1
|
Gong Z, Yang C, Dai W, Miao S, Liu Y, Jiao Z, Li B, Xie W, Zhao W, Han X, Yu S, Dong Z. Annexin A1 exerts analgesic effect in a mouse model of medication overuse headache. iScience 2023; 26:108153. [PMID: 37867938 PMCID: PMC10587614 DOI: 10.1016/j.isci.2023.108153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/30/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023] Open
Abstract
Medication overuse headache (MOH) is a serious global condition. The interaction between headache attacks and medication overuse complicates the understanding of its pathophysiology. In this study, we developed a preclinical MOH model that incorporates these two key factors by overusing rizatriptan benzoate (RIZ, 4 mg/kg, i.g.) in a glyceryl trinitrate (GTN, 10 mg/kg, i.p.) induced chronic migraine mouse model. We observed that RIZ overuse aggravated GTN-induced cutaneous allodynia and caused a prolonged state of latent sensitization. We also detected a significant upregulation of Annexin-A1 (ANXA1), a protein mainly expressed in the microglia of the spinal trigeminal nucleus caudalis (SPVC), in GTN+RIZ mice. Intracerebroventricular injection of ANXA1-derived peptide Ac2-26 trifluoroacetic acid (TFA) (5 μg/mouse) inhibited bright light stress (BLS) induced acute allodynia via the formyl peptide receptor (FPR) in GTN+RIZ mice. These results suggest that ANXA1 may have an analgesic effect in triptan-associated MOH and could potentially serve as a therapeutic target.
Collapse
Affiliation(s)
- Zihua Gong
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
- Department of Medical Oncology, Bethune International peace Hospital, Shijiazhuang, Hebei 050082, China
| | - Chunxiao Yang
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Wei Dai
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Shuai Miao
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yingyuan Liu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Zhiyang Jiao
- Department of Outpatient, Shijiazhuang Fourth Retired Cadre Sanatorium of Hebei province Military Region, Shijiazhuang, Hebei 050082, China
| | - Bozhi Li
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Xie
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wei Zhao
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Xun Han
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Shengyuan Yu
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| | - Zhao Dong
- Department of Neurology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
2
|
Resende F, de Araújo S, Tavares LP, Teixeira MM, Costa VV. The Multifaceted Role of Annexin A1 in Viral Infections. Cells 2023; 12:1131. [PMID: 37190040 PMCID: PMC10137178 DOI: 10.3390/cells12081131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Dysregulated inflammatory responses are often correlated with disease severity during viral infections. Annexin A1 (AnxA1) is an endogenous pro-resolving protein that timely regulates inflammation by activating signaling pathways that culminate with the termination of response, clearance of pathogen and restoration of tissue homeostasis. Harnessing the pro-resolution actions of AnxA1 holds promise as a therapeutic strategy to control the severity of the clinical presentation of viral infections. In contrast, AnxA1 signaling might also be hijacked by viruses to promote pathogen survival and replication. Therefore, the role of AnxA1 during viral infections is complex and dynamic. In this review, we provide an in-depth view of the role of AnxA1 during viral infections, from pre-clinical to clinical studies. In addition, this review discusses the therapeutic potential for AnxA1 and AnxA1 mimetics in treating viral infections.
Collapse
Affiliation(s)
- Filipe Resende
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Simone de Araújo
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Luciana Pádua Tavares
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Mauro Martins Teixeira
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Department of Biochemistry and Immunology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Post-Graduation Program of Cell Biology, Department of Morphology, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Center for Research and Development of Drugs, Biological Sciences Institute, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| |
Collapse
|
3
|
Zhang Y, Feng S, Cheng X, Lou K, Liu X, Zhuo M, Chen L, Ye J. The potential value of exosomes as adjuvants for novel biologic local anesthetics. Front Pharmacol 2023; 14:1112743. [PMID: 36778004 PMCID: PMC9909291 DOI: 10.3389/fphar.2023.1112743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
The side effects of anesthetic drugs are a key preoperative concern for anesthesiologists. Anesthetic drugs used for general anesthesia and regional blocks are associated with a potential risk of systemic toxicity. This prompted the use of anesthetic adjuvants to ameliorate these side effects and improve clinical outcomes. However, the adverse effects of anesthetic adjuvants, such as neurotoxicity and gastrointestinal reactions, have raised concerns about their clinical use. Therefore, the development of relatively safe anesthetic adjuvants with fewer side effects is an important area for future anesthetic drug research. Exosomes, which contain multiple vesicles with genetic information, can be released by living cells with regenerative and specific effects. Exosomes released by specific cell types have been found to have similar effects as many local anesthetic adjuvants. Due to their biological activity, carrier efficacy, and ability to repair damaged tissues, exosomes may have a better efficacy and safety profile than the currently used anesthetic adjuvants. In this article, we summarize the contemporary literature about local anesthetic adjuvants and highlight their potential side effects, while discussing the potential of exosomes as novel local anesthetic adjuvant drugs.
Collapse
Affiliation(s)
- Yunmeng Zhang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Shangzhi Feng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xin Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Kecheng Lou
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xin Liu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Ming Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Li Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,*Correspondence: Li Chen, ; Junming Ye,
| | - Junming Ye
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China,Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China,*Correspondence: Li Chen, ; Junming Ye,
| |
Collapse
|
4
|
New Insights on Formyl Peptide Receptor Type 2 Involvement in Nociceptive Processes in the Spinal Cord. Life (Basel) 2022; 12:life12040500. [PMID: 35454990 PMCID: PMC9026467 DOI: 10.3390/life12040500] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/02/2022] [Accepted: 03/09/2022] [Indexed: 12/19/2022] Open
Abstract
Formyl peptide receptor type 2 (FPR2/ALX) belongs to the formyl peptide receptors (FPRs) family clustered on chromosome 19 and encodes a family of three Class A of G protein-coupled receptors (GPCRs). A short N-terminal region, an NPXXY motif in transmembrane (TM) region 7 and an E/DRY motif that bridges TM3 and TM6 stabilizing inactive receptor conformations characterize this class of receptors. In recognizing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), FPRs play a crucial role in innate immune responses. FPR2/ALX is highly expressed in myeloid cells, as well as in chondrocytes, fibroblasts, endothelial, epithelial and smooth muscle cells. FPR2/ALX mRNA expression was recently reported in the rat brainstem, spinal cord, thalamus/hypothalamus, cerebral neocortex, hippocampus, cerebellum and striatum. The central nervous system (CNS) distribution of FPR2/ALX suggests important functions in nociception. Thus, the present study was carried out to investigate the possible role of FPR2/ALX in nociception in mice. Intrathecal administration of the formyl peptide receptor type 1 (FPR1) agonist fMLF and the FPR2/ALX agonist BML-111 relieved nociception and these effects were reduced by contemporary administration of the FPR2/ALX antagonist WRW4. Furthermore, measurement of cytokines and brain-derived neurotrophic factor (BDNF) in the spinal cord of neuropathic mice demonstrated that the antinociceptive effects of BML-111 might depend on the reduction in cytokine release and BDNF in the spinal cord. These results suggest a possible role of FPR2/ALX for pain control in the spinal cord.
Collapse
|
5
|
Yu C, Zhang L. Methylprednisolone up-regulates annexin A1 (ANXA1) to inhibit the inflammation, apoptosis and oxidative stress of cigarette smoke extract (CSE)-induced bronchial epithelial cells, a chronic obstructive pulmonary disease in vitro model, through the formyl peptide receptor 2 (FPR2) receptors and the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway. Bioengineered 2022; 13:4028-4038. [PMID: 35129068 PMCID: PMC8973914 DOI: 10.1080/21655979.2022.2031769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/03/2022] [Accepted: 01/03/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive degenerative disease, of which smoking is the main causer. We carried out this study with the aim of exploring the underlying mechanism of methylprednisolone (MP) treating the COPD. To stimulate COPD in vitro, cigarette smoke extract (CSE)was employed to induce human bronchial epithelial cells BEAS-2B. With the help of MTT and Tunel assays, the viability and apoptosis of BEAS-2B cells after indicated treatment were assessed. The levels of inflammatory response and oxidative stress were determined by the changes of markers basing on their commercial kits. Additionally, annexin A1 (ANXA1) expressions at both protein and mRNA levels were assessed with Western blot and Reverse transcription‑quantitative PCR (RT-qPCR). Moreover, the expressions of apoptosis- and formyl peptide receptor 2 (FPR2) receptors and the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway-related proteins were determined with Western blot., related proteins and proteins. As a result, MP up-regulated the ANXA1 expression in CSE-induced BEAS-2B cells. MP enhanced the viability but suppressed the apoptosis, inflammatory response and oxidative stress of CSE-induced BEAS-2B cells via regulating FPR2/AMPK pathway, while ANXA1 knockdown exhibited oppositive effects on them. In conclusion, MP up-regulated ANXA1 to inhibit the inflammation, apoptosis and oxidative stress of BEAS-2B cells induced by CSE, alleviating COPD through suppressing the FPR2/AMPK pathway.
Collapse
Affiliation(s)
- Chan Yu
- The First Department of Respiratory Medicine, Chengdu Eighth People’s Hospital (Geriatric Hospital of Chengdu Medical College), Chengdu City, China
| | - Linghui Zhang
- Department of Internal Medicine, Department of Clinical Medicine, Shijiazhuang Medical College, Shijiazhuang City, China
| |
Collapse
|
6
|
Zhang Y, Ma S, Ke X, Yi Y, Yu H, Yu D, Li Q, Shang Y, Lu Y, Pei L. The mechanism of Annexin A1 to modulate TRPV1 and nociception in dorsal root ganglion neurons. Cell Biosci 2021; 11:167. [PMID: 34446102 PMCID: PMC8393810 DOI: 10.1186/s13578-021-00679-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Annexin A1 (ANXA1) exerts anti-nociceptive effect through ANXA1 receptor formyl peptide receptor 2 (FPR2/ALX (receptor for lipoxin A4), FPR2) at the dorsal root ganglia (DRG) level. However, the mechanisms remain elucidated. By using radiant heat, hot/cold plate, tail flick, von Frey, and Randall-Selitto tests to detect nociception in intact and chemical (capsaicin, menthol, mustard oil, formalin or CFA) injected AnxA1 conditional knockout (AnxA1-/-) mice, applying calcium imaging and patch clamp recordings in cultured DRG neurons to measure neuronal excitability, conducting immunofluorescence and western blotting to detect the protein levels of TRPV1, FPR2 and its downstream molecules, and performing double immunofluorescence and co-immunoprecipitation to investigate the interaction between Calmodulin (CaM) and TRPV1; we aim to uncover the molecular and cellular mechanisms of ANXA1's role in antinociception. RESULTS AnxA1-/- mice exhibited significant sensitivity to noxious heat (mean ± SD, 6.2 ± 1.0 s vs. 9.9 ± 1.6 s in Hargreaves test; 13.6 ± 1.5 s vs. 19.0 ± 1.9 s in hot plate test; n = 8; P < 0.001), capsaicin (101.0 ± 15.3 vs. 76.2 ± 10.9; n = 8; P < 0.01), formalin (early phase: 169.5 ± 32.8 s vs. 76.0 ± 21.9 s; n = 8; P < 0.05; late phase: 444.6 ± 40.1 s vs. 320.4 ± 33.6 s; n = 8; P < 0.01) and CFA (3.5 ± 0.8 s vs. 5.9 ± 1.4 s; n = 8; P < 0.01). In addition, we found significantly increased capsaicin induced Ca2+ response, TRPV1 currents and neuronal firing in AnxA1 deficient DRG neurons. Furthermore, ANXA1 mimic peptide Ac2-26 robustly increased intracellular Ca2+, inhibited TRPV1 current, activated PLCβ and promoted CaM-TRPV1 interaction. And these effects of Ac2-26 could be attenuated by FPR2 antagonist Boc2. CONCLUSIONS Selective deletion of AnxA1 in DRG neurons enhances TRPV1 sensitivity and deteriorates noxious heat or capsaicin induced nociception, while ANXA1 mimic peptide Ac2-26 desensitizes TRPV1 via FPR2 and the downstream PLCβ-Ca2+-CaM signal. This study may provide possible target for developing new analgesic drugs in inflammatory pain.
Collapse
Affiliation(s)
- Yufen Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Anesthesiology, School of Medicine, Washington University in Saint Loius, St. Loius, MO, 63110, USA
| | - Sehui Ma
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao Ke
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yao Yi
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hongyan Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dian Yu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiang Li
- Exchange, Development & Service Center for Science & Technology Talents, The Ministry of Science and Technology (Most), Beijing, 100045, China
| | - You Shang
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Youming Lu
- Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China.,Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lei Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Collaborative Innovation Center for Brain Science, The Institute for Brain Research (IBR), Huazhong University of Science and Technology, Wuhan, 430030, China. .,Department of Anesthesiology, School of Medicine, Washington University in Saint Loius, St. Loius, MO, 63110, USA.
| |
Collapse
|
7
|
Pogatzki-Zahn EM, Gomez-Varela D, Erdmann G, Kaschube K, Segelcke D, Schmidt M. A proteome signature for acute incisional pain in dorsal root ganglia of mice. Pain 2021; 162:2070-2086. [PMID: 33492035 PMCID: PMC8208099 DOI: 10.1097/j.pain.0000000000002207] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 12/03/2020] [Accepted: 12/21/2020] [Indexed: 01/04/2023]
Abstract
ABSTRACT After surgery, acute pain is still managed insufficiently and may lead to short-term and long-term complications including chronic postsurgical pain and an increased prescription of opioids. Thus, identifying new targets specifically implicated in postoperative pain is of utmost importance to develop effective and nonaddictive analgesics. Here, we used an integrated and multimethod workflow to reveal unprecedented insights into proteome dynamics in dorsal root ganglia (DRG) of mice after plantar incision (INC). Based on a detailed characterization of INC-associated pain-related behavior profiles, including a novel paradigm for nonevoked pain, we performed quantitative mass-spectrometry-based proteomics in DRG 1 day after INC. Our data revealed a hitherto unknown INC-regulated protein signature in DRG with changes in distinct proteins and cellular signaling pathways. In particular, we show the differential regulation of 44 protein candidates, many of which are annotated with pathways related to immune and inflammatory responses such as MAPK/extracellular signal-regulated kinases signaling. Subsequent orthogonal assays comprised multiplex Western blotting, bioinformatic protein network analysis, and immunolabeling in independent mouse cohorts to validate (1) the INC-induced regulation of immune/inflammatory pathways and (2) the high priority candidate Annexin A1. Taken together, our results propose novel potential targets in the context of incision and, therefore, represent a highly valuable resource for further mechanistic and translational studies of postoperative pain.
Collapse
Affiliation(s)
- Esther M. Pogatzki-Zahn
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - David Gomez-Varela
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| | | | - Katharina Kaschube
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Daniel Segelcke
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University Hospital Muenster, University of Muenster, Muenster, Germany
| | - Manuela Schmidt
- Max-Planck Institute of Experimental Medicine, Somatosensory Signaling and Systems Biology Group, Goettingen, Germany
| |
Collapse
|
8
|
Cata JP, Uhelski ML, Gorur A, Dougherty PM. Nociception and Pain: New Roles for Exosomes. Neuroscientist 2021; 28:349-363. [PMID: 34166130 DOI: 10.1177/10738584211027105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The interchange of information from one cell to another relies on the release of hundreds of different molecules including small peptides, amino acids, nucleotides, RNA, steroids, retinoids, or fatty acid metabolites. Many of them are released to the extracellular matrix as free molecules and others can be part of the cargo of cellular vesicles. Small extracellular vesicles (30-150 nm), also known as exosomes, are a known mechanism of cell-to-cell communication in the nervous system. Exosomes participate in the pathogenesis of several neurological conditions including Alzheimer's and Parkinson's disease. However, exciting emerging evidence demonstrates that exosomes also regulate mechanisms of the sensory process including nociception. The goal of this review is to summarize the literature on exosome biogenesis, methods of small vesicle isolation and purification, and their role in nociception. We also provide insights on the potential applications of exosomes as pain biomarkers or as novel therapeutics.
Collapse
Affiliation(s)
- Juan P Cata
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Megan L Uhelski
- Department of Pain Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Aysegul Gorur
- Department of Anesthesiology and Perioperative Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA.,Anesthesiology and Surgical Oncology Research Group, Houston, TX, USA
| | - Patrick M Dougherty
- Department of Pain Medicine, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
9
|
Ayoub SS. Paracetamol (acetaminophen): A familiar drug with an unexplained mechanism of action. Temperature (Austin) 2021; 8:351-371. [PMID: 34901318 PMCID: PMC8654482 DOI: 10.1080/23328940.2021.1886392] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 02/02/2023] Open
Abstract
Paracetamol (acetaminophen) is undoubtedly one of the most widely used drugs worldwide. As an over-the-counter medication, paracetamol is the standard and first-line treatment for fever and acute pain and is believed to remain so for many years to come. Despite being in clinical use for over a century, the precise mechanism of action of this familiar drug remains a mystery. The oldest and most prevailing theory on the mechanism of analgesic and antipyretic actions of paracetamol relates to the inhibition of CNS cyclooxygenase (COX) enzyme activities, with conflicting views on the COX isoenzyme/variant targeted by paracetamol and on the nature of the molecular interactions with these enzymes. Paracetamol has been proposed to selectively inhibit COX-2 by working as a reducing agent, despite the fact that in vitro screens demonstrate low potency on the inhibition of COX-1 and COX-2. In vivo data from COX-1 transgenic mice suggest that paracetamol works through inhibition of a COX-1 variant enzyme to mediate its analgesic and particularly thermoregulatory actions (antipyresis and hypothermia). A separate line of research provides evidence on potentiation of the descending inhibitory serotonergic pathway to mediate the analgesic action of paracetamol, but with no evidence of binding to serotonergic molecules. AM404 as a metabolite for paracetamol has been proposed to activate the endocannabinoid and the transient receptor potential vanilloid-1 (TRPV1) systems. The current review gives an update and in some cases challenges the different theories on the pharmacology of paracetamol and raises questions on some of the inadequately explored actions of paracetamol. List of Abbreviations: AM404, N-(4-hydroxyphenyl)-arachidonamide; CB1R, Cannabinoid receptor-1; Cmax, Maximum concentration; CNS, Central nervous system; COX, Cyclooxygenase; CSF, Cerebrospinal fluid; ED50, 50% of maximal effective dose; FAAH, Fatty acid amidohydrolase; IC50, 50% of the maximal inhibitor concentration; LPS, Lipopolysaccharide; NSAIDs, Non-steroidal anti-inflammatory drugs; PGE2, Prostaglandin E2; TRPV1, Transient receptor potential vanilloid-1.
Collapse
Affiliation(s)
- Samir S Ayoub
- School of Health, Sport and Bioscience, Medicines Research Group, University of East London, London, UK
| |
Collapse
|
10
|
Vieira C, Salm DC, Horewicz VV, Ludtke DD, Emer AA, Koerich JF, Mazzardo G, Elias S, Moré AOO, Mazzardo-Martins L, Cidral-Filho FJ, Reed WR, Piovezan AP, Martins DF. Electroacupuncture decreases inflammatory pain through a pro-resolving mechanism involving the peripheral annexin A1-formyl peptide receptor 2/ALX-opioid receptor pathway. Pflugers Arch 2021; 473:683-695. [PMID: 33474635 DOI: 10.1007/s00424-020-02502-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/28/2020] [Accepted: 12/03/2020] [Indexed: 12/31/2022]
Abstract
The pro-resolving mechanism is a recently described endogenous process that controls inflammation. The present study evaluated components of this mechanism, including annexin 1 (ANXA1) and the formyl peptide receptor 2/ALX (FPR2/ALX) receptor, in the antihyperalgesic effect induced by electroacupuncture (EA) in an animal model of persistent peripheral inflammation. Male Swiss mice underwent intraplantar (i.pl.) injection with complete Freund's adjuvant (CFA). Mechanical hyperalgesia was assessed with von Frey monofilaments. Animals were treated with EA (2-10 Hz, ST36-SP6) or subcutaneous BML-111 injection (FPR2/ALX agonist) for 5 consecutive days. In a separate set of experiments, on the first and fifth days after CFA injection, animals received i.pl. WRW4 (FPR2/ALX antagonist) or naloxone (non-selective opioid receptor antagonist) before EA or BML-111 injection. Paw protein levels of FPR2/ALX and ANXA1 were evaluated on the second day after CFA injection by western blotting technique. EA and BML-111 reduced mechanical hyperalgesia. I.pl. naloxone or WRW4 prevented the antihyperalgesic effect induced by either EA or BML-111. EA increased ANXA1 but did not alter FPR2/ALX receptor levels in the paw. Furthermore, i.pl. pretreatment with WRW4 prevented the increase of ANXA1 levels induced by EA. This work demonstrates that the EA antihyperalgesic effect on inflammatory pain involves the ANXA1/FPR2/ALX pro-resolution pathway. This effect appears to be triggered by the activation of FPR2/ALX receptors and crosstalk communication with the opioid system.
Collapse
Affiliation(s)
- Cintia Vieira
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Daiana C Salm
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Verônica V Horewicz
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Daniela D Ludtke
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Aline A Emer
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Júlia F Koerich
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Gustavo Mazzardo
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Sayron Elias
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Ari O O Moré
- Integrative Medicine and Acupuncture Division, University Hospital, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Leidiane Mazzardo-Martins
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Neuroscience, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Francisco J Cidral-Filho
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - William R Reed
- Department of Physical Therapy, University of Alabama at Birmingham, Birmingham, AL, USA.,Rehabilitation Science Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna Paula Piovezan
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil
| | - Daniel F Martins
- Experimental Neuroscience Laboratory (LaNEx), University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil. .,Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Palhoça, Santa Catarina, Brazil.
| |
Collapse
|
11
|
Xi Y, Ju R, Wang Y. Roles of Annexin A protein family in autophagy regulation and therapy. Biomed Pharmacother 2020; 130:110591. [PMID: 32763821 DOI: 10.1016/j.biopha.2020.110591] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/08/2023] Open
Abstract
Annexin A is a kind of calcium-dependent phospholipid-binding proteins, which contributes to the formation of the cell membranes and cytoskeleton and played a part as a membrane skeleton to stabilize lipid bilayer. Autophagy is one of the most important programmed cell death mechanisms. And recently some reports suggest that annexin A family protein is associated with autophagy for annexin A can regulate the formation of vesicular lipid membranes and promote cell exocytosis. In this review, we summarized the roles of annexin A protein family in autophagy regulation and targeted medical treatment for better diagnoses and therapies.
Collapse
Affiliation(s)
- Yufeng Xi
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China.
| | - Yujia Wang
- Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Schröder N, Schaffrath A, Welter JA, Putzka T, Griep A, Ziegler P, Brandt E, Samer S, Heneka MT, Kaddatz H, Zhan J, Kipp E, Pufe T, Tauber SC, Kipp M, Brandenburg LO. Inhibition of formyl peptide receptors improves the outcome in a mouse model of Alzheimer disease. J Neuroinflammation 2020; 17:131. [PMID: 32331524 PMCID: PMC7181500 DOI: 10.1186/s12974-020-01816-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background An important hallmark of Alzheimer’s disease (AD) is the increase of Aβ1-42 burden and its accumulation to senile plaques, leading the reactive gliosis and neurodegeneration. The modulation of glia cell function represents an attractive therapeutic strategy, but is currently limited by an incomplete understanding of its relevance for AD. The chemotactic G-protein coupled formyl peptide receptor (FPR), which is known to modulate Aβ1-42 uptake and signal transduction, might be one candidate molecule regulating glia function in AD. Here, we investigate whether the modulation of FPR exerts beneficial effects in an AD preclinical model. Methods To address this question, APP/PS1 double-transgenic AD mice were treated for 20 weeks with either the pro-inflammatory FPR agonist fMLF, the FPR1/2 antagonist Boc2 or the anti-inflammatory FPR2 agonist Ac2-26. Spatial learning and memory were evaluated using a Morris water maze test. Immunohistological staining, gene expression studies, and flow cytometry analyses were performed to study neuronal loss, gliosis, and Aß-load in the hippocampus and cortex, respectively. Results FPR antagonism by Boc2-treatment significantly improved spatial memory performance, reduced neuronal pathology, induced the expression of homeostatic growth factors, and ameliorated microglia, but not astrocyte, reactivity. Furthermore, the elevated levels of amyloid plaques in the hippocampus were reduced by Boc2-treatment, presumably by an induction of amyloid degradation. Conclusions We suggest that the modulation of FPR signaling cascades might be considered as a promising therapeutic approach for alleviating the cognitive deficits associated with early AD. Additional studies are now needed to address the downstream effectors as well as the safety profile of Boc2.
Collapse
Affiliation(s)
- Nicole Schröder
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Anja Schaffrath
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Josua A Welter
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Tim Putzka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Angelika Griep
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany
| | - Patrick Ziegler
- Institute for Occupational and Social Medicine, RWTH Aachen University, Aachen, Germany
| | - Elisa Brandt
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Sebastian Samer
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Gerontopsychiatry, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Hannes Kaddatz
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Jiangshan Zhan
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Eugenia Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany.,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Thomas Pufe
- Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany
| | - Lars-Ove Brandenburg
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057, Rostock, Germany. .,Department of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
13
|
Loss of hypothermic and anti-pyretic action of paracetamol in cyclooxygenase-1 knockout mice is indicative of inhibition of cyclooxygenase-1 variant enzymes. Eur J Pharmacol 2019; 861:172609. [DOI: 10.1016/j.ejphar.2019.172609] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 02/02/2023]
|
14
|
Annexin 1 inhibits remifentanil-induced hyperalgesia and NMDA receptor phosphorylation via regulating spinal CXCL12/CXCR4 in rats. Neurosci Res 2019; 144:48-55. [DOI: 10.1016/j.neures.2018.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/13/2018] [Accepted: 07/23/2018] [Indexed: 02/01/2023]
|
15
|
Ismail C, Hussin C, Mohamed M, Aziz C. Preemptive Effects of Administration of Tualang Honey on Inflammatory Responses in Adult Male Rats. ACTA ACUST UNITED AC 2017. [DOI: 10.6000/1927-5951.2017.07.01.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
|
16
|
Lu Y, Ni S, He LN, Gao YJ, Jiang BC. Annexin A10 is involved in the development and maintenance of neuropathic pain in mice. Neurosci Lett 2016; 631:1-6. [PMID: 27507697 DOI: 10.1016/j.neulet.2016.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/04/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022]
Abstract
ANXA10 (annexin A10) is a member of the annexin family, and its biological effects are mediated primarily through the calcium-dependent phospholipid-binding and calcium ion binding. We examined the gene expressions of the L5 spinal cord after spinal nerve ligation (SNL)-induced neuropathic pain in mice by gene chip. The results showed that Anxa10 mRNA was the most upregulated gene in annexin family with 73.7-fold increase. Although previous studies have reported that several annexins are involved in nociceptive pain, the role of Anxa10 in pain remains undefined. We aimed to evaluate the role of ANXA10 in mediating injury-induced heat hyperalgesia and mechanical allodynia. We found that SNL induced persistent upregulation of Anxa10 mRNA and protein in the spinal cord of mice. Moreover, ANXA10 was colocalized with the neuronal marker MAP2 and astrocytic marker glial fibrillary acidic protein (GFAP), but not with microglial marker CD11b. Finally, pretreatment with Anxa10 siRNA partially prevented SNL-induced mechanical allodynia and heat hyperalgesia. Posttreatment with Anxa10 siRNA attenuated SNL-induced neuropathic pain. These findings suggest that ANXA10 might be a novel target in the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Ying Lu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Jiangsu 226001, China; Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Sujie Ni
- Department of Medical Oncology, The affiliated Hospital of Nantong University, Jiangsu 226001, China
| | - Li-Na He
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yong-Jing Gao
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Bao-Chun Jiang
- Pain Research Laboratory, Institute of Nautical Medicine, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
17
|
Yamanaka H, Kobayashi K, Okubo M, Noguchi K. Annexin A2 in primary afferents contributes to neuropathic pain associated with tissue type plasminogen activator. Neuroscience 2016; 314:189-99. [DOI: 10.1016/j.neuroscience.2015.11.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/21/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023]
|
18
|
Ong WY, Farooqui T, Kokotos G, Farooqui AA. Synthetic and natural inhibitors of phospholipases A2: their importance for understanding and treatment of neurological disorders. ACS Chem Neurosci 2015; 6:814-31. [PMID: 25891385 DOI: 10.1021/acschemneuro.5b00073] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Phospholipases A2 (PLA2) are a diverse group of enzymes that hydrolyze membrane phospholipids into arachidonic acid and lysophospholipids. Arachidonic acid is metabolized to eicosanoids (prostaglandins, leukotrienes, thromboxanes), and lysophospholipids are converted to platelet-activating factors. These lipid mediators play critical roles in the initiation, maintenance, and modulation of neuroinflammation and oxidative stress. Neurological disorders including excitotoxicity; traumatic nerve and brain injury; cerebral ischemia; Alzheimer's disease; Parkinson's disease; multiple sclerosis; experimental allergic encephalitis; pain; depression; bipolar disorder; schizophrenia; and autism are characterized by oxidative stress, inflammatory reactions, alterations in phospholipid metabolism, accumulation of lipid peroxides, and increased activities of brain phospholipase A2 isoforms. Several old and new synthetic inhibitors of PLA2, including fatty acid trifluoromethyl ketones; methyl arachidonyl fluorophosphonate; bromoenol lactone; indole-based inhibitors; pyrrolidine-based inhibitors; amide inhibitors, 2-oxoamides; 1,3-disubstituted propan-2-ones and polyfluoroalkyl ketones as well as phytochemical based PLA2 inhibitors including curcumin, Ginkgo biloba and Centella asiatica extracts have been discovered and used for the treatment of neurological disorders in cell culture and animal model systems. The purpose of this review is to summarize information on selective and potent synthetic inhibitors of PLA2 as well as several PLA2 inhibitors from plants, for treatment of oxidative stress and neuroinflammation associated with the pathogenesis of neurological disorders.
Collapse
Affiliation(s)
- Wei-Yi Ong
- Department
of Anatomy, National University of Singapore, Singapore 119260, Singapore
| | - Tahira Farooqui
- Department
of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio 43210, United States
| | - George Kokotos
- Laboratory
of Organic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis,
Athens 15771, Greece
| | - Akhlaq A. Farooqui
- Department
of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
19
|
The resolution of inflammation: Principles and challenges. Semin Immunol 2015; 27:149-60. [PMID: 25911383 DOI: 10.1016/j.smim.2015.03.014] [Citation(s) in RCA: 263] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022]
Abstract
The concept that chemokines, cytokines and pro-inflammatory mediators act in a co-ordinated fashion to drive the initiation of the inflammatory reaction is well understood. The significance of such networks acting during the resolution of inflammation however is poorly appreciated. In recent years, specific pro-resolving mediators were discovered which activate resolution pathways to return tissues to homeostasis. These mediators are diverse in nature, and include specialized lipid mediators (lipoxins, resolvins, protectins and maresins) proteins (annexin A1, galectins) and peptides, gaseous mediators including hydrogen sulphide, a purine (adenosine), as well as neuromodulator release under the control of the vagus nerve. Functionally, they can act to limit further leukocyte recruitment, induce neutrophil apoptosis and enhance efferocytosis by macrophages. They can also switch macrophages from classical to alternatively activated cells, promote the return of non-apoptotic cells to the lymphatics and help initiate tissue repair mechanisms and healing. Within this review we highlight the essential cellular aspects required for successful tissue resolution, briefly discuss the pro-resolution mediators that drive these processes and consider potential challenges faced by researchers in the quest to discover how inflammation resolves and why chronic inflammation persists.
Collapse
|
20
|
Abstract
The resolution of inflammation (RoI), once believed to be a passive process, has lately been shown to be an active and delicately orchestrated process. During the resolution phase of acute inflammation, novel mediators, including lipoxins and resolvins, which are members of the specialized pro-resolving mediators of inflammation, are produced. FPR2/ALXR, a receptor modulated by some of these lipids as well as by peptides (e.g., annexin A1), has been shown to be one of the receptors involved in the RoI. The aim of this perspective is to present the concept of the RoI from a medicinal chemistry point of view and to highlight the effort of the research community to discover and develop anti-inflammatory/pro-resolution small molecules to orchestrate inflammation by activation of FPR2/ALXR.
Collapse
Affiliation(s)
- Olivier Corminboeuf
- Actelion Pharmaceuticals Ltd. , Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | |
Collapse
|
21
|
Min SH, Soh JS, Park JY, Choi SU, Lee HW, Lee JJ, Kim JH. Epidural dexamethasone decreased inflammatory hyperalgesia and spinal cPLA₂ expression in a rat formalin test. Yonsei Med J 2014; 55:1631-9. [PMID: 25323902 PMCID: PMC4205705 DOI: 10.3349/ymj.2014.55.6.1631] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the effect of epidural dexamethasone on analgesia and cytosolic phospholipase A₂ (cPLA₂) expression in the spinal cord in a rat formalin test. MATERIALS AND METHODS Epidural dexamethasone injection was performed to Sprague-Dawley rats with a 25 gauge needle under fluoroscopy. Following the epidural injection, a formalin induced pain behavior test was performed. Next, the spinal cords corresponding to L4 dorsal root ganglion was extracted to observe the cPLA₂ expression. RESULTS There were no differences in pain response during phase I among the groups. The phase II pain response in 300 μg of epidural dexamethasone group decreased as compared to control, 30 μg of epidural dexamethasone, 100 μg of epidural dexamethasone, and 300 μg of systemic dexamethasone groups. The expression of cPLA₂ decreased in Rexed laminae I-II in 300 μg of the epidural dexamethasone group compared with the ones in the control group. CONCLUSION Taken together, these results suggest that 300 μg of epidural dexamethasone has an attenuating effect on the peripheral inflammatory tissue injury induced hyperalgesia and this effect is mediated through the inhibition of intraspinal cPLA₂ expression and the primary site of action is the laminae I-II of the spinal cord.
Collapse
Affiliation(s)
- Sam-Hong Min
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jung-Sub Soh
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji-Yong Park
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sung-Uk Choi
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hye-Won Lee
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jae-Jin Lee
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea
| | - Jae-Hwan Kim
- Department of Anesthesiology and Pain Medicine, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
22
|
Somma T, Cinci L, Formicola G, Pini A, Thurmond R, Ennis M, Bani D, Masini E. A selective antagonist of histamine H₄ receptors prevents antigen-induced airway inflammation and bronchoconstriction in guinea pigs: involvement of lipocortin-1. Br J Pharmacol 2014; 170:200-13. [PMID: 23734568 DOI: 10.1111/bph.12264] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 05/20/2013] [Accepted: 05/24/2013] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Among the pathogenic mechanisms of asthma, a role for oxidative/nitrosative stress has been well documented. Recent evidence suggests that histamine H₄ receptors play a modulatory role in allergic inflammation. Here we report the effects of compound JNJ 7777120 (JNJ), a selective H4 receptor antagonist, on antigen-induced airway inflammation, paying special attention to its effects on lipocortin-1 (LC-1/annexin-A1), a 37 kDA anti-inflammatory protein that plays a key role in the production of inflammatory mediators. EXPERIMENTAL APPROACH Ovalbumin (OA)-sensitized guinea pigs placed in a respiratory chamber were challenged with antigen. JNJ (5, 7.5 and 10 mg.kg⁻¹) was given i.p. for 4 days before antigen challenge. Respiratory parameters were recorded. Bronchoalveolar lavage (BAL) fluid was collected and lung specimens taken for further analyses 1 h after antigen challenge. In BAL fluid, levels of LC-1, PGD2 , LTB4 and TNF-α were measured. In lung tissue samples, myeloperoxidase, caspase-3 and Mn-superoxide dismutase activities and 8-hydroxy-2-deoxyguanosine levels were measured. KEY RESULTS OA challenge decreased LC-1 levels in BAL fluid, induced cough, dyspnoea and bronchoconstriction and increased PGD2 , LTB4 and TNF-α levels in lung tissue. Treatment with JNJ dose-dependently increased levels of LC-1, reduced respiratory abnormalities and lowered levels of PGD2 , LTB4 and TNF-α in BAL fluid. CONCLUSIONS AND IMPLICATIONS Antigen-induced asthma-like reactions in guinea pigs decreased levels of LC-1 and increased TNF-α and eicosanoid production. JNJ pretreatment reduced allergic asthmatic responses and airway inflammation, an effect associated with LC-1 up-regulation.
Collapse
Affiliation(s)
- T Somma
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Chen L, Lv F, Pei L. Annexin 1: a glucocorticoid-inducible protein that modulates inflammatory pain. Eur J Pain 2013; 18:338-47. [PMID: 23904250 DOI: 10.1002/j.1532-2149.2013.00373.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2013] [Indexed: 12/16/2022]
Abstract
Annexin 1, a glucocorticoid (GC)-inducible protein, can play an important role via formyl peptide receptor like 1 (FPR2/ALX, also known as FPRL1) in inflammatory pain modulation. The aim of this review is to analyze different lines of evidence for the role of ANXA1 with different mechanisms on inflammatory pain and describe the profile of ANXA1 as a potential analgesic. A Medline (PUBMED) search using the terms 'Annexin 1 distribution OR expression, FPR2/ALX distribution OR expression, Annexin 1 AND pain, Annexin 1 AND FPR2/ALX AND pain' was performed. Articles with a publication date up to Nov. 1st, 2012 were included. The antinociception of ANXA1 has been evaluated in diverse pain models. It has been suggested that ANXA1 may exerts its action via: (1) inhibiting vital cytokines involved in pain transmission, (2) inhibiting neutrophil accumulation through preventing transendothelial migration via an interaction with formyl peptide receptors, (3) facilitating tonic opioid release from neutrophil in inflammatory site, (4) interrupting the peripheral nociceptive transmission by suppressing neuronal excitability. In general, ANXA1 is a potential mediator for anti-nociception and the role with its receptor constitute attractive targets for developing anesthesia and analgesic drugs, and their interaction may prove to be a useful strategy to treat inflammatory pain.
Collapse
Affiliation(s)
- L Chen
- Department of Neurology of the First People's Hospital of Jingzhou, The first affiliated hospital of Yangtze University, Jingzhou, China
| | | | | |
Collapse
|
24
|
Dalli J, Consalvo AP, Ray V, Di Filippo C, D'Amico M, Mehta N, Perretti M. Proresolving and tissue-protective actions of annexin A1-based cleavage-resistant peptides are mediated by formyl peptide receptor 2/lipoxin A4 receptor. THE JOURNAL OF IMMUNOLOGY 2013; 190:6478-87. [PMID: 23686496 DOI: 10.4049/jimmunol.1203000] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endogenous mechanisms regulating the host response during inflammation resolution are critical in ensuring disposal of noxious stimuli and return to homeostasis. In this article, we engineered novel Annexin A1 (AnxA1)-based peptides, AnxA1(2-50), that displayed specific binding to the AnxA1 receptor (formyl peptide receptor 2/Lipoxin A4 receptor [FPR2/ALX]; IC50 ∼4 nM). Intravenous administration of AnxA1(2-50) markedly reduced (>60%) leukocyte adhesion to postcapillary venules in wild type and Fpr1(-/-), but not Fpr2/Alx(-/-), mice. Generation of a metabolically stable form of this peptide (CR-AnxA1(2-50)), engineered by substituting a cleavage site shared by human proteinase 3 and neutrophil elastase, yielded an agonist that was resistant to neutrophil-mediated cleavage and displayed enhanced proresolving actions: accelerated resolution of self-limited inflammation and enhanced macrophage efferocytosis after sterile injury, when compared with AnxA1(2-50). These actions were retained with human primary leukocytes where CR-AnxA1(2-50) decreased neutrophil-endothelial interactions (∼25-45%), and stimulated neutrophil apoptosis and macrophage efferocytosis (∼45%). In murine cardiac ischemia/reperfusion injury, CR-AnxA1(2-50) elicited tissue-protective actions reducing infarct size (∼60%) and incidence of 24-h death. These results identify AnxA1(2-50) and CR-AnxA1(2-50) as FPR2/ALX agonists that harness the proresolving actions of AnxA1, and thus may represent therapeutic tools for treatment of inflammatory conditions.
Collapse
Affiliation(s)
- Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
25
|
Girol AP, Mimura KKO, Drewes CC, Bolonheis SM, Solito E, Farsky SHP, Gil CD, Oliani SM. Anti-inflammatory mechanisms of the annexin A1 protein and its mimetic peptide Ac2-26 in models of ocular inflammation in vivo and in vitro. THE JOURNAL OF IMMUNOLOGY 2013; 190:5689-701. [PMID: 23645879 DOI: 10.4049/jimmunol.1202030] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Annexin A1 (AnxA1) is a protein that displays potent anti-inflammatory properties, but its expression in eye tissue and its role in ocular inflammatory diseases have not been well studied. We investigated the mechanism of action and potential uses of AnxA1 and its mimetic peptide (Ac2-26) in the endotoxin-induced uveitis (EIU) rodent model and in human ARPE-19 cells activated by LPS. In rats, analysis of untreated EIU after 24 and 48 h or EIU treated with topical applications or with a single s.c. injection of Ac2-26 revealed the anti-inflammatory actions of Ac2-26 on leukocyte infiltration and on the release of inflammatory mediators; the systemic administration of Boc2, a formylated peptide receptor (fpr) antagonist, abrogated the peptide's protective effects. Moreover, AnxA1(-/-) mice exhibited exacerbated EIU compared with wild-type animals. Immunohistochemical studies of ocular tissue showed a specific AnxA1 posttranslational modification in EIU and indicated that the fpr2 receptor mediated the anti-inflammatory actions of AnxA1. In vitro studies confirmed the roles of AnxA1 and fpr2 and the protective effects of Ac2-26 on the release of chemical mediators in ARPE-19 cells. Molecular analysis of NF-κB translocation and IL-6, IL-8, and cyclooxygenase-2 gene expression indicated that the protective effects of AnxA1 occur independently of the NF-κB signaling pathway and possibly in a posttranscriptional manner. Together, our data highlight the role of AnxA1 in ocular inflammation, especially uveitis, and suggest the use of AnxA1 or its mimetic peptide Ac2-26 as a therapeutic approach.
Collapse
Affiliation(s)
- Ana P Girol
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University, São José do Rio Preto 15054-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Peshavariya HM, Taylor CJ, Goh C, Liu GS, Jiang F, Chan EC, Dusting GJ. Annexin peptide Ac2-26 suppresses TNFα-induced inflammatory responses via inhibition of Rac1-dependent NADPH oxidase in human endothelial cells. PLoS One 2013; 8:e60790. [PMID: 23637767 PMCID: PMC3634803 DOI: 10.1371/journal.pone.0060790] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/03/2013] [Indexed: 11/25/2022] Open
Abstract
The anti-inflammatory peptide annexin-1 binds to formyl peptide receptors (FPR) but little is known about its mechanism of action in the vasculature. Here we investigate the effect of annexin peptide Ac2-26 on NADPH oxidase activity induced by tumour necrosis factor alpha (TNFα) in human endothelial cells. Superoxide release and intracellular reactive oxygen species (ROS) production from NADPH oxidase was measured with lucigenin-enhanced chemiluminescence and 2′,7′-dichlorodihydrofluorescein diacetate, respectively. Expression of NADPH oxidase subunits and intracellular cell adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1) were determined by real-time PCR and Western blot analysis. Promoter activity of nuclear factor kappa B (NFκB) was measured by luciferase activity assay. TNFα stimulated NADPH-dependent superoxide release, total ROS formation and expression of ICAM-1and VCAM-1. Pre-treatment with N-terminal peptide of annexin-1 (Ac2-26, 0.5–1.5 µM) reduced all these effects, and the inhibition was blocked by the FPRL-1 antagonist WRW4. Furthermore, TNFα-induced NFκB promoter activity was attenuated by both Ac2-26 and NADPH oxidase inhibitor diphenyliodonium (DPI). Surprisingly, Nox4 gene expression was reduced by TNFα whilst expression of Nox2, p22phox and p67phox remained unchanged. Inhibition of NADPH oxidase activity by either dominant negative Rac1 (N17Rac1) or DPI significantly attenuated TNFα-induced ICAM-1and VCAM-1 expression. Ac2-26 failed to suppress further TNFα-induced expression of ICAM-1 and VCAM-1 in N17Rac1-transfected cells. Thus, Ac2-26 peptide inhibits TNFα-activated, Rac1-dependent NADPH oxidase derived ROS formation, attenuates NFκB pathways and ICAM-1 and VCAM-1 expression in endothelial cells. This suggests that Ac2-26 peptide blocks NADPH oxidase activity and has anti-inflammatory properties in the vasculature which contributes to modulate in reperfusion injury inflammation and vascular disease.
Collapse
Affiliation(s)
- Hitesh M. Peshavariya
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
- * E-mail: (GJD); (HMP)
| | - Caroline J. Taylor
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Faculty of Health Sciences, The Australian Catholic University, Victoria, Australia
| | - Celeste Goh
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
| | - Guei-Sheung Liu
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Fan Jiang
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Elsa C. Chan
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
| | - Gregory J. Dusting
- O’Brien Institute, University of Melbourne, Fitzroy, Victoria, Australia
- Centre for Eye Research Australia, University of Melbourne, East Melbourne, Victoria, Australia
- * E-mail: (GJD); (HMP)
| |
Collapse
|
27
|
Seidel S, Neymeyer H, Kahl T, Röschel T, Mutig K, Flower R, Schnermann J, Bachmann S, Paliege A. Annexin A1 modulates macula densa function by inhibiting cyclooxygenase 2. Am J Physiol Renal Physiol 2012; 303:F845-54. [DOI: 10.1152/ajprenal.00704.2011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Annexin A1 (ANXA1) exerts anti-inflammatory effects through multiple mechanisms including inhibition of prostaglandin synthesis. Once secreted, ANXA1 can bind to G protein-coupled formyl peptide receptors (Fpr) and activate diverse cellular signaling pathways. ANXA1 is known to be expressed in cells of the juxtaglomerular apparatus, but its relation to the expression of cyclooxygenase 2 (COX-2) in thick ascending limb and macula densa cells has not been elucidated. We hypothesized that ANXA1 regulates the biosynthesis of COX-2. ANXA1 abundance in rat kidney macula densa was extensively colocalized with COX-2 (95%). Furosemide, an established stimulus for COX-2 induction, caused enhanced expression of both ANXA1 and COX-2 with maintained colocalization (99%). In ANXA1-deficient mice, COX-2-positive cells were more numerous than in control mice (+107%; normalized to glomerular number; P < 0.05) and renin expression was increased (+566%; normalized to glomerular number; P < 0.05). Cultured macula densa cells transfected with full-length rat ANXA1 revealed downregulation of COX-2 mRNA (−59%; P < 0.05). Similarly, treatment with dexamethasone suppressed COX-2 mRNA in the cells (−49%; P < 0.05), while inducing ANXA1 mRNA (+56%; P < 0.05) and ANXA1 protein secretion. Inhibition of the ANXA-1 receptor Fpr1 with cyclosporin H blunted the effect of dexamethasone on COX-2 expression. These data show that ANXA1 exerts an inhibitory effect on COX-2 expression in the macula densa. ANXA1 may be a novel intrinsic modulator of renal juxtaglomerular regulation by inhibition of PGE2 synthesis.
Collapse
Affiliation(s)
- S. Seidel
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - H. Neymeyer
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - T. Kahl
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - T. Röschel
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - K. Mutig
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - R. Flower
- Department of Biochemical Pharmacology, William Harvey Research Institute, Queen Mary, University of London, London, United Kingdom; and
| | - J. Schnermann
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - S. Bachmann
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| | - A. Paliege
- Department of Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany; and
| |
Collapse
|
28
|
Bandorowicz-Pikula J, Wos M, Pikula S. Do annexins participate in lipid messenger mediated intracellular signaling? A question revisited. Mol Membr Biol 2012; 29:229-42. [PMID: 22694075 DOI: 10.3109/09687688.2012.693210] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Annexins are physiologically important proteins that play a role in calcium buffering but also influence membrane structure, participate in Ca²⁺-dependent membrane repair events and in remodelling of the cytoskeleton. Thirty years ago several peptides isolated from lung perfusates, peritoneal leukocytes, neutrophiles and renal cells were proven inhibitory to the activity of phospholipase A₂. Those peptides were found to derive from structurally related proteins: annexins AnxA1 and AnxA2. These findings raised the question whether annexins may participate in regulation of the production of lipid second messengers and, therefore, modulate numerous lipid mediated signaling pathways in the cell. Recent advances in the field of annexins made also with the use of knock-out animal models revealed that these proteins are indeed important constituents of specific signaling pathways. In this review we provide evidence supporting the hypothesis that annexins, as membrane-binding proteins and organizers of the membrane lateral heterogeneity, may participate in lipid mediated signaling pathways by affecting the distribution and activity of lipid metabolizing enzymes (most of the reports point to phospholipase A₂) and of protein kinases regulating activity of these enzymes. Moreover, some experimental data suggest that annexins may directly interact with lipid metabolizing enzymes and, in a calcium-dependent or independent manner, with some of their substrates and products. On the basis of these observations, many investigators suggest that annexins are capable of linking Ca²⁺, redox and lipid signaling to coordinate vital cellular responses to the environmental stimuli.
Collapse
Affiliation(s)
- Joanna Bandorowicz-Pikula
- Laboratory of Cellular Metabolism, Department of Biochemistry, Nencki Institute of Experimental Biology, PL 02-093 Warsaw, Poland.
| | | | | |
Collapse
|
29
|
Sui W, Tang D, Zou G, Chen J, Ou M, Zhang Y, Dai Y. Differential proteomic analysis of renal tissue in lupus nephritis using iTRAQ reagent technology. Rheumatol Int 2011; 32:3537-43. [DOI: 10.1007/s00296-011-2207-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 10/22/2011] [Indexed: 10/15/2022]
|
30
|
Pei L, Zhang J, Zhao F, Su T, Wei H, Tian J, Li M, Shi J. Annexin 1 exerts anti-nociceptive effects after peripheral inflammatory pain through formyl-peptide-receptor-like 1 in rat dorsal root ganglion. Br J Anaesth 2011; 107:948-58. [PMID: 21990306 DOI: 10.1093/bja/aer299] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Annexin 1 (ANXA1) has analgesic effects in inflammatory pain. We aimed to investigate the anti-nociceptive role of ANXA1, at the dorsal root ganglion (DRG) level, through an interaction with formyl-peptide-receptor-like 1 (FPR2/ALX). METHODS Inflammatory pain was evoked by injecting complete Freund's adjuvant (CFA, 50 μl) into the hindpaw of male Sprague-Dawley rats. The distribution of ANXA1 and FPR2/ALX in L4/5 DRGs was evaluated by immunofluorescence. The expression of ANXA1 was measured by western blot. The involvement of FPR2/ALX in the anti-nociception of ANXA1 was investigated by thermal (irradiant heat) and mechanical (von Frey filament) pain tests with intrathecal (i.t.) ANXA1-derived peptide (Anxa1(2-26)), FPR2/ALX agonist 5(S)-6(R)-7-trihydroxyheptanoic-acid-methyl-ester (BML-111), and antagonist N-t-Boc-Phe-Leu-Phe-Leu-Phe (Boc1). RESULTS ANXA1 and FPR2/ALX localized in the satellite glial cells and neurones in L4/5 DRGs. CFA treatment (n=20) increased ANXA1 expression in L4/5 DRGs within 7 days (P<0.01). I.T. Anxa1(2-26) (20 and 100 µg µl(-1)) and BML-111 (10 and 100 nmol) reduced CFA-induced thermal and mechanical nociception within 48 h (n=40) (P<0.05). However, i.t. Boc1 10 µg intensified inflammatory pain (P<0.05) and reversed the anti-nociceptive effect of Anxa1(2-26) (n=25) (P<0.05). Moreover, ANXA1 expression increased in L4/5 DRGs after i.t. Anxa1(2-26) (20 µg µl(-1)) (P<0.05) and BML-111 (10 nmol) (P<0.01) but decreased after i.t. Boc1 (10 and 100 µg) alone (P<0.01) or Boc1 (10 µg) co-injection with Anxa1(2-26) (20 µg µl(-1)) (P<0.05). CONCLUSIONS Endogenous ANXA1 expression at the DRG level is involved in CFA-induced inflammatory pain, and i.t. ANXA1 20 µg µl(-1) produces its anti-nociceptive effect through FPR2/ALX.
Collapse
Affiliation(s)
- L Pei
- Department of Neurobiology and Key Laboratory of Neurological Diseases of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan 430030, PR China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Ayoub SS, Wood EG, Hassan SU, Bolton C. Cyclooxygenase expression and prostaglandin levels in central nervous system tissues during the course of chronic relapsing experimental autoimmune encephalomyelitis (EAE). Inflamm Res 2011; 60:919-28. [PMID: 21667309 DOI: 10.1007/s00011-011-0352-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 04/27/2011] [Accepted: 05/27/2011] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE Multiple sclerosis (MS) and its animal counterpart experimental autoimmune encephalomyelitis (EAE) have a major inflammatory component that drives and orchestrates both diseases. One particular group of mediators are the prostaglandins (PGs), which we have previously shown, through quantitation and pharmacological intervention, to be closely involved in the pathology of MS and EAE. The aim of the current study was to determine the expression of the PG-generating cyclooxygenase (COX) enzymes and the profile of PGE(2) and PGD(2), in selected central nervous system (CNS) tissues, with the development of the chronic relapsing (CR) form of EAE. In particular, the work investigates the possible relationship between the expression of COX isoenzymes and PG levels during the neurological phases of CR EAE. METHODS CR EAE was induced in Biozzi mice with inoculum containing lyophilised, syngeneic spinal cord emulsified in complete Freund's adjuvant. The cerebral cortex, cerebellum and spinal cord were dissected from mice during the acute, remission and relapse stages of disease with a minimum of five animals per treatment. The expression of COX-1, COX-1b variant and COX-2, in pooled samples, was determined by Western blotting. PGE(2) and PGD(2) levels in extracted samples were measured using commercial enzyme immunoassay kits. RESULTS COX-2 expression in spinal cords during acute disease remained unaltered and was in contrast to an enhancement of the enzyme, together with COX-1 and COX-1b, in all other sampled areas. PGE(2) and PGD(2) levels remained unchanged during the acute phase and the subsequent remission of symptoms. COX-1 and COX-1b expression was elevated in tissues during the relapse stage of CR EAE and concentrations of the prostanoids were markedly increased. CONCLUSIONS The study examines the implications of COX isoenzyme expression over the course of CR EAE and discusses the reported relationship between PGE(2) and PGD(2) in the instigation and resolution of CNS inflammation. Consideration is also given to the treatment of CR EAE and suggests that drugs designed to limit the inflammatory effects of the PGs should be administered prior to or during the relapse phase of the disease.
Collapse
Affiliation(s)
- Samir S Ayoub
- Centre for Biochemical Pharmacology, William Harvey Research Institute, St. Bartholomew's and London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | | | | | | |
Collapse
|
32
|
Abstract
IMPORTANCE OF THE FIELD Local injections of steroids have been widely practiced to provide regional analgesia for the treatment of a wide variety of musculoskeletal pain syndromes. However, evidence regarding the effectiveness of steroid injections is not substantial. Also, there have been reports of catastrophic complications associated with their use. AREAS COVERED IN THIS REVIEW The evidence currently available in the literature (Database: Ovid MEDLINE 1950 to 2010) is reviewed. The areas covered include the analgesic mechanisms of steroids, indications for steroid injections and their effectiveness, as well as the risks and precautions for steroid injections. WHAT THE READER WILL GAIN This is an up-to-date review on the clinical application of steroid injections for regional analgesia, which will give the reader an insight on how to maximize the benefits of steroids while minimizing their side effects and complications. TAKE HOME MESSAGE Although steroid injections are generally considered effective and safe in the treatment of painful condition of limbs, their use in the treatment of chronic back pain is still controversial and serious complications have been reported. More studies on outcome and safety are warranted.
Collapse
Affiliation(s)
- Steven H S Wong
- Queen Elizabeth Hospital, Department of Anaesthesiology, Hong Kong.
| | | | | |
Collapse
|
33
|
Abstract
The glucocorticoids are the most potent anti-inflammatory drugs that we possess and are effective in a wide variety of diseases. Although their action is known to involve receptor mediated changes in gene transcription, the exact mechanisms whereby these bring about their pleiotropic action in inflammation are yet to be totally understood. Whilst many different genes are regulated by the glucocorticoids, we have identified one particular protein-annexin A1 (Anx-A1)-whose synthesis and release is strongly regulated by the glucocorticoids in many cell types. The biology of this protein, as revealed by studies using transgenic animals, peptide mimetics and neutralizing antibodies, speaks to its role as a key modulator of both of the innate and adaptive immune systems. The mechanism whereby this protein exerts its effects is likely to be through the FPR receptor family-a hitherto rather enigmatic family of G protein coupled receptors, which are increasingly implicated in the regulation of many inflammatory processes. Here we review some of the key findings that have led up to the elucidation of this key pathway in inflammatory resolution.
Collapse
|