1
|
Khan KN, Guo SW, Ogawa K, Fujishita A, Mori T. The role of innate and adaptive immunity in endometriosis. J Reprod Immunol 2024; 163:104242. [PMID: 38503076 DOI: 10.1016/j.jri.2024.104242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/13/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
The innate and adaptive immune systems are the two key branches that determine host protection at all mucosal surfaces in human body, including the female reproductive tract. The pattern recognition receptors within the host that recognize pathogen-associated molecular patterns are expressed on the cells of the innate immune system. Rapidly reactive, theinnate immune system, responds immediately to the presence of infectious or other non-self agents, thereby launching an inflammatory response to protect the host until the activation of slower adaptive immune system. Macrophages, dendritic cells, and toll-like receptors are integral components of the innate immune system. In contrast, T-helper (Th1/Th2/Th17) cells and regulatory T (Treg) cells are the primary components of adaptive immune system. Studies showed that the growth and progression of endometriosis continue even in unilateral ovariectomized animal suggesting that besides ovarian steroid hormones, the growth of endometriosis could be regulated by innate/adaptive immune systems in pelvic environment. Recent reports demonstrated a potential role of Th1/Th2/Th17/Treg cells either individually or collectively in the initiation, maintenance, and progression of endometriosis. Herewe review the fundamental knowledge of innate and adaptive immunity and elaborate the role of innate and adaptive immunity in endometriosis based on both human and experimental data.
Collapse
Affiliation(s)
- Khaleque N Khan
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.
| | - Sun-Wei Guo
- Shanghai Obstetrics and Gynecology Hospital, Shanghai 200011, China.
| | - Kanae Ogawa
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Akira Fujishita
- Department of Gynecology, Saiseikai Nagasaki Hospital, Nagasaki 850-0003, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
2
|
Chakraborty A, Kamat SS. Lysophosphatidylserine: A Signaling Lipid with Implications in Human Diseases. Chem Rev 2024; 124:5470-5504. [PMID: 38607675 DOI: 10.1021/acs.chemrev.3c00701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Lysophosphatidylserine (lyso-PS) has emerged as yet another important signaling lysophospholipid in mammals, and deregulation in its metabolism has been directly linked to an array of human autoimmune and neurological disorders. It has an indispensable role in several biological processes in humans, and therefore, cellular concentrations of lyso-PS are tightly regulated to ensure optimal signaling and functioning in physiological settings. Given its biological importance, the past two decades have seen an explosion in the available literature toward our understanding of diverse aspects of lyso-PS metabolism and signaling and its association with human diseases. In this Review, we aim to comprehensively summarize different aspects of lyso-PS, such as its structure, biodistribution, chemical synthesis, and SAR studies with some synthetic analogs. From a biochemical perspective, we provide an exhaustive coverage of the diverse biological activities modulated by lyso-PSs, such as its metabolism and the receptors that respond to them in humans. We also briefly discuss the human diseases associated with aberrant lyso-PS metabolism and signaling and posit some future directions that may advance our understanding of lyso-PS-mediated mammalian physiology.
Collapse
Affiliation(s)
- Arnab Chakraborty
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| |
Collapse
|
3
|
Huang Z, Gao Y, Han Y, Yang J, Yang C, Li S, Zhou D, Huang Q, Yang J. Revealing the roles of TLR7, a nucleic acid sensor for COVID-19 in pan-cancer. BIOSAFETY AND HEALTH 2023:S2590-0536(23)00054-X. [PMID: 37362864 PMCID: PMC10167782 DOI: 10.1016/j.bsheal.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 06/28/2023] Open
Abstract
Recent studies suggested that cancer was a risk factor for coronavirus disease 2019 (COVID-19). Toll-like receptor 7 (TLR7), a severe acute respiratory syndrome 2 (SARS-CoV-2) virus's nucleic acid sensor, was discovered to be aberrantly expressed in many types of cancers. However, its expression pattern across cancers and association with COVID-19 (or its causing virus SARS-CoV-2) has not been systematically studied. In this study, we proposed a computational framework to comprehensively study the roles of TLR7 in COVID-19 and pan-cancers at genetic, gene expression, protein, epigenetic, and single-cell levels. We applied the computational framework in a few databases, including The Cancer Genome Atlas (TCGA), The Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), Human Protein Atlas (HPA), lung gene expression data of mice infected with SARS-CoV-2, and the like. As a result, TLR7 expression was found to be higher in the lung of mice infected with SARS-CoV-2 than that in the control group. The analysis in the Opentargets database also confirmed the association between TLR7 and COVID-19. There are also a few exciting findings in cancers. First, the most common type of TLR7 was "Missense" at the genomic level. Second, TLR7 mRNA expression was significantly up-regulated in 6 cancer types and down-regulated in 6 cancer types compared to normal tissues, further validated in the HPA database at the protein level. The genes significantly co-expressed with TLR7 were mainly enriched in the toll-like receptor signaling pathway, endolysosome, and signaling pattern recognition receptor activity. In addition, the abnormal TLR7 expression was associated with mismatch repair (MMR), microsatellite instability (MSI), and tumor mutational burden (TMB) in various cancers. Mined by the ESTIMATE algorithm, the expression of TLR7 was also closely linked to various immune infiltration patterns in pan-cancer, and TLR7 was mainly enriched in macrophages, as revealed by single-cell RNA sequencing. Third, abnormal expression of TLR7 could predict the survival of Brain Lower Grade Glioma (LGG), Lung adenocarcinoma (LUAD), Skin Cutaneous Melanoma (SKCM), Stomach adenocarcinoma (STAD), and Testicular Germ Cell Tumors (TGCT) patients, respectively. Finally, TLR7 expressions were very sensitive to a few targeted drugs, such as Alectinib and Imiquimod. In conclusion, TLR7 might be essential in the pathogenesis of COVID-19 and cancers.
Collapse
Affiliation(s)
- Zhijian Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yaoxin Gao
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuanyuan Han
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650000, China
| | - Jingwen Yang
- Department of Clinical Pharmacy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Can Yang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Shixiong Li
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Decong Zhou
- Geriatric Hospital of Hainan Medical Education Department, Haikou 571100, China
| | - Qiuyan Huang
- Department of Breast Surgical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd, Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| |
Collapse
|
4
|
Qi K, Yi X, Wang M, Wang J, Sun H, Liang P, Xu J, Zheng H. Streptococcus parasuis, an Emerging Zoonotic Pathogen, Possesses the Capacity to Induce Cerebral Inflammatory Responses. Pathogens 2023; 12:pathogens12040600. [PMID: 37111486 PMCID: PMC10141694 DOI: 10.3390/pathogens12040600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/17/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
To date, three Streptococcus parasuis strains, BS26, BS27, and NN1, have been isolated from the blood cultures of patients with peritonitis, pneumonia, and arthritis, indicating that S. parasuis is an emerging threat to susceptible people. There is thus an urgent need to further evaluate the pathogenesis of S. parasuis clinical strains in order to design efficient anti-inflammatory strategies. Our previous study demonstrated the capacity of S. parasuis clinical strains to enter the central nervous system (CNS) of infected mice. However, the characteristics and inflammatory mechanism of CNS infections caused by S. parasuis are still non-available. In the present study, we investigated the proportion and time of two clinical S. parasuis strains NN1 and BS26 infected mice that developed neurological symptoms. The characteristics of histopathological changes and the cerebral immune response in mice with neurological symptoms were analyzed. Furthermore, we evaluated the roles of microglia and astrocytes in the S. parasuis clinical strain-induced cerebral inflammation. Our data indicated that S. parasuis clinical strains possess a high potential to induce cerebral inflammation in susceptible people at the early phase of infection. Our study contributes to increasing the understanding of the pathogenicity of S. parasuis and the inflammatory mechanisms of the brain against infection caused by S. parasuis.
Collapse
Affiliation(s)
- Kexin Qi
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xueli Yi
- Center for Clinical Laboratory Diagnosis and Research, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Mingliu Wang
- Guangxi Zhuang Autonomous Region Center for Disease Prevention and Control, Nanning 530021, China
| | - Jianping Wang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Hui Sun
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Pujun Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin 541002, China
| | - Jianguo Xu
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Han Zheng
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
5
|
Abstract
Succinate is a circulating metabolite, and the relationship between abnormal changes in the physiological concentration of succinate and inflammatory diseases caused by the overreaction of certain immune cells has become a research focus. Recent investigations have shown that succinate produced by the gut microbiota has the potential to regulate host homeostasis and treat diseases such as inflammation. Gut microbes are important for maintaining intestinal homeostasis. Microbial metabolites serve as nutrients in energy metabolism, and act as signal molecules that stimulate host cell and organ function and affect the structural balance between symbiotic gut microorganisms. This review focuses on succinate as a metabolite of both host cells and gut microbes and its involvement in regulating the gut - immune tissue axis by activating intestinal mucosal cells, including macrophages, dendritic cells, and intestinal epithelial cells. We also examined its role as the mediator of microbiota - host crosstalk and its potential function in regulating intestinal microbiota homeostasis. This review explores feasible ways to moderate succinate levels and provides new insights into succinate as a potential target for microbial therapeutics for humans.
Collapse
Affiliation(s)
- Yi-Han Wei
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiang-Chao Zhao
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR, USA
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University, Guangdong Laboratory for Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangzhou, China
| |
Collapse
|
6
|
Shahinuzzaman ADA, Kamal AHM, Chakrabarty JK, Rahman A, Chowdhury SM. Identification of Inflammatory Proteomics Networks of Toll-like Receptor 4 through Immunoprecipitation-Based Chemical Cross-Linking Proteomics. Proteomes 2022; 10:proteomes10030031. [PMID: 36136309 PMCID: PMC9506174 DOI: 10.3390/proteomes10030031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/14/2022] [Accepted: 08/20/2022] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptor 4 (TLR4) is a receptor on an immune cell that can recognize the invasion of bacteria through their attachment with bacterial lipopolysaccharides (LPS). Hence, LPS is a pro-immune response stimulus. On the other hand, statins are lipid-lowering drugs and can also lower immune cell responses. We used human embryonic kidney (HEK 293) cells engineered to express HA-tagged TLR-4 upon treatment with LPS, statin, and both statin and LPS to understand the effect of pro- and anti-inflammatory responses. We performed a monoclonal antibody (mAb) directed co-immunoprecipitation (CO-IP) of HA-tagged TLR4 and its interacting proteins in the HEK 293 extracted proteins. We utilized an ETD cleavable chemical cross-linker to capture weak and transient interactions with TLR4 protein. We tryptic digested immunoprecipitated and cross-linked proteins on beads, followed by liquid chromatography–mass spectrometry (LC-MS/MS) analysis of the peptides. Thus, we utilized the label-free quantitation technique to measure the relative expression of proteins between treated and untreated samples. We identified 712 proteins across treated and untreated samples and performed protein network analysis using Ingenuity Pathway Analysis (IPA) software to reveal their protein networks. After filtering and evaluating protein expression, we identified macrophage myristoylated alanine-rich C kinase substrate (MARCKSL1) and creatine kinase proteins as a potential part of the inflammatory networks of TLR4. The results assumed that MARCKSL1 and creatine kinase proteins might be associated with a statin-induced anti-inflammatory response due to possible interaction with the TLR4.
Collapse
Affiliation(s)
- A. D. A. Shahinuzzaman
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Pharmaceutical Sciences Research Division, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Abu Hena Mostafa Kamal
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Advanced Technology Cores, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jayanta K. Chakrabarty
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Quantitative Proteomics and Metabolomics Center, Columbia University, New York, NY 10027, USA
| | - Aurchie Rahman
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Saiful M. Chowdhury
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX 76019, USA
- Correspondence: ; Tel.: +1-817-272-5439
| |
Collapse
|
7
|
Al-Khatib S, Shabaneh A, Abdo N, AL-Eitan L, Al-Mistarehi AH, Khader Y. Association of TLR9-1237T>C; rs5743836 polymorphism with increased risk of Hodgkin’s lymphoma: A case-control study. PLoS One 2022; 17:e0272312. [PMID: 35905120 PMCID: PMC9337659 DOI: 10.1371/journal.pone.0272312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022] Open
Abstract
Mature B-cell neoplasms are typically divided into Hodgkin and Non-Hodgkin Lymphomas. Hodgkin Lymphoma is characterized by the neoplastic Reed-Sternberg cells, usually harbored in an inflammatory background, with a frequent clinical presentation of mediastinal lymphadenopathy. Many studies link between autoimmunity and lymphomagenesis, a large proportion of these studies evidently trace the pathogenesis back to the misdirected detection of self-derived nucleic acids by Toll-Like Receptors (TLRs), especially those of the intracellular type. In this study, we analyzed the relationship between a selected SNP in TLR9 (TLR9-1237T>C; rs5743836) and the risk and overall survival of HL patients in a Jordanian Arab population. A total of 374 subjects; 136 cases of Hodgkin lymphoma and 238 matched healthy controls were incorporated in this study. Genomic DNA was extracted from formalin-fixed paraffin-embedded tissues. Genotyping of the genetic polymorphisms was conducted using a sequencing protocol. The results show a statistically significant higher distribution of the rs5743836 (TLR9-1237T>C) allele among the case population, with a p-value of 0.031 (<0.05). This distribution proved significant when studied in the codominant (only significant in the T/C genotype, p-value = 0.030), dominant (p-value = 0.025), and overdominant (p-value = 0.035) models. None of the models showed any statistically significant difference in survival associated with the rs5743836 (TLR9-1237T>C) SNP.
Collapse
Affiliation(s)
- Sohaib Al-Khatib
- Faculty of Medicine, Department of Pathology and Laboratory Medicine, Jordan University of Science and Technology, Irbid, Jordan
- * E-mail:
| | - Amin Shabaneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Nour Abdo
- Faculty of Medicine, Department of Public Health, Jordan University of Science and Technology, Irbid, Jordan
| | - Laith AL-Eitan
- Faculty of Science and Arts, Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Abdel-Hameed Al-Mistarehi
- Faculty of Medicine, Department of Family Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Yousef Khader
- Faculty of Medicine, Department of Public Health and Community Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Chen RX, Dai MD, Zhang QZ, Lu MP, Wang ML, Yin M, Zhu XJ, Wu ZF, Zhang ZD, Cheng L. TLR Signaling Pathway Gene Polymorphisms, Gene-Gene and Gene-Environment Interactions in Allergic Rhinitis. J Inflamm Res 2022; 15:3613-3630. [PMID: 35769128 PMCID: PMC9234183 DOI: 10.2147/jir.s364877] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/04/2022] [Indexed: 01/01/2023] Open
Abstract
Background Allergic rhinitis (AR) is a nasal inflammatory disease resulting from a complex interplay between genetic and environmental factors. The association between Toll-like receptor (TLR) signaling pathway and environmental factors in AR pathogenesis remains to be explored. This study aims to assess the genetic association of AR with single nucleotide polymorphisms (SNPs) in TLR signaling pathway, and investigate the roles of gene–gene and gene–environment interactions in AR. Methods A total of 452 AR patients and 495 healthy controls from eastern China were enrolled in this hospital-based case–control study. We evaluated putatively functional genetic polymorphisms in TLR2, TLR4 and CD14 genes for their association with susceptibility to AR and related clinical phenotypes. Interactions between environmental factors (such as traffic pollution, residence, pet keeping) and polymorphisms with AR were examined using logistic regression. Models were stratified by genotype and interaction terms, and tested for the significance of gene–gene and gene–environment interactions. Results In the single-locus analysis, two SNPs in CD14, rs2563298 (A/C) and rs2569191 (C/T) were associated with a significantly decreased risk of AR. Compared with the GG genotype, the GT and GT/TT genotypes of TLR2 rs7656411 (G/T) were associated with a significantly increased risk of AR. Gene–gene interactions (eg, TLR2 rs7656411, TLR4 rs1927914, and CD14 rs2563298) was associated with AR. Gene–environment interactions (eg, TLR4 or CD14 polymorphisms and certain environmental exposures) were found in AR cases, but they were not significant after Bonferroni correction. Conclusion The genetic polymorphisms of TLR2 and CD14 and gene–gene interactions in TLR signaling pathway were associated with susceptibility to AR in this Han Chinese population. However, the present results were limited to support the association between gene–environment interactions and AR.
Collapse
Affiliation(s)
- Ruo-Xi Chen
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Meng-Di Dai
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qing-Zhao Zhang
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Mei-Ping Lu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Mei-Lin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Min Yin
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.,International Centre for Allergy Research, Nanjing Medical University, Nanjing, People's Republic of China
| | - Xin-Jie Zhu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zhong-Fei Wu
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China
| | - Zheng-Dong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lei Cheng
- Department of Otorhinolaryngology & Clinical Allergy Center, The First Affiliated Hospital, Nanjing Medical University, Nanjing, People's Republic of China.,International Centre for Allergy Research, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
9
|
Farahani M, Niknam Z, Mohammadi Amirabad L, Amiri-Dashatan N, Koushki M, Nemati M, Danesh Pouya F, Rezaei-Tavirani M, Rasmi Y, Tayebi L. Molecular pathways involved in COVID-19 and potential pathway-based therapeutic targets. Biomed Pharmacother 2022; 145:112420. [PMID: 34801852 PMCID: PMC8585639 DOI: 10.1016/j.biopha.2021.112420] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 01/08/2023] Open
Abstract
Deciphering the molecular downstream consequences of severe acute respiratory syndrome coronavirus (SARS-CoV)- 2 infection is important for a greater understanding of the disease and treatment planning. Furthermore, greater understanding of the underlying mechanisms of diagnostic and therapeutic strategies can help in the development of vaccines and drugs against COVID-19. At present, the molecular mechanisms of SARS-CoV-2 in the host cells are not sufficiently comprehended. Some of the mechanisms are proposed considering the existing similarities between SARS-CoV-2 and the other members of the β-CoVs, and others are explained based on studies advanced in the structure and function of SARS-CoV-2. In this review, we endeavored to map the possible mechanisms of the host response following SARS-CoV-2 infection and surveyed current research conducted by in vitro, in vivo and human observations, as well as existing suggestions. We addressed the specific signaling events that can cause cytokine storm and demonstrated three forms of cell death signaling following virus infection, including apoptosis, pyroptosis, and necroptosis. Given the elicited signaling pathways, we introduced possible pathway-based therapeutic targets; ADAM17 was especially highlighted as one of the most important elements of several signaling pathways involved in the immunopathogenesis of COVID-19. We also provided the possible drug candidates against these targets. Moreover, the cytokine-cytokine receptor interaction pathway was found as one of the important cross-talk pathways through a pathway-pathway interaction analysis for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Masoumeh Farahani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Niknam
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Nasrin Amiri-Dashatan
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehdi Koushki
- Department of Clinical Biochemistry, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohadeseh Nemati
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Fahima Danesh Pouya
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran.
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
10
|
Potential value of pharmacological agents acting on toll-like receptor (TLR) 7 and/or TLR8 in COVID-19. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100068. [PMID: 34870161 PMCID: PMC8562070 DOI: 10.1016/j.crphar.2021.100068] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 10/24/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic is an ongoing global pandemic of coronavirus disease 2019 as an atypical type of viral pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Many potential pharmacotherapies are currently being investigated against this disease. This article points to and justifies, the importance of investigating the potential therapeutic value of pharmacological agents acting on Toll-like Receptor (TLR) 7 and/or TLR8 as double-edged swords combating COVID-19. Induction of TLR7 and/or TLR8 may be investigated as a strategy to stimulate immunity and may be added to anti-COVID19 vaccines to cope with their current viral escape challenge. TLR7 stimulation may not only help viral clearance through Th1 antiviral responses but may also provide beneficial broncho- and vaso-dilatory, as well as, anti-inflammatory effects. Pharmacological compounds acting as TLR7 and/or TLR8 agonists may be of value if used by frontline healthcare workers with comorbidities who demonstrate mild symptoms of the disease. On the other hand, TLR7 and/or TLR8 antagonists may be used in combination with immune-modulatory/anti-inflammatory drugs in severe cases of the disease, with potential synergistic effects that could also help in reducing the doses of such therapies, and consequently their adverse effects. There is potential value in development of TLR7/8 agonists/antagonists as double-edged swords combating COVID-19. TLR7/8 agonists, as immune-stimulants in mild disease, may also be added to preventive vaccines acting against viral escape. TLR7/8 antagonists may be combined with immune-suppressants in severe cases for potential synergism and minimized toxicity.
Collapse
|
11
|
Reens AL, Cabral DJ, Liang X, Norton JE, Therien AG, Hazuda DJ, Swaminathan G. Immunomodulation by the Commensal Microbiome During Immune-Targeted Interventions: Focus on Cancer Immune Checkpoint Inhibitor Therapy and Vaccination. Front Immunol 2021; 12:643255. [PMID: 34054810 PMCID: PMC8155485 DOI: 10.3389/fimmu.2021.643255] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence in clinical and preclinical studies indicates that success of immunotherapies can be impacted by the state of the microbiome. Understanding the role of the microbiome during immune-targeted interventions could help us understand heterogeneity of treatment success, predict outcomes, and develop additional strategies to improve efficacy. In this review, we discuss key studies that reveal reciprocal interactions between the microbiome, the immune system, and the outcome of immune interventions. We focus on cancer immune checkpoint inhibitor treatment and vaccination as two crucial therapeutic areas with strong potential for immunomodulation by the microbiota. By juxtaposing studies across both therapeutic areas, we highlight three factors prominently involved in microbial immunomodulation: short-chain fatty acids, microbe-associate molecular patterns (MAMPs), and inflammatory cytokines. Continued interrogation of these models and pathways may reveal critical mechanistic synergies between the microbiome and the immune system, resulting in novel approaches designed to influence the efficacy of immune-targeted interventions.
Collapse
Affiliation(s)
- Abigail L. Reens
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Damien J. Cabral
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Xue Liang
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - James E. Norton
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Alex G. Therien
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| | - Daria J. Hazuda
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
- Infectious Disease and Vaccine Research, Merck & Co., Inc., West Point, PA, United States
| | - Gokul Swaminathan
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, United States
| |
Collapse
|
12
|
Phenotypic Features of Mesenchymal Stem Cell Subpopulations Obtained under the Influence of Various Toll-Like Receptors Ligands. Bull Exp Biol Med 2021; 170:555-559. [PMID: 33725247 DOI: 10.1007/s10517-021-05105-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Indexed: 01/05/2023]
Abstract
The paper characterizes phenotypic features of subpopulations of human adipose tissue mesenchymal stem cells (MSC) resulting from exposure to Toll-like receptors ligands. MSC1 and MSC2 phenotypes were induced by exposure to LPS and polyinosinic-polycytidylic acid, respectively. Different exposures to Toll-like receptors ligands, short-term (3 h) and long-term (24 h), were studied. The cytokine profile of cells with the MSC2 phenotype differed depending on the duration of exposure to the inductor, while the cytokine profile of cells with the MSC1 phenotype remained stable. Morphometric features of mesenchymal stem cells with the MSC1 and MSC2 phenotypes, as well as differences in the IDO gene expression were identified. These differences can be used to distinguish between these cell types.
Collapse
|
13
|
Díaz-Carballo D, Saka S, Acikelli AH, Homp E, Erwes J, Demmig R, Klein J, Schröer K, Malak S, D'Souza F, Noa-Bolaño A, Menze S, Pano E, Andrioff S, Teipel M, Dammann P, Klein D, Nasreen A, Tannapfel A, Grandi N, Tramontano E, Ochsenfarth C, Strumberg D. Enhanced antitumoral activity of TLR7 agonists via activation of human endogenous retroviruses by HDAC inhibitors. Commun Biol 2021; 4:276. [PMID: 33658617 PMCID: PMC7930250 DOI: 10.1038/s42003-021-01800-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 01/25/2021] [Indexed: 12/18/2022] Open
Abstract
In this work, we are reporting that "Shock and Kill", a therapeutic approach designed to eliminate latent HIV from cell reservoirs, is extrapolatable to cancer therapy. This is based on the observation that malignant cells express a spectrum of human endogenous retroviral elements (HERVs) which can be transcriptionally boosted by HDAC inhibitors. The endoretroviral gene HERV-V2 codes for an envelope protein, which resembles syncytins. It is significantly overexpressed upon exposure to HDAC inhibitors and can be effectively targeted by simultaneous application of TLR7/8 agonists, triggering intrinsic apoptosis. We demonstrated that this synergistic cytotoxic effect was accompanied by the functional disruption of the TLR7/8-NFκB, Akt/PKB, and Ras-MEK-ERK signalling pathways. CRISPR/Cas9 ablation of TLR7 and HERV-V1/V2 curtailed apoptosis significantly, proving the pivotal role of these elements in driving cell death. The effectiveness of this new approach was confirmed in ovarian tumour xenograft studies, revealing a promising avenue for future cancer therapies.
Collapse
Affiliation(s)
- David Díaz-Carballo
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany.
| | - Sahitya Saka
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Ali H Acikelli
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Ekaterina Homp
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Julia Erwes
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Rebecca Demmig
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Jacqueline Klein
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Katrin Schröer
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Sascha Malak
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Flevy D'Souza
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Adrien Noa-Bolaño
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Saskia Menze
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Emilio Pano
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Swetlana Andrioff
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Marc Teipel
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| | - Philip Dammann
- Central Animal Laboratory, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Diana Klein
- Institute of Cell Biology, Cancer Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Amber Nasreen
- Visceral Surgery Department, Marien Hospital Herne, Ruhr University Bochum Medical School, Herne, Germany
| | | | - Nicole Grandi
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Enzo Tramontano
- Department of Life and Environmental Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, Italy
| | - Crista Ochsenfarth
- Department of Anesthesia, Intensive Care, Pain and Palliative Medicine, Marien Hospital Herne, Ruhr-University Bochum Medical School, Herne, Germany
| | - Dirk Strumberg
- Ruhr University Bochum, Faculty of Medicine, Department of Haematology and Oncology, Institute of Molecular Oncology and Experimental Therapeutics, Marien Hospital Herne, Herne, Germany
| |
Collapse
|
14
|
Kimani FW, Ajit J, Galluppi A, Manna S, Howitz WJ, Tang S, Esser-Kahn AP. Receptor-Ligand Kinetics Influence the Mechanism of Action of Covalently Linked TLR Ligands. ACS Chem Biol 2021; 16:380-388. [PMID: 33523635 DOI: 10.1021/acschembio.0c00924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We report a mechanistic study comparing the immune activation of conjugated Toll-like receptor (TLR) agonists and their unlinked mixtures. Herein, we synthesized a set of six linked dual agonists with different ligands, molecular structures, receptor locations, and biophysical characteristics. With these dimers, we ran a series of in vitro cell-based assays, comparing initial and overall NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation, cytokine expression profiles, as well as time-resolved TNF-α (Tumor Necrosis Factor alpha) expression. We show that initial activation kinetics, ligand specificity, and the dose of the agonist influence the activity of these linked TLR systems. These results can help improve vaccine design by showing how linked TLR agonists can improve their potency with the appropriate selection of key criteria.
Collapse
Affiliation(s)
- Flora W. Kimani
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Jainu Ajit
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Alexander Galluppi
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Saikat Manna
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - William J. Howitz
- Department of Chemistry, University of California, Irvine, Irvine, California 92697-2025, United States
| | - Sophia Tang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
15
|
Lambert C, Zappia J, Sanchez C, Florin A, Dubuc JE, Henrotin Y. The Damage-Associated Molecular Patterns (DAMPs) as Potential Targets to Treat Osteoarthritis: Perspectives From a Review of the Literature. Front Med (Lausanne) 2021; 7:607186. [PMID: 33537330 PMCID: PMC7847938 DOI: 10.3389/fmed.2020.607186] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022] Open
Abstract
During the osteoarthritis (OA) process, activation of immune systems, whether innate or adaptive, is strongly associated with low-grade systemic inflammation. This process is initiated and driven in the synovial membrane, especially by synovium cells, themselves previously activated by damage-associated molecular patterns (DAMPs) released during cartilage degradation. These fragments exert their biological activities through pattern recognition receptors (PRRs) that, as a consequence, induce the activation of signaling pathways and beyond the release of inflammatory mediators, the latter contributing to the vicious cycle between cartilage and synovial membrane. The primary endpoint of this review is to provide the reader with an overview of these many molecules categorized as DAMPs and the contribution of the latter to the pathophysiology of OA. We will also discuss the different strategies to control their effects. We are convinced that a better understanding of DAMPs, their receptors, and associated pathological mechanisms represents a decisive issue for degenerative joint diseases such as OA.
Collapse
Affiliation(s)
- Cécile Lambert
- MusculoSKeletal Innovative Research Lab, University of Liège, Institute of Pathology, CHU Sart-Tilman, Liège, Belgium
| | - Jérémie Zappia
- MusculoSKeletal Innovative Research Lab, University of Liège, Institute of Pathology, CHU Sart-Tilman, Liège, Belgium
| | - Christelle Sanchez
- MusculoSKeletal Innovative Research Lab, University of Liège, Institute of Pathology, CHU Sart-Tilman, Liège, Belgium
| | - Antoine Florin
- MusculoSKeletal Innovative Research Lab, University of Liège, Institute of Pathology, CHU Sart-Tilman, Liège, Belgium
| | - Jean-Emile Dubuc
- Orthopaedic Department, University Clinics St. Luc, Brussels, Belgium
| | - Yves Henrotin
- MusculoSKeletal Innovative Research Lab, University of Liège, Institute of Pathology, CHU Sart-Tilman, Liège, Belgium.,Physical Therapy and Rehabilitation Department, Princess Paola Hospital, Vivalia, Marche-en-Famenne, Belgium
| |
Collapse
|
16
|
Francis M, Gopinathan G, Salapatas A, Nares S, Gonzalez M, Diekwisch T, Luan X. SETD1 and NF-κB Regulate Periodontal Inflammation through H3K4 Trimethylation. J Dent Res 2020; 99:1486-1493. [PMID: 32762504 PMCID: PMC7684838 DOI: 10.1177/0022034520939029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The inflammatory response to periodontal pathogens is dynamically controlled by the chromatin state on inflammatory gene promoters. In the present study, we have focused on the effect of the methyltransferase SETD1B on histone H3 lysine K4 (H3K4) histone trimethylation on inflammatory gene promoters. Experiments were based on 3 model systems: 1) an in vitro periodontal ligament (PDL) cell culture model for the study of SETD1 function as it relates to histone methylation and inflammatory gene expression using Porphyromonas gingivalis lipopolysaccharide (LPS) as a pathogen, 2) a subcutaneous implantation model to determine the relationship between SETD1 and nuclear factor κB (NF-κB) through its activation inhibitor BOT-64, and 3) a mouse periodontitis model to test whether the NF-κB activation inhibitor BOT-64 reverses the inflammatory tissue destruction associated with periodontal disease. In our PDL progenitor cell culture model, P. gingivalis LPS increased H3K4me3 histone methylation on IL-1β, IL-6, and MMP2 gene promoters, while SETD1B inhibition decreased H3K4me3 enrichment and inflammatory gene expression in LPS-treated PDL cells. LPS also increased SETD1 nuclear localization in a p65-dependent fashion and the nuclear translocation of p65 as mediated through SETD1, suggestive of a synergistic effect between SETD1 and p65 in the modulation of inflammation. Confirming the role of SETD1 in p65-mediated periodontal inflammation, BOT-64 reduced the number of SETD1-positive cells in inflamed periodontal tissues, restored periodontal tissue integrity, and enhanced osteogenesis in a periodontal inflammation model in vivo. Together, these results have established the histone lysine methyltransferase SETD1 as a key factor in the opening of the chromatin on inflammatory gene promoters through histone H3K4 trimethylation. Our studies also confirmed the role of BOT-64 as a potent molecular therapeutic for the restoration of periodontal health through the inhibition of NF-κB activity and the amelioration of SETD1-induced chromatin relaxation.
Collapse
Affiliation(s)
- M. Francis
- Department of Oral Biology, UIC College of Dentistry, Chicago, IL, USA
| | - G. Gopinathan
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - A. Salapatas
- Department of Oral Biology, UIC College of Dentistry, Chicago, IL, USA
| | - S. Nares
- Department of Periodontics, UIC College of Dentistry, Chicago, IL, USA
| | - M. Gonzalez
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - T.G.H. Diekwisch
- Department of Oral Biology, UIC College of Dentistry, Chicago, IL, USA
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - X. Luan
- Department of Oral Biology, UIC College of Dentistry, Chicago, IL, USA
- Department of Periodontics and Center for Craniofacial Research and Diagnosis, Texas A&M University College of Dentistry, Dallas, TX, USA
| |
Collapse
|
17
|
Cappelletti M, Doll JR, Stankiewicz TE, Lawson MJ, Sauer V, Wen B, Kalinichenko VV, Sun X, Tilburgs T, Divanovic S. Maternal regulation of inflammatory cues is required for induction of preterm birth. JCI Insight 2020; 5:138812. [PMID: 33208552 PMCID: PMC7710297 DOI: 10.1172/jci.insight.138812] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Infection-driven inflammation in pregnancy is a major cause of spontaneous preterm birth (PTB). Both systemic infection and bacterial ascension through the vagina/cervix to the amniotic cavity are strongly associated with PTB. However, the contribution of maternal or fetal inflammatory responses in the context of systemic or localized models of infection-driven PTB is not well defined. Here, using intraperitoneal or intraamniotic LPS challenge, we examined the necessity and sufficiency of maternal and fetal Toll-like receptor (TLR) 4 signaling in induction of inflammatory vigor and PTB. Both systemic and local LPS challenge promoted induction of inflammatory pathways in uteroplacental tissues and induced PTB. Restriction of TLR4 expression to the maternal compartment was sufficient for induction of LPS-driven PTB in either systemic or intraamniotic challenge models. In contrast, restriction of TLR4 expression to the fetal compartment failed to induce LPS-driven PTB. Vav1-Cre-mediated genetic deletion of TLR4 suggested a critical role for maternal immune cells in inflammation-driven PTB. Further, passive transfer of WT in vitro-derived macrophages and dendritic cells to TLR4-null gravid females was sufficient to induce an inflammatory response and drive PTB. Cumulatively, these findings highlight the critical role for maternal regulation of inflammatory cues in induction of inflammation-driven parturition.
Collapse
Affiliation(s)
- Monica Cappelletti
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jessica R. Doll
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Traci E. Stankiewicz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Matthew J. Lawson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Vivien Sauer
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Bingqiang Wen
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Lung Regenerative Medicine
| | - Vladimir V. Kalinichenko
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Lung Regenerative Medicine
| | | | - Tamara Tilburgs
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Center for Inflammation and Tolerance, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
18
|
Iqbal MS, Sardar N, Akmal W, Sultan R, Abdullah H, Qindeel M, Dhama K, Bilal M. ROLE OF TOLL-LIKE RECEPTORS IN CORONAVIRUS INFECTION AND IMMUNE RESPONSE. ACTA ACUST UNITED AC 2020. [DOI: 10.18006/2020.8(spl-1-sars-cov-2).s66.s78] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The emergence of a novel coronavirus referred to as SARS-CoV-2 has become a global health apprehension due to rapid transmission tendency, severity, and wide geographical spread. This emergence was started from Wuhan, China in 2019 from the zoonotic source and spread worldwide, infecting almost half of the community on this earth. Many of the receptors are involved in proceeding with this infection in the organism's body. Toll-like receptors (TLRs) play essential and protective functions from a wide range of microbial pathogens. Small setup of TLR adaptor proteins leads to activate nuclear factor kappa B (NF-kB) and interferon-regulatory factor (IRF). Consequently, various advanced inflammatory cytokines, chemokines, and interferon reaction properties can be up-regulated. Similarly, TLR flagging works on autophagy in macrophages. Autophagy is a cell response to starvation that helps to eliminate damaged cytosol organelles and persistent proteins. It is also able to prevent the replication of intracellular pathogens. Several microbes subvert the autophagy pathways to sustain their viability. This review investigates how TLRs can modulate a macrophagic system and analyze the role of natural resistance autophagy.
Collapse
|
19
|
Using the Past to Maximize the Success Probability of Future Anti-Viral Vaccines. Vaccines (Basel) 2020; 8:vaccines8040566. [PMID: 33019507 PMCID: PMC7712378 DOI: 10.3390/vaccines8040566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Rapid obtaining of safe, effective, anti-viral vaccines has recently risen to the top of the international agenda. To maximize the success probability of future anti-viral vaccines, the anti-viral vaccines successful in the past are summarized here by virus type and vaccine type. The primary focus is on viruses with both single-stranded RNA genomes and a membrane envelope, given the pandemic past of influenza viruses and coronaviruses. The following conclusion is reached, assuming that success of future strategies is positively correlated with strategies successful in the past. The primary strategy, especially for emerging pandemic viruses, should be development of vaccine antigens that are live-attenuated viruses; the secondary strategy should be development of vaccine antigens that are inactivated virus particles. Support for this conclusion comes from the complexity of immune systems. These conclusions imply the need for a revision in current strategic planning.
Collapse
|
20
|
Gopallawa I, Lee RJ. Targeting the phosphoinositide-3-kinase/protein kinase B pathway in airway innate immunity. World J Biol Chem 2020; 11:30-51. [PMID: 33024516 PMCID: PMC7520643 DOI: 10.4331/wjbc.v11.i2.30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
The airway innate immune system maintains the first line of defense against respiratory infections. The airway epithelium and associated immune cells protect the respiratory system from inhaled foreign organisms. These cells sense pathogens via activation of receptors like toll-like receptors and taste family 2 receptors (T2Rs) and respond by producing antimicrobials, inflammatory cytokines, and chemokines. Coordinated regulation of fluid secretion and ciliary beating facilitates clearance of pathogens via mucociliary transport. Airway cells also secrete antimicrobial peptides and radicals to directly kill microorganisms and inactivate viruses. The phosphoinositide-3-kinase/protein kinase B (Akt) kinase pathway regulates multiple cellular targets that modulate cell survival and proliferation. Akt also regulates proteins involved in innate immune pathways. Akt phosphorylates endothelial nitric oxide synthase (eNOS) enzymes expressed in airway epithelial cells. Activation of eNOS can have anti-inflammatory, anti-bacterial, and anti-viral roles. Moreover, Akt can increase the activity of the transcription factor nuclear factor erythroid 2 related factor-2 that protects cells from oxidative stress and may limit inflammation. In this review, we summarize the recent findings of non-cancerous functions of Akt signaling in airway innate host defense mechanisms, including an overview of several known downstream targets of Akt involved in innate immunity.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Department of Otorhinolaryngology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Robert J Lee
- Department of Otorhinolaryngology and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
21
|
Vyas N, Kurian SJ, Bagchi D, Manu MK, Saravu K, Unnikrishnan MK, Mukhopadhyay C, Rao M, Miraj SS. Vitamin D in Prevention and Treatment of COVID-19: Current Perspective and Future Prospects. J Am Coll Nutr 2020; 40:632-645. [PMID: 32870735 DOI: 10.1080/07315724.2020.1806758] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Vitamin D deficiency (VDD) partly explains geographical differences in COVID-19 susceptibility, severity, and mortality. VDD among African-Americans, diabetics, hypertensive, and aged populations possibly explain the higher death rate, aggravated by cocooning. Vitamin D is pleiotropic, mediating bone metabolism, calcium homeostasis, and immune functions, whereas VDD is associated with inflammatory reactions and immune dysfunction, predisposing individuals to severe infections. Vitamin D modulates innate and adaptive immunity via the expression of genes that code antimicrobial peptides (AMPs). And the expression of cluster of differentiation (CD)14, the co-receptor for epidermal toll-like receptor (TLR)4. AMPs stimulate TLR2 in macrophages, increasing the conversion of vitamin D into its active form by cytochrome P450 27B1. Antiviral properties of vitamin D-induced AMPs can shift the polarization of the adaptive immune response from helper T cells (Th)1 to the more regulatory Th2 responses that suppress immune over-reactivity by preventing cytokine storm, which is already demonstrated during the Spanish flu episode. Vitamin D induces antiviral effects by both direct and indirect mechanisms via AMPs, immunomodulation, the interplay between major cellular and viral elements, induction of autophagy and apoptosis, variation of genetic and epigenetic factors. The crosstalk between vitamin D and intracellular signaling pathways may operate as a primary regulatory action on viral gene transcription. VDD may increase the likelihood of infection with enveloped viruses, including retrovirus, hepatitis, and dengue. Global data correlates severe VDD with COVID-19 associated coagulopathy, disrupted immune response and mortality, reduced platelet count, and prolonged prothrombin time, suggesting benefits from supplementation.Key teaching pointsVitamin D induces antiviral effects by direct and indirect mechanisms via AMPs, immunomodulation, induction of autophagy, etc.Epidemiology of VDD partly explains geographical differences in COVID-19 susceptibility, severity, and mortality.Global data correlates severe VDD with COVID-19 associated coagulopathy, disrupted immune response and mortality, reduced platelet count, and prolonged prothrombin time, together suggesting benefits from supplementation.Many clinical trials are underway globally to delineate the role of vitamin D in both prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Navya Vyas
- Department of Health Policy, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shilia Jacob Kurian
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Debasis Bagchi
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Mohan K Manu
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Department of Respiratory Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kavitha Saravu
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Department of Infectious Diseases, Kasturba Medical College and Hospital, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | | - Chiranjay Mukhopadhyay
- Kasturba Medical College and Hospital, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sonal Sekhar Miraj
- Manipal Center for Infectious Diseases, Prasanna School of Public Health, Manipal Academy of Higher Education, Manipal, Karnataka, India.,Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
22
|
Role of Innate Immune Receptor TLR4 and its endogenous ligands in epileptogenesis. Pharmacol Res 2020; 160:105172. [PMID: 32871246 DOI: 10.1016/j.phrs.2020.105172] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 12/22/2022]
Abstract
Understanding the interplay between the innate immune system, neuroinflammation, and epilepsy might offer a novel perspective in the quest of exploring new treatment strategies. Due to the complex pathology underlying epileptogenesis, no disease-modifying treatment is currently available that might prevent epilepsy after a plausible epileptogenic insult despite the advances in pre-clinical and clinical research. Neuroinflammation underlies the etiopathogenesis of epilepsy and convulsive disorders with Toll-like receptor (TLR) signal transduction being highly involved. Among TLR family members, TLR4 is an innate immune system receptor and lipopolysaccharide (LPS) sensor that has been reported to contribute to epileptogenesis by regulating neuronal excitability. Herein, we discuss available evidence on the role of TLR4 and its endogenous ligands, the high mobility group box 1 (HMGB1) protein, the heat shock proteins (HSPs) and the myeloid related protein 8 (MRP8), in epileptogenesis and post-traumatic epilepsy (PTE). Moreover, we provide an account of the promising findings of TLR4 modulation/inhibition in experimental animal models with therapeutic impact on seizures.
Collapse
|
23
|
Revisiting Platelets and Toll-Like Receptors (TLRs): At the Interface of Vascular Immunity and Thrombosis. Int J Mol Sci 2020; 21:ijms21176150. [PMID: 32858930 PMCID: PMC7504402 DOI: 10.3390/ijms21176150] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/19/2022] Open
Abstract
While platelet function has traditionally been described in the context of maintaining vascular integrity, recent evidence suggests that platelets can modulate inflammation in a much more sophisticated and nuanced manner than previously thought. Some aspects of this expanded repertoire of platelet function are mediated via expression of Toll-like receptors (TLRs). TLRs are a family of pattern recognition receptors that recognize pathogen-associated and damage-associated molecular patterns. Activation of these receptors is crucial for orchestrating and sustaining the inflammatory response to both types of danger signals. The TLR family consists of 10 known receptors, and there is at least some evidence that each of these are expressed on or within human platelets. This review presents the literature on TLR-mediated platelet activation for each of these receptors, and the existing understanding of platelet-TLR immune modulation. This review also highlights unresolved methodological issues that potentially contribute to some of the discrepancies within the literature, and we also suggest several recommendations to overcome these issues. Current understanding of TLR-mediated platelet responses in influenza, sepsis, transfusion-related injury and cardiovascular disease are discussed, and key outstanding research questions are highlighted. In summary, we provide a resource—a “researcher’s toolkit”—for undertaking further research in the field of platelet-TLR biology.
Collapse
|
24
|
The Lysophosphatidylserines-An Emerging Class of Signalling Lysophospholipids. J Membr Biol 2020; 253:381-397. [PMID: 32767057 DOI: 10.1007/s00232-020-00133-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/24/2020] [Indexed: 12/30/2022]
Abstract
Lysophospholipids are potent hormone-like signalling biological lipids that regulate many important biological processes in mammals (including humans). Lysophosphatidic acid and sphingosine-1-phosphate represent the best studied examples for this lipid class, and their metabolic enzymes and/or cognate receptors are currently under clinical investigation for treatment of various neurological and autoimmune diseases in humans. Over the past two decades, the lysophsophatidylserines (lyso-PSs) have emerged as yet another biologically important lysophospholipid, and deregulation in its metabolism has been linked to various human pathophysiological conditions. Despite its recent emergence, an exhaustive review summarizing recent advances on lyso-PSs and the biological pathways that this bioactive lysophospholipid regulates has been lacking. To address this, here, we summarize studies that led to the discovery of lyso-PS as a potent signalling biomolecule, and discuss the structure, its detection in biological systems, and the biodistribution of this lysophospholipid in various mammalian systems. Further, we describe in detail the enzymatic pathways that are involved in the biosynthesis and degradation of this lipid and the putative lyso-PS receptors reported in the literature. Finally, we discuss the various biological pathways directly regulated by lyso-PSs in mammals and prospect new questions for this still emerging biomedically important signalling lysophospholipid.
Collapse
|
25
|
COVID-19 pandemic: is a gender-defined dosage effect responsible for the high mortality rate among males? Immunogenetics 2020; 72:275-277. [PMID: 32342146 PMCID: PMC7186185 DOI: 10.1007/s00251-020-01165-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
26
|
Calaf GM, Ponce-Cusi R, Aguayo F, Muñoz JP, Bleak TC. Endocrine disruptors from the environment affecting breast cancer. Oncol Lett 2020; 20:19-32. [PMID: 32565930 PMCID: PMC7286136 DOI: 10.3892/ol.2020.11566] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Evaluation of carcinogenic substances from the environment is a challenge for scientists. Recently, a novel approach based on 10 key characteristics of human carcinogens classified by the International Agency for Research on Cancer (IARC) has emerged. Carcinogenesis depends on different mechanisms and factors, including genetic, infectious (bacteria, viruses) and environmental (chemicals) factors. Endocrine disruptors are exogenous chemicals that can interfere and impair the function of the endocrine system due to their interaction with estrogen receptors or their estrogen signaling pathways inducing adverse effects in the normal mammary development, originating cancer. They are heterogeneous chemicals and include numerous synthetic substances used worldwide in agriculture, industry and consumer products. The most common are plasticizers, such as bisphenol A (BPA), pesticides, such as dichlorodiphenyltrichloroethane, and polychlorinated biphenyls (PCBs). Xenoestrogens appear to serve an important role in the increased incidence of breast cancer in the United States and numerous other countries. Several studies have demonstrated the role of organochlorine xenoestrogens in breast cancer. Therefore, the overall cumulative exposure of women to estrogens results in an increased risk for this type of cancer. Factors like lifestyle and diet also serve a role in the increased incidence of this disease. The aim of the present study was to analyze these chemical compounds based on the key characteristics given by the IARC, with a special focus on breast cancer, to establish whether these compounds are carcinogens, and to create a model for future analysis of other endocrine disruptors.
Collapse
Affiliation(s)
- Gloria M Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
- Center for Radiological Research, Columbia University Medical Center, New York, NY 10032, USA
| | - Richard Ponce-Cusi
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| | - Francisco Aguayo
- Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago 8380000, Chile
| | - Juan P Muñoz
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| | - Tammy C Bleak
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| |
Collapse
|
27
|
Wang X, Wang F, Chen G, Yang B, Chen J, Fang Y, Wang K, Hou Y. Edwardsiella tarda induces enteritis in farmed seahorses (Hippocampus erectus): An experimental model and its evaluation. FISH & SHELLFISH IMMUNOLOGY 2020; 98:391-400. [PMID: 31991232 DOI: 10.1016/j.fsi.2020.01.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/15/2020] [Accepted: 01/23/2020] [Indexed: 06/10/2023]
Abstract
Bacterial enteritis is an important deadly threat to farmed seahorses. However, its pathogenesis is obscure because of the paucity of reproducible experimental intestinal inflammation models. Herein, a strain of Edwardsiella tarda YT1 from farmed seahorse Hippocampus erectus was isolated and identified by morphological, phylogenetic, and biochemical analysis, and confirmed as a pathogen of enteritis for the first time by challenge experiment. Two E. tarda concentrations (1 × 105 and 1 × 107 colony forming units [cfu] ml-1) were confirmed suitable for an enteritis model by intraperitoneal injection. To develop and evaluate the experimental model, we challenged seahorses with E. tarda and found that (1) the infection inhibited body length increase, significantly decreased body weight (P < 0.05), and induced typical pathological features including anorexia, anal inflammation, and intestinal fluid retention; (2) 19 external (weight, height, anal inflammation, feeding status, and intestinal fluid retention), histological (goblet and inflammatory cell numbers and thickening of lamina propria and muscularis mucosae), and molecular (hepcidin, liver-expressed antimicrobial peptide, lysozyme, piscidin, interleukin [IL]-1β, IL-1β receptor, IL-2, IL-10, interferon1, tumor necrosis factor [TNF]-α, and toll-like receptor 5 [TLR5]) indicators were suitable for model evaluation, as they could sensitively respond and varied similarly throughout the experiment, indicating the high sensitivity of seahorses against pathogen invasion; (3) TLR5 may play an essential role in triggering host immune responses during E. tarda-induced chronic enteritis, and (4) the evaluating system could reflect the pattern and intensity of disease progression. Thus, we developed an experimental model and an evaluating system of bacterial enteritis in farmed seahorses, helping us to reveal the pathogenesis of bacterial enteritis, identify potential therapeutic drugs, and search suitable genetic markers for seahorse molecular breeding.
Collapse
Affiliation(s)
- Xiaomeng Wang
- School of Life Sciences, Ludong University, Yantai, 264025, China
| | - Fang Wang
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Guozhong Chen
- School of Life Sciences, Ludong University, Yantai, 264025, China
| | - Boya Yang
- School of Agriculture, Ludong University, Yantai, 264025, China
| | - Jun Chen
- School of Agriculture, Ludong University, Yantai, 264025, China
| | - Yan Fang
- School of Agriculture, Ludong University, Yantai, 264025, China
| | - Kai Wang
- School of Agriculture, Ludong University, Yantai, 264025, China.
| | - Yuping Hou
- School of Life Sciences, Ludong University, Yantai, 264025, China.
| |
Collapse
|
28
|
Gródecka-Szwajkiewicz D, Ulańczyk Z, Zagrodnik E, Łuczkowska K, Rogińska D, Kawa MP, Stecewicz I, Safranow K, Ustianowski P, Szymański S, Machaliński B. Comparative Analysis of Global Gene Expression and Complement Components Levels in Umbilical Cord Blood from Preterm and Term Neonates: Implications for Significant Downregulation of Immune Response Pathways related to Prematurity. Int J Med Sci 2020; 17:1840-1853. [PMID: 32714087 PMCID: PMC7378668 DOI: 10.7150/ijms.46339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/03/2020] [Indexed: 01/03/2023] Open
Abstract
Background: Preterm birth is the most frequent cause of neonatal death, but its aetiology remains unclear. It has been suggested that the imbalance of immunological mechanisms responsible for maintaining pregnancy is contributing to preterm birth pathogenesis. We aimed to investigate global gene expression and the levels of several complement system components in umbilical cord blood samples from preterm neonates and compare them to term newborns. We sought to examine how differentially expressed genes could affect various immune-related pathways that are believed to be crucial factors in preterm birth. Material and methods: We enrolled 27 preterm infants (<37 weeks GA) and 52 term infants (>37 weeks GA), from which umbilical cord blood samples were collected. From these samples, peripheral blood mononuclear cells were isolated and subsequent RNA isolation was performed. We used Affymetrix Human Gene 2.1 ST Array Strip for microarray experiment and DAVID resources for bioinformatics analysis of the obtained data. Concentrations of C2, C3a, C5/C5a, C9, FactorD, Properdin were measured in umbilical cord blood plasma samples using multiplex fluorescent bead-based immunoassays using Luminex technology. Results: The levels of C3a and C5/5a were significantly elevated in preterm neonates compared to term babies, whereas C9 concentration was evidently increased in term infants. The expression of 250 genes was upregulated at least 2-fold and 3781 genes were downregulated at least 2-fold in preterm neonates in comparison with term infants. Functional annotation analysis revealed that in preterm infants in comparison to term babies there was a significant downregulation of genes encoding several Toll-like receptors, interleukins and genes involved in major signalling pathways (e.g. NF-κB, MAPK, TNF, Notch, JAK) and vital cellular processes (e.g. intracellular signal transduction, protein ubiquitination, protein transport, RNA splicing, DNA-templated transcription). Conclusions: Preterm birth results in immediate and long-term complications. Our results indicate that infants born prematurely show significant differences in complement components concentration and a downregulation of over 3,000 genes, involved mainly in various immune-related pathways, including innate immune response, phagocytosis and TLR function, when compared to full-term babies. Further studies on larger cohorts are needed to elucidate the role of immunity in prematurity.
Collapse
Affiliation(s)
| | - Zofia Ulańczyk
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Edyta Zagrodnik
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Miłosz P Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Iwona Stecewicz
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Przemysław Ustianowski
- Department of Perinatology, Obstetrics and Gynecology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Sławomir Szymański
- Department of Obstetrics and Pathology of Pregnancy, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
29
|
Short KK, Miller SM, Walsh L, Cybulski V, Bazin H, Evans JT, Burkhart D. Co-encapsulation of synthetic lipidated TLR4 and TLR7/8 agonists in the liposomal bilayer results in a rapid, synergistic enhancement of vaccine-mediated humoral immunity. J Control Release 2019; 315:186-196. [PMID: 31654684 PMCID: PMC6980726 DOI: 10.1016/j.jconrel.2019.10.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/09/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
To increase vaccine immunogenicity, modern vaccines incorporate adjuvants, which serve to enhance immune cross-protection, improve humoral and cell-mediated immunity, and promote antigen dose sparing. Pattern recognition receptors (PRRs), including the Toll-like receptor (TLR) family are promising targets for development of agonist formulations for use as vaccine adjuvants. Combinations of co-delivered TLR4 and TLR7/8 ligands have been demonstrated to have synergistic effects on innate and adaptive immune response. Here, we create liposomes that stably co-encapsulate CRX-601, a synthetic TLR4 agonist, and UM-3004, a lipidated TLR7/8 agonist, within the liposomal bilayer in order to achieve co-delivery, allow tunable physical properties, and induce in vitro and in vivo immune synergy. Co-encapsulation demonstrates a synergistic increase in IL-12p70 cytokine output in vitro from treated human peripheral blood mononuclear cells (hPBMCs). Further, co-encapsulated formulations give significant improvement of early IgG2a antibody titers in BALB/c mice following primary vaccination when compared to single agonist or dual agonists delivered in separate liposomes. This work demonstrates that co-encapsulation of TLR4 and lipidated TLR7/8 agonists within the liposomal bilayer leads to innate and adaptive immune synergy which biases a Th1 immune response. Thus, liposomal co-encapsulation may be a useful and flexible tool for vaccine adjuvant formulation containing multiple TLR agonists.
Collapse
Affiliation(s)
- Kristopher K Short
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Shannon M Miller
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Lois Walsh
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Van Cybulski
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Hélène Bazin
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - David Burkhart
- Center for Translational Medicine, University of Montana, Missoula, MT 59812, USA; Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT 59812, USA.
| |
Collapse
|
30
|
McGowan DC. Latest Advances in Small Molecule TLR 7/8 Agonist Drug Research. Curr Top Med Chem 2019; 19:2228-2238. [DOI: 10.2174/1568026619666191009165418] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/23/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Toll-like receptors (TLRs) 7 and 8 play an important role in the activation of innate immune
cells in mammals. These evolutionarily conserved receptors serve as important sentinels in response to
infection. Activation of TLRs 7 and 8 triggers induction of a Th1 type innate immune response. The
emergence of new structural and small molecule information generated in the last decade has contributed
enormously to our understanding of this highly sophisticated process of innate immunity signaling.
This review will focus on recent developments in the small molecule activation of TLR 7 and 8.
Collapse
Affiliation(s)
- David C. McGowan
- Janssen Pharmaceutica, N.V., Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
31
|
Qiu Y, Zheng J, Yang J, Li F, Zhou X, Song X. The predictive role of toll-like receptor-4 genetic polymorphisms in susceptibility to and prognosis of prostatic hyperplasia. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:86-92. [PMID: 30944713 PMCID: PMC6437452 DOI: 10.22038/ijbms.2018.33173.7922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Objective(s): This study was aimed to evaluate whether single nucleotide polymorphisms (SNPs) of TLR4 and common living habits of prostate hyperplasia (BPH) patients would affect the subjects’ risk and prognosis. Materials and Methods: We totally recruited 501 BPH patients and 964 healthy controls. The patients’ international prostate symptom score (IPSS) and quality of life assessment (QoL) were designated as the prognostic indexes for BPH patients. Altogether 7 SNPs within TLR4 were selected, and the interactions among SNPs and living habits were explained with multi-factor dimensionality reduction (MDR) modeling. Results: The mutant alleles of rs10983755 (G>A) and rs1927907 (G>A) tended to put on risk of BPH, yet the wide alleles of rs4986791 (C>T) and rs115336889 (G>C) were associated with incremental susceptibility to BPH (P<0.05). The rs10983755 (GA) and rs1927907 (GA) were suggested as the marker of non-aggressive BPH, whereas rs4986791 (TT) could symbolize aggressive BPH (P<0.05). The homozygotes of rs4986791 (TT) and rs115336889 (CC) could improve the IPSS change, and rs115336889 (CC) was also correlated with more obviously ameliorated Qol change (P<0.05). Finally, MDR modeling suggested that rs4986791 (TT) and rs115336889 (GG) shaped the genotyping combination featured by the lowest risk of BPH when smoking or drinking history was also evaluated. Conclusion: The SNPs situated within TLR4 were potent candidates for predicting risk and prognosis of BPH patients, and their interactions within environmental parameters also helped to develop effective strategies for preventing and treating BPH.
Collapse
Affiliation(s)
- Yunhua Qiu
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Jinzhou Zheng
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Jianfeng Yang
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Feng Li
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Xiqiu Zhou
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| | - Xiaoyun Song
- Department of General Surgery, Pudong Branch of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P.R. China
| |
Collapse
|
32
|
Grayfer L, Kerimoglu B, Yaparla A, Hodgkinson JW, Xie J, Belosevic M. Mechanisms of Fish Macrophage Antimicrobial Immunity. Front Immunol 2018; 9:1105. [PMID: 29892285 PMCID: PMC5985312 DOI: 10.3389/fimmu.2018.01105] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
Overcrowding conditions and temperatures shifts regularly manifest in large-scale infections of farmed fish, resulting in economic losses for the global aquaculture industries. Increased understanding of the functional mechanisms of fish antimicrobial host defenses is an important step forward in prevention of pathogen-induced morbidity and mortality in aquaculture setting. Like other vertebrates, macrophage-lineage cells are integral to fish immune responses and for this reason, much of the recent fish immunology research has focused on fish macrophage biology. These studies have revealed notable similarities as well as striking differences in the molecular strategies by which fish and higher vertebrates control their respective macrophage polarization and functionality. In this review, we address the current understanding of the biological mechanisms of teleost macrophage functional heterogeneity and immunity, focusing on the key cytokine regulators that control fish macrophage development and their antimicrobial armamentarium.
Collapse
Affiliation(s)
- Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Baris Kerimoglu
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | - Amulya Yaparla
- Department of Biological Sciences, George Washington University, Washington, DC, United States
| | | | - Jiasong Xie
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
33
|
Liang Y, Shen Y, Kuang L, Zhou G, Zhang L, Zhong X, Zhang J, Liu J. Cigarette smoke exposure promotes differentiation of CD4 + T cells toward Th17 cells by CD40-CD40L costimulatory pathway in mice. Int J Chron Obstruct Pulmon Dis 2018; 13:959-968. [PMID: 29606863 PMCID: PMC5868633 DOI: 10.2147/copd.s155754] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose This study aimed to investigate the impact of cigarette smoke exposure upon CD40–CD40L ligation between bone marrow-derived dendritic cells (BMDCs)and CD4+T cells, and to examine the effects of cigarette smoke exposure upon differentiation of CD4+T cells toward Th17 cells through blockade of CD40-CD40L pathway in mice. Methods The study was processed in vivo and in vitro. In vivo, Th17 cells, CD40, interleukin (IL)-17A, and IL-27 in the lung tissues were quantified and compared between mice with and without cigarette smoke exposure. In vitro, Th17 cells, IL-17A, and IL-27 yielded by multiple cell cultivations in which BMDCs from mice with or without cigarette smoke exposure were fostered with CD4+ T cells from healthy mice spleens in the presence of antagonistic CD40 antibody and/or cigarette smoke extract (CSE) were quantified and compared. The flow cytometry was used to detect expressions of Th17 cells and CD40, and the liquid chip was used to detect levels of IL-17A and IL-27. Results Both in vivo exposed to cigarette smoke and in vitro to CSE, CD40 expressions noticeably escalated on the surfaces of BMDCs. The presence of Th17 cells, IL-17A, and IL-27 in the lung tissues prominently increased in mice exposed to cigarette smoke. The in vitro culture of CD4+ T cells and BMDCs significantly enhanced the differentiation of CD4+ T cells toward Th17 cells and secretions of IL-17A and IL-27 in the case that BMDCs were produced from mice exposed to cigarette smoke or the culture occurred in the presence of CSE. Usage of antagonistic CD40 antibody evidently reduced the number of Th17 cells, IL-17A, and IL-27 that increased due to cigarette smoke exposure. Conclusion The CD40–CD40L ligation is associated with the quantities of Th17 cells and relevant cytokines in the context of cigarette smoke exposure. Reducing the number of Th17 cells via the usage of antagonistic CD40 antibody can be an inspiration for pursuing a novel therapeutic target for immune inflammation in COPD.
Collapse
Affiliation(s)
- Yi Liang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Ying Shen
- Division of General Practice, General Practice School of Guangxi Medical University, Nanning, Guangxi, People's Republic of China
| | - Liangjian Kuang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Guang Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Longju Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Xiaoning Zhong
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Jianquan Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| | - Jifeng Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University
| |
Collapse
|
34
|
Khan KN, Fujishita A, Hiraki K, Kitajima M, Nakashima M, Fushiki S, Kitawaki J. Bacterial contamination hypothesis: a new concept in endometriosis. Reprod Med Biol 2018; 17:125-133. [PMID: 29692669 PMCID: PMC5902457 DOI: 10.1002/rmb2.12083] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/03/2017] [Indexed: 11/11/2022] Open
Abstract
Background Endometriosis is a multifactorial disease that mainly affects women of reproductive age. The exact pathogenesis of this disease is still debatable. The role of bacterial endotoxin (lipopolysaccharide, LPS) and Toll-like receptor 4 (TLR4) in endometriosis were investigated and the possible source of endotoxin in the pelvic environment was examined. Methods The limulus amoebocyte lysate test was used to measure the endotoxin levels in the menstrual fluid and peritoneal fluid and their potential role in the growth of endometriosis was investigated. Menstrual blood and endometrial samples were cultured for the presence of microbes. The effect of gonadotrophin-releasing hormone agonist (GnRHa) treatment on intrauterine microbial colonization (IUMC) and the occurrence of endometritis was investigated. Main findings Results Lipopolysaccharide regulates the pro-inflammatory response in the pelvis and growth of endometriosis via the LPS/TLR4 cascade. The menstrual blood was highly contaminated with Escherichea coli and the endometrial samples were colonized with other microbes. A cross-talk between inflammation and ovarian steroids or the stress reaction also was observed in the pelvis. Treatment with GnRHa further worsens intrauterine microbial colonization, with the consequent occurrence of endometritis in women with endometriosis. Conclusion For the first time, a new concept called the "bacterial contamination hypothesis" is proposed in endometriosis. This study's findings of IUMC in women with endometriosis could hold new therapeutic potential in addition to the conventional estrogen-suppressing agent.
Collapse
Affiliation(s)
- Khaleque N Khan
- Graduate School of Medical Science Department of Obstetrics and Gynecology Kyoto Prefectural University of Medicine Kyoto Japan
| | - Akira Fujishita
- Department of Gynecology Saiseikai Nagasaki Hospital Nagasaki Japan
| | - Koichi Hiraki
- Department of Gynecology Saiseikai Nagasaki Hospital Nagasaki Japan
| | - Michio Kitajima
- Department of Obstetrics and Gynecology Nagasaki University Graduate School of Biomedical Sciences Nagasaki Japan
| | - Masahiro Nakashima
- Department of Tumor and Diagnostic Pathology Atomic Bomb Disease Institute Nagasaki Japan
| | - Shinji Fushiki
- Center for Quality Assurance in Research and Development Kyoto Prefectural University of Medicine Kyoto Japan
| | - Jo Kitawaki
- Graduate School of Medical Science Department of Obstetrics and Gynecology Kyoto Prefectural University of Medicine Kyoto Japan
| |
Collapse
|
35
|
Kenny LC, Kell DB. Immunological Tolerance, Pregnancy, and Preeclampsia: The Roles of Semen Microbes and the Father. Front Med (Lausanne) 2018; 4:239. [PMID: 29354635 PMCID: PMC5758600 DOI: 10.3389/fmed.2017.00239] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Although it is widely considered, in many cases, to involve two separable stages (poor placentation followed by oxidative stress/inflammation), the precise originating causes of preeclampsia (PE) remain elusive. We have previously brought together some of the considerable evidence that a (dormant) microbial component is commonly a significant part of its etiology. However, apart from recognizing, consistent with this view, that the many inflammatory markers of PE are also increased in infection, we had little to say about immunity, whether innate or adaptive. In addition, we focused on the gut, oral and female urinary tract microbiomes as the main sources of the infection. We here marshall further evidence for an infectious component in PE, focusing on the immunological tolerance characteristic of pregnancy, and the well-established fact that increased exposure to the father's semen assists this immunological tolerance. As well as these benefits, however, semen is not sterile, microbial tolerance mechanisms may exist, and we also review the evidence that semen may be responsible for inoculating the developing conceptus (and maybe the placenta) with microbes, not all of which are benign. It is suggested that when they are not, this may be a significant cause of PE. A variety of epidemiological and other evidence is entirely consistent with this, not least correlations between semen infection, infertility and PE. Our view also leads to a series of other, testable predictions. Overall, we argue for a significant paternal role in the development of PE through microbial infection of the mother via insemination.
Collapse
Affiliation(s)
- Louise C. Kenny
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
- Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- School of Chemistry, The University of Manchester, Manchester, United Kingdom
- The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
36
|
Li L, Xu G, Duan C. TLR2 affects CD86 expression and inflammatory response in burn injury mice through regulation of p38. Biochem Cell Biol 2017; 95:549-555. [PMID: 28460187 DOI: 10.1139/bcb-2016-0210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to assess the effects of TLR2-p38-CD86 signaling pathways on the inflammatory response in a mouse model of burn injury. Wild-type (TLR2+/+) and mutant-type (TLR2-/-) mice were obtained, and a mouse burn injury model was constructed. Tissue samples were examined with hematoxylin and eosin staining and the transferase mediated nick end labeling (TUNEL) method. Macrophages were treated with TLR2 agonist and p38 inhibitor. The expression levels of TLR2, p38, CD86, IL-1β, and TNF-α were quantified by RT-qPCR, Western blot, and ELISA. When compared with the sham group, the burn group had a significantly higher rate of apoptosis as well as higher expressions of TLR2, p38, CD86, IL-1β, and TNF-α. Inhibiting TLR2 was shown to significantly reduce the expressions of p-p38, CD86, IL-1β, and TNF-α. In the results of in-vitro experiments, TLR2 agonist increased the expression of p-p38, CD86, IL-1β, and TNF-α, whereas a p38 inhibitor was shown to reduce the expression of CD86, IL-1β, and TNF-α. Our results suggest that the TLR2-p38-CD86 signaling pathway plays a vital role in inflammation associated with burn injury.
Collapse
Affiliation(s)
- Li Li
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| | - Gang Xu
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| | - Chenwang Duan
- Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China.,Department of Burn and Plastic Surgery, Tangshan Gongren Hospital, Tangshan 063000, Hebei, China
| |
Collapse
|
37
|
Najar M, Krayem M, Meuleman N, Bron D, Lagneaux L. Mesenchymal Stromal Cells and Toll-Like Receptor Priming: A Critical Review. Immune Netw 2017; 17:89-102. [PMID: 28458620 PMCID: PMC5407987 DOI: 10.4110/in.2017.17.2.89] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/24/2017] [Accepted: 02/25/2017] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal Stromal Cells (MSCs) are potential cellular candidates for several immunotherapy purposes. Their multilineage potential and immunomodulatory properties make them interesting tools for the treatment of various immunological diseases. However, depending on the local microenvironment, diverse biological functions of MSCs can be modulated. Indeed, during infections such as obtained following TLR-agonist engagement (called as TLR priming), the phenotype, multilineage potential, hematopoietic support and immunomodulatory capacity of MSCs can present critical changes, which could further affect their therapeutic potential. Thus, for appropriate clinical application of MSCs, it is important to well know and understand these effects in particular during infectious episodes and to find the suitable experimental settings to study that. Pre-stimulation of MSCs with a specific TLR ligand may serve as an effective priming step to modulate one of its function to achieve a desired therapeutic issue.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Mohammad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels 1000, Belgium
| | - Nathalie Meuleman
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles (ULB), Campus Erasme, Belgium
| |
Collapse
|
38
|
Ishikawa S, Ito S. Repeated whole cigarette smoke exposure alters cell differentiation and augments secretion of inflammatory mediators in air-liquid interface three-dimensional co-culture model of human bronchial tissue. Toxicol In Vitro 2017; 38:170-178. [PMID: 27596523 DOI: 10.1016/j.tiv.2016.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/12/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
In vitro models of human bronchial epithelium are useful for toxicological testing because of their resemblance to in vivo tissue. We constructed a model of human bronchial tissue which has a fibroblast layer embedded in a collagen matrix directly below a fully-differentiated epithelial cell layer. The model was applied to whole cigarette smoke (CS) exposure repeatedly from an air-liquid interface culture while bronchial epithelial cells were differentiating. The effects of CS exposure on differentiation were determined by histological and gene expression analyses on culture day 21. We found a decrease in ciliated cells and perturbation of goblet cell differentiation. We also analyzed the effects of CS exposure on the inflammatory response, and observed a significant increase in secretion of IL-8, GRO-α, IL-1β, and GM-CSF. Interestingly, secretion of these mediators was augmented with repetition of whole CS exposure. Our data demonstrate the usefulness of our bronchial tissue model for in vitro testing and the importance of exposure repetition in perturbing the differentiation and inflammation processes.
Collapse
Affiliation(s)
- Shinkichi Ishikawa
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa 227-8512, Japan.
| | - Shigeaki Ito
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa 227-8512, Japan
| |
Collapse
|
39
|
Lu WL, Song DZ, Yue JL, Wang TT, Zhou XD, Zhang P, Zhang L, Huang DM. NLRP3 inflammasome may regulate inflammatory response of human periodontal ligament fibroblasts in an apoptosis-associated speck-like protein containing a CARD (ASC)-dependent manner. Int Endod J 2017; 50:967-975. [PMID: 27864974 DOI: 10.1111/iej.12722] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 11/14/2016] [Indexed: 02/05/2023]
Affiliation(s)
- W. L. Lu
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - D. Z. Song
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - J. L. Yue
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - T. T. Wang
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - X. D. Zhou
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - P. Zhang
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - L. Zhang
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - D. M. Huang
- Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
- Department of Conservative Dentistry; West China Hospital of Stomatology; Sichuan University; Chengdu China
| |
Collapse
|
40
|
|
41
|
Lee B, Ko E, Lee J, Jo Y, Hwang H, Goh TS, Joo M, Hong C. Soluble common gamma chain exacerbates COPD progress through the regulation of inflammatory T cell response in mice. Int J Chron Obstruct Pulmon Dis 2017; 12:817-827. [PMID: 28331303 PMCID: PMC5352154 DOI: 10.2147/copd.s123405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoking (CS) is a major cause of considerable morbidity and mortality by inducing lung cancer and COPD. COPD, a smoking-related disorder, is closely related to the alteration of immune system and inflammatory processes that are specifically mediated by T cells. Soluble common gamma chain (sγc) has recently been identified as a critical regulator of the development and differentiation of T cells. We examined the effects of sγc in a cigarette smoke extract (CSE) mouse model. The sγc level in CSE mice serum is significantly downregulated, and the cellularity of lymph node (LN) is systemically reduced in the CSE group. Overexpression of sγc enhances the cellularity and IFNγ production of CD8 T cells in LN and also enhances Th1 and Th17 differentiation of CD4 T cells in the respiratory tract. Mechanistically, the downregulation of sγc expression mediated by CSE is required to prevent excessive inflammatory T cell responses. Therefore, our data suggest that sγc may be one of the target molecules for the control of immunopathogenic progresses in COPD.
Collapse
Affiliation(s)
- Byunghyuk Lee
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine
| | - Eunhee Ko
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine
| | - Jiyeon Lee
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan
| | - Yuna Jo
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine
| | - Hyunju Hwang
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine
| | - Tae Sik Goh
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine; Department of Orthopedic Surgery, Medical Research Institute, Pusan National University School of Medicine, Busan, South Korea
| | - Myungsoo Joo
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan
| | - Changwan Hong
- Department of Anatomy and Cell Biology, Pusan National University School of Medicine
| |
Collapse
|
42
|
Lim SM, Lee SY, Jeong JJ, Choi HS, Chang HB, Kim DH. DW2007 Ameliorates Colitis and Rheumatoid Arthritis in Mice by Correcting Th17/Treg Imbalance and Inhibiting NF-κB Activation. Biomol Ther (Seoul) 2016; 24:638-649. [PMID: 27302961 PMCID: PMC5098544 DOI: 10.4062/biomolther.2016.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 02/18/2016] [Accepted: 03/24/2016] [Indexed: 01/16/2023] Open
Abstract
In the previous study, the rhizome mixture of Anemarrhena asphodeloides and Coptis chinensis (DW2007), improved TNBS-, oxazolone-, or DSS-induced colitis in mice by regulating macrophage activation. Therefore, to understand the effect of DW2007 on the T cell differentiation involved in the adaptive immunity, we measured its effect on both Th17 and Treg cell differentiation in splenocytes, in the lamina propria of mice with DSS-induced colitis (DIC), and in the spleens of mice with collagen-induced arthritis (CIA). Results showed that DW2007 potently inhibited the differentiation of splenocytes into Th17 cells, but increased Treg cell differentiation in vitro. In the colon of wild type and TLR4-/- mice with DIC, DW2007 potently suppressed DSS-induced colon shortening and myeloperoxidase activity. DW2007 also suppressed collagen-induced paw thickening, clinical index, and myeloperoxidase activity in CIA mice. Overall, DW2007 potently suppressed Th17 cell differentiation in mice with CIA and DIC, but increased Treg cell differentiation. Moreover, DW2007 strongly inhibited the expression of TNF-α and IL-1β, as well as the activation of NF-κB. Based on these findings, DW2007 may ameliorate inflammatory diseases by regulating the innate immunity via the inhibition of macrophage activation and the adaptive immunity via the correction of disturbed Th17/Treg cells.
Collapse
Affiliation(s)
- Su-Min Lim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sang-Yun Lee
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jin-Ju Jeong
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Sik Choi
- DongWha Pharm Research Institute, Yongin 04637, Republic of Korea
| | - Hwan Bong Chang
- DongWha Pharm Research Institute, Yongin 04637, Republic of Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
43
|
The Role of TLR4 on B Cell Activation and Anti- β2GPI Antibody Production in the Antiphospholipid Syndrome. J Immunol Res 2016; 2016:1719720. [PMID: 27868072 PMCID: PMC5102736 DOI: 10.1155/2016/1719720] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/16/2016] [Indexed: 01/02/2023] Open
Abstract
High titer of anti-β2-glycoprotein I antibodies (anti-β2GPI Ab) plays a pathogenic role in antiphospholipid syndrome (APS). Numerous studies have focused on the pathological mechanism in APS; however, little attention is paid to the immune mechanism of production of anti-β2GPI antibodies in APS. Our previous study demonstrated that Toll-like receptor 4 (TLR4) plays a vital role in the maturation of bone marrow-derived dendritic cells (BMDCs) from the mice immunized with human β2-glycoprotein I (β2GPI). TLR4 is required for the activation of B cells and the production of autoantibody in mice treated with β2GPI. However, TLR4 provides a third signal for B cell activation and then promotes B cells better receiving signals from both B cell antigen receptor (BCR) and CD40, thus promoting B cell activation, surface molecules expression, anti-β2GPI Ab production, and cytokines secretion and making B cell functioning like an antigen presenting cell (APC). At the same time, TLR4 also promotes B cells producing antibodies by upregulating the expression of B-cell activating factor (BAFF). In this paper, we aim to review the functions of TLR4 in B cell immune response and antibody production in autoimmune disease APS and try to find a new way for the prevention and treatment of APS.
Collapse
|
44
|
Carlier FM, Sibille Y, Pilette C. The epithelial barrier and immunoglobulin A system in allergy. Clin Exp Allergy 2016; 46:1372-1388. [PMID: 27684559 DOI: 10.1111/cea.12830] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Airway and intestinal epithelial layers represent first-line physical barriers, playing a key role in mucosal immunity. Barrier dysfunction, characterized by alterations such as disruption of cell-cell apical junctions and aberrant epithelial responses, probably constitutes early and key events for chronic immune responses to environmental antigens in the skin and in the gut. For instance, barrier dysfunction drives Th2 responses in atopic disorders or eosinophilic esophagitis. Such epithelial impairment is also a salient feature of allergic asthma and growing evidence indicates that barrier alterations probably play a driving role in this disease. IgA has been identified as the most abundant immunoglobulin in mucosa, where it acts as an active barrier through immune exclusion of inhaled or ingested antigens or pathogens. Historically, it has been thought to represent the serum factor underlying reaginic activity before IgE was discovered. Despite several studies about regulation and major functions of IgA at mucosal surfaces, its role in allergy remains largely unclear. This review aims at summarizing findings about epithelial functions and IgA biology that are relevant to allergy, and to integrate the emerging concepts and the recent developments in mucosal immunology, and how these could translate to clinical observations in allergy.
Collapse
Affiliation(s)
- F M Carlier
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium. .,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium.
| | - Y Sibille
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Centre Hospitalier Universitaire Dinant-Godinne UCL Namur, Yvoir, Belgium
| | - C Pilette
- Institut de Recherche Expérimentale et Clinique, Pôle Pneumologie, ORL et dermatologie, Brussels, Belgium.,Department of Internal Medicine, Division of Pneumology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.,Walloon Excellence in Lifesciences and Biotechnology, Wavre, Belgium
| |
Collapse
|
45
|
Xu J, Huang G, Guo TL. Developmental Bisphenol A Exposure Modulates Immune-Related Diseases. TOXICS 2016; 4:toxics4040023. [PMID: 29051427 PMCID: PMC5606650 DOI: 10.3390/toxics4040023] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 08/31/2016] [Accepted: 09/12/2016] [Indexed: 02/06/2023]
Abstract
Bisphenol A (BPA), used in polycarbonate plastics and epoxy resins, has a widespread exposure to humans. BPA is of concern for developmental exposure resulting in immunomodulation and disease development due to its ability to cross the placental barrier and presence in breast milk. BPA can use various mechanisms to modulate the immune system and affect diseases, including agonistic and antagonistic effects on many receptors (e.g., estrogen receptors), epigenetic modifications, acting on cell signaling pathways and, likely, the gut microbiome. Immune cell populations and function from the innate and adaptive immune system are altered by developmental BPA exposure, including decreased T regulatory (Treg) cells and upregulated pro- and anti-inflammatory cytokines and chemokines. Developmental BPA exposure can also contribute to the development of type 2 diabetes mellitus, allergy, asthma and mammary cancer disease by altering immune function. Multiple sclerosis and type 1 diabetes mellitus may also be exacerbated by BPA, although more research is needed. Additionally, BPA analogs, such as bisphenol S (BPS), have been increasing in use, and currently, little is known about their immune effects. Therefore, more studies should be conducted to determine if developmental exposure BPA and its analogs modulate immune responses and lead to immune-related diseases.
Collapse
Affiliation(s)
- Joella Xu
- Department of Veterinary Biosciences and Diagnostic Imaging, Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602-7382, USA.
| | - Guannan Huang
- Department of Environmental Health Sciences, University of Georgia, Athens, GA 30602-7382, USA.
| | - Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, Interdisciplinary Toxicology Program, University of Georgia, Athens, GA 30602-7382, USA.
| |
Collapse
|
46
|
MyD88 Shapes Vaccine Immunity by Extrinsically Regulating Survival of CD4+ T Cells during the Contraction Phase. PLoS Pathog 2016; 12:e1005787. [PMID: 27542117 PMCID: PMC4991787 DOI: 10.1371/journal.ppat.1005787] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/05/2016] [Indexed: 12/01/2022] Open
Abstract
Soaring rates of systemic fungal infections worldwide underscore the need for vaccine prevention. An understanding of the elements that promote vaccine immunity is essential. We previously reported that Th17 cells are required for vaccine immunity to the systemic dimorphic fungi of North America, and that Card9 and MyD88 signaling are required for the development of protective Th17 cells. Herein, we investigated where, when and how MyD88 regulates T cell development. We uncovered a novel mechanism in which MyD88 extrinsically regulates the survival of activated T cells during the contraction phase and in the absence of inflammation, but is dispensable for the expansion and differentiation of the cells. The poor survival of activated T cells in Myd88-/- mice is linked to increased caspase3-mediated apoptosis, but not to Fas- or Bim-dependent apoptotic pathways, nor to reduced expression of the anti-apoptotic molecules Bcl-2 or Bcl-xL. Moreover, TLR3, 7, and/or 9, but not TLR2 or 4, also were required extrinsically for MyD88-dependent Th17 cell responses and vaccine immunity. Similar MyD88 requirements governed the survival of virus primed T cells. Our data identify unappreciated new requirements for eliciting adaptive immunity and have implications for designing vaccines. Despite several million new systemic fungal infections annually worldwide, there are no commercial vaccines available. The development of effective vaccines requires a fundamental understanding of how protective immune responses are induced. Using experimental vaccine strains, we previously demonstrated that populations of T helper cells producing interleukin 17 (Th17 cells) and interferon gamma (Th1 cells) mediate vaccine resistance to systemic dimorphic fungi of North America. Here, we report how the immune system recognizes the fungal vaccines and induces the development of protective T cells. We delineate the role of pathogen recognition receptors (PRRs) and their common signaling pathway in host immune cells that recognize the fungal vaccine. While the signaling pathway studied is essential for the development of vaccine-induced T cells, the mechanism of action is novel and included T cell death after activation. The findings could be extended to virus-specific T cells suggesting that the mechanism is conserved among the microbial kingdom. Our work sheds new light on how protective T cells are induced and can be harnessed by vaccine strategies tailored against fungal and other microbial infections.
Collapse
|
47
|
Zhang JH, Shangguan ZS, Chen C, Zhang HJ, Lin Y. Anti-inflammatory effects of guggulsterone on murine macrophage by inhibiting LPS-induced inflammatory cytokines in NF-κB signaling pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1829-35. [PMID: 27330276 PMCID: PMC4896467 DOI: 10.2147/dddt.s104602] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The present study was aimed to investigate the effects of guggulsterone (GS) on proinflammatory responses as well as the underlying molecular mechanisms in macrophage upon lipopolysaccharide (LPS) stimulation. Effects of GS on viability of Raw264.7 cells were examined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Real-time polymerase chain reaction (PCR) was employed to examine the mRNA expression of cytokines, including interleukin 1β (IL-1β), tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS). Phosphorylations of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinases (p38), and inhibitor of nuclear factor kappaB (IκB) were determined using immunoblotting. The results revealed that GS was not toxic to Raw264.7 cells at designated concentrations. We demonstrated that GS significantly suppressed the elevated mRNA expression of proinflammatory cytokines, including IL-1β, TNF-α, and iNOS in a dose-dependent manner. GS treatment reduced the level of IκB phosphorylation in LPS-stimulated macrophages in a dose-dependent manner. Use of BAY 11-7082, an inhibitor of nuclear factor-kappaB (NF-κB), led to significantly suppressing effects on IL-1β and TNF-α expression similar as that of GS-treated cells. Our findings suggest that GS possesses anti-inflammatory activity, which may be attributed to downregulation of iNOS and inhibition of NF-κB activity in LPS-stimulated Raw264.7 cells.
Collapse
Affiliation(s)
- Jin-Hua Zhang
- College of Chemical Engineering, Huaqiao University, Xiamen, People's Republic of China; Department of Pharmacy, Xiamen Medical College, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Zhao-Shui Shangguan
- Central Laboratory, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Chao Chen
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Hui-Jie Zhang
- Xiamen Diabetes Institute, The First Affiliated Hospital of Xiamen University, Xiamen, People's Republic of China
| | - Yi Lin
- College of Chemical Engineering, Huaqiao University, Xiamen, People's Republic of China
| |
Collapse
|
48
|
Quesada-García A, Encinas P, Valdehita A, Baumann L, Segner H, Coll JM, Navas JM. Thyroid active agents T3 and PTU differentially affect immune gene transcripts in the head kidney of rainbow trout (Oncorynchus mykiss). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 174:159-168. [PMID: 26963519 DOI: 10.1016/j.aquatox.2016.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 02/16/2016] [Accepted: 02/20/2016] [Indexed: 06/05/2023]
Abstract
In mammals, numerous reports describe an immunomodulating effect of thyroid-active compounds. In contrast, only few reports have been published on this subject in fish. We previously demonstrated that immune cells of rainbow trout (Oncorhynchus mykiss) possess thyroid hormone receptors (THRs) and that exposure of trout to the thyroid hormone 3,3',5-triiodo-l-thyronine (T3) or the antithyroid drug propylthiouracil (PTU) alters immune cell transcript levels of THR and several immune genes. The present study aims to further characterize the immunomodulating action of thyroid-active compounds in trout immune cells. We report here the use of a custom-designed 60-mer oligo immune-targeted microarray for rainbow trout to analyze the gene expression profiles induced in the head kidney by T3 and PTU. Morphometric analyses of the thyroid showed that PTU exposure increased the size of the epithelial cells, whereas T3 induced no significant effects. Both T3 and PTU had diverse and partly contrasting effects on immune transcript profiles. The strongest differential effects of T3 and PTU on gene expressions were those targeting the Mitogen Associated Protein Kinase (MAPK), NFkB, Natural Killer (NK) and Toll-Like Receptor (TLR) pathways, a number of multipath genes (MPG) such as those encoding pleiotropic transcription factors (atf1, junb, myc), as well as important pro-inflammatory genes (tnfa, tnf6, il1b) and interferon-related genes (ifng, irf10). With these results we show for the first time in a fish species that the in vivo thyroidal status modulates a diversity of immune genes and pathways. This knowledge provides the basis to investigate both mechanisms and consequences of thyroid hormone- and thyroid disruptor-mediated immunomodulation for the immunocompetence of fish.
Collapse
Affiliation(s)
- Alba Quesada-García
- Instituto Nacional Investigaciones Agrarias y Alimentarias. INIA, Dpto. Medio Ambiente, Ctra. De la Coruña Km 7.5., E-28040 Madrid, Spain
| | - Paloma Encinas
- Instituto Nacional Investigaciones Agrarias y Alimentarias. INIA, Dpto. Biotecnologia, Ctra. De la Coruña Km 7.5., E-28040 Madrid, Spain
| | - Ana Valdehita
- Instituto Nacional Investigaciones Agrarias y Alimentarias. INIA, Dpto. Medio Ambiente, Ctra. De la Coruña Km 7.5., E-28040 Madrid, Spain
| | - Lisa Baumann
- Faculty of Vetsuisse, Centre for Fish and Wildlife Health, University of Bern, Länggasstra. 122, CH-3001 Bern, Switzerland
| | - Helmut Segner
- Faculty of Vetsuisse, Centre for Fish and Wildlife Health, University of Bern, Länggasstra. 122, CH-3001 Bern, Switzerland
| | - Julio M Coll
- Instituto Nacional Investigaciones Agrarias y Alimentarias. INIA, Dpto. Biotecnologia, Ctra. De la Coruña Km 7.5., E-28040 Madrid, Spain
| | - José M Navas
- Instituto Nacional Investigaciones Agrarias y Alimentarias. INIA, Dpto. Medio Ambiente, Ctra. De la Coruña Km 7.5., E-28040 Madrid, Spain.
| |
Collapse
|
49
|
Yang J, Qin N, Zhang H, Yang R, Xiang B, Wei Q. Cellular uptake of exogenous calcineurin B is dependent on TLR4/MD2/CD14 complexes, and CnB is an endogenous ligand of TLR4. Sci Rep 2016; 6:24346. [PMID: 27090571 PMCID: PMC4835703 DOI: 10.1038/srep24346] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 03/15/2016] [Indexed: 12/21/2022] Open
Abstract
Our previous research showed that recombinant calcineurin B (rhCnB) stimulates cytokine secretion by immune cells, probably through TLR4. Exogenous CnB can be incorporated into many different tumour cells in vitro, but the mode of uptake and receptors required remain unknown. Here, we report that exogenous CnB is taken up by cells in a time- and concentration-dependent manner via clathrin-dependent receptor-mediated internalization. Our findings further confirm that uptake is mediated by the TLR4/MD2 complex together with the co-receptor CD14. The MST results revealed a high affinity between CnB and the TLR4 receptor complex. No binding was detected between CnB and LPS. CnB inhibited the uptake of LPS, and LPS also inhibited the uptake of CnB. These results indicate that the uptake of exogenous CnB did not occur through LPS and that CnB was not a chaperone of LPS. Thus, we conclude that TLR4 receptor complexes were required for the recognition and internalization of exogenous CnB. CnB could be a potential endogenous ligand of TLR4 and function as an agonist of TLR4. These properties of CnB support its potential for development as an anti-cancer drug.
Collapse
Affiliation(s)
- Jinju Yang
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| | - Nannan Qin
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| | - Hongwei Zhang
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| | - Rui Yang
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| | - Benqiong Xiang
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| | - Qun Wei
- Department of Biochemistry and Molecular Biology, Beijing Normal University, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing, 100875, P. R. of China
| |
Collapse
|
50
|
Iqbal SM, Leonard C, Regmi SC, De Rantere D, Tailor P, Ren G, Ishida H, Hsu C, Abubacker S, Pang DS, Salo PT, Vogel HJ, Hart DA, Waterhouse CC, Jay GD, Schmidt TA, Krawetz RJ. Lubricin/Proteoglycan 4 binds to and regulates the activity of Toll-Like Receptors In Vitro. Sci Rep 2016; 6:18910. [PMID: 26752378 PMCID: PMC4707532 DOI: 10.1038/srep18910] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 11/30/2015] [Indexed: 01/03/2023] Open
Abstract
Proteoglycan 4 (PRG4/lubricin) is secreted by cells that reside in articular cartilage and line the synovial joint. Lubricin may play a role in modulating inflammatory responses through interaction with CD44. This led us to examine if lubricin could be playing a larger role in the modulation of inflammation/immunity through interaction with Toll-like receptors (TLRs). Human Embryonic Kidney (HEK) cells overexpressing TLRs 2, 4 or 5 and surface plasmon resonance were employed to determine if full length recombinant human lubricin was able to bind to and activate TLRs. Primary human synovial fibroblasts were also examined using flow cytometry and Luminex multiplex ELISA. A rat destabilization model of osteoarthritis (OA) was used to determine if lubricin injections were able to regulate pain and/or inflammation in vivo. Lubricin can bind to and regulate the activity of TLRs, leading to downstream changes in inflammatory signalling independent of HA. We confirmed these findings in vivo through intra-articular injections of lubricin in a rat OA model where the inhibition of systemic inflammatory signaling and reduction in pain were observed. Lubricin plays an important role in regulating the inflammatory environment under both homeostatic and tissue injury states.
Collapse
Affiliation(s)
- S M Iqbal
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - C Leonard
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - S C Regmi
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - D De Rantere
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - P Tailor
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - G Ren
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - H Ishida
- Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - Cy Hsu
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - S Abubacker
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - D Sj Pang
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - P T Salo
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - H J Vogel
- Faculty of Science, University of Calgary, Calgary, Alberta, Canada
| | - D A Hart
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - C C Waterhouse
- Snyder Institute, Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - G D Jay
- Faculty of Medicine, Brown University, Providence, Rhode Island, United States
| | - T A Schmidt
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada.,Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - R J Krawetz
- McCaig Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|