1
|
Shi S, Jiang H, Ma W, Guan Z, Han M, Man S, Wu Z, He S. Preclinical studies of natural flavonoids in inflammatory bowel disease based on macrophages: a systematic review with meta-analysis and network pharmacology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03501-0. [PMID: 39422746 DOI: 10.1007/s00210-024-03501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Flavonoid is a category of bioactive polyphenolic compounds that are extensively distributed in plants with specific pharmacological properties, such as anti-inflammatory and anti-oxidant. Importantly, natural flavonoids have shown the protected function on the dextran sulfate sodium (DSS)-induced colitis in animals and lipopolysaccharides (LPS)-induced inflammatory response in macrophages. The purpose of this systematic review is to explore the efficacy of natural flavonoids in animal models of IBD (inflammatory bowel disease) and potential mechanisms in macrophages by meta-analysis and network pharmacology in preclinical studies. Relevant foundation studies were searched from January 2010 to November 2023 in databases like PubMed, Elsevier ScienceDirect, and Web of Science. Then, OriginPro software was used to extract values from images, and the analysis was performed using Review Manager 5.3. The retrieved data was analyzed according to the fixed-effects model and random-effects model. Subsequently, heterogeneity was evaluated using the I2 statistics. Lastly, network pharmacology was applied to confirm mechanisms of natural flavonoids on IBD. According to the results of meta-analysis, we found the natural flavonoids exhibited powerful therapeutic effects against IBD, which not only reversed colonic shortness (WMD = 1.33, 95% CI (1.07, 1.59), P < 0.00001), but also reduced histological score (SMD = - 2.66, 95% CI (- 3.77, - 1.95), P < 0.00001) between natural flavonoid treatment groups compared with the experimental IBD model. Furthermore, treatment with natural flavonoids decreased the levels of tumor necrosis factor-α (TNF-α) in macrophages. Mechanistically, our summarized data substantiate that natural flavonoids alleviate LPS-induced M1 macrophage polarization, anti-oxidant, anti-inflammatory, maintain intestinal barrier, and inhibit the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome in macrophages. Moreover, the results of network pharmacology also support this. This systematic review demonstrated the efficiency of natural flavonoids in treating IBD in preclinical research by meta-analysis and network pharmacology, which offered supporting evidence for clinical trial implementation. However, some limitations remain present, such as technique quality shortage, missed reports on account of negative results, failure to count sample size, and the risk of bias.
Collapse
Affiliation(s)
- Shasha Shi
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hao Jiang
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenke Ma
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zitong Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengxue Han
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhuzhu Wu
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Shan He
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Medina-Rodriguez EM, Han D, Zeltzer SE, Moraskie Alvarez-Tabío MP, O'Connor G, Daunert S, Beurel E. Stress-induced VIPergic activation mediates microbiota/Th17cell-dependent depressive-like behaviors. Brain Behav Immun 2024; 123:739-751. [PMID: 39419356 DOI: 10.1016/j.bbi.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/02/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic stress often has deleterious effects leading to the development of psychiatric diseases. The gut-brain axis represents a novel avenue for stress research. The negative effects of stress on the gut physiology have been well-described, whereas the pathways whereby stress controls microbial composition to modulate behaviors remains mainly unknown. We discovered that vasoactive intestinal peptide (VIP) activation promoted stress-induced microbial changes leading to increased infiltration of T helper (Th) 17 cells and microglial activation in the hippocampus and depressive-like behaviors, uncovering a close crosstalk between intestinal VIPergic release and the gut microbiota during stress and providing a new interaction between the nervous system and the gut microbiome after stress. Neutralization of the signature cytokine of Th17 cells, interleukin (IL)-17A, was sufficient to block depressive-like behaviors, reduce neuronal VIPergic activation and microglia activation induced by VIPergic activation after stress, opening new potential therapeutic targets for depression.
Collapse
Affiliation(s)
- Eva M Medina-Rodriguez
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Dongmei Han
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Shanie E Zeltzer
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Michael P Moraskie Alvarez-Tabío
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Gregory O'Connor
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, United States
| | - Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, United States; Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, United States.
| |
Collapse
|
3
|
Malovic E, Ealy A, Miller C, Jang A, Hsu PJ, Sarkar S, Rokad D, Goeser C, Hartman AK, Zhu A, Palanisamy B, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, He C, Kanthasamy AG. Epitranscriptomic reader YTHDF2 regulates SEK1( MAP2K4)-JNK-cJUN inflammatory signaling in astrocytes during neurotoxic stress. iScience 2024; 27:110619. [PMID: 39252959 PMCID: PMC11382029 DOI: 10.1016/j.isci.2024.110619] [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: 02/12/2024] [Revised: 06/13/2024] [Accepted: 07/26/2024] [Indexed: 09/11/2024] Open
Abstract
As the most abundant glial cells in the central nervous system (CNS), astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress are many and complex. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stressor, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, the neurotoxic stress-induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechanistically, YTHDF2 RIP-sequencing identified MAP2K4 (MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed that Mn-exposed astrocytes mediate proinflammatory responses by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves as a key upstream 'molecular switch' controlling SEK1(MAP2K4)-JNK-cJUN proinflammatory signaling in astrocytes.
Collapse
Affiliation(s)
- Emir Malovic
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Alyssa Ealy
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Cameron Miller
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Ahyoung Jang
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Phillip J Hsu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Souvarish Sarkar
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Dharmin Rokad
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Cody Goeser
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Aleah Kristen Hartman
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Allen Zhu
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Bharathi Palanisamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Arthi Kanthasamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, IL, USA
| | - Anumantha G Kanthasamy
- Parkinson's Disorder Research Laboratory, Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- Isakson Center for Neurological Disease Research, University of Georgia, Athens, GA, USA
- Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| |
Collapse
|
4
|
Azargoonjahromi A. The duality of amyloid-β: its role in normal and Alzheimer's disease states. Mol Brain 2024; 17:44. [PMID: 39020435 PMCID: PMC11256416 DOI: 10.1186/s13041-024-01118-1] [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: 04/29/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024] Open
Abstract
Alzheimer's disease (AD) is a degenerative neurological condition that gradually impairs cognitive abilities, disrupts memory retention, and impedes daily functioning by impacting the cells of the brain. A key characteristic of AD is the accumulation of amyloid-beta (Aβ) plaques, which play pivotal roles in disease progression. These plaques initiate a cascade of events including neuroinflammation, synaptic dysfunction, tau pathology, oxidative stress, impaired protein clearance, mitochondrial dysfunction, and disrupted calcium homeostasis. Aβ accumulation is also closely associated with other hallmark features of AD, underscoring its significance. Aβ is generated through cleavage of the amyloid precursor protein (APP) and plays a dual role depending on its processing pathway. The non-amyloidogenic pathway reduces Aβ production and has neuroprotective and anti-inflammatory effects, whereas the amyloidogenic pathway leads to the production of Aβ peptides, including Aβ40 and Aβ42, which contribute to neurodegeneration and toxic effects in AD. Understanding the multifaceted role of Aβ, particularly in AD, is crucial for developing effective therapeutic strategies that target Aβ metabolism, aggregation, and clearance with the aim of mitigating the detrimental consequences of the disease. This review aims to explore the mechanisms and functions of Aβ under normal and abnormal conditions, particularly in AD, by examining both its beneficial and detrimental effects.
Collapse
|
5
|
Abbasifard M, Khorramdelazad H. Harmonizing hope: navigating the osteoarthritis melody through the CCL2/CCR2 axis for innovative therapeutic avenues. Front Immunol 2024; 15:1387651. [PMID: 39076996 PMCID: PMC11284107 DOI: 10.3389/fimmu.2024.1387651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/31/2024] Open
Abstract
Osteoarthritis (OA) is characterized by a complex interplay of molecular signals orchestrated by the CCL2/CCR2 axis. The pathogenesis of OA has been revealed to be influenced by a multifaceted effect of CCL2/CCR2 signaling on inflammation, cartilage degradation, and joint homeostasis. The CCL2/CCR2 axis promotes immune cell recruitment and tips the balance toward degeneration by influencing chondrocyte behavior. Insights into these intricate pathways will offer novel therapeutic approaches, paving the way for targeted interventions that may redefine OA management in the future. This review article explores the molecular symphony through the lens of the CCL2/CCR2 axis, providing a harmonious blend of current knowledge and future directions on OA treatment. Furthermore, in this study, through a meticulous review of recent research, the key players and molecular mechanisms that amplify the catabolic cascade within the joint microenvironment are identified, and therapeutic approaches to targeting the CCL2/CCR axis are discussed.
Collapse
Affiliation(s)
- Mitra Abbasifard
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
6
|
Ball BK, Kuhn MK, Fleeman Bechtel RM, Proctor EA, Brubaker DK. Differential responses of primary neuron-secreted MCP-1 and IL-9 to type 2 diabetes and Alzheimer's disease-associated metabolites. Sci Rep 2024; 14:12743. [PMID: 38830911 PMCID: PMC11148169 DOI: 10.1038/s41598-024-62155-3] [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: 12/01/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Type 2 diabetes (T2D) is implicated as a risk factor for Alzheimer's disease (AD), the most common form of dementia. In this work, we investigated neuroinflammatory responses of primary neurons to potentially circulating, blood-brain barrier (BBB) permeable metabolites associated with AD, T2D, or both. We identified nine metabolites associated with protective or detrimental properties of AD and T2D in literature (lauric acid, asparagine, fructose, arachidonic acid, aminoadipic acid, sorbitol, retinol, tryptophan, niacinamide) and stimulated primary mouse neuron cultures with each metabolite before quantifying cytokine secretion via Luminex. We employed unsupervised clustering, inferential statistics, and partial least squares discriminant analysis to identify relationships between cytokine concentration and disease-associations of metabolites. We identified MCP-1, a cytokine associated with monocyte recruitment, as differentially abundant between neurons stimulated by metabolites associated with protective and detrimental properties of AD and T2D. We also identified IL-9, a cytokine that promotes mast cell growth, to be differentially associated with T2D. Indeed, cytokines, such as MCP-1 and IL-9, released from neurons in response to BBB-permeable metabolites associated with T2D may contribute to AD development by downstream effects of neuroinflammation.
Collapse
Affiliation(s)
- Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Madison K Kuhn
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
| | - Rebecca M Fleeman Bechtel
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA.
| |
Collapse
|
7
|
Tyumentseva A, Khilazheva E, Petrova V, Stolyar S. Effects of iron oxide nanoparticles on the gene expression profiles of cerebral endotheliocytes and astrocytes. Toxicol In Vitro 2024; 98:105829. [PMID: 38615722 DOI: 10.1016/j.tiv.2024.105829] [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: 12/04/2023] [Revised: 04/01/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Iron oxide nanoparticles (IONPs) are considered as the most biocompatible magnetic materials suitable for biomedical applications. Nevertheless, there are many evidences of their toxicity for living organisms and partially neurotoxicity. The central nervous system is protected from undesirable substances circulating in the bloodstream by the blood-brain barrier (BBB). And even if being small enough, some nanoparticles could be able to penetrate cell membranes in other cells but will often be delayed by the BBB cells. However, the neurotoxicity of iron oxide is described even in the cases when IONPs should not uptake to the nervous system by experimental design. The aim of this study was to investigate what molecular changes in the cells-components of BBB - endotheliocytes and underlying astrocytes - may be caused by IONPs in the blood vessels of the brain. For this, a two-layer in vitro BBB model was created, consisting of rat cerebral endothelial cells and astrocytes. It was revealed that 100 and 200 mg/L of the nanoparticles induce metabolism alteration in the cells under study. Using RNA-sequencing, the up-regulation of pro-inflammatory chemokines encoding genes and changes in the expression of genes associated with detoxification in the endotheliocytes were demonstrated under the influence of 100 mg/L IONPs.
Collapse
Affiliation(s)
- Anna Tyumentseva
- Federal Research Center, Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences, Akademgorodok st. 50, Krasnoyarsk 660036, Russia.
| | - Elena Khilazheva
- Krasnoyarsk State Medical University named after Professor V.F. Voino-Yasenetsky, Ministry of Health of the Russian Federation, 660022, Partizana Zheleznyaka St. 1, Krasnoyarsk, Russia
| | - Valeria Petrova
- Federal Research Center, Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences, Akademgorodok st. 50, Krasnoyarsk 660036, Russia
| | - Sergey Stolyar
- Federal Research Center, Krasnoyarsk Science Center of the Siberian Branch of the Russian Academy of Sciences, Akademgorodok st. 50, Krasnoyarsk 660036, Russia.
| |
Collapse
|
8
|
Kedlian VR, Wang Y, Liu T, Chen X, Bolt L, Tudor C, Shen Z, Fasouli ES, Prigmore E, Kleshchevnikov V, Pett JP, Li T, Lawrence JEG, Perera S, Prete M, Huang N, Guo Q, Zeng X, Yang L, Polański K, Chipampe NJ, Dabrowska M, Li X, Bayraktar OA, Patel M, Kumasaka N, Mahbubani KT, Xiang AP, Meyer KB, Saeb-Parsy K, Teichmann SA, Zhang H. Human skeletal muscle aging atlas. NATURE AGING 2024; 4:727-744. [PMID: 38622407 PMCID: PMC11108788 DOI: 10.1038/s43587-024-00613-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Skeletal muscle aging is a key contributor to age-related frailty and sarcopenia with substantial implications for global health. Here we profiled 90,902 single cells and 92,259 single nuclei from 17 donors to map the aging process in the adult human intercostal muscle, identifying cellular changes in each muscle compartment. We found that distinct subsets of muscle stem cells exhibit decreased ribosome biogenesis genes and increased CCL2 expression, causing different aging phenotypes. Our atlas also highlights an expansion of nuclei associated with the neuromuscular junction, which may reflect re-innervation, and outlines how the loss of fast-twitch myofibers is mitigated through regeneration and upregulation of fast-type markers in slow-twitch myofibers with age. Furthermore, we document the function of aging muscle microenvironment in immune cell attraction. Overall, we present a comprehensive human skeletal muscle aging resource ( https://www.muscleageingcellatlas.org/ ) together with an in-house mouse muscle atlas to study common features of muscle aging across species.
Collapse
Affiliation(s)
- Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Tianliang Liu
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoping Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Zhuojian Shen
- Department of Thoracic Surgery, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Eirini S Fasouli
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Jan Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John E G Lawrence
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Qin Guo
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinrui Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krzysztof Polański
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nana-Jane Chipampe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Dabrowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xiaobo Li
- Core Facilities for Medical Science, Sun Yat-sen University, Guangzhou, China
| | - Omer Ali Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Minal Patel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Natsuhiko Kumasaka
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Andy Peng Xiang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK.
- Collaborative Biorepository for Translational Medicine (CBTM), NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
- Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
9
|
Schäufele TJ, Kolbinger A, Friedel J, Gurke R, Geisslinger G, Weigert A, Pierre S, Scholich K. Meloxicam treatment disrupts the regional structure of innate inflammation sites by targeting the pro-inflammatory effects of prostanoids. Br J Pharmacol 2024; 181:1051-1067. [PMID: 37823675 DOI: 10.1111/bph.16261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/10/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Non-steroidal anti-inflammatory drugs (NSAIDs) are the most widely prescribed drugs in the world due to their analgesic, antipyretic and anti-inflammatory effects. However, NSAIDs inhibit prostanoid synthesis, interfering with their pro-inflammatory and anti-inflammatory functions and potentially prolonging acute inflammation. EXPERIMENTAL APPROACH We used high-content immunohistochemistry to define the impact of meloxicam treatment on spatially separated pro-inflammatory and anti-inflammatory processes during innate inflammation in mice induced by zymosan. This allowed us to determine the effect of meloxicam treatment on the organization of pro-inflammatory and anti-inflammatory microenvironments, thereby identifying relevant changes in immune cell localization, recruitment and activation. KEY RESULTS Meloxicam treatment reduced zymosan-induced thermal hypersensitivity at early time points but delayed its resolution. High-content immunohistochemistry revealed that the pro-inflammatory area was smaller after treatment, diminishing neutrophil recruitment, M1-like macrophage polarization, and especially phagocytosis by neutrophils and macrophages. The polarization of macrophages towards the M2-like anti-inflammatory phenotype was unaffected, and the number of anti-inflammatory eosinophils actually increased. CONCLUSION AND IMPLICATIONS High-content immunohistochemistry was able to identify relevant meloxicam-mediated effects on inflammatory processes based on alterations in the regional structure of inflammation sites. Meloxicam delayed the clearance of pathogens by inhibiting pro-inflammatory processes, causing prolonged inflammation. Our data suggest that the prescription of NSAIDs as a treatment during an acute pathogen-driven inflammation should be reconsidered in patients with compromised immune systems.
Collapse
Affiliation(s)
- Tim J Schäufele
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Anja Kolbinger
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Joschua Friedel
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe University Frankfurt, Frankfurt, Germany
| | - Sandra Pierre
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
| | - Klaus Scholich
- Institute of Clinical Pharmacology, Goethe University Frankfurt, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Frankfurt, Germany
| |
Collapse
|
10
|
Ciechanowska A, Mika J. CC Chemokine Family Members' Modulation as a Novel Approach for Treating Central Nervous System and Peripheral Nervous System Injury-A Review of Clinical and Experimental Findings. Int J Mol Sci 2024; 25:3788. [PMID: 38612597 PMCID: PMC11011591 DOI: 10.3390/ijms25073788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Despite significant progress in modern medicine and pharmacology, damage to the nervous system with various etiologies still poses a challenge to doctors and scientists. Injuries lead to neuroimmunological changes in the central nervous system (CNS), which may result in both secondary damage and the development of tactile and thermal hypersensitivity. In our review, based on the analysis of many experimental and clinical studies, we indicate that the mechanisms occurring both at the level of the brain after direct damage and at the level of the spinal cord after peripheral nerve damage have a common immunological basis. This suggests that there are opportunities for similar pharmacological therapeutic interventions in the damage of various etiologies. Experimental data indicate that after CNS/PNS damage, the levels of 16 among the 28 CC-family chemokines, i.e., CCL1, CCL2, CCL3, CCL4, CCL5, CCL6, CCL7, CCL8, CCL9, CCL11, CCL12, CCL17, CCL19, CCL20, CCL21, and CCL22, increase in the brain and/or spinal cord and have strong proinflammatory and/or pronociceptive effects. According to the available literature data, further investigation is still needed for understanding the role of the remaining chemokines, especially six of them which were found in humans but not in mice/rats, i.e., CCL13, CCL14, CCL15, CCL16, CCL18, and CCL23. Over the past several years, the results of studies in which available pharmacological tools were used indicated that blocking individual receptors, e.g., CCR1 (J113863 and BX513), CCR2 (RS504393, CCX872, INCB3344, and AZ889), CCR3 (SB328437), CCR4 (C021 and AZD-2098), and CCR5 (maraviroc, AZD-5672, and TAK-220), has beneficial effects after damage to both the CNS and PNS. Recently, experimental data have proved that blockades exerted by double antagonists CCR1/3 (UCB 35625) and CCR2/5 (cenicriviroc) have very good anti-inflammatory and antinociceptive effects. In addition, both single (J113863, RS504393, SB328437, C021, and maraviroc) and dual (cenicriviroc) chemokine receptor antagonists enhanced the analgesic effect of opioid drugs. This review will display the evidence that a multidirectional strategy based on the modulation of neuronal-glial-immune interactions can significantly improve the health of patients after CNS and PNS damage by changing the activity of chemokines belonging to the CC family. Moreover, in the case of pain, the combined administration of such antagonists with opioid drugs could reduce therapeutic doses and minimize the risk of complications.
Collapse
Affiliation(s)
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Str., 31-343 Kraków, Poland;
| |
Collapse
|
11
|
Chen X, Tang SJ. Neural Circuitry Polarization in the Spinal Dorsal Horn (SDH): A Novel Form of Dysregulated Circuitry Plasticity during Pain Pathogenesis. Cells 2024; 13:398. [PMID: 38474361 PMCID: PMC10930392 DOI: 10.3390/cells13050398] [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: 01/20/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Pathological pain emerges from nociceptive system dysfunction, resulting in heightened pain circuit activity. Various forms of circuitry plasticity, such as central sensitization, synaptic plasticity, homeostatic plasticity, and excitation/inhibition balance, contribute to the malfunction of neural circuits during pain pathogenesis. Recently, a new form of plasticity in the spinal dorsal horn (SDH), named neural circuit polarization (NCP), was discovered in pain models induced by HIV-1 gp120 and chronic morphine administration. NCP manifests as an increase in excitatory postsynaptic currents (EPSCs) in excitatory neurons and a decrease in EPSCs in inhibitory neurons, presumably facilitating hyperactivation of pain circuits. The expression of NCP is associated with astrogliosis. Ablation of reactive astrocytes or suppression of astrogliosis blocks NCP and, concomitantly, the development of gp120- or morphine-induced pain. In this review, we aim to compare and integrate NCP with other forms of plasticity in pain circuits to improve the understanding of the pathogenic contribution of NCP and its cooperation with other forms of circuitry plasticity during the development of pathological pain.
Collapse
Affiliation(s)
| | - Shao-Jun Tang
- Stony Brook University Pain and Anesthesia Research Center (SPARC), Department of Anesthesiology, Stony Brook University, Stony Brook, NY 11794, USA;
| |
Collapse
|
12
|
Hammer M, Krzyzaniak C, Bahramnejad E, Smelser K, Hack J, Watkins J, Ronaldson P. Sex differences in physiological response to increased neuronal excitability in a knockin mouse model of pediatric epilepsy. Clin Sci (Lond) 2024; 138:205-223. [PMID: 38348743 PMCID: PMC10881277 DOI: 10.1042/cs20231572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/01/2024] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Epilepsy is a common neurological disease; however, few if any of the currently marketed antiseizure medications prevent or cure epilepsy. Discovery of pathological processes in the early stages of epileptogenesis has been challenging given the common use of preclinical models that induce seizures in physiologically normal animals. Moreover, despite known sex dimorphism in neurological diseases, females are rarely included in preclinical epilepsy models. METHODS We characterized sex differences in mice carrying a pathogenic knockin variant (p.N1768D) in the Scn8a gene that causes spontaneous tonic-clonic seizures (TCs) at ∼3 months of age and found that heterozygous females are more resilient than males in mortality and morbidity. To investigate the cellular mechanisms that underlie female resilience, we utilized blood-brain barrier (BBB) and hippocampal transcriptomic analyses in heterozygous mice before seizure onset (pre-TC) and in mice that experienced ∼20 TCs (post-TC). RESULTS In the pre-TC latent phase, both sexes exhibited leaky BBB; however, patterns of gene expression were sexually dimorphic. Females exhibited enhanced oxidative phosphorylation and protein biogenesis, while males activated gliosis and CREB signaling. After seizure onset (chronic phase), females exhibited a metabolic switch to lipid metabolism, while males exhibited increased gliosis and BBB dysfunction and a strong activation of neuroinflammatory pathways. CONCLUSION The results underscore the central role of oxidative stress and BBB permeability in the early stages of epileptogenesis, as well as sex dimorphism in response to increasing neuronal hyperexcitability. Our results also highlight the need to include both sexes in preclinical studies to effectively translate results of drug efficacy studies.
Collapse
Affiliation(s)
- Michael F. Hammer
- BIO5 Institute, University of Arizona, Tucson, Arizona, U.S.A
- Department of Neurology, University of Arizona, Tucson, Arizona, U.S.A
| | | | - Erfan Bahramnejad
- BIO5 Institute, University of Arizona, Tucson, Arizona, U.S.A
- Department of Pharmacology, University of Arizona, Tucson, Arizona, U.S.A
| | | | - Joshua B. Hack
- BIO5 Institute, University of Arizona, Tucson, Arizona, U.S.A
| | - Joseph C. Watkins
- Department of Mathematics, University of Arizona, Tucson, Arizona, U.S.A
| | | |
Collapse
|
13
|
Malovic E, Ealy A, Hsu PJ, Sarkar S, Miller C, Rokad D, Goeser C, Hartman AK, Zhu A, Palanisamy B, Zenitsky G, Jin H, Anantharam V, Kanthasamy A, He C, Kanthasamy AG. Epitranscriptomic Reader YTHDF2 Regulates SEK1( MAP2K4 )-JNK-cJUN Inflammatory Signaling in Astrocytes during Neurotoxic Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577106. [PMID: 38328119 PMCID: PMC10849634 DOI: 10.1101/2024.01.26.577106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
As the most abundant glial cells in the CNS, astrocytes dynamically respond to neurotoxic stress, however, the key molecular regulators controlling the inflammatory status of these sentinels during neurotoxic stress have remained elusive. Herein, we demonstrate that the m6A epitranscriptomic mRNA modification tightly regulates the pro-inflammatory functions of astrocytes. Specifically, the astrocytic neurotoxic stresser, manganese (Mn), downregulated the m6A reader YTHDF2 in human and mouse astrocyte cultures and in the mouse brain. Functionally, YTHDF2 knockdown augmented, while its overexpression dampened, neurotoxic stress induced proinflammatory response, suggesting YTHDF2 serves as a key upstream regulator of inflammatory responses in astrocytes. Mechnistically, YTHDF2 RIP-sequencing identified MAP2K4 ( MKK4; SEK1) mRNA as a YTHDF2 target influencing inflammatory signaling. Our target validation revealed Mn-exposed astrocytes mediates proinflammatory response by activating the phosphorylation of SEK1, JNK, and cJUN signaling. Collectively, YTHDF2 serves a key upstream 'molecular switch' controlling SEK1( MAP2K4 )-JNK-cJUN proinflammatory signaling in astrocytes.
Collapse
|
14
|
Kim B, Zhao W, Tang SY, Levin MG, Ibrahim A, Yang Y, Roberts E, Lai L, Li J, Assoian RK, FitzGerald GA, Arany Z. Endothelial lipid droplets suppress eNOS to link high fat consumption to blood pressure elevation. J Clin Invest 2023; 133:e173160. [PMID: 37824206 PMCID: PMC10721151 DOI: 10.1172/jci173160] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023] Open
Abstract
Metabolic syndrome, today affecting more than 20% of the US population, is a group of 5 conditions that often coexist and that strongly predispose to cardiovascular disease. How these conditions are linked mechanistically remains unclear, especially two of these: obesity and elevated blood pressure. Here, we show that high fat consumption in mice leads to the accumulation of lipid droplets in endothelial cells throughout the organism and that lipid droplet accumulation in endothelium suppresses endothelial nitric oxide synthase (eNOS), reduces NO production, elevates blood pressure, and accelerates atherosclerosis. Mechanistically, the accumulation of lipid droplets destabilizes eNOS mRNA and activates an endothelial inflammatory signaling cascade that suppresses eNOS and NO production. Pharmacological prevention of lipid droplet formation reverses the suppression of NO production in cell culture and in vivo and blunts blood pressure elevation in response to a high-fat diet. These results highlight lipid droplets as a critical and unappreciated component of endothelial cell biology, explain how lipids increase blood pressure acutely, and provide a mechanistic account for the epidemiological link between obesity and elevated blood pressure.
Collapse
Affiliation(s)
- Boa Kim
- Department of Pathology and Lab Medicine, McAllister Heart Institute, Nutrition Obesity Research Center, and Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Wencao Zhao
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Soon Y. Tang
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and
| | - Michael G. Levin
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Ayon Ibrahim
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Yifan Yang
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Emilia Roberts
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ling Lai
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Jian Li
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| | - Richard K. Assoian
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Garret A. FitzGerald
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, and
| | - Zoltan Arany
- Department of Medicine, Cardiovascular Institute, and Institute of Diabetes Obesity and Metabolism, Perelman School of Medicine
| |
Collapse
|
15
|
Xiao Z, Zhang Y, She Y, Yuan G, Yang G. IPO7 promotes lipopolysaccharide-induced inflammatory responses in human dental pulp cells via p38 MAPK and NF-κB signaling pathways. Mol Immunol 2023; 163:116-126. [PMID: 37769576 DOI: 10.1016/j.molimm.2023.09.011] [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: 04/05/2023] [Revised: 07/19/2023] [Accepted: 09/14/2023] [Indexed: 10/03/2023]
Abstract
Pulpitis is a chronic inflammatory process that greatly affects the physical, mental health and life quality of patients. Human dental pulp cells (hDPCs) are essential components of dental pulp tissue and play a significant role in pulpitis. Lipopolysaccharide (LPS) is an initiator of pulpitis and can induce the production of inflammatory cytokines in hDPCs by activating p38 MAPK and NF-κB signaling pathways. Importin7 (IPO7), a member of the importin-β family, is widely expressed in many tissues. Previous studies have shown that IPO7 mediated nuclear translocation of p-p38 after stimulation, and IPO7 homologous protein IPO8 participated in human dental pulp inflammation. This research aims to investigate whether IPO7 is involved in pulpitis and explore its underlying mechanisms. In the current study, we found the expression of IPO7 was increased in pulpitis tissue. In vitro, hDPCs treated with LPS to mimic the inflammatory environment, the expression of IPO7 was increased. Knockdown of IPO7 significantly inhibited the production of inflammatory cytokines and suppressed the p38 MAPK and NF-κB signaling pathways. Activating the p38 MAPK and NF-κB signaling pathways by the p38 activator and p65 activator reversed the inflammatory responses. IPO7 interacted with p-p38 under LPS stimulation in hDPCs. In addition, the increased binding between IPO7 and p-p38 is associated with the decreased binding ability of IPO7 to Sirt2. In conclusion, we found that IPO7 was highly expressed in pulpitis and played a vital role in modulating human dental pulp inflammation.
Collapse
Affiliation(s)
- Ziqiu Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
| | - Yue Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
| | - Yawei She
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
| | - Guohua Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
| | - Guobin Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University.
| |
Collapse
|
16
|
Grabarczyk M, Ksiazek-Winiarek D, Glabinski A, Szpakowski P. Dietary Polyphenols Decrease Chemokine Release by Human Primary Astrocytes Responding to Pro-Inflammatory Cytokines. Pharmaceutics 2023; 15:2294. [PMID: 37765263 PMCID: PMC10537369 DOI: 10.3390/pharmaceutics15092294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Astrocytes are considered to be the dominant cell fraction of the central nervous system. They play a supportive and protective role towards neurons, and regulate inflammatory processes; they thus make suitable targets for drugs and supplements, such as polyphenolic compounds. However, due to their wide range, knowledge of their anti-inflammatory potential remains relatively incomplete. The aim of this study was therefore to determine whether myricetin and chrysin are able to decrease chemokine release in reactive astrocytes. To assess the antioxidant and anti-inflammatory potential of polyphenols, human primary astrocytes were cultured in the presence of a reactive and neurotoxic astrocyte-inducing cytokine mixture (TNF-α, IL-1a, C1q), either alone or in the presence of myricetin or chrysin. The examined polyphenols were able to modify the secretion of chemokines by human cortical astrocytes, especially CCL5 (chrysin), CCL1 (myricetin) and CCL2 (both), while cell viability was not affected. Surprisingly, the compounds did not demonstrate any antioxidant properties in the astrocyte cultures.
Collapse
|
17
|
Lund MC, Ellman DG, Nielsen PV, Raffaele S, Fumagalli M, Guzman R, Degn M, Brambilla R, Meyer M, Clausen BH, Lambertsen KL. Selective Inhibition of Soluble Tumor Necrosis Factor Alters the Neuroinflammatory Response following Moderate Spinal Cord Injury in Mice. BIOLOGY 2023; 12:845. [PMID: 37372129 DOI: 10.3390/biology12060845] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023]
Abstract
Clinical and animal model studies have implicated inflammation and glial and peripheral immune cell responses in the pathophysiology of spinal cord injury (SCI). A key player in the inflammatory response after SCI is the pleiotropic cytokine tumor necrosis factor (TNF), which exists both in both a transmembrane (tmTNF) and a soluble (solTNF) form. In the present study, we extend our previous findings of a therapeutic effect of topically blocking solTNF signaling after SCI for three consecutive days on lesion size and functional outcome to study the effect on spatio-temporal changes in the inflammatory response after SCI in mice treated with the selective solTNF inhibitor XPro1595 and compared to saline-treated mice. We found that despite comparable TNF and TNF receptor levels between XPro1595- and saline-treated mice, XPro1595 transiently decreased pro-inflammatory interleukin (IL)-1β and IL-6 levels and increased pro-regenerative IL-10 levels in the acute phase after SCI. This was complemented by a decrease in the number of infiltrated leukocytes (macrophages and neutrophils) in the lesioned area of the spinal cord and an increase in the number of microglia in the peri-lesion area 14 days after SCI, followed by a decrease in microglial activation in the peri-lesion area 21 days after SCI. This translated into increased myelin preservation and improved functional outcomes in XPro1595-treated mice 35 days after SCI. Collectively, our data suggest that selective targeting of solTNF time-dependently modulates the neuroinflammatory response by favoring a pro-regenerative environment in the lesioned spinal cord, leading to improved functional outcomes.
Collapse
Affiliation(s)
- Minna Christiansen Lund
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Ditte Gry Ellman
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Pernille Vinther Nielsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
| | - Stefano Raffaele
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Raphael Guzman
- Department of Biomedicine, University of Basel, 4031 Basel, Switzerland
| | - Matilda Degn
- Department of Paediatrics and Adolescent Medicine, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Roberta Brambilla
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Brain Research Inter-Disciplinary Guided Excellence (BRIDGE), Department of Clinical Research, 5000 Odense, Denmark
- Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- Brain Research Inter-Disciplinary Guided Excellence (BRIDGE), Department of Clinical Research, 5000 Odense, Denmark
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Brain Research Inter-Disciplinary Guided Excellence (BRIDGE), Department of Clinical Research, 5000 Odense, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense, Denmark
- Brain Research Inter-Disciplinary Guided Excellence (BRIDGE), Department of Clinical Research, 5000 Odense, Denmark
| |
Collapse
|
18
|
Lok HC, Katzeff JS, Hodges JR, Piguet O, Fu Y, Halliday GM, Kim WS. Elevated GRO-α and IL-18 in serum and brain implicate the NLRP3 inflammasome in frontotemporal dementia. Sci Rep 2023; 13:8942. [PMID: 37268663 DOI: 10.1038/s41598-023-35945-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023] Open
Abstract
Neuroinflammation is a hallmark of frontotemporal dementia (FTD), a heterogeneous group of proteinopathies characterized by the progressive degeneration of the frontal and temporal lobes. It is marked by microglial activation and subsequent cytokine release. Although cytokine levels in FTD brain and CSF have been examined, the number of cytokines measured in each study is limited and knowledge on cytokine concentrations in FTD serum is scarce. Here, we assessed 48 cytokines in FTD serum and brain. The aim was to determine common cytokine dysregulation pathways in serum and brain in FTD. Blood samples and brain tissue samples from the superior frontal cortex (SFC) were collected from individuals diagnosed with behavioral variant FTD (bvFTD) and healthy controls, and 48 cytokines were measured using a multiplex immunological assay. The data were evaluated by principal component factor analysis to determine the contribution from different components of the variance in the cohort. Levels of a number of cytokines were altered in serum and SFC in bvFTD compared to controls, with increases in GRO-α and IL-18 in both serum and SFC. These changes could be associated with NLRP3 inflammasome activation or the NFκB pathway, which activates NLRP3. The results suggest the possible importance of the NLRP3 inflammasome in FTD. An improved understanding of the role of inflammasomes in FTD could provide valuable insights into the pathogenesis, diagnosis and treatment of FTD.
Collapse
Affiliation(s)
- Hiu Chuen Lok
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Jared S Katzeff
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - John R Hodges
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
- School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - YuHong Fu
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| | - Woojin Scott Kim
- Brain and Mind Centre, The University of Sydney, Camperdown, Sydney, NSW, 2050, Australia.
- School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Zhang X, Liu Z, Li W, Kang Y, Xu Z, Li X, Gao Y, Qi Y. MAPKs/AP-1, not NF-κB, is responsible for MCP-1 production in TNF-α-activated adipocytes. Adipocyte 2022; 11:477-486. [PMID: 35941819 PMCID: PMC9367654 DOI: 10.1080/21623945.2022.2107786] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Obesity is associated with the infiltration of monocytes/macrophages into adipose
tissue in which MCP-1 plays a crucial role. But the regulatory mechanism of
MCP-1 expression in adipocytes is not well defined. Our results demonstrated that TNF-α induced abundant MCP-1 production in adipocytes, including 3T3-L1 pre- (≈ 9 to 18-fold), mature adipocytes (≈ 4 to 6-fold), and primary adipocytes(< 2-fold), among which 3T3-L1 pre-adipocytes showed the best reactiveness. Thus, 3T3-L1 pre-adipocytes were used for the most of following experiments. At the transcriptional level, TNF-α (20 ng/mL) also promoted the mRNA expression of MCP-1. It is well recognized that the engagement of TNF-α with its receptor can trigger both NF-κB and AP-1 signalling, which was also confirmed in our study (5-fold and 2-fold). Unexpectedly and counterintuitively, multiple NF-κB inhibitors with different mechanisms failed to suppress TNF-α-induced MCP-1 production, but rather the inhibitors for any one of MAPKs (JNK, ERK and p38) could do. This study, for the first time, reveals that MAPKs/AP-1 but not NF-κB signalling is responsible for MCP-1 production in TNF-α-activated adipocytes. These findings provide important insight into the role of AP-1 signalling in adipose tissue, and may lead to the development of therapeutical repositioning strategies in metaflammation. Abbreviations:
AP-1, activator protein-1; CHX, cycloheximide; IR, insulin resistance; MAPK, mitogen-activated protein kinase; NF-κB, nuclear factor κB; RT-qPCR, quantitative real-time PCR; T2DM, type 2 diabetes mellitus; TRE, triphorbol acetate-response element.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Zhuangzhuang Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Wenjing Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yuan Kang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Zhenlu Xu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, Haidian, China
| |
Collapse
|
20
|
Ao Z, Song S, Tian C, Cai H, Li X, Miao Y, Wu Z, Krzesniak J, Ning B, Gu M, Lee LP, Guo F. Understanding Immune-Driven Brain Aging by Human Brain Organoid Microphysiological Analysis Platform. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2200475. [PMID: 35908805 PMCID: PMC9507385 DOI: 10.1002/advs.202200475] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/17/2022] [Indexed: 05/09/2023]
Abstract
The aging of the immune system drives systemic aging and the pathogenesis of age-related diseases. However, a significant knowledge gap remains in understanding immune-driven aging, especially in brain aging, due to the limited current in vitro models of neuroimmune interaction. Here, the authors report the development of a human brain organoid microphysiological analysis platform (MAP) to discover the dynamic process of immune-driven brain aging. The organoid MAP is created by 3D printing that confines organoid growth and facilitates cell and nutrition perfusion, promoting organoid maturation and their committment to forebrain identity. Dynamic rocking flow is incorporated into the platform that allows to perfuse primary monocytes from young (20 to 30-year-old) and aged (>60-year-old) donors and culture human cortical organoids to model neuroimmune interaction. The authors find that the aged monocytes increase infiltration and promote the expression of aging-related markers (e.g., higher expression of p16) within the human cortical organoids, indicating that aged monocytes may drive brain aging. The authors believe that the organoid MAP may provide promising solutions for basic research and translational applications in aging, neural immunological diseases, autoimmune disorders, and cancer.
Collapse
Affiliation(s)
- Zheng Ao
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Sunghwa Song
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Chunhui Tian
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Hongwei Cai
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Xiang Li
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine (CuSTOM)Division of Pulmonary BiologyDivision of Developmental BiologyCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
- University of Cincinnati School of MedicineCincinnatiOH45229USA
| | - Zhuhao Wu
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Jonathan Krzesniak
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| | - Bo Ning
- Center for Cellular and Molecular DiagnosticsDepartment of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLA70112USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine (CuSTOM)Division of Pulmonary BiologyDivision of Developmental BiologyCincinnati Children's Hospital Medical CenterCincinnatiOH45229USA
- University of Cincinnati School of MedicineCincinnatiOH45229USA
| | - Luke P. Lee
- Harvard Institute of MedicineHarvard Medical SchoolHarvard UniversityBrigham and Women's HospitalBostonMA02115USA
- Department of BioengineeringDepartment of Electrical Engineering and Computer ScienceUniversity of California at BerkeleyBerkeleyCA94720USA
- Department of BiophysicsInstitute of Quantum BiophysicsSungkyunkwan UniversitySuwonGyeonggi‐do16419South Korea
| | - Feng Guo
- Department of Intelligent Systems EngineeringIndiana UniversityBloomingtonIN47405USA
| |
Collapse
|
21
|
Zhang Z, Cui Y, Liu S, Huang J, Liu Y, Zhou Y, Zhu Z. Short-term treatment with zingerone ameliorates dextran sulfate sodium-induced mouse experimental colitis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:4873-4882. [PMID: 35246845 DOI: 10.1002/jsfa.11850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 02/12/2022] [Accepted: 03/04/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Ulcerative colitis (UC) is a relapsing and chronic inflammatory disease of the gastrointestinal tract, which seriously threatens human health. Zingerone (ZO) has been proven to be effective for many diseases. The purpose of this study is to investigate the protective effects and potential mechanisms of ZO extracted from ginger on dextran sulfate sodium (DSS)-induced mouse ulcerative colitis (UC). RESULTS The results showed that ZO alleviated the weight loss of UC model mice, reduced the disease activity index scores, and inhibited the shortening of colon length. ZO also improved DSS-induced pathological changes in colon tissue and inhibited the secretion of pro-inflammatory cytokines in colon and mesenteric lymph nodes. Further mechanism analysis found that ZO inhibited DSS-induced nuclear factor-κB pathway activation, and regulated peroxisome proliferator-activated receptor γ (PPARγ) expression. To further explore whether PPARγ was involved in the anti-UC effect of ZO, PPARγ inhibitor GW9662 was used. Although ZO also showed a protective effect on GW9662-treated colitis mice, the protective role was significantly weakened. Importantly, the administration of GW9662 significantly aggravated UC compared with the ZO + DSS group. In addition, we preliminarily found that ZO had the effects of inhibiting DSS-induced oxidative stress, maintaining intestinal barrier, and inhibiting the content of LPS and the population of Escherichia coli. CONCLUSIONS These results indicated that supplementation with ZO might be a new dietary strategy for the treatment of UC. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Yueqi Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
| | - Siyu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
| | - Jiang Huang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Heilongjiang Provincial Technology Innovation Center for Bovine Disease Control and Prevention, Daqing, China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| |
Collapse
|
22
|
Maciejewska-Skrendo A, Tarnowski M, Kopytko P, Kochanowicz A, Mieszkowski J, Stankiewicz B, Sawczuk M. CCL2 Gene Expression and Protein Level Changes Observed in Response to Wingate Anaerobic Test in High-Trained Athletes and Non-Trained Controls. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9947. [PMID: 36011581 PMCID: PMC9408289 DOI: 10.3390/ijerph19169947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 06/15/2023]
Abstract
Intensive, acute exercise may bring a large systemic inflammatory response marked by substantial increases in inflammatory cytokines and chemokines. One such chemokines-CCL2-is a key factor involved in inflammatory reaction to exercise. The direct aim of the study was to describe the changes in the CCL2 expression levels after anaerobic exercise in well-trained athletes adapted to long-term training and in non-trained participants. The expression of CCL2 mRNA was evaluated in peripheral blood MNCs and CCL2 protein level was observed in blood plasma. The changes were assessed as the response to an acute, intensive bout of exercise (Wingate Anaerobic Test) in two groups of participants: well-trained soccer players and non-trained individuals. An increase of CCL2 expression inn both mRNA and protein levels was observed. The response was greater in non-trained individuals and elevated levels of CCL2 transcripts persisted for more than 24 h after exercise. Well-trained individuals responded more modestly and the effect was attenuated relatively quickly. This shows muscular adaptation to a continuous training regime in well-trained individuals and better control of immune reactions to muscular injury. In non-training individuals, the induction of the inflammatory response was greater, suggesting presence of more serious myotrauma.
Collapse
Affiliation(s)
- Agnieszka Maciejewska-Skrendo
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland or
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland or
| | - Maciej Tarnowski
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland or
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patrycja Kopytko
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland or
| | - Andrzej Kochanowicz
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland or
| | - Jan Mieszkowski
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland or
| | - Błażej Stankiewicz
- Institute of Physical Culture, Kazimierz Wielki University, 85-091 Bydgoszcz, Poland
| | - Marek Sawczuk
- Faculty of Physical Culture, Gdansk University of Physical Education and Sport, 80-336 Gdansk, Poland or
- Institute of Physical Culture Sciences, University of Szczecin, 71-065 Szczecin, Poland or
| |
Collapse
|
23
|
Li Y, Lei Z, Ritzel RM, He J, Li H, Choi HMC, Lipinski MM, Wu J. Impairment of autophagy after spinal cord injury potentiates neuroinflammation and motor function deficit in mice. Theranostics 2022; 12:5364-5388. [PMID: 35910787 PMCID: PMC9330534 DOI: 10.7150/thno.72713] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 01/25/2023] Open
Abstract
Autophagy is a catabolic process that degrades cytoplasmic constituents and organelles in the lysosome, thus serving an important role in cellular homeostasis and protection against insults. We previously reported that defects in autophagy contribute to neuronal cell damage in traumatic spinal cord injury (SCI). Recent data from other inflammatory models implicate autophagy in regulation of immune and inflammatory responses, with low levels of autophagic flux associated with pro-inflammatory phenotypes. In the present study, we examined the effects of genetically or pharmacologically manipulating autophagy on posttraumatic neuroinflammation and motor function after SCI in mice. Methods: Young adult male C57BL/6, CX3CR1-GFP, autophagy hypomorph Becn1+/- mice, and their wildtype (WT) littermates were subjected to moderate thoracic spinal cord contusion. Neuroinflammation and autophagic flux in the injured spinal cord were assessed using flow cytometry, immunohistochemistry, and NanoString gene expression analysis. Motor function was evaluated with the Basso Mouse Scale and horizontal ladder test. Lesion volume and spared white matter were evaluated by unbiased stereology. To stimulate autophagy, disaccharide trehalose, or sucrose control, was administered in the drinking water immediately after injury and for up to 6 weeks after SCI. Results: Flow cytometry demonstrated dysregulation of autophagic function in both microglia and infiltrating myeloid cells from the injured spinal cord at 3 days post-injury. Transgenic CX3CR1-GFP mice revealed increased autophagosome formation and inhibition of autophagic flux specifically in activated microglia/macrophages. NanoString analysis using the neuroinflammation panel demonstrated increased expression of proinflammatory genes and decreased expression of genes related to neuroprotection in Becn1+/- mice as compared to WT controls at 3 days post-SCI. These findings were further validated by qPCR, wherein we observed significantly higher expression of proinflammatory cytokines. Western blot analysis confirmed higher protein expression of the microglia/macrophage marker IBA-1, inflammasome marker, NLRP3, and innate immune response markers cGAS and STING in Becn1+/- mice at 3 day after SCI. Flow cytometry demonstrated that autophagy deficit did not affect either microglial or myeloid counts at 3 days post-injury, instead resulting in increased microglial production of proinflammatory cytokines. Finally, locomotor function showed significantly worse impairments in Becn1+/- mice up to 6 weeks after SCI, which was accompanied by worsening tissue damage. Conversely, treatment with a naturally occurring autophagy inducer trehalose, reduced protein levels of p62, an adaptor protein targeting cargo to autophagosomes as well as the NLRP3, STING, and IBA-1 at 3 days post-injury. Six weeks of trehalose treatment after SCI led to improved motor function recovery as compared to control group, which was accompanied by reduced tissue damage. Conclusions: Our data indicate that inhibition of autophagy after SCI potentiates pro-inflammatory activation in microglia and is associated with worse functional outcomes. Conversely, increasing autophagy with trehalose, decreased inflammation and improved outcomes. These findings highlight the importance of autophagy in spinal cord microglia and its role in secondary injury after SCI.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Hui Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Harry M C Choi
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Marta M Lipinski
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| |
Collapse
|
24
|
Serra A, del Giudice G, Kinaret PAS, Saarimäki LA, Poulsen SS, Fortino V, Halappanavar S, Vogel U, Greco D. Characterization of ENM Dynamic Dose-Dependent MOA in Lung with Respect to Immune Cells Infiltration. NANOMATERIALS 2022; 12:nano12122031. [PMID: 35745370 PMCID: PMC9228743 DOI: 10.3390/nano12122031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 02/01/2023]
Abstract
The molecular effects of exposures to engineered nanomaterials (ENMs) are still largely unknown. In classical inhalation toxicology, cell composition of bronchoalveolar lavage (BAL) is a toxicity indicator at the lung tissue level that can aid in interpreting pulmonary histological changes. Toxicogenomic approaches help characterize the mechanism of action (MOA) of ENMs by investigating the differentially expressed genes (DEG). However, dissecting which molecular mechanisms and events are directly induced by the exposure is not straightforward. It is now generally accepted that direct effects follow a monotonic dose-dependent pattern. Here, we applied an integrated modeling approach to study the MOA of four ENMs by retrieving the DEGs that also show a dynamic dose-dependent profile (dddtMOA). We further combined the information of the dddtMOA with the dose dependency of four immune cell populations derived from BAL counts. The dddtMOA analysis highlighted the specific adaptation pattern to each ENM. Furthermore, it revealed the distinct effect of the ENM physicochemical properties on the induced immune response. Finally, we report three genes dose-dependent in all the exposures and correlated with immune deregulation in the lung. The characterization of dddtMOA for ENM exposures, both for apical endpoints and molecular responses, can further promote toxicogenomic approaches in a regulatory context.
Collapse
Affiliation(s)
- Angela Serra
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (G.d.G.); (L.A.S.)
- BioMediTech Institute, Tampere University, 33520 Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), 33520 Tampere, Finland
| | - Giusy del Giudice
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (G.d.G.); (L.A.S.)
- BioMediTech Institute, Tampere University, 33520 Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), 33520 Tampere, Finland
| | | | - Laura Aliisa Saarimäki
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (G.d.G.); (L.A.S.)
- BioMediTech Institute, Tampere University, 33520 Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), 33520 Tampere, Finland
| | - Sarah Søs Poulsen
- National Research Centre for the Working Environment, 2100 Copenhagen, Denmark; (S.S.P.); (U.V.)
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON K1A 0K9, Canada;
| | - Ulla Vogel
- National Research Centre for the Working Environment, 2100 Copenhagen, Denmark; (S.S.P.); (U.V.)
| | - Dario Greco
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (A.S.); (G.d.G.); (L.A.S.)
- BioMediTech Institute, Tampere University, 33520 Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), 33520 Tampere, Finland
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland;
- Correspondence:
| |
Collapse
|
25
|
Huang LJ, Wang YM, Gong LQ, Hu C, Gui Y, Zhang C, Tan X, Yu XK, Liao YL, Luo Y, Tang YQ, Dai YF, Deng Y, Wang D, Guo DL. N-Acetyldopamine Dimer Attenuates DSS-Induced Ulcerative Colitis by Suppressing NF-κB and MAPK Pathways. Front Pharmacol 2022; 13:842730. [PMID: 35462925 PMCID: PMC9030057 DOI: 10.3389/fphar.2022.842730] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
Ulcerative Colitis (UC) is a major form of chronic inflammatory bowel disease of the colonic mucosa and exhibits progressive morbidity. There is still a substantial need of small molecules with greater efficacy and safety for UC treatment. Here, we report a N-acetyldopamine dimer (NADD) elucidated (2R,3S)-2-(3′,4′-dihydroxyphenyl)-3-acetylamino-7-(N-acetyl-2″-aminoethyl)-1,4-benzodioxane, which is derived from traditional Chinese medicine Isaria cicadae, exhibits significant therapeutic efficacy against dextran sulfate sodium (DSS)-induced UC. Functionally, NADD treatment effectively relieves UC symptoms, including weight loss, colon length shortening, colonic tissue damage and expression of pro-inflammatory factors in pre-clinical models. Mechanistically, NADD treatment significantly inhibits the expression of genes in inflammation related NF-κB and MAPK signaling pathways by transcriptome analysis and western blot, which indicates that NADD inhibits the inflammation in UC might through these two pathways. Overall, this study identifies an effective small molecule for UC therapy.
Collapse
Affiliation(s)
- Li-Jun Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Mei Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei-Qiang Gong
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Gui
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian-Kuo Yu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Le Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Qin Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Fei Dai
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yun Deng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Da-le Guo
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
26
|
Czarnek M, Stalińska K, Sarad K, Bereta J. shRNAs targeting mouse Adam10 diminish cell response to proinflammatory stimuli independently of Adam10 silencing. Biol Open 2022; 11:274200. [PMID: 35107128 PMCID: PMC8905717 DOI: 10.1242/bio.059092] [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: 10/14/2021] [Accepted: 01/20/2022] [Indexed: 11/23/2022] Open
Abstract
RNA interference is one of the common methods of studying protein functions. In recent years critical reports have emerged indicating that off-target effects may have a much greater impact on RNAi-based analysis than previously assumed. We studied the influence of Adam10 and Adam17 silencing on MC38CEA cell response to proinflammatory stimuli. Eight lentiviral vector-encoded shRNAs that reduced ADAM10 expression, including two that are specific towards ADAM17, caused inhibition of cytokine-induced Nos2 expression presumably via off-target effects. ADAM10 silencing was not responsible for this effect because: (i) CRISPR/Cas9 knockdown of ADAM10 did not affect Nos2 levels; (ii) ADAM10 inhibitor increased rather than decreased Nos2 expression; (iii) overexpression of ADAM10 in the cells with shRNA-silenced Adam10 did not reverse the effect induced by shRNA; (iv) shRNA targeting ADAM10 resulted in decrease of Nos2 expression even in ADAM10-deficient cells. The studied shRNAs influenced transcription of Nos2 rather than stability of Nos2 mRNA. They also affected stimulation of Ccl2 and Ccl7 expression. Additionally, we used vectors with doxycycline-inducible expression of chosen shRNAs and observed reduced activation of NF-κB and, to a lesser extent, AP-1 transcription factors. We discuss the requirements of strict controls and verification of results with complementary methods for reliable conclusions of shRNA-based experiments. Summary: Use of several specific shRNAs is not enough to escape a pitfall of their off-target activity: the case of Adam10 and Adam17 silencing.
Collapse
Affiliation(s)
- Maria Czarnek
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387 Kraków, Poland
| | - Krystyna Stalińska
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387 Kraków, Poland
| | - Katarzyna Sarad
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387 Kraków, Poland
| | - Joanna Bereta
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Kraków, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
27
|
Daghestani MH, Ambreen K, Hakami HH, Omair MA, Saleem AM, Aleisa NA, AlNeghery LM, Amin MH, Alobaid HM, Omair MA, Hassen LM. Venom of the desert black snake Walterinnesia aegyptia enhances anti-tumor immunity via its beneficial modulatory effects on pro- and anti-tumorigenic inflammatory mediators in cultured colon cancer cells. Toxicol Res (Camb) 2021; 10:1116-1128. [PMID: 34956615 DOI: 10.1093/toxres/tfab093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/14/2022] Open
Abstract
The role of inflammation in colon cancer is understood as a well-accepted factor that has the tendency to release multiple pro- and anti-tumorigenic inflammatory mediators. Inflammation-induced increased expression of anti-tumorigenic inflammatory mediators and decreased expression of pro-tumorigenic inflammatory mediators encourage beneficial inflammatory effects in terms of powerful anti-tumor immunity. The present study aims to screen the beneficial inflammatory effects of Walterinnesia aegyptia venom via determining its modulatory tendency on the expression of 40 pro- and anti-tumorigenic inflammatory mediators (cytokines/growth factors/chemokines) in LoVo human colon cancer cell line. LoVo-cells were treated with varying doses of crude venom of W. aegyptia. Cell viability was checked utilizing flow cytometry, and IC50 of venom was determined. Venom-induced inflammatory effects were evaluated on the expression of 40 different inflammatory mediators (12 anti-tumorigenic cytokines, 11 pro-tumorigenic cytokines, 7 pro-tumorigenic growth factors, 9 pro-tumorigenic chemokines and 1 anti-tumorigenic chemokine) in treated LoVo-cells [utilizing enzyme-linked immunosorbent assay (ELISA)] and compared with controls. Treatment of venom induced significant cytotoxic effects on inflamed LoVo-cells. IC50 treatment of venom caused significant modulations on the expression of 22 inflammatory mediators in treated LoVo-cells. The beneficial modulatory effects of venom were screened via its capability to significantly increase the expression of five powerful anti-tumorigenic mediators (IL-9, IL-12p40, IL-15, IL-1RA and Fractalkine) and decrease the expression of four major pro-tumorigenic mediators (IL-1β, VEGF, MCP-1 and MCP-3). Walterinnesia aegyptia venom-induced beneficial modulations on the expression of nine crucial pro/anti-tumorigenic inflammatory mediators can be effectively used to enhance powerful anti-tumor immunity against colon cancer.
Collapse
Affiliation(s)
- Maha H Daghestani
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Khushboo Ambreen
- Department of Biotechnology, Integral University, Lucknow, India
| | - Hana H Hakami
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed A Omair
- Division of Rheumatology, Department of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz M Saleem
- Department of Surgery, Medical College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nadia A Aleisa
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Lina M AlNeghery
- Department of Biology, College of Science, Al Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Mohannad H Amin
- College of Dentistry, Riyadh ELM University, Riyadh, Saudi Arabia
| | - Hussah M Alobaid
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| | - Maha A Omair
- Department of Statistics and Operations Research, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Lena M Hassen
- Department of Zoology, College of Science, Centre for Scientific and Medical Female Colleges, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
28
|
Yang Y, Shi K, Patel DM, Liu F, Wu T, Chai Z. How to inhibit transforming growth factor beta safely in diabetic kidney disease. Curr Opin Nephrol Hypertens 2021; 30:115-122. [PMID: 33229911 DOI: 10.1097/mnh.0000000000000663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW Diabetic kidney disease (DKD) is a leading cause of mortality and morbidity in diabetes. This review aims to discuss the major features of DKD, to identify the difficult barrier encountered in developing a therapeutic strategy and to provide a potentially superior novel approach to retard DKD. RECENT FINDINGS Renal inflammation and fibrosis are prominent features of DKD. Transforming growth factor beta (TGFβ) with its activity enhanced in DKD plays a key pathological profibrotic role in promoting renal fibrosis. However, TGFβ is a difficult drug target because it has multiple important physiological functions, such as immunomodulation. These physiological functions of TGFβ can be interrupted as a result of complete blockade of the TGFβ pathway if TGFβ is directly targeted, leading to catastrophic side-effects, such as fulminant inflammation. Cell division autoantigen 1 (CDA1) is recently identified as an enhancer of profibrotic TGFβ signaling and inhibitor of anti-inflammatory SIRT1. Renal CDA1 expression is elevated in human DKD as well as in rodent models of DKD. Targeting CDA1, by either genetic approach or pharmacological approach in mice, leads to concurrent attenuation of renal fibrosis and inflammation without any deleterious effects observed. SUMMARY Targeting CDA1, instead of directly targeting TGFβ, represents a superior approach to retard DKD.
Collapse
Affiliation(s)
- Yuxin Yang
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Pathology, Zunyi maternity and Child Healthcare Hospital, Zunyi
| | - Kexin Shi
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Devang M Patel
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Fang Liu
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, China
| | - Tieqiao Wu
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Zhonglin Chai
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Ducza L, Szücs P, Hegedűs K, Bakk E, Gajtkó A, Wéber I, Holló K. NLRP2 Is Overexpressed in Spinal Astrocytes at the Peak of Mechanical Pain Sensitivity during Complete Freund Adjuvant-Induced Persistent Pain. Int J Mol Sci 2021; 22:ijms222111408. [PMID: 34768839 PMCID: PMC8584130 DOI: 10.3390/ijms222111408] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 12/15/2022] Open
Abstract
Our earlier findings revealed that interleukin-1 receptor type-1 (IL-1R1) was overexpressed in spinal neurons, and IL-1R1-deficient mice showed significant attenuation of thermal and mechanical allodynia during the course of the Complete Freund adjuvant (CFA)-induced persistent pain model. In the present study, we found that a ligand of IL-1R1, termed interleukin-1β (IL-1β), is also significantly overexpressed at the peak of mechanical pain sensitivity in the CFA-evoked pain model. Analysis of cellular distribution and modeling using IMARIS software showed that in the lumbar spinal dorsal horn, IL-1β is significantly elevated by astrocytic expression. Maturation of IL-1β to its active form is facilitated by the formation of the multiprotein complex called inflammasome; thus, we tested the expression of NOD-like receptor proteins (NLRPs) in astrocytes. At the peak of mechanical allodynia, we found expression of the NLRP2 inflammasome sensor and its significantly elevated co-localization with the GFAP astrocytic marker, while NLRP3 was moderately present and NLRP1 showed total segregation from the astrocytic profiles. Our results indicate that peripheral CFA injection induces NLRP2 inflammasome and IL-1β expression in spinal astrocytes. The release of mature IL-1β can contribute to the maintenance of persistent pain by acting on its neuronally expressed receptor, which can lead to altered neuronal excitability.
Collapse
|
30
|
Jurga AM, Paleczna M, Kadluczka J, Kuter KZ. Beyond the GFAP-Astrocyte Protein Markers in the Brain. Biomolecules 2021; 11:biom11091361. [PMID: 34572572 PMCID: PMC8468264 DOI: 10.3390/biom11091361] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
The idea of central nervous system as one-man band favoring neurons is long gone. Now we all are aware that neurons and neuroglia are team players and constant communication between those various cell types is essential to maintain functional efficiency and a quick response to danger. Here, we summarize and discuss known and new markers of astroglial multiple functions, their natural heterogeneity, cellular interactions, aging and disease-induced dysfunctions. This review is focused on newly reported facts regarding astrocytes, which are beyond the old stereotypes. We present an up-to-date list of marker proteins used to identify a broad spectrum of astroglial phenotypes related to the various physiological and pathological nervous system conditions. The aim of this review is to help choose markers that are well-tailored for specific needs of further experimental studies, precisely recognizing differential glial phenotypes, or for diagnostic purposes. We hope it will help to categorize the functional and structural diversity of the astroglial population and ease a clear readout of future experimental results.
Collapse
|
31
|
CC Chemokine Ligand 7 Derived from Cancer-Stimulated Macrophages Promotes Ovarian Cancer Cell Invasion. Cancers (Basel) 2021; 13:cancers13112745. [PMID: 34206004 PMCID: PMC8198020 DOI: 10.3390/cancers13112745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
In the tumor microenvironment, macrophages have been suggested to be stimulated by tumor cells, becoming tumor-associated macrophages that promote cancer development and progression. We examined the effect of these macrophages on human ovarian cancer cell invasion and found that conditioned medium of macrophages stimulated by ovarian cancer cells (OC-MQs) significantly increased cell invasion. CC chemokine ligand 7 (CCL7) expression and production were significantly higher in OC-MQs than in the control macrophages. Peritoneal macrophages from patients with ovarian cancer showed higher CCL7 expression levels than those from healthy controls. Inhibition of CCL7 using siRNA and neutralizing antibodies reduced the OC-MQ-CM-induced ovarian cancer cell invasion. CC chemokine receptor 3 (CCR3) was highly expressed in human ovarian cancer cells, and a specific inhibitor of this receptor reduced the OC-MQ-CM-induced invasion. Specific signaling and transcription factors were associated with enhanced CCL7 expression in OC-MQs. CCL7-induced invasion required the expression of matrix metalloproteinase 9 via activation of extracellular signal-related kinase signaling in human ovarian cancer cells. These data suggest that tumor-associated macrophages can affect human ovarian cancer metastasis via the CCL7/CCR3 axis.
Collapse
|
32
|
Ye J, Wang H, Cui L, Chu S, Chen N. The progress of chemokines and chemokine receptors in autism spectrum disorders. Brain Res Bull 2021; 174:268-280. [PMID: 34077795 DOI: 10.1016/j.brainresbull.2021.05.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/16/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders and the main symptoms of ASD are impairments in social communication and abnormal behavioral patterns. Studies have shown that immune dysfunction and neuroinflammation play a key role in ASD patients and experimental models. Chemokines are groups of small proteins that regulate cell migration and mediate inflammation responses via binding to chemokine receptors. Thus, chemokines/chemokine receptors may be involved in neurodevelopmental disorders and associated with ASD. In this review, we summarize the research progress of chemokine aberrations in ASD and also review the recent progress of clinical treatment of ASD and pharmacological research related to chemokines/chemokine receptors. This review highlights the possible connection between chemokines/chemokine receptors and ASD, and provides novel potential targets for drug discovery of ASD.
Collapse
Affiliation(s)
- Junrui Ye
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hongyun Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Liyuan Cui
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
33
|
Lertnimitphun P, Zhang W, Fu W, Yang B, Zheng C, Yuan M, Zhou H, Zhang X, Pei W, Lu Y, Xu H. Safranal Alleviated OVA-Induced Asthma Model and Inhibits Mast Cell Activation. Front Immunol 2021; 12:585595. [PMID: 34093515 PMCID: PMC8173045 DOI: 10.3389/fimmu.2021.585595] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction Asthma is a chronic and recurring airway disease, which related to mast cell activation. Many compounds derived from Chinese herbal medicine has promising effects on stabilizing mast cells and decreasing inflammatory mediator production. Safranal, one of the active compounds from Crocus sativus, shows many anti-inflammatory properties. In this study, we evaluated the effect of safranal in ovalbumin (OVA)-induced asthma model. Furthermore, we investigate the effectiveness of safranal on stabilizing mast cell and inhibiting the production of inflammatory mediators in passive systemic anaphylaxis (PSA) model. Methods OVA-induced asthma and PSA model were used to evaluate the effect of safranal in vivo. Lung tissues were collected for H&E, TB, IHC, and PAS staining. ELISA were used to determine level of IgE and chemokines (IL-4, IL-5, TNF-α, and IFN-γ). RNA sequencing was used to uncovers genes that safranal regulate. Bone marrow-derived mast cells (BMMCs) were used to investigate the inhibitory effect and mechanism of safranal. Cytokine production (IL-6, TNF-α, and LTC4) and NF-κB and MAPKs signaling pathway were assessed. Results Safranal reduced the level of serum IgE, the number of mast cells in lung tissue were decreased and Th1/Th2 cytokine levels were normalized in OVA-induced asthma model. Furthermore, safranal inhibited BMMCs degranulation and inhibited the production of LTC4, IL-6, and TNF-α. Safranal inhibits NF-κB and MAPKs pathway protein phosphorylation and decreases NF-κB p65, AP-1 nuclear translocation. In the PSA model, safranal reduced the levels of histamine and LTC4 in serum. Conclusions Safranal alleviates OVA-induced asthma, inhibits mast cell activation and PSA reaction. The possible mechanism occurs through the inhibition of the MAPKs and NF-κB pathways.
Collapse
Affiliation(s)
- Peeraphong Lertnimitphun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Acupuncture and Moxibustion, Huachiew TCM Hospital, Bangkok, Thailand
| | - Wenhui Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenwei Fu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baican Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Changwu Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Yuan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Zhou
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Zhang
- Saffron Department and International Trade Department, Shanghai Traditional Chinese Medicine Co., Ltd., Shanghai, China
| | - Weizhong Pei
- Saffron Department and International Trade Department, Shanghai Traditional Chinese Medicine Co., Ltd., Shanghai, China
| | - Yue Lu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Blood-brain barrier opening by intracarotid artery hyperosmolar mannitol induces sterile inflammatory and innate immune responses. Proc Natl Acad Sci U S A 2021; 118:2021915118. [PMID: 33906946 DOI: 10.1073/pnas.2021915118] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracarotid arterial hyperosmolar mannitol (ICAHM) blood-brain barrier disruption (BBBD) is effective and safe for delivery of therapeutics for central nervous system malignancies. ICAHM osmotically alters endothelial cells and tight junction integrity to achieve BBBD. However, occurrence of neuroinflammation following hemispheric BBBD by ICAHM remains unknown. Temporal proteomic changes in rat brains following ICAHM included increased damage-associated molecular patterns, cytokines, chemokines, trophic factors, and cell adhesion molecules, indicative of a sterile inflammatory response (SIR). Proteomic changes occurred within 5 min of ICAHM infusion and returned to baseline by 96 h. Transcriptomic analyses following ICAHM BBBD further supported an SIR. Immunohistochemistry revealed activated astrocytes, microglia, and macrophages. Moreover, proinflammatory proteins were elevated in serum, and proteomic and histological findings from the contralateral hemisphere demonstrated a less pronounced SIR, suggesting neuroinflammation beyond regions of ICAHM infusion. Collectively, these results demonstrate ICAHM induces a transient SIR that could potentially be harnessed for neuroimmunomodulation.
Collapse
|
35
|
Nisar S, Yousuf P, Masoodi T, Wani NA, Hashem S, Singh M, Sageena G, Mishra D, Kumar R, Haris M, Bhat AA, Macha MA. Chemokine-Cytokine Networks in the Head and Neck Tumor Microenvironment. Int J Mol Sci 2021; 22:ijms22094584. [PMID: 33925575 PMCID: PMC8123862 DOI: 10.3390/ijms22094584] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are aggressive diseases with a dismal patient prognosis. Despite significant advances in treatment modalities, the five-year survival rate in patients with HNSCC has improved marginally and therefore warrants a comprehensive understanding of the HNSCC biology. Alterations in the cellular and non-cellular components of the HNSCC tumor micro-environment (TME) play a critical role in regulating many hallmarks of cancer development including evasion of apoptosis, activation of invasion, metastasis, angiogenesis, response to therapy, immune escape mechanisms, deregulation of energetics, and therefore the development of an overall aggressive HNSCC phenotype. Cytokines and chemokines are small secretory proteins produced by neoplastic or stromal cells, controlling complex and dynamic cell-cell interactions in the TME to regulate many cancer hallmarks. This review summarizes the current understanding of the complex cytokine/chemokine networks in the HNSCC TME, their role in activating diverse signaling pathways and promoting tumor progression, metastasis, and therapeutic resistance development.
Collapse
Affiliation(s)
- Sabah Nisar
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Parvaiz Yousuf
- Department of Zoology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Tariq Masoodi
- Department of Genomic Medicine, Genetikode 400102, India;
| | - Nissar A. Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal 191201, India;
| | - Sheema Hashem
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
| | - Mayank Singh
- Departmental of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi 110029, India;
| | | | - Deepika Mishra
- Centre for Dental Education and Research, Department of Oral Pathology and Microbiology, All India Institute of Medical Sciences, New Delhi 110029, India;
| | - Rakesh Kumar
- Centre for Advanced Research, School of Biotechnology and Indian Council of Medical Research, Shri Mata Vaishno Devi University, Katra 182320, India;
| | - Mohammad Haris
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Ajaz A. Bhat
- Molecular and Metabolic Imaging Laboratory, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar; (S.N.); (S.H.); (M.H.)
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora 192122, India
- Correspondence: (A.A.B.); or (M.A.M.); Tel.: +974-40037703 (A.A.B.); +91-8082326900 (M.A.M.)
| |
Collapse
|
36
|
EZH2 Promotes Extracellular Matrix Degradation via Nuclear Factor-κB (NF-κB) and p38 Signaling Pathways in Pulpitis. Inflammation 2021; 44:1927-1936. [PMID: 33884563 DOI: 10.1007/s10753-021-01470-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 02/05/2023]
Abstract
Pulpitis is a complicated chronic inflammatory process which can be in a dynamic balance between damage and repair. The extracellular matrix plays an important regulatory role in wound healing and tissue repair. The aim of this study was to explore the role of the epigenetic mark, enhancer of zeste homolog 2 (EZH2) on the degradation of extracellular matrix during pulpitis. Quantitative polymerase chain reaction was used to assess the expression of matrix metalloproteinases (MMPs) and type I collagen in human dental pulp cells (HDPCs) upon EZH2 and EI1 (EZH2 inhibitor) stimulation. The mechanism of EZH2 affecting extracellular matrix was explored through quantitative polymerase chain reaction and Western blot. A rat model of dental pulp inflammation was established, and the expression of type I collagen in dental pulp under EZH2 stimulation was detected by immunohistochemical staining. EZH2 upregulated the expression of MMP-1, MMP-3, MMP-8, and MMP-10 and decreased the production of type I collagen in HDPCs, while EI1 had the opposite effect. EZH2 activated the nuclear factor-kappa B (NF-κB) and p38 signaling pathways in HDPCs, the inhibition of which reversed the induction of MMPs and the suppression of type I collagen. EZH2 can downregulate the type I collagen levels in an experimental model of dental pulpitis in rats. EZH2 promotes extracellular matrix degradation via nuclear factor-κB (NF-κB) and P38 signaling pathways in pulpitis. EZH2 can decrease the type I collagen levels in vivo and in vitro.
Collapse
|
37
|
Liberale L, Ministrini S, Carbone F, Camici GG, Montecucco F. Cytokines as therapeutic targets for cardio- and cerebrovascular diseases. Basic Res Cardiol 2021; 116:23. [PMID: 33770265 PMCID: PMC7997823 DOI: 10.1007/s00395-021-00863-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Despite major advances in prevention and treatment, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. In this context, inflammation is involved in the chronic process leading atherosclerotic plaque formation and its complications, as well as in the maladaptive response to acute ischemic events. For this reason, modulation of inflammation is nowadays seen as a promising therapeutic strategy to counteract the burden of cardio- and cerebrovascular disease. Being produced and recognized by both inflammatory and vascular cells, the complex network of cytokines holds key functions in the crosstalk of these two systems and orchestrates the progression of atherothrombosis. By binding to membrane receptors, these soluble mediators trigger specific intracellular signaling pathways eventually leading to the activation of transcription factors and a deep modulation of cell function. Both stimulatory and inhibitory cytokines have been described and progressively reported as markers of disease or interesting therapeutic targets in the cardiovascular field. Nevertheless, cytokine inhibition is burdened by harmful side effects that will most likely prevent its chronic use in favor of acute administrations in well-selected subjects at high risk. Here, we summarize the current state of knowledge regarding the modulatory role of cytokines on atherosclerosis, myocardial infarction, and stroke. Then, we discuss evidence from clinical trials specifically targeting cytokines and the potential implication of these advances into daily clinical practice.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland. .,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Stefano Ministrini
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,Internal Medicine, Angiology and Atherosclerosis, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, 8952, Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
38
|
Feng B, Zhu Y, Yan L, Yan H, Huang X, Jiang D, Li Z, Hua L, Zhuo Y, Fang Z, Che L, Lin Y, Xu S, Huang C, Zou Y, Li L, Wu D. Ursolic acid induces the production of IL6 and chemokines in both adipocytes and adipose tissue. Adipocyte 2020; 9:523-534. [PMID: 32876525 PMCID: PMC7714451 DOI: 10.1080/21623945.2020.1814545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/12/2020] [Accepted: 08/20/2020] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue inflammation plays an important role in the regulation of glucose and lipids metabolism. It is unknown whether Ursolic acid (UA) could regulate adipose tissue inflammation, though it can regulate inflammation in many other tissues. In this study, 3T3-L1 adipocytes, DIO mice and lean mice were treated with UA or vehicle. Gene expression of inflammatory factors, chemokines and immune markers in adipocytes and adipose tissue, cytokines in cell culture medium and serum, and inflammation regulatory pathways in adipocytes were detected. Results showed that UA increased the expression of interleukins and chemokines, but not TNFα, in both adipocytes and adipose tissue. IL6 and MCP1 levels in the cell culture medium and mouse serum were induced by UA treatment. Cd14 expression level and number of CD14+ monocytes were higher in UA treated adipose tissue than those in the control group. Glucose tolerance test was impaired by UA treatment in DIO mice. Mechanistically, UA induced the expression of Tlr4 and the phosphorylation levels of ERK and NFκB in adipocytes. In conclusion, our study indicated that short-term UA administration could induce CD14+ monocytes infiltration by increasing the production of interleukins and chemokines in mouse adipose tissue, which might further impair glucose tolerance test.
Collapse
Affiliation(s)
- Bin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yingguo Zhu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lijun Yan
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hui Yan
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaohua Huang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dandan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhen Li
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lun Hua
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yong Zhuo
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Zhengfeng Fang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lianqiang Che
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Lin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Shengyu Xu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Chao Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yuanfeng Zou
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Lixia Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - De Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
39
|
Popiolek-Barczyk K, Ciechanowska A, Ciapała K, Pawlik K, Oggioni M, Mercurio D, De Simoni MG, Mika J. The CCL2/CCL7/CCL12/CCR2 pathway is substantially and persistently upregulated in mice after traumatic brain injury, and CCL2 modulates the complement system in microglia. Mol Cell Probes 2020; 54:101671. [PMID: 33160071 DOI: 10.1016/j.mcp.2020.101671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/15/2020] [Accepted: 11/01/2020] [Indexed: 12/28/2022]
Abstract
Traumatic brain injury (TBI) is the leading cause of death in the global population. Disturbed inflammatory processes after TBI exacerbate secondary brain injury and contribute to unfavorable outcomes. Multiple inflammatory events that accompany brain trauma, such as glial activation, chemokine release, or the initiation of the complement system cascade, have been identified as potential targets for TBI treatment. However, the participation of chemokines in the complement activation remains unknown. Our studies sought to determine the changes in the expression of the molecules involved in the CCL2/CCL7/CCL12/CCR2 pathway in the injured brain and the effect of CCL2, CCL7, and CCL12 (10, 100, and 500 ng/mL) on the classic and lectin complement pathways and inflammatory factors in microglial cell cultures. Brain injury in mice was modeled by controlled cortical impact (CCI). Our findings indicate a time-dependent upregulation of CCL2, CCL7, and CCL12 at the mRNA and protein levels within the cortex, striatum, and/or thalamus beginning 24 h after the trauma. The analysis of the expression of the receptor of the tested chemokines, CCR2, revealed its substantial upregulation within the injured brain areas mainly on the mRNA level. Using primary cortical microglial cell cultures, we observed a substantial increase in the expression of CCL2, CCL7, and CCL12 after 24 h of LPS (100 ng/mL) treatment. CCL2 stimulation of microglia increased the level of IL-1β mRNA but did not influence the expression of IL-18, IL-6, and IL-10. Moreover, CCL2 significantly increased the expression of Iba1, a marker of microglia activation. CCL2 and CCL12 upregulated the expression of C1qa but did not influence the expression of C1ra and C1s1 (classical pathway); moreover, CCL2 increased ficolin A expression and reduced collectin 11 expression (lectin pathway). Additionally, we observed the downregulation of pentraxin 3, a modulator of the complement cascade, after CCL2 and CCL12 treatment. We did not detect the expression of ficolin B, Mbl1, and Mbl2 in microglial cells. Our data identify CCL2 as a modulator of the classical and lectin complement pathways suggesting that CCL2 may be a promising target for pharmacological intervention after brain injury. Moreover, our study provides evidence that CCL2 and two other CCR2 ligands may play a role in the development of changes in TBI.
Collapse
Affiliation(s)
- Katarzyna Popiolek-Barczyk
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Agata Ciechanowska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Ciapała
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Katarzyna Pawlik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland
| | - Marco Oggioni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Domenico Mercurio
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Joanna Mika
- Maj Institute of Pharmacology, Polish Academy of Sciences, Department of Pain Pharmacology, 12 Smetna Str, 31-343, Krakow, Poland.
| |
Collapse
|
40
|
Kasemsuk T, Phuagkhaopong S, Yubolphan R, Rungreangplangkool N, Vivithanaporn P. Cadmium induces CCL2 production in glioblastoma cells via activation of MAPK, PI3K, and PKC pathways. J Immunotoxicol 2020; 17:186-193. [DOI: 10.1080/1547691x.2020.1829211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Thitima Kasemsuk
- Division of Pharmacology, Faculty of Pharmaceutical Sciences, Burapha University, Chonburi, Thailand
| | - Suttinee Phuagkhaopong
- Pharmacology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ruedeemars Yubolphan
- Pharmacology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| |
Collapse
|
41
|
Shin MR, Park HJ, Seo BI, Roh SS. New approach of medicinal herbs and sulfasalazine mixture on ulcerative colitis induced by dextran sodium sulfate. World J Gastroenterol 2020; 26:5272-5286. [PMID: 32994687 PMCID: PMC7504242 DOI: 10.3748/wjg.v26.i35.5272] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/29/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sulfasalazine has been used as a standard-of-care in ulcerative colitis for decades, however, it results in severe adverse symptoms, such as hepatotoxicity, blood disorders, male infertility, and hypospermia. Accordingly, the new treatment strategy has to enhance pharmacological efficacy and stimultaneously minimize side effects.
AIM To compare the anti-inflammatory action of sulfasalazine alone or in combination with herbal medicine for ulcerative colitis in a dextran sodium sulfate (DSS)-induced colitis mouse model.
METHODS To induce ulcerative colitis, mice received 5% DSS in drinking water for 7 d. Animals were divided into five groups (n = 9 each) for use as normal (non-DSS), DSS controls, DSS + sulfasalazine (30 mg/kg)-treatment experimentals, DSS + sulfasalazine (60 mg/kg)-treatment experimentals, DSS + sulfasalazine (30 mg/kg) + Citrus unshiu peel and Bupleuri radix mixture (30 mg/kg) (SCPB)-treatment experimentals.
RESULTS The SCPB treatment showed an outstanding effectiveness in counteracting the ulcerative colitis, as evidenced by reduction in body weight, improvement in crypt morphology, increase in antioxidant defenses, down-regulation of proinflammatory proteins and cytokines, and inhibition of proteins related to apoptosis.
CONCLUSION SCPB may represent a promising alternative therapeutic against ulcerative colitis, without inducing adverse effects.
Collapse
Affiliation(s)
- Mi-Rae Shin
- Department of Herbology, Korean Medicine College, Daegu Haany University, Suseong-gu, Deagu 42158, South Korea
| | - Hae-Jin Park
- DHU Bio Convergence Testing Center, Gyeongsan-si, Gyeongsangbuk-do 38610, South Korea
| | - Bu-Il Seo
- Department of Herbology, Korean Medicine College, Daegu Haany University, Suseong-gu, Deagu 42158, South Korea
| | - Seong-Soo Roh
- Department of Herbology, Korean Medicine College, Daegu Haany University, Suseong-gu, Deagu 42158, South Korea
| |
Collapse
|
42
|
Cellular Mechanisms of Rejection of Optic and Sciatic Nerve Transplants: An Observational Study. Transplant Direct 2020; 6:e589. [PMID: 32766437 PMCID: PMC7382554 DOI: 10.1097/txd.0000000000001012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background. Organ transplantation is a standard therapeutic strategy for irreversible organ damage, but the utility of nerve transplantation remains generally unexplored, despite its potential benefit to a large patient population. Here, we aimed to establish a feasible preclinical mouse model for understanding the cellular mechanisms behind the rejection of peripheral and optic nerves. Methods. We performed syngenic and allogenic transplantation of optic and sciatic nerves in mice by inserting the nerve grafts inside the kidney capsule, and we assessed the allografts for signs of rejection through 14 d following transplantation. Then, we assessed the efficacy of CTLA4 Ig, Rapamycin, and anti-CD3 antibody in suppressing immune cell infiltration of the nerve allografts. Results. By 3 d posttransplantation, both sciatic and optic nerves transplanted from BALB/c mice into C57BL/6J recipients contained immune cell infiltrates, which included more CD11b+ macrophages than CD3+ T cells or B220+ B cells. Ex vivo immunogenicity assays demonstrated that sciatic nerves demonstrated higher alloreactivity in comparison with optic nerves. Interestingly, optic nerves contained higher populations of anti-inflammatory PD-L1+ cells than sciatic nerves. Treatment with anti-CD3 antibody reduced immune cell infiltrates in the optic nerve allograft, but exerted no significant effect in the sciatic nerve allograft. Conclusions. These findings establish the feasibility of a preclinical allogenic nerve transplantation model and provide the basis for future testing of directed, high-intensity immunosuppression in these mice.
Collapse
|
43
|
Shukla M, Mani KV, Deepshikha, Shukla S, Kapoor N. Moderate noise associated oxidative stress with concomitant memory impairment, neuro-inflammation and neurodegeneration. Brain Behav Immun Health 2020; 5:100089. [PMID: 34589861 PMCID: PMC8474184 DOI: 10.1016/j.bbih.2020.100089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 05/24/2020] [Accepted: 05/29/2020] [Indexed: 01/16/2023] Open
Abstract
Noise, a disturbing and unwanted sound is currently being perceived as a widespread environmental stressor. In the present study we investigated the activation of oxidative stress as a mechanism involved in cognitive impairment through changes in neuro-inflammation. Sprague Dawley rats (200-220 g m) were exposed to moderate (100dB) sound pressure level (SPL) noise daily for 2 h s over a period of 15 and 30 days and the consequence on brain regions of hippocampus observed through behavioral studies by Morris Water Maze to assess effects on spatial memory coupled with biochemical evaluation of markers of oxidative stress and inflammation. Further, the underlying mechanism pertaining to apoptosis was investigated by immuno-histological studies through assessment of Caspase-3 and TUNEL assay as well as morphological parameters, namely Nissl bodies in CA1, CA3 and DG regions of hippocampus. Poorer performance in the MWM indicative of decrement in concept formation, attention, working memory, and reference memory was observed on 15 and 30 days of noise exposures. At the cellular level, increased oxidative stress and inflammation was noticed as evinced by elevated levels of TNF-α, IL-6, IL-1α and IFN-γ in both hippocampus and plasma. Exposure to noise also led to a gradual increase in the number of pyknotic and apoptotic neurons together with the increase in DNA fragmentation in hippocampus. Increased levels of inflammatory genes (i.g.) ccl2, ccr5, ifng, il13, il1a, tnfa coupled with decreased levels of bmp2 and il3 genes were found in both the noise exposure groups. Our findings revealed that moderate intensity noise exposure impaired early memory changes in expression of several gene families including genes associated with regulation of transcription, inflammatory response, and, response to oxidative stress.
Collapse
Affiliation(s)
- Manish Shukla
- Occupational Health Division, Defence Institute of Physiology & Allied Sciences (DIPAS), DRDO Lucknow Road, Timarpur, Delhi, India
| | - Kumar Vyonkesh Mani
- Occupational Health Division, Defence Institute of Physiology & Allied Sciences (DIPAS), DRDO Lucknow Road, Timarpur, Delhi, India
| | - Deepshikha
- Occupational Health Division, Defence Institute of Physiology & Allied Sciences (DIPAS), DRDO Lucknow Road, Timarpur, Delhi, India
| | - Sangeeta Shukla
- School of Studies in Zoology, Jiwaji University, Gwalior, M.P, India
| | - Neeru Kapoor
- Occupational Health Division, Defence Institute of Physiology & Allied Sciences (DIPAS), DRDO Lucknow Road, Timarpur, Delhi, India
| |
Collapse
|
44
|
Harb Z, Deckert V, Bressenot AM, Christov C, Guéant-Rodriguez RM, Raso J, Alberto JM, de Barros JPP, Umoret R, Peyrin-Biroulet L, Lagrost L, Bronowicki JP, Guéant JL. The deficit in folate and vitamin B12 triggers liver macrovesicular steatosis and inflammation in rats with dextran sodium sulfate-induced colitis. J Nutr Biochem 2020; 84:108415. [PMID: 32645655 DOI: 10.1016/j.jnutbio.2020.108415] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/21/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
The risks of nonalcoholic steatohepatitis (NASH) and deficiency in vitamin B12 and folate (methyl donor deficiency, MDD) are increased in inflammatory bowel disease (IBD). We investigated the influence of MDD on NASH in rats with DSS-induced colitis. Two-month-old male Wistar rats were subjected to MDD diet and/or ingestion of DSS and compared to control animals. We studied steatosis, inflammation, fibrosis, plasma levels of metabolic markers, cytokines and lipopolysaccharide, and inflammatory pathways in liver. MDD triggered a severe macrovesicular steatosis with inflammation in DSS animals that was not observed in animals subjected to DSS or MDD only. The macrovesicular steatosis was closely correlated to folate, vitamin B12, homocysteine plasma level and liver S-adenosyl methionine/S-adenosyl homocysteine (SAM/SAH) ratio. Liver inflammation was evidenced by activation of nuclear factor kappa B (NFκB) pathway and nuclear translocation of NFκB phospho-p65. MDD worsened the increase of interleukin 1-beta (IL-1β) and abolished the increase of IL10 produced by DSS colitis. It increased monocyte chemoattractant protein 1 (MCP-1). MDD triggers liver macrovesicular steatosis and inflammation through imbalanced expression of IL-1β vs. IL10 and increase of MCP-1 in DSS colitis. Our results suggest evaluating whether IBD patients with MDD and increase of MCP-1 are at higher risk of NASH.
Collapse
Affiliation(s)
- Zeinab Harb
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Valérie Deckert
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Aude Marchal Bressenot
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Anatomo-Pathology, Robert Debré University Hospital, Reims
| | - Christo Christov
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Biochemical and Molecular biology lab, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Jérémie Raso
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Jean Marc Alberto
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Jean-Paul Pais de Barros
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Remy Umoret
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Laurent Peyrin-Biroulet
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Laurent Lagrost
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Jean-Pierre Bronowicki
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Jean-Louis Guéant
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Biochemical and Molecular biology lab, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France.
| |
Collapse
|
45
|
Takizawa T, Qin T, Lopes de Morais A, Sugimoto K, Chung JY, Morsett L, Mulder I, Fischer P, Suzuki T, Anzabi M, Böhm M, Qu WS, Yanagisawa T, Hickman S, Khoury JE, Whalen MJ, Harriott AM, Chung DY, Ayata C. Non-invasively triggered spreading depolarizations induce a rapid pro-inflammatory response in cerebral cortex. J Cereb Blood Flow Metab 2020; 40:1117-1131. [PMID: 31242047 PMCID: PMC7181092 DOI: 10.1177/0271678x19859381] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cortical spreading depolarization (CSD) induces pro-inflammatory gene expression in brain tissue. However, previous studies assessing the relationship between CSD and inflammation have used invasive methods that directly trigger inflammation. To eliminate the injury confounder, we induced CSDs non-invasively through intact skull using optogenetics in Thy1-channelrhodopsin-2 transgenic mice. We corroborated our findings by minimally invasive KCl-induced CSDs through thinned skull. Six CSDs induced over 1 h dramatically increased cortical interleukin-1β (IL-1β), chemokine (C-C motif) ligand 2 (CCL2), and tumor necrosis factor-α (TNF-α) mRNA expression peaking around 1, 2 and 4 h, respectively. Interleukin-6 (IL-6) and intercellular adhesion molecule-1 (ICAM-1) were only modestly elevated. A single CSD also increased IL-1β, CCL2, and TNF-α, and revealed an ultra-early IL-1β response within 10 min. The response was blunted in IL-1 receptor-1 knockout mice, implicating IL-1β as an upstream mediator, and suppressed by dexamethasone, but not ibuprofen. CSD did not alter systemic inflammatory indices. In summary, this is the first report of pro-inflammatory gene expression after non-invasively induced CSDs. Altogether, our data provide novel insights into the role of CSD-induced neuroinflammation in migraine headache pathogenesis and have implications for the inflammatory processes in acute brain injury where numerous CSDs occur for days.
Collapse
Affiliation(s)
- Tsubasa Takizawa
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Tao Qin
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Andreia Lopes de Morais
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Kazutaka Sugimoto
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Joon Yong Chung
- Neuroscience Center, Massachusetts
General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pediatrics, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Liza Morsett
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Inge Mulder
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Paul Fischer
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Charité –
Universitätsmedizin Berlin, Berlin, Germany
| | - Tomoaki Suzuki
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Maryam Anzabi
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Maximilian Böhm
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Charité –
Universitätsmedizin Berlin, Berlin, Germany
| | - Wen-sheng Qu
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Takeshi Yanagisawa
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
| | - Suzanne Hickman
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Joseph El Khoury
- Center for Immunology & Inflammatory
Diseases, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA,
USA
| | - Michael J Whalen
- Neuroscience Center, Massachusetts
General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Pediatrics, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea M Harriott
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - David Y Chung
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cenk Ayata
- Neurovascular Research Laboratory,
Department of Radiology, Massachusetts General Hospital, Harvard Medical School,
Charlestown, MA, USA
- Department of Neurology, Massachusetts
General Hospital, Harvard Medical School, Boston, MA, USA
- Cenk Ayata, Massachusetts General Hospital,
149 13th Street, 6403, Charlestown, MA 02129, USA.
| |
Collapse
|
46
|
Martyniuk CJ, Martínez R, Kostyniuk DJ, Mennigen JA, Zubcevic J. Genetic ablation of bone marrow beta-adrenergic receptors in mice modulates miRNA-transcriptome networks of neuroinflammation in the paraventricular nucleus. Physiol Genomics 2020; 52:169-177. [PMID: 32089076 PMCID: PMC7191424 DOI: 10.1152/physiolgenomics.00001.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/07/2020] [Accepted: 02/16/2020] [Indexed: 12/21/2022] Open
Abstract
Elucidating molecular pathways regulating neuroimmune communication is critical for therapeutic interventions in conditions characterized by overactive immune responses and dysfunctional autonomic nervous system. We generated a bone marrow-specific adrenergic beta 1 and beta 2 knockout mouse chimera (AdrB1.B2 KO) to determine how sympathetic drive to the bone affects transcripts and miRNAs in the hypothalamic paraventricular nucleus (PVN). This model has previously exhibited a dampened systemic immune response and decreased blood pressure compared with control animals. Reduced sympathetic responsiveness of the bone marrow hematopoietic cells of AdrB1.B2 KO chimera led to suppression of transcriptional networks that included leukocyte cell adhesion and migration and T cell-activation and recruitment. Transcriptome responses related to IL-17a signaling and the renin-angiotensin system were also suppressed in the PVN. Based on the transcriptome response, we next computationally predicted miRNAs in the PVN that may underscore the reduced sympathetic responsiveness of the bone marrow cells. These included miR-27b-3p, miR-150, miR-223-3p, and miR-326. Using real-time PCR, we measured a downregulation in the expression of miR-150-5p, miR-205-5p, miR-223-3p, miR-375-5p, miR-499a-5p, miR-27b-3p, let-7a-5p, and miR-21a-5p in the PVN of AdrB1.B2 KO chimera, confirming computational predictions that these miRNAs are associated with reduced neuro-immune responses and the loss of sympathetic responsiveness in the bone marrow. Intriguingly, directional responses of the miRNA corresponded to mRNAs, suggesting complex temporal or circuit-dependent posttranscriptional control of gene expression in the PVN. This study identifies molecular pathways involved in neural-immune interactions that may act as targets of therapeutic intervention for a dysfunctional autonomic nervous system.
Collapse
Affiliation(s)
- Christopher J Martyniuk
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| | - Ruben Martínez
- Department of Environmental Chemistry, Institute of Environmental Assessment and Water Research, IDAEA-CSIC, Jordi Girona, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universidad de Barcelona (UB), Barcelona, Spain
| | | | - Jan A Mennigen
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
47
|
Mul Fedele ML, Aiello I, Caldart CS, Golombek DA, Marpegan L, Paladino N. Differential Thermoregulatory and Inflammatory Patterns in the Circadian Response to LPS-Induced Septic Shock. Front Cell Infect Microbiol 2020; 10:100. [PMID: 32226779 PMCID: PMC7080817 DOI: 10.3389/fcimb.2020.00100] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis is caused by a dysregulated host response to infection, and characterized by uncontrolled inflammation together with immunosuppression, impaired innate immune functions of phagocytes and complement activation. Septic patients develop fever or hypothermia, being the last one characteristic of severe cases. Both lipopolysaccharide (LPS) and Tumor Necrosis Factor (TNF)-α- induced septic shock in mice is dependent on the time of administration. In this study, we aimed to further characterize the circadian response to high doses of LPS. First, we found that mice injected with LPS at ZT11 developed a higher hypothermia than those inoculated at ZT19. This response was accompanied by higher neuronal activation of the preoptic, suprachiasmatic, and paraventricular nuclei of the hypothalamus. However, LPS-induced Tnf-α and Tnf-α type 1 receptor (TNFR1) expression in the preoptic area was time-independent. We also analyzed peritoneal and spleen macrophages, and observed an exacerbated response after ZT11 stimulation. The serum of mice inoculated with LPS at ZT11 induced deeper hypothermia in naïve animals than the one coming from ZT19-inoculated mice, related to higher TNF-α serum levels during the day. We also analyzed the response in TNFR1-deficient mice, and found that both the daily difference in the mortality rate, the hypothermic response and neuronal activation were lost. Moreover, mice subjected to circadian desynchronization showed no differences in the mortality rate throughout the day, and developed lower minimum temperatures than mice under light-dark conditions. Also, those injected at ZT11 showed increased levels of TNF-α in serum compared to standard light conditions. These results suggest a circadian dependency of the central thermoregulatory and peripheral inflammatory response to septic-shock, with TNF-α playing a central role in this circadian response.
Collapse
Affiliation(s)
- Malena Lis Mul Fedele
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| | - Ignacio Aiello
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| | - Carlos Sebastián Caldart
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| | - Diego Andrés Golombek
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| | - Luciano Marpegan
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| | - Natalia Paladino
- Laboratorio de Cronobiología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes/CONICET, Buenos Aires, Argentina
| |
Collapse
|
48
|
Li M, Hu FC, Qiao F, Du ZY, Zhang ML. Sodium acetate alleviated high-carbohydrate induced intestinal inflammation by suppressing MAPK and NF-κB signaling pathways in Nile tilapia (Oreochromis niloticus). FISH & SHELLFISH IMMUNOLOGY 2020; 98:758-765. [PMID: 31730927 DOI: 10.1016/j.fsi.2019.11.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 05/20/2023]
Abstract
With the development of aquaculture industry, high-carbohydrate diet is used to stimulate protein-sparing effect and reduce feed cost. However, fish utilize carbohydrates poorly in general, and instead, high level of carbohydrates in the diet influence the growth condition of fish. How to alleviate the side effects of high carbohydrate diet on fish health has attracted more and more attentions. In the present study, Nile tilapia (Oreochromis niloticus) were fed with 25% and 45% of carbohydrate diet for eight weeks. Higher body weight but lower resistance to pathogen was found in 45% carbohydrate diet group. Higher expression level of inflammation cytokines, increased expression of total NF-κB protein and phosphorylated NF-κB protein (p-NF-κB) were detected in higher carbohydrate group. Concentration of short-chain fatty acids (SCFAs) was measured and the results indicated that high-carbohydrate diet decreased acetate content in the intestine. In order to detect the relationship between the decreased concentration of acetate and lower resistance to pathogen in high-carbohydrate group, 45% of carbohydrate diets (HC) supplemented with different concentrations of sodium acetate (HC + LA, 100 mmol/L; HC + MA, 200 mmol/L; HC + HA, 400 mmol/L) were used to raise Nile Tilapia for eight weeks. The results indicated that addition of 200 mmol/L sodium acetate (HC + MA) reduced the mortality when fish were challenged with Aeromonas hydrophila. Furthermore, we also found that addition of 200 mmol/L sodium acetate mainly inhibited p38 mitogen-activated protein kinase (p38MAPK) and NF-κB phosphorylation to decrease the expression level of inflammation cytokines (IL-8, IL-12, TNF-α and IL-1β) in the intestine. The present study indicated that certain concentration of sodium acetate could alleviate high-carbohydrate induced intestinal inflammation mainly by suppressing MAPK activation and NF-κB phosphorylation.
Collapse
Affiliation(s)
- Miao Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang-Chao Hu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), College of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
49
|
Liu H, Osterburg AR, Flury J, Swank Z, McGraw DW, Gupta N, Wikenheiser-Brokamp KA, Kumar A, Tazi A, Inoue Y, Hirose M, McCormack FX, Borchers MT. MAPK mutations and cigarette smoke promote the pathogenesis of pulmonary Langerhans cell histiocytosis. JCI Insight 2020; 5:132048. [PMID: 31961828 DOI: 10.1172/jci.insight.132048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
Pulmonary Langerhans cell histiocytosis (PLCH) is a rare smoking-related lung disease characterized by dendritic cell (DC) accumulation, bronchiolocentric nodule formation, and cystic lung remodeling. Approximately 50% of patients with PLCH harbor somatic BRAF-V600E mutations in cells of the myeloid/monocyte lineage. However, the rarity of the disease and lack of animal models have impeded the study of PLCH pathogenesis. Here, we establish a cigarette smoke-exposed (CS-exposed) BRAF-V600E-mutant mouse model that recapitulates many hallmark characteristics of PLCH. We show that CD11c-targeted expression of BRAF-V600E increases DC responsiveness to stimuli, including the chemokine CCL20, and that mutant cell accumulation in the lungs of CS-exposed mice is due to both increased cellular viability and enhanced recruitment. Moreover, we report that the chemokine CCL7 is secreted from DCs and human peripheral blood monocytes in a BRAF-V600E-dependent manner, suggesting a possible mechanism for recruitment of cells known to dominate PLCH lesions. Inflammatory lesions and airspace dilation in BRAF-V600E mice in response to CS are attenuated by transitioning animals to filtered air and treatment with a BRAF-V600E inhibitor, PLX4720. Collectively, this model provides mechanistic insights into the role of myelomonocytic cells and the BRAF-V600E mutation and CS exposure in PLCH pathogenesis and provides a platform to develop biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Huan Liu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Andrew R Osterburg
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jennifer Flury
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Zulma Swank
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Dennis W McGraw
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Nishant Gupta
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Pathology and Laboratory Medicine and.,Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ashish Kumar
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Abdellatif Tazi
- INSERM UMR-S 976, University Paris-Diderot, Sorbonne Paris Cité, Paris, France
| | - Yoshikazu Inoue
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Masaki Hirose
- National Hospital Organization Kinki-Chuo Chest Medical Center, Osaka, Japan
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| | - Michael T Borchers
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Cincinnati Veterans Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
50
|
Improvement of Inflammation through Antioxidant Pathway of Gardeniae Fructus 50% EtOH Extract (GE) from Acute Reflux Esophagitis Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4826176. [PMID: 32185206 PMCID: PMC7060875 DOI: 10.1155/2020/4826176] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
Gardeniae Fructus 50% EtOH extract (GE) is a traditional herb that has been used to treat a variety of diseases. In this study, we investigate the antioxidant, anti-inflammatory, and antiapoptotic properties of GE on acute reflux-induced esophagitis (RE) model in rats. 2,2′-Azino-bis (3-ethylbenzothiazolin-6-sulfonic acid) (ABTS) and 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging assays were performed to determine the antioxidant activity of GE. GE was given orally at 50 and 100 mg/kg body weight 1h 30 min prior to RE induction. And its effect was assessed in comparison with RE control and normal groups. The administration of the extract of the GE showed remarkable protection of mucosal damage in esophageal tissue, and the histologic observation showed that the gastric lesion was improved. Increased reactive oxygen species (ROS) levels in the serum were diminished by GE treatment. The antioxidative biomarkers including nuclear factor-erythroid 2-related factor 2 (Nrf-2), heme oxygenase-1 (HO-1), superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX) were significantly increased. GE administration significantly reduced the inflammatory protein expression through MAPK-related signaling pathways and the nuclear factor-kappa B (NF-κB) pathway. These results suggest that GE protects the esophagus mucosal membrane by attenuating oxidative stress and inflammatory response under reflux esophagitis condition through the antioxidant pathway. Therefore, it is suggested that GE may be a potential remedy for the treatment of reflux esophagitis.
Collapse
|