1
|
Zhao D, Sha BX, Zeng LF, Liang GH, Huang HT, Pan JK, Liu J, Zhao S. Exploring and analyzing two aging related genes FPR1 and UCHL1 and their potential molecular mechanisms in aggravating lumbar disc herniation. J Orthop Surg Res 2024; 19:841. [PMID: 39695855 DOI: 10.1186/s13018-024-05257-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/10/2024] [Indexed: 12/20/2024] Open
Abstract
The aim of this research was to investigate dysregulated pivotal genes in individuals with lumbar disc herniation (LDH) to identify potential diagnostic biomarkers and treatment targets for LDH. Key aging-related genes in LDH were identified through multiple methods. Two dysregulated key genes (FPR1 and UCHL1) were finally identified, showing high diagnostic value in both training and external validation cohorts. Dysregulated expression of these hub genes established a detrimental cycle in LDH by promoting inflammatory response, immune infiltration, and aging progression. This highlights significant pathological alterations caused by these hub genes in LDH pathogenesis. The current study developed a novel genetic signature associated with aging that accurately diagnoses LDH while characterizing biological alterations in patients with this condition. And this genetic signature holds promise as an indicator to assist clinical decision-making. Moreover, identification of FPR1 and UCHL1 as pivotal genes presents potential prospects for targeted therapeutic interventions for LDH.
Collapse
Affiliation(s)
- Di Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Bang-Xin Sha
- The Fifth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ling-Feng Zeng
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Gui-Hong Liang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - He-Tao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Jian-Ke Pan
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China
| | - Jun Liu
- Bone and Joint Research Team of Degeneration and Injury, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510120, China.
- Guangdong Second Traditional Chinese Medicine Hospital (Guangdong Province Enginering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, 510095, China.
- The Fifth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Shuai Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou, University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
| |
Collapse
|
2
|
Peng J, He J, Lin L, Li Y, Xia Y. Neural Stem Cell Extracellular Vesicles Carrying YBX1 Inhibited Neuronal Pyroptosis Through Increasing m6A-modified GPR30 Stability and Expression in Ischemic Stroke. Transl Stroke Res 2023:10.1007/s12975-023-01210-z. [PMID: 37966628 DOI: 10.1007/s12975-023-01210-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Neural stem cell-derived extracellular vesicles (NSC-derived EVs) alleviated ischemic stroke (IS) by suppressing the activation of nucleotide-binding domain leucine-rich repeats family protein 3 (NLRP3) inflammasome and neuronal pyroptosis. However, the specific mechanism needs further investigation. qRT-qPCR, Western blotting, and immunofluorescence detected related gene expression. Immunofluorescent analyzed the expression of Ki-67, βIII-Tubulin (Tuj1), and GFAP. Lactate dehydrogenase (LDH) release and IL-1β and IL-18 levels were analyzed by LDH and ELISA kits. TTC staining evaluated the infarction of brain tissues. Flow cytometric analysis measured caspase-1 activity. M6A methylated RNA immunoprecipitation PCR (MeRIP-PCR) measured methylation levels of G protein-coupled receptor 30 (GPR30). RIP and Co-IP analyzed the interactions of Y box binding protein (YBX1)/GPR30, YBX1/IGF2BP1 and NLRP3/speckle-type POZ protein (SPOP), as well as the ubiquitination levels of NLRP3. NSC-derived EVs inhibited the ischemia-reperfusion (I/R) injury of rats and the neuronal pyroptosis induced by oxygen-glucose deprivation/reoxygenation (OGD/R). Knockdown of EVs carrying YBX1 or GPR30 silencing abolished these inhibiting effects. GPR30 mRNA and IGF2BP1 protein were enriched by YBX1 antibody. YBX1 enhanced the stability of m6A-modified GPR30 by interacting with IGF2BP1 and thus promoting GPR30 expression. Knockdown of IGF2BP1 suppressed the binding between YBX1 and GPR30 mRNA. GPR30 promoted NLRP3 ubiquitination by interacting with SPOP. EVs carrying YBX1 could reduce the infarction of brain tissues and inhibit neuronal pyroptosis in rats with I/R injury. NSC-derived EVs carrying YBX1 increased the stability of m6A-modified GPR30 by interacting with IGF2BP1; the upregulation of GPR30 inhibited the activation of NLRP3 inflammasome through promoting NLRP3 ubiquitination by SPOP, ultimately suppressing the neuronal pyroptosis in IS.
Collapse
Affiliation(s)
- Jun Peng
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Jun He
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Long Lin
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - You Li
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Hainan Province, Haikou, 570208, People's Republic of China.
| |
Collapse
|
3
|
Srivastava R, Singh K, Abouhashem AS, Kumar M, Kacar S, Verma SS, Mohanty SK, Sinha M, Ghatak S, Xuan Y, Sen CK. Human fetal dermal fibroblast-myeloid cell diversity is characterized by dominance of pro-healing Annexin1-FPR1 signaling. iScience 2023; 26:107533. [PMID: 37636079 PMCID: PMC10450526 DOI: 10.1016/j.isci.2023.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/06/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Fetal skin achieves scarless wound repair. Dermal fibroblasts play a central role in extracellular matrix deposition and scarring outcomes. Both fetal and gingival wound repair share minimal scarring outcomes. We tested the hypothesis that compared to adult skin fibroblasts, human fetal skin fibroblast diversity is unique and partly overlaps with gingival skin fibroblasts. Human fetal skin (FS, n = 3), gingiva (HGG, n = 13), and mature skin (MS, n = 13) were compared at single-cell resolution. Dermal fibroblasts, the most abundant cluster, were examined to establish a connectome with other skin cells. Annexin1-FPR1 signaling pathway was dominant in both FS as well as HGG fibroblasts and related myeloid cells while scanty in MS fibroblasts. Myeloid-specific FPR1-ORF delivered in murine wound edge using tissue nanotransfection (TNT) technology significantly enhanced the quality of healing. Pseudotime analyses identified the co-existence of an HGG fibroblast subset with FPR1high myeloid cells of fetal origin indicating common underlying biological processes.
Collapse
Affiliation(s)
- Rajneesh Srivastava
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kanhaiya Singh
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ahmed S. Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
- Sharkia Clinical Research Department, Ministry of Health, Zagazig, Egypt
| | - Manishekhar Kumar
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sedat Kacar
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sumit S. Verma
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sujit K. Mohanty
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mithun Sinha
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Xuan
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chandan K. Sen
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Indiana Center for Regenerative Medicine and Engineering, Indiana University Health Comprehensive Wound Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
4
|
Prevete N, Poto R, Marone G, Varricchi G. Unleashing the power of formyl peptide receptor 2 in cardiovascular disease. Cytokine 2023; 169:156298. [PMID: 37454543 DOI: 10.1016/j.cyto.2023.156298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/25/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
N-formyl peptide receptors (FPRs) are seven-transmembrane, G protein-coupled receptors with a wide distribution in immune and non-immune cells, recognizing N-formyl peptides from bacterial and mitochondrial origin and several endogenous signals. Three FPRs have been identified in humans: FPR1, FPR2, and FPR3. Most FPR ligands can activate a pro-inflammatory response, while a limited group of FPR agonists can elicit anti-inflammatory and homeostatic responses. Annexin A1 (AnxA1), a glucocorticoid-induced protein, its N-terminal peptide Ac2-26, and lipoxin A4 (LXA4), a lipoxygenase-derived eicosanoid mediator, exert significant immunomodulatory effects by interacting with FPR2 and/or FPR1. The ability of FPRs to recognize both ligands with pro-inflammatory or inflammation-resolving properties places them in a crucial position in the balance between activation against harmful events and maintaince of tissue integrity. A new field of investigation focused on the role of FPRs in the setting of heart injury. FPRs are expressed on cardiac macrophages, which are the predominant immune cells in the myocardium and play a key role in heart diseases. Several endogenous (AnxA1, LXA4) and synthetic compounds (compound 43, BMS-986235) reduced infarct size and promoted the resolution of inflammation via the activation of FPR2 on cardiac macrophages. Further studies should evaluate FPR2 role in other cardiovascular disorders.
Collapse
Affiliation(s)
- Nella Prevete
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology, National Research Council (CNR), 80131 Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy; World Allergy Organization (WAO), Center of Excellence (CoE), 80131 Naples, Italy.
| |
Collapse
|
5
|
Wickstead ES, Solito E, McArthur S. Promiscuous Receptors and Neuroinflammation: The Formyl Peptide Class. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122009. [PMID: 36556373 PMCID: PMC9786789 DOI: 10.3390/life12122009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/04/2022]
Abstract
Formyl peptide receptors, abbreviated as FPRs in humans, are G-protein coupled receptors (GPCRs) mainly found in mammalian leukocytes. However, they are also expressed in cell types crucial for homeostatic brain regulation, including microglia and blood-brain barrier endothelial cells. Thus, the roles of these immune-associated receptors are extensive, from governing cellular adhesion and directed migration through chemotaxis, to granule release and superoxide formation, to phagocytosis and efferocytosis. In this review, we will describe the similarities and differences between the two principal pro-inflammatory and anti-inflammatory FPRs, FPR1 and FPR2, and the evidence for their importance in the development of neuroinflammatory disease, alongside their potential as therapeutic targets.
Collapse
Affiliation(s)
- Edward S. Wickstead
- Department of Neurology, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence: (E.S.W.); (S.M.)
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
- Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples “Federico II”, 80131 Naples, Italy
| | - Simon McArthur
- Institute of Dentistry, Faculty of Medicine & Dentistry, Queen Mary University of London, Blizard Institute, 4, Newark Street, London E1 2AT, UK
- Correspondence: (E.S.W.); (S.M.)
| |
Collapse
|
6
|
Huang K, Wang Z, He Z, Li Y, Li S, Shen K, Zhu G, Liu Z, Lv S, Zhang C, Yang H, Yang X, Liu S. Downregulated formyl peptide receptor 2 expression in the epileptogenic foci of patients with focal cortical dysplasia type IIb and tuberous sclerosis complex. Immun Inflamm Dis 2022; 10:e706. [PMID: 36301030 PMCID: PMC9597500 DOI: 10.1002/iid3.706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Focal cortical dysplasia type IIb (FCDIIb) and tuberous sclerosis complex (TSC) show persistent neuroinflammation, which promotes epileptogenesis and epilepsy progression, suggesting that endogenous resolution of inflammation is inadequate to relieve neuronal network hyperexcitability. To explore the potential roles of formyl peptide receptor 2 (FPR2), which is a key regulator of inflammation resolution, in epilepsy caused by FCDIIb and TSC, we examined the expression and cellular distribution of FPR2. METHOD The expression of FPR2 and nuclear factor-κB (NF-κB) signaling pathway was examined by real-time PCR, western blots, and analyzed via one-way analysis of variance. The distribution of FPR2 was detected using immunostaining. The expression of resolvin D1 (RvD1, the endogenous ligand of FPR2) was observed via enzyme-linked immunosorbent assay. Pearson's correlation test was used to evaluate the correlation between the expression levels of FPR2 and RvD1 and the clinical variants. RESULTS The expression of FPR2 was significantly lower in FCDIIb (p = .0146) and TSC (p = .0006) cortical lesions than in controls, as was the expression of RvD1 (FCDIIb: p = .00431; TSC: p = .0439). Weak FPR2 immunoreactivity was observed in dysmorphic neurons (DNs), balloon cells (BCs), and giant cells (GCs) in FCDIIb and TSC tissues. Moreover, FPR2 was mainly distributed in dysplastic neurons; it was sparse in microglia and nearly absent in astrocytes. The NF-κB pathway was significantly activated in patients with FCDIIb and TSC, and the protein level of NF-κB was negatively correlated with the protein level of FPR2 (FCDIIb: p = .00395; TSC: p = .0399). In addition, the protein level of FPR2 was negatively correlated with seizure frequency in FCDIIb (p = .0434) and TSC (p = .0351) patients. CONCLUSION In summary, these results showed that the expression and specific distribution of FPR2 may be involved in epilepsy caused by FCDIIb and TSC, indicating that downregulation of FPR2 mediated the dysfunction of neuroinflammation resolution in FCDIIb and TSC.
Collapse
Affiliation(s)
- Kaixuan Huang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Zhongke Wang
- Department of NeurosurgeryArmed Police Hospital of ChongqingChongqingChina
| | - Zeng He
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yang Li
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shujing Li
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Kaifeng Shen
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Gang Zhu
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Zhonghong Liu
- Department of NeurosurgeryArmed Police Hospital of ChongqingChongqingChina
| | - Shengqing Lv
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Chunqing Zhang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Hui Yang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Xiaolin Yang
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Shiyong Liu
- Department of Neurosurgery, Epilepsy Research Center of PLA, Xinqiao HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
7
|
Ma H, Guo X, Wang Z, Han M, Liu H. Therapeutic potential of WKYMVm in diseases. Front Pharmacol 2022; 13:986963. [PMID: 36120322 PMCID: PMC9479759 DOI: 10.3389/fphar.2022.986963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/08/2022] [Indexed: 11/17/2022] Open
Abstract
The synthetic hexapeptide WKYMVm, screened from a synthetic peptide library, has been identified as an agonist of FPRs with the strongest activating effect on FPR2. WKYMVm plays an anti-inflammatory role in most inflammatory diseases by increasing the chemotaxis of phagocytes and regulating the secretion of inflammatory factors. WKYMVm can inhibit or promote the progression of different types of tumors, which depends on the regulation of WKYMVm on various components such as immune cells, inflammatory factors, chemokines, and tumor epithelial cells. Another major function of WKYMVm is to promote angiogenesis, which is reflected in its therapeutic value in ischemic diseases, wound healing and bone repair. In addition to the above functions, this paper also reviews the effects of WKYMVm on fibrosis, insulin resistance, osteolytic diseases and neurodegenerative diseases. By summarizing related studies, this review can increase people’s comprehensive understanding of WKYMVm, promote its broad and in-depth research, and help to exert its therapeutic value as soon as possible.
Collapse
Affiliation(s)
- Huan Ma
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoming Guo
- Department of Endoscopy, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiguo Wang
- Department of Endoscopy, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Mei Han
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- *Correspondence: Hui Liu, , Mei Han,
| | - Hui Liu
- Department of Gastroenterology, Second Hospital of Dalian Medical University, Dalian, Liaoning, China
- *Correspondence: Hui Liu, , Mei Han,
| |
Collapse
|
8
|
Rodocker HI, Bordbar A, Larson MJE, Biltz RG, Wangler L, Fadda P, Godbout JP, Tedeschi A. Breaking Mental Barriers Promotes Recovery After Spinal Cord Injury. Front Mol Neurosci 2022; 15:868563. [PMID: 35875670 PMCID: PMC9301320 DOI: 10.3389/fnmol.2022.868563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/14/2022] [Indexed: 11/23/2022] Open
Abstract
Functional recovery after spinal cord injury (SCI) often proves difficult as physical and mental barriers bar survivors from enacting their designated rehabilitation programs. We recently demonstrated that adult mice administered gabapentinoids, clinically approved drugs prescribed to mitigate chronic neuropathic pain, recovered upper extremity function following cervical SCI. Given that rehabilitative training enhances neuronal plasticity and promotes motor recovery, we hypothesized that the combination of an aerobic-based rehabilitation regimen like treadmill training with gabapentin (GBP) administration will maximize recovery in SCI mice by strengthening synaptic connections along the sensorimotor axis. Whereas mice administered GBP recovered forelimb functions over the course of weeks and months following SCI, no additive forelimb recovery as the result of voluntary treadmill training was noted in these mice. To our surprise, we also failed to find an additive effect in mice administered vehicle. As motivation is crucial in rehabilitation interventions, we scored active engagement toward the rehabilitation protocol and found that mice administered GBP were consistently participating in the rehabilitation program. In contrast, mice administered vehicle exhibited a steep decline in participation, especially at chronic time points. Whereas neuroinflammatory gene expression profiles were comparable between experimental conditions, we discovered that mice administered GBP had increased hippocampal neurogenesis and exhibited less anxiety-like behavior after SCI. We also found that an external, social motivator effectively rescues participation in mice administered vehicle and promotes forelimb recovery after chronic SCI. Thus, not only does a clinically relevant treatment strategy preclude the deterioration of mental health after chronic SCI, but group intervention strategies may prove to be physically and emotionally beneficial for SCI individuals.
Collapse
Affiliation(s)
- Haven I. Rodocker
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Arman Bordbar
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Molly J. E. Larson
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Rebecca G. Biltz
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Lynde Wangler
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Paolo Fadda
- Department of Cancer Biology, The Ohio State University, Columbus, OH, United States
| | - Jonathan P. Godbout
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
- Chronic Brain Injury Program, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Zhang J, Li Z, Fan M, Jin W. Lipoxins in the Nervous System: Brighter Prospects for Neuroprotection. Front Pharmacol 2022; 13:781889. [PMID: 35153778 PMCID: PMC8826722 DOI: 10.3389/fphar.2022.781889] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/07/2022] [Indexed: 12/28/2022] Open
Abstract
Lipoxins (LXs) are generated from arachidonic acid and are involved in the resolution of inflammation and confer protection in a variety of pathological processes. In the nervous system, LXs exert an array of protective effects against neurological diseases, including ischemic or hemorrhagic stroke, neonatal hypoxia-ischemia encephalopathy, brain and spinal cord injury, Alzheimer's disease, multiple sclerosis, and neuropathic pain. Lipoxin administration is a potential therapeutic strategy in neurological diseases due to its notable efficiency and unique superiority regarding safety. Here, we provide an overview of LXs in terms of their synthesis, signaling pathways and neuroprotective evidence. Overall, we believe that, along with advances in lipoxin-related drug design, LXs will bring brighter prospects for neuroprotection.
Collapse
Affiliation(s)
- Jiayu Zhang
- Graduate School of Hebei Medical University, Shijiazhuang, China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Zhe Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Mingyue Fan
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| | - Wei Jin
- Department of Neurology, Hebei General Hospital, Shijiazhuang, China
| |
Collapse
|
10
|
Stimulation of the Migration and Expansion of Adult Mouse Neural Stem Cells by the FPR2-Specific Peptide WKYMVm. Life (Basel) 2021; 11:life11111248. [PMID: 34833124 PMCID: PMC8622362 DOI: 10.3390/life11111248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/21/2022] Open
Abstract
Neural stem cells (NSCs) are multipotent cells capable of self-renewal and differentiation into different nervous system cells. Mouse NSCs (mNSCs) are useful tools for studying neurogenesis and the therapeutic applications of neurodegenerative diseases in mammals. Formyl peptide receptor 2 (FPR2), expressed in the central nervous system and brain, is involved in the migration and differentiation of murine embryonic-derived NSCs. In this study, we explored the effect of FPR2 activation in adult mNSCs using the synthetic peptide Trp-Lys-Tyr-Met-Val-D-Met-NH2 (WKYMVm), an agonist of FPR2. After isolation of NSCs from the subventricular zone of the adult mouse brain, they were cultured in two culture systems—neurospheres or adherent monolayers—to demonstrate the expression of NSC markers and phenotypes. Under different conditions, mNSCs differentiated into neurons and glial cells such as astrocytes, microglia, and oligodendrocytes. Treatment with WKYMVm stimulated the chemotactic migration of mNSCs. Moreover, WKYMVm-treated mNSCs were found to promote proliferation; this result was confirmed by the expansion of mNSCs in Matrigel and the increase in the number of Ki67-positive cells. Incubation of mNSCs with WKYMVm in a supplement-free medium enhanced the survival rate of the mNSCs. Together, these results suggest that WKYMVm-induced activation of FPR2 stimulates cellular responses in adult NSCs.
Collapse
|
11
|
Cao J, Gan H, Xiao H, Chen H, Jian D, Jian D, Zhai X. Key protein-coding genes related to microglia in immune regulation and inflammatory response induced by epilepsy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:9563-9578. [PMID: 34814358 DOI: 10.3934/mbe.2021469] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Several studies have shown a link between immunity, inflammatory processes, and epilepsy. Active neuroinflammation and marked immune cell infiltration occur in epilepsy of diverse etiologies. Microglia, as the first line of defense in the central nervous system, are the main effectors of neuroinflammatory processes. Discovery of new biomarkers associated with microglia activation after epileptogenesis indicates that targeting specific molecules may help control seizures. In this research, we used a combination of several bioinformatics approaches, including RNA sequencing, to explore differentially expressed genes (DEGs) in epileptic lesions and control samples, and to construct a protein-protein interaction (PPI) network for DEGs, which was examined utilizing plug-ins in Cytoscape software. Finally, we aimed to identify 10 hub genes in immune and inflammation-related sub-networks, which were subsequently validated in real-time quantitative polymerase chain reaction analysis in a mouse model of kainic acid-induced epilepsy. The expression patterns of nine genes were consistent with sequencing outcomes. Meanwhile, several genes, including CX3CR1, CX3CL1, GPR183, FPR1, P2RY13, P2RY12 and LPAR5, were associated with microglial activation and migration, providing novel candidate targets for immunotherapy in epilepsy and laying the foundation for further research.
Collapse
Affiliation(s)
- Jing Cao
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400010, China
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400010, China
| | - Hui Gan
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400010, China
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400010, China
| | - Han Xiao
- Ministry of Education Key Laboratory of Child Development and Disorders, Childrenӳ Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing 400010, China
| | - Hui Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, Childrenӳ Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing 400010, China
| | - Dan Jian
- Ministry of Education Key Laboratory of Child Development and Disorders, Childrenӳ Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing 400010, China
| | - Dan Jian
- Institute of Neuroscience, Chongqing Medical University, Chongqing 400010, China
- Department of Pathology, Chongqing Medical University, Chongqing 400010, China
| | - Xuan Zhai
- Ministry of Education Key Laboratory of Child Development and Disorders, Childrenӳ Hospital of Chongqing Medical University, Chongqing, P.R China, Chongqing 400010, China
| |
Collapse
|
12
|
Zhu J, Li L, Ding J, Huang J, Shao A, Tang B. The Role of Formyl Peptide Receptors in Neurological Diseases via Regulating Inflammation. Front Cell Neurosci 2021; 15:753832. [PMID: 34650406 PMCID: PMC8510628 DOI: 10.3389/fncel.2021.753832] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/02/2021] [Indexed: 01/02/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a group of G protein-coupled cell surface receptors that play important roles in host defense and inflammation. Owing to the ubiquitous expression of FPRs throughout different cell types and since they interact with structurally diverse chemotactic agonists, they have a dual function in inflammatory processes, depending on binding with different ligands so that accelerate or inhibit key intracellular kinase-based regulatory pathways. Neuroinflammation is closely associated with the pathogenesis of neurodegenerative diseases, neurogenic tumors and cerebrovascular diseases. From recent studies, it is clear that FPRs are important biomarkers for neurological diseases as they regulate inflammatory responses by monitoring glial activation, accelerating neural differentiation, regulating angiogenesis, and controlling blood brain barrier (BBB) permeability, thereby affecting neurological disease progression. Given the complex mechanisms of neurological diseases and the difficulty of healing, we are eager to find new and effective therapeutic targets. Here, we review recent research about various mechanisms of the effects generated after FPR binding to different ligands, role of FPRs in neuroinflammation as well as the development and prognosis of neurological diseases. We summarize that the FPR family has dual inflammatory functional properties in central nervous system. Emphasizing that FPR2 acts as a key molecule that mediates the active resolution of inflammation, which binds with corresponding receptors to reduce the expression and activation of pro-inflammatory composition, govern the transport of immune cells to inflammatory tissues, and restore the integrity of the BBB. Concurrently, FPR1 is essentially related to angiogenesis, cell proliferation and neurogenesis. Thus, treatment with FPRs-modulation may be effective for neurological diseases.
Collapse
Affiliation(s)
- Jiahui Zhu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lingfei Li
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiao Ding
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinyu Huang
- Department of Cardiology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Tang
- Department of Neurology, The Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Identification of Potential Core Genes in Parkinson's Disease Using Bioinformatics Analysis. PARKINSON'S DISEASE 2021; 2021:1690341. [PMID: 34580608 PMCID: PMC8464436 DOI: 10.1155/2021/1690341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/21/2021] [Accepted: 09/08/2021] [Indexed: 12/31/2022]
Abstract
Purpose This study aimed to explore new core genes related to the occurrence of Parkinson's disease (PD) and core genes that can lead to the progression of PD. Methods The expression profile data of GSE42966, which contained six substantia nigra tissues isolated from normal individuals and nine substantia nigra tissues isolated from patients with PD, were obtained from Gene Expression Omnibus. Differentially expressed genes (DEGs) were identified, followed by functional enrichment analysis and protein-protein interaction (PPI) network construction. We then identified 10 hub genes and analyzed their expression in different Braak stages. Results A total of 773 DEGs were identified that were significantly enriched in metabolic pathways. Ten hub genes were identified through the PPI network, namely, GNG3, MAPK1, FPR1, ATP5B, GNG2, PRKACA, HRAS, HSPA8, PSAP, and GABBR2. The expression of HRAS was different in patients with PD with Braak stages 3 and 4. Conclusion These 10 hub genes and the metabolic pathways they are enriched in may be involved in the pathogenesis of PD. HRAS may have potential value in predicting the progression of PD.
Collapse
|
14
|
Tylek K, Trojan E, Leśkiewicz M, Regulska M, Bryniarska N, Curzytek K, Lacivita E, Leopoldo M, Basta-Kaim A. Time-Dependent Protective and Pro-Resolving Effects of FPR2 Agonists on Lipopolysaccharide-Exposed Microglia Cells Involve Inhibition of NF-κB and MAPKs Pathways. Cells 2021; 10:cells10092373. [PMID: 34572022 PMCID: PMC8472089 DOI: 10.3390/cells10092373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 02/07/2023] Open
Abstract
Prolonged or excessive microglial activation may lead to disturbances in the resolution of inflammation (RoI). The importance of specialized pro-resolving lipid mediators (SPMs) in RoI has been highlighted. Among them, lipoxins (LXA4) and aspirin-triggered lipoxin A4 (AT-LXA4) mediate beneficial responses through the activation of N-formyl peptide receptor-2 (FPR2). We aimed to shed more light on the time-dependent protective and anti-inflammatory impact of the endogenous SPMs, LXA4, and AT-LXA4, and of a new synthetic FPR2 agonist MR-39, in lipopolysaccharide (LPS)-exposed rat microglial cells. Our results showed that LXA4, AT-LXA4, and MR-39 exhibit a protective and pro-resolving potential in LPS-stimulated microglia, even if marked differences were apparent regarding the time dependency and efficacy of inhibiting particular biomarkers. The LXA4 action was found mainly after 3 h of LPS stimulation, and the AT-LXA4 effect was varied in time, while MR-39′s effect was mainly observed after 24 h of stimulation by endotoxin. MR-39 was the only FPR2 ligand that attenuated LPS-evoked changes in the mitochondrial membrane potential and diminished the ROS and NO release. Moreover, the LPS-induced alterations in the microglial phenotype were modulated by LXA4, AT-LXA4, and MR-39. The anti-inflammatory effect of MR-39 on the IL-1β release was mediated through FPR2. All tested ligands inhibited TNF-α production, while AT-LXA4 and MR-39 also diminished IL-6 levels in LPS-stimulated microglia. The favorable action of LXA4 and MR-39 was mediated through the inhibition of ERK1/2 phosphorylation. AT-LXA4 and MR39 diminished the phosphorylation of the transcription factor NF-κB, while AT-LXA4 also affected p38 kinase phosphorylation. Our results suggest that new pro-resolving synthetic mediators can represent an attractive treatment option for the enhancement of RoI, and that FPR2 can provide a perspective as a target in immune-related brain disorders.
Collapse
Affiliation(s)
- Kinga Tylek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Ewa Trojan
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Monika Leśkiewicz
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Magdalena Regulska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Natalia Bryniarska
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Katarzyna Curzytek
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
| | - Enza Lacivita
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Marcello Leopoldo
- Department of Pharmacy—Drug Sciences, University of Bari, Via Orabona 4, 70125 Bari, Italy; (E.L.); (M.L.)
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland; (K.T.); (E.T.); (M.L.); (M.R.); (N.B.); (K.C.)
- Correspondence: ; Tel.: +48-12-662-32-73
| |
Collapse
|
15
|
Xu YW, Yang JS, Kang DZ, Yao PS. RETRACTED ARTICLE: Astrocytes Regulate Differentiation and Glutamate Uptake of Glioma Stem Cells via Formyl Peptide Receptor. Cell Mol Neurobiol 2021; 41:1389. [PMID: 32474726 DOI: 10.1007/s10571-020-00886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/25/2020] [Indexed: 11/30/2022]
Affiliation(s)
- Ya-Wen Xu
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, NO. 20 Chazhong Road, Taijiang District, Fuzhou, 350004, Fujian, China
| | - Jin-Shan Yang
- Department of Neurology, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - De-Zhi Kang
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, NO. 20 Chazhong Road, Taijiang District, Fuzhou, 350004, Fujian, China.
| | - Pei-Sen Yao
- Department of Neurosurgery, First Affiliated Hospital of Fujian Medical University, NO. 20 Chazhong Road, Taijiang District, Fuzhou, 350004, Fujian, China.
| |
Collapse
|
16
|
The effects of genotype on inflammatory response in hippocampal progenitor cells: A computational approach. Brain Behav Immun Health 2021; 15:100286. [PMID: 34345870 PMCID: PMC8261829 DOI: 10.1016/j.bbih.2021.100286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/08/2023] Open
Abstract
Cell culture models are valuable tools to study biological mechanisms underlying health and disease in a controlled environment. Although their genotype influences their phenotype, subtle genetic variations in cell lines are rarely characterised and taken into account for in vitro studies. To investigate how the genetic makeup of a cell line might affect the cellular response to inflammation, we characterised the single nucleotide variants (SNPs) relevant to inflammation-related genes in an established hippocampal progenitor cell line (HPC0A07/03C) that is frequently used as an in vitro model for hippocampal neurogenesis (HN). SNPs were identified using a genotyping array, and genes associated with chronic inflammatory and neuroinflammatory response gene ontology terms were retrieved using the AmiGO application. SNPs associated with these genes were then extracted from the genotyping dataset, for which a literature search was conducted, yielding relevant research articles for a total of 17 SNPs. Of these variants, 10 were found to potentially affect hippocampal neurogenesis whereby a majority (n=7) is likely to reduce neurogenesis under inflammatory conditions. Taken together, the existing literature seems to suggest that all stages of hippocampal neurogenesis could be negatively affected due to the genetic makeup in HPC0A07/03C cells under inflammation. Additional experiments will be needed to validate these specific findings in a laboratory setting. However, this computational approach already confirms that in vitro studies in general should control for cell lines subtle genetic variations which could mask or exacerbate findings.
Collapse
|
17
|
Wickstead ES, Irving MA, Getting SJ, McArthur S. Exploiting formyl peptide receptor 2 to promote microglial resolution: a new approach to Alzheimer's disease treatment. FEBS J 2021; 289:1801-1822. [PMID: 33811735 DOI: 10.1111/febs.15861] [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: 01/11/2021] [Revised: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease and dementia are among the most significant current healthcare challenges given the rapidly growing elderly population, and the almost total lack of effective therapeutic interventions. Alzheimer's disease pathology has long been considered in terms of accumulation of amyloid beta and hyperphosphorylated tau, but the importance of neuroinflammation in driving disease has taken greater precedence over the last 15-20 years. Inflammatory activation of the primary brain immune cells, the microglia, has been implicated in Alzheimer's pathogenesis through genetic, preclinical, imaging and postmortem human studies, and strategies to regulate microglial activity may hold great promise for disease modification. Neuroinflammation is necessary for defence of the brain against pathogen invasion or damage but is normally self-limiting due to the engagement of endogenous pro-resolving circuitry that terminates inflammatory activity, a process that appears to fail in Alzheimer's disease. Here, we discuss the potential for a major regulator and promoter of resolution, the receptor FPR2, to restrain pro-inflammatory microglial activity, and propose that it may serve as a valuable target for therapeutic investigation in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Murray A Irving
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| | - Stephen J Getting
- College of Liberal Arts & Sciences, School of Life Sciences, University of Westminster, London, UK
| | - Simon McArthur
- Institute of Dentistry, Barts and the London School of Medicine & Dentistry, Blizard Institute, Queen Mary, University of London, UK
| |
Collapse
|
18
|
Edwards JM, Roy S, Galla SL, Tomcho JC, Bearss NR, Waigi EW, Mell B, Cheng X, Saha P, Vijay-Kumar M, McCarthy CG, Joe B, Wenceslau CF. FPR-1 (Formyl Peptide Receptor-1) Activation Promotes Spontaneous, Premature Hypertension in Dahl Salt-Sensitive Rats. Hypertension 2021; 77:1191-1202. [PMID: 33641367 DOI: 10.1161/hypertensionaha.120.16237] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jonnelle M Edwards
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Shaunak Roy
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sarah L Galla
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jeremy C Tomcho
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Nicole R Bearss
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Emily W Waigi
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Blair Mell
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Xi Cheng
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Piu Saha
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Matam Vijay-Kumar
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cameron G McCarthy
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Bina Joe
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Camilla F Wenceslau
- From the Department of Pharmacology and Physiology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| |
Collapse
|
19
|
Xia W, Zhu J, Wang X, Tang Y, Zhou P, Hou M, Li S. ANXA1 directs Schwann cells proliferation and migration to accelerate nerve regeneration through the FPR2/AMPK pathway. FASEB J 2020; 34:13993-14005. [PMID: 32856352 DOI: 10.1096/fj.202000726rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
Many factors are involved in the process of nerve regeneration. Understanding the mechanisms regarding how these factors promote an efficient remyelination is crucial to deciphering the molecular and cellular processes required to promote nerve repair. Schwann cells (SCs) play a central role in the process of peripheral nerve repair/regeneration. Using a model of facial nerve crush injury and repair, we identified Annexin A1 (ANXA1) as the extracellular trigger of SC proliferation and migration. ANXA1 activated formyl peptide receptor 2 (FPR2) receptors and the downstream adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling cascade, leading to SC proliferation and migration in vitro. SCs lacking FPR2 or AMPK displayed a defect in proliferation and migration. After facial nerve injury (FNI), ANXA1 promoted the proliferation of SCs and nerve regeneration in vivo. Collectively, these data identified the ANXA1/FPR2/AMPK axis as an important pathway in SC proliferation and migration. ANXA1-induced remyelination and SC proliferation promotes FNI regeneration.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Jin Zhu
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyi Wang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Yinda Tang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhou
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shiting Li
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Diagnosis and Treatment of Cranial Nerve Diseases, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Formyl Peptide Receptor 1 Signaling in Acute Inflammation and Neural Differentiation Induced by Traumatic Brain Injury. BIOLOGY 2020; 9:biology9090238. [PMID: 32825368 PMCID: PMC7563302 DOI: 10.3390/biology9090238] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/31/2022]
Abstract
Traumatic brain injury (TBI) is a shocking disease frequently followed by behavioral disabilities, including risk of cerebral atrophy and dementia. N-formylpeptide receptor 1 (FPR1) is expressed in cells and neurons in the central nervous system. It is involved in inflammatory processes and during the differentiation process in the neural stem cells. We investigate the effect of the absence of Fpr1 gene expression in mice subjected to TBI from the early stage of acute inflammation to neurogenesis and systematic behavioral testing four weeks after injury. C57BL/6 animals and Fpr1 KO mice were subjected to TBI and sacrificed 24 h or four weeks after injury. Twenty-four hours after injury, TBI Fpr1 KO mice showed reduced histological impairment, tissue damage and acute inflammation (MAPK activation, NF-κB signaling induction, NRLP3 inflammasome pathway activation and oxidative stress increase). Conversely, four weeks after TBI, the Fpr1 KO mice showed reduced survival of the proliferated cells in the Dentate Gyrus compared to the WT group. Behavioral analysis confirmed this trend. Moreover, TBI Fpr1 KO animals displayed reduced neural differentiation (evaluated by beta-III tubulin expression) and upregulation of astrocyte differentiation (evaluated by GFAP expression). Collectively, our study reports that, immediately after TBI, Fpr1 increased acute inflammation, while after four weeks, Fpr1 promoted neurogenesis.
Collapse
|
21
|
Liu GJ, Tao T, Wang H, Zhou Y, Gao X, Gao YY, Hang CH, Li W. Functions of resolvin D1-ALX/FPR2 receptor interaction in the hemoglobin-induced microglial inflammatory response and neuronal injury. J Neuroinflammation 2020; 17:239. [PMID: 32795323 PMCID: PMC7429751 DOI: 10.1186/s12974-020-01918-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/04/2020] [Indexed: 02/09/2023] Open
Abstract
Background Early brain injury (EBI) has been thought to be a key factor affecting the prognosis of subarachnoid hemorrhage (SAH). Many pathologies are involved in EBI, with inflammation and neuronal death being crucial to this process. Resolvin D1 (RvD1) has shown superior anti-inflammatory properties by interacting with lipoxin A4 receptor/formyl peptide receptor 2 (ALX/FPR2) in various diseases. However, it remains not well described about its role in the central nervous system (CNS). Thus, the goal of the present study was to elucidate the potential functions of the RvD1-ALX/FPR2 interaction in the brain after SAH. Methods We used an in vivo model of endovascular perforation and an in vitro model of hemoglobin (Hb) exposure as SAH models in the current study. RvD1 was used at a concentration of 25 nM in our experiments. Western blotting, quantitative polymerase chain reaction (qPCR), immunofluorescence, and other chemical-based assays were performed to assess the cellular localizations and time course fluctuations in ALX/FPR2 expression, evaluate the effects of RvD1 on Hb-induced primary microglial activation and neuronal damage, and confirm the role of ALX/FPR2 in the function of RvD1. Results ALX/FPR2 was expressed on both microglia and neurons, but not astrocytes. RvD1 exerted a good inhibitory effect in the microglial pro-inflammatory response induced by Hb, possibly by regulating the IRAK1/TRAF6/NF-κB or MAPK signaling pathways. RvD1 could also potentially attenuate Hb-induced neuronal oxidative damage and apoptosis. Finally, the mRNA expression of IRAK1/TRAF6 in microglia and GPx1/bcl-xL in neurons was reversed by the ALX/FPR2-specific antagonist Trp-Arg-Trp-Trp-Trp-Trp-NH2 (WRW4), indicating that ALX/FPR2 could mediate the neuroprotective effects of RvD1. Conclusions The results of the present study indicated that the RvD1-ALX/FPR2 interaction could potentially play dual roles in the CNS, as inhibiting Hb promoted microglial pro-inflammatory polarization and ameliorating Hb induced neuronal oxidant damage and death. These results shed light on a good therapeutic target (ALX/FPR2) and a potential effective drug (RvD1) for the treatment of SAH and other inflammation-associated brain diseases.
Collapse
Affiliation(s)
- Guang-Jie Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Han Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Southern Medical University (Guangzhou), Nanjing, China
| | - Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuan Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
22
|
Sowmithra S, Jain NK, Datta I. Evaluating In Vitro Neonatal Hypoxic-Ischemic Injury Using Neural Progenitors Derived from Human Embryonic Stem Cells. Stem Cells Dev 2020; 29:929-951. [DOI: 10.1089/scd.2020.0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Sowmithra Sowmithra
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Nishtha Kusum Jain
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| | - Indrani Datta
- Department of Biophysics, National Institute of Mental Health and Neurosciences, Institute of National Importance, Bengaluru, India
| |
Collapse
|
23
|
Cussell PJ, Gomez Escalada M, Milton NG, Paterson AW. The N-formyl peptide receptors: contemporary roles in neuronal function and dysfunction. Neural Regen Res 2020; 15:1191-1198. [PMID: 31960798 PMCID: PMC7047793 DOI: 10.4103/1673-5374.272566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 09/20/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
N-formyl peptide receptors (FPRs) were first identified upon phagocytic leukocytes, but more than four decades of research has unearthed a plethora of non-myeloid roles for this receptor family. FPRs are expressed within neuronal tissues and markedly in the central nervous system, where FPR interactions with endogenous ligands have been implicated in the pathophysiology of several neurodegenerative diseases including Alzheimer's disease and Parkinson's disease, as well as neurological cancers such as neuroblastoma. Whilst the homeostatic function of FPRs in the nervous system is currently undefined, a variety of novel physiological roles for this receptor family in the neuronal context have been posited in both human and animal settings. Rapid developments in recent years have implicated FPRs in the process of neurogenesis and neuronal differentiation which, upon greater characterisation, could represent a novel pharmacological target for neuronal regeneration therapies that may be used in the treatment of brain/spinal cord injury, stroke and neurodegeneration. This review aims to summarize the recent progress made to determine the physiological role of FPRs in a neuronal setting, and to put forward a case for FPRs as a novel pharmacological target for conditions of the nervous system, and for their potential to open the door to novel neuronal regeneration therapies.
Collapse
Affiliation(s)
- Peter J.G. Cussell
- Centre for Biomedical Science Research, School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, UK
| | - Margarita Gomez Escalada
- Centre for Biomedical Science Research, School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, UK
| | - Nathaniel G.N. Milton
- Centre for Biomedical Science Research, School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, UK
| | - Andrew W.J. Paterson
- Centre for Biomedical Science Research, School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, UK
| |
Collapse
|
24
|
Krashia P, Cordella A, Nobili A, La Barbera L, Federici M, Leuti A, Campanelli F, Natale G, Marino G, Calabrese V, Vedele F, Ghiglieri V, Picconi B, Di Lazzaro G, Schirinzi T, Sancesario G, Casadei N, Riess O, Bernardini S, Pisani A, Calabresi P, Viscomi MT, Serhan CN, Chiurchiù V, D'Amelio M, Mercuri NB. Blunting neuroinflammation with resolvin D1 prevents early pathology in a rat model of Parkinson's disease. Nat Commun 2019; 10:3945. [PMID: 31477726 PMCID: PMC6718379 DOI: 10.1038/s41467-019-11928-w] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 08/12/2019] [Indexed: 12/31/2022] Open
Abstract
Neuroinflammation is one of the hallmarks of Parkinson’s disease (PD) and may contribute to midbrain dopamine (DA) neuron degeneration. Recent studies link chronic inflammation with failure to resolve early inflammation, a process operated by specialized pro-resolving mediators, including resolvins. However, the effects of stimulating the resolution of inflammation in PD – to modulate disease progression – still remain unexplored. Here we show that rats overexpressing human α-synuclein (Syn) display altered DA neuron properties, reduced striatal DA outflow and motor deficits prior to nigral degeneration. These early alterations are coupled with microglia activation and perturbations of inflammatory and pro-resolving mediators, namely IFN-γ and resolvin D1 (RvD1). Chronic and early RvD1 administration in Syn rats prevents central and peripheral inflammation, as well as neuronal dysfunction and motor deficits. We also show that endogenous RvD1 is decreased in human patients with early-PD. Our results suggest there is an imbalance between neuroinflammatory and pro-resolving processes in PD. Resolvins are endogenous lipids with pro-resolving activity. Here the authors find that rats overexpressing human α-synuclein show defects in dopamine signalling before dopamine cell loss, and that this is associated with low Resolvin D1 levels and neuroinflammation.
Collapse
Affiliation(s)
- Paraskevi Krashia
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Medicine and Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128, Rome, Italy
| | - Alberto Cordella
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Annalisa Nobili
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Medicine and Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128, Rome, Italy
| | - Livia La Barbera
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Mauro Federici
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Alessandro Leuti
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Medicine and Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128, Rome, Italy
| | - Federica Campanelli
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Giuseppina Natale
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Gioia Marino
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Valeria Calabrese
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Francescangelo Vedele
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Veronica Ghiglieri
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Philosophy, Human, Social and Educational Sciences, University of Perugia, 06123, Perugia, Italy
| | - Barbara Picconi
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Giulia Di Lazzaro
- Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Giulia Sancesario
- Department of Clinical and Behavioural Neurology, IRCCS Santa Lucia Foundation, 00143, Rome, Italy
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, 72076, Tübingen, Germany
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Antonio Pisani
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy
| | - Paolo Calabresi
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Neurology Clinic, Department of Medicine, University of Perugia, Santa Maria della Misericordia Hospital, 06156, Perugia, Italy
| | - Maria Teresa Viscomi
- Institute of Histology and Embryology, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Charles Nicholas Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anaesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, 02115, Boston, MA, USA
| | - Valerio Chiurchiù
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Medicine and Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128, Rome, Italy
| | - Marcello D'Amelio
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy.,Department of Medicine and Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Experimental Neurosciences, IRCCS Santa Lucia Foundation, 00143, Rome, Italy. .,Department of Systems Medicine, University of Rome 'Tor Vergata', 00133, Rome, Italy.
| |
Collapse
|
25
|
Yazdi A, Ghasemi‐Kasman M, Javan M. Possible regenerative effects of fingolimod (FTY720) in multiple sclerosis disease: An overview on remyelination process. J Neurosci Res 2019; 98:524-536. [DOI: 10.1002/jnr.24509] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 07/19/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Azadeh Yazdi
- Department of Physiology, School of Medicine Isfahan University of Medical Sciences Isfahan Iran
| | - Maryam Ghasemi‐Kasman
- Cellular and Molecular Biology Research Center Health Research Institute, Babol University of Medical Sciences Babol Iran
- Neuroscience Research Center Health Research Institute, Babol University of Medical Sciences Babol Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences Tarbiat Modares University Tehran Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center Royan Institute for Stem Cell Biology and Technology, ACECR Tehran Iran
| |
Collapse
|
26
|
Romano M, Patruno S, Pomilio A, Recchiuti A. Proresolving Lipid Mediators and Receptors in Stem Cell Biology: Concise Review. Stem Cells Transl Med 2019; 8:992-998. [PMID: 31187940 PMCID: PMC6766599 DOI: 10.1002/sctm.19-0078] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/19/2019] [Indexed: 12/29/2022] Open
Abstract
Accumulating evidence indicates that stem cells (SCs) possess immunomodulatory, anti‐inflammatory, and prohealing properties. The mechanisms underlying these functions are being investigated with the final goal to set a solid background for the clinical use of SCs and/or their derivatives. Specialized proresolving lipid mediators (SPMs) are small lipids formed by the enzymatic metabolism of polyunsaturated fatty acids. They represent a leading class of molecules that actively and timely regulate the resolution of inflammation and promote tissue/organ repair. SC formation of these mediators as well as expression of their receptors has been recently reported, suggesting that SPMs may be involved in the immunomodulatory, proresolving functions of SCs. In the present review, we summarize the current knowledge on SPMs in SCs, focusing on biosynthetic pathways, receptors, and bioactions, with the intent to provide an integrated view of SPM impact on SC biology. stem cells translational medicine2019;8:992–998
Collapse
Affiliation(s)
- Mario Romano
- Department of Medical, Oral, and Biotechnological Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,StemTech Group, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center on Aging Sciences and Translational Medicine (CeSI-MeT), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Sara Patruno
- Department of Medical, Oral, and Biotechnological Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,StemTech Group, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center on Aging Sciences and Translational Medicine (CeSI-MeT), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Antonella Pomilio
- Department of Medical, Oral, and Biotechnological Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,StemTech Group, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center on Aging Sciences and Translational Medicine (CeSI-MeT), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Antonio Recchiuti
- Department of Medical, Oral, and Biotechnological Sciences, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy.,Center on Aging Sciences and Translational Medicine (CeSI-MeT), "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
27
|
Cussell PJG, Howe MS, Illingworth TA, Gomez Escalada M, Milton NGN, Paterson AWJ. The formyl peptide receptor agonist FPRa14 induces differentiation of Neuro2a mouse neuroblastoma cells into multiple distinct morphologies which can be specifically inhibited with FPR antagonists and FPR knockdown using siRNA. PLoS One 2019; 14:e0217815. [PMID: 31170199 PMCID: PMC6553754 DOI: 10.1371/journal.pone.0217815] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 05/20/2019] [Indexed: 12/11/2022] Open
Abstract
The N-formyl peptide receptors (FPRs) have been identified within neuronal tissues and may serve as yet undetermined functions within the nervous system. The FPRs have been implicated in the progression and invasiveness of neuroblastoma and other cancers. In this study the effects of the synthetic FPR agonist FPRa14, FPR antagonists and FPR knockdown using siRNA on mouse neuroblastoma neuro2a (N2a) cell differentiation plus toxicity were examined. The FPRa14 (1-10μM) was found to induce a significant dose-dependent differentiation response in mouse neuroblastoma N2a cells. Interestingly, three distinct differentiated morphologies were observed, with two non-archetypal forms observed at the higher FPRa14 concentrations. These three forms were also observed in the human neuroblastoma cell-lines IMR-32 and SH-SY5Y when exposed to 100μM FPRa14. In N2a cells combined knockdown of FPR1 and FPR2 using siRNA inhibited the differentiation response to FPRa14, suggesting involvement of both receptor subtypes. Pre-incubating N2a cultures with the FPR1 antagonists Boc-MLF and cyclosporin H significantly reduced FPRa14-induced differentiation to near baseline levels. Meanwhile, the FPR2 antagonist WRW4 had no significant effect on FPRa14-induced N2a differentiation. These results suggest that the N2a differentiation response observed has an FPR1-dependent component. Toxicity of FPRa14 was only observed at higher concentrations. All three antagonists used blocked FPRa14-induced toxicity, whilst only siRNA knockdown of FPR2 reduced toxicity. This suggests that the toxicity and differentiation involve different mechanisms. The demonstration of neuronal differentiation mediated via FPRs in this study represents a significant finding and suggests a role for FPRs in the CNS. This finding could potentially lead to novel therapies for a range of neurological conditions including neuroblastoma, Alzheimer's disease, Parkinson's disease and neuropathic pain. Furthermore, this could represent a potential avenue for neuronal regeneration therapies.
Collapse
Affiliation(s)
- Peter J. G. Cussell
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, United Kingdom
| | - Michael S. Howe
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, United Kingdom
| | - Thomas A. Illingworth
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, United Kingdom
| | | | - Nathaniel G. N. Milton
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, United Kingdom
| | - Andrew W. J. Paterson
- School of Clinical and Applied Sciences, Leeds Beckett University, Leeds, United Kingdom
- * E-mail:
| |
Collapse
|
28
|
Ong WY, Chua JJE. Role of formyl peptide receptor 2 (FPR2) in the normal brain and in neurological conditions. Neural Regen Res 2019; 14:2071-2072. [PMID: 31397336 PMCID: PMC6788234 DOI: 10.4103/1673-5374.262575] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore; Life Sciences Institute Neurobiology Research Programme, National University of Singapore, Singapore
| | - John Jia En Chua
- Life Sciences Institute Neurobiology Research Programme; Department of Physiology, National University of Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
29
|
Korimová A, Klusáková I, Hradilová-Svíženská I, Kohoutková M, Joukal M, Dubový P. Mitochondrial Damage-Associated Molecular Patterns of Injured Axons Induce Outgrowth of Schwann Cell Processes. Front Cell Neurosci 2018; 12:457. [PMID: 30542268 PMCID: PMC6277938 DOI: 10.3389/fncel.2018.00457] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/12/2018] [Indexed: 01/13/2023] Open
Abstract
Activated Schwann cells put out cytoplasmic processes that play a significant role in cell migration and axon regeneration. Following nerve injury, axonal mitochondria release mitochondrial damage-associated molecular patterns (mtDAMPs), including formylated peptides and mitochondrial DNA (mtDNA). We hypothesize that mtDAMPs released from disintegrated axonal mitochondria may stimulate Schwann cells to put out cytoplasmic processes. We investigated RT4-D6P2T schwannoma cells (RT4) in vitro treated with N-formyl-L-methionyl-L-leucyl-phenylalanine (fMLP) or cytosine-phospho-guanine oligodeoxynucleotide (CpG ODN) for 1, 6 and 24 h. We also used immunohistochemical detection to monitor the expression of formylpeptide receptor 2 (FPR2) and toll-like receptor 9 (TLR9), the canonical receptors for formylated peptides and mtDNA, in RT4 cells and Schwann cells distal to nerve injury. RT4 cells treated with fMLP put out a significantly higher number of cytoplasmic processes compared to control cells. Preincubation with PBP10, a selective inhibitor of FPR2 resulted in a significant reduction of cytoplasmic process outgrowth. A significantly higher number of cytoplasmic processes was also found after treatment with CpG ODN compared to control cells. Pretreatment with inhibitory ODN (INH ODN) resulted in a reduced number of cytoplasmic processes after subsequent treatment with CpG ODN only at 6 h, but 1 and 24 h treatment with CpG ODN demonstrated an additive effect of INH ODN on the development of cytoplasmic processes. Immunohistochemistry and western blot detected increased levels of tyrosine-phosphorylated paxillin in RT4 cells associated with cytoplasmic process outgrowth after fMLP or CpG ODN treatment. We found increased immunofluorescence of FPR2 and TLR9 in RT4 cells treated with fMLP or CpG ODN as well as in activated Schwann cells distal to the nerve injury. In addition, activated Schwann cells displayed FPR2 and TLR9 immunostaining close to GAP43-immunopositive regenerated axons and their growth cones after nerve crush. Increased FPR2 and TLR9 immunoreaction was associated with activation of p38 and NFkB, respectively. Surprisingly, the growth cones displayed also FPR2 and TLR9 immunostaining. These results present the first evidence that potential mtDAMPs may play a key role in the induction of Schwann cell processes. This reaction of Schwann cells can be mediated via FPR2 and TLR9 that are canonical receptors for formylated peptides and mtDNA. The possible role for FPR2 and TLR9 in growth cones is also discussed.
Collapse
Affiliation(s)
- Andrea Korimová
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Ilona Klusáková
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Ivana Hradilová-Svíženská
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marcela Kohoutková
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marek Joukal
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Petr Dubový
- Department of Anatomy, Division of Neuroanatomy, Faculty of Medicine, Masaryk University, Brno, Czechia
| |
Collapse
|
30
|
Gravina AG, Prevete N, Tuccillo C, De Musis C, Romano L, Federico A, de Paulis A, D’Argenio G, Romano M. Peptide Hp(2-20) accelerates healing of TNBS-induced colitis in the rat. United European Gastroenterol J 2018; 6:1428-1436. [PMID: 30386616 PMCID: PMC6206543 DOI: 10.1177/2050640618793564] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/26/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND AIMS Hp(2-20), a Helicobacter pylori-derived peptide interacting with N-formyl peptide receptors (FPRs), accelerates the healing of gastric injury in rats. Whether Hp(2-20) affects the recovery of inflamed colonic mucosa is unknown. We evaluated whether Hp(2-20) accelerated the healing of 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis and explored the mechanism(s) underlying any such effect. METHODS Fifteen rats underwent rectal administration of Hp(2-20) 250-500 µg/kg/day, or of its control peptide Hp1 for 10 days, following induction of colitis with TNBS. Macroscopic and histological damage was quantified using predetermined injury scores. FPR1, COX-2, TNF-α, TGF-β, HB-EGF and tissue transglutaminase (t-TG) messenger RNA (mRNA) expression in colonic tissue was determined by quantitative polymerase chain reaction; FPR1, TNF-α and COX-2 protein levels by Western blotting. RESULTS (1) Hp(2-20) accelerated healing of TNBS-induced colitis compared to controls consistently with the expression of FPRs in colonic mucosa; (2) TNBS upregulated mRNA mucosal expression of COX-2, TNF-α, TGF-β, HB-EGF and t-TG and (3) this, with the exception of HB-EGF, was significantly counteracted by Hp(2-20). CONCLUSIONS Hp(2-20), an FPR agonist, accelerates the healing of TNBS-induced colitis in the rat. This effect is associated with a significant reduction in colonic tissue levels of COX-2, TGF-β, TNF-α and t-TG. We postulate that FPR-dependent pathways may be involved in the repair of inflamed colonic mucosa.
Collapse
Affiliation(s)
- AG Gravina
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| | - N Prevete
- Department of Translational Medical
Sciences (DiSMeT)-University of Naples Federico II and Institute of Endocrinology
and Experimental Oncology (IEOS) “G. Salvatore,” CNR, Naples, Italy
| | - C Tuccillo
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| | - C De Musis
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| | - L Romano
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| | - A Federico
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| | - A de Paulis
- Department of Translational Medical
Sciences (DiSMeT) and Center for Basic and Clinical Immunologic Research (CISI),
University of Naples Federico II, Naples, Italy
| | - G D’Argenio
- Gastroenterology Unit, Department of
Clinical and Experimental Medicine, University of Naples Federico II, Naples,
Italy
| | - M Romano
- Hepatogastroenterology Unit, Department
of Precision Medicine, University of Campania “Luigi Vanvitelli,” Naples,
Italy
| |
Collapse
|
31
|
Ho CFY, Ismail NB, Koh JKZ, Gunaseelan S, Low YH, Ng YK, Chua JJE, Ong WY. Localisation of Formyl-Peptide Receptor 2 in the Rat Central Nervous System and Its Role in Axonal and Dendritic Outgrowth. Neurochem Res 2018; 43:1587-1598. [PMID: 29948727 PMCID: PMC6061218 DOI: 10.1007/s11064-018-2573-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/06/2018] [Accepted: 06/09/2018] [Indexed: 01/03/2023]
Abstract
Arachidonic acid and docosahexaenoic acid (DHA) released by the action of phospholipases A2 (PLA2) on membrane phospholipids may be metabolized by lipoxygenases to the anti-inflammatory mediators lipoxin A4 (LXA4) and resolvin D1 (RvD1), and these can bind to a common receptor, formyl-peptide receptor 2 (FPR2). The contribution of this receptor to axonal or dendritic outgrowth is unknown. The present study was carried out to elucidate the distribution of FPR2 in the rat CNS and its role in outgrowth of neuronal processes. FPR2 mRNA expression was greatest in the brainstem, followed by the spinal cord, thalamus/hypothalamus, cerebral neocortex, hippocampus, cerebellum and striatum. The brainstem and spinal cord also contained high levels of FPR2 protein. The cerebral neocortex was moderately immunolabelled for FPR2, with staining mostly present as puncta in the neuropil. Dentate granule neurons and their axons (mossy fibres) in the hippocampus were very densely labelled. The cerebellar cortex was lightly stained, but the deep cerebellar nuclei, inferior olivary nucleus, vestibular nuclei, spinal trigeminal nucleus and dorsal horn of the spinal cord were densely labelled. Electron microscopy of the prefrontal cortex showed FPR2 immunolabel mostly in immature axon terminals or ‘pre-terminals’, that did not form synapses with dendrites. Treatment of primary hippocampal neurons with the FPR2 inhibitors, PBP10 or WRW4, resulted in reduced lengths of axons and dendrites. The CNS distribution of FPR2 suggests important functions in learning and memory, balance and nociception. This might be due to an effect of FPR2 in mediating arachidonic acid/LXA4 or DHA/RvD1-induced axonal or dendritic outgrowth.
Collapse
Affiliation(s)
| | - Nadia Binte Ismail
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore
| | - Joled Kong-Ze Koh
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore
| | - Saravanan Gunaseelan
- Department of Physiology, National University of Singapore, Singapore, 119260, Singapore
| | - Yi-Hua Low
- Institute of Neurology, University College London, London, UK
| | - Yee-Kong Ng
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore
| | - John Jia-En Chua
- Department of Physiology, National University of Singapore, Singapore, 119260, Singapore. .,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore, 138673. .,Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 117456, Singapore.
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore, 119260, Singapore. .,Neurobiology and Ageing Research Programme, National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
32
|
Lipoxin A 4 Attenuates the Inflammatory Response in Stem Cells of the Apical Papilla via ALX/FPR2. Sci Rep 2018; 8:8921. [PMID: 29892010 PMCID: PMC5995968 DOI: 10.1038/s41598-018-27194-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/25/2018] [Indexed: 01/01/2023] Open
Abstract
Similar to the onset phase of inflammation, its resolution is a process that unfolds in a manner that is coordinated and regulated by a panel of mediators. Lipoxin A4 (LXA4) has been implicated as an anti-inflammatory, pro-resolving mediator. We hypothesized that LXA4 attenuates or prevents an inflammatory response via the immunosuppressive activity of Stem Cells of the Apical Papilla (SCAP). Here, we report for the first time in vitro that in a SCAP population, lipoxin receptor ALX/FPR2 was constitutively expressed and upregulated after stimulation with lipopolysaccharide and/or TNF-α. Moreover, LXA4 significantly enhanced proliferation, migration, and wound healing capacity of SCAP through the activation of its receptor, ALX/FPR2. Cytokine, chemokine and growth factor secretion by SCAP was inhibited in a dose dependent manner by LXA4. Finally, LXA4 enhanced immunomodulatory properties of SCAP towards Peripheral Blood Mononuclear Cells. These findings provide the first evidence that the LXA4-ALX/FPR2 axis in SCAP regulates inflammatory mediators and enhances immunomodulatory properties. Such features of SCAP may also support the role of these cells in the resolution phase of inflammation and suggest a novel molecular target for ALX/FPR2 receptor to enhance a stem cell-mediated pro-resolving pathway.
Collapse
|
33
|
High-mobility group box 1 facilitates migration of neural stem cells via receptor for advanced glycation end products signaling pathway. Sci Rep 2018. [PMID: 29540727 PMCID: PMC5852130 DOI: 10.1038/s41598-018-22672-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
High-mobility group box 1 (HMGB1) facilitates neural stem cells (NSCs) proliferation and differentiation into neuronal linage. However, the effect of HMGB1 on NSCs migration is still elusive. The present study is to investigate the corelation between HMGB1 and NSCs migration and the potential mechanism. The results indicated that 1 ng/ml HMGB1 promoted NSCs proliferation using CCK8 assays. Moreover, data showed that 1 ng/ml HMGB1 facilitated NSCs migration via filopodia formation using phase-contrast and transwell assays. Furthermore, 1 ng/ml HMGB1 upregulated the expression of RAGE, one of the HMGB1 receptor, using western blotting assays and immunofluorescence staining. In addition, 1 ng/ml HMGB1 increased the percentage of filopodia formation using phalloidin staining. Meanwhile, the enhanced migration effect could be abrogated by 50 nM FPS-ZM1, one of the RAGE antagonist, and RAGE-specific siRNA through immunofluorescence and phalloidin staining. Together, our data demonstrate that HMGB1/RAGE axis facilitates NSCs migration via promoting filopodia formation, which might serve as a candidate for central nervous system (CNS) injury treatment and/or a preconditioning method for NSCs implantation.
Collapse
|
34
|
Bisicchia E, Sasso V, Catanzaro G, Leuti A, Besharat ZM, Chiacchiarini M, Molinari M, Ferretti E, Viscomi MT, Chiurchiù V. Resolvin D1 Halts Remote Neuroinflammation and Improves Functional Recovery after Focal Brain Damage Via ALX/FPR2 Receptor-Regulated MicroRNAs. Mol Neurobiol 2018; 55:6894-6905. [PMID: 29357041 DOI: 10.1007/s12035-018-0889-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/08/2018] [Indexed: 12/31/2022]
Abstract
Remote damage is a secondary phenomenon that usually occurs after a primary brain damage in regions that are distant, yet functionally connected, and that is critical for determining the outcomes of several CNS pathologies, including traumatic brain and spinal cord injuries. The understanding of remote damage-associated mechanisms has been mostly achieved in several models of focal brain injury such as the hemicerebellectomy (HCb) experimental paradigm, which helped to identify the involvement of many key players, such as inflammation, oxidative stress, apoptosis and autophagy. Currently, few interventions have been shown to successfully limit the progression of secondary damage events and there is still an unmet need for new therapeutic options. Given the emergence of the novel concept of resolution of inflammation, mediated by the newly identified ω3-derived specialized pro-resolving lipid mediators, such as resolvins, we reported a reduced ability of HCb-injured animals to produce resolvin D1 (RvD1) and an increased expression of its target receptor ALX/FPR2 in remote brain regions. The in vivo administration of RvD1 promoted functional recovery and neuroprotection by reducing the activation of Iba-1+ microglia and GFAP+ astrocytes as well as by impairing inflammatory-induced neuronal cell death in remote regions. These effects were counteracted by intracerebroventricular neutralization of ALX/FPR2, whose activation by RvD1 also down-regulated miR-146b- and miR-219a-1-dependent inflammatory markers. In conclusion, we propose that innovative therapies based on RvD1-ALX/FPR2 axis could be exploited to curtail remote damage and enable neuroprotective effects after acute focal brain damage.
Collapse
Affiliation(s)
- Elisa Bisicchia
- IRCCS Santa Lucia Foundation, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Valeria Sasso
- IRCCS Santa Lucia Foundation, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | | | - Alessandro Leuti
- IRCCS Santa Lucia Foundation, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | | | | | - Marco Molinari
- IRCCS Santa Lucia Foundation, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | | | - Valerio Chiurchiù
- IRCCS Santa Lucia Foundation, via del Fosso di Fiorano 64, 00143, Rome, Italy. .,Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy.
| |
Collapse
|
35
|
Stama ML, Lacivita E, Kirpotina LN, Niso M, Perrone R, Schepetkin IA, Quinn MT, Leopoldo M. Functional N-Formyl Peptide Receptor 2 (FPR2) Antagonists Based on the Ureidopropanamide Scaffold Have Potential To Protect Against Inflammation-Associated Oxidative Stress. ChemMedChem 2017; 12:1839-1847. [PMID: 28922577 PMCID: PMC5909973 DOI: 10.1002/cmdc.201700429] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/06/2017] [Indexed: 12/11/2022]
Abstract
Formyl peptide receptor 2 (FPR2) is a G protein coupled receptor belonging to the N-formyl peptide receptor (FPR) family that plays critical roles in peripheral and brain inflammatory responses. FPR2 has been proposed as a target for the development of drugs that could facilitate the resolution of chronic inflammatory reactions by enhancing endogenous anti-inflammation systems. Starting from lead compounds previously identified in our laboratories, we designed a new series of ureidopropanamide derivatives with the goal of converting functional activity from agonism into antagonism and to develop new FPR2 antagonists. Although none of the compounds behaved as antagonists, some of the compounds were able to induce receptor desensitization and, thus, functionally behaved as antagonists. Evaluation of these compounds in an in vitro model of neuroinflammation showed that they decreased the production of reactive oxygen species in mouse microglial N9 cells after stimulation with lipopolysaccharide. These FPR2 ligands may protect cells from damage due to inflammation-associated oxidative stress.
Collapse
Affiliation(s)
- Madia L. Stama
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Enza Lacivita
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Liliya N. Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Mauro Niso
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Roberto Perrone
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| | - Igor A. Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Mark T. Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
| | - Marcello Leopoldo
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125 Bari, Italy
| |
Collapse
|
36
|
Stama ML, Ślusarczyk J, Lacivita E, Kirpotina LN, Schepetkin IA, Chamera K, Riganti C, Perrone R, Quinn MT, Basta-Kaim A, Leopoldo M. Novel ureidopropanamide based N-formyl peptide receptor 2 (FPR2) agonists with potential application for central nervous system disorders characterized by neuroinflammation. Eur J Med Chem 2017; 141:703-720. [PMID: 29102463 DOI: 10.1016/j.ejmech.2017.09.023] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/01/2017] [Accepted: 09/14/2017] [Indexed: 12/25/2022]
Abstract
Formyl peptide receptor2 (FPR2) is a G-protein coupled receptor that plays critical roles in inflammatory reactions. FPR2-specific interaction can be possibly used to facilitate the resolution of pathological inflammatory responses by enhancing endogenous anti-inflammation systems. Starting from our lead agonist 5, we designed new ureidopropanamides derivatives able to activate FPR2 in transfected cells and human neutrophils. The new FPR2 agonists showed good stability towards oxidative metabolism in vitro. Moreover, selected compounds showed anti-inflammatory properties in LPS-stimulated rat primary microglial cells. (S)-3-(4-Cyanophenyl)-N-[[1-(3-chloro-4-fluorophenyl)cyclopropyl]methyl]-2-[3-(4-fluorophenyl)ureido]propanamide ((S)-17) emerged as prospective pharmacological tool to study the effects of FPR2 activation in the central nervous system (CNS) being able to reduce IL-1β and TNF-α levels in LPS-stimulated microglial cells and showing good permeation rate in hCMEC/D3 cells, an in vitro model of blood brain barrier. These results are very promising and can open new therapeutic perspectives in the treatment of CNS disorders characterized by neuroinflammation.
Collapse
Affiliation(s)
- Madia Letizia Stama
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125, Bari, Italy
| | - Joanna Ślusarczyk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Enza Lacivita
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125, Bari, Italy.
| | - Liliya N Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - Igor A Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - Katarzyna Chamera
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Chiara Riganti
- Dipartimento di Oncologia, Università di Torino, via Santena, 5/bis, 10126, Torino, Italy
| | - Roberto Perrone
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125, Bari, Italy
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, 59717, USA
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna St, 31-343, Krakow, Poland
| | - Marcello Leopoldo
- Dipartimento di Farmacia - Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, via Orabona, 4, 70125, Bari, Italy
| |
Collapse
|
37
|
Formyl peptide receptors promotes neural differentiation in mouse neural stem cells by ROS generation and regulation of PI3K-AKT signaling. Sci Rep 2017; 7:206. [PMID: 28303030 PMCID: PMC5428260 DOI: 10.1038/s41598-017-00314-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/20/2017] [Indexed: 01/12/2023] Open
Abstract
This study aimed to determine whether formyl peptide receptors (FPRs) regulated the differentiation of neural stem cells (NSCs). FPRs promote the migration of NSCs both in vitro and in vivo. However, the role of FPRs during differentiation of NSCs is unknown. Analysis by Western blot showed significantly increased expression of FPR1 and FPR2 during differentiation of NSCs. The activation of FPRs promotes NSCs to differentiate into neurons with more primary neurites and branch points and longer neurites per cell. Meanwhile, this activation also inhibits the differentiation of NSC into astrocytes. This bidirectional effect can be inhibited by the FPRs-specific inhibitor. Moreover, it was found that the activation of FPRs increased the generation of reactive oxygen species (ROS) and phosphorylation of AKT in the NSCs, while N-acetylcysteine and LY294002 inhibited the FPRs-stimulated increase in ROS generation and AKT phosphorylation, and blocked the FPRs-stimulated neural differentiation into neurons. Therefore, FPRs-stimulated neural differentiation was mediated via ROS and PI3K-AKT signaling pathways. Collectively, the present findings provided a novel insight into the functional role of FPRs in neurogenesis, with important implications for its potential use as a candidate for treating brain or spinal cord injury.
Collapse
|
38
|
Lee HY, Lee M, Bae YS. Formyl Peptide Receptors in Cellular Differentiation and Inflammatory Diseases. J Cell Biochem 2017; 118:1300-1307. [PMID: 28075050 DOI: 10.1002/jcb.25877] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/15/2022]
Abstract
Formyl peptide receptors (FPRs) are a family of classical chemoattractant receptors. Although FPRs are mainly expressed in phagocytic innate immune cells including monocytes/macrophages and neutrophils, recent reports demonstrated that additional different cell types such as T-lymphocytes and several non-immune cells also express functional FPRs. FPRs were first reported as a specific receptor to detect bacteria-derived N-formyl peptides. However, accumulating evidence has shown that FPRs can recognize various ligands derived from pathogens, mitochondria, and host. This review summarizes studies on some interesting endogenous agonists for FPRs. Here, we discuss functional roles of FPRs and their ligands concerning the regulation of cellular differentiation focusing on myeloid lineage cells. Accumulating evidence also suggests that FPRs may contribute to the control of inflammatory diseases. Here, we briefly review the current understanding of the functional role of FPRs and their ligands in inflammatory disorders in some animal disease models. J. Cell. Biochem. 118: 1300-1307, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ha Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, Republic of Korea
| |
Collapse
|
39
|
Ge H, Yu A, Chen J, Yuan J, Yin Y, Duanmu W, Tan L, Yang Y, Lan C, Chen W, Feng H, Hu R. Poly-L-ornithine enhances migration of neural stem/progenitor cells via promoting α-Actinin 4 binding to actin filaments. Sci Rep 2016; 6:37681. [PMID: 27874083 PMCID: PMC5118728 DOI: 10.1038/srep37681] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 11/01/2016] [Indexed: 12/21/2022] Open
Abstract
The recruitment of neural stem/progenitor cells (NSPCs) for brain restoration after injury is a promising regenerative therapeutic strategy. This strategy involves enhancing proliferation, migration and neuronal differentation of NSPCs. To date, the lack of biomaterials, which facilitate these processes to enhance neural regeneration, is an obstacle for the cell replacement therapies. Our previous study has shown that NSPCs grown on poly-L-ornithine (PO) could proliferate more vigorously and differentiate into more neurons than that on Poly-L-Lysine (PLL) and Fibronectin (FN). Here, we demonstrate that PO could promote migration of NSPCs in vitro, and the underlying mechanism is PO activates α-Actinins 4 (ACTN4), which is firstly certified to be expessed in NSPCs, to promote filopodia formation and therefore enhances NSPCs migration. Taken together, PO might serve as a better candidate for transplanted biomaterials in the regenerative therapeutic strategy, compared with PLL and FN.
Collapse
Affiliation(s)
- Hongfei Ge
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Anyong Yu
- Department of Emergency, The First Affiliated Hospital of Zunyi Medical College, Guizhou 563003, China
| | - Jingyu Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jichao Yuan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yi Yin
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wangsheng Duanmu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Liang Tan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yang Yang
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Chuan Lan
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Weixiang Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Hua Feng
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Rong Hu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
40
|
Effects of FTY720 (Fingolimod) on Proliferation, Differentiation, and Migration of Brain-Derived Neural Stem Cells. Stem Cells Int 2016; 2016:9671732. [PMID: 27829841 PMCID: PMC5088305 DOI: 10.1155/2016/9671732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 08/31/2016] [Accepted: 09/20/2016] [Indexed: 12/27/2022] Open
Abstract
Insufficient proliferation, differentiation, and migration are the main pitfalls of neural stem cells (NSCs) in reparative therapeutics for the central nervous system (CNS) diseases. The potent lipid mediator sphingosine-1-phosphate (S1P) regulates cells' biological behavior broadly in the CNS. However, the effects of activating S1P on NSCs are not quite clear. In the current study, FTY720 (Fingolimod), an analog of S1P, was employed to induce the proliferation, differentiation, and migration of cultured brain-derived NSCs. The results indicated that proliferation and migration ability of NSCs were promoted by FTY720. Though we observed no obvious neuron prefers differentiation of NSCs, there were more protoplasmic astrocytes developed in the presence of certain concentration of FTY720. This work gives more comprehensive understanding of how FTY720 affects NSCs.
Collapse
|