1
|
Rodrigues e-Lacerda R, Barra NG, Fang H, Anhê GF, Schertzer JD. NOD2 protects against allergic lung inflammation in obese female mice. iScience 2024; 27:111130. [PMID: 39507249 PMCID: PMC11539594 DOI: 10.1016/j.isci.2024.111130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 08/07/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
Obesity is associated with compartmentalized changes in immune responses that can be protective or pathogenic. It has been proposed that obesity-related changes in the microbiota influence allergic lung inflammation. We hypothesized that sensors of the bacterial cell wall influenced allergenic lung inflammation during obesity. Ovalbumin (OVA)-induced lung inflammation was similar in female Nod1-/- and wild-type mice during high-fat-diet-induced obesity, but allergic lung inflammation was higher in obese, high-fat-diet-fed female Nod2-/- mice. Obese Nod2-/- mice had higher inflammatory cell infiltration in the bronchial alveolar lavage (BAL) and lungs, pulmonary fibrosis, mucus levels, hypertrophy and hyperplasia of goblet cells, M2 alveolar macrophage infiltration, interleukin-4 (IL-4), IL-5, IL-6, and lower CXCL1 and IL-22. Therefore, Nod2 protects against excessive lung inflammation and is a bacterial sensor that relays protective responses to allergenic lung inflammation in obese female mice.
Collapse
Affiliation(s)
- Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Nicole G. Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| | - Gabriel Forato Anhê
- Department of Translational Medicine, University of Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz, Campinas, SP CEP 13083-887, Brazil
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, 1200 Main Street West, Hamilton, ON L8N 3Z5, Canada
| |
Collapse
|
2
|
Yao D, Shao J, Jia D, Sun W. Immunotoxicity of legacy and alternative per- and polyfluoroalkyl substances on zebrafish larvae. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 358:124511. [PMID: 38977121 DOI: 10.1016/j.envpol.2024.124511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/04/2024] [Accepted: 07/06/2024] [Indexed: 07/10/2024]
Abstract
Hexafluoropropylene oxide dimer acid (HFPO-DA) and perfluoroethylcyclohexane sulfonate (PFECHS) are increasingly used as alternatives for perfluorooctanoic acid (PFOA) and perfluorooctane sulfonate (PFOS). However, their immunotoxicity and underlying molecular mechanisms remain poorly understood. Here, to assess immunotoxic effects, zebrafish embryos were exposed to environmentally relevant concentrations of PFOA, PFOS, HFPO-DA, and PFECHS for four days. Results revealed that all four per- and polyfluoroalkyl substances (PFAS) resulted in decreased heart rate and spontaneous movement, and induced oxidative stress in zebrafish larvae. Notably, HFPO-DA exhibited more severe oxidative stress than PFOA. Immune dysfunction was observed, characterized by elevated cytokine, complement factor, nitric oxide, and neutrophil content, along with a significant decrease in lysozyme content. Transcriptomic analysis revealed the activation of Toll-like receptor (TLR)/NOD-like receptor (NLR)/RIG-I-like receptor (RLR) and associated downstream genes, indicating their pivotal role in PFAS-induced immunomodulation. Molecular docking simulations demonstrated stable interactions between PFAS and key receptors (TLR2, NOD2 and RIG-I). Overall, HFPO-DA and PFECHS exhibited immunotoxic effects in zebrafish larvae similar to legacy PFAS, providing important information for understanding the toxic mode of action of these emerging alternatives.
Collapse
Affiliation(s)
- Dengdiao Yao
- College of Animal Science, Guizhou University, The Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, Guizhou, 550025, China.
| | - Jian Shao
- College of Animal Science, Guizhou University, The Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guiyang, Guizhou, 550025, China
| | - Dantong Jia
- Key Laboratory of Water and Sediment Sciences, Ministry of Education, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, Beijing, 100871, China
| | - Weiling Sun
- Key Laboratory of Water and Sediment Sciences, Ministry of Education, College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China; State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, Beijing, 100871, China.
| |
Collapse
|
3
|
Garcia-Vidal E, Calba I, Riveira-Muñoz E, García E, Clotet B, Serra-Mitjà P, Cabrera C, Ballana E, Badia R. Nucleotide-Binding Oligomerization Domain 1 (NOD1) Agonists Prevent SARS-CoV-2 Infection in Human Lung Epithelial Cells through Harnessing the Innate Immune Response. Int J Mol Sci 2024; 25:5318. [PMID: 38791357 PMCID: PMC11121681 DOI: 10.3390/ijms25105318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The lung is prone to infections from respiratory viruses such as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). A challenge in combating these infections is the difficulty in targeting antiviral activity directly at the lung mucosal tract. Boosting the capability of the respiratory mucosa to trigger a potent immune response at the onset of infection could serve as a potential strategy for managing respiratory infections. This study focused on screening immunomodulators to enhance innate immune response in lung epithelial and immune cell models. Through testing various subfamilies and pathways of pattern recognition receptors (PRRs), the nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family was found to selectively activate innate immunity in lung epithelial cells. Activation of NOD1 and dual NOD1/2 by the agonists TriDAP and M-TriDAP, respectively, increased the number of IL-8+ cells by engaging the NF-κB and interferon response pathways. Lung epithelial cells showed a stronger response to NOD1 and dual NOD1/2 agonists compared to control. Interestingly, a less-pronounced response to NOD1 agonists was noted in PBMCs, indicating a tissue-specific effect of NOD1 in lung epithelial cells without inducing widespread systemic activation. The specificity of the NOD agonist pathway was confirmed through gene silencing of NOD1 (siRNA) and selective NOD1 and dual NOD1/2 inhibitors in lung epithelial cells. Ultimately, activation induced by NOD1 and dual NOD1/2 agonists created an antiviral environment that hindered SARS-CoV-2 replication in vitro in lung epithelial cells.
Collapse
Affiliation(s)
| | - Ignasi Calba
- IrsiCaixa, 08916 Badalona, Barcelona, Spain (E.G.)
- Health Research Institute Germans Trias i Pujol (IGTP), Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Barcelona, Spain
| | | | | | - Bonaventura Clotet
- IrsiCaixa, 08916 Badalona, Barcelona, Spain (E.G.)
- University of Vic—Central University of Catalonia (UVic-UCC), 08500 Vic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, 28029 Madrid, Spain
| | - Pere Serra-Mitjà
- Pulmonology and Allergy Unit, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08041 Barcelona, Barcelona, Spain;
| | - Cecilia Cabrera
- IrsiCaixa, 08916 Badalona, Barcelona, Spain (E.G.)
- Health Research Institute Germans Trias i Pujol (IGTP), Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Barcelona, Spain
| | - Ester Ballana
- IrsiCaixa, 08916 Badalona, Barcelona, Spain (E.G.)
- Health Research Institute Germans Trias i Pujol (IGTP), Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, 28029 Madrid, Spain
| | - Roger Badia
- IrsiCaixa, 08916 Badalona, Barcelona, Spain (E.G.)
- Health Research Institute Germans Trias i Pujol (IGTP), Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, 08916 Badalona, Barcelona, Spain
| |
Collapse
|
4
|
Ding Y, Sun W, Han M, Liu Z, Kang H, Ma X, Wang J, Mu H, Huang Y, Hou S, Sun D, Shen X, Wu X, Liu R. NLRX1: a key regulator in mitochondrial respiration and colorectal cancer progression. Med Oncol 2024; 41:131. [PMID: 38683455 DOI: 10.1007/s12032-024-02364-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024]
Abstract
Colorectal cancer (CRC) is a prevalent and aggressive malignancy with high mortality rates and significant risks to human well-being. Population-wide screening for tumor suppressor genes and oncogenes shows promise for reducing the incidence and fatality of CRC. Recent studies have suggested that NLRX1, an innate immunity suppressor, may play a role in regulating chronic inflammation and tumorigenesis. However, further investigation is needed to understand the specific role of NLRX1 in CRC. To evaluate the impact of NLRX1 on migration, invasion, and metastasis, two human colon cancer cell lines were studied in vitro. Additionally, a knockout mouse tumor-bearing model was used to validate the inhibitory effect of NLRX1 on tumor emergence and progression. The Seahorse XF96 technology was employed to assess mitochondrial function and glycolysis in colorectal cancer cells overexpressing NLRX1. Moreover, public databases were consulted to analyze gene and protein expression levels of NLRX1. Finally, the results were validated using a series of CRC patient samples. Our findings demonstrate that downregulation of NLRX1 enhances proliferation, colony formation, and tumor-forming capacity in HCT116 and LoVo cells. Conversely, overexpression of NLRX1 negatively impacts basal respiration and mitochondrial ATP-linked respiration in both cell lines, resulting in a notable decrease in maximal respiration during the standard mitochondrial stress test. Furthermore, analysis of data from the TCGA database reveals a significant reduction in NLRX1 expression in colon and rectal cancer tissues compared to normal tissues. This result was validated using clinical samples, where immunohistochemistry staining and western blotting demonstrated a notable reduction in NLRX1 protein levels in CRC compared to adjacent normal tissues. The decreased expression of NLRX1 may serve as a significant prognostic indicator and diagnostic biomarker for CRC patients.
Collapse
Affiliation(s)
- Yaxin Ding
- College of Life Science, Northwest University, Xi'an, China
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wenjie Sun
- College of Life Science, Northwest University, Xi'an, China
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Mingwei Han
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ziyu Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Huarui Kang
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xiaohan Ma
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jiayu Wang
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hongrui Mu
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yuxiao Huang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shiyuan Hou
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Danni Sun
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xing Shen
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
5
|
Vinţeler N, Feurdean CN, Petkes R, Barabas R, Boşca BA, Muntean A, Feștilă D, Ilea A. Biomaterials Functionalized with Inflammasome Inhibitors-Premises and Perspectives. J Funct Biomater 2024; 15:32. [PMID: 38391885 PMCID: PMC10889089 DOI: 10.3390/jfb15020032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
This review aimed at searching literature for data regarding the inflammasomes' involvement in the pathogenesis of oral diseases (mainly periodontitis) and general pathologies, including approaches to control inflammasome-related pathogenic mechanisms. The inflammasomes are part of the innate immune response that activates inflammatory caspases by canonical and noncanonical pathways, to control the activity of Gasdermin D. Once an inflammasome is activated, pro-inflammatory cytokines, such as interleukins, are released. Thus, inflammasomes are involved in inflammatory, autoimmune and autoinflammatory diseases. The review also investigated novel therapies based on the use of phytochemicals and pharmaceutical substances for inhibiting inflammasome activity. Pharmaceutical substances can control the inflammasomes by three mechanisms: inhibiting the intracellular signaling pathways (Allopurinol and SS-31), blocking inflammasome components (VX-765, Emricasan and VX-740), and inhibiting cytokines mediated by the inflammasomes (Canakinumab, Anakinra and Rilonacept). Moreover, phytochemicals inhibit the inflammasomes by neutralizing reactive oxygen species. Biomaterials functionalized by the adsorption of therapeutic agents onto different nanomaterials could represent future research directions to facilitate multimodal and sequential treatment in oral pathologies.
Collapse
Affiliation(s)
- Norina Vinţeler
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Claudia Nicoleta Feurdean
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Regina Petkes
- Department of Chemistry and Chemical Engineering of Hungarian Line of Study, Faculty of Chemistry and Chemical Engineering, Babeș-Bolyai University, 400028 Cluj-Napoca, Romania
| | - Reka Barabas
- Department of Chemistry and Chemical Engineering of Hungarian Line of Study, Faculty of Chemistry and Chemical Engineering, Babeș-Bolyai University, 400028 Cluj-Napoca, Romania
| | - Bianca Adina Boşca
- Department of Histology, Faculty of Medicine, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandrina Muntean
- Department of Paediatric, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania
| | - Dana Feștilă
- Department of Orthodontics, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca 400012, Romania
| | - Aranka Ilea
- Department of Oral Rehabilitation, Faculty of Dentistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
6
|
Huang S, Huang Y, Su T, Huang R, Su L, Wu Y, Weng S, He J, Xie J. Orange-spotted grouper nervous necrosis virus-encoded protein A induces interferon expression via RIG-I/MDA5-MAVS-TBK1-IRF3 signaling in fish cells. Microbiol Spectr 2024; 12:e0453222. [PMID: 38095472 PMCID: PMC10783131 DOI: 10.1128/spectrum.04532-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE As a major pathogen, nervous necrosis virus (NNV) infects more than 120 fish species worldwide and is virulent to larvae and juvenile fish, hampering the development of the fish fry industry. Understanding virus-host interaction and underlying mechanisms is an important but largely unknown issue in fish virus studies. Here, using channel catfish ovary and fathead minnow cells as models for the study of innate immunity signaling, we found that NNV-encoded ProA activated interferon signaling via the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) pathway which was still suppressed by the infection of wild-type NNV. This finding has important implications for the comprehension of NNV protein function and the immune response from different cells. First, RIG-I is the key node for anti-NNV innate immunity. Second, the response intensity of RLR signaling determines the degree of NNV proliferation. This study expands our knowledge regarding the overview of signal pathways affected by NNV-encoded protein and also highlights potential directions for the control of aquatic viruses.
Collapse
Affiliation(s)
- Siyou Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Yi Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Taowen Su
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Runqing Huang
- School of Life Science, Huizhou University, Huizhou, China
| | - Lianpan Su
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Yujia Wu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Jianguo He
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| | - Junfeng Xie
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), China-ASEAN Belt and Road Joint Laboratory on Mariculture Technology, Guangdong Provincial Key Laboratory of Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Liu R, Guo Y, Yu J, Wei X, Zhou F, Yuan X, Cai L, Yu C. Protective effect of N-(E)-p-coumaroyltyrosine on LPS-induced acute inflammatory injury and signaling pathway analysis. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109242. [PMID: 37995893 DOI: 10.1016/j.fsi.2023.109242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 09/13/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
N-trans-p-coumaroyltyrosine (N-(E)-p-coumaroyltyrosine, NPCT), extracted and purified from Abri Mollis Herba, is an amino acid amide. The defense mechanism of NPCT against inflammatory response is still unknown. In this study, lipopolysaccharide (LPS)-induced zebrafish acute inflammatory injury model was established to observe the inhibitory effect of NPCT on the aggregation of inflammatory cells in the yolk sac of zebrafish, as well as the inhibitory effect of NPCT on inflammatory and gas signaling factors. Results show that NPCT could inhibit inflammatory cell infiltration in zebrafish yolk sac, the migration and aggregation of macrophages and neutrophils to the site of inflammation, and the release of Nitric Oxide (NO) and Reactive Oxygen Species (ROS) in zebrafish, indicating that NPCT could substantially significantly prevent the development of LPS-induced acute systemic inflammation. In addition, the analysis results of RNA-seq showed that in the model group versus the administered group, the differentially expressed genes were mainly enriched to inflammatory signaling pathways, such as the NOD-like receptor signaling pathway and Toll-like receptor signaling pathway, which were down-regulated in the administered group. The TLR4, MyD88, IRAK4, NF-κB, IκB, NLRP3, Caspase-1, ASC, IL-1β, and IL-6 genes were significantly different in the transcripts, and the overall trend of the qPCR results was consistent with the transcriptome sequencing results. Therefore, NPCT had a significant inhibitory effect on LPS-induced acute inflammatory injury in zebrafish, and its anti-inflammatory mechanism may be through the regulation of key genes on the NOD-like receptor signaling pathway and Toll-like receptor signaling pathway, thereby affecting the release of relevant inflammatory cytokines.
Collapse
Affiliation(s)
- Roujia Liu
- Centre for Drug Research and Development, Guangdong Pharmaceutical University; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, East Waihuan Road 280, Guangzhou, PR China; Center of Human Microecology Engineering and Technology of Guangdong Province, Guangdong Longsee Biomedical Corporation, Guangzhou, PR China
| | - Yuhai Guo
- Department of Orthopedics, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, PR China
| | | | - Xinru Wei
- Centre for Drug Research and Development, Guangdong Pharmaceutical University; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, East Waihuan Road 280, Guangzhou, PR China
| | - Feirong Zhou
- Centre for Drug Research and Development, Guangdong Pharmaceutical University; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, East Waihuan Road 280, Guangzhou, PR China
| | - Xujiang Yuan
- Centre for Drug Research and Development, Guangdong Pharmaceutical University; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, East Waihuan Road 280, Guangzhou, PR China
| | - Lei Cai
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, PR China.
| | - Chuqin Yu
- Centre for Drug Research and Development, Guangdong Pharmaceutical University; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University; Guangdong Engineering & Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, East Waihuan Road 280, Guangzhou, PR China.
| |
Collapse
|
8
|
Dixon CL, Martin NR, Niphakis MJ, Cravatt BF, Fairn GD. Attenuating ABHD17 enhances S- palmitoylation, membrane localization and signal transduction of NOD2 and Crohn's disease-associated variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572362. [PMID: 38187608 PMCID: PMC10769251 DOI: 10.1101/2023.12.20.572362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
NOD2 is an intracellular innate immune receptor that senses bacterial peptidoglycans. Although soluble in the cytosol, a portion of the protein is associated with the plasma membrane and endosomal compartments for microbial surveillance. Palmitoylation of NOD2 by zDHHC5 promotes its membrane recruitment to drive proinflammatory and antimicrobial responses to pathogenic invasion. A depalmitoylation step by an unknown protein, thioesterase, releases NOD2 from membranes into the cytosol, where the protein can then enter a new cycle of palmitoylation-depalmitoylation. Here, we identify α/β -hydrolase domain-containing protein 17 isoforms (ABHD17A, 17B, 17C) as the thioesterases responsible for depalmitoylation of NOD2. Inhibiting ABHD17 increased the plasmalemmal localization of both wild-type NOD2 and a subset of hypo-palmitoylated Crohn's disease-associated variants, resulting in increased NF-κB activation and production of pro-inflammatory cytokines in epithelial cells. These results suggest that targeted inhibition of ABHD17 may rescue some Crohn's disease-associated NOD2 variants.
Collapse
|
9
|
Tao J, Liu G, Gu K, Jia G, Zhao H, Chen X, Tian G, Cai J, Wang J. Tryptophan alleviates intestinal damage through regulating necroptosis and TLR4/NOD signaling pathways in pigs after lipopolysaccharide challenge. Anim Biotechnol 2023; 34:4938-4946. [PMID: 37199152 DOI: 10.1080/10495398.2023.2213278] [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] [Indexed: 05/19/2023]
Abstract
This study aimed to test the hypothesis that necroptosis, toll-like receptor 4 (TLR4)/nucleotide-binding oligomerization domain (NOD) signaling pathway in the jejunum of lipopolysaccharide (LPS)-challenged piglets are involved in the alleviation of intestinal injury and inflammation by tryptophan supplementation. Tryptophan supplementation has improved intestinal morphology. Also, tryptophan has been found to increase the mRNA and protein expression of tight junction proteins and decrease the expression of pro-inflammatory cytokines. Dietary tryptophan decreased the mRNA expression of heat shock protein 70, TLR4, NOD1, NOD2, myeloid differentiation primary response gene 88, interleukin 1 receptor-associated kinase 1, TNF receptor-associated factor 6, receptor-interacting serine/threonine-protein kinase 2-like, nuclear factor-kappaB transcription factor P65 in the jejunum of piglets. Tryptophan alleviated LPS-induced necroptosis and decreased the mRNA expression of mixed lineage kinase domain-like, receptor-interacting serine/threonine kinase 1, receptor-interacting serine/threonine-protein kinase 3-like, Fas (TNFRSF6)-associated via death domain, PGAM family member 5. Collectively, our results suggest that tryptophan supplementation helps in the attenuation of intestinal injury and inflammation by alleviating necroptosis and TLR4/NOD in lipopolysaccharide-challenged pigs.
Collapse
Affiliation(s)
- Jingyuan Tao
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guangmang Liu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ke Gu
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Jia
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hua Zhao
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Tian
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jingyi Cai
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jing Wang
- Maize Research Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Zou C, Zan X, Jia Z, Zheng L, Gu Y, Liu F, Han Y, Xu C, Wu A, Zhi Q. Crosstalk between alternative splicing and inflammatory bowel disease: Basic mechanisms, biotechnological progresses and future perspectives. Clin Transl Med 2023; 13:e1479. [PMID: 37983927 PMCID: PMC10659771 DOI: 10.1002/ctm2.1479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 10/07/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Alternative splicing (AS) is an omnipresent regulatory mechanism of gene expression that enables the generation of diverse splice isoforms from a single gene. Recently, AS events have gained considerable momentum in the pathogenesis of inflammatory bowel disease (IBD). METHODS Our review has summarized the complex process of RNA splicing, and firstly highlighted the potential involved molecules that target aberrant splicing events in IBD. The quantitative transcriptome analyses such as microarrays, next-generation sequencing (NGS) for AS events in IBD have been also discussed. RESULTS Available evidence suggests that some abnormal splicing RNAs can lead to multiple intestinal disorders during the onset of IBD as well as the progression to colitis-associated cancer (CAC), including gut microbiota perturbations, intestinal barrier dysfunctions, innate/adaptive immune dysregulations, pro-fibrosis activation and some other risk factors. Moreover, current data show that the advanced technologies, including microarrays and NGS, have been pioneeringly employed to screen the AS candidates and elucidate the potential regulatory mechanisms of IBD. Besides, other biotechnological progresses such as the applications of third-generation sequencing (TGS), single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST), will be desired with great expectations. CONCLUSIONS To our knowledge, the current review is the first one to evaluate the potential regulatory mechanisms of AS events in IBD. The expanding list of aberrantly spliced genes in IBD along with the developed technologies provide us new clues to how IBD develops, and how these important AS events can be explored for future treatment.
Collapse
Affiliation(s)
- Chentao Zou
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xinquan Zan
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Zhenyu Jia
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lu Zheng
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yijie Gu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fei Liu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Ye Han
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chunfang Xu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Airong Wu
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Qiaoming Zhi
- Department of General SurgeryThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
11
|
Chen S, Lei Q, Zou X, Ma D. The role and mechanisms of gram-negative bacterial outer membrane vesicles in inflammatory diseases. Front Immunol 2023; 14:1157813. [PMID: 37398647 PMCID: PMC10313905 DOI: 10.3389/fimmu.2023.1157813] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Outer membrane vesicles (OMVs) are spherical, bilayered, and nanosized membrane vesicles that are secreted from gram-negative bacteria. OMVs play a pivotal role in delivering lipopolysaccharide, proteins and other virulence factors to target cells. Multiple studies have found that OMVs participate in various inflammatory diseases, including periodontal disease, gastrointestinal inflammation, pulmonary inflammation and sepsis, by triggering pattern recognition receptors, activating inflammasomes and inducing mitochondrial dysfunction. OMVs also affect inflammation in distant organs or tissues via long-distance cargo transport in various diseases, including atherosclerosis and Alzheimer's disease. In this review, we primarily summarize the role of OMVs in inflammatory diseases, describe the mechanism through which OMVs participate in inflammatory signal cascades, and discuss the effects of OMVs on pathogenic processes in distant organs or tissues with the aim of providing novel insights into the role and mechanism of OMVs in inflammatory diseases and the prevention and treatment of OMV-mediated inflammatory diseases.
Collapse
|
12
|
Cui T, Liu P, Chen X, Liu Z, Wang B, Gao C, Wang Z, Li C, Yang N. Identification and functional characterization of caspases in turbot (Scophthalmus maximus) in response to bacterial infection. FISH & SHELLFISH IMMUNOLOGY 2023; 137:108757. [PMID: 37084854 DOI: 10.1016/j.fsi.2023.108757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Apoptosis is the autonomous and orderly death of cells under genetic control to maintain the stability of the internal environment, and is a programmed cell death process with unique morphological and biochemical properties that is regulated by a variety of factors. Caspase gene family has a significant function in the process of apoptosis. However, the knowledge of caspases in turbot remains largely unknown. In present study, a total of nine turbot caspase genes were identified. The mRNA length of these caspase genes was ranged from 1149 bp (caspase-1) to 3216 bp (caspase-2), and the protein length was ranged from 281 aa (caspase-3a) to 507 aa (caspase-10). Phylogenetic analysis showed these caspase genes were divided into three subfamilies. The qRT-PCR results showed that turbot caspase genes were expressed in all the examined organs, especially the intestine, kidney, blood and gills. Meanwhile, we explored the expression patterns of caspase genes in the intestine, skin and gills after Vibrio anguillarum and Aeromonas salmonids infections. The results showed that caspase genes showed different expression patterns in mucosal tissues after bacterial infection, demonstrating the critical role of caspase genes in mucosal immune responses. In addition, protein-protein interaction analysis showed that caspase proteins interacted with immune molecules such as NLR, IL-1β, and birc. The results of interference and overexpression experiments showed that caspase-1 might play key roles in the regulation of the IL-1β production, but the detailed mechanism needs to be further studied. The results of this study provide valuable information for further study the roles of caspase genes in turbot, which could help us to further understand the inflammatory pathways in teleost.
Collapse
Affiliation(s)
- Tong Cui
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Peng Liu
- Yantai Marine Economic Research Institute, Yantai, China
| | - Xuan Chen
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhe Liu
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Beibei Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chengbin Gao
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhongyi Wang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chao Li
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| | - Ning Yang
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
13
|
Murphy MJ, Edemobi P, Leasure AC, Gulati M, Miller EJ, Damsky W, Cohen JM. Autoimmune comorbidities associated with sarcoidosis: a case-control study in the All of Us research program. Rheumatol Adv Pract 2023; 7:rkad030. [PMID: 38606002 PMCID: PMC11007907 DOI: 10.1093/rap/rkad030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/22/2023] [Indexed: 04/13/2024] Open
Abstract
Objective The degree to which sarcoidosis patients are affected by autoimmune diseases is poorly understood. Prior studies of autoimmune co-morbidities in sarcoidosis have focused on populations outside the USA or have been impeded by small sample sizes and limited scope. This case-control study evaluated the association between sarcoidosis and autoimmune diseases in a large, diverse cohort based in the USA. Methods We used data from the All of Us research programme to conduct a case-control study involving patients ≥18 years old, from 2018 to the present, diagnosed with sarcoidosis. Sarcoidosis cases and age-, sex- and race-matched controls were identified in a 1:4 ratio. Autoimmune co-morbidities were compared between sarcoidosis patients and controls in univariable and multivariable analyses using logistic regression. The degree of association was measured using the odds ratio (OR). Results A total of 1408 sarcoidosis cases and 5632 controls were included in this study. Seven of 24 examined autoimmune diseases were significantly associated with sarcoidosis in our multivariable analysis (P < 0.05). The composite variable of any autoimmune disease was also significantly associated with sarcoidosis (OR = 2.29, P < 0.001). Conclusion We demonstrate an association between sarcoidosis and multiple autoimmune diseases in a large and diverse cohort based in the USA. These results underscore the need for careful screening of sarcoidosis patients for concomitant autoimmune disease.
Collapse
Affiliation(s)
| | | | - Audrey C Leasure
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mridu Gulati
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Edward J Miller
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey M Cohen
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Song Y, He C, Jiang Y, Yang M, Xu Z, Yuan L, Zhang W, Xu Y. Bulk and single-cell transcriptome analyses of islet tissue unravel gene signatures associated with pyroptosis and immune infiltration in type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1132194. [PMID: 36967805 PMCID: PMC10034023 DOI: 10.3389/fendo.2023.1132194] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
INTRODUCTION Type 2 diabetes (T2D) is a common chronic heterogeneous metabolic disorder. However, the roles of pyroptosis and infiltrating immune cells in islet dysfunction of patients with T2D have yet to be explored. In this study, we aimed to explore potential crucial genes and pathways associated with pyroptosis and immune infiltration in T2D. METHODS To achieve this, we performed a conjoint analysis of three bulk RNA-seq datasets of islets to identify T2D-related differentially expressed genes (DEGs). After grouping the islet samples according to their ESTIMATE immune scores, we identified immune- and T2D-related DEGs. A clinical prediction model based on pyroptosis-related genes for T2D was constructed. Weighted gene co-expression network analysis was performed to identify genes positively correlated with pyroptosis-related pathways. A protein-protein interaction network was established to identify pyroptosis-related hub genes. We constructed miRNA and transcriptional networks based on the pyroptosis-related hub genes and performed functional analyses. Single-cell RNA-seq (scRNA-seq) was conducted using the GSE153885 dataset. Dimensionality was reduced using principal component analysis and t-distributed statistical neighbor embedding, and cells were clustered using Seurat. Different cell types were subjected to differential gene expression analysis and gene set enrichment analysis (GSEA). Cell-cell communication and pseudotime trajectory analyses were conducted using the samples from patients with T2D. RESULTS We identified 17 pyroptosis-related hub genes. We determined the abundance of 13 immune cell types in the merged matrix and found that these cell types were correlated with the 17 pyroptosis-related hub genes. Analysis of the scRNA-seq dataset of 1892 islet samples from patients with T2D and controls revealed 11 clusters. INS and IAPP were determined to be pyroptosis-related and candidate hub genes among the 11 clusters. GSEA of the 11 clusters demonstrated that the myc, G2M checkpoint, and E2F pathways were significantly upregulated in clusters with several differentially enriched pathways. DISCUSSION This study elucidates the gene signatures associated with pyroptosis and immune infiltration in T2D and provides a critical resource for understanding of islet dysfunction and T2D pathogenesis.
Collapse
Affiliation(s)
- Yaxian Song
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chen He
- Department of Geriatric Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yan Jiang
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mengshi Yang
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhao Xu
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lingyan Yuan
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenhua Zhang
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yushan Xu
- Department of Endocrinology, Yunnan Province Clinical Medical Center for Endocrine and Metabolic Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yushan Xu,
| |
Collapse
|
15
|
Kocaaga A, Cakmak Genc G, Karakas Celik S, Piskin İE, Calik M, Dursun A. Association of NOD1 and NOD2 Polymorphisms With Susceptibility to Subacute Sclerosing Panencephalitis. J Child Neurol 2022; 38:38-43. [PMID: 36544356 DOI: 10.1177/08830738221144081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background: Subacute sclerosing panencephalitis is a progressive neurodegenerative disease that is a late complication of measles infection. However, to date, the pathogenesis of subacute sclerosing panencephalitis is still not explained; both viral and host factors seem to be associated. The present study aimed to investigate the relationship between NOD1 and NOD2 gene variants and subacute sclerosing panencephalitis. Methods: The gene variants of NOD1 (rs2075820 and rs2075818) and NOD2 (R334Q and R334W) were explored in 64 subacute sclerosing panencephalitis patients and 70 controls using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Results: The frequencies of the AA genotype and A allele of rs2075820 (NOD1; c.796G>A) polymorphism were lower in patients compared with controls (P = .022 and .014, respectively). The presence of the A allele of rs2075820 may be considered as a protective factor for subacute sclerosing panencephalitis. There was a significant difference between the groups in rs2075818 (NOD1 G/C) polymorphism, and the CC genotype increased the risk of subacute sclerosing panencephalitis by 3.471-fold. The carriers of the C allele of rs2075818 (G/C) had a 1.855-fold susceptibility to subacute sclerosing panencephalitis (P = .018). The GC genotype might be associated with subacute sclerosing panencephalitis susceptibility in the patients compared with patients without having that haplotype (P = .03). Conclusions: Thus, we identified an association between subacute sclerosing panencephalitis and the rs2075820 (NOD1 G/A) and rs2075818 (NOD1 G/C) polymorphisms. These findings implicate a possible effect of this genetic polymorphism in susceptibility to subacute sclerosing panencephalitis, which needs to be confirmed in bigger populations.
Collapse
Affiliation(s)
- Ayça Kocaaga
- Department of Medical Genetics, Health Ministery Eskisehir City Hospital, Eskişehir, Turkey
| | - Gunes Cakmak Genc
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Sevim Karakas Celik
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - İbrahim E Piskin
- Department of Pediatrics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Mustafa Calik
- Department of Pediatric Neurology, Harran Univercity School of Medicine, Sanlıurfa, Turkey
| | - Ahmet Dursun
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| |
Collapse
|
16
|
Cosme D, Soares-da-Silva P, Magro F. Effect of Toll-like receptor-2, -4, -5, -7, and NOD2 stimulation on potassium channel conductance in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2022; 323:G410-G419. [PMID: 36040119 DOI: 10.1152/ajpgi.00139.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Disproportionate activation of pattern recognition receptors plays a role in inflammatory bowel disease (IBD) pathophysiology. Diarrhea is a hallmark symptom of IBD, resulting at least in part from an electrolyte imbalance that may be caused by changes in potassium channel activity. We evaluated the impact of Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain 2 (NOD2) stimulation on potassium conductance of the basolateral membrane in human intestinal epithelial cells (IECs) and the role of potassium channels through electrophysiological assays under short-circuit current in Ussing chambers. TLRs and NOD2 were stimulated using specific agonists, and potassium channels were selectively blocked using triarylmethane-34 (TRAM-34), adenylyl-imidodiphosphate (AMP-PNP), and BaCl2. Potassium conductance of the basolateral membrane decreased upon activation of TLR2, TLR4, and TLR7 in T84 cells (means ± SE, -11.2 ± 4.5, -40.4 ± 7.2, and -19.4 ± 5.9, respectively) and in Caco-2 cells (-13.1 ± 5.7, -55.7 ± 7.4, and -29.1 ± 7.2, respectively). In contrast, activation of TLR5 and NOD2 increased basolateral potassium conductance, both in T84 cells (18.0 ± 4.1 and 18.4 ± 2.8, respectively) and in Caco-2 cells (21.2 ± 8.4 and 16.0 ± 3.6, respectively). TRAM-34 and AMP-PNP induced a decrease in basolateral potassium conductance upon TLR4 stimulation in both cell lines. Both KCa3.1- and Kir6-channels appear to be important mediators of this effect in IECs and could be potential targets for therapeutic agent development.NEW & NOTEWORTHY This study highlights that PRRs stimulation directly influences K+-channel conductance in IECs. TLR-2, -4, -7 stimulation decreased K+ conductance, whereas TLR5 and NOD2 stimulation had the opposite effect, leading to an increase of it instead. This study reports for the first time that KCa3.1- and Kir6-channels play a role in K+ transport pathways triggered by TLR4 stimulation. These findings suggest that KCa3.1- and Kir6-channels modulation may be a potential target for new therapeutic agents in IBD.
Collapse
Affiliation(s)
- Dina Cosme
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP, Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Patrício Soares-da-Silva
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP, Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Fernando Magro
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Gastroenterology, São João Hospital University Centre, Porto, Portugal.,Center for Health Technology and Services Research, Porto, Portugal.,Clinical Pharmacology Unit, São João Hospital University Centre, Porto, Portugal.,Portuguese Inflammatory Bowel Disease Group, Porto, Portugal
| |
Collapse
|
17
|
M 6A RNA Methylation Mediates NOD1/NF-kB Signaling Activation in the Liver of Piglets Challenged with Lipopolysaccharide. Antioxidants (Basel) 2022; 11:antiox11101954. [PMID: 36290677 PMCID: PMC9598714 DOI: 10.3390/antiox11101954] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal modification that widely participates in various immune and inflammatory responses; however, its regulatory mechanisms in the inflammation of liver induced by lipopolysaccharide in piglets remain largely unknown. In the present study, piglets were intraperitoneally injected with 80 μg/kg LPS or an equal dose of sterile saline. Results indicated that LPS administration increased activities of serum alanine aminotransferase (ALT), induced M1 macrophage polarization and promoted secretion of inflammatory cytokines, and finally led to hepatic lesions in piglets. The NOD1/NF-κB signaling pathway was activated in the livers of the LPS group. Moreover, the total m6A level was significantly elevated after LPS treatment. MeRIP-seq showed that 1166 and 1344 transcripts contained m6A methylation in control and LPS groups, respectively. The m6A methylation sites of these transcripts mainly distributes in the 5′ untranslated region (5′UTR), the coding sequence (CDS), and the 3′ untranslated region (3′UTR). Interestingly, these genes were mostly enriched in the NF-κB signaling pathway, and LPS treatment significantly changed the m6A modification in NOD1, RIPK2, NFKBIA, NFKBIB, and TNFAIP3 mRNAs. In addition, knockdown of METTL3 or overexpression of FTO both changed gene levels in the NOD1/NF-κB pathway, suggesting that activation of this pathway was regulated by m6A RNA methylation. Moreover, the alteration of m6A RNA methylation profile may be associated with the increase of reactive oxygen species (ROS), HIF-1α, and MAT2A. In conclusion, LPS activated the NOD1/NF-κB pathway at post-transcriptional regulation through changing m6A RNA methylation, and then promoted the overproduction of proinflammatory cytokines, ultimately resulting in liver inflammation and damage.
Collapse
|
18
|
Mao L, Dhar A, Meng G, Fuss I, Montgomery-Recht K, Yang Z, Xu Q, Kitani A, Strober W. Blau syndrome NOD2 mutations result in loss of NOD2 cross-regulatory function. Front Immunol 2022; 13:988862. [PMID: 36189261 PMCID: PMC9520668 DOI: 10.3389/fimmu.2022.988862] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/29/2022] [Indexed: 12/02/2022] Open
Abstract
The studies described here provide an analysis of the pathogenesis of Blau syndrome and thereby the function of NOD2 as seen through the lens of its dysfunction resulting from Blau-associated NOD2 mutations in its nucleotide-binding domain (NBD). As such, this analysis also sheds light on the role of NOD2 risk polymorphisms in the LRR domain occurring in Crohn’s disease. The main finding was that Blau NOD2 mutations precipitate a loss of canonical NOD2 signaling via RIPK2 and that this loss has two consequences: first, it results in defective NOD2 ligand (MDP)-mediated NF-κB activation and second, it disrupts NOD2-mediated cross-regulation whereby NOD2 downregulates concomitant innate (TLR) responses. Strong evidence is also presented favoring the view that NOD2-mediated cross-regulation is under mechanistic control by IRF4 and that failure to up-regulate this factor because of faulty NOD2 signaling is the proximal cause of defective cross-regulation and the latter’s effect on Blau syndrome inflammation. Overall, these studies highlight the role of NOD2 as a regulatory factor and thus provide additional insight into its function in inflammatory disease. Mutations in the nucleotide binding domain of the CARD15 (NOD2) gene underlie the granulomatous inflammation characterizing Blau syndrome (BS). In studies probing the mechanism of this inflammation we show here that NOD2 plasmids expressing various Blau mutations in HEK293 cells result in reduced NOD2 activation of RIPK2 and correspondingly reduced NOD2 activation of NF-κB. These in vitro studies of NOD2 signaling were accompanied by in vivo studies showing that BS-NOD2 also exhibit defects in cross-regulation of innate responses underlying inflammation. Thus, whereas over-expressed intact NOD2 suppresses TNBS-colitis, over-expressed BS-NOD2 does not; in addition, whereas administration of NOD2 ligand (muramyl dipeptide, MDP) suppresses DSS-colitis in Wild Type (WT) mice it fails to do so in homozygous or heterozygous mice bearing a NOD2 Blau mutation. Similarly, mice bearing a Blau mutation exhibit enhanced anti-collagen antibody-induced arthritis. The basis of such cross-regulatory failure was revealed in studies showing that MDP-stimulated cells bearing BS-NOD2 exhibit a reduced capacity to signal via RIPK2 as well as a reduced capacity to up-regulate IRF4, a factor shown previously to mediate NOD2 suppression of NF-κB activation. Indeed, TLR-stimulated cells bearing a Blau mutation exhibited enhanced in vitro cytokine responses that are quieted by lentivirus transduction of IRF4. In addition, enhanced anti-collagen-induced joint inflammation in mice bearing a Blau mutation was accompanied by reduced IRF4 expression in inflamed joint tissue and IRF4 expression was reduced in MDP-stimulated cells from BS patients. Thus, inflammation characterizing Blau syndrome are caused, at least in part, by faulty canonical signaling and reduce IRF4-mediated cross-regulation.
Collapse
Affiliation(s)
- Liming Mao
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Atika Dhar
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Guangxun Meng
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai, China
- Pasteurien College, Soochow University, Suzhou, China
| | - Ivan Fuss
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kim Montgomery-Recht
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute (NCI) Campus at Frederick, Frederick, MD, United States
| | - Zhiqiong Yang
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Atsushi Kitani
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Warren Strober
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Warren Strober,
| |
Collapse
|
19
|
Kozyra P, Pitucha M. Terminal Phenoxy Group as a Privileged Moiety of the Drug Scaffold-A Short Review of Most Recent Studies 2013-2022. Int J Mol Sci 2022; 23:8874. [PMID: 36012142 PMCID: PMC9408176 DOI: 10.3390/ijms23168874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/03/2022] [Accepted: 08/07/2022] [Indexed: 11/16/2022] Open
Abstract
The terminal phenoxy group is a moiety of many drugs in use today. Numerous literature reports indicated its crucial importance for biological activity; thus, it is a privileged scaffold in medicinal chemistry. This review focuses on the latest achievements in the field of novel potential agents bearing a terminal phenoxy group in 2013-2022. The article provided information on neurological, anticancer, potential lymphoma agent, anti-HIV, antimicrobial, antiparasitic, analgesic, anti-diabetic as well as larvicidal, cholesterol esterase inhibitors, and antithrombotic or agonistic activities towards the adrenergic receptor. Additionally, for selected agents, the Structure-Activity-Relationship (SAR) is also discussed. Thus, this study may help the readers to better understand the nature of the phenoxy group, which will translate into rational drug design and the development of a more efficient drug. To the best of our knowledge, this is the first review devoted to an in-depth analysis of the various activities of compounds bearing terminal phenoxy moiety.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
20
|
Tang Y, Zhang P, Liu Q, Cao L, Xu J. Pyroptotic Patterns in Blood Leukocytes Predict Disease Severity and Outcome in COVID-19 Patients. Front Immunol 2022; 13:888661. [PMID: 35928821 PMCID: PMC9343985 DOI: 10.3389/fimmu.2022.888661] [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: 03/03/2022] [Accepted: 06/17/2022] [Indexed: 12/04/2022] Open
Abstract
The global coronavirus disease 2019 (COVID-19) pandemic has lasted for over 2 years now and has already caused millions of deaths. In COVID-19, leukocyte pyroptosis has been previously associated with both beneficial and detrimental effects, so its role in the development of this disease remains controversial. Using transcriptomic data (GSE157103) of blood leukocytes from 126 acute respiratory distress syndrome patients (ARDS) with or without COVID-19, we found that COVID-19 patients present with enhanced leukocyte pyroptosis. Based on unsupervised clustering, we divided 100 COVID-19 patients into two clusters (PYRcluster1 and PYRcluster2) according to the expression of 35 pyroptosis-related genes. The results revealed distinct pyroptotic patterns associated with different leukocytes in these PYRclusters. PYRcluster1 patients were in a hyperinflammatory state and had a worse prognosis than PYRcluster2 patients. The hyperinflammation of PYRcluster1 was validated by the results of gene set enrichment analysis (GSEA) of proteomic data (MSV000085703). These differences in pyroptosis between the two PYRclusters were confirmed by the PYRscore. To improve the clinical treatment of COVID-19 patients, we used least absolute shrinkage and selection operator (LASSO) regression to construct a prognostic model based on differentially expressed genes between PYRclusters (PYRsafescore), which can be applied as an effective prognosis tool. Lastly, we explored the upstream transcription factors of different pyroptotic patterns, thereby identifying 112 compounds with potential therapeutic value in public databases.
Collapse
Affiliation(s)
- Yingkui Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peidong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyu Liu
- Department of Critical Care Medicine, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Luyang Cao
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
- *Correspondence: Jingsong Xu, ; Luyang Cao,
| | - Jingsong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou, China
- *Correspondence: Jingsong Xu, ; Luyang Cao,
| |
Collapse
|
21
|
Zveik O, Rechtman A, Haham N, Adini I, Canello T, Lavon I, Brill L, Vaknin-Dembinsky A. Sera of Neuromyelitis Optica Patients Increase BID-Mediated Apoptosis in Astrocytes. Int J Mol Sci 2022; 23:ijms23137117. [PMID: 35806122 PMCID: PMC9266359 DOI: 10.3390/ijms23137117] [Citation(s) in RCA: 3] [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: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/30/2022] Open
Abstract
Neuromyelitis optica (NMO) is a rare disease usually presenting with bilateral or unilateral optic neuritis with simultaneous or sequential transverse myelitis. Autoantibodies directed against aquaporin-4 (AQP4-IgG) are found in most patients. They are believed to cross the blood−brain barrier, target astrocytes, activate complement, and eventually lead to astrocyte destruction, demyelination, and axonal damage. However, it is still not clear what the primary pathological event is. We hypothesize that the interaction of AQP4-IgG and astrocytes leads to DNA damage and apoptosis. We studied the effect of sera from seropositive NMO patients and healthy controls (HCs) on astrocytes’ immune gene expression and viability. We found that sera from seropositive NMO patients led to higher expression of apoptosis-related genes, including BH3-interacting domain death agonist (BID), which is the most significant differentiating gene (p < 0.0001), and triggered more apoptosis in astrocytes compared to sera from HCs. Furthermore, NMO sera increased DNA damage and led to a higher expression of immunological genes that interact with BID (TLR4 and NOD-1). Our findings suggest that sera of seropositive NMO patients might cause astrocytic DNA damage and apoptosis. It may be one of the mechanisms implicated in the primary pathological event in NMO and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Omri Zveik
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Ariel Rechtman
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Nitzan Haham
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Irit Adini
- Department of Surgery, Harvard Medical School, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, 51 Blossom Street, Boston, MA 02114, USA;
| | - Tamar Canello
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Iris Lavon
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Leslie and Michael Gaffin Center for Neuro-Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Livnat Brill
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (O.Z.); (A.R.); (N.H.); (T.C.); (I.L.); (L.B.)
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91120, Israel
- Correspondence: ; Tel.: +972-2-677-7741
| |
Collapse
|
22
|
Li T, Liu H, Dong C, Lyu J. Prognostic Implications of Pyroptosis-Related Gene Signatures in Lung Squamous Cell Carcinoma. Front Pharmacol 2022; 13:806995. [PMID: 35153782 PMCID: PMC8829032 DOI: 10.3389/fphar.2022.806995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/10/2022] [Indexed: 01/10/2023] Open
Abstract
Background: Lung squamous cell carcinoma (LUSC) has been a highly malignant tumor with very poor prognosis. It is confirmed that pyroptosis refers to the deaths of cells in a programmed and inflammatory manner. Nevertheless, the correlation between expression of genes related with pyroptosis and their prognosis remains uncertain in LUSC. Methods: Utilization of The Cancer Genome Atlas (TCGA) cohort has been done for evaluating the prognostics of pyroptosis-related genes for survival and constructing a signature with multiple genes. The least absolute shrinkage and selection operator (LASSO) Cox regression was performed for establishing such pyroptosis-related gene signature. Results: Eventually, identification of 28 genes in relation to pyroptosis was made in LUSC and healthy lung tissues. Upon the basis of these differentially-expressed genes (DEGs), the patients of LUSC can be divided into two subtypes. Nine gene signatures were established using LASSO. The surviving rate for low-risk group was apparently greater in contrast with the high-risk group (p < .001). According to our finding, risk score worked as an independent predictive factor of OS among LUSC sufferers in combination with clinical characteristics. In line with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, the enrichment of immunity-related genes and decreasing immunity status among the high-risk group. Conclusion: Genes in relation with pyroptosis played an essential role in tumor immunity, which is capable of predicting the prognosis for LUSCs.
Collapse
Affiliation(s)
- Tingting Li
- Department of Pharmacy, Xi'an Chest Hospital, Xi'an, China
| | - Huanqing Liu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chunsheng Dong
- School of Computer Science, Shaanxi Normal University, Xi'an, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
23
|
Toll-Like Receptors (TLRs), NOD-Like Receptors (NLRs), and RIG-I-Like Receptors (RLRs) in Innate Immunity. TLRs, NLRs, and RLRs Ligands as Immunotherapeutic Agents for Hematopoietic Diseases. Int J Mol Sci 2021; 22:ijms222413397. [PMID: 34948194 PMCID: PMC8704656 DOI: 10.3390/ijms222413397] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023] Open
Abstract
The innate immune system plays a pivotal role in the first line of host defense against infections and is equipped with patterns recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Several classes of PRRS, including Toll-like receptors (TLRs), NOD-like receptors (NLRs), and RIG-I-like receptors (RLRs) recognize distinct microbial components and directly activate immune cells. TLRs are transmembrane receptors, while NLRs and RLRs are intracellular molecules. Exposure of immune cells to the ligands of these receptors activates intracellular signaling cascades that rapidly induce the expression of a variety of overlapping and unique genes involved in the inflammatory and immune responses. The innate immune system also influences pathways involved in cancer immunosurveillance. Natural and synthetic agonists of TLRs, NLRs, or RLRs can trigger cell death in malignant cells, recruit immune cells, such as DCs, CD8+ T cells, and NK cells, into the tumor microenvironment, and are being explored as promising adjuvants in cancer immunotherapies. In this review, we provide a concise overview of TLRs, NLRs, and RLRs: their structure, functions, signaling pathways, and regulation. We also describe various ligands for these receptors and their possible application in treatment of hematopoietic diseases.
Collapse
|
24
|
Chen L, Cao SQ, Lin ZM, He SJ, Zuo JP. NOD-like receptors in autoimmune diseases. Acta Pharmacol Sin 2021; 42:1742-1756. [PMID: 33589796 PMCID: PMC8564530 DOI: 10.1038/s41401-020-00603-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/18/2020] [Indexed: 01/31/2023] Open
Abstract
Autoimmune diseases are chronic immune diseases characterized by dysregulation of immune system, which ultimately results in a disruption in self-antigen tolerance. Cumulative data show that nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) play essential roles in various autoimmune diseases, such as inflammatory bowel disease (IBD), rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), psoriasis, multiple sclerosis (MS), etc. NLR proteins, consisting of a C-terminal leucine-rich repeat (LRR), a central nucleotide-binding domain, and an N-terminal effector domain, form a group of pattern recognition receptors (PRRs) that mediate the immune response by specifically recognizing cellular pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) and triggering numerous signaling pathways, including RIP2 kinase, caspase-1, nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (MAPK) and so on. Based on their N-terminal domain, NLRs are divided into five subfamilies: NLRA, NLRB, NLRC, NLRP, and NLRX1. In this review, we briefly describe the structures and signaling pathways of NLRs, summarize the recent progress on NLR signaling in the occurrence and development of autoimmune diseases, as well as highlight numerous natural products and synthetic compounds targeting NLRs for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Li Chen
- grid.9227.e0000000119573309Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Shi-qi Cao
- grid.9227.e0000000119573309Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Ze-min Lin
- grid.9227.e0000000119573309Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Shi-jun He
- grid.9227.e0000000119573309Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jian-ping Zuo
- grid.9227.e0000000119573309Laboratory of Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.412540.60000 0001 2372 7462Laboratory of Immunology and Virology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203 China
| |
Collapse
|
25
|
P R, Ramireddy S, Chakraborty S, Mukherjee S, J S, C S. Structural localization of pathogenic mutations in the central nucleotide-binding domain (NBD) of nucleotide-binding oligomerization domain-2 (NOD2) protein and their inference in inflammatory disorders. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2021; 40:1198-1219. [PMID: 34622739 DOI: 10.1080/15257770.2021.1986719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The human NBD domain which is centrally located in the NOD2 protein displays an essential role in oligomerization and initiates the immune response via CARD-RIPK2 interaction. The mutations associated with the NBD domain have been largely implicated in inflammatory disorders such as Blau syndrome and sarcoidosis. This study aims to determine the structural and phenotypic effect of a lethal mutation that occurs in the NBD domain which has an axiomatic impact on protein dysfunction. Initially, the most deleterious missense mutations were screened through various in silico analysis. Out of 33 variants, I-Mutant 3.0, SIFT, PolyPhen 2, Align GVGD, PHD SNP and SNP&GO have statistically identified 5 variants (R42W, D90E, E91K, G189D & W198L) as less stable, deleterious and damaging. Our predicted models have paved the way to understand the various structural properties such as physiochemical, secondary structural arrangements and stabilizing residues in folding associated with the native and mutant NBD domain especially of the functionally important regions. From the aforementioned results, R42W and G189D were found to be the more predominant among the mutants. Precisely, through molecular simulation, we have strongly justified the significant conformational disruption of R42W and G189D through the stabilization factors, folding and essential dynamics. Conclusively, these regions (α341-44, α13185-191 and β6133-143β7) seem to adopt such structures that are not conducive to wild-type-like functionality. Our prediction and validation of lethal mutations based on structural stability may be useful for conducting experimental studies in detail to uncover the protein deregulation leading to inflammatory disorders.
Collapse
Affiliation(s)
- Raghuraman P
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Sriroopreddy Ramireddy
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Sulagno Chakraborty
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Sayani Mukherjee
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Sreeshma J
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Sudandiradoss C
- Department of Biotechnology, School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
26
|
Eshleman EM, Alenghat T. Epithelial sensing of microbiota-derived signals. Genes Immun 2021; 22:237-246. [PMID: 33824498 PMCID: PMC8492766 DOI: 10.1038/s41435-021-00124-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 02/01/2023]
Abstract
The gastrointestinal tract harbors trillions of microbial species, collectively termed the microbiota, which establish a symbiotic relationship with the host. Decades of research have emphasized the necessity of microbial signals in the development, maturation, and function of host physiology. However, changes in the composition or containment of the microbiota have been linked to the development of several chronic inflammatory diseases, including inflammatory bowel diseases. Intestinal epithelial cells (IECs) are in constant contact with the microbiota and are critical for maintaining intestinal homeostasis. Signals from the microbiota are directly sensed by IECs and influence intestinal health by calibrating immune cell responses and fortifying intestinal barrier function. IECs detect commensal microbes through engagement of common pattern recognition receptors or by sensing the production of microbial-derived metabolites. Deficiencies in these microbial-detecting pathways in IECs leads to impaired epithelial barrier function and altered intestinal homeostasis. This Review aims to highlight the pathways by which IECs sense microbiota-derived signals and the necessity of these detection pathways in maintaining epithelial barrier integrity.
Collapse
Affiliation(s)
- Emily M Eshleman
- Division of Immunobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Theresa Alenghat
- Division of Immunobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
27
|
Kocaaga A, Cakmak Genc G, Karakas Celık S, Koca R, Dursun A. Association of NOD1, NOD2, PYDC1 and PYDC2 genes with Behcet's disease susceptibility and clinical manifestations. Ophthalmic Genet 2021; 42:691-697. [PMID: 34294014 DOI: 10.1080/13816810.2021.1955273] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Purpose: Behçet's disease (BD) is an autoinflammatory disease with clinical manifestations such as mucocutaneous, ocular, vascular, gastrointestinal, musculoskeletal and central nervous system involvement. Features of innate and adaptive immunity and inflammasome pathways have been claimed in the pathogenesis of BD. We aimed to investigate the roles of NOD1, NOD2, PYDC1 and PYDC2 genes in the genetic predisposition of BD.Materials and Methods: Genetic variations of NOD1 (rs2075820 and rs2075818) and NOD2 (R334Q and R334W) genes were explored in 68 BD patients and 70 controls with PCR-RFLP (polymerase chain reaction-restriction fragment length polymorphism) approach. PYDC1 and PYDC2 gene variants were investigated by Sanger sequencing.Results: The polymorphism of rs2075820 (NOD1 G/A) had a statistically significant difference between the BD and controls, AA genotype was 2.460-fold protective. When compared in terms of cardiovascular involvement in BD patients, AA genotype was increased the risk of cardiovascular involvement 4.286-fold. There was a significant difference between BD and controls in rs2075818 (NOD1 G/C) polymorphism and CC genotype increased the risk of BD by 3.780-fold. In terms of rs2075818 variants, there was a statistically significant difference between BD patients with ocular lesions, joints, cardiovascular and gastrointestinal involvement and controls. There was a significant difference between the patients with joint involvement and controls and the risk increased of 3.310-fold.Conclusion: The data shed new light on the association between polymorphisms of NOD1 gene and BD and clinicial manifestations. However, NOD2, PYDC1 and PYDC2 genes were not associated with BD in the Turkish population.
Collapse
Affiliation(s)
- Ayca Kocaaga
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Gunes Cakmak Genc
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Sevim Karakas Celık
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Rafet Koca
- Department of Dermatology, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| | - Ahmet Dursun
- Department of Medical Genetics, Zonguldak Bülent Ecevit University Health Practice and Research Center, Zonguldak, Turkey
| |
Collapse
|
28
|
Suresh M, Li B, Murreddu MG, Gudima SO, Menne S. Involvement of Innate Immune Receptors in the Resolution of Acute Hepatitis B in Woodchucks. Front Immunol 2021; 12:713420. [PMID: 34367179 PMCID: PMC8340647 DOI: 10.3389/fimmu.2021.713420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022] Open
Abstract
The antiviral property of small agonist compounds activating pattern recognition receptors (PRRs), including toll-like and RIG-I receptors, have been preclinically evaluated and are currently tested in clinical trials against chronic hepatitis B (CHB). The involvement of other PRRs in modulating hepatitis B virus infection is less known. Thus, woodchucks with resolving acute hepatitis B (AHB) after infection with woodchuck hepatitis virus (WHV) were characterized as animals with normal or delayed resolution based on their kinetics of viremia and antigenemia, and the presence and expression of various PRRs were determined in both outcomes. While PRR expression was unchanged immediately after infection, most receptors were strongly upregulated during resolution in liver but not in blood. Besides well-known PRRs, including TLR7/8/9 and RIG-I, other less-characterized receptors, such as IFI16, ZBP1/DAI, AIM2, and NLRP3, displayed comparable or even higher expression. Compared to normal resolution, a 3-4-week lag in peak receptor expression and WHV-specific B- and T-cell responses were noted during delayed resolution. This suggested that PRR upregulation in woodchuck liver occurs when the mounting WHV replication reaches a certain level, and that multiple receptors are involved in the subsequent induction of antiviral immune responses. Liver enzyme elevations occurred early during normal resolution, indicating a faster induction of cytolytic mechanisms than in delayed resolution, and correlated with an increased expression of NK-cell and CD8 markers and cytolytic effector molecules. The peak liver enzyme level, however, was lower during delayed resolution, but hepatic inflammation was more pronounced and associated with a higher expression of cytolytic markers. Further comparison of PRR expression revealed that most receptors were significantly reduced in woodchucks with established and progressing CHB, and several RNA sensors more so than DNA sensors. This correlated with a lower expression of receptor adaptor and effector molecules, suggesting that persistent, high-level WHV replication interferes with PRR activation and is associated with a diminished antiviral immunity based on the reduced expression of immune cell markers, and absent WHV-specific B- and T-cell responses. Overall, the differential expression of PRRs during resolution and persistence of WHV infection emphasizes their importance in the ultimate viral control during AHB that is impaired during CHB.
Collapse
Affiliation(s)
- Manasa Suresh
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Bin Li
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Marta G. Murreddu
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Severin O. Gudima
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Stephan Menne
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
29
|
Shehata WA, Shoeib M, Shoeib MM, Shokhba H, Shams A. Nucleotide binding and oligomerization domain 2 in psoriasis: a clinical and immunohistochemical study. J Immunoassay Immunochem 2021; 43:43-53. [PMID: 34137669 DOI: 10.1080/15321819.2021.1941095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Psoriasis is a chronic immune-mediated inflammatory disease, affecting about 2 to 3% of the population worldwide. Nucleotide-binding and oligomerization domain 2-like receptor has been implicated in the pathogenesis of different inflammatory diseases. The current work aims to investigate the expression of nucleotide-binding and oligomerization domain 2-like receptor in psoriatic skin through an immunohistochemical study. This cross-sectional case-control study included 20 patients with chronic plaque psoriasis and 20 age- and sex-matched normal subjects as controls. Psoriasis severity was assessed through the use of Psoriasis Area Severity Index (PASI) score. Skin biopsies were taken under local anesthesia from cases and from matched sites of controls. Expression of nucleotide-binding and oligomerization domain 2 in epidermis of studied cases and controls showed positive epidermal expression of nucleotide-binding and oligomerization domain 2 in all cases (100%) versus 6 (30%) controls with a significant increase (χ2 = 21.54, P˂0.001). Moreover, dermal expression of nucleotide-binding and oligomerization domain 2 was higher in psoriatic skin lesion (95%) compared to controls (15%) with a significant difference (χ2 = 25.86, P˂0.001). We concluded that nucleotide-binding and oligomerization domain 2 may be implicated in psoriasis pathogenesis being higher in cases in comparison to controls.
Collapse
Affiliation(s)
- Wafaa Ahmed Shehata
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufia University, Shibn Al Kawm, Egypt
| | - Mohamed Shoeib
- Department of Dermatology, Andrology and STDs, Faculty of Medicine, Menoufia University, Shibn Al Kawm, Egypt
| | - May Mohamed Shoeib
- Department of Dermatology, Researcher at the National Research Center, Cairo, Egypt
| | - Hend Shokhba
- General Practitioner in Health Sector, Cairo, Egypt
| | - Asmaa Shams
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibn Al Kawm, Egypt
| |
Collapse
|
30
|
Kaur G, Iyer LM, Burroughs AM, Aravind L. Bacterial death and TRADD-N domains help define novel apoptosis and immunity mechanisms shared by prokaryotes and metazoans. eLife 2021; 10:70394. [PMID: 34061031 PMCID: PMC8195603 DOI: 10.7554/elife.70394] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 05/23/2021] [Indexed: 12/12/2022] Open
Abstract
Several homologous domains are shared by eukaryotic immunity and programmed cell-death systems and poorly understood bacterial proteins. Recent studies show these to be components of a network of highly regulated systems connecting apoptotic processes to counter-invader immunity, in prokaryotes with a multicellular habit. However, the provenance of key adaptor domains, namely those of the Death-like and TRADD-N superfamilies, a quintessential feature of metazoan apoptotic systems, remained murky. Here, we use sensitive sequence analysis and comparative genomics methods to identify unambiguous bacterial homologs of the Death-like and TRADD-N superfamilies. We show the former to have arisen as part of a radiation of effector-associated α-helical adaptor domains that likely mediate homotypic interactions bringing together diverse effector and signaling domains in predicted bacterial apoptosis- and counter-invader systems. Similarly, we show that the TRADD-N domain defines a key, widespread signaling bridge that links effector deployment to invader-sensing in multicellular bacterial and metazoan counter-invader systems. TRADD-N domains are expanded in aggregating marine invertebrates and point to distinctive diversifying immune strategies probably directed both at RNA and retroviruses and cellular pathogens that might infect such communities. These TRADD-N and Death-like domains helped identify several new bacterial and metazoan counter-invader systems featuring underappreciated, common functional principles: the use of intracellular invader-sensing lectin-like (NPCBM and FGS), transcription elongation GreA/B-C, glycosyltransferase-4 family, inactive NTPase (serving as nucleic acid receptors), and invader-sensing GTPase switch domains. Finally, these findings point to the possibility of multicellular bacteria-stem metazoan symbiosis in the emergence of the immune/apoptotic systems of the latter.
Collapse
Affiliation(s)
- Gurmeet Kaur
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - Lakshminarayan M Iyer
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - A Maxwell Burroughs
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| | - L Aravind
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, United States
| |
Collapse
|
31
|
Nod-Like Receptors in Host Defence and Disease at the Epidermal Barrier. Int J Mol Sci 2021; 22:ijms22094677. [PMID: 33925158 PMCID: PMC8124564 DOI: 10.3390/ijms22094677] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/18/2021] [Accepted: 04/27/2021] [Indexed: 12/16/2022] Open
Abstract
The nucleotide-binding domain and leucine-rich-repeat-containing family (NLRs) (sometimes called the NOD-like receptors, though the family contains few bona fide receptors) are a superfamily of multidomain-containing proteins that detect cellular stress and microbial infection. They constitute a critical arm of the innate immune response, though their functions are not restricted to pathogen recognition and members engage in controlling inflammasome activation, antigen-presentation, transcriptional regulation, cell death and also embryogenesis. NLRs are found from basal metazoans to plants, to zebrafish, mice and humans though functions of individual members can vary from species to species. NLRs also display highly wide-ranging tissue expression. Here, we discuss the importance of NLRs to the immune response at the epidermal barrier and summarise the known role of individual family members in the pathogenesis of skin disease.
Collapse
|
32
|
He J, Meng Z, Lu D, Liu X, Lin H. Recognition of DAP and activation of NF-κB by cytosolic sensor NOD1 in Oreochromis niloticus. FISH & SHELLFISH IMMUNOLOGY 2021; 110:75-85. [PMID: 33444736 DOI: 10.1016/j.fsi.2020.11.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/12/2020] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
As a lower vertebrate, the immune defense mechanism of fish mainly depends on the innate immune system. Nucleotide-binding oligomerization domain-like receptors (NLRs) are an important class of pattern recognition receptors in the innate immune system. In this study, NOD1 gene was cloned and characterized in Nile tilapia (Oreochromis niloticus). The ORF of Nile tilapia NOD1 gene was 2826 bp long and encoded 941 amino acid residues with a structure of CARD-NACHT-LRRs that was similar to the other counterparts in mammals and fishes. Phylogenetic and synteny analysis showed that NOD1 was conserved among different fishes and existed at least in the early stage of fish evolution. Expression pattern revealed that NOD1 mRNA was constitutively expressed in the tested tissues, while had high expression level in main immune organs and mucosal immune tissues (liver, head kidney, spleen, blood, gill, and intestine). Following Streptococcus agalactiae challenge, Nile tilapia NOD1 mRNA expression levels were altered in immune organs (liver, head kidney, spleen, blood), and the expression pattern was similar in liver, spleen and blood. Furthermore, the ligand recognition and signaling pathway of Nile tilapia NOD1 were also analyzed, it showed that NOD1 could recognize Tri-DAP intracellularly and activated NF-κB signaling pathway. In summary, our results indicated that the Nile tilapia NOD1 may play an important role in innate immune system and provided a basis for the functional study of NOD1 in teleost.
Collapse
Affiliation(s)
- Jianan He
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Zining Meng
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China; Southern Laboratory of Ocean Science and Engineering, Zhuhai, 519000, China
| | - Danqi Lu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| | - Xiaochun Liu
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China; Southern Laboratory of Ocean Science and Engineering, Zhuhai, 519000, China.
| | - Haoran Lin
- State Key Laboratory of Biocontrol, Guangdong Province Key Laboratory for Improved Variety Reproduction of Aquatic Economic Animals, Institute of Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| |
Collapse
|
33
|
Kozlov EM, Ivanova E, Grechko AV, Wu WK, Starodubova AV, Orekhov AN. Involvement of Oxidative Stress and the Innate Immune System in SARS-CoV-2 Infection. Diseases 2021; 9:diseases9010017. [PMID: 33668325 PMCID: PMC8005963 DOI: 10.3390/diseases9010017] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/13/2022] Open
Abstract
The emergence of the novel coronavirus in December 2019 in China marked the beginning of a pandemic that impacted healthcare systems and economic life all over the world. The virus primarily targets the respiratory system causing severe acute respiratory syndrome (SARS) in some patients, and therefore received the name of SARS-CoV-2. The pathogen stands out among other coronaviruses by its rapid transmission from human to human, with the majority of infected individuals being asymptomatic or presenting with only minor illness, therefore facilitating the pathogen spread. At the same time, people from the risk groups, such as the elderly, patients suffering from chronic diseases, or obese individuals, have increased chances of developing a severe or even fatal disease. The search for risk factors explaining this phenomenon continues. In this review, we focus on the known mechanisms of SARS-CoV-2 infection affecting the functioning of the immune system and discuss potential risk factors responsible for the severe disease course. Oxidative stress is one of such factors, which plays a prominent role in innate immunity activity, and recent research has revealed its tight involvement in SARS-CoV-2 infection. We discuss these recent findings and the development of excessive inflammation and cytokine storm observed during SARS-CoV-2 infection. Finally, we consider potential use of antioxidant drugs for alleviating the severe symptoms in affected patients.
Collapse
Affiliation(s)
- Evgenii M. Kozlov
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Ekaterina Ivanova
- Department of Basic Research, Institute of Atherosclerosis Research, 121609 Moscow, Russia
- Correspondence: ; Tel./Fax: +7-(495)4159594
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Wei-Kai Wu
- Department of Medical Research, National Taiwan University Hospital, Taipei 10617, Taiwan;
| | - Antonina V. Starodubova
- Federal Research Centre for Nutrition, Biotechnology and Food Safety, 2/14 Ustinsky Passage, 109240 Moscow, Russia;
- Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, 117997 Moscow, Russia
| | - Alexander N. Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Laboratory of Infectious Pathology and Molecular Microecology, Institute of Human Morphology, 117418 Moscow, Russia
| |
Collapse
|
34
|
Integrated Full-Length Transcriptome and RNA-Seq to Identify Immune System Genes from the Skin of Sperm Whale ( Physeter macrocephalus). Genes (Basel) 2021; 12:genes12020233. [PMID: 33562637 PMCID: PMC7914425 DOI: 10.3390/genes12020233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/22/2022] Open
Abstract
Cetaceans are a group of secondary aquatic mammals whose ancestors returned to the ocean from land, and during evolution, their immune systems adapted to the aquatic environment. Their skin, as the primary barrier to environmental pathogens, supposedly evolved to adapt to a new living environment. However, the immune system in the skin of cetaceans and the associated molecular mechanisms are still largely unknown. To better understand the immune system, we extracted RNA from the sperm whale's (Physeter macrocephalus) skin and performed PacBio full-length sequencing and RNA-seq sequencing. We obtained a total of 96,350 full-length transcripts with an average length of 1705 bp and detected 5150 genes that were associated with 21 immune-related pathways by gene annotation enrichment analysis. Moreover, we found 89 encoding genes corresponding to 33 proteins were annotated in the NOD-like receptor (NLR)-signaling pathway, including NOD1, NOD2, RIP2, and NF-kB genes, which were discussed in detail and predicted to play essential roles in the immune system of the sperm whale. Furthermore, NOD1 was highly conservative during evolution by the sequence comparison and phylogenetic tree. These results provide new information about the immune system in the skin of cetaceans, as well as the evolution of immune-related genes.
Collapse
|
35
|
Liu F, Shao M, Xu F, Rong F. Inhibition of NOD1 Attenuates Neonatal Hypoxia-Ischemia Induced Long-Term Cognitive Impairments in Mice Through Modulation of Autophagy-Related Proteins. Neuropsychiatr Dis Treat 2021; 17:2659-2669. [PMID: 34421301 PMCID: PMC8373312 DOI: 10.2147/ndt.s314884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/12/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Autophagy is implicated in neonatal hypoxia-ischemia (HI) induced cognitive impairment. The nucleotide-oligomerizing domain-1 (NOD1), a protein involved in inflammatory responses, has been shown to activate autophagy to promote progression of other diseases. We aimed to investigate whether and how NOD1 is involved in HI-induced brain injury using an HI mouse model. METHODS We induced HI in neonatal mice and examined levels of NOD1 and genes associated with autophagy. We then inhibited NOD1 by intracerebroventricular injection of si-NOD1 following HI induction and tested the effects on autophagy, inflammatory responses and long-term behavioral outcomes through Morris water maze and open field tests. RESULTS We found that HI induction significantly elevated mRNA levels of NOD1 (3.54 folds change) and autophagy-related genes including Atg5 (3.89 folds change) and Beclin-1 (3.34 folds change). NOD1 inhibition following HI induction suppressed autophagy signaling as well as HI induced proinflammatory cytokine production. Importantly, NOD1 inhibition after HI improved long-term cognitive function, without impacting exploratory and locomotor activities. CONCLUSION We show here that NOD1 is involved in the pathogenesis of HI-induced brain injury through modulation of autophagy-related proteins and inflammatory responses. Our findings suggest that NOD1 may be a potent target for developing therapeutic strategies for treating HI-induced brain injury.
Collapse
Affiliation(s)
- Fang Liu
- Department of Child Health Care, Zibo Central Hospital, Zibo, 255000, Shandong, People's Republic of China
| | - Mingyu Shao
- Department of Child Health Care, Zibo Central Hospital, Zibo, 255000, Shandong, People's Republic of China
| | - Feng Xu
- Department of Pediatrics, Zibo Central Hospital, Zibo, 255000, Shandong, People's Republic of China
| | - Fang Rong
- The Community Clinic of Overseas Chinese Town, Zibo Central Hospital, North Gate of Zhongrun Overseas Chinese Town, Zibo, 255000, Shandong, People's Republic of China
| |
Collapse
|
36
|
Crump GM, Zhou J, Mashayekh S, Grimes CL. Revisiting peptidoglycan sensing: interactions with host immunity and beyond. Chem Commun (Camb) 2020; 56:13313-13322. [PMID: 33057506 PMCID: PMC7642115 DOI: 10.1039/d0cc02605k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The interaction between host immunity and bacterial cells plays a pivotal role in a variety of human diseases. The bacterial cell wall component peptidoglycan (PG) is known to stimulate an immune response, which makes PG a distinctive recognition element for unveiling these complicated molecular interactions. Pattern recognition receptor (PRR) proteins are among the critical components of this system that initially recognize molecular patterns associated with microorganisms such as bacteria and fungi. These molecular patterns are mostly embedded in the bacterial or fungal cell wall structure and can be released and presented to the immune system in various situations. Nonetheless, detailed knowledge of this recognition is limited due to the diversity among the PG polymer and its fragments; the subsequent responses by multiple hosts add more complexity. Here, we discuss how our understanding of the role and molecular mechanisms of the well-studied PRR, the NOD-like receptors (NLRs), in the human immune system has evolved in recent years. We highlight the instances of other classes of proteins with similar behavior in the recognition of PG that have been identified in other microorganisms such as yeasts. These proteins are particularly interesting because a network of cellular interactions exists between human host cells, bacteria and yeast as a part of the normal human flora. To support our understanding of these interactions, we provide insight into the chemist's toolbox of peptidoglycan probes that aid in the investigations of the behaviors of these proteins and other biological contexts relevant to the sensing and recognition of peptidoglycan. The importance of these interactions in human health for the development of biomarkers and biotherapy is highlighted.
Collapse
Affiliation(s)
- Geneva Maddison Crump
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA.
| | | | | | | |
Collapse
|
37
|
Cawthon CR, Kirkland RA, Pandya S, Brinson NA, de La Serre CB. Non-neuronal crosstalk promotes an inflammatory response in nodose ganglia cultures after exposure to byproducts from gram positive, high-fat-diet-associated gut bacteria. Physiol Behav 2020; 226:113124. [PMID: 32763334 PMCID: PMC7530053 DOI: 10.1016/j.physbeh.2020.113124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
Vagal afferent neurons (VAN) projecting to the lamina propria of the digestive tract are the primary source of gut-originating signals to the central nervous system (CNS). VAN cell bodies are found in the nodose ganglia (NG). Responsiveness of VAN to gut-originating signals is altered by feeding status with sensitivity to satiety signals such as cholecystokinin (CCK) increasing in the fed state. Chronic high-fat (HF) feeding results in inflammation at the level of the NG associated with a loss of VAN ability to switch phenotype from the fasted to the fed state. HF feeding also leads to compositional changes in the gut microbiota. HF diet consumption notably drives increased Firmicutes to Bacteroidetes phyla ratio and increased members of the Actinobacteria phylum. Firmicutes and Actinobacteria are largely gram positive (GP). In this study, we aimed to determine if byproducts from GP bacteria can induce an inflammatory response in cultured NG and to characterize the mechanism and cell types involved in the response. NG were collected from male Wistar rats and cultured for a total of 72 hours. At 48-68 hours after plating, cultures were treated with neuronal culture media in which Serinicoccus chungangensis had been grown and removed (SUP), lipoteichoic acid (LTA), or meso-diaminopimelic acid (meso-DAP). Some treatments included the glial inhibitors minocycline (MINO) and/or fluorocitrate (FC). The responses were evaluated using immunocytochemistry, qPCR, and electrochemiluminescence. We found that SUP induced an inflammatory response characterized by increased interleukin (IL)-6 staining and increased expression of genes for IL-6, interferon (IFN)γ, and tumor necrosis factor (TNF)α along with genes associated with cell-to-cell communication such as C-C motif chemokine ligand-2 (CCL2). Inclusion of inhibitors attenuated some responses but failed to completely normalize all indications of response, highlighting the role of immunocompetent cellular crosstalk in regulating the inflammatory response. LTA and meso-DAP produced responses that shared characteristics with SUP but were not identical. Our results support a role for HF associated GP bacterial byproducts' ability to contribute to vagal inflammation and to engage signaling from nonneuronal cells.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Rebecca A Kirkland
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Shreya Pandya
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Nigel A Brinson
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States
| | - Claire B de La Serre
- Department of Foods and Nutrition, The University of Georgia, Athens, Georgia30602, United States.
| |
Collapse
|
38
|
Golbabapour S, Bagheri-Lankarani K, Ghavami S, Geramizadeh B. Autoimmune Hepatitis and Stellate Cells: An Insight into the Role of Autophagy. Curr Med Chem 2020; 27:6073-6095. [PMID: 30947648 DOI: 10.2174/0929867326666190402120231] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/11/2019] [Accepted: 03/15/2019] [Indexed: 02/08/2023]
Abstract
Autoimmune hepatitis is a necroinflammatory process of liver, featuring interface hepatitis
by T cells, macrophages and plasma cells that invade to periportal parenchyma. In this process, a
variety of cytokines are secreted and liver tissues undergo fibrogenesis, resulting in the apoptosis of
hepatocytes. Autophagy is a complementary mechanism for restraining intracellular pathogens to
which the innate immune system does not provide efficient endocytosis. Hepatocytes with their
particular regenerative features are normally in a quiescent state, and, autophagy controls the accumulation
of excess products, therefore the liver serves as a basic model for the study of autophagy.
Impairment of autophagy in the liver causes the accumulation of damaged organelles, misfolded
proteins and exceeded lipids in hepatocytes as seen in metabolic diseases. In this review, we introduce
autoimmune hepatitis in association with autophagy signaling. We also discuss some genes and
proteins of autophagy, their regulatory roles in the activation of hepatic stellate cells and the importance
of lipophagy and tyrosine kinase in hepatic fibrogenesis. In order to provide a comprehensive
overview of the regulatory role of autophagy in autoimmune hepatitis, the pathway analysis of autophagy
in autoimmune hepatitis is also included in this article.
Collapse
Affiliation(s)
- Shahram Golbabapour
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Birmingham, B15 2WB, United Kingdom
| | - Kamran Bagheri-Lankarani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Ghavami
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bita Geramizadeh
- Department of Pathology, Medical school of Shiraz University, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
39
|
Shrungeswara AH, Unnikrishnan MK. Energy Provisioning and Inflammasome Activation: The Pivotal Role of AMPK in Sterile Inflammation and Associated Metabolic Disorders. Antiinflamm Antiallergy Agents Med Chem 2020; 20:107-117. [PMID: 32938355 DOI: 10.2174/1871523019666200916115034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/02/2020] [Accepted: 08/19/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Body defenses and metabolic processes probably co-evolved in such a way that rapid, energy-intensive acute inflammatory repair is functionally integrated with energy allocation in a starvation/ infection / injury-prone primitive environment. Disruptive metabolic surplus, aggravated by sedentary lifestyle induces chronic under-activation of AMPK, the master regulator of intracellular energy homeostasis. Sudden increase in chronic, dysregulated 'sterile' inflammatory disorders probably results from a shift towards calorie rich, sanitized, cushioned, injury/ infection free environment, repositioning inflammatory repair pathways towards chronic, non-microbial, 'sterile', 'low grade', and 'parainflammation'. AMPK, (at the helm of energy provisioning) supervises the metabolic regulation of inflammasome activation, a common denominator in lifestyle disorders. DISCUSSION In this review, we discuss various pathways linking AMPK under-activation and inflammasome activation. AMPK under-activation, the possible norm in energy-rich sedentary lifestyle, could be the central agency that stimulates inflammasome activation by multiple pathways such as 1: decreasing autophagy, and accumulation of intracellular DAMPs, (particulate crystalline molecules, advanced glycation end-products, oxidized lipids, etc.) 2: stimulating a glycolytic shift (pro-inflammatory) in metabolism, 3: promoting NF-kB activation and decreasing Nrf2 activation, 4: increasing reactive oxygen species (ROS) formation, Unfolded Protein Response (UPR) and Endoplasmic Reticulum (ER) stress. CONCLUSION The 'inverse energy crisis' associated with calorie-rich, sedentary lifestyle, advocates dietary and pharmacological interventions for treating chronic metabolic disorders by overcoming / reversing AMPK under-activation.
Collapse
Affiliation(s)
- Akhila H Shrungeswara
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | | |
Collapse
|
40
|
Ma Y, Yang J, Wei X, Pei Y, Ye J, Li X, Si G, Tian J, Dong Y, Liu G. Nonpeptidic quinazolinone derivatives as dual nucleotide-binding oligomerization domain-like receptor 1/2 antagonists for adjuvant cancer chemotherapy. Eur J Med Chem 2020; 207:112723. [PMID: 32920426 DOI: 10.1016/j.ejmech.2020.112723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 12/17/2022]
Abstract
Nucleotide-binding oligomerization domain-containing protein 1 and 2 (NOD1/2) receptors are potential immune checkpoints. In this article, a quinazolinone derivative (36b) as a NOD1/2 dual antagonist was identified that significantly sensitizes B16 tumor-bearing mice to paclitaxel treatment by inhibiting both nuclear factor κB (NF-κB) and mitogen-activated protein kinase inflammatory signaling that mediated by NOD1/2.
Collapse
Affiliation(s)
- Yao Ma
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 2A Nanwei Rd, Xicheng Dist, Beijing, 100050, PR China
| | - Jingshu Yang
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Xiduan Wei
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Yameng Pei
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Jingjia Ye
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Xueyuan Li
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Guangxu Si
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Jingyuan Tian
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China
| | - Yi Dong
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 2A Nanwei Rd, Xicheng Dist, Beijing, 100050, PR China.
| | - Gang Liu
- School of Pharmaceutical Sciences, Tsinghua University, Haidian Dist, Beijing, 100084, PR China.
| |
Collapse
|
41
|
Root-Bernstein R. Synergistic Activation of Toll-Like and NOD Receptors by Complementary Antigens as Facilitators of Autoimmune Disease: Review, Model and Novel Predictions. Int J Mol Sci 2020; 21:ijms21134645. [PMID: 32629865 PMCID: PMC7369971 DOI: 10.3390/ijms21134645] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/29/2022] Open
Abstract
Persistent activation of toll-like receptors (TLR) and nucleotide-binding oligomerization domain-containing proteins (NOD) in the innate immune system is one necessary driver of autoimmune disease (AD), but its mechanism remains obscure. This study compares and contrasts TLR and NOD activation profiles for four AD (autoimmune myocarditis, myasthenia gravis, multiple sclerosis and rheumatoid arthritis) and their animal models. The failure of current AD theories to explain the disparate TLR/NOD profiles in AD is reviewed and a novel model is presented that explains innate immune support of persistent chronic inflammation in terms of unique combinations of complementary AD-specific antigens stimulating synergistic TLRs and/or NODs. The potential explanatory power of the model is explored through testable, novel predictions concerning TLR- and NOD-related AD animal models and therapies.
Collapse
|
42
|
Wang G, Fu Y, Ma K, Liu J, Liu X. NOD2 regulates microglial inflammation through the TAK1-NF-κB pathway and autophagy activation in murine pneumococcal meningitis. Brain Res Bull 2020; 158:20-30. [PMID: 32109527 DOI: 10.1016/j.brainresbull.2020.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 01/16/2020] [Accepted: 02/11/2020] [Indexed: 10/24/2022]
Abstract
Streptococcus pneumoniae is responsible for pneumococcal meningitis, with significant mortality and morbidity worldwide. Microglial inflammation plays a vital role in meningitis. The peptidoglycan sensor NOD2 (nucleotide-binding oligomerization domain 2) has been identified to promote microglia activation, but the role in autophagy following pneumococcal meningitis remains unclear. In the present study, we investigated the role of NOD2 in microglial inflammation and autophagy, as well as related signaling pathways, during S. pneumonia infection. NOD2 expression was knocked down by the injection of lentivirus-mediated short-hairpin RNA (shRNA). Our results revealed that NOD2 promotes microglial inflammation by increasing inflammatory mediators. We also showed that the TAK1-NF-κB pathway is involved in this process. In addition, NOD2 increased the expression of autophagy-related proteins and induced autophagosome formation. Rapamycin and 3-MA were utilized to assess the role of autophagy in microglial inflammation induced by S. pneumonia. We demonstrated that autophagy serves as a cellular defense mechanism to reduce inflammatory mediators. Similar to the in vitro results, NOD2 induced inflammation and autophagy in the brain in a mouse meningitis model. Moreover, NOD2 silencing significantly reduced brain edema and improved the neurological function of pneumococcal meningitis mice. Taken together, these data demonstrate that NOD2 promotes microglial inflammation and autophagy in murine pneumococcal meningitis, and the TAK1-NF-κB pathway is involved in microglial activation.
Collapse
Affiliation(s)
- Guan Wang
- Department of Pediatrics, Qilu Hospital, Shandong University, 107#, Wen Hua Xi Road, Jinan, Shandong, 250012, PR China
| | - Yanan Fu
- Qilu Hospital, Shandong University, 107#, Wen Hua Xi Road, Jinan, Shandong, 250012, PR China
| | - Kun Ma
- Department of Pediatrics, Shandong Provincial Qianfoshan Hospital, The Frist Hospital Affiliated with Shandong First Medical University, 16766#, Jing Shi Road, Jinan, Shandong, 250014, PR China
| | - Junli Liu
- Department of Pediatrics, Taian Central Hospital, 29#, Long Tan Road, Taian, Shandong, 271000, PR China
| | - Xinjie Liu
- Department of Pediatrics, Qilu Hospital, Shandong University, 107#, Wen Hua Xi Road, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
43
|
Guzelj S, Gobec M, Urbančič D, Mlinarič-Raščan I, Corsini E, Jakopin Ž. Structural features and functional activities of benzimidazoles as NOD2 antagonists. Eur J Med Chem 2020; 190:112089. [PMID: 32014680 DOI: 10.1016/j.ejmech.2020.112089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
Abstract
NOD1 and NOD2 are pattern recognition receptors that have important roles in innate immune responses. Although their overactivation has been linked to a number of diseases, NOD2 in particular remains a virtually unexploited target in this respect, with only one structural class of antagonist reported. To gain insight into the structure-activity relationships of NOD2 antagonists, a series of novel analogs was designed and synthesized, and then screened for antagonist activity versus NOD2, and counter-screened versus NOD1. Compounds 32 and 38 were identified as potent and moderately selective NOD2 antagonists, and 33 and 42 as dual NOD1/NOD2 antagonists, with balanced activities against both targets in the low micromolar range. These data enable in-depth exploration of their structure-activity relationships and provide deeper understanding of the structural features required for NOD2 antagonism.
Collapse
Affiliation(s)
- Samo Guzelj
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Martina Gobec
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Dunja Urbančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Irena Mlinarič-Raščan
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Environmental Science and Policy, University of Milan, Via Balzaretti 9, 20133, Milan, Italy
| | - Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
44
|
Jiang HY, Najmeh S, Martel G, MacFadden-Murphy E, Farias R, Savage P, Leone A, Roussel L, Cools-Lartigue J, Gowing S, Berube J, Giannias B, Bourdeau F, Chan CHF, Spicer JD, McClure R, Park M, Rousseau S, Ferri LE. Activation of the pattern recognition receptor NOD1 augments colon cancer metastasis. Protein Cell 2020; 11:187-201. [PMID: 31956962 PMCID: PMC7026222 DOI: 10.1007/s13238-019-00687-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/13/2019] [Indexed: 01/02/2023] Open
Abstract
While emerging data suggest nucleotide oligomerization domain receptor 1 (NOD1), a cytoplasmic pattern recognition receptor, may play an important and complementary role in the immune response to bacterial infection, its role in cancer metastasis is entirely unknown. Hence, we sought to determine the effects of NOD1 on metastasis. NOD1 expression in paired human primary colon cancer, human and murine colon cancer cells were determined using immunohistochemistry and immunoblotting (WB). Clinical significance of NOD1 was assessed using TCGA survival data. A series of in vitro and in vivo functional assays, including adhesion, migration, and metastasis, was conducted to assess the effect of NOD1. C12-iE-DAP, a highly selective NOD1 ligand derived from gram-negative bacteria, was used to activate NOD1. ML130, a specific NOD1 inhibitor, was used to block C12-iE-DAP stimulation. Stable knockdown (KD) of NOD1 in human colon cancer cells (HT29) was constructed with shRNA lentiviral transduction and the functional assays were thus repeated. Lastly, the predominant signaling pathway of NOD1-activation was identified using WB and functional assays in the presence of specific kinase inhibitors. Our data demonstrate that NOD1 is highly expressed in human colorectal cancer (CRC) and human and murine CRC cell lines. Clinically, we demonstrate that this increased NOD1 expression negatively impacts survival in patients with CRC. Subsequently, we identify NOD1 activation by C12-iE-DAP augments CRC cell adhesion, migration and metastasis. These effects are predominantly mediated via the p38 mitogen activated protein kinase (MAPK) pathway. This is the first study implicating NOD1 in cancer metastasis, and thus identifying this receptor as a putative therapeutic target.
Collapse
Affiliation(s)
- Henry Y Jiang
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Sara Najmeh
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Guy Martel
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Elyse MacFadden-Murphy
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Raquel Farias
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Paul Savage
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, QC, H3A 1A3, Canada
| | - Arielle Leone
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Lucie Roussel
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Jonathan Cools-Lartigue
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Stephen Gowing
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Julie Berube
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Betty Giannias
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada
| | - France Bourdeau
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada
| | - Carlos H F Chan
- Department of Surgery, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Jonathan D Spicer
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Rebecca McClure
- Department of Pathology, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, Canada
| | - Morag Park
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue, Montreal, QC, H3A 1A3, Canada
| | - Simon Rousseau
- Meakins-Christie Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, QC, H4A 3J1, Canada
| | - Lorenzo E Ferri
- Thoracic and Upper GI Cancer Research Laboratories, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Block E, Lab #E02-4134, Montreal, QC, H4A 3J1, Canada. .,Department of Experimental Surgery and Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.
| |
Collapse
|
45
|
Ahn MY, Kang JK, Kwon SM, Yoon HE, Yoon JH. Expression of nucleotide-binding oligomerization domain 1 and 2 in oral lichen planus. J Dent Sci 2020; 15:1-8. [PMID: 32256993 PMCID: PMC7109494 DOI: 10.1016/j.jds.2019.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 11/28/2019] [Indexed: 11/16/2022] Open
Abstract
Background/purpose Oral lichen planus (OLP) is a chronic inflammatory disease of oral mucosa. The present study investigated the expression of nucleotide-binding oligomerization domain (NOD), a pivotal sensor protein of the innate immune system, in OLP. Materials and methods Oral mucosal biopsies were collected from 20 patients with OLP and 6 individuals with normal oral mucosa (NOM). The expression of NOD1 and NOD2 was determined using RT-PCR and immunohistochemistry in OLP and NOM samples. Results The mRNA expression of NOD1 and NOD2 was significantly higher in the OLP group than in the NOM group. The protein expression of NOD1 was marginally upregulated in all mucosal layers in the OLP group compared with that of the NOM group; however, the differences were not significant. The expression of NOD2 was elevated in infiltrating lymphocytes of the submucosal layer in the OLP group compared with the NOM group, but was undetected in other inflammatory disease, inflammatory fibrous hyperplasia (IFH). This study revealed the upregulation of NOD2 mRNA and protein in the OLP group, but not in the NOM group. Conclusion These findings suggest that NOD2 may play an important role in the pathogenesis of OLP and represents a new diagnostic and treatment target.
Collapse
Affiliation(s)
- Mee-Young Ahn
- Major in Pharmaceutical Engineering, Division of Bio-industry, College of Medical and Life Sciences, Silla University, Busan, Republic of Korea
| | - Jin-Kyu Kang
- Department of Oral Medicine and Orofacial Pain, College of Dentistry, Daejeon Dental Hospital, Wonkwang University, Daejeon, Republic of Korea
| | - Seong-Min Kwon
- Department of Oral and Maxillofacial Pathology, College of Dentistry, Wonkwang Bone Regeneration Research Institute, Daejeon Dental Hospital, Wonkwang University, Daejeon, Republic of Korea
| | - Hyo-Eun Yoon
- Department of Oral and Maxillofacial Pathology, College of Dentistry, Wonkwang Bone Regeneration Research Institute, Daejeon Dental Hospital, Wonkwang University, Daejeon, Republic of Korea
| | - Jung-Hoon Yoon
- Department of Oral and Maxillofacial Pathology, College of Dentistry, Wonkwang Bone Regeneration Research Institute, Daejeon Dental Hospital, Wonkwang University, Daejeon, Republic of Korea
| |
Collapse
|
46
|
Gharagozloo M, Mahmoud S, Simard C, Yamamoto K, Bobbala D, Ilangumaran S, Smith MD, Lamontagne A, Jarjoura S, Denault JB, Blais V, Gendron L, Vilariño-Güell C, Sadovnick AD, Ting JP, Calabresi PA, Amrani A, Gris D. NLRX1 inhibits the early stages of CNS inflammation and prevents the onset of spontaneous autoimmunity. PLoS Biol 2019; 17:e3000451. [PMID: 31525189 PMCID: PMC6762215 DOI: 10.1371/journal.pbio.3000451] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 09/26/2019] [Accepted: 08/28/2019] [Indexed: 12/20/2022] Open
Abstract
Nucleotide-binding, leucine-rich repeat containing X1 (NLRX1) is a mitochondria-located innate immune sensor that inhibits major pro-inflammatory pathways such as type I interferon and nuclear factor-κB signaling. We generated a novel, spontaneous, and rapidly progressing mouse model of multiple sclerosis (MS) by crossing myelin-specific T-cell receptor (TCR) transgenic mice with Nlrx1−/− mice. About half of the resulting progeny developed spontaneous experimental autoimmune encephalomyelitis (spEAE), which was associated with severe demyelination and inflammation in the central nervous system (CNS). Using lymphocyte-deficient mice and a series of adoptive transfer experiments, we demonstrate that genetic susceptibility to EAE lies within the innate immune compartment. We show that NLRX1 inhibits the subclinical stages of microglial activation and prevents the generation of neurotoxic astrocytes that induce neuronal and oligodendrocyte death in vitro. Moreover, we discovered several mutations within NLRX1 that run in MS-affected families. In summary, our findings highlight the importance of NLRX1 in controlling the early stages of CNS inflammation and preventing the onset of spontaneous autoimmunity. NLRX1 is a guardian protein that inhibits the inflammatory response of glial cells within the central nervous system and prevents the onset of a spontaneous multiple sclerosis–like disease in mice. This study uses a novel mouse model to provide mechanistic insights into the neurodegenerative origin of multiple sclerosis.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shaimaa Mahmoud
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Camille Simard
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Kenzo Yamamoto
- Department of Chemical Engineering and Biotechnological Engineering, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Diwakar Bobbala
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Subburaj Ilangumaran
- Department of Anatomy and Cell Biology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Albert Lamontagne
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Samir Jarjoura
- Department of Neurology, Faculty of Medicine, MS Clinic, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jean-Bernard Denault
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Véronique Blais
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Louis Gendron
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | - A. Dessa Sadovnick
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Jenny P. Ting
- Department of Microbiology and Immunology, Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Abdelaziz Amrani
- Department of Pediatrics, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Denis Gris
- Department of Pharmacology and Physiology, Faculty of Medicine, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- * E-mail:
| |
Collapse
|
47
|
Heat Shock Proteins and Inflammasomes. Int J Mol Sci 2019; 20:ijms20184508. [PMID: 31547225 PMCID: PMC6771073 DOI: 10.3390/ijms20184508] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 01/23/2023] Open
Abstract
Heat shock proteins (HSP) regulate inflammation in many physiological contexts. However, inflammation is a broad process, involving numerous cytokines produced by different molecular pathways with multiple functions. In this review, we focused on the particular role of HSP on the inflammasomes intracellular platforms activated by danger signals and that enable activation of inflammatory caspases, mainly caspase-1, leading to the production of the pro-inflammatory cytokine IL-1β. Interestingly, some members of the HSP family favor inflammasomes activation whereas others inhibit it, suggesting that HSP modulators for therapeutic purposes, must be carefully chosen.
Collapse
|
48
|
Jakopin Ž, Corsini E. THP-1 Cells and Pro-inflammatory Cytokine Production: An in Vitro Tool for Functional Characterization of NOD1/NOD2 Antagonists. Int J Mol Sci 2019; 20:ijms20174265. [PMID: 31480368 PMCID: PMC6747088 DOI: 10.3390/ijms20174265] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/26/2019] [Accepted: 08/28/2019] [Indexed: 02/06/2023] Open
Abstract
THP-1 cells express high levels of native functional nucleotide-binding oligomerization domain 1 (NOD1), NOD2, and Toll-like receptor 4 (TLR4) receptors, and have often been used for investigating the immunomodulatory effects of small molecules. We postulated that they would represent an ideal cell-based model for our study, the aim of which was to develop a new in vitro tool for functional characterization of NOD antagonists. NOD antagonists were initially screened for their effect on NOD agonist-induced interleukin-8 (IL-8) release. Next, we examined the extent to which the selected NOD antagonists block the NOD-TLR4 synergistic crosstalk by measuring the effect of NOD antagonism on tumor necrosis factor-α (TNF-α) secretion from doubly activated THP-1 cells. Overall, the results obtained indicate that pro-inflammatory cytokine secretion from THP-1 provides a valuable, simple and reproducible in vitro tool for functional characterization of NOD antagonists.
Collapse
Affiliation(s)
- Žiga Jakopin
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Emanuela Corsini
- Laboratory of Toxicology, Department of Pharmacological Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milan, Italy
| |
Collapse
|
49
|
Takano M, Takeuchi T, Kuriyama S, Yumoto R. Role of peptide transporter 2 and MAPK signaling pathways in the innate immune response induced by bacterial peptides in alveolar epithelial cells. Life Sci 2019; 229:173-179. [DOI: 10.1016/j.lfs.2019.05.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023]
|
50
|
Rivers SL, Klip A, Giacca A. NOD1: An Interface Between Innate Immunity and Insulin Resistance. Endocrinology 2019; 160:1021-1030. [PMID: 30807635 PMCID: PMC6477778 DOI: 10.1210/en.2018-01061] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/19/2019] [Indexed: 12/17/2022]
Abstract
Insulin resistance is driven, in part, by activation of the innate immune system. We have discussed the evidence linking nucleotide-binding oligomerization domain (NOD)1, an intracellular pattern recognition receptor, to the onset and progression of obesity-induced insulin resistance. On a molecular level, crosstalk between downstream NOD1 effectors and the insulin receptor pathway inhibits insulin signaling, potentially through reduced insulin receptor substrate action. In vivo studies have demonstrated that NOD1 activation induces peripheral, hepatic, and whole-body insulin resistance. Also, NOD1-deficient models are protected from high-fat diet (HFD)-induced insulin resistance. Moreover, hematopoietic NOD1 deficiency prevented HFD-induced changes in proinflammatory macrophage polarization status, thus protecting against the development of metabolic inflammation and insulin resistance. Serum from HFD-fed mice activated NOD1 signaling ex vivo; however, the molecular identity of the activating factors remains unclear. Many have proposed that an HFD changes the gut permeability, resulting in increased translocation of bacterial fragments and increased circulating NOD1 ligands. In contrast, others have suggested that NOD1 ligands are endogenous and potentially lipid-derived metabolites produced during states of nutrient overload. Nevertheless, that NOD1 contributes to the development of insulin resistance, and that NOD1-based therapy might provide benefit, is an exciting advancement in metabolic research.
Collapse
Affiliation(s)
- Sydney L Rivers
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Amira Klip
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Correspondence: Adria Giacca, MD, Department of Physiology, Faculty of Medicine, University of Toronto, Medical Sciences Building, 1 King’s College Circle, No. 3336, Toronto, Ontario M5S 1A8, Canada. E-mail:
| |
Collapse
|