and 8 play an important role in the immune system activation, and their agonists may therefore serve as promising candidate vaccine adjuvants. However, the chronic immune activation by excessive TLR stimulation is a hallmark of several clinically important infectious and autoimmune diseases, which warrants the search for TLR antagonists. In this study, we have synthesized and characterized a variety of compounds belonging to three heterocyclic chemical series: imidazo[1,2-a]pyrazine, imidazo[1,5-a]quinoxaline, and pyrazolo[1,5-a]quinoxaline. These compounds have been tested for their TLR7 or TLR8 agonistic and antagonistic activities. Several of them are shown to be selective TLR7 antagonists without any TLR7 or TLR8 agonistic activity. The selectivity was confirmed by a comparative ligand-docking study in TLR7 antagonist pocket. Two compounds of the pyrazolo[1,5-a]quinoxaline series (10a and 10b) are potent selective TLR7 antagonists and may be considered as promising starting points for the development of new therapeutic agents.
Collapse
Affiliation(s)
- Nour Bou Karroum
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, CNRS, Université de Montpellier , Faculté de Pharmacie , 15 avenue Charles Flahault , BP 14491, Montpellier 34093 Cedex 5 , France.,Tumorigenèse et Pharmacologie Antitumorale , Lebanese University, EDST , BP 90656, Fanar Jdeideh 1202 , Lebanon
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale , Lebanese University, EDST , BP 90656, Fanar Jdeideh 1202 , Lebanon
| | - Jean-François Guichou
- CNRS, UMR 5048, INSERM, U105, Université de Montpellier, Centre de Biochimie Structurale , Montpellier F-34090 , France
| | - Pierre-Antoine Bonnet
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, CNRS, Université de Montpellier , Faculté de Pharmacie , 15 avenue Charles Flahault , BP 14491, Montpellier 34093 Cedex 5 , France
| | - Cindy Patinote
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, CNRS, Université de Montpellier , Faculté de Pharmacie , 15 avenue Charles Flahault , BP 14491, Montpellier 34093 Cedex 5 , France
| | - Hasnaa Bouharoun-Tayoun
- Laboratory of Immunology and Vector-Borne Diseases, Faculty of Public Health , Lebanese University , Fanar Jdeideh 1202 , Lebanon
| | - Soulaima Chamat
- Laboratory of Immunology and Vector-Borne Diseases, Faculty of Public Health , Lebanese University , Fanar Jdeideh 1202 , Lebanon.,Faculty of Medical Sciences , Lebanese University , Hadath 1500 , Lebanon
| | - Pierre Cuq
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, CNRS, Université de Montpellier , Faculté de Pharmacie , 15 avenue Charles Flahault , BP 14491, Montpellier 34093 Cedex 5 , France
| | - Mona Diab-Assaf
- Tumorigenèse et Pharmacologie Antitumorale , Lebanese University, EDST , BP 90656, Fanar Jdeideh 1202 , Lebanon
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale , Lebanese University, EDST , BP 90656, Fanar Jdeideh 1202 , Lebanon
| | - Carine Deleuze-Masquefa
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 F16, CNRS, Université de Montpellier , Faculté de Pharmacie , 15 avenue Charles Flahault , BP 14491, Montpellier 34093 Cedex 5 , France
| |
Collapse
|
57
|
Seo SU, Kweon MN. Virome-host interactions in intestinal health and disease. Curr Opin Virol 2019; 37:63-71. [PMID: 31295677 DOI: 10.1016/j.coviro.2019.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023]
Abstract
The enteric virome consists largely of bacteriophages and prophages related to commensal bacteria. Bacteriophages indirectly affect the host immune system by targeting their associated bacteria; however, studies suggest that bacteriophages also have distinct pathways that enable them to interact directly with the host. Eukaryotic viruses are less abundant than bacteriophages but are more efficient in the stimulation of host immune responses. Acute, permanent, and latent viral infections are detected by different types of pattern recognition receptors and induce host immune responses, including the antiviral type I interferon response. Understanding the complex interplay between commensal microorganisms and the host immune system is a prerequisite to elucidating their role in intestinal diseases.
Collapse
Affiliation(s)
- Sang-Uk Seo
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul 05505, South Korea.
| | - Mi-Na Kweon
- Mucosal Immunology Laboratory, Department of Convergence Medicine, University of Ulsan College of Medicine/Asan Medical Center, Seoul 05505, South Korea.
| |
Collapse
|
58
|
Besednova N, Zaporozhets T, Kuznetsova T, Makarenkova I, Fedyanina L, Kryzhanovsky S, Malyarenko O, Ermakova S. Metabolites of Seaweeds as Potential Agents for the Prevention and Therapy of Influenza Infection. Mar Drugs 2019; 17:E373. [PMID: 31234532 PMCID: PMC6627559 DOI: 10.3390/md17060373] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023] Open
Abstract
CONTEXT Seaweed metabolites (fucoidans, carrageenans, ulvans, lectins, and polyphenols) are biologically active compounds that target proteins or genes of the influenza virus and host components that are necessary for replication and reproduction of the virus. OBJECTIVE This review gathers the information available in the literature regarding to the useful properties of seaweeds metabolites as potential agents for the prevention and therapy of influenza infection. MATERIALS AND METHODS The sources of scientific literature were found in various electronic databases (i.e., PubMed, Web of Science, and ScienceDirect) and library search. The retrospective search depth is 25 years. RESULTS Influenza is a serious medical and social problem for humanity. Recently developed drugs are quite effective against currently circulating influenza virus strains, but their use can lead to the selection of resistant viral strains. In this regard, new therapeutic approaches and drugs with a broad spectrum of activity are needed. Metabolites of seaweeds fulfill these requirements. This review presents the results of in vitro and in vivo experimental and clinical studies about the effectiveness of these compounds in combating influenza infection and explains the necessity of their use as a potential basis for the creation of new drugs with a broad spectrum of activity.
Collapse
Affiliation(s)
- Natalia Besednova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, Vladivostok 690087, Russia.
| | - Tatiana Zaporozhets
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, Vladivostok 690087, Russia.
| | - Tatiana Kuznetsova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, Vladivostok 690087, Russia.
| | - Ilona Makarenkova
- Federal State Budgetary Scientific Institution, Somov Research Institute of Epidemiology and Microbiology, Sel'skaya street, 1, Vladivostok 690087, Russia.
| | - Lydmila Fedyanina
- Far Eastern Federal University, School of Biomedicine, bldg. M25 FEFU Campus, Ajax Bay, Russky Isl., Vladivostok 690922, Russia.
| | - Sergey Kryzhanovsky
- Far Eastern Federal University, School of Biomedicine, bldg. M25 FEFU Campus, Ajax Bay, Russky Isl., Vladivostok 690922, Russia.
| | - Olesya Malyarenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Pr. 100-letiya Vladivostoka, 159, Vladivostok 690022, Russia.
| | - Svetlana Ermakova
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, Pr. 100-letiya Vladivostoka, 159, Vladivostok 690022, Russia.
| |
Collapse
|
59
|
Patra MC, Shah M, Choi S. Toll-like receptor-induced cytokines as immunotherapeutic targets in cancers and autoimmune diseases. Semin Cancer Biol 2019; 64:61-82. [PMID: 31054927 DOI: 10.1016/j.semcancer.2019.05.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/27/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
Abstract
Immune cells of the myeloid and lymphoid lineages express Toll-like receptors (TLRs) to recognize pathogenic components or cellular debris and activate the immune system through the secretion of cytokines. Cytokines are signaling molecules that are structurally and functionally distinct from one another, although their secretion profiles and signaling cascades often overlap. This situation gives rise to pleiotropic cell-to-cell communication pathways essential for protection from infections as well as cancers. Nonetheless, deregulated signaling can have detrimental effects on the host, in the form of inflammatory or autoimmune diseases. Because cytokines are associated with numerous autoimmune and cancerous conditions, therapeutic strategies to modulate these molecules or their biological responses have been immensely beneficial over the years. There are still challenges in the regulation of cytokine function in patients, even in those who take approved biological therapeutics. In this review, our purpose is to discuss the differential expression patterns of TLR-regulated cytokines and their cell type specificity that is associated with cancers and immune-system-related diseases. In addition, we highlight key structural features and molecular recognition of cytokines by receptors; these data have facilitated the development and approval of several biologics for the treatment of autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Mahesh Chandra Patra
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea.
| |
Collapse
|
60
|
Henrick BM, Yao XD, Zahoor MA, Abimiku A, Osawe S, Rosenthal KL. TLR10 Senses HIV-1 Proteins and Significantly Enhances HIV-1 Infection. Front Immunol 2019; 10:482. [PMID: 30930906 PMCID: PMC6430187 DOI: 10.3389/fimmu.2019.00482] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/22/2019] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptors (TLRs) play a crucial role in innate immunity and provide a first line of host defense against invading pathogens. Of the identified human TLRs, TLR10 remains an orphan receptor whose ligands and functions are poorly understood. In the present study, we sought to evaluate the level of TLR10 expression in breast milk (BM) and explore its potential function in the context of HIV-1 infection. We evaluated HIV-1-infected (Nigerian: n = 40) and uninfected (Nigerian: n = 27; Canadian: n = 15) BM samples for TLR expression (i.e., TLR10, TLR2, and TLR1) and report here that HIV-1-infected BM from Nigerian women showed significantly higher levels of TLR10, TLR1, and TLR2 expression. Moreover, the level of TLR10 expression in HIV-1-infected BM was upregulated by over 100-fold compared to that from uninfected control women. In vitro studies using TZMbl cells demonstrated that TLR10 overexpression contributes to higher HIV-1 infection and proviral DNA integration. Conversely, TLR10 inhibition significantly decreased HIV-1 infection. Notably, HIV-1 gp41 was recognized as a TLR10 ligand, leading to the induction of IL-8 and NF-κBα activation. The identification of a TLR10 ligand and its involvement in HIV-1 infection enhances our current understanding of HIV-1 replication and may assist in the development of improved future therapeutic strategies.
Collapse
Affiliation(s)
- Bethany M Henrick
- Evolve Biosystems, Davis, CA, United States.,Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Xiao-Dan Yao
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Muhammad Atif Zahoor
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Sophia Osawe
- Institue of Human Virology-Nigeria, Abuja, Nigeria
| | - Kenneth L Rosenthal
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
61
|
Sun Y, Su J, Yang S, Liu Z, Liu D, Gan F, Chen X, Huang K. Mannan Oligosaccharide Protects against the Aflatoxin-B 1-Promoted Influenza Replication and Tissue Damages in a Toll-Like-Receptor-4-Dependent Manner. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:735-745. [PMID: 30586993 DOI: 10.1021/acs.jafc.8b05829] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Our previous study reported that aflatoxin B1 (AFB1) promoted influenza replication. Mannan oligosaccharide (MOS), derived from the cell walls of yeast, is a potent immunomodulator. Here, we investigated the role of MOS in AFB1-promoted influenza replication and further explored the underlying mechanisms. In vitro and in vivo, the exposure to AFB1 alone resulted in significantly decreased weight gain and increased viral replication as well as lung and spleen damages. Increased influenza replication coupled with increases in toll-like receptor 4 (TLR4), phosphorylated nuclear factor κB, and tumor necrosis factor α (TNF-α) levels. However, MOS given in conjunction with exposure to AFB1 significantly reversed these above changes. A further study indicated that MOS activity was abolished by TLR4 knockout or TLR4 overexpression. Surprisingly, TNF-α played no role in the MOS-mediated protective effects. Collectively, our data suggest that MOS alleviates the AFB1-promoted influenza replication, inflammation, and tissue damages in a TLR4-dependent manner.
Collapse
|
62
|
Nagashima H, Yamaoka Y. Importance of Toll-like Receptors in Pro-inflammatory and Anti-inflammatory Responses by Helicobacter pylori Infection. Curr Top Microbiol Immunol 2019; 421:139-158. [PMID: 31123888 DOI: 10.1007/978-3-030-15138-6_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Infectious diseases have been paramount among the threats to human health and survival throughout evolutionary history. Bacterial cell-surface molecules are key factors in the microorganism-host crosstalk, as they can interact with host pattern-recognition receptors (PRRs) of the gastrointestinal mucosa. The best-studied PRRs are toll-like receptors (TLRs). Because TLRs play an important key role in host defense, they have received increasing interest in the evolutionary and population genetics literature, and their variation represents a potential target of adaptive evolution. Helicobacter pylori is one of the commensal bacteria in our body and can have pathogenic properties in a subset of infected people. The history of H. pylori research indicated that humans and bacteria co-evolved during evolution. A genome-wide association study (GWAS) has opened the way for investigating the genomic evolution of bacterial pathogens during the colonization and infection of humans. Recent GWAS research emphasized the importance of TLRs, especially TLR10 during pathogenesis in H. pylori infection. We demonstrated that TLR10, whose ligand was unknown for a long time, can recognize H. pylori LPS. Our results of H. pylori research suggest that TLR10 might play an important role to also recognize other commensal bacteria. In this review, we discuss the importance of TLRs in pro-inflammatory and anti-inflammatory responses by H. pylori infection. Especially, we highlight the TLR10 interaction with H. pylori infection, providing new insights about TLR10 signaling.
Collapse
Affiliation(s)
- Hiroyuki Nagashima
- Department of Gastroenterology, Hokkaido Cancer Center, Sapporo, Hokkaido, Japan.,Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita, 879-5593, Japan
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita, 879-5593, Japan. .,Department of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
63
|
Increased Expression of TLR10 in B Cell Subsets Correlates with Disease Activity in Rheumatoid Arthritis. Mediators Inflamm 2018; 2018:9372436. [PMID: 30686934 PMCID: PMC6327257 DOI: 10.1155/2018/9372436] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/10/2018] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptor (TLR) 10, mainly expressed on B cells, has emerged as a modulatory receptor in inflammation. Nonetheless, the clinical significance of TLR10 in rheumatoid arthritis (RA) remains unclear. In this study, we explored the expression of TLR10 in B cells and B cell subsets in RA subjects and healthy controls (HCs) and determined its relevance to disease activity and inflammatory biomarkers. TLR10 levels in B cells and B cell subsets (CD19+CD27+, CD19+CD27−, CD27+IgD−, CD27+IgD+, CD27−IgD+, D27−IgD−, CD19+CD5+, and CD19+CD5−) and inflammatory biomarker concentrations in peripheral blood (PB) obtained from RA subjects and HCs were detected by flow cytometry and enzyme-linked immunosorbent assay (ELISA), respectively. The correlations of TLR10 expression with disease activity and inflammatory biomarkers were then analysed. Similar levels of TLR10 in all CD19+ B cells were observed in the RA subjects and HCs. Compared to that in the HCs, TLR10 was elevated significantly in the CD19+CD27−IgD− and CD19+CD5+ subsets in the RA subjects. In addition, almost all subsets expressing TLR10 were increased with disease activity. The present study reveals that enhanced TLR10 in B cell subsets is positively correlated with disease activity in RA subjects.
Collapse
|
64
|
Heil M, Vega-Muñoz I. Nucleic Acid Sensing in Mammals and Plants: Facts and Caveats. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 345:225-285. [PMID: 30904194 DOI: 10.1016/bs.ircmb.2018.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The accumulation of nucleic acids in aberrant compartments is a signal of danger: fragments of cytosolic or extracellular self-DNA indicate cellular dysfunctions or disruption, whereas cytosolic fragments of nonself-DNA or RNA indicate infections. Therefore, nucleic acids trigger immunity in mammals and plants. In mammals, endosomal Toll-like receptors (TLRs) sense single-stranded (ss) or double-stranded (ds) RNA or CpG-rich DNA, whereas various cytosolic receptors sense dsDNA. Although a self/nonself discrimination could favor targeted immune responses, no sequence-specific sensing of nucleic acids has been reported for mammals. Specific immune responses to extracellular self-DNA versus DNA from related species were recently reported for plants, but the underlying mechanism remains unknown. The subcellular localization of mammalian receptors can favor self/nonself discrimination based on the localization of DNA fragments. However, autoantibodies and diverse damage-associated molecular patterns (DAMPs) shuttle DNA through membranes, and most of the mammalian receptors share downstream signaling elements such as stimulator of interferon genes (STING) and the master transcription regulators, nuclear factor (NF)-κB, and interferon regulatory factor 3 (IRF3). The resulting type I interferon (IFN) response stimulates innate immunity against multiple threats-from infection to physical injury or endogenous DNA damage-all of which lead to the accumulation of eDNA or cytoplasmatic dsDNA. Therefore, no or only low selective pressures might have favored a strict self/nonself discrimination in nucleic acid sensing. We conclude that the discrimination between self- and nonself-DNA is likely to be less strict-and less important-than assumed originally.
Collapse
Affiliation(s)
- Martin Heil
- Departmento de Ingeniería Genética, CINVESTAV-Irapuato, Irapuato, Guanajuato, Mexico.
| | - Isaac Vega-Muñoz
- Departmento de Ingeniería Genética, CINVESTAV-Irapuato, Irapuato, Guanajuato, Mexico
| |
Collapse
|
65
|
Crosstalk of toll-like receptors signaling and Nrf2 pathway for regulation of inflammation. Biomed Pharmacother 2018; 108:1866-1878. [PMID: 30372892 DOI: 10.1016/j.biopha.2018.10.019] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/09/2018] [Accepted: 10/03/2018] [Indexed: 12/20/2022] Open
Abstract
Inflammation as a second line of defense of innate immunity plays a crucial role in eliminating invading pathogens (bacteria, viruses, fungi as well as other parasites). The inflammatory response may also activate adaptive immune system involving lymphocytes to mount either antibody dependent or cell-mediated immune responses to clear pathogenic insult. However, if continued, the inflammatory processes may become uncontrolled culminating in cellular injury and tissue destruction, thereby manifesting itself in chronic form. The chronic inflammation has been associated with numerous human pathological conditions like allergies and autoimmune diseases, atherosclerosis, arthritis, Alzheimer's disease, cancer, obesity, type 2 diabetes, schizophrenia, neuro-degenerative diseases and numerous others. The dysregulated inflammatory process is associated with overproduction of free radicals leading to oxidative stress and activation of different cell signaling pathways. The regulation of inflammation by TLR signaling as well as Nrf2 pathways separately is widely documented. Since both these major signaling pathways modulate inflammation, they may crosstalk to bring about coordinated inflammatory responses. The linkage between TLR signaling and Nrf2-Keap1 pathway may serve as a bridge between immune regulation and oxidative stress responses through regulation of inflammation. Also, inflammation is reportedly responsible for the plethora of diseased conditions; a study of its regulation by targeting the TLR-Nrf2 cross-talks may also be beneficial for the development of therapeutic therapies or prophylactic treatments. Hence, present review focuses on the crosstalk between TLR signaling and Nrf2 pathway with respect to their role in modulation of inflammation in normal as well as pathologic conditions.
Collapse
|
66
|
Duan L, Zhang XD, Miao WY, Sun YJ, Xiong G, Wu Q, Li G, Yang P, Yu H, Li H, Wang Y, Zhang M, Hu LY, Tong X, Zhou WH, Yu X. PDGFRβ Cells Rapidly Relay Inflammatory Signal from the Circulatory System to Neurons via Chemokine CCL2. Neuron 2018; 100:183-200.e8. [PMID: 30269986 DOI: 10.1016/j.neuron.2018.08.030] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 03/30/2018] [Accepted: 08/20/2018] [Indexed: 01/19/2023]
Abstract
Acute infection, if not kept in check, can lead to systemic inflammatory responses in the brain. Here, we show that within 2 hr of systemic inflammation, PDGFRβ mural cells of blood vessels rapidly secrete chemokine CCL2, which in turn increases total neuronal excitability by promoting excitatory synaptic transmission in glutamatergic neurons of multiple brain regions. By single-cell RNA sequencing, we identified Col1a1 and Rgs5 subgroups of PDGFRβ cells as the main source of CCL2. Lipopolysaccharide (LPS)- or Poly(I:C)-treated pericyte culture medium induced similar effects in a CCL2-dependent manner. Importantly, in Pdgfrb-Cre;Ccl2fl/fl mice, LPS-induced increase in excitatory synaptic transmission was significantly attenuated. These results demonstrate in vivo that PDGFRβ cells function as initial sensors of external insults by secreting CCL2, which relays the signal to the central nervous system. Through their gateway position in the brain, PDGFRβ cells are ideally positioned to respond rapidly to environmental changes and to coordinate responses.
Collapse
Affiliation(s)
- Lihui Duan
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Di Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wan-Ying Miao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun-Jun Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoliang Xiong
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuzi Wu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangying Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ping Yang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Hang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Humingzhu Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yue Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Min Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li-Yuan Hu
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiaoping Tong
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wen-Hao Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
67
|
Yin F, Liu J, Gao S, Liu A, Zhao S, Li S, Wang J, Li Y, Luo J, Guan G, Yin H. Exploring the TLR and NLR signaling pathway relevant molecules induced by the Theileria annulata infection in calves. Parasitol Res 2018; 117:3269-3276. [PMID: 30084033 DOI: 10.1007/s00436-018-6026-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/24/2018] [Indexed: 01/28/2023]
Abstract
Theileria annulata is the pathogen of bovine tropical theileriosis. It is extremely harmful to the cattle industry, with huge economic losses. The toll-like receptor (TLR) and NOD-like receptor (NLR) signaling pathways are crucial for resistance to infection of the protozoa, such as Plasmodium falciparum, Toxoplasma gondii, and Trypanosoma cruzi. However, the role of these immune-related pathways is unclear during T. annulata infection. In the present study, peripheral blood mononuclear cells and serum were separated from blood samples of calves infected with homogenized tick supernatants carrying T. annulata sporozoites at 12 h, 24 h, 36 h, 48 h, 72 h, 96 h, 120 h, 144 h and 168 h postinoculation. The Custom RT2 Profiler PCR Array was used to explore the mRNA levels of 42 TLR and NLR signaling pathway relevant genes. The TLR1, TLR6, TLR10, NLRP1, and MyD88 genes and their downstream signaling molecules significantly differed after the T. annulata infection in comparison with that of preinfection from 72 h to 168 h postinoculation. The serum concentrations of IL-6, IL-1β, and TNFα were significantly increased at 96 h and 168 h postinfection. These findings provided novel information to help determine the mechanisms of TLR and NLR signaling pathway involvement in protection against T. annulata infection.
Collapse
Affiliation(s)
- Fangyuan Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Junlong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Shandian Gao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Aihong Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Shuaiyang Zhao
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Sitong Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Jinming Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Youquan Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China
| | - Guiquan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China.
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Xujiaping 1, Lanzhou, 730046, Gansu, People's Republic of China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
68
|
Yip TF, Selim ASM, Lian I, Lee SMY. Advancements in Host-Based Interventions for Influenza Treatment. Front Immunol 2018; 9:1547. [PMID: 30042762 PMCID: PMC6048202 DOI: 10.3389/fimmu.2018.01547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/22/2018] [Indexed: 12/15/2022] Open
Abstract
Influenza is a major acute respiratory infection that causes mortality and morbidity worldwide. Two classes of conventional antivirals, M2 ion channel blockers and neuraminidase inhibitors, are mainstays in managing influenza disease to lessen symptoms while minimizing hospitalization and death in patients with severe influenza. However, the development of viral resistance to both drug classes has become a major public health concern. Vaccines are prophylaxis mainstays but are limited in efficacy due to the difficulty in matching predicted dominant viral strains to circulating strains. As such, other potential interventions are being explored. Since viruses rely on host cellular functions to replicate, recent therapeutic developments focus on targeting host factors involved in virus replication. Besides controlling virus replication, potential targets for drug development include controlling virus-induced host immune responses such as the recently suggested involvement of innate lymphoid cells and NADPH oxidases in influenza virus pathogenesis and immune cell metabolism. In this review, we will discuss the advancements in novel host-based interventions for treating influenza disease.
Collapse
Affiliation(s)
- Tsz-Fung Yip
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Aisha Sami Mohammed Selim
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ida Lian
- School of Life Sciences and Chemical Technology, Ngee Ann Polytechnic, Singapore, Singapore
| | - Suki Man-Yan Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
69
|
Franko J, McCall JL, Barnett JB. Evaluating Macrophages in Immunotoxicity Testing. Methods Mol Biol 2018; 1803:255-296. [PMID: 29882145 DOI: 10.1007/978-1-4939-8549-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Macrophages are a heterogeneous group of cells that have a multitude of functions depending on their differentiation state. While classically known for their phagocytic and antigen presentation abilities, it is now evident that these cells fulfill homeostatic functions beyond the elimination of invading pathogens. In addition, macrophages have also been implicated in the downregulation of inflammatory responses following pathogen removal, tissue remodeling, repair, and angiogenesis. Alterations in macrophage differentiation and/or activity due to xenobiotic exposure can have grave consequences on organismal homeostasis, potentially contributing to disease due to immunosuppression or chronic inflammatory responses, depending upon the pathways affected. In this chapter, we provide an overview of the macrophages subtypes, their origin and a general discussion of several different assays used to assess their functional status.
Collapse
Affiliation(s)
- Jennifer Franko
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jamie L McCall
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - John B Barnett
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
70
|
Zhou S, Qi Q, Wang X, Zhang L, Xu L, Dong L, Zhu J, Li Y, Wang X, Xu Z, Liu F, Hu W, Zhou L, Chen X, Su C. SjHSP60 induces CD4 + CD25 + Foxp3 + Tregs via TLR4-Mal-drived production of TGF-β in macrophages. Immunol Cell Biol 2018; 96:958-968. [PMID: 29697865 DOI: 10.1111/imcb.12160] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 03/25/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) play a pivotal role in limiting immunopathological damage to host organs after schistosome infection. Transforming growth factor-β (TGF-β) is an essential factor for the periphery conversion of CD4+ CD25- T cells into CD4+ CD25+ Foxp3+ Tregs by inducing the key transcription factor Foxp3. Antigen presenting cells (APCs), which highly express TGF-β, are involved in parasite antigen-induced Treg conversion in peripheral. However, the mechanisms underlying high TGF-β induction in APCs by parasite antigens remain to be clarified during schistosome infection. Here, we demonstrated that Schistosoma japonicum stress protein, heat shock protein 60 (SjHSP60), promoted TGF-β production in macrophages (Mφ). Furthermore, we showed that activation of TLR4-Mal (MyD88 adaptor-like protein) signaling by SjHSP60 is necessary for induction of TGF-β expression in Mφ, which subsequently promoted Treg induction. Our results not only demonstrate a novel mechanism of TGF-β production in Mφ for inducing Tregs in mice with schistosomiasis, but also allude to the possibility of targeting parasite stress protein for potential therapeutics.
Collapse
Affiliation(s)
- Sha Zhou
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qianqian Qi
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofan Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lina Zhang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liyang Dong
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jifeng Zhu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yalin Li
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuefeng Wang
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhipeng Xu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Liu
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Hu
- Department of Microbiology and Microbial Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Liang Zhou
- Department of Infectious Diseases and Immunology, University of Florida, Gainesville, FL, USA
| | - Xiaojun Chen
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuan Su
- State Key Lab of Reproductive Medicine, Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
71
|
Figueiredo Borgognoni C, Kim JH, Zucolotto V, Fuchs H, Riehemann K. Human macrophage responses to metal-oxide nanoparticles: a review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:694-703. [PMID: 29726285 DOI: 10.1080/21691401.2018.1468767] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nanomaterials have been widely used in our daily lives in medicine, cosmetics, paints, textiles and food products. Many studies aim to determine their biological effects in different types of cells. The interaction of these materials with the immune system leads to reactions by modifying the susceptibility or resistance of the host body which could induce adverse health effects. Macrophages, as specific cells of the innate immune response, play a crucial role in the human defence system to foreign agents. They can be used as a reliable test object for the investigation of immune responses under nanomaterials exposure displayed by expression of a variety of receptors and active secretion of key signalling substances for these processes. This report covers studies of human macrophage behaviours upon exposure of nanomaterials. We focused on their interaction with metal-oxide nanoparticles as these are largely used in medical and cosmetics applications. The discussion and summary of these studies can guide the development of new nanomaterials, which are, at the same time, safe and useful for new purposes, especially for health applications.
Collapse
Affiliation(s)
- Camila Figueiredo Borgognoni
- a Center for Nanotechnology (CeNTech) , Münster , Germany.,b Physics Institute , University of Sao Paulo , Sao Carlos , Brazil
| | - Joo Hyoung Kim
- a Center for Nanotechnology (CeNTech) , Münster , Germany.,c Institute of Physics , University of Münster , Münster , Germany
| | | | - Harald Fuchs
- a Center for Nanotechnology (CeNTech) , Münster , Germany.,c Institute of Physics , University of Münster , Münster , Germany
| | - Kristina Riehemann
- a Center for Nanotechnology (CeNTech) , Münster , Germany.,c Institute of Physics , University of Münster , Münster , Germany
| |
Collapse
|
72
|
Li P, Xu J, Rao HM, Li X, Zhang YK, Jiang F, Wu WX. Mechanism of Apoptosis Induction by Mycoplasmal Nuclease MGA_0676 in Chicken Embryo Fibroblasts. Front Cell Infect Microbiol 2018; 8:105. [PMID: 29670864 PMCID: PMC5893762 DOI: 10.3389/fcimb.2018.00105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/19/2018] [Indexed: 12/22/2022] Open
Abstract
MGA_0676 has been characterized as a Mycoplasma gallisepticum nuclease that can induce apoptosis of chicken cells. However, the mechanism by which MGA_0676 induces apoptosis has remained unclear. In this study, we evaluated MGA_0676-induced apoptosis and internalization in immortalized chicken embryo fibroblasts (DF-1) and cancer cell lines. The internalization of MGA_0676 was proven through caveolin-mediated endocytosis by blocking the endocytosis with specific inhibitors or with siRNA. We identified the Thif domain of NEDD8-activating enzyme E1 regulatory subunit (NAE) in DF-1 as the target region interacting with the SNC domain of MGA_0676. The interaction between the Thif and SNC domains was observed co-located in the perinuclear and nuclear of DF-1. We found that the interaction between NAE and MGA_0676 increased the ability of apoptosis and accelerated the process of cullin neddylation in DF-1 cells, in turn activating NF-κB. This resulted in the observed aggregation of NF-κB in the nuclei of DF-1 cells. Moreover, the apoptosis induced by MGA_0676 decreased significantly when NF-κB was inhibited by siRNA or BAY 11-7082 or when NAE was silenced by siRNA. Overall, our results demonstrate that MGA_0676 is internalized through caveolin-mediated endocytosis, interacts with SNC-dependent Thif to accelerate the process of cullin neddylation and activates NF-κB in DF-1 cells, ultimately playing a key role in apoptosis in chicken cells. Our results indicate MGA_0676 constitutes a critical etiological virulence factor of the respiratory disease caused by M. gallisepticum. This study also opens a venue to investigate MGA_0676 as a potential candidate as pro-apoptotic drug in cancer studies.
Collapse
Affiliation(s)
- Peng Li
- Key Laboratory of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jian Xu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agricultural and Forestry Sciences, Beijing, China
| | - Hong-Mei Rao
- Key Laboratory of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xia Li
- Key Laboratory of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yun-Ke Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Fei Jiang
- Veterinary Diagnostic Laboratory, China Animal Disease Control Center, Beijing, China
| | - Wen-Xue Wu
- Key Laboratory of Animal Epidemiology and Zoonosis, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
73
|
Enteric Virome Sensing-Its Role in Intestinal Homeostasis and Immunity. Viruses 2018; 10:v10040146. [PMID: 29570694 PMCID: PMC5923440 DOI: 10.3390/v10040146] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/18/2018] [Accepted: 03/22/2018] [Indexed: 12/18/2022] Open
Abstract
Pattern recognition receptors (PRRs) sensing commensal microorganisms in the intestine induce tightly controlled tonic signaling in the intestinal mucosa, which is required to maintain intestinal barrier integrity and immune homeostasis. At the same time, PRR signaling pathways rapidly trigger the innate immune defense against invasive pathogens in the intestine. Intestinal epithelial cells and mononuclear phagocytes in the intestine and the gut-associated lymphoid tissues are critically involved in sensing components of the microbiome and regulating immune responses in the intestine to sustain immune tolerance against harmless antigens and to prevent inflammation. These processes have been mostly investigated in the context of the bacterial components of the microbiome so far. The impact of viruses residing in the intestine and the virus sensors, which are activated by these enteric viruses, on intestinal homeostasis and inflammation is just beginning to be unraveled. In this review, we will summarize recent findings indicating an important role of the enteric virome for intestinal homeostasis as well as pathology when the immune system fails to control the enteric virome. We will provide an overview of the virus sensors and signaling pathways, operative in the intestine and the mononuclear phagocyte subsets, which can sense viruses and shape the intestinal immune response. We will discuss how these might interact with resident enteric viruses directly or in context with the bacterial microbiome to affect intestinal homeostasis.
Collapse
|
74
|
Lee SMY, Yip TF, Yan S, Jin DY, Wei HL, Guo RT, Peiris JSM. Recognition of Double-Stranded RNA and Regulation of Interferon Pathway by Toll-Like Receptor 10. Front Immunol 2018; 9:516. [PMID: 29616030 PMCID: PMC5865411 DOI: 10.3389/fimmu.2018.00516] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/27/2018] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR)-10 remains an orphan receptor without well-characterized ligands or functions. Here, we reveal that TLR10 is predominantly localized to endosomes and binds dsRNA in vitro at endosomal pH, suggesting that dsRNA is a ligand of TLR10. Recognition of dsRNA by TLR10 activates recruitment of myeloid differentiation primary response gene 88 for signal transduction and suppression of interferon regulatory factor-7 dependent type I IFN production. We also demonstrate crosstalk between TLR10 and TLR3, as they compete with each other for dsRNA binding. Our results suggest for the first time that dsRNA is a ligand for TLR10 and propose novel dual functions of TLR10 in regulating IFN signaling: first, recognition of dsRNA as a nucleotide-sensing receptor and second, sequestration of dsRNA from TLR3 to inhibit TLR3 signaling in response to dsRNA stimulation.
Collapse
Affiliation(s)
- Suki Man-Yan Lee
- HKU-Pasteur Research Pole and Center of Influenza Research, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tsz-Fung Yip
- HKU-Pasteur Research Pole and Center of Influenza Research, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sheng Yan
- HKU-Pasteur Research Pole and Center of Influenza Research, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hong-Li Wei
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Rey-Ting Guo
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Joseph Sriyal Malik Peiris
- HKU-Pasteur Research Pole and Center of Influenza Research, Li Ka Shing Faculty of Medicine, School of Public Health, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
75
|
Patra MC, Kwon HK, Batool M, Choi S. Computational Insight Into the Structural Organization of Full-Length Toll-Like Receptor 4 Dimer in a Model Phospholipid Bilayer. Front Immunol 2018; 9:489. [PMID: 29593733 PMCID: PMC5857566 DOI: 10.3389/fimmu.2018.00489] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/26/2018] [Indexed: 01/07/2023] Open
Abstract
Toll-like receptors (TLRs) are a unique category of pattern recognition receptors that recognize distinct pathogenic components, often utilizing the same set of downstream adaptors. Specific molecular features of extracellular, transmembrane (TM), and cytoplasmic domains of TLRs are crucial for coordinating the complex, innate immune signaling pathway. Here, we constructed a full-length structural model of TLR4-a widely studied member of the interleukin-1 receptor/TLR superfamily-using homology modeling, protein-protein docking, and molecular dynamics simulations to understand the differential domain organization of TLR4 in a membrane-aqueous environment. Results showed that each functional domain of the membrane-bound TLR4 displayed several structural transitions that are biophysically essential for plasma membrane integration. Specifically, the extracellular and cytoplasmic domains were partially immersed in the upper and lower leaflets of the membrane bilayer. Meanwhile, TM domains tilted considerably to overcome the hydrophobic mismatch with the bilayer core. Our analysis indicates an alternate dimerization or a potential oligomerization interface of TLR4-TM. Moreover, the helical properties of an isolated TM dimer partly agree with that of the full-length receptor. Furthermore, membrane-absorbed or solvent-exposed surfaces of the toll/interleukin-1 receptor domain are consistent with previous X-ray crystallography and biochemical studies. Collectively, we provided a complete structural model of membrane-bound TLR4 that strengthens our current understanding of the complex mechanism of receptor activation and adaptor recruitment in the innate immune signaling pathway.
Collapse
Affiliation(s)
- Mahesh Chandra Patra
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Hyuk-Kwon Kwon
- Department of Orthopaedics and Rehabilitation, Yale School of Medicine, New Haven, CT, United States
| | - Maria Batool
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
76
|
Chan CP, Yuen CK, Cheung PHH, Fung SY, Lui PY, Chen H, Kok KH, Jin DY. Antiviral activity of double-stranded RNA-binding protein PACT against influenza A virus mediated via suppression of viral RNA polymerase. FASEB J 2018. [PMID: 29513570 DOI: 10.1096/fj.201701361r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PACT is a double-stranded RNA-binding protein that has been implicated in host-influenza A virus (IAV) interaction. PACT facilitates the action of RIG-I in the activation of the type I IFN response, which is suppressed by the viral nonstructural protein NS1. PACT is also known to interact with the IAV RNA polymerase subunit PA. Exactly how PACT exerts its antiviral activity during IAV infection remains to be elucidated. In the current study, we demonstrated the interplay between PACT and IAV polymerase. Induction of IFN-β by the IAV RNP complex was most robust when both RIG-I and PACT were expressed. PACT-dependent activation of IFN-β production was suppressed by the IAV polymerase subunits, polymerase acidic protein, polymerase basic protein 1 (PB1), and PB2. PACT associated with PA, PB1, and PB2. Compromising PACT in IAV-infected A549 cells resulted in the augmentation of viral RNA (vRNA) transcription and replication and IFN-β production. Furthermore, vRNA replication was boosted by knockdown of PACT in both A549 cells and IFN-deficient Vero cells. Thus, the antiviral activity of PACT is mediated primarily via its interaction with and inhibition of IAV polymerase. Taken together, our findings reveal a new facet of the host-IAV interaction in which the interplay between PACT and IAV polymerase affects the outcome of viral infection and antiviral response.-Chan, C.-P., Yuen, C.-K., Cheung, P.-H. H., Fung, S.-Y., Lui, P.-Y., Chen, H., Kok, K.-H., Jin, D.-Y. Antiviral activity of double-stranded RNA-binding protein PACT against influenza A virus mediated via suppression of viral RNA polymerase.
Collapse
Affiliation(s)
- Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Chun-Kit Yuen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Sin-Yee Fung
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Pak-Yin Lui
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Honglin Chen
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Kin-Hang Kok
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
77
|
Abstract
Innate immunity is traditionally thought of as the first line of defense against pathogens that enter the body. It is typically characterized as a rather weak defense mechanism, designed to restrict pathogen replication until the adaptive immune response generates a tailored response and eliminates the infectious agent. However, intensive research in recent years has resulted in better understanding of innate immunity as well as the discovery of many effector proteins, revealing its numerous powerful mechanisms to defend the host. Furthermore, this research has demonstrated that it is simplistic to strictly separate adaptive and innate immune functions since these two systems often work synergistically rather than sequentially. Here, we provide a broad overview of innate pattern recognition receptors in antiviral defense, with a focus on the TRIM family, and discuss their signaling pathways and mechanisms of action with special emphasis on the intracellular antibody receptor TRIM21.
Collapse
Affiliation(s)
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
78
|
Patra MC, Choi S. Insight into Phosphatidylinositol-Dependent Membrane Localization of the Innate Immune Adaptor Protein Toll/Interleukin 1 Receptor Domain-Containing Adaptor Protein. Front Immunol 2018; 9:75. [PMID: 29434596 PMCID: PMC5796906 DOI: 10.3389/fimmu.2018.00075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 01/11/2018] [Indexed: 01/23/2023] Open
Abstract
The toll/interleukin 1 receptor (TIR) domain-containing adaptor protein (TIRAP) plays an important role in the toll-like receptor (TLR) 2, TLR4, TLR7, and TLR9 signaling pathways. TIRAP anchors to phosphatidylinositol (PI) 4,5-bisphosphate (PIP2) on the plasma membrane and PI (3,4,5)-trisphosphate (PIP3) on the endosomal membrane and assists in recruitment of the myeloid differentiation primary response 88 protein to activated TLRs. To date, the structure and mechanism of TIRAP’s membrane association are only partially understood. Here, we modeled an all-residue TIRAP dimer using homology modeling, threading, and protein–protein docking strategies. Molecular dynamics simulations revealed that PIP2 creates a stable microdomain in a dipalmitoylphosphatidylcholine bilayer, providing TIRAP with its physiologically relevant orientation. Computed binding free energy values suggest that the affinity of PI-binding domain (PBD) for PIP2 is stronger than that of TIRAP as a whole for PIP2 and that the short PI-binding motif (PBM) contributes to the affinity between PBD and PIP2. Four PIP2 molecules can be accommodated by distinct lysine-rich surfaces on the dimeric PBM. Along with the known PI-binding residues (K15, K16, K31, and K32), additional positively charged residues (K34, K35, and R36) showed strong affinity toward PIP2. Lysine-to-alanine mutations at the PI-binding residues abolished TIRAP’s affinity for PIP2; however, K34, K35, and R36 consistently interacted with PIP2 headgroups through hydrogen bond (H-bond) and electrostatic interactions. TIRAP exhibited a PIP2-analogous intermolecular contact and binding affinity toward PIP3, aided by an H-bond network involving K34, K35, and R36. The present study extends our understanding of TIRAP’s membrane association, which could be helpful in designing peptide decoys to block TLR2-, TLR4-, TLR7-, and TLR9-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Mahesh Chandra Patra
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, South Korea
| |
Collapse
|
79
|
Pickens JA, Tripp RA. Verdinexor Targeting of CRM1 is a Promising Therapeutic Approach against RSV and Influenza Viruses. Viruses 2018; 10:E48. [PMID: 29361733 PMCID: PMC5795461 DOI: 10.3390/v10010048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/11/2022] Open
Abstract
Two primary causes of respiratory tract infections are respiratory syncytial virus (RSV) and influenza viruses, both of which remain major public health concerns. There are a limited number of antiviral drugs available for the treatment of RSV and influenza, each having limited effectiveness and each driving selective pressure for the emergence of drug-resistant viruses. Novel broad-spectrum antivirals are needed to circumvent problems with current disease intervention strategies, while improving the cytokine-induced immunopathology associated with RSV and influenza infections. In this review, we examine the use of Verdinexor (KPT-335, a novel orally bioavailable drug that functions as a selective inhibitor of nuclear export, SINE), as an antiviral with multifaceted therapeutic potential. KPT-335 works to (1) block CRM1 (i.e., Chromosome Region Maintenance 1; exportin 1 or XPO1) mediated export of viral proteins critical for RSV and influenza pathogenesis; and (2) repress nuclear factor κB (NF-κB) activation, thus reducing cytokine production and eliminating virus-associated immunopathology. The repurposing of SINE compounds as antivirals shows promise not only against RSV and influenza virus but also against other viruses that exploit the nucleus as part of their viral life cycle.
Collapse
Affiliation(s)
- Jennifer A Pickens
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| | - Ralph A Tripp
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
80
|
Musella M, Manic G, De Maria R, Vitale I, Sistigu A. Type-I-interferons in infection and cancer: Unanticipated dynamics with therapeutic implications. Oncoimmunology 2017. [PMID: 28638743 DOI: 10.1080/2162402x.2017.1314424] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
If there is a great new hope in the treatment of cancer, the immune system is it. Innate and adaptive immunity either promote or attenuate tumorigenesis and so can have opposing effects on the therapeutic outcome. Originally described as potent antivirals, Type-I interferons (IFNs) were quickly recognized as central coordinators of tumor-immune system interactions. Type-I-IFNs are produced by, and act on, both tumor and immune cells being either host-protecting or tumor-promoting. Here, we discuss Type-I-IFNs in infectious and cancer diseases highlighting their dichotomous role and raising the importance to deeply understand the underlying mechanisms so to reshape the way we can exploit Type-I-IFNs therapeutically.
Collapse
Affiliation(s)
- Martina Musella
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Gwenola Manic
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Ruggero De Maria
- Department of General Pathology and Physiopathology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ilio Vitale
- Unit of Cellular Networks and Molecular Therapeutic Targets, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy.,Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Antonella Sistigu
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
81
|
Tavares LP, Teixeira MM, Garcia CC. The inflammatory response triggered by Influenza virus: a two edged sword. Inflamm Res 2017; 66:283-302. [PMID: 27744631 DOI: 10.1007/s00011-016-0996-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV) is a relevant respiratory tract pathogen leading to a great number of deaths and hospitalizations worldwide. Secondary bacterial infections are a very common cause of IAV associated morbidity and mortality. The robust inflammatory response that follows infection is important for the control of virus proliferation but is also associated with lung damage, morbidity and death. The role of the different components of immune response underlying protection or disease during IAV infection is not completely elucidated. Overall, in the context of IAV infection, inflammation is a 'double edge sword' necessary to control infection but causing disease. Therefore, a growing number of studies suggest that immunomodulatory strategies may improve disease outcome without affecting the ability of the host to deal with infection. This review summarizes recent aspects of the inflammatory responses triggered by IAV that are preferentially involved in causing severe pulmonary disease and the anti-inflammatory strategies that have been suggested to treat influenza induced immunopathology.
Collapse
Affiliation(s)
- Luciana P Tavares
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mauro M Teixeira
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristiana C Garcia
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, ICB Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Laboratório de Vírus Respiratórios e do Sarampo, Instituto Oswaldo Cruz, Fiocruz, Avenida Brasil, 4365, 21040360, Rio de Janeiro, Brazil.
| |
Collapse
|
82
|
Hess NJ, Felicelli C, Grage J, Tapping RI. TLR10 suppresses the activation and differentiation of monocytes with effects on DC-mediated adaptive immune responses. J Leukoc Biol 2017; 101:1245-1252. [PMID: 28235773 DOI: 10.1189/jlb.3a1116-492r] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/16/2017] [Accepted: 01/30/2017] [Indexed: 11/24/2022] Open
Abstract
TLRs are important pattern-recognition receptors involved in the activation of innate immune responses against foreign pathogens. TLR10 is the only TLR family member without a known ligand, signaling pathway, or clear cellular function. Previous work has shown that TLR10 suppresses proinflammatory cytokine production in response to TLR agonists in a mixed human mononuclear cell population. We report that TLR10 is preferentially expressed on monocytes and suppresses proinflammatory cytokine production resulting from either TLR or CD40 stimulation. TLR10 engagement affects both the MAPK and Akt signaling pathways, leading to changes in the transcriptome of isolated human monocytes. Differentiation of monocytes into dendritic cells in the presence of an αTLR10 mAb reduced the expression of maturation markers and the induction of proinflammatory cytokines, again in response to either TLR or CD40 stimulation. Finally, in coculture experiments, TLR10 differentiated dendritic cells exhibited a decreased capacity to activate T cells as measured by IL-2 and IFN-γ production. These data demonstrate that TLR10 is a novel regulator of innate immune responses and of the differentiation of primary human monocytes into effective dendritic cells.
Collapse
Affiliation(s)
- Nicholas J Hess
- Department of Microbiology, University of Illinois, Urbana-Champaign, Illinois, USA; and
| | - Christopher Felicelli
- Department of Microbiology, University of Illinois, Urbana-Champaign, Illinois, USA; and
| | - Jennifer Grage
- Department of Microbiology, University of Illinois, Urbana-Champaign, Illinois, USA; and
| | - Richard I Tapping
- Department of Microbiology, University of Illinois, Urbana-Champaign, Illinois, USA; and .,College of Medicine, University of Illinois, Urbana-Champaign, Illinois, USA
| |
Collapse
|
83
|
Review on Toll-Like Receptor Activation in Myasthenia Gravis: Application to the Development of New Experimental Models. Clin Rev Allergy Immunol 2017; 52:133-147. [PMID: 27207173 DOI: 10.1007/s12016-016-8549-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Abnormal toll-like receptor (TLR) activation and uncontrolled resolution of inflammation are suspected to play a key role in the development of autoimmune diseases. Acquired myasthenia gravis (MG) is an invalidating neuromuscular disease leading to muscle weaknesses. MG is mainly mediated by anti-acetylcholine receptor (AChR) autoantibodies, and thymic hyperplasia characterized by ectopic germinal centers is a common feature in MG. An abnormal expression of certain TLRs is observed in the thymus of MG patients associated with the overexpression of interferon (IFN)-β, the orchestrator of thymic changes in MG. Experimental models have been developed for numerous autoimmune diseases. These models are induced by animal immunization with a purified antigen solubilized in complete Freund's adjuvant (CFA) containing heat-inactivated mycobacterium tuberculosis (MTB). Sensitization against the antigen is mainly due to the activation of TLR signaling pathways by the pathogen motifs displayed by MTB, and attempts have been made to substitute the use of CFA by TLR agonists. AChR emulsified in CFA is used to induce the classical experimental autoimmune MG model (EAMG). However, the TLR4 activator lipopolysaccharide (LPS) has proved to be efficient to replace MTB and induce a sensitization against purified AChR. Poly(I:C), the well-known TLR3 agonist, is also able by itself to induce MG symptoms in mice associated with early thymic changes as observed in human MG. In this review, we discuss the abnormal expression of TLRs in MG patients and we describe the use of TLR agonists to induce EAMG in comparison with other autoimmune experimental models.
Collapse
|
84
|
Comparative and temporal transcriptome analysis of peste des petits ruminants virus infected goat peripheral blood mononuclear cells. Virus Res 2017; 229:28-40. [DOI: 10.1016/j.virusres.2016.12.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 11/22/2022]
|
85
|
Mortaz E, Adcock IM, Tabarsi P, Darazam IA, Movassaghi M, Garssen J, Jamaati H, Velayati A. Pattern recognitions receptors in immunodeficiency disorders. Eur J Pharmacol 2017; 808:49-56. [PMID: 28095323 DOI: 10.1016/j.ejphar.2017.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 01/04/2017] [Accepted: 01/13/2017] [Indexed: 01/13/2023]
Abstract
Pattern recognition receptors (PRRs) recognize common microbial or host-derived macromolecules and have important roles in early activation and response of the immune system. Initiation of the innate immune response starts with the recognition of microbial structures called pathogen associated molecular patterns (PAMPs). Recognition of PAMPs is performed by germline-encoded receptors expressed mainly on immune cells termed pattern recognition receptors (PRRs). Several classes of pattern recognition receptors (PRRs) are involved in the pathogenesis of diseases, including Toll-like receptors (TLRs), C-type lectin receptors (CLRs), and Nod-like receptors (NLRs). Patients with primary immune deficiencies (PIDs) affecting TLR signaling can elucidate the importance of these proteins in the human immune system. Defects in interleukin-1 receptor-associated kinase-4 and myeloid differentiation factor 88 (MyD88) lead to susceptibility to infections with bacteria, while mutations in nuclear factor-κB essential modulator (NEMO) and other downstream mediators generally induce broader susceptibility to bacteria, viruses, and fungi. In contrast, TLR3 signaling defects are associated with susceptibility to herpes simplex virus type 1 encephalitis. Other PIDs induce functional alterations of TLR signaling pathways, such as common variable immunodeficiency in which plasmacytoid dendritic cell defects enhance defective responses of B cells to shared TLR agonists. Altered TLR responses to TLR2 and 4 agonists are seen in chronic granulomatous disease (CGD) and X-linked agammaglobulinemia (XLA). Enhanced TLR responses, meanwhile, are seen for TLRs 5 and 9 in CGD, TLRs 4, 7/8, and 9 in XLA, TLRs 2 and 4 in hyper IgE syndrome (HIES), and for most TLRs in adenosine deaminase deficiency. In this review we provide the reader with an update on the role of TLRs and downstream signaling pathways in PID disorders.
Collapse
Affiliation(s)
- Esameil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ilad Alavi Darazam
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti, University of Medical Sciences,Tehran, Iran
| | - Masoud Movassaghi
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles (UCLA), USA
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Sciences, Utrecht University, Utrecht, The Netherlands; Department of Immunology, Nutricia Research, Utrecht, the Netherlands
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center and National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Aliakbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
86
|
Shirjang S, Mansoori B, Solali S, Hagh MF, Shamsasenjan K. Toll-like receptors as a key regulator of mesenchymal stem cell function: An up-to-date review. Cell Immunol 2016; 315:1-10. [PMID: 28284487 DOI: 10.1016/j.cellimm.2016.12.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/07/2016] [Accepted: 12/24/2016] [Indexed: 02/07/2023]
Abstract
Understanding the role of toll-like receptors (TLRs) in the immunomodulation potential, differentiation, migration, and survival of mesenchymal stem cells (MSCs) is absolutely vital to fully exploiting their MSC-based therapeutic potential. Furthermore, through recognition of exogenous or endogenous ligands produced upon injury, TLRs have been linked to allograft rejection and maintenance of chronic inflammatory diseases, including Crohn's disease, rheumatoid arthritis. Characterizing the effect of TLRs in biological control of MSCs fate and function could improve our knowledge about the MSC-based cell therapy and immunotherapy. In this paper, we outline the impacts of TLR activation and mechanisms on MSCs immunomodulatory functions, differentiation, migration, and survivability. Moreover, we indicate that the expression patterns of TLRs in MSCs from different sources.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
87
|
Kieckens E, Rybarczyk J, Li RW, Vanrompay D, Cox E. Potential immunosuppressive effects of Escherichia coli O157:H7 experimental infection on the bovine host. BMC Genomics 2016; 17:1049. [PMID: 28003017 PMCID: PMC5178093 DOI: 10.1186/s12864-016-3374-y] [Citation(s) in RCA: 9] [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: 06/21/2016] [Accepted: 12/05/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Enterohaemorrhagic Escherichia coli (EHEC), like E. coli O157:H7 are frequently detected in bovine faecal samples at slaughter. Cattle do not show clinical symptoms upon infection, but for humans the consequences after consuming contaminated beef can be severe. The immune response against EHEC in cattle cannot always clear the infection as persistent colonization and shedding in infected animals over a period of months often occurs. In previous infection trials, we observed a primary immune response after infection which was unable to protect cattle from re-infection. These results may reflect a suppression of certain immune pathways, making cattle more prone to persistent colonization after re-infection. To test this, RNA-Seq was used for transcriptome analysis of recto-anal junction tissue and ileal Peyer's patches in nine Holstein-Friesian calves in response to a primary and secondary Escherichia coli O157:H7 infection with the Shiga toxin (Stx) negative NCTC12900 strain. Non-infected calves served as controls. RESULTS In tissue of the recto-anal junction, only 15 genes were found to be significantly affected by a first infection compared to 1159 genes in the ileal Peyer's patches. Whereas, re-infection significantly changed the expression of 10 and 17 genes in the recto-anal junction tissue and the Peyer's patches, respectively. A significant downregulation of 69 immunostimulatory genes and a significant upregulation of seven immune suppressing genes was observed. CONCLUSIONS Although the recto-anal junction is a major site of colonization, this area does not seem to be modulated upon infection to the same extent as ileal Peyer's patches as the changes in gene expression were remarkably higher in the ileal Peyer's patches than in the recto-anal junction during a primary but not a secondary infection. We can conclude that the main effect on the transcriptome was immunosuppression by E. coli O157:H7 (Stx-) due to an upregulation of immune suppressive effects (7/12 genes) or a downregulation of immunostimulatory effects (69/94 genes) in the ileal Peyer's patches. These data might indicate that a primary infection promotes a re-infection with EHEC by suppressing the immune function.
Collapse
Affiliation(s)
- E. Kieckens
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - J. Rybarczyk
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - R. W. Li
- USDA-ARS, Bovine Functional Genomics Laboratory, Beltsville, MD USA
| | - D. Vanrompay
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - E. Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
88
|
CLEC5A-Mediated Enhancement of the Inflammatory Response in Myeloid Cells Contributes to Influenza Virus Pathogenicity In Vivo. J Virol 2016; 91:JVI.01813-16. [PMID: 27795434 PMCID: PMC5165214 DOI: 10.1128/jvi.01813-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Human infections with influenza viruses exhibit mild to severe clinical outcomes as a result of complex virus-host interactions. Induction of inflammatory mediators via pattern recognition receptors may dictate subsequent host responses for pathogen clearance and tissue damage. We identified that human C-type lectin domain family 5 member A (CLEC5A) interacts with the hemagglutinin protein of influenza viruses expressed on lentiviral pseudoparticles through lectin screening. Silencing CLEC5A gene expression, blocking influenza-CLEC5A interactions with anti-CLEC5A antibodies, or dampening CLEC5A-mediated signaling using a spleen tyrosine kinase inhibitor consistently reduced the levels of proinflammatory cytokines produced by human macrophages without affecting the replication of influenza A viruses of different subtypes. Infection of bone marrow-derived macrophages from CLEC5A-deficient mice showed reduced levels of tumor necrosis factor alpha (TNF-α) and IP-10 but elevated alpha interferon (IFN-α) compared to those of wild-type mice. The heightened type I IFN response in the macrophages of CLEC5A-deficient mice was associated with upregulated TLR3 mRNA after treatment with double-stranded RNA. Upon lethal challenges with a recombinant H5N1 virus, CLEC5A-deficient mice showed reduced levels of proinflammatory cytokines, decreased immune cell infiltration in the lungs, and improved survival compared to the wild-type mice, despite comparable viral loads noted throughout the course of infection. The survival difference was more prominent at a lower dose of inoculum. Our results suggest that CLEC5A-mediated enhancement of the inflammatory response in myeloid cells contributes to influenza pathogenicity in vivo and may be considered a therapeutic target in combination with effective antivirals. Well-orchestrated host responses together with effective viral clearance are critical for optimal clinical outcome after influenza infections.
IMPORTANCE Multiple pattern recognition receptors work in synergy to sense viral RNA or proteins synthesized during influenza replication and mediate host responses for viral control. Well-orchestrated host responses may help to maintain the inflammatory response to minimize tissue damage while inducing an effective adaptive immune response for viral clearance. We identified that CLEC5A, a C-type lectin receptor which has previously been reported to mediate flavivirus-induced inflammatory responses, enhanced induction of proinflammatory cytokines and chemokines in myeloid cells after influenza infections. CLEC5A-deficient mice infected with influenza virus showed reduced inflammation in the lungs and improved survival compared to that of the wild-type mice despite comparable viral loads. The survival difference was more prominent at a lower dose of inoculum. Collectively, our results suggest that dampening CLEC5A-mediated inflammatory responses in myeloid cells reduces immunopathogenesis after influenza infections.
Collapse
|
89
|
Elshabrawy HA, Essani AE, Szekanecz Z, Fox DA, Shahrara S. TLRs, future potential therapeutic targets for RA. Autoimmun Rev 2016; 16:103-113. [PMID: 27988432 DOI: 10.1016/j.autrev.2016.12.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 01/27/2023]
Abstract
Toll like receptors (TLR)s have a central role in regulating innate immunity and in the last decade studies have begun to reveal their significance in potentiating autoimmune diseases such as rheumatoid arthritis (RA). Earlier investigations have highlighted the importance of TLR2 and TLR4 function in RA pathogenesis. In this review, we discuss the newer data that indicate roles for TLR5 and TLR7 in RA and its preclinical models. We evaluate the pathogenicity of TLRs in RA myeloid cells, synovial tissue fibroblasts, T cells, osteoclast progenitor cells and endothelial cells. These observations establish that ligation of TLRs can transform RA myeloid cells into M1 macrophages and that the inflammatory factors secreted from M1 and RA synovial tissue fibroblasts participate in TH-17 cell development. From the investigations conducted in RA preclinical models, we conclude that TLR-mediated inflammation can result in osteoclastic bone erosion by interconnecting the myeloid and TH-17 cell response to joint vascularization. In light of emerging unique aspects of TLR function, we summarize the novel approaches that are being tested to impair TLR activation in RA patients.
Collapse
Affiliation(s)
- Hatem A Elshabrawy
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA
| | - Abdul E Essani
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA
| | - Zoltán Szekanecz
- Department of Rheumatology, Institute of Medicine, University of Debrecen Faculty of Medicine, Nagyerdei Str 98, Debrecen H-4004, Hungary
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shiva Shahrara
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA.
| |
Collapse
|
90
|
Hess NJ, Jiang S, Li X, Guan Y, Tapping RI. TLR10 Is a B Cell Intrinsic Suppressor of Adaptive Immune Responses. THE JOURNAL OF IMMUNOLOGY 2016; 198:699-707. [PMID: 27956526 DOI: 10.4049/jimmunol.1601335] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/11/2016] [Indexed: 12/29/2022]
Abstract
Toll-like receptors play a central role in the initiation of adaptive immune responses with several TLR agonists acting as known B cell mitogens. Despite thousands of publications on TLRs, the function of TLR10 remains unknown. We have found that Ab-mediated engagement of TLR10 on primary human B cells suppresses B cell proliferation, cytokine production, and signal transduction. When challenged with either a T independent or T dependent Ag, TLR10 transgenic mice exhibit diminished Ab responses. Adoptive transfer of splenic B cells into B cell-deficient mice revealed that the suppressive effects on Ag-specific humoral immune responses are entirely B cell intrinsic. Our results demonstrate that TLR10 has a functional role within the B cell lineage that is distinct from that of other TLR family members and may provide a potential therapeutic target for diseases characterized by dysregulated B cell activity.
Collapse
Affiliation(s)
- Nicholas J Hess
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; and
| | - Song Jiang
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; and.,College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Xinyan Li
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; and
| | - Yue Guan
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; and
| | - Richard I Tapping
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; and .,College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
91
|
Behzadi E, Behzadi P. The role of toll-like receptors (TLRs) in urinary tract infections (UTIs). Cent European J Urol 2016; 69:404-410. [PMID: 28127459 PMCID: PMC5260452 DOI: 10.5173/ceju.2016.871] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/01/2016] [Accepted: 09/27/2016] [Indexed: 12/16/2022] Open
Abstract
Introduction Urinary Tract Infections (UTIs) are caused by different types of microbial agents such as uropathogenic Escherichia coli (UPEC) and Candida albicans. The presence of strong physical barriers may prevent the breach of pathogens into the urinary tract. However, sometimes the pathogenic microorganisms may pass through the barriers and stimulate the innate and adaptive responses. Among a variety of innate immune responses, Toll-Like Receptors (TLRs) are one of the most unique and interesting molecules regarding UTIs. Thus, the authors have focused their attention on the role of TLRs in urinary tract defense against pathogenic microbial agents such as UPEC and C.albicans through this literature review. Material and methods Several papers regarding UTIs and TLRs including original and review articles were searched by PubMed and Google Scholar. They were studied and the most important aspects in association with the role of TLRs in UTIs were extracted. Additionally, this paper was prepared using the experience of the authors. Results The TLRs 2, 4 and 5 are the most functional molecules that contribute to urinary tract defense system and UTIs. It is incredible that TLRs are able to detect and recognize different parts of microbial components relating to the same pathogen. Besides, the flexibility of the TLR molecules may lead to identification of different types of microorganisms with different signaling pathways. Conclusions Our knowledge associated with TLRs and their activities against microbial causative agents of UTIs may help us to prevent, control and treat UTIs at a higher quality level.
Collapse
Affiliation(s)
- Elham Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Payam Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
92
|
Gittelman RM, Schraiber JG, Vernot B, Mikacenic C, Wurfel MM, Akey JM. Archaic Hominin Admixture Facilitated Adaptation to Out-of-Africa Environments. Curr Biol 2016; 26:3375-3382. [PMID: 27839976 DOI: 10.1016/j.cub.2016.10.041] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/26/2016] [Accepted: 10/19/2016] [Indexed: 12/31/2022]
Abstract
As modern humans dispersed from Africa throughout the world, they encountered and interbred with archaic hominins, including Neanderthals and Denisovans [1, 2]. Although genome-scale maps of introgressed sequences have been constructed [3-6], considerable gaps in knowledge remain about the functional, phenotypic, and evolutionary significance of archaic hominin DNA that persists in present-day individuals. Here, we describe a comprehensive set of analyses that identified 126 high-frequency archaic haplotypes as putative targets of adaptive introgression in geographically diverse populations. These loci are enriched for immune-related genes (such as OAS1/2/3, TLR1/6/10, and TNFAIP3) and also encompass genes (including OCA2 and BNC2) that influence skin pigmentation phenotypes. Furthermore, we leveraged existing and novel large-scale gene expression datasets to show many positively selected archaic haplotypes act as expression quantitative trait loci (eQTLs), suggesting that modulation of transcript abundance was a common mechanism facilitating adaptive introgression. Our results demonstrate that hybridization between modern and archaic hominins provided an important reservoir of advantageous alleles that enabled adaptation to out-of-Africa environments.
Collapse
Affiliation(s)
- Rachel M Gittelman
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Joshua G Schraiber
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Benjamin Vernot
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Carmen Mikacenic
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98195, USA
| | - Mark M Wurfel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98195, USA
| | - Joshua M Akey
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
93
|
Semik E, Gurgul A, Ząbek T, Ropka-Molik K, Koch C, Mählmann K, Bugno-Poniewierska M. Transcriptome analysis of equine sarcoids. Vet Comp Oncol 2016; 15:1370-1381. [PMID: 27779365 DOI: 10.1111/vco.12279] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/23/2016] [Accepted: 09/24/2016] [Indexed: 01/28/2023]
Abstract
Equine sarcoids are the most commonly detected skin tumours in Equidae. In the present research, a comparative transcriptomic analysis was performed which aimed at looking inside a tumour biology and identification of the expression profile as a potential source of cancer specific genes useful as biomarkers. We have used Horse Gene Expression Microarray data from matched equine sarcoids and tumour-distant skin samples. In total, 901 significantly differentially expressed genes (DEGs) between lesional and healthy skin samples have been identified (fold change ≥ 2; P < 0.05). The large subset of DEGs, with decreased expression, was associated with a suppression of malignant transformation, whereas several overexpressed genes were involved in the processes associated with growth and progression of a tumour or immune system activity. Our results, as a first to date, showed comprehensive transcriptome analysis of skin tumour in horses and pinpointed significant pathways and genes related with oncogenesis processes.
Collapse
Affiliation(s)
- E Semik
- Department of Genomics and Molecular Biology of Animals, National Research Institute of Animal Production, Balice, Poland
| | - A Gurgul
- Department of Genomics and Molecular Biology of Animals, National Research Institute of Animal Production, Balice, Poland
| | - T Ząbek
- Department of Genomics and Molecular Biology of Animals, National Research Institute of Animal Production, Balice, Poland
| | - K Ropka-Molik
- Department of Genomics and Molecular Biology of Animals, National Research Institute of Animal Production, Balice, Poland
| | - C Koch
- ISME - Equine Clinic Bern, Department of Clinical Veterinary Medicine, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - K Mählmann
- Equine Clinic, General Surgery and Radiology, Freie Universität Berlin, Berlin, Germany
| | - M Bugno-Poniewierska
- Department of Genomics and Molecular Biology of Animals, National Research Institute of Animal Production, Balice, Poland
| |
Collapse
|
94
|
Systematic Identification and Bioinformatic Analysis of MicroRNAs in Response to Infections of Coxsackievirus A16 and Enterovirus 71. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4302470. [PMID: 27843944 PMCID: PMC5098103 DOI: 10.1155/2016/4302470] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
Abstract
Hand, foot, and mouth disease (HFMD), mainly caused by coxsackievirus A16 (CVA16) and enterovirus 71 (EV71) infections, remains a serious public health issue with thousands of newly diagnostic cases each year since 2008 in China. The mechanisms underlying viral infection, however, are elusive to date. In the present study, we systematically investigated the host cellular microRNA (miRNA) expression patterns in response to CVA16 and EV71 infections. Through microarray examination, 27 miRNAs (15 upregulated and 12 downregulated) were found to be coassociated with the replication process of two viruses, while the expression levels of 15 and 5 miRNAs were significantly changed in CVA16- and EV71-infected cells, respectively. A great number of target genes of 27 common differentially expressed miRNAs were predicted by combined use of two computational target prediction algorithms, TargetScan and MiRanda. Comprehensive bioinformatic analysis of target genes in GO categories and KEGG pathways indicated the involvement of diverse biological functions and signaling pathways during viral infection. These results provide an overview of the roles of miRNAs in virus-host interaction, which will contribute to further understanding of HFMD pathological mechanisms.
Collapse
|
95
|
Transcriptome analysis reveals regional and temporal differences in mucosal immune system development in the small intestine of neonatal calves. BMC Genomics 2016; 17:602. [PMID: 27515123 PMCID: PMC4981982 DOI: 10.1186/s12864-016-2957-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 07/20/2016] [Indexed: 02/07/2023] Open
Abstract
Background Postnatal development of the mammalian mucosal immune system is crucial for responding to the rapid colonization by commensal bacteria and possible exposure to pathogens. This study analyzed expression patterns for mRNAs and their relationship with microRNAs (miRNAs) in the bovine small intestine during the critical neonatal period (0 to 42 days). This analysis revealed molecular mechanisms regulating the postnatal development of the intestinal mucosal immune system. Results Small intestine samples (jejunum and ileum) were collected from newborn male, Holstein calves immediately post-partum (n = 3) and at 7 (n = 5), 21 (n = 5), and 42 (n = 5) days of age and the transcriptomes were profiled using RNA-Seq. When analyzing all time points collectively, greater expression of genes encoding the complement functional pathway, as well as lower expression of genes encoding Toll-like receptors and NOD-like receptors were observed in the jejunum when compared to the ileum. In addition, significant changes in the expression of immune-related genes were detected within the first week post-partum in both jejunum and ileum. For example, increased expression of genes encoding tight junction proteins (claudin 1, claudin 4 and occludin), an antimicrobial peptide (Regenerating Islet-Derived 3-γ), NOD-like receptors (NACHT, LRR and PYD domain-containing protein 3), regulatory T cell marker (forkhead box P3), and both anti-inflammatory (interleukin 10) and pro-inflammatory (interleukin 8) cytokines was observed throughout the small intestine of 7-day-old calves when compared to newborn calves. Moreover, the expression of mucosal immune-related genes were either positively or negatively correlated with total bacterial population depending on both intestinal region and age. The integrated analysis of miRNAs and mRNAs supported the conclusion that miRNAs may regulate temporal changes in the expression of genes encoding tight junction proteins (miR-335), cytokines (miR-335) and bacterial recognition (miR-100) during the first week of small intestine development. Conclusion The rapid development of transcriptional differences between jejunum and ileum reveal that these two intestinal regions make distinct contributions to the intestinal mucosal immune system during the early neonatal period. In addition, transcriptome analysis indicates that the first week after birth is a very dynamic developmental period for the intestinal mucosal immune system and these changes may be regulated by both miRNAs and microbial colonization. Findings from this study indicate that a detailed analysis of both the abundance and diversity of the colonizing microbiome may be necessary to understand factors regulating the rapid development of the mucosal immune system during the first week of life. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2957-y) contains supplementary material, which is available to authorized users.
Collapse
|
96
|
Toll-like receptors signaling: A complex network for NF-κB activation in B-cell lymphoid malignancies. Semin Cancer Biol 2016; 39:15-25. [DOI: 10.1016/j.semcancer.2016.07.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 11/17/2022]
|
97
|
Lim YX, Ng YL, Tam JP, Liu DX. Human Coronaviruses: A Review of Virus-Host Interactions. Diseases 2016; 4:E26. [PMID: 28933406 PMCID: PMC5456285 DOI: 10.3390/diseases4030026] [Citation(s) in RCA: 390] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/18/2016] [Accepted: 07/18/2016] [Indexed: 12/19/2022] Open
Abstract
Human coronaviruses (HCoVs) are known respiratory pathogens associated with a range of respiratory outcomes. In the past 14 years, the onset of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV) have thrust HCoVs into spotlight of the research community due to their high pathogenicity in humans. The study of HCoV-host interactions has contributed extensively to our understanding of HCoV pathogenesis. In this review, we discuss some of the recent findings of host cell factors that might be exploited by HCoVs to facilitate their own replication cycle. We also discuss various cellular processes, such as apoptosis, innate immunity, ER stress response, mitogen-activated protein kinase (MAPK) pathway and nuclear factor kappa B (NF-κB) pathway that may be modulated by HCoVs.
Collapse
Affiliation(s)
- Yvonne Xinyi Lim
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Yan Ling Ng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Ding Xiang Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
98
|
Yu D, Wu Y, Xu L, Fan Y, Peng L, Xu M, Yao YG. Identification and characterization of toll-like receptors (TLRs) in the Chinese tree shrew (Tupaia belangeri chinensis). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 60:127-138. [PMID: 26923770 DOI: 10.1016/j.dci.2016.02.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 02/23/2016] [Indexed: 06/05/2023]
Abstract
In mammals, the toll-like receptors (TLRs) play a major role in initiating innate immune responses against pathogens. Comparison of the TLRs in different mammals may help in understanding the TLR-mediated responses and developing of animal models and efficient therapeutic measures for infectious diseases. The Chinese tree shrew (Tupaia belangeri chinensis), a small mammal with a close relationship to primates, is a viable experimental animal for studying viral and bacterial infections. In this study, we characterized the TLRs genes (tTLRs) in the Chinese tree shrew and identified 13 putative TLRs, which are orthologs of mammalian TLR1-TLR9 and TLR11-TLR13, and TLR10 was a pseudogene in tree shrew. Positive selection analyses using the Maximum likelihood (ML) method showed that tTLR8 and tTLR9 were under positive selection, which might be associated with the adaptation to the pathogen challenge. The mRNA expression levels of tTLRs presented an overall low and tissue-specific pattern, and were significantly upregulated upon Hepatitis C virus (HCV) infection. tTLR4 and tTLR9 underwent alternative splicing, which leads to different transcripts. Phylogenetic analysis and TLR structure prediction indicated that tTLRs were evolutionarily conserved, which might reflect an ancient mechanism and structure in the innate immune response system. Taken together, TLRs had both conserved and unique features in the Chinese tree shrew.
Collapse
Affiliation(s)
- Dandan Yu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yong Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Ling Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China
| | - Yu Fan
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Li Peng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Min Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Yong-Gang Yao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China; Kunming Primate Research Center of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
99
|
Medvedev AE, Murphy M, Zhou H, Li X. E3 ubiquitin ligases Pellinos as regulators of pattern recognition receptor signaling and immune responses. Immunol Rev 2016; 266:109-22. [PMID: 26085210 DOI: 10.1111/imr.12298] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pellinos are a family of E3 ubiquitin ligases discovered for their role in catalyzing K63-linked polyubiquitination of Pelle, an interleukin-1 (IL-1) receptor-associated kinase homolog in the Drosophila Toll pathway. Subsequent studies have revealed the central and non-redundant roles of mammalian Pellino-1, Pellino-2, and Pelino-3 in signaling pathways emanating from IL-1 receptors, Toll-like receptors, NOD-like receptors, T- and B-cell receptors. While Pellinos ability to interact with many signaling intermediates suggested their scaffolding roles, recent findings in mice expressing ligase-inactive Pellinos demonstrated the importance of Pellino ubiquitin ligase activity. Cell-specific functions of Pellinos have emerged, e.g. Pellino-1 being a negative regulator in T lymphocytes and a positive regulator in myeloid cells, and details of molecular regulation of receptor signaling by various members of the Pellino family have been revealed. In this review, we summarize current information about Pellino-mediated regulation of signaling by pattern recognition receptors, T-cell and B-cell receptors and tumor necrosis factor receptors, and discuss Pellinos roles in sepsis and infectious diseases, as well as in autoimmune, inflammatory, and allergic disorders. We also provide our perspective on the potential of targeting Pellinos with peptide- or small molecule-based drug compounds as a new therapeutic approach for septic shock and autoimmune pathologies.
Collapse
Affiliation(s)
- Andrei E Medvedev
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Michael Murphy
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Hao Zhou
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Xiaoxia Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
100
|
Stable Toll-Like Receptor 10 Knockdown in THP-1 Cells Reduces TLR-Ligand-Induced Proinflammatory Cytokine Expression. Int J Mol Sci 2016; 17:ijms17060859. [PMID: 27258267 PMCID: PMC4926393 DOI: 10.3390/ijms17060859] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 05/24/2016] [Accepted: 05/26/2016] [Indexed: 12/28/2022] Open
Abstract
Toll-like receptor 10 (TLR10) is the only orphan receptor whose natural ligand and function are unknown among the 10 human TLRs. In this study, to test whether TLR10 recognizes some known TLR ligands, we established a stable TLR10 knockdown human monocytic cell line THP-1 using TLR10 short hairpin RNA lentiviral particle and puromycin selection. Among 60 TLR10 knockdown clones that were derived from each single transduced cell, six clones were randomly selected, and then one of those clones, named E7, was chosen for the functional study. E7 exhibited approximately 50% inhibition of TLR10 mRNA and protein expression. Of all the TLRs, only the expression of TLR10 changed significantly in this cell line. Additionally, phorbol 12-myristate 13-acetate-induced macrophage differentiation of TLR10 knockdown cells was not affected in the knockdown cells. When exposed to TLR ligands, such as synthetic diacylated lipoprotein (FSL-1), lipopolysaccharide (LPS), and flagellin, significant induction of proinflammatory cytokine gene expression including Interleukin-8 (IL-8), Interleukin-1 beta (IL-1β), Tumor necrosis factor-alpha (TNF-α) and Chemokine (C–C Motif) Ligand 20 (CCL20) expression, was found in the control THP-1 cells, whereas the TLR10 knockdown cells exhibited a significant reduction in the expression of IL-8, IL-1β, and CCL20. TNF-α was the only cytokine for which the expression did not decrease in the TLR10 knockdown cells from that measured in the control cells. Analysis of putative binding sites for transcription factors using a binding-site-prediction program revealed that the TNF-α promoter does not have putative binding sites for AP-1 or c-Jun, comprising a major transcription factor along with NF-κB for TLR signaling. Our results suggest that TLR10 is involved in the recognition of FSL-1, LPS, and flagellin and TLR-ligand-induced expression of TNF-α does not depend on TLR10.
Collapse
|