101
|
Donertas B, Unel CC, Erol K. Cannabinoids and agmatine as potential therapeutic alternatives for cisplatin-induced peripheral neuropathy. J Exp Pharmacol 2018; 10:19-28. [PMID: 29950907 PMCID: PMC6018893 DOI: 10.2147/jep.s162059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cisplatin is a widely used antineoplastic agent in the treatment of various cancers. Peripheral neuropathy is a well-known side effect of cisplatin and has the potential to result in limiting and/or reducing the dose, decreasing the quality of life. Unfortunately, the mechanism for cisplatin-induced neuropathy has not been completely elucidated. Currently, available treatments for neuropathic pain (NP) are mostly symptomatic, insufficient and are often linked with several detrimental side effects; thus, effective treatments are needed. Cannabinoids and agmatine are endogenous modulators that are implicated in painful states. This review explains the cisplatin-induced neuropathy and antinociceptive effects of cannabinoids and agmatine in animal models of NP and their putative therapeutic potential in cisplatin-induced neuropathy.
Collapse
Affiliation(s)
- Basak Donertas
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Cigdem Cengelli Unel
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Kevser Erol
- Department of Medical Pharmacology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir, Turkey
| |
Collapse
|
102
|
Funes SC, Rios M, Escobar‐Vera J, Kalergis AM. Implications of macrophage polarization in autoimmunity. Immunology 2018; 154:186-195. [PMID: 29455468 PMCID: PMC5980179 DOI: 10.1111/imm.12910] [Citation(s) in RCA: 611] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/31/2018] [Accepted: 02/09/2018] [Indexed: 12/12/2022] Open
Abstract
Macrophages are extremely heterogeneous and plastic cells with an important role not only in physiological conditions, but also during inflammation (both for initiation and resolution). In the early 1990s, two different phenotypes of macrophages were described: one of them called classically activated (or inflammatory) macrophages (M1) and the other alternatively activated (or wound-healing) macrophages (M2). Currently, it is known that functional polarization of macrophages into only two groups is an over-simplified description of macrophage heterogeneity and plasticity; indeed, it is necessary to consider a continuum of functional states. Overall, the current available data indicate that macrophage polarization is a multifactorial process in which a huge number of factors can be involved producing different activation scenarios. Once a macrophage adopts a phenotype, it still retains the ability to continue changing in response to new environmental influences. The reversibility of polarization has a critical therapeutic value, especially in diseases in which an M1/M2 imbalance plays a pathogenic role. In this review, we assess the high plasticity of macrophages and their potential to be exploited to reduce chronic/detrimental inflammation. On the whole, the evidence detailed in this review underscores macrophage polarization as a target of interest for immunotherapy.
Collapse
Affiliation(s)
- Samanta C. Funes
- Facultad de Ciencias BiológicasDepartamento de Genética Molecular y MicrobiologíaMillennium Institute on Immunology and ImmunotherapyPontificia Universidad Católica de ChileSantiagoChile
| | - Mariana Rios
- Facultad de Ciencias BiológicasDepartamento de Genética Molecular y MicrobiologíaMillennium Institute on Immunology and ImmunotherapyPontificia Universidad Católica de ChileSantiagoChile
| | - Jorge Escobar‐Vera
- Facultad de Ciencias de la SaludDepartamento BiomédicoLaboratorio de GenéticaUniversidad de AntofagastaAntofagastaChile
| | - Alexis M. Kalergis
- Facultad de Ciencias BiológicasDepartamento de Genética Molecular y MicrobiologíaMillennium Institute on Immunology and ImmunotherapyPontificia Universidad Católica de ChileSantiagoChile
- Facultad de MedicinaDepartamento de EndocrinologíaEscuela de MedicinaPontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
103
|
Guo J, Wang H, Li L, Yuan Y, Shi X, Hou S. Treatment with IL-19 improves locomotor functional recovery after contusion trauma to the spinal cord. Br J Pharmacol 2018; 175:2611-2621. [PMID: 29500933 DOI: 10.1111/bph.14193] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE IL-19 skews the immune response towards a Th2 type and appears to stimulate angiogenesis. In the current study, we tested if IL-19 treatment could reduce secondary injury and improve functional recovery after contusion spinal cord injury (SCI). EXPERIMENTAL APPROACH Firstly, mice were given a moderate-severe thoracic SCI at the T9-10 level and expression of IL-19 and its receptor was measured in the injured spinal cord. Then SCI mice were treated with mouse recombinant IL-19 and its blocking antibody to investigate the therapeutic effect of IL-19. KEY RESULTS Protein expression of IL-19 and its receptor IL-20R1 and IL-20R2 was up-regulated in the injured spinal cord of mice. IL-19 treatment promoted the recovery of locomotor function dose-dependently and reduced loss of motor neurons and microglial and glial activation following SCI. Treatment of SCI mice with IL-19 attenuated macrophage accumulation, reduced protein levels of TNF-α and CCL2 and promoted Th2 response and M2 macrophage activation in the injured region. Treatment of SCI mice with IL-19 promoted angiogenesis through up-regulating VEGF in the injured region. Treatment of SCI mice with IL-19 up-regulated HO-1 expression and decreased oxidative stress in the injured region. The beneficial effect of IL-19 was abolished by coadministration of the blocking antibody. Additionally, IL-19 deficiency in mice delayed the recovery of locomotor function following SCI. CONCLUSIONS AND IMPLICATIONS IL-19 treatment reduced secondary injuries and improved locomotor functional recovery after contusion SCI, through diverse mechanisms including immune cell polarization, angiogenesis and anti-oxidative responses.
Collapse
Affiliation(s)
- Jidong Guo
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| | - Huadong Wang
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| | - Li Li
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| | - Yanli Yuan
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| | - Xiuxiu Shi
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| | - Shuxun Hou
- Institute of Orthopaedics, First Affiliated Hospital of CPLA General Hospital, Beijing, China
| |
Collapse
|
104
|
Dyck S, Kataria H, Alizadeh A, Santhosh KT, Lang B, Silver J, Karimi-Abdolrezaee S. Perturbing chondroitin sulfate proteoglycan signaling through LAR and PTPσ receptors promotes a beneficial inflammatory response following spinal cord injury. J Neuroinflammation 2018; 15:90. [PMID: 29558941 PMCID: PMC5861616 DOI: 10.1186/s12974-018-1128-2] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/12/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) results in upregulation of chondroitin sulfate proteoglycans (CSPGs) by reactive glia that impedes repair and regeneration in the spinal cord. Degradation of CSPGs is known to be beneficial in promoting endogenous repair mechanisms including axonal sprouting/regeneration, oligodendrocyte replacement, and remyelination, and is associated with improvements in functional outcomes after SCI. Recent evidence suggests that CSPGs may regulate secondary injury mechanisms by modulating neuroinflammation after SCI. To date, the role of CSPGs in SCI neuroinflammation remains largely unexplored. The recent discovery of CSPG-specific receptors, leukocyte common antigen-related (LAR) and protein tyrosine phosphatase-sigma (PTPσ), allows unraveling the cellular and molecular mechanisms of CSPGs in SCI. In the present study, we have employed parallel in vivo and in vitro approaches to dissect the role of CSPGs and their receptors LAR and PTPσ in modulating the inflammatory processes in the acute and subacute phases of SCI. METHODS In a clinically relevant model of compressive SCI in female Sprague Dawley rats, we targeted LAR and PTPσ by two intracellular functionally blocking peptides, termed ILP and ISP, respectively. We delivered ILP and ISP treatment intrathecally to the injured spinal cord in a sustainable manner by osmotic mini-pumps for various time-points post-SCI. We employed flow cytometry, Western blotting, and immunohistochemistry in rat SCI, as well as complementary in vitro studies in primary microglia cultures to address our questions. RESULTS We provide novel evidence that signifies a key immunomodulatory role for LAR and PTPσ receptors in SCI. We show that blocking LAR and PTPσ reduces the population of classically activated M1 microglia/macrophages, while promoting alternatively activated M2 microglia/macrophages and T regulatory cells. This shift was associated with a remarkable elevation in pro-regenerative immune mediators, interleukin-10 (IL-10), and Arginase-1. Our parallel in vitro studies in microglia identified that while CSPGs do not induce an M1 phenotype per se, they promote a pro-inflammatory phenotype. Interestingly, inhibiting LAR and PTPσ in M1 and M2 microglia positively modulates their inflammatory response in the presence of CSPGs, and harnesses their ability for phagocytosis and mobilization. Interestingly, our findings indicate that CSPGs regulate microglia, at least in part, through the activation of the Rho/ROCK pathway downstream of LAR and PTPσ. CONCLUSIONS We have unveiled a novel role for LAR and PTPσ in regulating neuroinflammation in traumatic SCI. Our findings provide new insights into the mechanisms by which manipulation of CSPG signaling can promote recovery from SCI. More importantly, this work introduces the potential of ILP/ISP as a viable strategy for modulating the immune response following SCI and other neuroinflammatory conditions of the central nervous system.
Collapse
Affiliation(s)
- Scott Dyck
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Kallivalappil T Santhosh
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Bradley Lang
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jerry Silver
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, the Regenerative Medicine Program, the Spinal Cord Research Center, University of Manitoba, 629-Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
105
|
Wu Y, Shen L, Wang R, Tang J, Ding SQ, Wang SN, Guo XY, Hu JG, Lü HZ. Increased ceruloplasmin expression caused by infiltrated leukocytes, activated microglia, and astrocytes in injured female rat spinal cords. J Neurosci Res 2018; 96:1265-1276. [PMID: 29377294 DOI: 10.1002/jnr.24221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/21/2017] [Accepted: 01/11/2018] [Indexed: 12/17/2022]
Abstract
Ceruloplasmin (Cp), an enzyme containing six copper atoms, has important roles in iron homeostasis and antioxidant defense. After spinal cord injury (SCI), the cellular components in the local microenvironment are very complex and include functional changes of resident cells and the infiltration of leukocytes. It has been confirmed that Cp is elevated primarily in astrocytes and to a lesser extent in macrophages following SCI in mice. However, its expression in other cell types is still not very clear. In this manuscript, we provide a sensible extension of these findings by examining this system within a female Sprague-Dawley rat model and expanding the scope of inquiry to include additional cell types. Quantitative reverse transcription polymerase chain reaction and Western blot analysis revealed that the Cp mRNA and protein in SCI tissue homogenates were quite consistent with prior publications. However, we observed that Cp was expressed not only in GFAP+ astrocytes (consistent with prior reports) but also in CD11b+ microglia, CNPase+ oligodendrocytes, NeuN+ neurons, CD45+ leukocytes, and CD68+ activated microglia/macrophages. Quantitative analysis proved that infiltrated leukocytes, activated microglia/macrophages, and astrocytes should be the major sources of increased Cp.
Collapse
Affiliation(s)
- Yan Wu
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, People's Republic of China
| | - Lin Shen
- Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Jie Tang
- Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, People's Republic of China
| | - Shu-Qin Ding
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Sai-Nan Wang
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, People's Republic of China
| | - Xue-Yan Guo
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, People's Republic of China
| | - Jian-Guo Hu
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - He-Zuo Lü
- Clinical Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Bengbu, People's Republic of China
| |
Collapse
|
106
|
Sun G, Yang S, Cao G, Wang Q, Hao J, Wen Q, Li Z, So KF, Liu Z, Zhou S, Zhao Y, Yang H, Zhou L, Yin Z. γδ T cells provide the early source of IFN-γ to aggravate lesions in spinal cord injury. J Exp Med 2017; 215:521-535. [PMID: 29282251 PMCID: PMC5789408 DOI: 10.1084/jem.20170686] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/22/2017] [Accepted: 11/28/2017] [Indexed: 02/05/2023] Open
Abstract
Immune responses and neuroinflammation are critically involved in spinal cord injury (SCI). γδ T cells, a small subset of T cells, regulate the inflammation process in many diseases, yet their function in SCI is still poorly understood. In this paper, we demonstrate that mice deficient in γδ T cells (TCRδ-/- ) showed improved functional recovery after SCI. γδ T cells are detected at the lesion sites within 24 hours after injury and are predominantly of the Vγ4 subtype and express the inflammatory cytokine IFN-γ. Inactivating IFN-γ signaling in macrophages results in a significantly reduced production of proinflammatory cytokines in the cerebrospinal fluid (CSF) of mice with SCIs and improves functional recovery. Furthermore, treatment of SCI with anti-Vγ4 antibodies has a beneficial effect, similar to that obtained with anti-TNF-α. In SCI patients, γδ T cells are detected in the CSF, and most of them are IFN-γ positive. In conclusion, manipulation of γδ T cell functions may be a potential approach for future SCI treatment.
Collapse
Affiliation(s)
- Guodong Sun
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Shuxian Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Guangchao Cao
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Qianghua Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Jianlei Hao
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Qiong Wen
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Zonghua Liu
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China.,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Sufang Zhou
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, China
| | - Hengwen Yang
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China.,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Libing Zhou
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China .,Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, China.,Key Laboratory of Neuroscience, School of Basic Medical Sciences, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhinan Yin
- The First Affiliated Hospital, Biomedical Translational Research Institute and Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China .,State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
107
|
Anti-inflammatory (M2) macrophage media reduce transmission of oligomeric amyloid beta in differentiated SH-SY5Y cells. Neurobiol Aging 2017; 60:173-182. [DOI: 10.1016/j.neurobiolaging.2017.08.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/16/2017] [Accepted: 08/19/2017] [Indexed: 01/28/2023]
|
108
|
Spatio-temporal expression of Hexokinase-3 in the injured female rat spinal cords. Neurochem Int 2017; 113:23-33. [PMID: 29196144 DOI: 10.1016/j.neuint.2017.11.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/22/2017] [Accepted: 11/27/2017] [Indexed: 11/20/2022]
Abstract
Hexokinase-3 (HK3) is a member of hexokinase family, which can catalyze the first step of glucose metabolism. It can increase ATP levels, reduce the production of reactive oxygen species, increase mitochondrial biogenesis, protect mitochondrial membrane potential and play an antioxidant role. However, the change of its expression in spinal cord after injury is still unknown. In this study, we investigated the spatio-temporal expression of HK3 in the spinal cords by using a spinal cord injury (SCI) model in adult female Sprague-Dawley rats. Quantitative reverse transcription-PCR and western blot analysis revealed that HK3 could be detected in sham-opened spinal cords. After SCI, it gradually increased, reached a peak at 7 days post-injury (dpi), and then gradually decreased with the prolonging of injury time, but still maintained at a higher level for up to 28 dpi (the longest time evaluated in this study). Immunofluorescence staining showed that HK3 was found in GFAP+, β-tubulin III+ and IBA-1+ cells in sham-opened spinal cords. After SCI, in addition to the above-mentioned cells, it could also be found in CD45+ and CD68+ cells. These results demonstrate that HK3 is mainly expressed in astrocytes, neurons and microglia in normal spinal cords, and could rapidly increase in infiltrated leukocytes, activated microglia/macrophages and astrocytes after SCI. These data suggest that HK3 may be involved in the pathologic process of SCI by promoting glucose metabolism.
Collapse
|
109
|
Wang C, Wang Q, Lou Y, Xu J, Feng Z, Chen Y, Tang Q, Zheng G, Zhang Z, Wu Y, Tian N, Zhou Y, Xu H, Zhang X. Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation. J Cell Mol Med 2017; 22:1148-1166. [PMID: 29148269 PMCID: PMC5783886 DOI: 10.1111/jcmm.13368] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/03/2017] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a severe neurological disease; however, few drugs have been proved to treat SCI effectively. Neuroinflammation is the major pathogenesis of SCI secondary injury and considered to be the therapeutic target of SCI. Salidroside (Sal) has been reported to exert anti‐inflammatory effects in airway, adipose and myocardial tissue; however, the role of Sal in SCI therapeutics has not been clarified. In this study, we showed that Sal could improve the functional recovery of spinal cord in rats as revealed by increased BBB locomotor rating scale, angle of incline, and decreased cavity of spinal cord injury and apoptosis of neurons in vivo. Immunofluorescence double staining of microglia marker and M1/M2 marker demonstrated that Sal could suppress M1 microglia polarization and activate M2 microglia polarization in vivo. To verify how Sal exerts its effects on microglia polarization and neuron protection, we performed the mechanism study in vitro in microglia cell line BV‐2 and neuron cell line PC12. The results showed that Sal prevents apoptosis of PC12 cells in coculture with LPS‐induced M1 BV‐2 microglia, also the inflammatory secretion phenotype of M1 BV‐2 microglia was suppressed by Sal, and further studies demonstrated that autophagic flux regulation through AMPK/mTOR pathway was involved in Sal regulated microglia polarization after SCI. Overall, our study illustrated that Sal could promote spinal cord injury functional recovery in rats, and the mechanism may relate to its microglia polarization modulation through AMPK‐/mTOR‐mediated autophagic flux stimulation.
Collapse
Affiliation(s)
- Chenggui Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Qingqing Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yiting Lou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Jianxiang Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zhenhua Feng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yu Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Qian Tang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Gang Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Zengjie Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.,Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China.,Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
| |
Collapse
|
110
|
Lima R, Monteiro S, Lopes JP, Barradas P, Vasconcelos NL, Gomes ED, Assunção-Silva RC, Teixeira FG, Morais M, Sousa N, Salgado AJ, Silva NA. Systemic Interleukin-4 Administration after Spinal Cord Injury Modulates Inflammation and Promotes Neuroprotection. Pharmaceuticals (Basel) 2017; 10:ph10040083. [PMID: 29064422 PMCID: PMC5748640 DOI: 10.3390/ph10040083] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/17/2017] [Accepted: 10/22/2017] [Indexed: 12/18/2022] Open
Abstract
Traumatic spinal cord injury (SCI) causes dramatic disability and dysfunction in the motor, sensory and autonomic systems. The severe inflammatory reaction that occurs after SCI is strongly associated with further tissue damage. As such, immunomodulatory strategies have been developed, aimed at reducing inflammation, but also at shaping the immune response in order to protect, repair and promote regeneration of spared neural tissue. One of those promising strategies is the intraspinal administration of the cytokine interleukin-4 (IL-4) that was shown to promote a phenotype on specific immune cells associated with neuroprotection and repair. In this work, we evaluated if a systemic delivery of IL-4 for a 7-days period was also capable of promoting neuroprotection after SCI by analyzing different neural cells populations and motor recovery. IL-4 treatment promoted an elevation of the anti-inflammatory cytokine IL-10 in the serum both at 24 h and 7 days after injury. Locally, treatment with IL-4 led to a reduction on cells expressing markers associated with inflammation, CD11b/c and iNOS. Importantly, IL-4 treatment increased the neuronal markers βIII-tubulin and NeuN, and the oligodendrocyte marker O4, suggesting a neuroprotective effect. Moreover, 100% of the animals treated with IL-4 were able to recover weight support against only 33% of saline treated animals. Overall, these results show that systemic administration of IL-4 positively impacts different aspects of spinal cord injury, creating a more favorable environment for recovery to take place.
Collapse
Affiliation(s)
- Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - José P Lopes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Pedro Barradas
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Natália L Vasconcelos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Eduardo D Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Rita C Assunção-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Fábio G Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Mónica Morais
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Portugal.
| |
Collapse
|
111
|
Kim JH, Kim JY, Mun CH, Suh M, Lee JE. Agmatine Modulates the Phenotype of Macrophage Acute Phase after Spinal Cord Injury in Rats. Exp Neurobiol 2017; 26:278-286. [PMID: 29093636 PMCID: PMC5661060 DOI: 10.5607/en.2017.26.5.278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 08/29/2017] [Accepted: 09/24/2017] [Indexed: 12/26/2022] Open
Abstract
Agmatine is a decarboxylated arginine by arginine decarboxylase. Agmatine is known to be a neuroprotective agent. It has been reported that agmatine works as a NMDA receptor blocker or a competitive nitric oxide synthase inhibitor in CNS injuries. In spinal cord injury, agmatine showed reduction of neuropathic pain, improvement of locomotor function, and neuroprotection. Macrophage is a key cellular component in neuroinflammation, a major cause of impairment after spinal cord injury. Macrophage has subtypes, M1 and M2 macrophages. M1 macrophage induces a pro-inflammatory response, but M2 inspires an anti-inflammatory response. In this study, it was clarified whether the neuroprotective effect of agmatine is related with the modulation of macrophage subdivision after spinal cord injury. Spinal cord injury was induced in rats with contusion using MASCIS. Animals received agmatine (100 mg/kg, IP) daily for 6 days beginning the day after spinal cord injury. The proportion of M1 and M2 macrophages are confirmed with immunohistochemistry and FACS. CD206+ & ED1+ cells were counted as M2 macrophages. The systemic treatment of agmatine increased M2 macrophages caudal side to epicenter 1 week after spinal cord injury in immunohistochemistry. M2 macrophage related markers, Arginase-1 and CD206 mRNA, were increased in the agmatine treatment group and M2 macrophage expressing and stimulated cytokine, IL-10 mRNA, also was significantly overexpressed by agmatine injection. Among BMPs, BMP2/4/7, agmatine significantly increased only the expression of BMP2 known to reduce M1 macrophage under inflammatory status. These results suggest that agmatine reduces impairment after spinal cord injury through modulating the macrophage phenotype.
Collapse
Affiliation(s)
- Jae Hwan Kim
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Sungkyunkwan University (SKKU), Suwon 16419, Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea.,Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jae Young Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Chin Hee Mun
- Division of Rheumatology, Department of Internal Medicine, Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Minah Suh
- Center for Neuroscience Imaging Research, Institute for Basic Science (IBS), Sungkyunkwan University (SKKU), Suwon 16419, Korea.,Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul 03722, Korea.,BK21 PLUS Project for Medical Science and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
112
|
Liu M, Deng M, Ma Y. Differently polarized macrophages affect the viability and growth of NSPCs by regulating the expression of PACAP. Neuropeptides 2017; 65:114-119. [PMID: 28751044 DOI: 10.1016/j.npep.2017.07.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 04/29/2017] [Accepted: 07/05/2017] [Indexed: 01/07/2023]
Abstract
UNLABELLED Objective To explore the influence of differently polarized macrophages, M1 or M2, to viability and growth of NSPCs and its possible mechanism, especially the role of pituitary adenylate cyclase-activating polypeptide (PACAP) in it. METHOD Spinal cord-derived NSPCs were co-cultured with M1 or M2 through a transwell system. Growth of NSPCs in both groups was observed through an inverted microscope within 3days. NSPCs viability of each group, represented as intracellular ATP levels, was measured using the Cellular ATP Kit HTS following co-culture for 24h. PACAP levels in the co-cultured NSPCs was alleviated with immunofluorescence and Western blot analysis. RESULTS Morphologically NSPCs demonstrated a long spindle shape with short sprout on 3rd day when cultured together with M2. NSPCs cultured with M1 showed a small circle or oval shape with no obvious sprout. Expression of PACAP was observed in NSPCs co-cultured with M2 through immunofluorescence. In contrast, NSPCs did not demonstrate PACAP staining in the presence of M1 or cultured alone. PACAP in the NSPCs co-cultured with M2 was upregulated significantly compared with that co-cultured with M1 according to Western blot method. The relative ATP level of NSPCs co-cultured with M1 was markedly decreased while that with M2 was elevated significantly. That trend could be relieved by exogenous PACAP or PACAP 6-38. Viability change of NSPCs by M1/M2 correlated with apoptosis. CONCLUSION Differently polarized macrophages could affect the growth and viability of NSPCs by regulating the expression of PACAP.
Collapse
Affiliation(s)
- Min Liu
- Department of Immunology, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Ming Deng
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan City 430060, China
| | - Yonggang Ma
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan City 430060, China.
| |
Collapse
|
113
|
Pires LR, Lopes CDF, Salvador D, Rocha DN, Pêgo AP. Ibuprofen-loaded fibrous patches-taming inhibition at the spinal cord injury site. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:157. [PMID: 28894995 DOI: 10.1007/s10856-017-5967-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/19/2017] [Indexed: 06/07/2023]
Abstract
It is now widely accepted that a therapeutic strategy for spinal cord injury (SCI) demands a multi-target approach. Here we propose the use of an easily implantable bilayer polymeric patch based on poly(trimethylene carbonate-co-ε-caprolactone) (P(TMC-CL)) that combines physical guidance cues provided by electrospun aligned fibres and the delivery of ibuprofen, as a mean to reduce the inhibitory environment at the lesion site by taming RhoA activation. Bilayer patches comprised a solvent cast film onto which electrospun aligned fibres have been deposited. Both layers were loaded with ibuprofen. In vitro release (37°C, in phosphate buffered saline) of the drug from the loaded scaffolds under sink condition was found to occur in the first 24 h. The released ibuprofen was shown to retain its bioactivity, as indicated by the reduction of RhoA activation when the neuronal-like cell line ND7/23 was challenged with lysophosphatidic acid. Ibuprofen-loaded P(TMC-CL) bilayer scaffolds were successfully implanted in vivo in a dorsal hemisection rat SCI model mediating the reduction of RhoA activation after 5 days of implantation in comparison to plain P(TMC-CL) scaffolds. Immunohistochemical analysis of the tissue shows βIII tubulin positive cells close to the ibuprofen-loaded patches further supporting the use of this strategy in the context of regeneration after a lesion in the spinal cord.
Collapse
Affiliation(s)
- Liliana R Pires
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INL- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Cátia D F Lopes
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Daniela Salvador
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Daniela N Rocha
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Ana Paula Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS), Porto, Portugal.
- Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.
| |
Collapse
|
114
|
Truettner JS, Bramlett HM, Dietrich WD. Posttraumatic therapeutic hypothermia alters microglial and macrophage polarization toward a beneficial phenotype. J Cereb Blood Flow Metab 2017; 37:2952-2962. [PMID: 27864465 PMCID: PMC5536802 DOI: 10.1177/0271678x16680003] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Posttraumatic inflammatory processes contribute to pathological and reparative processes observed after traumatic brain injury (TBI). Recent findings have emphasized that these divergent effects result from subsets of proinflammatory (M1) or anti-inflammatory (M2) microglia and macrophages. Therapeutic hypothermia has been tested in preclinical and clinical models of TBI to limit secondary injury mechanisms including proinflammatory processes. This study evaluated the effects of posttraumatic hypothermia (PTH) on phenotype patterns of microglia/macrophages. Sprague-Dawley rats underwent moderate fluid percussion brain injury with normothermia (37℃) or hypothermia (33℃). Cortical and hippocampal regions were analyzed using flow cytometry and reverse transcription-polymerase chain reaction (RT-PCR) at several periods after injury. Compared to normothermia, PTH attenuated infiltrating cortical macrophages positive for CD11b+ and CD45high. At 24 h, the ratio of iNOS+ (M1) to arginase+ (M2) cells after hypothermia showed a decrease compared to normothermia. RT-PCR of M1-associated genes including iNOS and IL-1β was significantly reduced with hypothermia while M2-associated genes including arginase and CD163 were significantly increased compared to normothermic conditions. The injury-induced increased expression of the chemokine Ccl2 was also reduced with PTH. These studies provide a link between temperature-sensitive alterations in macrophage/microglia activation and polarization toward a M2 phenotype that could be permissive for cell survival and repair.
Collapse
Affiliation(s)
- Jessie S Truettner
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| | - W Dalton Dietrich
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
115
|
Lin T, Pajarinen J, Nabeshima A, Lu L, Nathan K, Yao Z, Goodman SB. Establishment of NF-κB sensing and interleukin-4 secreting mesenchymal stromal cells as an "on-demand" drug delivery system to modulate inflammation. Cytotherapy 2017; 19:1025-1034. [PMID: 28739167 DOI: 10.1016/j.jcyt.2017.06.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/12/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022]
Abstract
Chronic inflammation is associated with up-regulation of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and excessive inflammatory cytokine secretion by M1 macrophages. The anti-inflammatory cytokine interleukin (IL)-4 converts pro-inflammatory M1 macrophages into an anti-inflammatory and tissue-regenerative M2 phenotype, thus reducing inflammation and enhancing tissue regeneration. We have generated NF-κB responsive, or constitutively active IL-4 expression lentiviral vectors transduced into murine bone marrow-derived mesenchymal stromal cells (MSCs). MSCs with a constitutively active IL-4 expression vector produced large quantities of IL-4 continuously, whereas IL-4 secretion was significantly induced by lipopolysaccharide (LPS) in the NF-κB sensing MSCs. In contrast, LPS had no effect on MSCs with IL-4 secretion driven by a constitutively active promoter. We also found that intermittent and continuous LPS treatment displayed distinct NF-κB activation profiles, and this regulation was independent of IL-4 signaling. The supernatant containing IL-4 from the LPS-treated MSCs suppressed M1 marker (inducible nitric oxide synthase [iNOS] and tumor necrosis factor alpha [TNFα]) expression and enhanced M2 marker (Arginase 1, CD206 and IL1 receptor antagonist [IL1Ra]) expression in primary murine macrophages. The IL-4 secretion at the basal, non-LPS induced level was sufficient to suppress TNFα and enhance Arginase 1 at a lower level, but had no significant effects on iNOS, CD206 and IL1Ra expression. Finally, IL-4 secretion at basal or LPS-induced levels significantly suppressed osteogenic differentiation of MSCs. Our findings suggest that the IL-4 secreting MSCs driven by NF-κB sensing or constitutive active promoter have great potential for mitigating the effects of chronic inflammation and promoting earlier tissue regeneration.
Collapse
Affiliation(s)
- Tzuhua Lin
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Jukka Pajarinen
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Akira Nabeshima
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Laura Lu
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Karthik Nathan
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Zhenyu Yao
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA; Department of Bioengineering, Stanford University, Stanford, California, USA.
| |
Collapse
|
116
|
Alternative activation-skewed microglia/macrophages promote hematoma resolution in experimental intracerebral hemorrhage. Neurobiol Dis 2017; 103:54-69. [PMID: 28365213 DOI: 10.1016/j.nbd.2017.03.016] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 02/24/2017] [Accepted: 03/28/2017] [Indexed: 12/27/2022] Open
Abstract
Microglia/macrophages (MMΦ) are highly plastic phagocytes that can promote both injury and repair in diseased brain through the distinct function of classically activated and alternatively activated subsets. The role of MMΦ polarization in intracerebral hemorrhage (ICH) is unknown. Herein, we comprehensively characterized MMΦ dynamics after ICH in mice and evaluated the relevance of MMΦ polarity to hematoma resolution. MMΦ accumulated within the hematoma territory until at least 14days after ICH induction. Microglia rapidly reacted to the hemorrhagic insult as early as 1-1.5h after ICH and specifically presented a "protective" alternatively activated phenotype. Substantial numbers of activated microglia and newly recruited monocytes also assumed an early alternatively activated phenotype, but the phenotype gradually shifted to a mixed spectrum over time. Ultimately, markers of MMΦ classic activation dominated at the chronic stage of ICH. We enhanced MMΦ alternative activation by administering intraperitoneal injections of rosiglitazone, and subsequently observed elevations in CD206 expression on brain-isolated CD11b+ cells and increases in IL-10 levels in serum and perihematomal tissue. Enhancement of MMΦ alternative activation correlated with hematoma volume reduction and improvement in neurologic deficits. Intraventricular injection of alternative activation signature cytokine IL-10 accelerated hematoma resolution, whereas microglial phagocytic ability was abolished by IL-10 receptor neutralization. Our results suggest that MMΦ respond dynamically to brain hemorrhage by exhibiting diverse phenotypic changes at different stages of ICH. Alternative activation-skewed MMΦ aid in hematoma resolution, and IL-10 signaling might contribute to regulation of MMΦ phagocytosis and hematoma clearance in ICH.
Collapse
|
117
|
Barrett JP, Henry RJ, Villapol S, Stoica BA, Kumar A, Burns MP, Faden AI, Loane DJ. NOX2 deficiency alters macrophage phenotype through an IL-10/STAT3 dependent mechanism: implications for traumatic brain injury. J Neuroinflammation 2017; 14:65. [PMID: 28340575 PMCID: PMC5366128 DOI: 10.1186/s12974-017-0843-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND NADPH oxidase (NOX2) is an enzyme system that generates reactive oxygen species (ROS) in microglia and macrophages. Excessive ROS production is linked with neuroinflammation and chronic neurodegeneration following traumatic brain injury (TBI). Redox signaling regulates macrophage/microglial phenotypic responses (pro-inflammatory versus anti-inflammatory), and NOX2 inhibition following moderate-to-severe TBI markedly reduces pro-inflammatory activation of macrophages/microglia resulting in concomitant increases in anti-inflammatory responses. Here, we report the signaling pathways that regulate NOX2-dependent macrophage/microglial phenotype switching in the TBI brain. METHODS Bone marrow-derived macrophages (BMDMs) prepared from wildtype (C57Bl/6) and NOX2 deficient (NOX2-/-) mice were treated with lipopolysaccharide (LPS; 10 ng/ml), interleukin-4 (IL-4; 10 ng/ml), or combined LPS/IL-4 to investigate signal transduction pathways associated with macrophage activation using western immunoblotting and qPCR analyses. Signaling pathways and activation markers were evaluated in ipsilateral cortical tissue obtained from adult male wildtype and NOX2-/- mice that received moderate-level controlled cortical impact (CCI). A neutralizing anti-IL-10 approach was used to determine the effects of IL-10 on NOX2-dependent transitions from pro- to anti-inflammatory activation states. RESULTS Using an LPS/IL-4-stimulated BMDM model that mimics the mixed pro- and anti-inflammatory responses observed in the injured cortex, we show that NOX2-/- significantly reduces STAT1 signaling and markers of pro-inflammatory activation. In addition, NOX2-/- BMDMs significantly increase anti-inflammatory marker expression; IL-10-mediated STAT3 signaling, but not STAT6 signaling, appears to be critical in regulating this anti-inflammatory response. Following moderate-level CCI, IL-10 is significantly increased in microglia/macrophages in the injured cortex of NOX2-/- mice. These changes are associated with increased STAT3 activation, but not STAT6 activation, and a robust anti-inflammatory response. Neutralization of IL-10 in NOX2-/- BMDMs or CCI mice blocks STAT3 activation and the anti-inflammatory response, thereby demonstrating a critical role for IL-10 in regulating NOX2-dependent transitions between pro- and anti-inflammatory activation states. CONCLUSIONS These studies indicate that following TBI NOX2 inhibition promotes a robust anti-inflammatory response in macrophages/microglia that is mediated by the IL-10/STAT3 signaling pathway. Thus, therapeutic interventions that inhibit macrophage/microglial NOX2 activity may improve TBI outcomes by not only limiting pro-inflammatory neurotoxic responses, but also enhancing IL-10-mediated anti-inflammatory responses that are neuroprotective.
Collapse
Affiliation(s)
- James P Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Rebecca J Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Sonia Villapol
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Alok Kumar
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - Mark P Burns
- Laboratory for Brain Injury and Dementia, Department of Neuroscience, Georgetown University, Washington, DC, USA
| | - Alan I Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA
| | - David J Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, 655 West Baltimore Street, #6-011, Baltimore, MD, 21201, USA.
| |
Collapse
|
118
|
Shi LL, Zhang N, Xie XM, Chen YJ, Wang R, Shen L, Zhou JS, Hu JG, Lü HZ. Transcriptome profile of rat genes in injured spinal cord at different stages by RNA-sequencing. BMC Genomics 2017; 18:173. [PMID: 28201982 PMCID: PMC5312572 DOI: 10.1186/s12864-017-3532-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 02/01/2017] [Indexed: 12/15/2022] Open
Abstract
Background Spinal cord injury (SCI) results in fatal damage and currently has no effective treatment. The pathological mechanisms of SCI remain unclear. In this study, genome-wide transcriptional profiling of spinal cord samples from injured rats at different time points after SCI was performed by RNA-Sequencing (RNA-Seq). The transcriptomes were systematically characterized to identify the critical genes and pathways that are involved in SCI pathology. Results RNA-Seq results were obtained from total RNA harvested from the spinal cords of sham control rats and rats in the acute, subacute, and chronic phases of SCI (1 day, 6 days and 28 days after injury, respectively; n = 3 in every group). Compared with the sham-control group, the number of differentially expressed genes was 1797 in the acute phase (1223 upregulated and 574 downregulated), 6590 in the subacute phase (3460 upregulated and 3130 downregulated), and 3499 in the chronic phase (1866 upregulated and 1633 downregulated), with an adjusted P-value <0.05 by DESeq. Gene ontology (GO) enrichment analysis showed that differentially expressed genes were most enriched in immune response, MHC protein complex, antigen processing and presentation, translation-related genes, structural constituent of ribosome, ion gated channel activity, small GTPase mediated signal transduction and cytokine and/or chemokine activity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that the most enriched pathways included ribosome, antigen processing and presentation, retrograde endocannabinoid signaling, axon guidance, dopaminergic synapses, glutamatergic synapses, GABAergic synapses, TNF, HIF-1, Toll-like receptor, NF-kappa B, NOD-like receptor, cAMP, calcium, oxytocin, Rap1, B cell receptor and chemokine signaling pathway. Conclusions This study has not only characterized changes in global gene expression through various stages of SCI progression in rats, but has also systematically identified the critical genes and signaling pathways in SCI pathology. These results will expand our understanding of the complex molecular mechanisms involved in SCI and provide a foundation for future studies of spinal cord tissue damage and repair. The sequence data from this study have been deposited into Sequence Read Archive (http://www.ncbi.nlm.nih.gov/sra; accession number PRJNA318311). Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3532-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ling-Ling Shi
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Department of Immunology, Bengbu Medical College, Anhui, 233030, People's Republic of China
| | - Nan Zhang
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Xiu-Mei Xie
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Yue-Juan Chen
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Lin Shen
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Jian-Sheng Zhou
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China
| | - Jian-Guo Hu
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China.
| | - He-Zuo Lü
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, People's Republic of China. .,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, 287 Chang Huai Road, Anhui, 233004, People's Republic of China. .,Department of Immunology, Bengbu Medical College, Anhui, 233030, People's Republic of China.
| |
Collapse
|
119
|
Neuroinflammation as Fuel for Axonal Regeneration in the Injured Vertebrate Central Nervous System. Mediators Inflamm 2017; 2017:9478542. [PMID: 28203046 PMCID: PMC5288536 DOI: 10.1155/2017/9478542] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/05/2016] [Accepted: 12/25/2016] [Indexed: 01/19/2023] Open
Abstract
Damage to the central nervous system (CNS) is one of the leading causes of morbidity and mortality in elderly, as repair after lesions or neurodegenerative disease usually fails because of the limited capacity of CNS regeneration. The causes underlying this limited regenerative potential are multifactorial, but one critical aspect is neuroinflammation. Although classically considered as harmful, it is now becoming increasingly clear that inflammation can also promote regeneration, if the appropriate context is provided. Here, we review the current knowledge on how acute inflammation is intertwined with axonal regeneration, an important component of CNS repair. After optic nerve or spinal cord injury, inflammatory stimulation and/or modification greatly improve the regenerative outcome in rodents. Moreover, the hypothesis of a beneficial role of inflammation is further supported by evidence from adult zebrafish, which possess the remarkable capability to repair CNS lesions and even restore functionality. Lastly, we shed light on the impact of aging processes on the regenerative capacity in the CNS of mammals and zebrafish. As aging not only affects the CNS, but also the immune system, the regeneration potential is expected to further decline in aged individuals, an element that should definitely be considered in the search for novel therapeutic strategies.
Collapse
|
120
|
Chen J, Ning R, Zacharek A, Cui C, Cui X, Yan T, Venkat P, Zhang Y, Chopp M. MiR-126 Contributes to Human Umbilical Cord Blood Cell-Induced Neurorestorative Effects After Stroke in Type-2 Diabetic Mice. Stem Cells 2016; 34:102-13. [PMID: 26299579 DOI: 10.1002/stem.2193] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/16/2015] [Accepted: 07/25/2015] [Indexed: 12/30/2022]
Abstract
Diabetes mellitus (DM) is a high risk factor for stroke and leads to more severe vascular and white-matter injury than stroke in non-DM. We tested the neurorestorative effects of delayed human umbilical cord blood cell (HUCBC) treatment of stroke in type-2 diabetes (T2DM). db/db-T2DM and db/+-non-DM mice were subjected to distal middle cerebral artery occlusion (dMCAo) and were treated 3 days after dMCAo with: (a) non-DM + Phosphate buffered saline (PBS); (b) T2DM + PBS; (c) T2DM + naïve-HUCBC; (d) T2DM + miR-126(-/-) HUCBC. Functional evaluation, vascular and white-matter changes, neuroinflammation, and miR-126 effects were measured in vivo and in vitro. T2DM mice exhibited significantly decreased serum and brain tissue miR-126 expression compared with non-DM mice. T2DM + HUCBC mice exhibited increased miR-126 expression, increased tight junction protein expression, axon/myelin, vascular density, and M2-macrophage polarization. However, decreased blood-brain barrier leakage, brain hemorrhage, and miR-126 targeted gene vascular cell adhesion molecule-1 and monocyte chemotactic protein 1 expression in the ischemic brain as well as improved functional outcome were present in HUCBC-treated T2DM mice compared with control T2DM mice. MiR-126(-/-) HUCBC-treatment abolished the benefits of naïve-HUCBC-treatment in T2DM stroke mice. In vitro, knock-in of miR-126 in primary cultured brain endothelial cells (BECs) or treatment of BECs with naïve-HUCBCs significantly increased capillary-like tube formation, and increased axonal outgrowth in primary cultured cortical neurons; whereas treatment of BECs or cortical neurons with miR-126(-/-) HUCBC attenuated HUCBC-treatment-induced capillary tube formation and axonal outgrowth. Our data suggest delayed HUCBC-treatment of stroke increases vascular/white-matter remodeling and anti-inflammatory effects; MiR-126 may contribute to HUCBC-induced neurorestorative effects in T2DM mice.
Collapse
Affiliation(s)
- Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, People's Republic of China
| | - Ruizhuo Ning
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Chengcheng Cui
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Xu Cui
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Tao Yan
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| | - Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan, USA.,Department of Physics, Oakland University, Rochester, Michigan, USA
| |
Collapse
|
121
|
Kong X, Gao J. Macrophage polarization: a key event in the secondary phase of acute spinal cord injury. J Cell Mol Med 2016; 21:941-954. [PMID: 27957787 PMCID: PMC5387136 DOI: 10.1111/jcmm.13034] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/29/2016] [Indexed: 01/18/2023] Open
Abstract
Acute spinal cord injury (SCI) has become epidemic in modern society. Despite advances made in the understanding of the pathogenesis and improvements in early recognition and treatment, it remains a devastating event, often producing severe and permanent disability. SCI has two phases: acute and secondary. Although the acute phase is marked by severe local and systemic events such as tissue contusion, ischaemia, haemorrhage and vascular damage, the outcome of SCI are mainly influenced by the secondary phase. SCI causes inflammatory responses through the activation of innate immune responses that contribute to secondary injury, in which polarization‐based macrophage activation is a hallmarker. Macrophages accumulated within the epicentre and the haematoma of the injured spinal cord play a significant role in this inflammation. Depending on their phenotype and activation status, macrophages may initiate secondary injury mechanisms and/or promote CNS regeneration and repair. When it comes to therapies for SCI, very few can be performed in the acute phase. However, as macrophage activation and polarization switch are exquisitely sensitive to changes in microenvironment, some trials have been conducted to modulate macrophage polarization towards benefiting the recovery of SCI. Given this, it is important to understand how macrophages and SCI interrelate and interact on a molecular pathophysiological level. This review provides a comprehensive overview of the immuno‐pathophysiological features of acute SCI mainly from the following perspectives: (i) the overview of the pathophysiology of acute SCI, (ii) the roles of macrophage, especially its polarization switch in acute SCI, and (iii) newly developed neuroprotective therapies modulating macrophage polarization in acute SCI.
Collapse
Affiliation(s)
- Xiangyi Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
122
|
Wu Y, Lin YH, Shi LL, Yao ZF, Xie XM, Jiang ZS, Tang J, Hu JG, Lü HZ. Temporal kinetics of CD8 + CD28 + and CD8 + CD28 - T lymphocytes in the injured rat spinal cord. J Neurosci Res 2016; 95:1666-1676. [PMID: 27898179 DOI: 10.1002/jnr.23993] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/09/2016] [Accepted: 10/31/2016] [Indexed: 01/13/2023]
Abstract
This study aims to explore the temporal changes of cytotoxic CD8+ CD28+ and regulatory CD8+ CD28- T-cell subsets in the lesion microenvironment after spinal cord injury (SCI) in rats, by combination of immunohistochemistry (IHC) and flow cytometry (FCM). In the sham-opened spinal cord, few CD8+ T cells were found. After SCI, the CD8+ T cells were detected at one day post-injury (dpi), then markedly increased and were significantly higher at 3, 7, and 14 dpi compared with one dpi (p < 0.01), the highest being seven dpi. In CD8+ T cells, more than 90% were CD28+ , and there were only small part of CD28- ( < 10%). After 14 days, the infiltrated CD8+ T cells were significantly decreased, and few could be found in good condition at 21 and 28 dpi. Annexin V and propidium iodide (PI) staining showed that the percentages of apoptotic/necrotic CD8+ cells at 14 dpi and 21 dpi were significantly higher than those of the other early time-points (p < 0.01). These results indicate that CD8+ T cells could rapidly infiltrate into the injured spinal cords and survive two weeks, however, cytotoxic CD8+ T cells were dominant. Therefore, two weeks after injury might be the "time window" for treating SCI by prolonging survival times and increasing the fraction of CD8+ regulatory T-cells. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yan Wu
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| | - Yu-Hong Lin
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| | - Ling-Ling Shi
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| | - Zong-Feng Yao
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China
| | - Xiu-Mei Xie
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China
| | - Zheng-Song Jiang
- Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| | - Jie Tang
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| | - Jian-Guo Hu
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China
| | - He-Zuo Lü
- Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, the First Affiliated Hospital of Bengbu Medical College, Anhui, 233004, P.R. China.,Department of Immunology, Bengbu Medical College, and Anhui Key Laboratory of Infection and Immunity at Bengbu Medical College, Anhui, 233030, P.R. China
| |
Collapse
|
123
|
Activation of the niacin receptor HCA2 reduces demyelination and neurofilament loss, and promotes functional recovery after spinal cord injury in mice. Eur J Pharmacol 2016; 791:124-136. [DOI: 10.1016/j.ejphar.2016.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 11/17/2022]
|
124
|
Pannell M, Labuz D, Celik MÖ, Keye J, Batra A, Siegmund B, Machelska H. Adoptive transfer of M2 macrophages reduces neuropathic pain via opioid peptides. J Neuroinflammation 2016; 13:262. [PMID: 27717401 PMCID: PMC5055715 DOI: 10.1186/s12974-016-0735-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022] Open
Abstract
Background During the inflammation which occurs following nerve damage, macrophages are recruited to the site of injury. Phenotypic diversity is a hallmark of the macrophage lineage and includes pro-inflammatory M1 and anti-inflammatory M2 populations. Our aim in this study was to investigate the ability of polarized M0, M1, and M2 macrophages to secrete opioid peptides and to examine their relative contribution to the modulation of neuropathic pain. Methods Mouse bone marrow-derived cells were cultured as unstimulated M0 macrophages or were stimulated into an M1 phenotype using lipopolysaccharide and interferon-γ or into an M2 phenotype using interleukin-4. The macrophage phenotypes were verified using flow cytometry for surface marker analysis and cytokine bead array for cytokine profile assessment. Opioid peptide levels were measured by radioimmunoassay and enzyme immunoassay. As a model of neuropathic pain, a chronic constriction injury (CCI) of the sciatic nerve was employed. Polarized M0, M1, and M2 macrophages (5 × 105 cells) were injected perineurally twice, on days 14 and 15 following CCI or sham surgery. Mechanical and heat sensitivity were measured using the von Frey and Hargreaves tests, respectively. To track the injected macrophages, we also transferred fluorescently stained polarized cells and analyzed the surface marker profile of endogenous and injected cells in the nerves ex vivo. Results Compared to M0 and M1 cells, M2 macrophages contained and released higher amounts of opioid peptides, including Met-enkephalin, dynorphin A (1–17), and β-endorphin. M2 cells transferred perineurally at the nerve injury site reduced mechanical, but not heat hypersensitivity following the second injection. The analgesic effect was reversed by the perineurally applied opioid receptor antagonist naloxone methiodide. M2 cells did not affect sensitivity following sham surgery. Neither M0 nor M1 cells altered mechanical and heat sensitivity in CCI or sham-operated animals. Tracing the fluorescently labeled M0, M1, and M2 cells ex vivo showed that they remained in the nerve and preserved their phenotype. Conclusions Perineural transplantation of M2 macrophages resulted in opioid-mediated amelioration of neuropathy-induced mechanical hypersensitivity, while M1 macrophages did not exacerbate pain. Therefore, rather than focusing on macrophage-induced pain generation, promoting opioid-mediated M2 actions may be more relevant for pain control.
Collapse
Affiliation(s)
- Maria Pannell
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Dominika Labuz
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Melih Ö Celik
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Jacqueline Keye
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Arvind Batra
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Britta Siegmund
- Medizinische Klinik für Gastroenterologie, Infektiologie und Rheumatologie, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Halina Machelska
- Department of Anesthesiology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203, Berlin, Germany.
| |
Collapse
|
125
|
Guglielmetti C, Le Blon D, Santermans E, Salas-Perdomo A, Daans J, De Vocht N, Shah D, Hoornaert C, Praet J, Peerlings J, Kara F, Bigot C, Mai Z, Goossens H, Hens N, Hendrix S, Verhoye M, Planas AM, Berneman Z, van der Linden A, Ponsaerts P. Interleukin-13 immune gene therapy prevents CNS inflammation and demyelination via alternative activation of microglia and macrophages. Glia 2016; 64:2181-2200. [PMID: 27685637 DOI: 10.1002/glia.23053] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 07/01/2016] [Accepted: 08/11/2016] [Indexed: 02/06/2023]
Abstract
Detrimental inflammatory responses in the central nervous system are a hallmark of various brain injuries and diseases. With this study we provide evidence that lentiviral vector-mediated expression of the immune-modulating cytokine interleukin 13 (IL-13) induces an alternative activation program in both microglia and macrophages conferring protection against severe oligodendrocyte loss and demyelination in the cuprizone mouse model for multiple sclerosis (MS). First, IL-13 mediated modulation of cuprizone induced lesions was monitored using T2 -weighted magnetic resonance imaging and magnetization transfer imaging, and further correlated with quantitative histological analyses for inflammatory cell influx, oligodendrocyte death, and demyelination. Second, following IL-13 immune gene therapy in cuprizone-treated eGFP+ bone marrow chimeric mice, we provide evidence that IL-13 directs the polarization of both brain-resident microglia and infiltrating macrophages towards an alternatively activated phenotype, thereby promoting the conversion of a pro-inflammatory environment toward an anti-inflammatory environment, as further evidenced by gene expression analyses. Finally, we show that IL-13 immune gene therapy is also able to limit lesion severity in a pre-existing inflammatory environment. In conclusion, these results highlight the potential of IL-13 to modulate microglia/macrophage responses and to improve disease outcome in a mouse model for MS. GLIA 2016;64:2181-2200.
Collapse
Affiliation(s)
- Caroline Guglielmetti
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Eva Santermans
- Center for Statistics, I-Biostat, Hasselt University, Hasselt, Belgium
| | - Angelica Salas-Perdomo
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Nathalie De Vocht
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Disha Shah
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Chloé Hoornaert
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jelle Praet
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jurgen Peerlings
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Firat Kara
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Christian Bigot
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Zhenhua Mai
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Icometrix, Leuven, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.,Center for Statistics, I-Biostat, Hasselt University, Hasselt, Belgium.,Centre for Health Economic Research and Modelling Infectious Diseases (Chermid), University of Antwerp, Antwerp, Belgium
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Marleen Verhoye
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Annemie van der Linden
- Bio-Imaging Laboratory, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium. .,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
126
|
Li J, Liu Y, Xu H, Fu Q. Nanoparticle-Delivered IRF5 siRNA Facilitates M1 to M2 Transition, Reduces Demyelination and Neurofilament Loss, and Promotes Functional Recovery After Spinal Cord Injury in Mice. Inflammation 2016; 39:1704-17. [DOI: 10.1007/s10753-016-0405-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
127
|
McLean NA, Verge VMK. Dynamic impact of brief electrical nerve stimulation on the neural immune axis-polarization of macrophages toward a pro-repair phenotype in demyelinated peripheral nerve. Glia 2016; 64:1546-61. [PMID: 27353566 DOI: 10.1002/glia.23021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 06/06/2016] [Accepted: 06/07/2016] [Indexed: 12/13/2022]
Abstract
Demyelinating peripheral nerves are infiltrated by cells of the monocyte lineage, including macrophages, which are highly plastic, existing on a continuum from pro-inflammatory M1 to pro-repair M2 phenotypic states. Whether one can therapeutically manipulate demyelinated peripheral nerves to promote a pro-repair M2 phenotype remains to be elucidated. We previously identified brief electrical nerve stimulation (ES) as therapeutically beneficial for remyelination, benefits which include accelerated clearance of macrophages, making us theorize that ES alters the local immune response. Thus, the impact of ES on the immune microenvironment in the zone of demyelination was examined. Adult male rat tibial nerves were focally demyelinated via 1% lysophosphatidyl choline (LPC) injection. Five days later, half underwent 1 hour 20 Hz sciatic nerve ES proximal to the LPC injection site. ES had a remarkable and significant impact, shifting the macrophage phenotype from predominantly pro-inflammatory/M1 toward a predominantly pro-repair/M2 one, as evidenced by an increased incidence of expression of M2-associated phenotypic markers in identified macrophages and a decrease in M1-associated marker expression. This was discernible at 3 days post-ES (8 days post-LPC) and continued at the 5 day post-ES (10 days post-LPC) time point examined. ES also affected chemokine (C-C motif) ligand 2 (CCL2; aka MCP-1) expression in a manner that correlated with increases and decreases in macrophage numbers observed in the demyelination zone. The data establish that briefly increasing neuronal activity favorably alters the immune microenvironment in demyelinated nerve, rapidly polarizing macrophages toward a pro-repair phenotype, a beneficial therapeutic concept that may extend to other pathologies. GLIA 2016;64:1546-1561.
Collapse
Affiliation(s)
- Nikki A McLean
- Department of Anatomy and Cell Biology, CMSNRC (Cameco MS Neuroscience Research Center), University of Saskatchewan, Saskatoon, SK, Canada
| | - Valerie M K Verge
- Department of Anatomy and Cell Biology, CMSNRC (Cameco MS Neuroscience Research Center), University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
128
|
Wang S, Zhang H, Xu Y. Crosstalk between microglia and T cells contributes to brain damage and recovery after ischemic stroke. Neurol Res 2016; 38:495-503. [DOI: 10.1080/01616412.2016.1188473] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
129
|
Schmidt A, Zhang XM, Joshi RN, Iqbal S, Wahlund C, Gabrielsson S, Harris RA, Tegnér J. Human macrophages induce CD4(+)Foxp3(+) regulatory T cells via binding and re-release of TGF-β. Immunol Cell Biol 2016; 94:747-62. [PMID: 27075967 DOI: 10.1038/icb.2016.34] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 12/12/2022]
Abstract
While pro-inflammatory immune responses are a requirement to combat microbes, uncontrolled self-directed inflammatory immune responses are the hallmark of autoimmune diseases. Restoration of immunological tolerance involves both suppression of ongoing tissue-destructive immune responses and re-education of the host immune system. Both functionally immunosuppressive macrophages (M2) and regulatory T cells (Tregs) are implicated in these processes. Their mutual interaction is synergistic in this context and adoptive transfer of each cell type has been functioning as immunotherapy in experimental models, being particularly effective when using M2 macrophages generated with an optimized interleukin-4 (IL-4)/interleukin-10 (IL-10)/transforming growth factor-β (TGF-β) combination. As a prerequisite for eventual translation of M2 therapy into clinical settings we herein studied the induction, stability and mechanism of generation of human induced Tregs (iTregs) by M2 macrophages generated with IL-4/IL-10/TGF-β. The supernatants of monocyte-derived human M2 macrophages robustly induced FOXP3 and other Treg signature molecules such as CTLA-4 and IKZF4 in human naïve CD4 T cells. M2-induced iTregs displayed enhanced FOXP3 stability and low expression of pro-inflammatory cytokines interferon-γ and IL-17, as well as functional immunosuppressive activity compared with control T cells. The FOXP3-inducing activity was dependent on TGF-β, which was both expressed and captured with re-release by M2 macrophages into the soluble supernatant fraction, in which the TGF-β was not confined to extracellular vesicles such as exosomes. We propose that adoptive transfer of human M2 macrophages may be exploited in the future to induce Tregs in situ by delivering TGF-β, which could be developed as a therapeutic strategy to target autoimmune and other inflammatory diseases.
Collapse
Affiliation(s)
- Angelika Schmidt
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, & Science for Life Laboratory, Stockholm, Sweden
| | - Xing-Mei Zhang
- Applied Immunology & Immunotherapy, Center for Molecular Medicine, Karolinska University Hospital at Solna, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Rubin N Joshi
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, & Science for Life Laboratory, Stockholm, Sweden
| | - Shasina Iqbal
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, & Science for Life Laboratory, Stockholm, Sweden
| | - Casper Wahlund
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet & Karolinska University Hospital, Stockholm, Sweden
| | - Susanne Gabrielsson
- Translational Immunology Unit, Department of Medicine Solna, Karolinska Institutet & Karolinska University Hospital, Stockholm, Sweden
| | - Robert A Harris
- Applied Immunology & Immunotherapy, Center for Molecular Medicine, Karolinska University Hospital at Solna, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, & Science for Life Laboratory, Stockholm, Sweden
| |
Collapse
|
130
|
Hu JG, Shi LL, Chen YJ, Xie XM, Zhang N, Zhu AY, Jiang ZS, Feng YF, Zhang C, Xi J, Lü HZ. Differential effects of myelin basic protein-activated Th1 and Th2 cells on the local immune microenvironment of injured spinal cord. Exp Neurol 2016; 277:190-201. [PMID: 26772636 DOI: 10.1016/j.expneurol.2016.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/01/2016] [Accepted: 01/04/2016] [Indexed: 01/22/2023]
Abstract
Myelin basic protein (MBP) activated T cells (MBP-T) play an important role in the damage and repair process of the central nervous system (CNS). However, whether these cells play a beneficial or detrimental role is still a matter of debate. Although some studies showed that MBP-T cells are mainly helper T (Th) cells, their subtypes are still not very clear. One possible explanation for MBP-T immunization leading to conflicting results may be the different subtypes of T cells are responsible for distinct effects. In this study, the Th1 and Th2 type MBP-T cells (MBP-Th1 and -Th2) were polarized in vitro, and their effects on the local immune microenvironment and tissue repair of spinal cord injury (SCI) after adoptive immunization were investigated. In MBP-Th1 cell transferred rats, the high levels of pro-inflammatory cells (Th1 cells and M1 macrophages) and cytokines (IFN-γ, TNF-α, -β, IL-1β) were detected in the injured spinal cord; however, the anti-inflammatory cells (Th2 cells, regulatory T cells, and M2 macrophages) and cytokines (IL-4, -10, and -13) were found in MBP-Th2 cell transferred animals. MBP-Th2 cell transfer resulted in decreased lesion volume, increased myelination of axons, and preservation of neurons. This was accompanied by significant locomotor improvement. These results indicate that MBP-Th2 adoptive transfer has beneficial effects on the injured spinal cord, in which the increased number of Th2 cells may alter the local microenvironment from one primarily populated by Th1 and M1 cells to another dominated by Th2, Treg, and M2 cells and is conducive for SCI repair.
Collapse
Affiliation(s)
- Jian-Guo Hu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Ling-Ling Shi
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Yue-Juan Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Xiu-Mei Xie
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Nan Zhang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - An-You Zhu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Zheng-Song Jiang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China
| | - Yi-Fan Feng
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Chen Zhang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - Jin Xi
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Anhui 233004, PR China; Department of Immunology, Bengbu Medical College, Anhui 233030, PR China.
| |
Collapse
|
131
|
Papa S, Caron I, Erba E, Panini N, De Paola M, Mariani A, Colombo C, Ferrari R, Pozzer D, Zanier ER, Pischiutta F, Lucchetti J, Bassi A, Valentini G, Simonutti G, Rossi F, Moscatelli D, Forloni G, Veglianese P. Early modulation of pro-inflammatory microglia by minocycline loaded nanoparticles confers long lasting protection after spinal cord injury. Biomaterials 2016; 75:13-24. [DOI: 10.1016/j.biomaterials.2015.10.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 12/30/2022]
|
132
|
Cao XJ, Feng SQ, Fu CF, Gao K, Guo JS, Guo XD, He XJ, Huang ZW, Li ZH, Liu L, Liu RH, Lü HZ, Mei XF, Ning B, Ning GZ, Qian CH, Qin J, Qu YZ, Saijilafu, Shi B, Sui T, Sun TS, Wang J, Wen JK, Xiao J, Xu B, Xu HD, Yu PP, Zhang ZC, Zhou Y, Zhou YL. Repair, protection and regeneration of spinal cord injury. Neural Regen Res 2015; 10:1953-75. [PMID: 26889184 PMCID: PMC4730820 DOI: 10.4103/1673-5374.172314] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
133
|
Azithromycin drives alternative macrophage activation and improves recovery and tissue sparing in contusion spinal cord injury. J Neuroinflammation 2015; 12:218. [PMID: 26597676 PMCID: PMC4657208 DOI: 10.1186/s12974-015-0440-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/18/2015] [Indexed: 01/26/2023] Open
Abstract
Background Macrophages persist indefinitely at sites of spinal cord injury (SCI) and contribute to both pathological and reparative processes. While the alternative, anti-inflammatory (M2) phenotype is believed to promote cell protection, regeneration, and plasticity, pro-inflammatory (M1) macrophages persist after SCI and contribute to protracted cell and tissue loss. Thus, identifying non-invasive, clinically viable, pharmacological therapies for altering macrophage phenotype is a challenging, yet promising, approach for treating SCI. Azithromycin (AZM), a commonly used macrolide antibiotic, drives anti-inflammatory macrophage activation in rodent models of inflammation and in humans with cystic fibrosis. Methods We hypothesized that AZM treatment can alter the macrophage response to SCI and reduce progressive tissue pathology. To test this hypothesis, mice (C57BL/6J, 3-month-old) received daily doses of AZM (160 mg/kg) or vehicle treatment via oral gavage for 3 days prior and up to 7 days after a moderate-severe thoracic contusion SCI (75-kdyn force injury). Fluorescent-activated cell sorting was used in combination with real-time PCR (rtPCR) to evaluate the disposition and activation status of microglia, monocytes, and neutrophils, as well as macrophage phenotype in response to AZM treatment. An open-field locomotor rating scale (Basso Mouse Scale) and gridwalk task were used to determine the effects of AZM treatment on SCI recovery. Bone marrow-derived macrophages (BMDMs) were used to determine the effect of AZM treatment on macrophage phenotype in vitro. Results In accordance with our hypothesis, SCI mice exhibited significantly increased anti-inflammatory and decreased pro-inflammatory macrophage activation in response to AZM treatment. In addition, AZM treatment led to improved tissue sparing and recovery of gross and coordinated locomotor function. Furthermore, AZM treatment altered macrophage phenotype in vitro and lowered the neurotoxic potential of pro-inflammatory, M1 macrophages. Conclusions Taken together, these data suggest that pharmacologically intervening with AZM can alter SCI macrophage polarization toward a beneficial phenotype that, in turn, may potentially limit secondary injury processes. Given that pro-inflammatory macrophage activation is a hallmark of many neurological pathologies and that AZM is non-invasive and clinically viable, these data highlight a novel approach for treating SCI and other maladaptive neuroinflammatory conditions. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0440-3) contains supplementary material, which is available to authorized users.
Collapse
|
134
|
Loane DJ, Kumar A. Microglia in the TBI brain: The good, the bad, and the dysregulated. Exp Neurol 2015; 275 Pt 3:316-327. [PMID: 26342753 DOI: 10.1016/j.expneurol.2015.08.018] [Citation(s) in RCA: 493] [Impact Index Per Article: 54.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 08/05/2015] [Accepted: 08/25/2015] [Indexed: 01/24/2023]
Abstract
As the major cellular component of the innate immune system in the central nervous system (CNS) and the first line of defense whenever injury or disease occurs, microglia play a critical role in neuroinflammation following a traumatic brain injury (TBI). In the injured brain microglia can produce neuroprotective factors, clear cellular debris and orchestrate neurorestorative processes that are beneficial for neurological recovery after TBI. However, microglia can also become dysregulated and can produce high levels of pro-inflammatory and cytotoxic mediators that hinder CNS repair and contribute to neuronal dysfunction and cell death. The dual role of microglial activation in promoting beneficial and detrimental effects on neurons may be accounted for by their polarization state and functional responses after injury. In this review article we discuss emerging research on microglial activation phenotypes in the context of acute brain injury, and the potential role of microglia in phenotype-specific neurorestorative processes such as neurogenesis, angiogenesis, oligodendrogenesis and regeneration. We also describe some of the known molecular mechanisms that regulate phenotype switching, and highlight new therapeutic approaches that alter microglial activation state balance to enhance long-term functional recovery after TBI. An improved understanding of the regulatory mechanisms that control microglial phenotypic shifts may advance our knowledge of post-injury recovery and repair, and provide opportunities for the development of novel therapeutic strategies for TBI.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States; Shock, Trauma, and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States.
| | - Alok Kumar
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, United States; Shock, Trauma, and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
135
|
Zhang B, Bailey WM, Braun KJ, Gensel JC. Age decreases macrophage IL-10 expression: Implications for functional recovery and tissue repair in spinal cord injury. Exp Neurol 2015; 273:83-91. [PMID: 26263843 DOI: 10.1016/j.expneurol.2015.08.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 07/31/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022]
Abstract
Macrophages with different activation states are present after spinal cord injury (SCI). M1 macrophages purportedly promote secondary injury processes while M2 cells support axon growth. The average age at the time of SCI has increased in recent decades, however, little is known about how different physiological factors contribute to macrophage activation states after SCI. Here we investigate the effect of age on IL-10, a key indicator of M2 macrophage activation. Following mild-moderate SCI in 4 and 14 month old (MO) mice we detected significantly reduced IL-10 expression with age in the injured spinal cord. Specifically, CD86/IL-10 positive macrophages, also known as M2b or regulatory macrophages, were reduced in 14 vs. 4 MO SCI animals. This age-dependent shift in macrophage phenotype was associated with impaired functional recovery and enhanced tissue damage in 14-month-old SCI mice. In vitro, M2b macrophages release anti-inflammatory cytokines without causing neurotoxicity, suggesting that imbalances in the M2b response in 14-month-old mice may be contributing to secondary injury processes. Our data indicate that age is an important factor that regulates SCI inflammation and recovery even to mild-moderate injury. Further, alterations in macrophage activation states may contribute to recovery and we have identified the M2b phenotype as a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Bei Zhang
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536, United States
| | - William M Bailey
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536, United States
| | - Kaitlyn J Braun
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536, United States
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|