1
|
Xu J, Li Y, Yang X, Li H, Xiao X, You J, Li H, Zheng L, Yi C, Li Z, Huang Y. Quercetin inhibited LPS-induced cytokine storm by interacting with the AKT1-FoxO1 and Keap1-Nrf2 signaling pathway in macrophages. Sci Rep 2024; 14:20913. [PMID: 39245773 PMCID: PMC11381534 DOI: 10.1038/s41598-024-71569-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: 05/10/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
Cytokine storm (CS) emerges as an exacerbated inflammatory response triggered by various factors such as pathogens and excessive immunotherapy, posing a significant threat to life if left unchecked. Quercetin, a monomer found in traditional Chinese medicine, exhibits notable anti-inflammatory and antiviral properties. This study endeavors to explore whether quercetin intervention could mitigate CS through a combination of network pharmacology analysis and experimental validation. First, common target genes and potential mechanisms affected by quercetin and CS were identified through network pharmacology, and molecular docking experiments confirmed quercetin and core targets. Subsequently, in vitro experiments of Raw264.7 cells stimulated by lipopolysaccharide (LPS) showed that quercetin could effectively inhibit the overexpression of pro-inflammatory mediators and regulate the AKT1-FoxO1 signaling pathway. At the same time, quercetin can reduce ROS through the Keap1-Nrf2 signaling pathway. In addition, in vivo studies of C57BL/6 mice injected with LPS further confirmed quercetin's inhibitory effect on CS. In conclusion, this investigation elucidated novel target genes and signaling pathways implicated in the therapeutic effects of quercetin on CS. Moreover, it provided compelling evidence supporting the efficacy of quercetin in reversing LPS-induced CS, primarily through the regulation of the AKT1-FoxO1 and Keap1-Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Jingyi Xu
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Yue Li
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Xi Yang
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Hong Li
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Xi Xiao
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Jia You
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Huawei Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Medicine, Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Lingnan Zheng
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Cheng Yi
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China.
| | - Zhaojun Li
- Department of Radiation Oncology, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), No.31, Longhua Road, Haikou, 570100, China.
| | - Ying Huang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China.
| |
Collapse
|
2
|
Xu F, Zheng H, Dong X, Zhou A, Emu Q. miRNA expression signatures induced by pasteurella multocida infection in goats lung. Sci Rep 2024; 14:19626. [PMID: 39179681 PMCID: PMC11343864 DOI: 10.1038/s41598-024-69654-3] [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: 01/04/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024] Open
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression and are involved in bacterial pathogenesis and host-pathogen interactions. In this study, we investigated the function of miRNAs in the regulation of host responses to Pasteurella multocida infection. Using next-generation sequencing, we analyzed miRNA expression pattern and identified differentially expressed miRNAs in Pasteurella multocida-infected goat lungs. In addition, we investigated the function of differentially expressed miRNAs andtheir targeted signaling pathways in bacterial infection processes. The results showed that Pasteurella multocida infection led to 69 significantly differentially expressed miRNAs, including 28 known annotated miRNAs with miR-497-3p showing the most significant difference. Gene target prediction and functional enrichment analyses showed that the target genes were mainly involved in cell proliferation, regulation of the cellular metabolic process, positive regulation of cellular process, cellular senescence, PI3K-Akt signaling pathway, FoxO signaling pathway and infection-related pathways. In conclusion, these data provide a new perspective on the roles of miRNAs in Pasteurella multocida infection.
Collapse
Affiliation(s)
- Feng Xu
- Animal Genetics and Breeding Key Laboratory of Sichuan Province, Animal Science Academy of Sichuan Province, Chengdu, China
| | - Hao Zheng
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan, China
| | - Xia Dong
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan, China
| | - Ao Zhou
- Laboratory of Genetic Breeding, Reproduction and Precision Livestock Farming, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Hubei Provincial Center of Technology Innovation for Domestic Animal Breeding, Wuhan, China.
| | - Quzhe Emu
- Animal Genetics and Breeding Key Laboratory of Sichuan Province, Animal Science Academy of Sichuan Province, Chengdu, China.
| |
Collapse
|
3
|
Zheng L, Cui X, Jiang Z, Li H, Zhu Z, Dai X, Liu X, Zhang L, Huang X, Ren Q. Differential expression of sNPF in male and female eyestalk leading to sex dimorphism of AMP expression in Procambarus clarkii intestine. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109735. [PMID: 38945414 DOI: 10.1016/j.fsi.2024.109735] [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: 02/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Antimicrobial peptide (AMP) is an important component of crustaceans' innate immune system. In this study, a short neuropeptide F (sNPF) gene (Pc-sNPF) and a Forkhead box O (FOXO) gene (PcFOXO) from Procambarus clarkii were identified. Analysis findings showed that the expression level of AMP genes differed between male and female P. clarkii. Furthermore, Pc-sNPF and PcFOXO were related to the sex dimorphism of AMP. Knockdown of Pc-sNPF in the eyestalk significantly upregulated the expression of PcFOXO and two anti-lipopolysaccharide factors (PcALF4 and PcALFL) in the intestine of P. clarkii. The expression of PcFOXO in the intestine of female P. clarkii was higher than in that of males. Results from RNA interference revealed that PcFOXO positively regulated the expression of PcALF4 and PcALFL in the intestine of male and female P. clarkii. In summary, our study showed that differences in Pc-sNPF expression in eyestalk of male and female P. clarkii leading to sex dimorphism of AMP expression in the intestine are mediated by the sNPF-FOXO-AMP signal pathway called the eyestalk-intestine axis.
Collapse
Affiliation(s)
- Liangmin Zheng
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Xinyi Cui
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Zilin Jiang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Hao Li
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Ziyue Zhu
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Xiaoling Dai
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Xiaohan Liu
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Lihua Zhang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China
| | - Xin Huang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing 210023, Jiangsu Province, China.
| | - Qian Ren
- School of Marine Sciences, Nanjing University of Information Science and Technology, Nanjing 210044, Jiangsu Province, China.
| |
Collapse
|
4
|
Hajdú G, Szathmári C, Sőti C. Modeling Host-Pathogen Interactions in C. elegans: Lessons Learned from Pseudomonas aeruginosa Infection. Int J Mol Sci 2024; 25:7034. [PMID: 39000143 PMCID: PMC11241598 DOI: 10.3390/ijms25137034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Infections, such as that by the multiresistant opportunistic bacterial pathogen Pseudomonas aeruginosa, may pose a serious health risk, especially on vulnerable patient populations. The nematode Caenorhabditis elegans provides a simple organismal model to investigate both pathogenic mechanisms and the emerging role of innate immunity in host protection. Here, we review the virulence and infection strategies of P. aeruginosa and host defenses of C. elegans. We summarize the recognition mechanisms of patterns of pathogenesis, including novel pathogen-associated molecular patterns and surveillance immunity of translation, mitochondria, and lysosome-related organelles. We also review the regulation of antimicrobial and behavioral defenses by the worm's neuroendocrine system. We focus on how discoveries in this rich field align with well-characterized evolutionary conserved protective pathways, as well as on potential crossovers to human pathogenesis and innate immune responses.
Collapse
Affiliation(s)
- Gábor Hajdú
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csenge Szathmári
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| | - Csaba Sőti
- Department of Molecular Biology, Semmelweis University, 1094 Budapest, Hungary
| |
Collapse
|
5
|
Kole A, Bag AK, Pal AJ, De D. Generic model to unravel the deeper insights of viral infections: an empirical application of evolutionary graph coloring in computational network biology. BMC Bioinformatics 2024; 25:74. [PMID: 38365632 PMCID: PMC10874019 DOI: 10.1186/s12859-024-05690-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
PURPOSE Graph coloring approach has emerged as a valuable problem-solving tool for both theoretical and practical aspects across various scientific disciplines, including biology. In this study, we demonstrate the graph coloring's effectiveness in computational network biology, more precisely in analyzing protein-protein interaction (PPI) networks to gain insights about the viral infections and its consequences on human health. Accordingly, we propose a generic model that can highlight important hub proteins of virus-associated disease manifestations, changes in disease-associated biological pathways, potential drug targets and respective drugs. We test our model on SARS-CoV-2 infection, a highly transmissible virus responsible for the COVID-19 pandemic. The pandemic took significant human lives, causing severe respiratory illnesses and exhibiting various symptoms ranging from fever and cough to gastrointestinal, cardiac, renal, neurological, and other manifestations. METHODS To investigate the underlying mechanisms of SARS-CoV-2 infection-induced dysregulation of human pathobiology, we construct a two-level PPI network and employed a differential evolution-based graph coloring (DEGCP) algorithm to identify critical hub proteins that might serve as potential targets for resolving the associated issues. Initially, we concentrate on the direct human interactors of SARS-CoV-2 proteins to construct the first-level PPI network and subsequently applied the DEGCP algorithm to identify essential hub proteins within this network. We then build a second-level PPI network by incorporating the next-level human interactors of the first-level hub proteins and use the DEGCP algorithm to predict the second level of hub proteins. RESULTS We first identify the potential crucial hub proteins associated with SARS-CoV-2 infection at different levels. Through comprehensive analysis, we then investigate the cellular localization, interactions with other viral families, involvement in biological pathways and processes, functional attributes, gene regulation capabilities as transcription factors, and their associations with disease-associated symptoms of these identified hub proteins. Our findings highlight the significance of these hub proteins and their intricate connections with disease pathophysiology. Furthermore, we predict potential drug targets among the hub proteins and identify specific drugs that hold promise in preventing or treating SARS-CoV-2 infection and its consequences. CONCLUSION Our generic model demonstrates the effectiveness of DEGCP algorithm in analyzing biological PPI networks, provides valuable insights into disease biology, and offers a basis for developing novel therapeutic strategies for other viral infections that may cause future pandemic.
Collapse
Affiliation(s)
- Arnab Kole
- Department of Computer Application, The Heritage Academy, Kolkata, W.B., 700107, India.
| | - Arup Kumar Bag
- Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | | | - Debashis De
- Department of Computer Science and Engineering, Maulana Abul Kalam Azad University of Technology, Nadia, W.B., 741249, India
| |
Collapse
|
6
|
Dai X, Quan D, Wang L, Cui D, Wan X, Ren Q. FOXO is involved in antimicrobial peptides expression during WSSV infection in Exopalaemon carinicauda. FISH & SHELLFISH IMMUNOLOGY 2024; 144:109286. [PMID: 38097095 DOI: 10.1016/j.fsi.2023.109286] [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: 09/26/2023] [Revised: 11/24/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023]
Abstract
The forkhead box transcription factor O family protein (FOXO) acts as a transcription factor that regulates biological processes regarding DNA repair, immunity, cell cycle regulation, and other biological processes. In this study, EcFOXO was identified from the ridgetail white prawn, Exopalaemon carinicauda. EcFOXO protein contains multiple low-complexity regions and a forkhead (FH) domain. Phylogenetic tree showed that EcFOXO is clustered with crustacean FOXOs. The amino acid sequences of its FH domain are highly similar to the FH domain of FOXOs from other crustaceans. The expression of EcFOXO is altered after white spot syndrome virus (WSSV) stimulation in hepatopancreas and gills. The relationship between EcFOXO and EcRelish was explored by RNA interference (RNAi). Results showed that EcFOXO and EcRelish could positively regulate each other's expression. The expression levels of various antimicrobial peptides (AMPs) significantly reduced after interfering with EcFOXO or EcRelish. These results suggest a positive regulatory loop between EcFOXO and EcRelish, which participates in the innate immunity of ridgetail white prawn by regulating the expression of AMPs during WSSV infection. This study enriches the knowledge about the regulatory mechanism of FOXO in the innate immunity of crustaceans.
Collapse
Affiliation(s)
- Xiaoling Dai
- School of Marine Sciences, Nanjing University of Information Science & Technology, Nanjing, Jiangsu Province, 210044, China; Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Derun Quan
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Libao Wang
- Institute of Oceanology & Marine Fisheries, Nantong, Jiangsu, 226007, China
| | - Di Cui
- College of Agricultural and Biological Engineering, Heze University, Heze, Shandong Province, 274015, China.
| | - Xihe Wan
- Institute of Oceanology & Marine Fisheries, Nantong, Jiangsu, 226007, China.
| | - Qian Ren
- School of Marine Sciences, Nanjing University of Information Science & Technology, Nanjing, Jiangsu Province, 210044, China.
| |
Collapse
|
7
|
Kim S, Lee SK, Son A, Lee J, Kim HG. A Comparative Inflammation-on-a-Chip with a Complete 3D Interface: Pharmacological Applications in COPD-Induced Neutrophil Migration. Adv Healthc Mater 2023; 12:e2301673. [PMID: 37505448 PMCID: PMC11469264 DOI: 10.1002/adhm.202301673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/06/2023] [Indexed: 07/29/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a slow-progressing inflammatory lung disease that is associated with high mortality and disability. There is a lack of appropriate preclinical models of COPD, which hampers drug discovery efforts. Herein, a comparative inflammation-on-a-chip (IoC) is developed with a complete 3D interface without the formation of any micropillar and phaseguide structures that replicated chemoattractant-induced neutrophil transendothelial migration (NTEM), a key feature of COPD. The IoC model is used to evaluate the pharmacological effects of CXCR2 inhibitors (MK-7123, AZD5069, and SB225002) on the migration of neutrophil-like cells in the presence of plasma samples from patients with COPD. This is the first study to evaluate inhibitors of CXCR2-dependent NTEM in a comparative IoC model that mimics the physiological 3D microenvironment, consisting of an endothelial barrier, extracellular compartment, and inflammatory conditions. This IoC model will be useful to investigate COPD severity using patient samples, and will aid basic and translational research involving NTEM.
Collapse
Affiliation(s)
- Soohyun Kim
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Sung Kyun Lee
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Ahryeong Son
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Jong‐Hwan Lee
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| | - Hong Gi Kim
- Center for Infectious Disease Vaccine and Diagnosis InnovationKorea Research Institute of Chemical TechnologyDaejeon34114Republic of Korea
| |
Collapse
|
8
|
Ruperti F, Becher I, Stokkermans A, Wang L, Marschlich N, Potel C, Maus E, Stein F, Drotleff B, Schippers K, Nickel M, Prevedel R, Musser JM, Savitski MM, Arendt D. Molecular profiling of sponge deflation reveals an ancient relaxant-inflammatory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551666. [PMID: 37577507 PMCID: PMC10418225 DOI: 10.1101/2023.08.02.551666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
A hallmark of animals is the coordination of whole-body movement. Neurons and muscles are central to this, yet coordinated movements also exist in sponges that lack these cell types. Sponges are sessile animals with a complex canal system for filter-feeding. They undergo whole-body movements resembling "contractions" that lead to canal closure and water expulsion. Here, we combine 3D optical coherence microscopy, pharmacology, and functional proteomics to elucidate anatomy, molecular physiology, and control of these movements. We find them driven by the relaxation of actomyosin stress fibers in epithelial canal cells, which leads to whole-body deflation via collapse of the incurrent and expansion of the excurrent system, controlled by an Akt/NO/PKG/A pathway. A concomitant increase in reactive oxygen species and secretion of proteinases and cytokines indicate an inflammation-like state reminiscent of vascular endothelial cells experiencing oscillatory shear stress. This suggests an ancient relaxant-inflammatory response of perturbed fluid-carrying systems in animals.
Collapse
Affiliation(s)
- Fabian Ruperti
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Collaboration for joint Ph.D. degree between EMBL and Heidelberg University, Faculty of Biosciences 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | | | - Ling Wang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Nick Marschlich
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| | - Clement Potel
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Emanuel Maus
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Bernhard Drotleff
- Metabolomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Klaske Schippers
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Michael Nickel
- Bionic Consulting Dr. Michael Nickel, 71686 Remseck am Neckar, Germany
| | - Robert Prevedel
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jacob M Musser
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Mikhail M Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Detlev Arendt
- Developmental Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Centre for Organismal Studies (COS), University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
9
|
Lyu B, Li J, Niemeyer B, Anderson DM, Beerntsen B, Song Q. Integrative analysis highlights molecular and immune responses of tick Amblyomma americanum to Escherichia coli challenge. Front Cell Infect Microbiol 2023; 13:1236785. [PMID: 37583446 PMCID: PMC10424933 DOI: 10.3389/fcimb.2023.1236785] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 07/10/2023] [Indexed: 08/17/2023] Open
Abstract
Ticks are ectoparasites that can transmit various pathogens capable of causing life-threatening illnesses in people and animals, making them a severe public health threat. Understanding how ticks respond to bacterial infection is crucial for deciphering their immune defense mechanisms and identifying potential targets for controlling tick-borne diseases. In this study, an in-depth transcriptome analysis was used to investigate the molecular and immune responses of Amblyomma americanum to infection caused by the microinjection of Escherichia coli. With an abundance of differentially expressed genes discovered at different times, the analysis demonstrated significant changes in gene expression profiles in response to E. coli challenge. Notably, we found alterations in crucial immune markers, including the antimicrobial peptides defensin and microplusin, suggesting they may play an essential role in the innate immune response. Furthermore, KEGG analysis showed that following E. coli exposure, a number of key enzymes, including lysosomal alpha-glucosidase, fibroblast growth factor, legumain, apoptotic protease-activating factor, etc., were altered, impacting the activity of the lysosome, mitogen-activated protein kinase, antigen processing and presentation, bacterial invasion, apoptosis, and the Toll and immune deficiency pathways. In addition to the transcriptome analysis, we constructed protein interaction networks to elucidate the molecular interactions underlying the tick's response to E. coli challenge. Hub genes were identified, and their functional enrichment provided insights into the regulation of cytoskeleton rearrangement, apoptotic processes, and kinase activity that may occur in infected cells. Collectively, the findings shed light on the potential immune responses in A. americanum that control E. coli infection.
Collapse
Affiliation(s)
- Bo Lyu
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Jingjing Li
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Brigid Niemeyer
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Deborah M. Anderson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Brenda Beerntsen
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
10
|
Altered Expression of Antimicrobial Peptides in the Upper Gastrointestinal Tract of Patients with Diabetes Mellitus. Nutrients 2023; 15:nu15030754. [PMID: 36771460 PMCID: PMC9919831 DOI: 10.3390/nu15030754] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Antimicrobial peptides (AMP) are essential components of innate immunity with a broad range of antimicrobial activities against bacteria, viruses, and fungi. The aim of this study was to investigate AMP expression in the upper gastrointestinal tract in normal and pathological metabolic states in humans. Furthermore, we examined the correlation between vitamin D levels and AMP expression in the same cohort. Serum concentrations of 25-hydroxyvitamin D3 were measured, and mRNA expression of β-defensins HBD-1, -2, -3, -4, α-defensins HD-5 and -6 and cathelicidin in the upper gastrointestinal tract epithelia were determined by quantitative RT-PCR in 31 individuals (10 with type 2 diabetes, 10 with insulin resistance, and 11 healthy controls). The majority of the cohort showed low vitamin D concentrations, which were negatively correlated with mRNA expression levels of HBD-3 in corpus mucosa. HBD-1 and HBD-3 mRNA were expressed in corpus mucosa, with the former significantly decreased in patients with diabetes. Hence, we conclude that type 2 diabetes is associated with reduced AMP expression in the upper gastrointestinal tract, which might contribute towards epithelial barrier dysfunction and increased bacterial translocation in these patients.
Collapse
|
11
|
Eckhardt CM, Gambazza S, Bloomquist TR, De Hoff P, Vuppala A, Vokonas PS, Litonjua AA, Sparrow D, Parvez F, Laurent LC, Schwartz J, Baccarelli AA, Wu H. Extracellular Vesicle-Encapsulated microRNAs as Novel Biomarkers of Lung Health. Am J Respir Crit Care Med 2023; 207:50-59. [PMID: 35943330 PMCID: PMC9952856 DOI: 10.1164/rccm.202109-2208oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 08/08/2022] [Indexed: 02/03/2023] Open
Abstract
Rationale: Early detection of respiratory diseases is critical to facilitate delivery of disease-modifying interventions. Extracellular vesicle-enriched microRNAs (EV-miRNAs) may represent reliable markers of early lung injury. Objectives: Evaluate associations of plasma EV-miRNAs with lung function. Methods: The prospective NAS (Normative Aging Study) collected plasma EV-miRNA measurements from 1996-2015 and spirometry every 3-5 years through 2019. Associations of EV-miRNAs with baseline lung function were modeled using linear regression. To complement the individual miRNA approach, unsupervised machine learning was used to identify clusters of participants with distinct EV-miRNA profiles. Associations of EV-miRNA profiles with multivariate latent longitudinal lung function trajectories were modeled using log binomial regression. Biological functions of significant EV-miRNAs were explored using pathway analyses. Results were replicated in an independent sample of NAS participants and in the HEALS (Health Effects of Arsenic Longitudinal Study). Measurements and Main Results: In the main cohort of 656 participants, 51 plasma EV-miRNAs were associated with baseline lung function (false discovery rate-adjusted P value < 0.05), 28 of which were replicated in the independent NAS sample and/or in the HEALS cohort. A subset of participants with distinct EV-miRNA expression patterns had increased risk of declining lung function over time, which was replicated in the independent NAS sample. Significant EV-miRNAs were shown in pathway analyses to target biological pathways that regulate respiratory cellular immunity, the lung inflammatory response, and airway structural integrity. Conclusions: Plasma EV-miRNAs may represent a robust biomarker of subclinical lung injury and may facilitate early identification and treatment of patients at risk of developing overt lung disease.
Collapse
Affiliation(s)
- Christina M. Eckhardt
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Irving Medical Center, New York, New York
| | - Simone Gambazza
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Healthcare Professions Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Tessa R. Bloomquist
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Peter De Hoff
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Aishwarya Vuppala
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Pantel S. Vokonas
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Augusto A. Litonjua
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York; and
| | - David Sparrow
- Veterans Affairs Normative Aging Study, Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Faruque Parvez
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California
| | - Joel Schwartz
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| | - Haotian Wu
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
12
|
Chen Y, Peng M, Li W, Zhao M, Cao X, Li C, Zhang H, Yang M, Liang L, Yue Y, Xia T, Zhong R, Wang Y, Shu Z. Inhibition of inflammasome activation via sphingolipid pathway in acute lung injury by Huanglian Jiedu decoction: An integrative pharmacology approach. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154469. [PMID: 36202056 DOI: 10.1016/j.phymed.2022.154469] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/21/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a serious health issue which causes significant morbidity and mortality. Inflammation is an important factor in the pathogenesis of ALI. Even though ALI has been successfully managed using a traditiomal Chinese medicine (TCM), Huanglian Jiedu Decoction (HLD), its mechanism of action remains unknown. PURPOSE This study explored the therapeutic potential of HLD in lipopolysaccharide (LPS)-induced ALI rats by utilizing integrative pharmacology. METHODS Here, the therapeutic efficacy of HLD was evaluated using lung wet/dry weight ratio (W/D), myeloperoxide (MPO) activity, and levels of tumor necrosis factor (TNF-α), interleukin (IL)-1β and IL-6. Network pharmacology predictd the active components of HLD in ALI. Lung tissues were subjected to perform Hematoxylin-eosin (H&E) staining, metabolomics, and transcriptomics. The acid ceramidase (ASAH1) inhibitor, carmofur, was employedto suppress the sphingolipid signaling pathway. RESULTS HLD reduced pulmonary edema and vascular permeability, and suppressed the levels of TNF-α, IL-6, and IL-1β in lung tissue, Bronchoalveolar lavage fluid (BALF), and serum. Network pharmacology combined with transcriptomics and metabolomics showed that sphingolipid signaling was the main regulatory pathway for HLD to ameliorate ALI, as confirmed by immunohistochemical analysis. Then, we reverse verified that the sphingolipid signaling pathway was the main pathway involed in ALI. Finally, berberine, baicalein, obacunone, and geniposide were docked with acid ceramidase to further explore the mechanisms of interaction between the compound and protein. CONCLUSION HLD does have a better therapeutic effect on ALI, and its molecular mechanism is better elucidated from the whole, which is to balance lipid metabolism, energy metabolism and amino acid metabolism, and inhibit NLRP3 inflammasome activation by regulating the sphingolipid pathway. Therefore, HLD and its active components can be used to develop new therapies for ALI and provide a new model for exploring complex TCM systems for treating ALI.
Collapse
Affiliation(s)
- Ying Chen
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mingming Peng
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Wei Li
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mantong Zhao
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xia Cao
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chuanqiu Li
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Han Zhang
- School of Pharmacy, Jiamusi University, Jiamusi 154000, China
| | - Mengru Yang
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lanyuan Liang
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yiming Yue
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tianyi Xia
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Renxing Zhong
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Wang
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zunpeng Shu
- Department of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
13
|
Huang P, Zhang J, Duan W, Jiao J, Leng A, Qu J. Plant polysaccharides with anti-lung injury effects as a potential therapeutic strategy for COVID-19. Front Pharmacol 2022; 13:982893. [DOI: 10.3389/fphar.2022.982893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
When coronavirus disease 2019 (COVID-19) develops into the severe phase, lung injury, acute respiratory distress syndrome, and/or respiratory failure could develop within a few days. As a result of pulmonary tissue injury, pathomorphological changes usually present endothelial dysfunction, inflammatory cell infiltration of the lung interstitium, defective gas exchange, and wall leakage. Consequently, COVID-19 may progress to tremendous lung injury, ongoing lung failure, and death. Exploring the treatment drugs has important implications. Recently, the application of traditional Chinese medicine had better performance in reducing fatalities, relieving symptoms, and curtailing hospitalization. Through constant research and study, plant polysaccharides may emerge as a crucial resource against lung injury with high potency and low side effects. However, the absence of a comprehensive understanding of lung-protective mechanisms impedes further investigation of polysaccharides. In the present article, a comprehensive review of research into plant polysaccharides in the past 5 years was performed. In total, 30 types of polysaccharides from 19 kinds of plants have shown lung-protective effects through the pathological processes of inflammation, oxidative stress, apoptosis, autophagy, epithelial–mesenchymal transition, and immunomodulation by mediating mucin and aquaporins, macrophage, endoplasmic reticulum stress, neutrophil, TGF-β1 pathways, Nrf2 pathway, and other mechanisms. Moreover, the deficiencies of the current studies and the future research direction are also tentatively discussed. This research provides a comprehensive perspective for better understanding the mechanism and development of polysaccharides against lung injury for the treatment of COVID-19.
Collapse
|
14
|
Ehrhardt B, El-Merhie N, Kovacevic D, Schramm J, Bossen J, Roeder T, Krauss-Etschmann S. Airway remodeling: The Drosophila model permits a purely epithelial perspective. FRONTIERS IN ALLERGY 2022; 3:876673. [PMID: 36187164 PMCID: PMC9520053 DOI: 10.3389/falgy.2022.876673] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Airway remodeling is an umbrella term for structural changes in the conducting airways that occur in chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). The pathobiology of remodeling involves multiple mesenchymal and lymphoid cell types and finally leads to a variety of hardly reversible changes such as hyperplasia of goblet cells, thickening of the reticular basement membrane, deposition of collagen, peribronchial fibrosis, angiogenesis and hyperplasia of bronchial smooth muscle cells. In order to develop solutions for prevention or innovative therapies, these complex processes must be understood in detail which requires their deconstruction into individual building blocks. In the present manuscript we therefore focus on the role of the airway epithelium and introduce Drosophila melanogaster as a model. The simple architecture of the flies’ airways as well as the lack of adaptive immunity allows to focus exclusively on the importance of the epithelium for the remodeling processes. We will review and discuss genetic and environmentally induced changes in epithelial structures and molecular responses and propose an integrated framework of research for the future.
Collapse
Affiliation(s)
- Birte Ehrhardt
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Natalia El-Merhie
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Draginja Kovacevic
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Juliana Schramm
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
| | - Judith Bossen
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Thomas Roeder
- Division of Molecular Physiology, Institute of Zoology, Christian-Albrechts University Kiel, Kiel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Kiel, Germany
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany
- Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
- Correspondence: Susanne Krauss-Etschmann
| |
Collapse
|
15
|
Zhao Y, Nie X, Han Z, Liu P, Xu H, Huang X, Ren Q. The forkhead box O transcription factor regulates lipase and anti-microbial peptide expressions to promote lipid catabolism and improve innate immunity in the Eriocheir sinensis with hepatopancreatic necrosis disease. FISH & SHELLFISH IMMUNOLOGY 2022; 124:107-117. [PMID: 35378309 DOI: 10.1016/j.fsi.2022.03.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 06/14/2023]
Abstract
Eriocheir sinensis is a crustacean with great economic value, but the occurrence of hepatopancreatic necrosis disease (HPND) severely restricts the development of crab aquaculture. Study on the survival mechanism of crabs with HPND is beneficial to provide new strategies for disease prevention and control. The Forkhead box O (FOXO) transcription factor family is involved in various key biological processes of organisms. In this study, a FOXO gene (named as EsFOXO) from E. sinensis was cloned. The full-length cDNA of EsFOXO is 2592 bp containing a 2133 bp open reading frame that encodes 710 amino acids. EsFOXO was widely distributed in multiple immune tissues. Further study found that the expression of EsFOXO in the intestine of crabs with HPND was significantly upregulated compared with that in the normal crabs. However, whether EsFOXO is involved in the immune and metabolic regulation of crabs remains unknown. RNA interference analysis showed that EsFOXO participates in the positive regulation of the expression of two pancreatic lipases, three anti-lipopolysaccharide factors, and three crustins. Results from our research suggest that two strategies are adopted by crabs with HPND for survival under starvation: on the one hand, the synthesis of antimicrobial peptides is increased to improve the innate immunity; on the other hand, the expression of enzymes correlated with lipid catabolism is up-regulated that mobilizes the fat in the crab, going through catabolism. Our study provides more evidence for an in-depth understanding of the survival mechanism of crabs with HPND.
Collapse
Affiliation(s)
- Yuqi Zhao
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Ximei Nie
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Zhengxiao Han
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Peng Liu
- Nanjing Forestry University, Nanjing, Jiangsu Province, 210037, China
| | - Hao Xu
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China
| | - Xin Huang
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China.
| | - Qian Ren
- Jiangsu Province Engineering Research Center for Aquatic Animals Breeding and Green Efficient Aquacultural Technology, College of Marine Science and Engineering, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, China.
| |
Collapse
|
16
|
Nath AS, Parsons BD, Makdissi S, Chilvers RL, Mu Y, Weaver CM, Euodia I, Fitze KA, Long J, Scur M, Mackenzie DP, Makrigiannis AP, Pichaud N, Boudreau LH, Simmonds AJ, Webber CA, Derfalvi B, Hammon Y, Rachubinski RA, Di Cara F. Modulation of the cell membrane lipid milieu by peroxisomal β-oxidation induces Rho1 signaling to trigger inflammatory responses. Cell Rep 2022; 38:110433. [PMID: 35235794 DOI: 10.1016/j.celrep.2022.110433] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 12/21/2021] [Accepted: 02/01/2022] [Indexed: 12/11/2022] Open
Abstract
Phagocytosis, signal transduction, and inflammatory responses require changes in lipid metabolism. Peroxisomes have key roles in fatty acid homeostasis and in regulating immune function. We find that Drosophila macrophages lacking peroxisomes have perturbed lipid profiles, which reduce host survival after infection. Using lipidomic, transcriptomic, and genetic screens, we determine that peroxisomes contribute to the cell membrane glycerophospholipid composition necessary to induce Rho1-dependent signals, which drive cytoskeletal remodeling during macrophage activation. Loss of peroxisome function increases membrane phosphatidic acid (PA) and recruits RhoGAPp190 during infection, inhibiting Rho1-mediated responses. Peroxisome-glycerophospholipid-Rho1 signaling also controls cytoskeleton remodeling in mouse immune cells. While high levels of PA in cells without peroxisomes inhibit inflammatory phenotypes, large numbers of peroxisomes and low amounts of cell membrane PA are features of immune cells from patients with inflammatory Kawasaki disease and juvenile idiopathic arthritis. Our findings reveal potential metabolic markers and therapeutic targets for immune diseases and metabolic disorders.
Collapse
Affiliation(s)
- Anu S Nath
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Brendon D Parsons
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Stephanie Makdissi
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Rebecca L Chilvers
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Yizhu Mu
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Ceileigh M Weaver
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Irene Euodia
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Katherine A Fitze
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Juyang Long
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Michal Scur
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Duncan P Mackenzie
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Andrew P Makrigiannis
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada
| | - Nicolas Pichaud
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Luc H Boudreau
- Université de Moncton, Department of Chemistry and Biochemistry, Moncton, NB E1A 3E9, Canada; New Brunswick Centre for Precision Medicine (NBCPM), Moncton, NB E1A 3E9, Canada
| | - Andrew J Simmonds
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Christine A Webber
- University of Alberta, Department of Cell Biology, Edmonton, AB T6G 2H7, Canada
| | - Beata Derfalvi
- Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada
| | - Yannick Hammon
- INSERM au Centre d'Immunologie de Marseille Luminy, Marseille 13288, France
| | | | - Francesca Di Cara
- Dalhousie University, Department of Microbiology and Immunology, Halifax, NS B3K 6R8, Canada; Dalhousie University, Department of Pediatrics, Halifax, NS B3K 6R8, Canada.
| |
Collapse
|
17
|
Xiang M, Liu T, Tian C, Ma K, Gou J, Huang R, Li S, Li Q, Xu C, Li L, Lee CH, Zhang Y. Kinsenoside attenuates liver fibro-inflammation by suppressing dendritic cells via the PI3K-AKT-FoxO1 pathway. Pharmacol Res 2022; 177:106092. [PMID: 35066108 PMCID: PMC8776354 DOI: 10.1016/j.phrs.2022.106092] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/25/2022]
Abstract
Kinsenoside (KD) exhibits anti-inflammatory and immunosuppressive effects. Dendritic cells (DCs) are critical regulators of the pathologic inflammatory milieu in liver fibrosis (LF). Herein, we explored whether and how KD repressed development of LF via DC regulation and verified the pathway involved in the process. Given our analysis, both KD and adoptive transfer of KD-conditioned DCs conspicuously reduced hepatic histopathological damage, proinflammatory cytokine release and extracellular matrix deposition in CCl4-induced LF mice. Of note, KD restrained the LF-driven rise in CD86, MHC-II, and CCR7 levels and, simultaneously, upregulated PD-L1 expression on DCs specifically, which blocked CD8+T cell activation. Additionally, KD reduced DC glycolysis, maintained DCs immature, accompanied by IL-12 decrease in DCs. Inhibiting DC function by KD disturbed the communication of DCs and HSCs with the expression or secretion of α-SMA and Col-I declined in the liver. Mechanistically, KD suppressed the phosphorylation of PI3K-AKT driven by LF or PI3K agonist, followed by enhanced nuclear transport of FoxO1 and upregulated interaction of FoxO1 with the PD-L1 promoter in DCs. PI3K inhibitor or si-IL-12 acting on DC could relieve LF, HSC activation and diminish the effect of KD. In conclusion, KD suppressed DC maturation with promoted PD-L1 expression via PI3K-AKT-FoxO1 and decreased IL-12 secretion, which blocked activation of CD8+T cells and HSCs, thereby alleviating liver injury and fibro-inflammation in LF.
Collapse
Affiliation(s)
- Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tingting Liu
- Department of Pharmacy, the First Affiliated Hospital of Anhui Medical University, the Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kun Ma
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jing Gou
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rongrong Huang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Senlin Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chuanrui Xu
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Li
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Yonghui Zhang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Jia X, Huang J, Wu B, Yang M, Xu W. A Competitive Endogenous RNA Network Based on Differentially Expressed lncRNA in Lipopolysaccharide-Induced Acute Lung Injury in Mice. Front Genet 2021; 12:745715. [PMID: 34917127 PMCID: PMC8669720 DOI: 10.3389/fgene.2021.745715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/14/2021] [Indexed: 12/03/2022] Open
Abstract
Non-coding RNAs have remarkable roles in acute lung injury (ALI) initiation. Nevertheless, the significance of long non-coding RNAs (lncRNAs) in ALI is still unknown. Herein, we purposed to identify potential key genes in ALI and create a competitive endogenous RNA (ceRNA) modulatory network to uncover possible molecular mechanisms that affect lung injury. We generated a lipopolysaccharide-triggered ALI mouse model, whose lung tissue was subjected to RNA sequencing, and then we conducted bioinformatics analysis to select genes showing differential expression (DE) and to build a lncRNA-miRNA (microRNA)- mRNA (messenger RNA) modulatory network. Besides, GO along with KEGG assessments were conducted to identify major biological processes and pathways, respectively, involved in ALI. Then, RT-qPCR assay was employed to verify levels of major RNAs. A protein-protein interaction (PPI) network was created using the Search Tool for the Retrieval of Interacting Genes (STRING) database, and the hub genes were obtained with the Molecular Complex Detection plugin. Finally, a key ceRNA subnetwork was built from major genes and their docking sites. Overall, a total of 8,610 lncRNAs were identified in the normal and LPS groups. Based on the 308 DE lncRNAs [p-value < 0.05, |log2 (fold change) | > 1] and 3,357 DE mRNAs [p-value < 0.05, |log2 (fold change) | > 1], lncRNA-miRNA and miRNA-mRNA pairs were predicted using miRanda. The lncRNA-miRNA-mRNA network was created from 175 lncRNAs, 22 miRNAs, and 209 mRNAs in ALI. The RT-qPCR data keep in step with the RNA sequencing data. GO along with KEGG analyses illustrated that DE mRNAs in this network were mainly bound up with the inflammatory response, developmental process, cell differentiation, cell proliferation, apoptosis, and the NF-kappa B, PI3K-Akt, HIF-1, MAPK, Jak-STAT, and Notch signaling pathways. A PPI network on the basis of the 209 genes was established, and three hub genes (Nkx2-1, Tbx2, and Atf5) were obtained from the network. Additionally, a lncRNA-miRNA-hub gene subnetwork was built from 15 lncRNAs, 3 miRNAs, and 3 mRNAs. Herein, novel ideas are presented to expand our knowledge on the regulation mechanisms of lncRNA-related ceRNAs in the pathogenesis of ALI.
Collapse
Affiliation(s)
- Xianxian Jia
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinhui Huang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bo Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Miao Yang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Xu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
19
|
Salomão R, Neto IVDS, Ramos GV, Tibana RA, Durigan JQ, Pereira GB, Franco OL, Royer C, Neves FDAR, de Carvalho ACA, Nóbrega OT, Haddad R, Prestes J, Marqueti RDC. Paternal Resistance Exercise Modulates Skeletal Muscle Remodeling Pathways in Fathers and Male Offspring Submitted to a High-Fat Diet. Front Physiol 2021; 12:706128. [PMID: 34646148 PMCID: PMC8503191 DOI: 10.3389/fphys.2021.706128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/02/2021] [Indexed: 11/24/2022] Open
Abstract
Although some studies have shown that a high-fat diet (HFD) adversely affects muscle extracellular matrix remodeling, the mechanisms involved in muscle trophism, inflammation, and adipogenesis have not been fully investigated. Thus, we investigated the effects of 8 weeks of paternal resistance training (RT) on gene and protein expression/activity of critical factors involved in muscle inflammation and remodeling of fathers and offspring (offspring exposed to standard chow or HFD). Animals were randomly distributed to constitute sedentary fathers (SF; n = 7; did not perform RT) or trained fathers (TF n = 7; performed RT), with offspring from mating with sedentary females. After birth, 28 male pups were divided into four groups (n = 7 per group): offspring from sedentary father submitted either to control diet (SFO-C) or high-fat diet (SFO-HF) and offspring from trained father submitted to control diet (TFO-C) or high-fat diet (TFO-HF). Our results show that an HFD downregulated collagen mRNA levels and upregulated inflammatory and atrophy pathways and adipogenic transcription factor mRNA levels in offspring gastrocnemius muscle. In contrast, paternal RT increased MMP-2 activity and decreased IL-6 levels in offspring exposed to a control diet. Paternal RT upregulated P70s6k and Ppara mRNA levels and downregulated Atrogin1 mRNA levels, while decreasing NFκ-B, IL-1β, and IL-8 protein levels in offspring exposed to an HFD. Paternal physical training influences key skeletal muscle remodeling pathways and inflammatory profiles relevant for muscle homeostasis maintenance in offspring submitted to different diets.
Collapse
Affiliation(s)
- Rebecca Salomão
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program in Rehabilitation Sciences, Universidade de Brasília, Brasília, Brazil
| | - Ivo Vieira de Sousa Neto
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program of Sciences and Technology of Health, Universidade de Brasília, Brasília, Brazil
| | | | - Ramires Alsamir Tibana
- Graduate Program in Health Sciences, Faculdade de Medicine, Universidade Federal do Mato Grosso (UFTM), Cuiabá, Brazil
| | | | - Guilherme Borges Pereira
- Interinstitutional Program of Post-Graduation in Physiological Sciences (UFSCar/UNESP), Department of Physiological Sciences, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Octávio Luiz Franco
- Graduate Program in Genomics Science and Biotechnology, Universidade Católica de Brasília, Brasília, Brazil.,S-Inova Biotech, Graduate Program in Biotechnology, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Carine Royer
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Laboratory of Molecular Pharmacology, Faculty of Health Sciences, Universidade de Brasília, Brasília, Brazil
| | | | | | - Otávio Toledo Nóbrega
- Graduate Program of Medical Sciences, Universidade de Brasília, Brasília, Brazil.,Center for Tropical Medicine, Universidade de Brasília, Brasília, Brazil
| | - Rodrigo Haddad
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Center for Tropical Medicine, Universidade de Brasília, Brasília, Brazil
| | - Jonato Prestes
- Graduate Program of Physical Education, Universidade Católica de Brasilia, Brasília, Brazil
| | - Rita de Cássia Marqueti
- Laboratory of Molecular Analysis, Faculty of Ceilândia, Universidade de Brasília, Brasília, Brazil.,Graduate Program in Rehabilitation Sciences, Universidade de Brasília, Brasília, Brazil.,Graduate Program of Sciences and Technology of Health, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|
20
|
Chi Q, Hu X, Liu Z, Han Y, Tao D, Xu S, Li S. H 2S exposure induces cell death in the broiler thymus via the ROS-initiated JNK/MST1/FOXO1 pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 222:112488. [PMID: 34246945 DOI: 10.1016/j.ecoenv.2021.112488] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
Hydrogen sulfide (H2S) is a common toxic gas in chicken houses that endangers the health of poultry. Harbin has a cold climate in winter, and the conflict between heat preservation and ventilation in poultry houses is obvious. In this study, we investigated the H2S content in chicken houses during winter in Harbin and found that the H2S concentration exceeded the national standard in individual chicken houses. Then, a model of H2S exposure was established in an environmental simulation chamber. We also developed a NaHS exposure model of chicken peripheral blood lymphocytes in vitro. Proteomics analysis was used to reveal the toxicology of thymus injury in broilers, the FOXO signaling pathway was determined to be significantly enriched, ROS bursts and JNK/MST1/FOXO1 pathway activation induced by H2S exposure were detected, and ROS played an important switch role in the JNK/MST1/FOXO1 pathway. In addition, H2S exposure-induced thymus cell death involved immune dysregulation. Overall, the present study adds data for H2S contents in chicken houses, provides new findings for the mechanism of H2S poisoning and reveals a new regulatory pathway in immune injury.
Collapse
Affiliation(s)
- Qianru Chi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Xueyuan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Zhaoyi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yanfei Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Dayong Tao
- College of Animal Science, Tarim University, Alar, Xinjiang Uygur Autonomous Region 843300, China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China; College of Animal Science, Tarim University, Alar, Xinjiang Uygur Autonomous Region 843300, China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| |
Collapse
|
21
|
Cheema PS, Nandi D, Nag A. Exploring the therapeutic potential of forkhead box O for outfoxing COVID-19. Open Biol 2021; 11:210069. [PMID: 34102081 PMCID: PMC8187014 DOI: 10.1098/rsob.210069] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/27/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has wreaked unprecedented societal havoc worldwide. The infected individuals may present mild to severe symptoms, with nearly 20% of the confirmed patients impaired with significant complications, including multi-organ failure. Acute respiratory distress imposed by SARS-CoV-2 largely results from an aggravated cytokine storm and deregulated immune response. The forkhead box O (FoxO) transcription factors are reported to play a significant role in maintaining normal cell physiology by regulating survival, apoptosis, oxidative stress, development and maturation of T and B lymphocytes, secretion of inflammatory cytokines, etc. We propose a potent anti-inflammatory approach based on activation of the FoxO as an attractive strategy against the novel coronavirus. This regime will be focused on restoring redox and inflammatory homeostasis along with repair of the damaged tissue, activation of lymphocyte effector and memory cells. Repurposing FoxO activators as a means to alleviate the inflammatory burst following SARS-CoV-2 infection can prove immensely valuable in the ongoing pandemic and provide a reliable groundwork for enriching our repertoire of antiviral modalities for any such complication in the future. Altogether, our review highlights the possible efficacy of FoxO activation as a novel arsenal for clinical management of COVID-19.
Collapse
Affiliation(s)
- Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Deeptashree Nandi
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi, South Campus, Biotech Building, 2nd Floor, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| |
Collapse
|
22
|
Künzi L, Easter M, Hirsch MJ, Krick S. Cystic Fibrosis Lung Disease in the Aging Population. Front Pharmacol 2021; 12:601438. [PMID: 33935699 PMCID: PMC8082404 DOI: 10.3389/fphar.2021.601438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/15/2021] [Indexed: 01/02/2023] Open
Abstract
The demographics of the population with cystic fibrosis (CF) is continuously changing, with nowadays adults outnumbering children and a median predicted survival of over 40 years. This leads to the challenge of treating an aging CF population, while previous research has largely focused on pediatric and adolescent patients. Chronic inflammation is not only a hallmark of CF lung disease, but also of the aging process. However, very little is known about the effects of an accelerated aging pathology in CF lungs. Several chronic lung disease pathologies show signs of chronic inflammation with accelerated aging, also termed “inflammaging”; the most notable being chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In these disease entities, accelerated aging has been implicated in the pathogenesis via interference with tissue repair mechanisms, alterations of the immune system leading to impaired defense against pulmonary infections and induction of a chronic pro-inflammatory state. In addition, CF lungs have been shown to exhibit increased expression of senescence markers. Sustained airway inflammation also leads to the degradation and increased turnover of cystic fibrosis transmembrane regulator (CFTR). This further reduces CFTR function and may prevent the novel CFTR modulator therapies from developing their full efficacy. Therefore, novel therapies targeting aging processes in CF lungs could be promising. This review summarizes the current research on CF in an aging population focusing on accelerated aging in the context of chronic airway inflammation and therapy implications.
Collapse
Affiliation(s)
- Lisa Künzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Epidemiology, Biostatistics and Prevention Institute, Department of Public and Global Health, University of Zürich, Zürich, Switzerland
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Gregory Fleming Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL, United States.,Comprehensive Center for Healthy Aging, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
23
|
Wagner C, Uliczka K, Bossen J, Niu X, Fink C, Thiedmann M, Knop M, Vock C, Abdelsadik A, Zissler UM, Isermann K, Garn H, Pieper M, Wegmann M, Koczulla AR, Vogelmeier CF, Schmidt-Weber CB, Fehrenbach H, König P, Silverman N, Renz H, Pfefferle P, Heine H, Roeder T. Constitutive immune activity promotes JNK- and FoxO-dependent remodeling of Drosophila airways. Cell Rep 2021; 35:108956. [PMID: 33826881 DOI: 10.1016/j.celrep.2021.108956] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/29/2020] [Accepted: 03/17/2021] [Indexed: 01/07/2023] Open
Abstract
Extensive remodeling of the airways is a major characteristic of chronic inflammatory lung diseases such as asthma or chronic obstructive pulmonary disease (COPD). To elucidate the importance of a deregulated immune response in the airways for remodeling processes, we established a matching Drosophila model. Here, triggering the Imd (immune deficiency) pathway in tracheal cells induced organ-wide remodeling. This structural remodeling comprises disorganization of epithelial structures and comprehensive epithelial thickening. We show that these structural changes do not depend on the Imd pathway's canonical branch terminating on nuclear factor κB (NF-κB) activation. Instead, activation of a different segment of the Imd pathway that branches off downstream of Tak1 and comprises activation of c-Jun N-terminal kinase (JNK) and forkhead transcription factor of the O subgroup (FoxO) signaling is necessary and sufficient to mediate the observed structural changes of the airways. Our findings imply that targeting JNK and FoxO signaling in the airways could be a promising strategy to interfere with disease-associated airway remodeling processes.
Collapse
Affiliation(s)
- Christina Wagner
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Division of Invertebrate Models, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Karin Uliczka
- Division of Invertebrate Models, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Division of Innate Immunity, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Judith Bossen
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Xiao Niu
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Christine Fink
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Marcus Thiedmann
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Mirjam Knop
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Christina Vock
- Division of Experimental Pneumology, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany
| | - Ahmed Abdelsadik
- Zoology, Aswan University, Aswan 81528, Egypt; Molecular Biotechnology Program, Faculty of Advanced Basic Sciences, Galala University, 43552 New Galala, Egypt
| | - Ulrich M Zissler
- Center of Allergy and Environment (ZAUM), Technical University Munich and Helmholtz Center Munich, German Research Center for Environmental Health, 80802 Munich, Germany; CPC-M, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kerstin Isermann
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Mario Pieper
- University Lübeck, Anatomical Institute, 23538 Lübeck, Germany
| | - Michael Wegmann
- Division of Asthma Exacerbation & Regulation, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Andreas R Koczulla
- Pulmonary and Critical Care Medicine, Department of Medicine, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Claus F Vogelmeier
- Pulmonary and Critical Care Medicine, Department of Medicine, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University Munich and Helmholtz Center Munich, German Research Center for Environmental Health, 80802 Munich, Germany; CPC-M, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Heinz Fehrenbach
- Division of Experimental Pneumology, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Peter König
- University Lübeck, Anatomical Institute, 23538 Lübeck, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Neil Silverman
- University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Harald Renz
- Molecular Diagnostics, Institute of Laboratory Medicine and Pathobiochemistry, Medical Faculty, Philipps University of Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Petra Pfefferle
- Comprehensive Biobank Marburg, University Medical Center Giessen and Marburg, Medical Faculty, Philipps University Marburg, 35043 Marburg, Germany; UGMLC, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Holger Heine
- Division of Innate Immunity, Priority Research Area Asthma and Allergy, Research Center Borstel, 23845 Borstel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Thomas Roeder
- Zoology, Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany.
| |
Collapse
|
24
|
Li C, Hong PP, Yang MC, Zhao XF, Wang JX. FOXO regulates the expression of antimicrobial peptides and promotes phagocytosis of hemocytes in shrimp antibacterial immunity. PLoS Pathog 2021; 17:e1009479. [PMID: 33798239 PMCID: PMC8046353 DOI: 10.1371/journal.ppat.1009479] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/14/2021] [Accepted: 03/16/2021] [Indexed: 01/11/2023] Open
Abstract
Invertebrates rely on innate immunity, including humoral and cellular immunity, to resist pathogenic infection. Previous studies showed that forkhead box transcription factor O (FOXO) participates in mucosal immune responses of mammals and the gut humoral immune regulation of invertebrates. However, whether FOXO is involved in systemic and cellular immunity regulation in invertebrates remains unknown. In the present study, we identified a FOXO from shrimp (Marsupenaeus japonicus) and found that it was expressed at relatively basal levels in normal shrimp, but was upregulated significantly in shrimp challenged by Vibrio anguillarum. FOXO played a critical role in maintaining hemolymph and intestinal microbiota homeostasis by promoting the expression of Relish, the transcription factor of the immune deficiency (IMD) pathway for expression of antimicrobial peptides (AMPs) in shrimp. We also found that pathogen infection activated FOXO and induced its nuclear translocation by reducing serine/threonine kinase AKT activity. In the nucleus, activated FOXO directly regulated the expression of its target Amp and Relish genes against bacterial infection. Furthermore, FOXO was identified as being involved in cellular immunity by promoting the phagocytosis of hemocytes through upregulating the expression of the phagocytotic receptor scavenger receptor C (Src), and two small GTPases, Rab5 and Rab7, which are related to phagosome trafficking to the lysosome in the cytoplasm. Taken together, our results indicated that FOXO exerts its effects on homeostasis of hemolymph and the enteric microbiota by activating the IMD pathway in normal shrimp, and directly or indirectly promoting AMP expression and enhancing phagocytosis of hemocytes against pathogens in bacteria-infected shrimp. This study revealed the different functions of FOXO in the mucosal (local) and systemic antibacterial immunity of invertebrates.
Collapse
Affiliation(s)
- Cang Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Pan-Pan Hong
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Ming-Chong Yang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
| | - Jin-Xing Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, Shandong, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, China
| |
Collapse
|
25
|
Farhan M, Silva M, Li S, Yan F, Fang J, Peng T, Hu J, Tsao M, Little P, Zheng W. The role of FOXOs and autophagy in cancer and metastasis-Implications in therapeutic development. Med Res Rev 2020; 40:2089-2113. [PMID: 32474970 PMCID: PMC7586888 DOI: 10.1002/med.21695] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 12/17/2022]
Abstract
Autophagy is a highly conserved intracellular degradation process that plays a crucial role in cell survival and stress reactions as well as in cancer development and metastasis. Autophagy process involves several steps including sequestration, fusion of autophagosomes with lysosomes and degradation. Forkhead box O (FOXO) transcription factors regulate the expression of genes involved in cellular metabolic activity and signaling pathways of cancer growth and metastasis. Recent evidence suggests that FOXO proteins are also involved in autophagy regulation. The relationship among FOXOs, autophagy, and cancer has been drawing attention of many who work in the field. This study summarizes the role of FOXO proteins and autophagy in cancer growth and metastasis and analyzes their potential roles in cancer disease management.
Collapse
Affiliation(s)
- Mohd Farhan
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Marta Silva
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Shuai Li
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Fengxia Yan
- Department of MedicineJinan UniversityGuangzhouChina
| | - Jiankang Fang
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Tangming Peng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| | - Jim Hu
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Ming‐Sound Tsao
- Department of Laboratory Medicine and PathobiologyUniversity of TorontoTorontoOntarioCanada
| | - Peter Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Wenhua Zheng
- Faculty of Health SciencesCentre of Reproduction, Development and Aging, Institute of Translational Medicine, University of MacauTaipaMacau SARChina
| |
Collapse
|
26
|
Al-Mukh H, Baudoin L, Bouaboud A, Sanchez-Salgado JL, Maraqa N, Khair M, Pagesy P, Bismuth G, Niedergang F, Issad T. Lipopolysaccharide Induces GFAT2 Expression to Promote O-Linked β- N-Acetylglucosaminylation and Attenuate Inflammation in Macrophages. THE JOURNAL OF IMMUNOLOGY 2020; 205:2499-2510. [PMID: 32978282 DOI: 10.4049/jimmunol.2000345] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/29/2020] [Indexed: 12/14/2022]
Abstract
Glycosylation with O-linked β-N-acetylglucosamine (O-GlcNAcylation) is a reversible posttranslational modification that regulates the activity of intracellular proteins according to glucose availability and its metabolism through the hexosamine biosynthesis pathway. This modification has been involved in the regulation of various immune cell types, including macrophages. However, little is known concerning the mechanisms that regulate the protein O-GlcNAcylation level in these cells. In the present work, we demonstrate that LPS treatment induces a marked increase in protein O-GlcNAcylation in RAW264.7 cells, bone marrow-derived and peritoneal mouse macrophages, as well as human monocyte-derived macrophages. Targeted deletion of OGT in macrophages resulted in an increased effect of LPS on NOS2 expression and cytokine production, suggesting that O-GlcNAcylation may restrain inflammatory processes induced by LPS. The effect of LPS on protein O-GlcNAcylation in macrophages was associated with an increased expression and activity of glutamine fructose 6-phosphate amidotransferase (GFAT), the enzyme that catalyzes the rate-limiting step of the hexosamine biosynthesis pathway. More specifically, we observed that LPS potently stimulated GFAT2 isoform mRNA and protein expression. Genetic or pharmacological inhibition of FoxO1 impaired the LPS effect on GFAT2 expression, suggesting a FoxO1-dependent mechanism. We conclude that GFAT2 should be considered a new LPS-inducible gene involved in regulation of protein O-GlcNAcylation, which permits limited exacerbation of inflammation upon macrophage activation.
Collapse
Affiliation(s)
- Hasanain Al-Mukh
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | - Léa Baudoin
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | | | | | - Nabih Maraqa
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | - Mostafa Khair
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | - Patrick Pagesy
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | - Georges Bismuth
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| | | | - Tarik Issad
- Université de Paris, Institut Cochin, CNRS, INSERM, F-75014 Paris, France
| |
Collapse
|
27
|
Kerschner JL, Paranjapye A, Yin S, Skander DL, Bebek G, Leir SH, Harris A. A functional genomics approach to investigate the differentiation of iPSCs into lung epithelium at air-liquid interface. J Cell Mol Med 2020; 24:9853-9870. [PMID: 32692488 PMCID: PMC7520342 DOI: 10.1111/jcmm.15568] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 01/24/2023] Open
Abstract
The availability of robust protocols to differentiate induced pluripotent stem cells (iPSCs) into many human cell lineages has transformed research into the origins of human disease. The efficacy of differentiating iPSCs into specific cellular models is influenced by many factors including both intrinsic and extrinsic features. Among the most challenging models is the generation of human bronchial epithelium at air‐liquid interface (HBE‐ALI), which is the gold standard for many studies of respiratory diseases including cystic fibrosis. Here, we perform open chromatin mapping by ATAC‐seq and transcriptomics by RNA‐seq in parallel, to define the functional genomics of key stages of the iPSC to HBE‐ALI differentiation. Within open chromatin peaks, the overrepresented motifs include the architectural protein CTCF at all stages, while motifs for the FOXA pioneer and GATA factor families are seen more often at early stages, and those regulating key airway epithelial functions, such as EHF, are limited to later stages. The RNA‐seq data illustrate dynamic pathways during the iPSC to HBE‐ALI differentiation, and also the marked functional divergence of different iPSC lines at the ALI stages of differentiation. Moreover, a comparison of iPSC‐derived and lung donor‐derived HBE‐ALI cultures reveals substantial differences between these models.
Collapse
Affiliation(s)
- Jenny L Kerschner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Alekh Paranjapye
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Shiyi Yin
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Dannielle L Skander
- Systems Biology and Bioinformatics Graduate Program, Case Western Reserve University, Cleveland, OH, USA
| | - Gurkan Bebek
- Systems Biology and Bioinformatics Graduate Program, Case Western Reserve University, Cleveland, OH, USA.,Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, OH, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA.,Electrical Engineering and Computer Science Department, Case Western Reserve University, Cleveland, OH, USA
| | - Shih-Hsing Leir
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Ann Harris
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
28
|
Sazzini M, Abondio P, Sarno S, Gnecchi-Ruscone GA, Ragno M, Giuliani C, De Fanti S, Ojeda-Granados C, Boattini A, Marquis J, Valsesia A, Carayol J, Raymond F, Pirazzini C, Marasco E, Ferrarini A, Xumerle L, Collino S, Mari D, Arosio B, Monti D, Passarino G, D'Aquila P, Pettener D, Luiselli D, Castellani G, Delledonne M, Descombes P, Franceschi C, Garagnani P. Genomic history of the Italian population recapitulates key evolutionary dynamics of both Continental and Southern Europeans. BMC Biol 2020; 18:51. [PMID: 32438927 PMCID: PMC7243322 DOI: 10.1186/s12915-020-00778-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/01/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The cline of human genetic diversity observable across Europe is recapitulated at a micro-geographic scale by variation within the Italian population. Besides resulting from extensive gene flow, this might be ascribable also to local adaptations to diverse ecological contexts evolved by people who anciently spread along the Italian Peninsula. Dissecting the evolutionary history of the ancestors of present-day Italians may thus improve the understanding of demographic and biological processes that contributed to shape the gene pool of European populations. However, previous SNP array-based studies failed to investigate the full spectrum of Italian variation, generally neglecting low-frequency genetic variants and examining a limited set of small effect size alleles, which may represent important determinants of population structure and complex adaptive traits. To overcome these issues, we analyzed 38 high-coverage whole-genome sequences representative of population clusters at the opposite ends of the cline of Italian variation, along with a large panel of modern and ancient Euro-Mediterranean genomes. RESULTS We provided evidence for the early divergence of Italian groups dating back to the Late Glacial and for Neolithic and distinct Bronze Age migrations having further differentiated their gene pools. We inferred adaptive evolution at insulin-related loci in people from Italian regions with a temperate climate, while possible adaptations to pathogens and ultraviolet radiation were observed in Mediterranean Italians. Some of these adaptive events may also have secondarily modulated population disease or longevity predisposition. CONCLUSIONS We disentangled the contribution of multiple migratory and adaptive events in shaping the heterogeneous Italian genomic background, which exemplify population dynamics and gene-environment interactions that played significant roles also in the formation of the Continental and Southern European genomic landscapes.
Collapse
Affiliation(s)
- Marco Sazzini
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy.
- Interdepartmental Centre Alma Mater Research Institute on Global Challenges and Climate Change, University of Bologna, Bologna, Italy.
| | - Paolo Abondio
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Stefania Sarno
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | | | - Matteo Ragno
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Cristina Giuliani
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Sara De Fanti
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudia Ojeda-Granados
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
- Department of Molecular Biology in Medicine, Civil Hospital of Guadalajara "Fray Antonio Alcalde" and Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Alessio Boattini
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Julien Marquis
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
- Current Address: Lausanne Genomic Technologies Facility, University of Lausanne, Lausanne, Switzerland
| | - Armand Valsesia
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | - Jerome Carayol
- Nestlé Research, EPFL Innovation Park, Lausanne, Switzerland
| | | | - Chiara Pirazzini
- IRCCS Bologna Institute of Neurological Sciences, Bologna, Italy
| | - Elena Marasco
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
- Applied Biomedical Research Center (CRBA), S. Orsola-Malpighi Polyclinic, Bologna, Italy
| | - Alberto Ferrarini
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
- Current Address: Menarini Silicon Biosystems SpA, Castel Maggiore, Bologna, Italy
| | - Luciano Xumerle
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
| | | | - Daniela Mari
- Geriatric Unit, Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Beatrice Arosio
- Geriatric Unit, Fondazione Ca' Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniela Monti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Patrizia D'Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Davide Pettener
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Donata Luiselli
- Department of Cultural Heritage, University of Bologna, Ravenna, Italy
| | - Gastone Castellani
- Interdepartmental Centre Alma Mater Research Institute on Global Challenges and Climate Change, University of Bologna, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Massimo Delledonne
- Functional Genomics Laboratory, Department of Biotechnology, University of Verona, Verona, Italy
| | | | - Claudio Franceschi
- Department of Applied Mathematics, Institute of Information Technology, Lobachevsky University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Paolo Garagnani
- Interdepartmental Centre Alma Mater Research Institute on Global Challenges and Climate Change, University of Bologna, Bologna, Italy.
- Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy.
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden.
| |
Collapse
|
29
|
Ponton F, Morimoto J, Robinson K, Kumar SS, Cotter SC, Wilson K, Simpson SJ. Macronutrients modulate survival to infection and immunity in Drosophila. J Anim Ecol 2019; 89:460-470. [PMID: 31658371 PMCID: PMC7027473 DOI: 10.1111/1365-2656.13126] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 07/17/2019] [Indexed: 12/21/2022]
Abstract
Immunity and nutrition are two essential modulators of individual fitness. However, while the implications of immune function and nutrition on an individual's lifespan and reproduction are well established, the interplay between feeding behaviour, infection and immune function remains poorly understood. Asking how ecological and physiological factors affect immune responses and resistance to infections is a central theme of eco‐immunology. In this study, we used the fruit fly, Drosophila melanogaster, to investigate how infection through septic injury modulates nutritional intake and how macronutrient balance affects survival to infection by the pathogenic Gram‐positive bacterium Micrococcus luteus. Our results show that infected flies maintain carbohydrate intake, but reduce protein intake, thereby shifting from a protein‐to‐carbohydrate (P:C) ratio of ~1:4 to ~1:10 relative to non‐infected and sham‐infected flies. Strikingly, the proportion of flies dying after M. luteus infection was significantly lower when flies were fed a low‐P high‐C diet, revealing that flies shift their macronutrient intake as means of nutritional self‐medication against bacterial infection. These results are likely due to the effects of the macronutrient balance on the regulation of the constitutive expression of innate immune genes, as a low‐P high‐C diet was linked to an upregulation in the expression of key antimicrobial peptides. Together, our results reveal the intricate relationship between macronutrient intake and resistance to infection and integrate the molecular cross‐talk between metabolic and immune pathways into the framework of nutritional immunology.
Collapse
Affiliation(s)
- Fleur Ponton
- Department of Biological Sciences, Macquarie University, Sydney, NSW, Australia
| | - Juliano Morimoto
- Department of Biological Sciences, Macquarie University, Sydney, NSW, Australia
| | - Katie Robinson
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Sheemal S Kumar
- Department of Biological Sciences, Macquarie University, Sydney, NSW, Australia
| | | | - Kenneth Wilson
- Lancaster Environment Centre, Lancaster University, Lancaster, UK
| | - Stephen J Simpson
- Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
30
|
FOXO3a regulates rhinovirus-induced innate immune responses in airway epithelial cells. Sci Rep 2019; 9:18180. [PMID: 31796819 PMCID: PMC6890790 DOI: 10.1038/s41598-019-54567-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/15/2019] [Indexed: 12/27/2022] Open
Abstract
Forkhead transcription factor class O (FOXO)3a, which plays a critical role in a wide variety of cellular processes, was also found to regulate cell-type-specific antiviral responses. Airway epithelial cells express FOXO3a and play an important role in clearing rhinovirus (RV) by mounting antiviral type I and type III interferon (IFN) responses. To elucidate the role of FOXO3a in regulating antiviral responses, we generated airway epithelial cell-specific Foxo3a knockout (Scga1b1-Foxo3a−/−) mice and a stable FOXO3a knockout human airway epithelial cell line. Compared to wild-type, Scga1b1-Foxo3a−/− mice show reduced IFN-α, IFN-β, IFN-λ2/3 in response to challenge with RV or double-stranded (ds)RNA mimic, Poly Inosinic-polycytidylic acid (Poly I:C) indicating defective dsRNA receptor signaling. RV-infected Scga1b1-Foxo3a−/− mice also show viral persistence, enhanced lung inflammation and elevated pro-inflammatory cytokine levels. FOXO3a K/O airway epithelial cells show attenuated IFN responses to RV infection and this was associated with conformational change in mitochondrial antiviral signaling protein (MAVS) but not with a reduction in the expression of dsRNA receptors under unstimulated conditions. Pretreatment with MitoTEMPO, a mitochondrial-specific antioxidant corrects MAVS conformation and restores antiviral IFN responses to subsequent RV infection in FOXO3a K/O cells. Inhibition of oxidative stress also reduces pro-inflammatory cytokine responses to RV in FOXO3a K/O cells. Together, our results indicate that FOXO3a plays a critical role in regulating antiviral responses as well as limiting pro-inflammatory cytokine expression. Based on these results, we conclude that FOXO3a contributes to optimal viral clearance and prevents excessive lung inflammation following RV infection.
Collapse
|
31
|
Martínez M, Polizzotto A, Flores N, Semorile L, Maffía PC. Antibacterial, anti-biofilm and in vivo activities of the antimicrobial peptides P5 and P6.2. Microb Pathog 2019; 139:103886. [PMID: 31778756 DOI: 10.1016/j.micpath.2019.103886] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 10/22/2019] [Accepted: 11/21/2019] [Indexed: 01/03/2023]
Abstract
Cationic antimicrobial peptides (AMPs) are short linear amino acid sequences, which display antimicrobial activity against a wide range of bacterial species. They are promising novel antimicrobials since they have shown bactericidal effects against multiresistant bacteria. Their amphiphilic structure with hydrophobic and cationic regions drives their interaction with anionic bacterial cytoplasmic membranes, which leads to their disruption. In this work two synthetic designed AMPs, P5 and P6.2, which have been previously analyzed in their ability to interact with bacterial or eukaryotic membranes, were evaluated in their anti-biofilm and in vivo antibacterial activity. In a first step, a time-kill kinetic assay against P. aeruginosa and S. aureus and a curve for hemolytic activity were performed in order to determine the killing rate and the possible undesirable toxic effect, respectively, for both peptides. The biofilm inhibitory activity was quantified at sub MIC concentrations of the peptides and the results showed that P5 displayed antibiofilm activity on both strains while P6.2 only on S. aureus. Scanning electron microscopy (SEM) of bacteria treated with peptides at their MIC revealed protruding blisters on Gam-negative P. aeruginosa strain, but almost no visible surface alteration on Gram-positive S. aureus. These micrographs highlighted different manifestations of the membrane-disrupting activity that these kinds of peptides possess. Finally, both peptides were analyzed in vivo, in the lungs of neutropenic mice previously instilled with P. aeruginosa. Mice lungs were surgically extracted and bacteria and pro-inflammatory cytokines (IL-β, IL-6 and TNF-α) were quantified by colony forming units and ELISA, respectively. Results showed that instillation of the peptides produced a significant decrease in the number of living bacteria in the lungs, concomitant with a decrease in pro-inflammatory cytokines. Overall, the results presented here suggest that these two new peptides could be good candidates for future drug development for anti-biofilm and anti-infective therapy.
Collapse
Affiliation(s)
- Melina Martínez
- Laboratorio de Microbiología Molecular, Instituto de Microbiología Básica y Aplicada, Universidad Nacional de Quilmes, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Axel Polizzotto
- Laboratorio de Microbiología Molecular, Instituto de Microbiología Básica y Aplicada, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Naiquén Flores
- Laboratorio de Microbiología Molecular, Instituto de Microbiología Básica y Aplicada, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Liliana Semorile
- Laboratorio de Microbiología Molecular, Instituto de Microbiología Básica y Aplicada, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Paulo César Maffía
- Laboratorio de Microbiología Molecular, Instituto de Microbiología Básica y Aplicada, Universidad Nacional de Quilmes, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina.
| |
Collapse
|
32
|
Atherton LJ, Jorquera PA, Bakre AA, Tripp RA. Determining Immune and miRNA Biomarkers Related to Respiratory Syncytial Virus (RSV) Vaccine Types. Front Immunol 2019; 10:2323. [PMID: 31649663 PMCID: PMC6794384 DOI: 10.3389/fimmu.2019.02323] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) causes serious respiratory tract illness and substantial morbidity and some mortality in populations at the extremes of age, i.e., infants, young children, and the elderly. To date, RSV vaccine development has been unsuccessful, a feature linked to the lack of biomarkers available to assess the safety and efficacy of RSV vaccine candidates. We examined microRNAs (miR) as potential biomarkers for different types of RSV vaccine candidates. In this study, mice were vaccinated with a live attenuated RSV candidate that lacks the small hydrophobic (SH) and attachment (G) proteins (CP52), an RSV G protein microparticle (GA2-MP) vaccine, a formalin-inactivated RSV (FI-RSV) vaccine or were mock-treated. Several immunological endpoints and miR expression profiles were determined in mouse serum and bronchoalveolar lavage (BAL) following vaccine priming, boost, and RSV challenge. We identified miRs that were linked with immunological parameters of disease and protection. We show that miRs are potential biomarkers providing valuable insights for vaccine development.
Collapse
Affiliation(s)
- Lydia J Atherton
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Patricia A Jorquera
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Abhijeet A Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
33
|
Finlay BB, Pettersson S, Melby MK, Bosch TCG. The Microbiome Mediates Environmental Effects on Aging. Bioessays 2019; 41:e1800257. [PMID: 31157928 DOI: 10.1002/bies.201800257] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/26/2019] [Indexed: 12/14/2022]
Abstract
Humans' indigenous microbes strongly influence organ functions in an age- and diet-dependent manner, adding an important dimension to aging biology that remains poorly understood. Although age-related differences in the gut microbiota composition correlate with age-related loss of organ function and diseases, including inflammation and frailty, variation exists among the elderly, especially centenarians and people living in areas of extreme longevity. Studies using short-lived as well as nonsenescent model organisms provide surprising functional insights into factors affecting aging and implicate attenuating effects of microbes as well as a crucial role for certain transcription factors like forkhead box O. The unexpected beneficial effects of microbes on aged animals imply an even more complex interplay between the gut microbiome and the host. The microbiome constitutes the major interface between humans and the environment, is influenced by biosocial stressors and behaviors, and mediates effects on health and aging processes, while being moderated by sex and developmental stages.
Collapse
Affiliation(s)
- Brett B Finlay
- Canadian Institute for Advanced Research (CIFAR), MaRS Centre, West Tower, 661 University Avenue, Suite 505, Toronto, M5G 1M1, ON, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Sven Pettersson
- Canadian Institute for Advanced Research (CIFAR), MaRS Centre, West Tower, 661 University Avenue, Suite 505, Toronto, M5G 1M1, ON, Canada.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 639798, Singapore.,Department of Immunology, Weizmann Institute of Science, 7610001, Rehovot, Israel.,Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Melissa K Melby
- Canadian Institute for Advanced Research (CIFAR), MaRS Centre, West Tower, 661 University Avenue, Suite 505, Toronto, M5G 1M1, ON, Canada.,Department of Anthropology, College of Arts and Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Thomas C G Bosch
- Canadian Institute for Advanced Research (CIFAR), MaRS Centre, West Tower, 661 University Avenue, Suite 505, Toronto, M5G 1M1, ON, Canada.,Zoological Institute, University of Kiel, Kiel, 24118, Germany
| |
Collapse
|
34
|
Benayoun BA, Pollina EA, Singh PP, Mahmoudi S, Harel I, Casey KM, Dulken BW, Kundaje A, Brunet A. Remodeling of epigenome and transcriptome landscapes with aging in mice reveals widespread induction of inflammatory responses. Genome Res 2019; 29:697-709. [PMID: 30858345 PMCID: PMC6442391 DOI: 10.1101/gr.240093.118] [Citation(s) in RCA: 205] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 01/25/2019] [Indexed: 12/20/2022]
Abstract
Aging is accompanied by the functional decline of tissues. However, a systematic study of epigenomic and transcriptomic changes across tissues during aging is missing. Here, we generated chromatin maps and transcriptomes from four tissues and one cell type from young, middle-aged, and old mice—yielding 143 high-quality data sets. We focused on chromatin marks linked to gene expression regulation and cell identity: histone H3 trimethylation at lysine 4 (H3K4me3), a mark enriched at promoters, and histone H3 acetylation at lysine 27 (H3K27ac), a mark enriched at active enhancers. Epigenomic and transcriptomic landscapes could easily distinguish between ages, and machine-learning analysis showed that specific epigenomic states could predict transcriptional changes during aging. Analysis of data sets from all tissues identified recurrent age-related chromatin and transcriptional changes in key processes, including the up-regulation of immune system response pathways such as the interferon response. The up-regulation of the interferon response pathway with age was accompanied by increased transcription and chromatin remodeling at specific endogenous retroviral sequences. Pathways misregulated during mouse aging across tissues, notably innate immune pathways, were also misregulated with aging in other vertebrate species—African turquoise killifish, rat, and humans—indicating common signatures of age across species. To date, our data set represents the largest multitissue epigenomic and transcriptomic data set for vertebrate aging. This resource identifies chromatin and transcriptional states that are characteristic of young tissues, which could be leveraged to restore aspects of youthful functionality to old tissues.
Collapse
Affiliation(s)
- Bérénice A Benayoun
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Elizabeth A Pollina
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Param Priya Singh
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Salah Mahmoudi
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Itamar Harel
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Kerriann M Casey
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Ben W Dulken
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA.,Department of Computer Science, Stanford University, Stanford, California 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA.,Paul F. Glenn Laboratories for the Biology of Aging, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
35
|
Plasma cathelicidin and longitudinal lung function in current and former smokers. PLoS One 2019; 14:e0212628. [PMID: 30811465 PMCID: PMC6392327 DOI: 10.1371/journal.pone.0212628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/06/2019] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Cathelicidin (also known as LL-37 in humans) is an antimicrobial peptide secreted by epithelial and immune cells and regulated by vitamin D. The immunological roles of cathelicidin make it a putative biomarker to identify individuals at risk for reduced lung function. The objective of this study is to determine potential independent associations between low plasma cathelicidin and longitudinal lung function in current or former smokers without COPD. METHODS In a nested analysis of 308 participants from an observational cohort study, plasma cathelicidin and serum 25-hydroxy-vitamin D measurements were obtained at baseline, years three and five. The independent association between lowest quartile cathelicidin (<35 ng/ml) and forced-expiratory-volume-in-1-second (FEV1) at baseline, six and 18 months from each cathelicidin measurement was assessed with generalized estimating equations after adjusting for age, sex, race, smoking status and intensity. The long-term stability of cathelicidin and relationship with vitamin D was evaluated. RESULTS The cohort was 91% African-American, mean age 48.6 years, 32% female, and 81% current smokers. Participants with low cathelicidin were more likely to be female and have lower FEV1. Low cathelicidin was not independently associated with baseline FEV1. There was an independent association between low cathelicidin and reduced FEV1 at six months [-72 ml (95% CI, -140 to -8ml); p = 0.027] and 18 months [-103 ml (95% CI, -180 to -27 ml); p = 0.007]. Cathelicidin was stable over time and not correlated with vitamin D level. CONCLUSION In current and former smokers with preserved lung function, low cathelicidin is associated with sustained lung function reductions at six and 18 months, suggesting that cathelicidin may be an informative biomarker to predict persistent lung function disparities among at-risk individuals.
Collapse
|
36
|
Zhang J, Yang W, Xu J, Yang W, Li Q, Zhong Y, Cao Y, Yu XQ, Deng X. Regulation of antimicrobial peptide genes via insulin-like signaling pathway in the silkworm Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 103:12-21. [PMID: 30321587 DOI: 10.1016/j.ibmb.2018.10.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 09/29/2018] [Accepted: 10/11/2018] [Indexed: 06/08/2023]
Abstract
Antimicrobial peptides (AMPs) are important effector molecules of insect humoral immunity, and expression of AMPs is mainly regulated by the Toll and immune deficiency (IMD) pathways. FoxO, a key downstream regulator of the insulin-like signaling (ILS) pathway, has been recently reported to be involved in the regulation of AMPs in Drosophila melanogaster. In the present study, we investigated AMP gene expression and the regulation pathway controlled by the starvation in the silkworm Bombyx mori. We discovered that antibacterial activity in the hemolymph of B. mori larvae was increased by starvation, and expression of AMP genes (BmCecB6, BmAtta1, BmLeb3 and BmDefB) as well as the ILS target genes (FoxO, InR and Brummer) were strongly activated in the fat body by starvation. Moreover, phosphorylation of Akt kinase was reduced in the Bm-12 cells after starvation, suggesting that the ILS pathway was inhibited by starvation. We then showed that more FoxO protein was present in the cytoplasm than in the nucleus of Bm-12 cells under normal conditions, but more FoxO was detected in the nucleus after cells were starved for 8 h, indicating that FoxO was activated by starvation. In summary, our results indicated that starvation can activate AMP gene expression in B. mori via the ILS/FoxO signaling pathway.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China; Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, 5007 Rockhill Road, Kansas City, MO, 64110, USA
| | - Weike Yang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Junfeng Xu
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Wanying Yang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qingrong Li
- The Sericultural and Agri-Food Research Institute of the Guangdong Academy of Agricultural Sciences, Guangzhou, 510610, China
| | - Yangjin Zhong
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Yang Cao
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| | - Xiao-Qiang Yu
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, 5007 Rockhill Road, Kansas City, MO, 64110, USA.
| | - Xiaojuan Deng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding/Guangdong Provincial Sericulture and Mulberry Engineering Research Center, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
37
|
Mortzfeld BM, Taubenheim J, Fraune S, Klimovich AV, Bosch TCG. Stem Cell Transcription Factor FoxO Controls Microbiome Resilience in Hydra. Front Microbiol 2018; 9:629. [PMID: 29666616 PMCID: PMC5891625 DOI: 10.3389/fmicb.2018.00629] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/19/2018] [Indexed: 01/18/2023] Open
Abstract
The aging process is considered to be the result of accumulating cellular deterioration in an individual organism over time. It can be affected by the combined influence of genetic, epigenetic, and environmental factors including life-style-associated events. In the non-senescent freshwater polyp Hydra, one of the classical model systems for evolutionary developmental biology and regeneration, transcription factor FoxO modulates both stem cell proliferation and innate immunity. This provides strong support for the role of FoxO as a critical rate-of-aging regulator. However, how environmental factors interact with FoxO remains unknown. Here, we find that deficiency in FoxO signaling in Hydra leads to dysregulation of antimicrobial peptide expression and that FoxO loss-of-function polyps are impaired in selection for bacteria resembling the native microbiome and more susceptible to colonization of foreign bacteria. These findings reveal a key role of FoxO signaling in the communication between host and microbiota and embed the evolutionary conserved longevity factor FoxO into the holobiont concept.
Collapse
Affiliation(s)
| | - Jan Taubenheim
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Sebastian Fraune
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | | | - Thomas C G Bosch
- Zoological Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
38
|
Yu Y, Guo M, Wei Y, Yu S, Li H, Wang Y, Xu X, Cui Y, Tian J, Liang L, Peng K, Liu T. FoxO3a confers cetuximab resistance in RAS wild-type metastatic colorectal cancer through c-Myc. Oncotarget 2018; 7:80888-80900. [PMID: 27825133 PMCID: PMC5348362 DOI: 10.18632/oncotarget.13105] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 10/14/2016] [Indexed: 12/26/2022] Open
Abstract
Resistance to epidermal growth factor receptor (EGFR) targeted monoclonal antibody therapy represents a clinical challenge in patients suffered from RAS wild-type (WT) metastatic colorectal cancer (mCRC). However, the molecular mechanisms and key factors conferring this resistance are largely unknown. Forkhead transcription factors of the O class 3a (FoxO3a), an important regulator of cell survival, has been reported with dual functions in tumor recently. In this study, we found that FoxO3a was highly expressed in cetuximab resistant CRC tissues compared with cetuximab sensitive tissues. We therefore further analyzed its function in induced cetuximab resistant RAS-WT CRC cells (Caco2-CR) and intrinsic resistant cells with BRAF mutation (HT29). We found that FoxO3a was significantly up-regulated in Caco2-CR as well as in cetuximab treated HT29 cells. Knockdown of FoxO3a could sensitize these cells to cetuximab treatment with reduced cell proliferation and migration ability. Further, biochemical experiments demonstrated that FoxO3a directly bind to c-Myc promoter and activated the transcription of the c-Myc gene, thus participated in regulating of c-Myc downstream genes, including ACO2, LARS2, MRPL12 and PKM2 in these resistant cells. Moreover, knockdown of c-Myc elevated cell apoptosis to cetuximab treatment and suppressed cell proliferation and migration ability consistently. Altogether, our study indicates that FoxO3a might be a key regulator in cetuximab resistance through up-regulating c-Myc in colorectal cancer targeted therapy.
Collapse
Affiliation(s)
- Yiyi Yu
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Mengzhou Guo
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Ye Wei
- Department of General Surgery, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Hong Li
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Xiaojing Xu
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Yuehong Cui
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Jiawen Tian
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Li Liang
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Ke Peng
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhong Shan Hospital, Fu Dan University, Shanghai, China
| |
Collapse
|
39
|
A Complex Relationship between Immunity and Metabolism in Drosophila Diet-Induced Insulin Resistance. Mol Cell Biol 2017; 38:MCB.00259-17. [PMID: 29084810 DOI: 10.1128/mcb.00259-17] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/18/2017] [Indexed: 12/31/2022] Open
Abstract
Both systemic insulin resistance and tissue-specific insulin resistance have been described in Drosophila and are accompanied by many indicators of metabolic disease. The downstream mediators of insulin-resistant pathophysiology remain unclear. We analyzed insulin signaling in the fat body studying loss and gain of function. When expression of the sole Drosophila insulin receptor (InR) was reduced in larval fat bodies, animals exhibited developmental delay and reduced size in a diet-dependent manner. Fat body InR knockdown also led to reduced survival on high-sugar diets. To look downstream of InR at potential mediators of insulin resistance, transcriptome sequencing (RNA-seq) studies in insulin-resistant fat bodies revealed differential expression of genes, including those involved in innate immunity. Obesity-associated insulin resistance led to increased susceptibility of flies to infection, as in humans. Reduced innate immunity was dependent on fat body InR expression. The peptidoglycan recognition proteins (PGRPs) PGRP-SB2 and PGRP-SC2 were selected for further study based on differential expression studies. Downregulating PGRP-SB2 selectively in the fat body protected animals from the deleterious effects of overnutrition, whereas downregulating PGRP-SC2 produced InR-like phenotypes. These studies extend earlier work linking the immune and insulin signaling pathways and identify new targets of insulin signaling that could serve as potential drug targets to treat type 2 diabetes.
Collapse
|
40
|
Xiao Q, Ye Q, Wang W, Xiao J, Fu B, Xia Z, Zhang X, Liu Z, Zeng X. Mild hypothermia pretreatment protects against liver ischemia reperfusion injury via the PI3K/AKT/FOXO3a pathway. Mol Med Rep 2017; 16:7520-7526. [PMID: 28944825 PMCID: PMC5865885 DOI: 10.3892/mmr.2017.7501] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Mild hypothermia is known to protect against ischemia and reperfusion (IR) injury. The exact mechanisms of the protection are not fully understood. Forkhead box O3 (FOXO3a) has been defined as a critical mediator in cellular processes, including oxidative stress, apoptosis, inflammation, cell death and DNA repair; however, the protection function in mild hypothermia has not been reported previously. The current study was designed to investigate the function of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/FOXO3a pathway in pretreatment with mild hypothermia during IR injury. Additionally, PI3K/AKT/FOXO3a signaling was inhibited using Ly294002 and the effect on the protective function of mild hypothermia pretreatment was evaluated. Furthermore, the apoptotic and inflammatory response induced by the IR injury was evaluated. Liver IR injury induced a significant increase in the level of apoptosis and inflammatory responses. However, pretreatment with mild hypothermia increased phospho (p)-AKT and p-FOXO3a following IR injury, and significantly reduced apoptosis and inflammatory cytokines release. However, inhibiting p-AKT and p-FOXO3a using Ly294002 suppressed the liver protection produced by mild hypothermia. In conclusion, these findings indicated that mild hypothermia pretreatment exhibited liver protective effects against IR injury associated with suppressing inflammatory cytokine release and apoptosis via the PI3K/AKT/FOXO3a pathway.
Collapse
Affiliation(s)
- Qi Xiao
- Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Central South University, Changsha, Hunan 410013, P.R. China
| | - Qifa Ye
- Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Wang
- Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jiansheng Xiao
- Department of Transplant Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Biqi Fu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhiping Xia
- Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xingjian Zhang
- Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhongzhong Liu
- Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xianpeng Zeng
- Hubei Key Laboratory of Medical Technology on Transplantation, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
41
|
Mortzfeld BM, Bosch TCG. Eco-Aging: stem cells and microbes are controlled by aging antagonist FoxO. Curr Opin Microbiol 2017; 38:181-187. [DOI: 10.1016/j.mib.2017.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 01/10/2023]
|
42
|
Oteiza A, Mechti N. FoxO4 negatively controls Tat-mediated HIV-1 transcription through the post-transcriptional suppression of Tat encoding mRNA. J Gen Virol 2017; 98:1864-1878. [PMID: 28699853 DOI: 10.1099/jgv.0.000837] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The connection between the repression of human immunodeficiency virus type 1(HIV-1) transcription and the resting CD4+ T cell state suggests that the host transcription factors involved in the active maintenance of lymphocyte quiescence are likely to repress the viral transactivator, Tat, thereby restricting HIV-1 transcription. In this study, we analysed the interplay between Tat and the forkhead box transcription factors, FoxO1 and FoxO4. We show that FoxO1 and FoxO4 antagonize Tat-mediated transactivation of HIV-1 promoter through the repression of Tat protein expression. No effect was observed on the expression of two HIV-1 accessory proteins, Vif and Vpr. Unexpectedly, we found that FoxO1 and FoxO4 expression causes a strong dose-dependent post-transcriptional suppression of Tat mRNA, indicating that FoxO should effectively inhibit HIV-1 replication by destabilizing Tat mRNA and suppressing Tat-mediated HIV-1 transcription. In accordance with this, we observed that the Tat mRNA half-life is reduced by FoxO4 expression. The physiological relevance of our findings was validated using the J-Lat 10.6 model of latently infected cells. We demonstrated that the overexpression of a constitutively active FoxO4-TM mutant antagonized HIV-1 transcription reactivation in response to T cell activators, such as TNF-α or PMA. Altogether, our findings demonstrate that FoxO factors can control HIV-1 transcription and provide new insights into their potential role during the establishment of HIV-1 latency.
Collapse
Affiliation(s)
- Alexandra Oteiza
- CNRS UMR5235, DIMNP, Université de Montpellier, Bat 24, CC107, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Nadir Mechti
- CNRS UMR5235, DIMNP, Université de Montpellier, Bat 24, CC107, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| |
Collapse
|
43
|
Link W, Fernandez-Marcos PJ. FOXO transcription factors at the interface of metabolism and cancer. Int J Cancer 2017. [PMID: 28631330 DOI: 10.1002/ijc.30840] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetes refers to a group of metabolic diseases characterized by impaired insulin signalling and high blood glucose. A growing body of epidemiological evidence links diabetes to several types of cancer but the underlying molecular mechanisms are poorly understood. The signalling cascade connecting insulin and FOXO proteins provides a compelling example for a conserved pathway at the interface between insulin signalling and cancer. FOXOs are transcription factors that orchestrate programs of gene expression known to control a variety of processes in response to cellular stress. Genes regulated by this family of proteins are involved in the regulation of cellular energy production, oxidative stress resistance and cell viability and proliferation. Accordingly, FOXO factors have been shown to play an important role in the suppression of tumour growth and in the regulation of metabolic homeostasis. There is emerging evidence that deregulation of FOXO factors might account for the association between insulin resistance-related metabolic disorders and cancer.
Collapse
Affiliation(s)
- Wolfgang Link
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, room 2.22, Faro, 8005-139, Portugal.,Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Campus de Gambelas, Faro, 8005-139, Portugal.,Algarve Biomedical Center (ABC), University of Algarve, Campus de Gambelas, Faro, 8005-139, Portugal
| | | |
Collapse
|
44
|
Polke M, Seiler F, Lepper PM, Kamyschnikow A, Langer F, Monz D, Herr C, Bals R, Beisswenger C. Hypoxia and the hypoxia-regulated transcription factor HIF-1α suppress the host defence of airway epithelial cells. Innate Immun 2017; 23:373-380. [PMID: 28409544 DOI: 10.1177/1753425917698032] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Chronic diseases of the respiratory tract, such as cystic fibrosis, are associated with mucosal and systemic hypoxia. Innate immune functions of airway epithelial cells are required to prevent and control infections of the lung parenchyma. The transcription factor hypoxia-inducible factor 1α (HIF-1α) regulates cellular adaptation to low oxygen conditions. Here, we show that hypoxia and HIF-1α regulate innate immune mechanisms of cultured human bronchial epithelial cells (HBECs). Exposure of primary HBECs to hypoxia or the prolyl hydroxylase inhibitor dimethyloxaloylglycine (DMOG) resulted in a significantly decreased expression of inflammatory mediators (IL-6, IFN-γ-induced protein 10) in response to ligands for TLRs (flagellin, polyI:C) and Pseudomonas aeruginosa, whereas the expression of inflammatory mediators was not affected by hypoxia or DMOG in the absence of microbial factors. Small interfering RNA-mediated knockdown of HIF-1α in HBECs and in the bronchial epithelial cell line Calu-3 resulted in increased expression of inflammatory mediators. The inflammatory response was decreased in lungs of mice stimulated with inactivated P. aeruginosa under hypoxia. These data suggest that hypoxia suppresses the innate immune response of airway epithelial cells via HIF-1α.
Collapse
Affiliation(s)
- Markus Polke
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frederik Seiler
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Philipp M Lepper
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Andreas Kamyschnikow
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frank Langer
- 2 Department of Thoracic and Cardiovascular Surgery, Saarland University, Homburg, Germany
| | - Dominik Monz
- 3 Department of Pediatrics and Neonatology, Saarland University, Homburg, Germany
| | - Christian Herr
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Robert Bals
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christoph Beisswenger
- 1 Department of Internal Medicine V - Pulmonology, Allergology and Critical Care Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
45
|
FoxO1 regulates allergic asthmatic inflammation through regulating polarization of the macrophage inflammatory phenotype. Oncotarget 2017; 7:17532-46. [PMID: 27007158 PMCID: PMC4951231 DOI: 10.18632/oncotarget.8162] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 03/01/2016] [Indexed: 01/18/2023] Open
Abstract
Inflammatory monocyte and tissue macrophages influence the initiation, progression, and resolution of type 2 immune responses, and alveolar macrophages are the most prevalent immune-effector cells in the lung. While we were characterizing the M1- or M2-like macrophages in type 2 allergic inflammation, we discovered that FoxO1 is highly expressed in alternatively activated macrophages. Although several studies have been focused on the fundamental role of FoxOs in hematopoietic and immune cells, the exact role that FoxO1 plays in allergic asthmatic inflammation in activated macrophages has not been investigated. Growing evidences indicate that FoxO1 acts as an upstream regulator of IRF4 and could have a role in a specific inflammatory phenotype of macrophages. Therefore, we hypothesized that IRF4 expression regulated by FoxO1 in alveolar macrophages is required for established type 2 immune mediates allergic lung inflammation. Our data indicate that targeted deletion of FoxO1 using FoxO1-selective inhibitor AS1842856 and genetic ablation of FoxO1 in macrophages significantly decreases IRF4 and various M2 macrophage-associated genes, suggesting a mechanism that involves FoxO1-IRF4 signaling in alveolar macrophages that works to polarize macrophages toward established type 2 immune responses. In response to the challenge of DRA (dust mite, ragweed, and Aspergillus) allergens, macrophage specific FoxO1 overexpression is associated with an accentuation of asthmatic lung inflammation, whereas pharmacologic inhibition of FoxO1 by AS1842856 attenuates the development of asthmatic lung inflammation. Thus, our study identifies a role for FoxO1-IRF4 signaling in the development of alternatively activated alveolar macrophages that contribute to type 2 allergic airway inflammation.
Collapse
|
46
|
|
47
|
Lu Y, Zhu Y, Wang X, Wang F, Peng J, Hou H, Sun Z. FOXO3 rs12212067: T > G Association with Active Tuberculosis in Han Chinese Population. Inflammation 2016. [PMID: 26223437 DOI: 10.1007/s10753-015-0217-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well known that the human innate immune and adaptive immune response play important role in tuberculosis (TB) infection and progress. Emerging evidence shows that FOXO3 plays an important role in the human immune system. Recent research has shown that the FOXO3 genetic variants are associated malaria infection. In this study, 268 confirmed TB patients, 321 patients with latent tuberculosis infection (LTBI), and 475 TB-free controls were recruited; the single-nucleotide polymorphism (SNP) rs12212067: T > G in FOXO3 was genotyped using predesigned TaqMan® allelic discrimination assays. The results showed that the G allele of rs12212067 in FOXO3 was more common in health control and the latent TB group compared with the active TB group (p = 0.048, odds ratio (OR) 95 % confidence intervals (CI) = 1.37 (1.00-1.89); p = 0.042, OR 95 % CI = 1.42 (1.01-1.99), respectively); furthermore, within active TB patients, the G allele of rs12212067 in FOXO3 was more frequent in extra-pulmonary tuberculosis (EPTB) group compared to pulmonary tuberculosis (PTB) group (p = 0.035, OR 95 % CI = 0.57 (0.33-0.97). In conclusion, this study found that rs12212067 in FOXO3 was associated with increased risk of active TB. The minor G allele might be a protection factor which was found more common in latent TB patients and healthy controls than active TB patients.
Collapse
Affiliation(s)
- Yanjun Lu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Yaowu Zhu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Feng Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Jing Peng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Hongyan Hou
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China
| | - Ziyong Sun
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095# Jiefangdadao, Wuhan, 430030, China.
| |
Collapse
|
48
|
Gupta P, Srivastav S, Saha S, Das PK, Ukil A. Leishmania donovani inhibits macrophage apoptosis and pro-inflammatory response through AKT-mediated regulation of β-catenin and FOXO-1. Cell Death Differ 2016; 23:1815-1826. [PMID: 27662364 DOI: 10.1038/cdd.2016.101] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/18/2016] [Accepted: 08/17/2016] [Indexed: 12/25/2022] Open
Abstract
In order to establish infection, intra-macrophage parasite Leishmania donovani needs to inhibit host defense parameters like inflammatory cytokine production and apoptosis. In the present study, we demonstrate that the parasite achieves both by exploiting a single host regulator AKT for modulating its downstream transcription factors, β-catenin and FOXO-1. L. donovani-infected RAW264.7 and bone marrow-derived macrophages (BMDM) treated with AKT inhibitor or dominant negative AKT constructs showed decreased anti-inflammatory cytokine production and increased host cell apoptosis resulting in reduced parasite survival. Infection-induced activated AKT triggered phosphorylation-mediated deactivation of its downstream target, GSK-3β. Inactivated GSK-3β, in turn, could no longer sequester cytosolic β-catenin, an anti-apoptotic transcriptional regulator, as evidenced from its nuclear translocation during infection. Constitutively active GSK-3β-transfected L. donovani-infected cells mimicked the effects of AKT inhibition and siRNA-mediated silencing of β-catenin led to disruption of mitochondrial potential along with increased caspase-3 activity and IL-12 production leading to decreased parasite survival. In addition to activating anti-apoptotic β-catenin, phospho-AKT inhibits activation of FOXO-1, a pro-apoptotic transcriptional regulator. Nuclear retention of FOXO-1, inhibited during infection, was reversed when infected cells were transfected with dominant negative AKT constructs. Overexpression of FOXO-1 in infected macrophages not only documented increased apoptosis but promoted enhanced TLR4 expression and NF-κB activity along with an increase in IL-1β and decrease in IL-10 secretion. In vivo administration of AKT inhibitor significantly decreased liver and spleen parasite burden and switched cytokine balance in favor of host. In contrast, GSK-3β inhibitor did not result in any significant change in infectivity parameters. Collectively our findings revealed that L. donovani triggered AKT activation to regulate GSK-3β/β-catenin/FOXO-1 axis, thus ensuring inhibition of both host cell apoptosis and immune response essential for its intra-macrophage survival.
Collapse
Affiliation(s)
- Purnima Gupta
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Supriya Srivastav
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shriya Saha
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| | - Pijush K Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Anindita Ukil
- Department of Biochemistry, University of Calcutta, Kolkata 700019, India
| |
Collapse
|
49
|
Inhibition of ROS and upregulation of inflammatory cytokines by FoxO3a promotes survival against Salmonella typhimurium. Nat Commun 2016; 7:12748. [PMID: 27599659 PMCID: PMC5023958 DOI: 10.1038/ncomms12748] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Virulent intracellular pathogens, such as the Salmonella species, engage numerous virulence factors to subvert host defence mechanisms to induce a chronic infection that leads to typhoid or exacerbation of other chronic inflammatory conditions. Here we show the role of the forkhead transcription factor FoxO3a during infection of mice with Salmonella typhimurium (ST). Although FoxO3a signalling does not affect the development of CD8+ T cell responses to ST, FoxO3a has an important protective role, particularly during the chronic stage of infection, by limiting the persistence of oxidative stress. Furthermore, FoxO3a signalling regulates ERK signalling in macrophages, which results in the maintenance of a proinflammatory state. FoxO3a signalling does not affect cell proliferation or cell death. Thus, these results reveal mechanisms by which FoxO3a promotes host survival during infection with chronic, virulent intracellular bacteria. FoxO3a signalling has limited influence over acute bacterial infection. Here the authors show that FoxO3a promotes survival of mice in response to chronic Salmonella typhimurium infection by restraining oxidative stress and ERK signalling.
Collapse
|
50
|
McLaughlin CN, Nechipurenko IV, Liu N, Broihier HT. A Toll receptor-FoxO pathway represses Pavarotti/MKLP1 to promote microtubule dynamics in motoneurons. J Cell Biol 2016; 214:459-74. [PMID: 27502486 PMCID: PMC4987293 DOI: 10.1083/jcb.201601014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 07/07/2016] [Indexed: 02/06/2023] Open
Abstract
McLaughlin et al. uncover a motoneuronal Toll-6–directed pathway that functions via dSARM and FoxO to attenuate microtubule stability through repression of Pavarotti/MKLP1 transcription. Genetic and pharmacological strategies reveal a novel requirement for dynamic synaptic microtubules in structural plasticity, which are established by Toll-6–FoxO signaling. FoxO proteins are evolutionarily conserved regulators of neuronal structure and function, yet the neuron-specific pathways within which they act are poorly understood. To elucidate neuronal FoxO function in Drosophila melanogaster, we first screened for FoxO’s upstream regulators and downstream effectors. On the upstream side, we present genetic and molecular pathway analyses indicating that the Toll-6 receptor, the Toll/interleukin-1 receptor domain adaptor dSARM, and FoxO function in a linear pathway. On the downstream side, we find that Toll-6–FoxO signaling represses the mitotic kinesin Pavarotti/MKLP1 (Pav-KLP), which itself attenuates microtubule (MT) dynamics. We next probed in vivo functions for this novel pathway and found that it is essential for axon transport and structural plasticity in motoneurons. We demonstrate that elevated expression of Pav-KLP underlies transport and plasticity phenotypes in pathway mutants, indicating that Toll-6–FoxO signaling promotes MT dynamics by limiting Pav-KLP expression. In addition to uncovering a novel molecular pathway, our work reveals an unexpected function for dynamic MTs in enabling rapid activity-dependent structural plasticity.
Collapse
Affiliation(s)
- Colleen N McLaughlin
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Inna V Nechipurenko
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Nan Liu
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| | - Heather T Broihier
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|