1
|
Lu ZJ, Pan QL, Lin FX. Epigenetic modifications of inflammation in spinal cord injury. Biomed Pharmacother 2024; 179:117306. [PMID: 39153436 DOI: 10.1016/j.biopha.2024.117306] [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: 05/19/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Spinal cord injury (SCI) is a central nervous system injury that leads to neurological dysfunction or paralysis, which seriously affects patients' quality of life and causes a heavy social and economic burden. The pathological mechanism of SCI has not been fully revealed, resulting in unsatisfactory clinical treatment. Therefore, more research is urgently needed to reveal its precise pathological mechanism. Numerous studies have shown that inflammation is closely related to various pathological processes in SCI. Inflammatory response is an important pathological process leading to secondary injury, and sustained inflammatory response can exacerbate the injury and hinder the recovery of neurological function after injury. Epigenetic modification is considered to be an important regulatory mechanism in the pathological process of many diseases. Epigenetic modification mainly affects the function and characteristics of genes through the reversibility of mechanisms such as DNA methylation, histone modification, and regulation of non-coding RNA, thus having a significant impact on the pathological process of diseases and the survival state of the body. Recently, the role of epigenetic modification in the inflammatory response of SCI has gradually entered the field of view of researchers, and epigenetic modification may be a potential means to treat SCI. In this paper, we review the effects and mechanisms of different types of epigenetic modifications (including histone modifications, DNA methylation, and non-coding RNAs) on post-SCI inflammation and their potential therapeutic effects on inflammation to improve our understanding of the secondary SCI stage. This review aims to help identify new markers, signaling pathways and targeted drugs, and provide theoretical basis and new strategies for the diagnosis and treatment of SCI.
Collapse
Affiliation(s)
- Zhi-Jun Lu
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| | - Qi-Lin Pan
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China
| | - Fei-Xiang Lin
- Department of Spine Surgery, Ganzhou People's Hospital, 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China; Department of Spine Surgery, The Affiliated Ganzhou Hospital of Nanchang University (Ganzhou Hospital-Nanfang Hospital, Southern Medical University), 16 Meiguan Avenue, Ganzhou, Jiangxi Province 341000, PR China.
| |
Collapse
|
2
|
KDM6B promotes gastric carcinogenesis and metastasis via upregulation of CXCR4 expression. Cell Death Dis 2022; 13:1068. [PMID: 36564369 PMCID: PMC9789124 DOI: 10.1038/s41419-022-05458-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 12/24/2022]
Abstract
KDM6B (Lysine-specific demethylase 6B) is a histone lysine demethyltransferase that plays a key role in many types of cancers. However, its potential role in gastric cancer (GC) remains unclear. Here, we focused on the clinical significance and potential role of KDM6B in GC. We found that the KDM6B expression is upregulated in GC tissues and that its high expression in patients is related to poor prognosis. KDM6B ectopic expression promotes GC cells' proliferation and metastasis, while its inhibition has opposite effects in vitro and in vivo. Mechanistically, KDM6B promotes GC cells proliferation and metastasis through its enzymatic activity through the induction of H3K27me3 demethylation near the CXCR4 (C-X-C chemokine receptor type 4) promoter region, resulting in the upregulation of CXCR4 expression. Furthermore, H. pylori was found to induce KDM6B expression. In conclusion, our results suggest that KDM6B is aberrantly expressed in GC and plays a key role in gastric carcinogenesis and metastasis through CXCR4 upregulation. Our work also suggests that KDM6B may be a potential oncogenic factor and a therapeutic target for GC.
Collapse
|
3
|
Qiu M, Xu E, Zhan L. Epigenetic Regulations of Microglia/Macrophage Polarization in Ischemic Stroke. Front Mol Neurosci 2021; 14:697416. [PMID: 34707480 PMCID: PMC8542724 DOI: 10.3389/fnmol.2021.697416] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. Microglia/macrophages (MMs)-mediated neuroinflammation contributes significantly to the pathological process of ischemic brain injury. Microglia, serving as resident innate immune cells in the central nervous system, undergo pro-inflammatory phenotype or anti-inflammatory phenotype in response to the microenvironmental changes after cerebral ischemia. Emerging evidence suggests that epigenetics modifications, reversible modifications of the phenotype without changing the DNA sequence, could play a pivotal role in regulation of MM polarization. However, the knowledge of the mechanism of epigenetic regulations of MM polarization after cerebral ischemia is still limited. In this review, we present the recent advances in the mechanisms of epigenetics involved in regulating MM polarization, including histone modification, non-coding RNA, and DNA methylation. In addition, we discuss the potential of epigenetic-mediated MM polarization as diagnostic and therapeutic targets for ischemic stroke. It is valuable to identify the underlying mechanisms between epigenetics and MM polarization, which may provide a promising treatment strategy for neuronal damage after cerebral ischemia.
Collapse
Affiliation(s)
- Meiqian Qiu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - En Xu
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neurosciences and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China
| |
Collapse
|
4
|
Wrede AH, McNamara MC, Baldwin R, Luo J, Montazami R, Kanthasamy A, Hashemi NN. Characterization of Astrocytic Response after Experiencing Cavitation In Vitro. GLOBAL CHALLENGES (HOBOKEN, NJ) 2020; 4:1900014. [PMID: 32642072 PMCID: PMC7330500 DOI: 10.1002/gch2.201900014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/06/2020] [Indexed: 06/11/2023]
Abstract
When a traumatic brain injury (TBI) occurs, low-pressure regions inside the skull can cause vapor contents in the cerebral spinal fluid (CSF) to expand and collapse, a phenomenon known as cavitation. When these microbubbles (MBs) collapse, shock waves are radiated outward and are known to damage surrounding materials in other applications, like the steel foundation of boat propellers, so it is alarming to realize the damage that cavitation inflicts on vulnerable brain tissue. Using cell-laden microfibers, the longitudinal morphological response that mouse astrocytes have to surrounding cavitation in vitro is visually analyzed. Astrocytic damage is evident immediately after cavitation when compared to a control sample, as their processes retract. Forty-eight hours later, the astrocytes appeared to spread across the fibers, as normal. This study also analyzes the gene expression changes that occur post-cavitation via quantitative polymerase chain reaction (qPCR) methods. After cavitation a number of pro-inflammatory genes are upregulated, including TNFα, IL-1β, C1q, Serping1, NOS1, IL-6, and JMJD3. Taken together, these results confirm that surrounding cavitation is detrimental to astrocytic function, and yield opportunities to further the understanding of how protective headgear can minimize or eliminate the occurrence of cavitation.
Collapse
Affiliation(s)
- Alex H. Wrede
- Department of Mechanical EngineeringIowa State UniversityAmesIA50011USA
| | | | - Rodger Baldwin
- Department of Mechanical EngineeringIowa State UniversityAmesIA50011USA
| | - Jie Luo
- Department of Biomedical SciencesIowa State UniversityAmesIA50011USA
| | - Reza Montazami
- Department of Mechanical EngineeringIowa State UniversityAmesIA50011USA
| | | | - Nicole N. Hashemi
- Department of Mechanical EngineeringIowa State UniversityAmesIA50011USA
- Department of Biomedical SciencesIowa State UniversityAmesIA50011USA
| |
Collapse
|
5
|
Zhao XN, Bai ZZ, Li CH, Sheng CL, Li HY. The NK-1R Antagonist Aprepitant Prevents LPS-Induced Oxidative Stress and Inflammation in RAW264.7 Macrophages. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1943-1952. [PMID: 32546961 PMCID: PMC7246327 DOI: 10.2147/dddt.s244099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
Abstract
Background The macrophage is one of the most important types of immune cells that protect against harmful stimuli. Macrophage activation plays a pivotal role in the progression and development of various inflammatory diseases. The neurokinin 1 receptor (NK-1R) is a G protein-coupled receptor that plays an important role in inflammatory diseases. Aprepitant is a kind of NK-1R antagonist. The purpose of this study is to determine the protective effect of aprepitant in lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Methods We examined the anti-inflammatory and anti-oxidant effects of aprepitant in LPS-treated RAW264.7 macrophages by using real-time PCR, ELISA, and Western blot analysis. We also assessed cellular oxidative stress signaling by measuring the levels of cellular MDA, total ROS, and NADPH oxidase expression. Cellular NO production was measured by DAF-FM DA staining. The inhibitory effect of aprepitant against NF-κB signaling was evaluated by luciferase assay and Western blot analysis. Results The expression of NK-1R is increased in LPS-induced macrophages, suggesting a potential role of the receptor in the inflammatory response. We show that aprepitant protects macrophages against oxidative stress by reducing the generation of ROS and the expression of NOX-4. Furthermore, aprepitant inhibits the secretion of pro-inflammatory cytokines and chemotactic factors by mediating the NF-κB signaling pathway. Conclusion The NK-1R receptor antagonist aprepitant acts as an anti-inflammatory agent, indicating that the blockage of the NK-1R pathway in macrophages has the potential to suppress inflammation.
Collapse
Affiliation(s)
- Xiao-Nan Zhao
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Zhen-Zi Bai
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Cheng-Hua Li
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Chuan-Lun Sheng
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| | - Hong-Yan Li
- Department of Infectious, The Third Hospital of Jilin University, Changchun City, Jilin Province 130033, People's Republic of China
| |
Collapse
|
6
|
Ye Y, Wang Y, Yang Y, Tao L. Aloperine suppresses LPS-induced macrophage activation through inhibiting the TLR4/NF-κB pathway. Inflamm Res 2020; 69:375-383. [PMID: 32144444 DOI: 10.1007/s00011-019-01313-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/13/2019] [Accepted: 12/26/2019] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE The currently available anti-inflammatory drugs often cause diverse side effects with long-term use. Exploring anti-inflammatory drugs with better efficacy and lower toxicity presents an ongoing challenge. Aloperine is an alkaloid extracted from the leaves and seeds of Sophora alopecuroides L. However, the anti-inflammatory effects of Aloperine have not been fully elucidated. This study aimed to investigate whether Aloperine suppresses lipopolysaccharide (LPS)-induced inflammatory responses in RAW264.7 macrophages. METHODS RAW264.7 macrophages were stimulated with LPS (1 μg/mL) in the presence or absence of Aloperine (50 and 100 μM). mRNA expression was measured by real-time PCR, and protein expression was assessed by western blot analysis. The secretion of pro-inflammatory cytokines was measured by ELISA. The levels of nitric oxide (NO) and reactive oxygen species (ROS) were measured by staining. The transcriptional activity of NF-κB was assayed by a luciferase activity assay. RESULTS The results proved that Aloperine inhibited the expression of LPS-induced pro-inflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-17A (IL-17A)] in macrophages. Treatment with Aloperine inhibited NO production through suppressing inducible nitric oxide synthase (iNOS) expression and the secretion of prostaglandin E2 (PGE2) by inhibiting cyclooxygenase 2 (COX-2) expression. Aloperine prevented LPS-induced oxidative stress by reducing the generation of ROS. Furthermore, aloperine significantly reduced Toll-like receptor 4 (TLR4) and myeloid differentiation factor (Myd-88) levels and prevented the nuclear translocation of nuclear factor-κB (NF-κB) in LPS-treated macrophages. CONCLUSION Taken together, our findings show that Aloperine could suppress LPS-induced macrophage activation by inhibiting the TLR4/Myd-88/NF-κB pathway.
Collapse
Affiliation(s)
- Yinyin Ye
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China.
| | - Yuwei Wang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China
| | - Yanlang Yang
- Department of Nephrology, Yijishan Hospital of Wannan Medical College, # 92 Zheshan Road, Wuhu, 241002, Anhui Province, People's Republic of China
| | - Liangfei Tao
- Medical Emergency Center, #158 Yueming Road, Baoshan District, Shanghai, 201900, People's Republic of China.
| |
Collapse
|
7
|
Zhang X, Liu L, Yuan X, Wei Y, Wei X. JMJD3 in the regulation of human diseases. Protein Cell 2019; 10:864-882. [PMID: 31701394 PMCID: PMC6881266 DOI: 10.1007/s13238-019-0653-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
In recent years, many studies have shown that histone methylation plays an important role in maintaining the active and silent state of gene expression in human diseases. The Jumonji domain-containing protein D3 (JMJD3), specifically demethylate di- and trimethyl-lysine 27 on histone H3 (H3K27me2/3), has been widely studied in immune diseases, infectious diseases, cancer, developmental diseases, and aging related diseases. We will focus on the recent advances of JMJD3 function in human diseases, and looks ahead to the future of JMJD3 gene research in this review.
Collapse
Affiliation(s)
- Xiangxian Zhang
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Liu
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xia Yuan
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuquan Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiawei Wei
- Laboratory of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Lee JY, Mehrazarin S, Alshaikh A, Kim S, Chen W, Lux R, Gwack Y, Kim RH, Kang MK. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-κB signaling and osteoclastogenesis. FASEB J 2019; 33:10515-10527. [PMID: 31251083 DOI: 10.1096/fj.201900154rr] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Histone Lys-specific demethylases (KDMs) play a key role in many biological processes through epigenetic mechanisms. However, the role of KDMs in inflammatory responses to oral bacterial infection is poorly understood. Here, we show a novel regulatory role of KDM3C in inflammatory responses to oral bacterial infection. KDM3C expression is transiently suppressed in human and mouse macrophages exposed to LPS from Porphyromonas gingivalis (Pg LPS). Loss of KDM3C in both human and mouse macrophages led to notable induction of proinflammatory cytokines in response to Pg LPS stimulation. Also, KDM3C depletion led to strong induction of p65 phosphorylation and accelerated nuclear translocation in cells exposed to Pg LPS. Kdm3C knockout (KO) in mice led to increased alveolar bone destruction upon induction of experimental periodontitis or pulp exposure compared with those of the wild-type (WT) littermates. The Kdm3C KO mice also revealed an increased number of osteoclasts juxtaposed to the bony lesions. We also confirmed enhanced osteoclastogenesis by bone marrow-derived macrophages isolated from the Kdm3C KO compared with the WT controls. These findings suggest an anti-inflammatory function of KDM3C in regulating the inflammatory responses against oral bacterial infection through suppression of NF-κB signaling and osteoclastogenesis.-Lee, J. Y., Mehrazarin, S., Alshaikh, A., Kim, S., Chen, W., Lux, R., Gwack, Y., Kim, R. H., Kang, M. K. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-κB signaling and osteoclastogenesis.
Collapse
Affiliation(s)
- Jae Young Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA.,Section of Endodontics, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Shebli Mehrazarin
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Abdullah Alshaikh
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Sol Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Wei Chen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA.,Section of Endodontics, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Renate Lux
- Section of Periodontics, Division of Constitutive and Regenerative Sciences, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA; and
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Reuben H Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| | - Mo K Kang
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA.,Section of Endodontics, University of California-Los Angeles (UCLA) School of Dentistry, Los Angeles, California, USA
| |
Collapse
|
9
|
Cheray M, Joseph B. Epigenetics Control Microglia Plasticity. Front Cell Neurosci 2018; 12:243. [PMID: 30123114 PMCID: PMC6085560 DOI: 10.3389/fncel.2018.00243] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 01/31/2023] Open
Abstract
Microglia, resident immune cells of the central nervous system, fulfill multiple functions in the brain throughout life. These microglial functions range from participation in innate and adaptive immune responses, involvement in the development of the brain and its homeostasis maintenance, to contribution to degenerative, traumatic, and proliferative diseases; and take place in the developing, the aging, the healthy, or the diseased brain. Thus, an impressive level of cellular plasticity, appears as a requirement for the pleiotropic biological functions of microglia. Epigenetic changes, including histone modifications or DNA methylation as well as microRNA expression, are important modifiers of gene expression, and have been involved in cell phenotype regulation and reprogramming and are therefore part of the mechanisms regulating cellular plasticity. Here, we review and discuss the epigenetic mechanisms, which are emerging as contributors to this microglial cellular plasticity and thereby can constitute interesting targets to modulate microglia associated brain diseases, including developmental diseases, neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
10
|
Penas C, Navarro X. Epigenetic Modifications Associated to Neuroinflammation and Neuropathic Pain After Neural Trauma. Front Cell Neurosci 2018; 12:158. [PMID: 29930500 PMCID: PMC5999732 DOI: 10.3389/fncel.2018.00158] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/22/2018] [Indexed: 12/20/2022] Open
Abstract
Accumulating evidence suggests that epigenetic alterations lie behind the induction and maintenance of neuropathic pain. Neuropathic pain is usually a chronic condition caused by a lesion, or pathological change, within the nervous system. Neuropathic pain appears frequently after nerve and spinal cord injuries or diseases, producing a debilitation of the patient and a decrease of the quality of life. At the cellular level, neuropathic pain is the result of neuronal plasticity shaped by an increase in the sensitivity and excitability of sensory neurons of the central and peripheral nervous system. One of the mechanisms thought to contribute to hyperexcitability and therefore to the ontogeny of neuropathic pain is the altered expression, trafficking, and functioning of receptors and ion channels expressed by primary sensory neurons. Besides, neuronal and glial cells, such as microglia and astrocytes, together with blood borne macrophages, play a critical role in the induction and maintenance of neuropathic pain by releasing powerful neuromodulators such as pro-inflammatory cytokines and chemokines, which enhance neuronal excitability. Altered gene expression of neuronal receptors, ion channels, and pro-inflammatory cytokines and chemokines, have been associated to epigenetic adaptations of the injured tissue. Within this review, we discuss the involvement of these epigenetic changes, including histone modifications, DNA methylation, non-coding RNAs, and alteration of chromatin modifiers, that have been shown to trigger modification of nociception after neural lesions. In particular, the function on these processes of EZH2, JMJD3, MeCP2, several histone deacetylases (HDACs) and histone acetyl transferases (HATs), G9a, DNMT, REST and diverse non-coding RNAs, are described. Despite the effort on developing new therapies, current treatments have only produced limited relief of this pain in a portion of patients. Thus, the present review aims to contribute to find novel targets for chronic neuropathic pain treatment.
Collapse
Affiliation(s)
- Clara Penas
- Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Xavier Navarro
- Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
11
|
Liu Y, Fang S, Li X, Feng J, Du J, Guo L, Su Y, Zhou J, Ding G, Bai Y, Wang S, Wang H, Liu Y. Aspirin inhibits LPS-induced macrophage activation via the NF-κB pathway. Sci Rep 2017; 7:11549. [PMID: 28912509 PMCID: PMC5599518 DOI: 10.1038/s41598-017-10720-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 08/14/2017] [Indexed: 12/26/2022] Open
Abstract
Aspirin (acetylsalicylic acid, ASA) has been shown to improve bone marrow mesenchymal stem cell-based calvarial bone regeneration by promoting osteogenesis and inhibiting osteoclastogenesis. However, it remains unknown whether aspirin influences other immune cells during bone formation. In the present study, we investigated whether ASA treatment influenced macrophage activation during the LPS inducement. We found that ASA could downregulate the expressions of iNOS and TNF-α both in mouse peritoneum macrophages and RAW264.7 cells induced by LPS via the IκK/IκB/NF-κB pathway and a COX2/PGE2/EP2/NF-κB feedback loop, without affecting the expressions of FIZZ/YM-1/ARG1 induced by IL-4. Furthermore, we created a rat mandibular bone defect model and showed that ASA treatment improved bone regeneration by inhibiting LPS-induced macrophage activation in the early stages of inflammation. Taken together, our results indicated that ASA treatment was a feasible strategy for improving bone regeneration, particularly in inflammatory conditions.
Collapse
Affiliation(s)
- Yitong Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Silian Fang
- Department of Oral and Maxillofacial Surgery, the Sixth affiliated Hospital of Sun Yat-sen University, Beijing, P. R. China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Jie Feng
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Lijia Guo
- Department of Oral and Maxillofacial Surgery, the Sixth affiliated Hospital of Sun Yat-sen University, Beijing, P. R. China
| | - Yingying Su
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, P. R. China
| | - Jian Zhou
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Gang Ding
- Department of Stomatology, Yidu Central Hospital, Weifang Medical University, Weifang, P. R. China
| | - Yuxing Bai
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Songling Wang
- Salivary Gland Disease Center and Molecular Laboratory for Gene Therapy and Tooth Regeneration, School of Stomatology, Capital Medical University, Beijing, P. R. China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, P. R. China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, P. R. China.
| |
Collapse
|
12
|
Xuan D, Han Q, Tu Q, Zhang L, Yu L, Murry D, Tu T, Lian J, Stein GS, Zhang J, Chen J. Epigenetic Modulation in Periodontitis: Interaction of Adiponectin and JMJD3-IRF4 Axis in Macrophages. J Cell Physiol 2016; 231:1090-6. [PMID: 26399931 PMCID: PMC5298882 DOI: 10.1002/jcp.25201] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 01/26/2023]
Abstract
Emerging evidence suggests an important role for epigenetic mechanisms in modulating signals during macrophage polarization and inflammation. JMJD3, a JmjC family histone demethylase necessary for M2 polarization is also required for effective induction of multiple M1 genes by lipopolysaccharide (LPS). However, the effects of JMJD3 to inflammation in the context of obesity remains unknown. To address this deficiency, we firstly examined the expression of JMJD3 in macrophage isolated from bone marrow and adipose tissue of diet induced obesity (DIO) mice. The results indicated that JMJD3 was down-regulated in obesity. Adiponectin (APN), a factor secreted by adipose tissue which is down-regulated in obesity, functions to switch macrophage polarization from M1 to M2, thereby attenuating chronic inflammation. Intriguingly, our results indicated that APN contributed to JMJD3 up-regulation, reduced macrophage infiltration in obese adipose tissue, and abolished the up-regulation of JMJD3 in peritoneal macrophages isolated from DIO mice when challenged with Porphyromonas gingivalis LPS (pg.lps). To elucidate the interaction of APN and JMJD3 involved in macrophage transformation in the context of inflammation, we designed the loss and gain-function experiments of APN in vivo with APN(-/-) mice with experimental periodontitis and in vitro with macrophage isolated from APN(-/-) mice. For the first time, we found that APN can help to reduce periodontitis-related bone loss, modulate JMJD3 and IRF4 expression, and macrophage infiltration. Therefore, it can be inferred that APN may contribute to anti-inflammation macrophage polarization by regulating JMJD3 expression, which provides a basis for macrophage-centered epigenetic therapeutic strategies.
Collapse
Affiliation(s)
- Dongying Xuan
- Department of Periodontology, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou, China
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Qianqian Han
- Department of Periodontology, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou, China
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Qisheng Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Lan Zhang
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Liming Yu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Dana Murry
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Tianchi Tu
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| | - Jane Lian
- Department of Biochemistry, University of Vermont College of Medicine, C401 Given Building, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | - Gary S. Stein
- Department of Biochemistry, University of Vermont College of Medicine, C401 Given Building, 89 Beaumont Avenue, Burlington, VT 05405, USA
| | - Jincai Zhang
- Department of Periodontology, Guangdong Provincial Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Jake Chen
- Division of Oral Biology, Tufts University School of Dental Medicine, Boston, MA, 02111, USA
| |
Collapse
|
13
|
Kapellos TS, Iqbal AJ. Epigenetic Control of Macrophage Polarisation and Soluble Mediator Gene Expression during Inflammation. Mediators Inflamm 2016; 2016:6591703. [PMID: 27143818 PMCID: PMC4842078 DOI: 10.1155/2016/6591703] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/28/2016] [Indexed: 12/14/2022] Open
Abstract
Macrophages function as sentinel cells, which constantly monitor the host environment for infection or injury. Macrophages have been shown to exhibit a spectrum of activated phenotypes, which can often be categorised under the M1/M2 paradigm. M1 macrophages secrete proinflammatory cytokines and chemokines, such as TNF-α, IL-6, IL-12, CCL4, and CXCL10, and induce phagocytosis and oxidative dependent killing mechanisms. In contrast, M2 macrophages support wound healing and resolution of inflammation. In the past decade, interest has grown in understanding the mechanisms involved in regulating macrophage activation. In particular, epigenetic control of M1 or M2 activation states has been shown to rely on posttranslational modifications of histone proteins adjacent to inflammatory-related genes. Changes in methylation and acetylation of histones by methyltransferases, demethylases, acetyltransferases, and deacetylases can all impact how macrophage phenotypes are generated. In this review, we summarise the latest advances in the field of epigenetic regulation of macrophage polarisation to M1 or M2 states, with particular focus on the cytokine and chemokine profiles associated with these phenotypes.
Collapse
Affiliation(s)
| | - Asif J. Iqbal
- Sir William Dunn school of Pathology, South Parks Road, Oxford OX1 3RE, UK
| |
Collapse
|
14
|
Jiang J, Jia T, Gong W, Ning B, Wooley PH, Yang SY. Macrophage Polarization in IL-10 Treatment of Particle-Induced Inflammation and Osteolysis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:57-66. [DOI: 10.1016/j.ajpath.2015.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 01/11/2023]
|
15
|
Abstract
Microglia are considered the brain's resident immune cell involved in immune defense, immunocompetence, and phagocytosis. They maintain tissue homeostasis within the brain and spinal cord under normal condition and serves as its initial host defense system. However, when the central nervous system (CNS) faces injury, microglia respond through signaling molecules expressed or released by neighboring cells. Microglial responses are dual in nature. They induce a nonspecific immune response that may exacerbate CNS injury, especially in the acute stages, but are also essential to CNS recovery and repair. The full range of microglial mechanisms have yet to be clarified, but there is accumulating knowledge about microglial activation in acute CNS injury. Microglial responses require hours to days to fully develop, and may present a therapeutic target for intervention with a much longer window of opportunity compare to other neurological treatments. The challenge will be to find ways to selectively suppress the deleterious effects of microglial activation without compromising its beneficial functions. This review aims to provide an overview of the recent progress relating on the deleterious and beneficial effect of microglia in the setting of acute CNS injury and the potential therapeutic intervention against microglial activation to CNS injury.
Collapse
Affiliation(s)
- Masahito Kawabori
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, 4150 Clement Street, San Francisco, CA, 94121, USA
| | | |
Collapse
|
16
|
Nixon SE, González-Peña D, Lawson MA, McCusker RH, Hernandez AG, O’Connor JC, Dantzer R, Kelley KW, Rodriguez-Zas SL. Analytical workflow profiling gene expression in murine macrophages. J Bioinform Comput Biol 2015; 13:1550010. [PMID: 25708305 PMCID: PMC4539142 DOI: 10.1142/s0219720015500109] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Comprehensive and simultaneous analysis of all genes in a biological sample is a capability of RNA-Seq technology. Analysis of the entire transcriptome benefits from summarization of genes at the functional level. As a cellular response of interest not previously explored with RNA-Seq, peritoneal macrophages from mice under two conditions (control and immunologically challenged) were analyzed for gene expression differences. Quantification of individual transcripts modeled RNA-Seq read distribution and uncertainty (using a Beta Negative Binomial distribution), then tested for differential transcript expression (False Discovery Rate-adjusted p-value < 0.05). Enrichment of functional categories utilized the list of differentially expressed genes. A total of 2079 differentially expressed transcripts representing 1884 genes were detected. Enrichment of 92 categories from Gene Ontology Biological Processes and Molecular Functions, and KEGG pathways were grouped into 6 clusters. Clusters included defense and inflammatory response (Enrichment Score = 11.24) and ribosomal activity (Enrichment Score = 17.89). Our work provides a context to the fine detail of individual gene expression differences in murine peritoneal macrophages during immunological challenge with high throughput RNA-Seq.
Collapse
Affiliation(s)
- Scott E. Nixon
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Dianelys González-Peña
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Marcus A. Lawson
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Robert H. McCusker
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Alvaro G. Hernandez
- Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jason C. O’Connor
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Robert Dantzer
- Department of Symptom Research, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Keith W. Kelley
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Sandra L. Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Statistics, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|