51
|
Park J, Chang JY, Kim JY, Lee JE. Monocyte Transmodulation: The Next Novel Therapeutic Approach in Overcoming Ischemic Stroke? Front Neurol 2020; 11:578003. [PMID: 33193029 PMCID: PMC7642685 DOI: 10.3389/fneur.2020.578003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
The immune response following neuroinflammation is a vital element of ischemic stroke pathophysiology. After the onset of ischemic stroke, a specialized vasculature system that effectively protects central nervous system tissues from the invasion of blood cells and other macromolecules is broken down within minutes, thereby triggering the inflammation cascade, including the infiltration of peripheral blood leukocytes. In this series of processes, blood-derived monocytes have a significant effect on the outcome of ischemic stroke through neuroinflammatory responses. As neuroinflammation is a necessary and pivotal component of the reparative process after ischemic stroke, understanding the role of infiltrating monocytes in the modulation of inflammatory responses may offer a great opportunity to explore new therapies for ischemic stroke. In this review, we discuss and highlight the function and involvement of monocytes in the brain after ischemic injury, as well as their impact on tissue damage and repair.
Collapse
Affiliation(s)
- Joohyun Park
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Young Chang
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Youl Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Brain Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
52
|
The role of peripheral monocytes and macrophages in ischemic stroke. Neurol Sci 2020; 41:3589-3607. [PMID: 33009963 DOI: 10.1007/s10072-020-04777-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
Abstract
After acute ischemic stroke (AIS), peripheral monocytes infiltrate into the lesion site within 24 h, peak at 3 to 7 days, and then differentiate into macrophages. Traditionally, monocytes/macrophages (MMs) are thought to play a deleterious role in AIS. Depletion of MMs in the acute phase can alleviate brain injury induced by ischemia. However, several studies have shown that MMs have anti-inflammatory functions, participate in angiogenesis, phagocytose necrotic neurons, and promote neurovascular repair. Therefore, MMs play dual roles in ischemic stroke, depending mainly upon the MM microenvironment and the window of time post-stroke. Because activated microglia and MMs are similar in morphology and function, previous studies have often investigated them together. However, recent studies have used special methods to distinguish MMs from microglia and have found that MMs have properties which differ from microglia. Here, we review the unique role of MMs and the interaction between MMs and neurovascular units, including neurons, astrocytes, microglia, and microvessels. Future therapeutics targeting MMs should regulate the polarization and subset transformation of the MMs at different stages of AIS rather than comprehensively suppressing MM infiltration and differentiation. In addition, more studies are needed to elucidate the cellular and molecular mechanisms of MM subsets and polarization during ischemic stroke.
Collapse
|
53
|
Sariol A, Mackin S, Allred MG, Ma C, Zhou Y, Zhang Q, Zou X, Abrahante JE, Meyerholz DK, Perlman S. Microglia depletion exacerbates demyelination and impairs remyelination in a neurotropic coronavirus infection. Proc Natl Acad Sci U S A 2020; 117:24464-24474. [PMID: 32929007 PMCID: PMC7533697 DOI: 10.1073/pnas.2007814117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microglia are considered both pathogenic and protective during recovery from demyelination, but their precise role remains ill defined. Here, using an inhibitor of colony stimulating factor 1 receptor (CSF1R), PLX5622, and mice infected with a neurotropic coronavirus (mouse hepatitis virus [MHV], strain JHMV), we show that depletion of microglia during the time of JHMV clearance resulted in impaired myelin repair and prolonged clinical disease without affecting the kinetics of virus clearance. Microglia were required only during the early stages of remyelination. Notably, large deposits of extracellular vesiculated myelin and cellular debris were detected in the spinal cords of PLX5622-treated and not control mice, which correlated with decreased numbers of oligodendrocytes in demyelinating lesions in drug-treated mice. Furthermore, gene expression analyses demonstrated differential expression of genes involved in myelin debris clearance, lipid and cholesterol recycling, and promotion of oligodendrocyte function. The results also demonstrate that microglial functions affected by depletion could not be compensated by infiltrating macrophages. Together, these results demonstrate that microglia play key roles in debris clearance and in the initiation of remyelination following infection with a neurotropic coronavirus but are not necessary during later stages of remyelination.
Collapse
Affiliation(s)
- Alan Sariol
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Samantha Mackin
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Merri-Grace Allred
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| | - Chen Ma
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Yu Zhou
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Qinran Zhang
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, 430072 Wuhan, China
| | - Juan E Abrahante
- University of Minnesota Informatics Institute (UMII), Minneapolis, MN 55455
| | | | - Stanley Perlman
- Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
54
|
Murata Y, Sugimoto K, Yang C, Harada K, Gono R, Harada T, Miyashita Y, Higashisaka K, Katada R, Tanaka J, Matsumoto H. Activated microglia-derived macrophage-like cells exacerbate brain edema after ischemic stroke correlate with astrocytic expression of aquaporin-4 and interleukin-1 alpha release. Neurochem Int 2020; 140:104848. [PMID: 32920036 DOI: 10.1016/j.neuint.2020.104848] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023]
Abstract
Brain edema following brain infarction affects mobility and mortality. The mechanisms underlying this process remain to be elucidated. Animal studies have shown that aquaporin-4 (AQP4) expression in astrocytes increases after stroke, and its deletion significantly reduces brain swelling. Recently, two kinds of cells, resident microglia-derived macrophage-like cells (MG-MΦ) and bone marrow-derived macrophages (BM-MΦ), have been reported to accumulate in the ischemic core and stimulate adjacent astrocytes. Therefore, we hypothesized that these cells play crucial roles in the expression of AQP4 and ultimately lead to exacerbated brain edema. To verify this hypothesis, we investigated the role of MG- or BM-MΦ in brain edema using a rat model of transient middle cerebral artery occlusion and rat astrocyte primary cultures. AQP4 expression significantly increased in the peri-infarct tissue at 3-7 days post-reperfusion (dpr) and in the core tissue at 5 and 7 dpr, which synchronized with the expression of Iba1, Il1a, Tnf, and C1qa mRNA. Interleukin (IL)-1α treatment or coculture with MG- and BM-MΦ increased AQP4 expression in astrocytes, while an IL-1 receptor type I antagonist reduced these effects. Furthermore, aggravated animals exhibited high expression of Aqp4 and Il1a mRNA in the ischemic core at 7 dpr, which led to the exacerbation of brain edema. MG-MΦ signature genes were highly expressed in the ischemic core in aggravated rats, while BM-MΦ signature genes were weakly expressed. These findings suggest that IL-1α produced by MG-MΦ induces astrocytic AQP4 expression in the peri-infarct and ischemic core tissues, thereby exacerbating brain edema. Therefore, the regulation of MG-MΦ may prevent the exacerbation of brain edema.
Collapse
Affiliation(s)
- Yukie Murata
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Kana Sugimoto
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Chihpin Yang
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Kazuo Harada
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Rina Gono
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Teiji Harada
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Yohei Miyashita
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Kazuma Higashisaka
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Ryuichi Katada
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, 791-0295, Japan.
| | - Hiroshi Matsumoto
- Department of Legal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
55
|
Gan YM, Liu DL, Chen C, Duan W, Yang YX, Du JR. Phthalide derivative CD21 alleviates cerebral ischemia-induced neuroinflammation: Involvement of microglial M2 polarization via AMPK activation. Eur J Pharmacol 2020; 886:173552. [PMID: 32926919 DOI: 10.1016/j.ejphar.2020.173552] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 09/02/2020] [Accepted: 09/10/2020] [Indexed: 12/17/2022]
Abstract
Microglia can be activated to become the classic phenotype (M1) or alternative phenotype (M2), which play an important role in regulating neuroinflammatory response and tissue repair after ischemic stroke. CD21, a novel phthalide derivative, is a potential neuroprotectant against ischemic brain injury. The present study further investigated the effects of CD21 on post-ischemic microglial polarization and the underlying mechanisms. Transient middle cerebral artery occlusion (tMCAO) was used as a mouse model of ischemic stroke, while BV2 cells stimulated with conditioned medium collected from oxygen-glucose deprivation-treated HT22 cells were used in in vitro ischemic studies. The current results showed that CD21 dose-dependently and significantly improved neurological outcomes in tMCAO mice. Biochemical analyses revealed that CD21 decreased the expression of M1 phenotype markers (CD86, interleukin-1β and inducible nitric oxide synthase) and increased the expression of M2 phenotype markers (CD206, interleukin-10 and YM1/2) in both ischemic brain tissues and BV2 cells. Meanwhile, CD21 decreased the production of proinflammatory cytokines (interleukin-1β, interleukin-6 and tumor necrosis factor-α), promoted the release of the antiinflammatory cytokine (interleukin-10), and enhanced the phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK) in ischemic brain tissue and BV2 cells. Furthermore, the AMPK inhibitor (compound C) reversed these effects of CD21 in BV2 cells. These findings indicate that CD21 alleviates post-ischemic neuroinflammation through induction of microglial M2 polarization that is at least in part medicated by AMPK activation, suggesting that CD21 may be a promising candidate for protecting against ischemic brain injury.
Collapse
Affiliation(s)
- Yu-Miao Gan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Dong-Ling Liu
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Chu Chen
- Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610041, PR China
| | - Wei Duan
- School of Medicine and Centre for Molecular and Medical Research, Deakin University, Waurn Ponds, Victoria, Australia
| | - Yu-Xin Yang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China
| | - Jun-Rong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
56
|
Wang D, Liu F, Zhu L, Lin P, Han F, Wang X, Tan X, Lin L, Xiong Y. FGF21 alleviates neuroinflammation following ischemic stroke by modulating the temporal and spatial dynamics of microglia/macrophages. J Neuroinflammation 2020; 17:257. [PMID: 32867781 PMCID: PMC7457364 DOI: 10.1186/s12974-020-01921-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022] Open
Abstract
Background Resident microglia and macrophages are the predominant contributors to neuroinflammation and immune reactions, which play a critical role in the pathogenesis of ischemic brain injury. Controlling inflammatory responses is considered a promising therapeutic approach for stroke. Recombinant human fibroblast growth factor 21 (rhFGF21) presents anti-inflammatory properties by modulating microglia and macrophages; however, our knowledge of the inflammatory modulation of rhFGF21 in focal cerebral ischemia is lacking. Therefore, we investigated whether rhFGF21 improves ischemic outcomes in experimental stroke by targeting microglia and macrophages. Methods C57BL/6 mice were subjected to middle cerebral artery occlusion (MCAO) and randomly divided into groups that received intraperitoneal rhFGF21 or vehicle daily starting at 6 h after reperfusion. Behavior assessments were monitored for 14 days after MCAO, and the gene expression levels of inflammatory cytokines were analyzed via qRT-PCR. The phenotypic variation of microglia/macrophages and the presence of infiltrated immune cells were examined by flow cytometry and immunostaining. Additionally, magnetic cell sorting (MACS) in combination with fluorescence-activated cell sorting (FACS) was used to purify microglia and macrophages. Results rhFGF21 administration ameliorated neurological deficits in behavioral tests by regulating the secretion of pro-inflammatory and anti-inflammatory cytokines. rhFGF21 also attenuated the polarization of microglia/macrophages toward the M1 phenotype and the accumulation of peripheral immune cells after stroke, accompanied by a temporal evolution of the phenotype of microglia/macrophages and infiltration of peripheral immune cells. Furthermore, rhFGF21 treatment inhibited M1 polarization of microglia and pro-inflammatory cytokine expression through its actions on FGF receptor 1 (FGFR1) by suppressing nuclear factor-kappa B (NF-κB) and upregulating peroxisome proliferator-activated receptor-γ (PPAR-γ). Conclusions rhFGF21 treatment promoted functional recovery in experimental stroke by modulating microglia/macrophage-mediated neuroinflammation via the NF-κB and PPAR-γ signaling pathways, making it a potential anti-inflammatory agent for stroke treatment.
Collapse
Affiliation(s)
- Dongxue Wang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fei Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Liyun Zhu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ping Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xue Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Xianxi Tan
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Li Lin
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China. .,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Ye Xiong
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
57
|
Battaglini D, Pimentel-Coelho PM, Robba C, dos Santos CC, Cruz FF, Pelosi P, Rocco PRM. Gut Microbiota in Acute Ischemic Stroke: From Pathophysiology to Therapeutic Implications. Front Neurol 2020; 11:598. [PMID: 32670191 PMCID: PMC7330114 DOI: 10.3389/fneur.2020.00598] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/22/2020] [Indexed: 12/16/2022] Open
Abstract
The microbiota-gut-brain axis is considered a central regulator of the immune system after acute ischemic stroke (AIS), with a potential role in determining outcome. Several pathways are involved in the evolution of gut microbiota dysbiosis after AIS. Brain-gut and gut-brain signaling pathways involve bidirectional communication between the hypothalamic-pituitary-adrenal axis, the autonomic nervous system, the enteric nervous system, and the immune cells of the gut. Alterations in gut microbiome can be a risk factor and may also lead to AIS. Both risk factors for AIS and gut-microbiome composition are influenced by similar factors, including diabetes, hypertension, hyperlipidemia, obesity, and vascular dysfunction. Furthermore, the systemic inflammatory response after AIS may yield liver, renal, respiratory, gastrointestinal, and cardiovascular impairment, including the multiple organ dysfunction syndrome. This review focus on biochemical, immunological, and neuroanatomical modulation of gut microbiota and its possible systemic harmful effects after AIS, as well as the role of ischemic stroke on microbiota composition. Finally, we highlight the role of gut microbiota as a potential novel therapeutic target in acute ischemic stroke.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Pedro Moreno Pimentel-Coelho
- Laboratório de Neurobiologia Comparada e do Desenvolvimento, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Claudia C. dos Santos
- Keenan and Li Ka Shing Knowledge Institute, University Health Toronto—St. Michael's Hospital, Toronto, ON, Canada
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Network on Neuroinflammation, Carlos Chagas Filho Foundation for Supporting Research in the State of Rio de Janeiro (FAPERJ), Rio de Janeiro, Brazil
| |
Collapse
|
58
|
Sharmin O, Abir AH, Potol A, Alam M, Banik J, Rahman AFMT, Tarannum N, Wadud R, Habib ZF, Rahman M. Activation of GPR35 protects against cerebral ischemia by recruiting monocyte-derived macrophages. Sci Rep 2020; 10:9400. [PMID: 32523084 PMCID: PMC7287103 DOI: 10.1038/s41598-020-66417-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
Pamoic acid is a potent ligand for G protein Coupled Receptor 35 (GPR35) and exhibits antinociceptive property. GPR35 activation leads to increased energy utilization and the expression of anti-inflammatory genes. However, its role in brain disorders, especially in stroke, remains unexplored. Here we show in a mouse model of stroke that GPR35 activation by pamoic acid is neuroprotective. Pharmacological inhibition of GPR35 reveals that pamoic acid reduces infarcts size in a GPR35 dependent manner. The flowcytometric analysis shows the expression of GPR35 on the infiltrating monocytes/macrophages and neutrophils in the ischemic brain. Pamoic acid treatment results in a preferential increment of noninflammatory Ly-6CLo monocytes/macrophages in the ischemic brain along with the reduced neutrophil counts. The neuroprotective effect of GPR35 activation depends on protein kinase B (Akt) and p38 MAPK. Together we conclude that GPR35 activation by pamoic acid reprograms Ly-6CLo monocytes/macrophages to relay a neuroprotective signal into the ischemic brain.
Collapse
Affiliation(s)
- Ozayra Sharmin
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh
| | - Ariful Haque Abir
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh
| | - Abdullah Potol
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.,Faculty of Medicine, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Mahabub Alam
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh
| | - Jewel Banik
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.,Deptartment of Neurobiology & Developmental Sciences, College of Medicine, UAMS, 4301W. Markham St., Little Rock, AR, 72205, USA
| | - A F M Towheedur Rahman
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.,Milwaukee Institute of Drug Discovery, Department of chemistry and Biochemistry, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Nuzhat Tarannum
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh
| | - Rasiqh Wadud
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.,Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Zaki Farhad Habib
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.,Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge, UK
| | - Mahbubur Rahman
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health & Life Sciences, North South University, Bashundhra R/A, Dhaka, 1229, Bangladesh.
| |
Collapse
|
59
|
Wang Y, Luo Y, Yao Y, Ji Y, Feng L, Du F, Zheng X, Tao T, Zhai X, Li Y, Han P, Xu B, Zhao H. Silencing the lncRNA Maclpil in pro-inflammatory macrophages attenuates acute experimental ischemic stroke via LCP1 in mice. J Cereb Blood Flow Metab 2020; 40:747-759. [PMID: 30895879 PMCID: PMC7168792 DOI: 10.1177/0271678x19836118] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Long noncoding RNAs (lncRNA) expression profiles change in the ischemic brain after stroke, but their roles in specific cell types after stroke have not been studied. We tested the hypothesis that lncRNA modulates brain injury by altering macrophage functions. Using RNA deep sequencing, we identified 73 lncRNAs that were differentially expressed in monocyte-derived macrophages (MoDMs) and microglia-derived macrophages (MiDMs) isolated in the ischemic brain three days after stroke. Among these, the lncRNA, GM15628, is highly expressed in pro-inflammatory MoDMs but not in MiDMs, and are functionally related to its neighbor gene, lymphocyte cytosolic protein 1 (LCP1), which plays a role in maintaining cell shape and cell migration. We termed this lncRNA as Macrophage contained LCP1 related pro-inflammatory lncRNA, Maclpil. Using cultured macrophages polarized by LPS, M(LPS), we found that downregulation of Maclpil in M(LPS) decreased pro-inflammatory gene expression while promoting anti-inflammatory gene expression. Maclpil inhibition also reduced the migration and phagocytosis ability of MoDMs by inhibiting LCP1. Furthermore, adoptive transfer of Maclpil silenced M(LPS), reduced ischemic brain infarction, improved behavioral performance and attenuated penetration of MoDMs in the ischemic hemisphere. We conclude that by blocking macrophage, Maclpil protects against acute ischemic stroke by inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Ying Luo
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yang Yao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhua Ji
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Liangshu Feng
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Fang Du
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaoya Zheng
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Tao Tao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xuan Zhai
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yaning Li
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Pei Han
- Department of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
60
|
Eng J, Orf J, Perez K, Sawant D, DeVoss J. Generation of bone marrow chimeras using X-ray irradiation: comparison to cesium irradiation and use in immunotherapy. J Biol Methods 2020; 7:e125. [PMID: 32206674 PMCID: PMC7082502 DOI: 10.14440/jbm.2020.314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/28/2019] [Accepted: 11/28/2019] [Indexed: 01/06/2023] Open
Abstract
Bone marrow chimeras represent a key tool employed to understand biological contributions stemming from the hematopoietic versus the stromal compartment. In most institutions, cesium irradiators are used to lethally irradiate recipient animals prior to the injection of donor bone marrow. Cesium irradiators, however, have significant liabilities—including concerns around domestic security. Recently, X-ray irradiators have been implemented as a potential alternative to cesium sources. Only a small number of publications in the literature have attempted to compare these two modalities and, in most cases, the emphasis was on irradiation of human blood productions. We were able to find only a single study that directly compared X-ray and cesium technologies in the generation of murine bone marrow chimeras, a standard laboratory practice. This study focused on chimerism in the blood of recipient animals. In the present study, we begin by comparing cesium and X-ray based sources for irradiation, then transition to using X-ray-based systems for immunology models with an emphasis on immunotherapy of cancer in immunocompetent mouse models—specifically evaluating chimerism in the blood, spleen, and tumor microenvironment. While our data demonstrate that the two platforms are functionally comparable and suggest that X-ray based technology is a suitable alternative to cesium sources. We also highlight a difference in chimerism between the peripheral (blood, spleen) and tumor compartments that is observed using both technologies. While the overall degree of chimerism in the peripheral tissues is very high, the degree of chimerism in the tumor is cell type specific with T and NK cells showing lower chimerism than other cell types.
Collapse
Affiliation(s)
- Jason Eng
- Amgen Research, Department of Oncology, South San Francisco, CA 94080, USA
| | - Jessica Orf
- Amgen Research, Department of Oncology, South San Francisco, CA 94080, USA
| | - Kristy Perez
- Amgen Research, Department of Oncology, South San Francisco, CA 94080, USA
| | - Deepali Sawant
- Amgen Research, Department of Oncology, South San Francisco, CA 94080, USA
| | - Jason DeVoss
- Amgen Research, Department of Oncology, South San Francisco, CA 94080, USA
| |
Collapse
|
61
|
Gong Z, Wang C, Ni L, Ying L, Shu J, Wang J, Yu C, Xia K, Cheng F, Shi K, Xu G, Yu Q, Shen J, Chen Q, Li F, Liang C. An injectable recombinant human milk fat globule-epidermal growth factor 8-loaded copolymer system for spinal cord injury reduces inflammation through NF-κB and neuronal cell death. Cytotherapy 2020; 22:193-203. [PMID: 32173261 DOI: 10.1016/j.jcyt.2020.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/19/2020] [Accepted: 01/30/2020] [Indexed: 01/17/2023]
Abstract
Spinal cord injury (SCI) is a common disease and a major cause of paralysis, carrying much burden around the world. Despite the progress made with growth factors therapy, the response rate of acute SCI treatment still remains unsatisfactory, due largely to complex and severe inflammatory reactions. Herein, we prepare a MFG-E8-loaded copolymer system-based anti-inflammation therapy for SCI treatment. It is shown that the MFG-E8-loaded copolymer system can decrease pro-inflammatory cytokine expression and neuron death. In a rat model of crush-caused SCI, the copolymer system shows significant therapeutic efficacy by ameliorating inflammation, decreasing fibrotic scar, promoting myelin regeneration and suppressing overall SCI severity.
Collapse
Affiliation(s)
- Zhe Gong
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chenggui Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Licheng Ni
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Liwei Ying
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jiawei Shu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jingkai Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chao Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Feng Cheng
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Kesi Shi
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Guoping Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Qunfei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | | | - Qixin Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Fangcai Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China; Orthopedics Research Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
62
|
Keilhoff G, Titze M, Rathert H, Lucas B, Esser T, Ebmeyer U. Normoxic post-ROSC ventilation delays hippocampal CA1 neurodegeneration in a rat cardiac arrest model, but does not prevent it. Exp Brain Res 2020; 238:807-824. [PMID: 32125470 DOI: 10.1007/s00221-020-05746-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/03/2020] [Indexed: 01/21/2023]
Abstract
The European Resuscitation Guidelines recommend that survivors of cardiac arrest (CA) be resuscitated with 100% O2 and undergo subsequent-post-return of spontaneous circulation (ROSC)-reduction of O2 supply to prevent hyperoxia. Hyperoxia produces a "second neurotoxic hit," which, together with the initial ischemic insult, causes ischemia-reperfusion injury. However, heterogeneous results from animal studies suggest that normoxia can also be detrimental. One clear reason for these inconsistent results is the considerable heterogeneity of the models used. In this study, the histological outcome of the hippocampal CA1 region following resuscitation with 100% O2 combined with different post-ROSC ventilation regimes (21%, 50%, and 100% O2) was investigated in a rat CA/resuscitation model with survival times of 7 and 21 days. Immunohistochemical stainings of NeuN, MAP2, GFAP, and IBA1 revealed a neuroprotective potency of post-ROSC ventilation with 21% O2, although it was only temporary. This limitation should be because of the post-ROSC intervention targeting only processes of ischemia-induced secondary injury. There were no ventilation-dependent effects on either microglial activation, reduction of which is accepted as being neuroprotective, or astroglial activation, which is accepted as being able to enhance neurons' resistance to ischemia/reperfusion injury. Furthermore, our findings verify the limited comparability of animal studies because of the individual heterogeneity of the animals, experimental regimes, and evaluation procedures used.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany.
| | - Maximilian Titze
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - Henning Rathert
- Institute of Biochemistry and Cell Biology, Medical Faculty, University of Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - Benjamin Lucas
- Department of Trauma Surgery, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| | - Torben Esser
- Department of Anesthesiology, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| | - Uwe Ebmeyer
- Department of Anesthesiology, Medical Faculty, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
63
|
Werner Y, Mass E, Ashok Kumar P, Ulas T, Händler K, Horne A, Klee K, Lupp A, Schütz D, Saaber F, Redecker C, Schultze JL, Geissmann F, Stumm R. Cxcr4 distinguishes HSC-derived monocytes from microglia and reveals monocyte immune responses to experimental stroke. Nat Neurosci 2020; 23:351-362. [PMID: 32042176 PMCID: PMC7523735 DOI: 10.1038/s41593-020-0585-y] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/02/2020] [Indexed: 12/26/2022]
Abstract
Monocyte-derived and tissue-resident macrophages are ontogenetically distinct components of the innate immune system. Assessment of their respective functions in pathology is complicated by changes to the macrophage phenotype during inflammation. Here we find that Cxcr4-CreER enables permanent genetic labeling of hematopoietic stem cells (HSCs) and distinguishes HSC-derived monocytes from microglia and other tissue-resident macrophages. By combining Cxcr4-CreER-mediated lineage tracing with Cxcr4 inhibition or conditional Cxcr4 ablation in photothrombotic stroke, we find that Cxcr4 promotes initial monocyte infiltration and subsequent territorial restriction of monocyte-derived macrophages to infarct tissue. After transient focal ischemia, Cxcr4 deficiency reduces monocyte infiltration and blunts the expression of pattern recognition and defense response genes in monocyte-derived macrophages. This is associated with an altered microglial response and deteriorated outcomes. Thus, Cxcr4 is essential for an innate-immune-system-mediated defense response after cerebral ischemia. We further propose Cxcr4-CreER as a universal tool to study functions of HSC-derived cells.
Collapse
Affiliation(s)
- Yves Werner
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Elvira Mass
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Developmental Biology of the Immune System, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany.
| | - Praveen Ashok Kumar
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Kristian Händler
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Arik Horne
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Kathrin Klee
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Dagmar Schütz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Friederike Saaber
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | | | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ralf Stumm
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany.
| |
Collapse
|
64
|
Fang YY, Zhang JH. MFG-E8 alleviates oxygen-glucose deprivation-induced neuronal cell apoptosis by STAT3 regulating the selective polarization of microglia. Int J Neurosci 2020; 131:15-24. [PMID: 32098538 DOI: 10.1080/00207454.2020.1732971] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Background: Ischemic stroke is a complex pathological process, involving inflammatory reaction, energy metabolism disorder, free radical injury, cell apoptosis and other aspects. Accumulating evidences have revealed that MFG-E8 had a protective effect on multiple organ injuries. However, the comprehensive function and mechanism of MFG-E8 in ischemic brain remain largely unclear.Methods: BV-2 cells were treated with recombinant murine MFG-E8 (rmMFG-E8) or/and Colivelin TFA after exposing for 4 h with oxygen glucose deprivation (OGD). Cell viability and apoptosis were assessed by MTT assay and Flow cytometry. RT-qPCR and Western blot assays were applied to examine the expression levels of MFG-E8, apoptosis-related proteins and M1/M2 polarization markers.Results: Our results demonstrated that OGD significantly inhibited microglial viability and facilitated apoptosis. In addition, we found that OGD downregulated MFG-E8 expression, and MFG-E8 inhibited OGD-induced microglial apoptosis and promoted microglial M2 polarization. In terms of mechanism, we proved that MFG-E8 regulated OGD-induced microglial M1/M2 polarization by inhibiting p-STAT3 and SOCS3 expressions, which was reversed by STAT3 activator (Colivelin TFA). Finally, we verified MFG-E8 alleviated OGD-induced neuronal cell apoptosis by M2 polarization of BV-2 cells.Conclusions: We demonstrated that MFG-E8 reduced neuronal cell apoptosis by enhancing activation of microglia via STAT3 signaling. Therefore, we suggested that MFG-E8 might provide a novel mechanism for ischemic stroke.
Collapse
Affiliation(s)
- Ying-Ying Fang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, P.R. China
| | - Jing-Hui Zhang
- Department of Rehabilitation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, P.R. China.,Guangdong Association of Rehabilitation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, P.R. China
| |
Collapse
|
65
|
Rabenstein M, Vay SU, Blaschke S, Walter HL, Ladwig A, Fink GR, Rueger MA, Schroeter M. Crosstalk between stressed brain cells: direct and indirect effects of ischemia and aglycemia on microglia. J Neuroinflammation 2020; 17:33. [PMID: 31980036 PMCID: PMC6982395 DOI: 10.1186/s12974-020-1697-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In cerebral ischemia, microglia have a dichotomous role in keeping the balance between pro- and anti-inflammatory mediators to avoid deleterious chronic inflammation and to leverage repair processes. METHODS We examined functional and inflammatory markers in primary rat microglia in vitro after oxygen-glucose deprivation (OGD) or glucose deprivation (aglycemia). We then investigated the preconditioning effect of OGD or aglycemia upon a subsequent strong inflammatory stimulus, here lipopolysaccharides (LPS). Moreover, an "in vitro brain model" of neurons and glia, differentiated from primary rat neural stem cells, was exposed to OGD or aglycemia. Conditioned medium (CM) of this neuronal/glial co-culture was then used to condition microglia, followed by LPS as a "second hit." RESULTS OGD or aglycemia at sublethal doses did not significantly affect microglia function, including the expression of inflammatory markers. However, preconditioning with either OGD or aglycemia led to a decreased pro-inflammatory response to a subsequent stimulus with LPS. Interestingly, the anti-inflammatory markers IGF-1 and IL-10 were additionally reduced after such preconditioning, while expression of CD206 remained unaffected. Treatment with CM from the neuronal/glial co-culture alone did not affect the expression of inflammatory markers in microglia. In contrast, treatment with CM increased the expression of both pro- and anti-inflammatory markers in microglia upon a second hit with LPS. Interestingly, this effect could be attenuated in microglia treated with CM from neuronal/glia co-cultures preconditioned with OGD or aglycemia. CONCLUSIONS Data suggest specific and distinct microglia signatures in response to metabolic stress. While metabolic stress directly and indirectly applied to microglia did not mitigate their subsequent response to inflammation, preconditioning with metabolic stress factors such as OGD and aglycemia elicited a decreased inflammatory response to a subsequent inflammation stimulus.
Collapse
Affiliation(s)
- Monika Rabenstein
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
| | - Sabine Ulrike Vay
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
| | - Stefan Blaschke
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
- Research Centre Juelich, Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Helene Luise Walter
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
| | - Anne Ladwig
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
| | - Gereon Rudolf Fink
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
- Research Centre Juelich, Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Maria Adele Rueger
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany
- Research Centre Juelich, Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich, Germany
| | - Michael Schroeter
- Department of Neurology, Faculty of Medicine and University Hospital, University of Cologne, Kerpener Strasse 62, 50924, Cologne, Germany.
- Research Centre Juelich, Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Juelich, Germany.
| |
Collapse
|
66
|
Wang Y, Zhang JH, Sheng J, Shao A. Immunoreactive Cells After Cerebral Ischemia. Front Immunol 2019; 10:2781. [PMID: 31849964 PMCID: PMC6902047 DOI: 10.3389/fimmu.2019.02781] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
The immune system is rapidly activated after ischemic stroke. As immune cells migrate and infiltrate across the blood-brain barrier into the ischemic region, a cascade of cellular and molecular biological reactions occur, involving migrated immune cells, resident glial cells, and the vascular endothelium. These events regulate infarction evolution and thus influence the outcome of ischemic stroke. Most immune cells exert dual effects on cerebral ischemia, and some crucial cells may become central targets in ischemic stroke treatment and rehabilitation.
Collapse
Affiliation(s)
- Yijie Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
67
|
Quarta A, Le Blon D, D'aes T, Pieters Z, Hamzei Taj S, Miró-Mur F, Luyckx E, Van Breedam E, Daans J, Goossens H, Dewilde S, Hens N, Pasque V, Planas AM, Hoehn M, Berneman Z, Ponsaerts P. Murine iPSC-derived microglia and macrophage cell culture models recapitulate distinct phenotypical and functional properties of classical and alternative neuro-immune polarisation. Brain Behav Immun 2019; 82:406-421. [PMID: 31525508 DOI: 10.1016/j.bbi.2019.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/24/2022] Open
Abstract
The establishment and validation of reliable induced pluripotent stem cell (iPSC)-derived in vitro models to study microglia and monocyte/macrophage immune function holds great potential for fundamental and translational neuro-immunology research. In this study, we first demonstrate that ramified CX3CR1+ iPSC-microglia (cultured within a neural environment) and round-shaped CX3CR1- iPSC-macrophages can easily be differentiated from newly established murine CX3CR1eGFP/+CCR2RFP/+ iPSC lines. Furthermore, we show that obtained murine iPSC-microglia and iPSC-macrophages are distinct cell populations, even though iPSC-macrophages may upregulate CX3CR1 expression when cultured within a neural environment. Next, we characterized the phenotypical and functional properties of murine iPSC-microglia and iPSC-macrophages following classical and alternative immune polarisation. While iPSC-macrophages could easily be triggered to adopt a classically-activated or alternatively-activated phenotype following, respectively, lipopolysaccharide + interferon γ or interleukin 13 (IL13) stimulation, iPSC-microglia and iPSC-macrophages cultured within a neural environment displayed a more moderate activation profile as characterised by the absence of MHCII expression upon classical immune polarisation and the absence of Ym1 expression upon alternative immune polarisation. Finally, extending our preceding in vivo studies, this striking phenotypical divergence was also observed for resident microglia and infiltrating monocytes within highly inflammatory cortical lesions in CX3CR1eGFP/+CCR2RFP/+ mice subjected to middle cerebral arterial occlusion (MCAO) stroke and following IL13-mediated therapeutic intervention thereon. In conclusion, our study demonstrates that the applied murine iPSC-microglia and iPSC-macrophage culture models are able to recapitulate in vivo microglia and monocyte/macrophage ontogeny and corresponding phenotypical/functional properties upon classical and alternative immune polarisation, and therefore represent a valuable in vitro platform to further study and modulate microglia and (infiltrating) monocyte immune responses under neuro-inflammatory conditions within a neural environment.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Tine D'aes
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zoë Pieters
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Belgium; Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Belgium
| | - Somayyeh Hamzei Taj
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Francesc Miró-Mur
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Evi Luyckx
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Protein Chemistry, Proteomics and Epigenetic Signaling, University of Antwerp, Antwerp, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Sylvia Dewilde
- Protein Chemistry, Proteomics and Epigenetic Signaling, University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Belgium; Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Belgium
| | - Vincent Pasque
- Department of Development and Regeneration, Leuven Stem Cell Institute, Leuven Cancer Institute, KU Leuven - University of Leuven, Belgium
| | - Anna M Planas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Zwi Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
68
|
Jayaraj RL, Azimullah S, Beiram R, Jalal FY, Rosenberg GA. Neuroinflammation: friend and foe for ischemic stroke. J Neuroinflammation 2019; 16:142. [PMID: 31291966 PMCID: PMC6617684 DOI: 10.1186/s12974-019-1516-2] [Citation(s) in RCA: 901] [Impact Index Per Article: 150.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/10/2019] [Indexed: 12/13/2022] Open
Abstract
Stroke, the third leading cause of death and disability worldwide, is undergoing a change in perspective with the emergence of new ideas on neurodegeneration. The concept that stroke is a disorder solely of blood vessels has been expanded to include the effects of a detrimental interaction between glia, neurons, vascular cells, and matrix components, which is collectively referred to as the neurovascular unit. Following the acute stroke, the majority of which are ischemic, there is secondary neuroinflammation that both promotes further injury, resulting in cell death, but conversely plays a beneficial role, by promoting recovery. The proinflammatory signals from immune mediators rapidly activate resident cells and influence infiltration of a wide range of inflammatory cells (neutrophils, monocytes/macrophages, different subtypes of T cells, and other inflammatory cells) into the ischemic region exacerbating brain damage. In this review, we discuss how neuroinflammation has both beneficial as well as detrimental roles and recent therapeutic strategies to combat pathological responses. Here, we also focus on time-dependent entry of immune cells to the ischemic area and the impact of other pathological mediators, including oxidative stress, excitotoxicity, matrix metalloproteinases (MMPs), high-mobility group box 1 (HMGB1), arachidonic acid metabolites, mitogen-activated protein kinase (MAPK), and post-translational modifications that could potentially perpetuate ischemic brain damage after the acute injury. Understanding the time-dependent role of inflammatory factors could help in developing new diagnostic, prognostic, and therapeutic neuroprotective strategies for post-stroke inflammation.
Collapse
Affiliation(s)
- Richard L. Jayaraj
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Sheikh Azimullah
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Rami Beiram
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Fakhreya Y. Jalal
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, UAE
| | - Gary A. Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131 USA
| |
Collapse
|
69
|
Microglia, Monocytes, and the Recurrence of Anxiety in Stress-Sensitized Mice. Biol Psychiatry 2019; 85:e67-e68. [PMID: 30857640 DOI: 10.1016/j.biopsych.2018.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/30/2019] [Indexed: 11/22/2022]
|
70
|
Collmann FM, Pijnenburg R, Hamzei-Taj S, Minassian A, Folz-Donahue K, Kukat C, Aswendt M, Hoehn M. Individual in vivo Profiles of Microglia Polarization After Stroke, Represented by the Genes iNOS and Ym1. Front Immunol 2019; 10:1236. [PMID: 31214190 PMCID: PMC6558167 DOI: 10.3389/fimmu.2019.01236] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 05/15/2019] [Indexed: 12/26/2022] Open
Abstract
Microglia are the brain-innate immune cells which actively surveil their environment and mediate multiple aspects of neuroinflammation, due to their ability to acquire diverse activation states and phenotypes. Simplified, M1-like microglia are defined as pro-inflammatory cells, while the alternative M2-like cells promote neuroprotection. The modulation of microglia polarization is an appealing neurotherapeutic strategy for stroke and other brain lesions, as well as neurodegenerative diseases. However, the activation profile and change of phenotype during experimental stroke is not well understood. With a combined magnetic resonance imaging (MRI) and optical imaging approach and genetic targeting of two key genes of the M1- and M2-like phenotypes, iNOS and Ym1, we were able to monitor in vivo the dynamic adaption of the microglia phenotype in response to experimental stroke.
Collapse
Affiliation(s)
- Franziska M Collmann
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Rory Pijnenburg
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Somayyeh Hamzei-Taj
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Anuka Minassian
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Kat Folz-Donahue
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Christian Kukat
- FACS & Imaging Core Facility, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Markus Aswendt
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany.,Department of Neurology, University Hospital Cologne, Cologne, Germany
| | - Mathias Hoehn
- In-vivo-NMR, Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany.,Radiology Department, Leiden University Medical Center, Leiden, Netherlands.,PERCUROS, Enschede, Netherlands
| |
Collapse
|
71
|
Haenseler W, Rajendran L. Concise Review: Modeling Neurodegenerative Diseases with Human Pluripotent Stem Cell-Derived Microglia. Stem Cells 2019; 37:724-730. [PMID: 30801863 PMCID: PMC6849818 DOI: 10.1002/stem.2995] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/25/2019] [Accepted: 02/03/2019] [Indexed: 12/11/2022]
Abstract
Inflammation of the brain and the consequential immunological responses play pivotal roles in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia (FTD). Microglia, the resident macrophage cells of the brain, have also emerged as key players in neuroinflammation. As primary human microglia from living subjects are normally not accessible to researchers, there is a pressing need for an alternative source of authentic human microglia which allows modeling of neurodegeneration in vitro. Several protocols for induced pluripotent stem cell (iPSC)‐derived microglia have recently been developed and provide unlimited access to patient‐derived material. In this present study, we give an overview of iPSC‐derived microglia models in monoculture and coculture systems, their advantages and limitations, and how they have already been used for disease phenotyping. Furthermore, we outline some of the gene engineering tools to generate isogenic controls, the creation of gene knockout iPSC lines, as well as covering reporter cell lines, which could help to elucidate complex cell interaction mechanisms in the microglia/neuron coculture system, for example, microglia‐induced synapse loss. Finally, we deliberate on how said cocultures could aid in personalized drug screening to identify patient‐specific therapies against neurodegeneration. stem cells2019;37:724–730
Collapse
Affiliation(s)
- Walther Haenseler
- Systems and Cell Biology of Neurodegeneration, IREM, University of Zurich, Schlieren, Switzerland
| | - Lawrence Rajendran
- Systems and Cell Biology of Neurodegeneration, IREM, University of Zurich, Schlieren, Switzerland.,UK-Dementia Research Institute (UK-DRI), Maurice Wohl Basic & Clinical Neuroscience Institute, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
72
|
Han J, Zhu K, Zhang X, Harris RA. Enforced microglial depletion and repopulation as a promising strategy for the treatment of neurological disorders. Glia 2019; 67:217-231. [PMID: 30378163 PMCID: PMC6635749 DOI: 10.1002/glia.23529] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/17/2018] [Accepted: 08/22/2018] [Indexed: 01/18/2023]
Abstract
Microglia are prominent immune cells in the central nervous system (CNS) and are critical players in both neurological development and homeostasis, and in neurological diseases when dysfunctional. Our previous understanding of the phenotypes and functions of microglia has been greatly extended by a dearth of recent investigations. Distinct genetically defined subsets of microglia are now recognized to perform their own independent functions in specific conditions. The molecular profiling of single microglial cells indicates extensively heterogeneous reactions in different neurological disorders, resulting in multiple potentials for crosstalk with other kinds of CNS cells such as astrocytes and neurons. In settings of neurological diseases it could thus be prudent to establish effective cell-based therapies by targeting entire microglial networks. Notably, activated microglial depletion through genetic targeting or pharmacological therapies within a suitable time window can stimulate replenishment of the CNS niche with new microglia. Additionally, enforced repopulation through provision of replacement cells also represents a potential means of exchanging dysfunctional with functional microglia. In each setting the newly repopulated microglia might have the potential to resolve ongoing neuroinflammation. In this review, we aim to summarize the most recent knowledge of microglia and to highlight microglial depletion and subsequent repopulation as a promising cell replacement therapy. Although glial cell replacement therapy is still in its infancy and future translational studies are still required, the approach is scientifically sound and provides new optimism for managing the neurotoxicity and neuroinflammation induced by activated microglia.
Collapse
Affiliation(s)
- Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Xing‐Mei Zhang
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical NeuroscienceKarolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital at SolnaStockholmSweden
| |
Collapse
|
73
|
Rajan WD, Wojtas B, Gielniewski B, Gieryng A, Zawadzka M, Kaminska B. Dissecting functional phenotypes of microglia and macrophages in the rat brain after transient cerebral ischemia. Glia 2018; 67:232-245. [PMID: 30485549 DOI: 10.1002/glia.23536] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 01/30/2023]
Abstract
Ischemic brain injury causes local inflammation, which involves activation of resident microglia, leukocyte, and monocyte infiltration. Involvement of peripheral immune cells in ischemia-induced damage and repair is debatable. Using flow cytometry, gene expression profiling, and immunocytochemistry, we show that microglia predominate in the ischemic brain and express inflammation mediators at Day 1 after transient middle cerebral artery occlusion (MCAo) in rats. At Day 3, both resident microglia and bone marrow (BM)-derived macrophages are detected in the ischemic hemispheres and display unique transcriptomic profiles. Functional groups enriched in BM-macrophages are indicative of the pro-regenerative, immunosuppressive phenotype. Transient depletion of peripheral macrophages with clodronate-filled liposomes reduced the number of Arg1+ Iba1+ expressing cells in the ischemic brain. The analysis of microglia and macrophage signature genes shows that each cell type maintains the expression of their identity genes, even if gene expression is modified in a response to environmental clues. At Day 7, infiltrating BM-macrophages exhibit the reduced expression of Arg1, the elevated expression of iNos and many inflammatory genes, as shown by RNA sequencing. This is consistent with their switch toward a pro-inflammatory phenotype. We propose that BM-macrophages recruited to the injured brain early after ischemia could contribute to functional recovery after stroke, but they switch toward a pro-inflammatory phenotype in the ischemic parenchyma. Our results point to the detrimental role of microglia in an ischemic brain and the primarily pro-regenerative role of infiltrating BM-macrophages.
Collapse
Affiliation(s)
- Wenson D Rajan
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartlomiej Gielniewski
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Anna Gieryng
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Zawadzka
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, The Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
74
|
Store-operated calcium entry in thrombosis and thrombo-inflammation. Cell Calcium 2018; 77:39-48. [PMID: 30530092 DOI: 10.1016/j.ceca.2018.11.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/31/2018] [Accepted: 11/14/2018] [Indexed: 01/03/2023]
Abstract
Cytosolic free calcium (Ca2+) is a second messenger regulating a wide variety of functions in blood cells, including adhesion, activation, proliferation and migration. Store-operated Ca2+ entry (SOCE), triggered by depletion of Ca2+ from the endoplasmic reticulum, provides a main mechanism of regulated Ca2+ influx in blood cells. SOCE is mediated and regulated by isoforms of the ion channel proteins ORAI and TRP, and the transmembrane Ca2+ sensors stromal interaction molecules (STIMs), respectively. This report provides an overview of the (patho)physiological importance of SOCE in blood cells implicated in thrombosis and thrombo-inflammation, i.e. platelets and immune cells. We also discuss the physiological consequences of dysregulated SOCE in platelets and immune cells and the potential of SOCE inhibition as a therapeutic option to prevent or treat arterial thrombosis as well as thrombo-inflammatory disease states such as ischemic stroke.
Collapse
|
75
|
Wimmer I, Zrzavy T, Lassmann H. Neuroinflammatory responses in experimental and human stroke lesions. J Neuroimmunol 2018; 323:10-18. [DOI: 10.1016/j.jneuroim.2018.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 02/07/2023]
|
76
|
Fouda AY, Xu Z, Shosha E, Lemtalsi T, Chen J, Toque HA, Tritz R, Cui X, Stansfield BK, Huo Y, Rodriguez PC, Smith SB, Caldwell RW, Narayanan SP, Caldwell RB. Arginase 1 promotes retinal neurovascular protection from ischemia through suppression of macrophage inflammatory responses. Cell Death Dis 2018; 9:1001. [PMID: 30254218 PMCID: PMC6156564 DOI: 10.1038/s41419-018-1051-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/24/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
The lack of effective therapies to limit neurovascular injury in ischemic retinopathy is a major clinical problem. This study aimed to examine the role of ureohydrolase enzyme, arginase 1 (A1), in retinal ischemia-reperfusion (IR) injury. A1 competes with nitric oxide synthase (NOS) for their common substrate l-arginine. A1-mediated l-arginine depletion reduces nitric oxide (NO) formation by NOS leading to vascular dysfunction when endothelial NOS is involved but prevents inflammatory injury when inducible NOS is involved. Studies were performed using wild-type (WT) mice, global A1+/− knockout (KO), endothelial-specific A1 KO, and myeloid-specific A1 KO mice subjected to retinal IR injury. Global as well as myeloid-specific A1 KO mice showed worsened IR-induced neuronal loss and retinal thinning. Deletion of A1 in endothelial cells had no effect, while treatment with PEGylated (PEG) A1 improved neuronal survival in WT mice. In addition, A1+/− KO mice showed worsened vascular injury manifested by increased acellular capillaries. Western blotting analysis of retinal tissue showed increased inflammatory and necroptotic markers with A1 deletion. In vitro experiments showed that macrophages lacking A1 exhibit increased inflammatory response upon LPS stimulation. PEG-A1 treatment dampened this inflammatory response and decreased the LPS-induced metabolic reprogramming. Moreover, intravitreal injection of A1 KO macrophages or systemic macrophage depletion with clodronate liposomes increased neuronal loss after IR injury. These results demonstrate that A1 reduces IR injury-induced retinal neurovascular degeneration via dampening macrophage inflammatory responses. Increasing A1 offers a novel strategy for limiting neurovascular injury and promoting macrophage-mediated repair.
Collapse
Affiliation(s)
- Abdelrahman Y Fouda
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Zhimin Xu
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Esraa Shosha
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Tahira Lemtalsi
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Haroldo A Toque
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Rebekah Tritz
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Xuezhi Cui
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - Brian K Stansfield
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Yuqing Huo
- Vascular Biology Center, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA
| | | | - Sylvia B Smith
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA.,Department of Ophthalmology, Augusta University, Augusta, GA, USA
| | - R William Caldwell
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - S Priya Narayanan
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Vascular Biology Center, Augusta University, Augusta, GA, USA.,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA.,Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, USA
| | - Ruth B Caldwell
- Charlie Norwood VA Medical Center, Augusta, GA, USA. .,Vascular Biology Center, Augusta University, Augusta, GA, USA. .,James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA, USA. .,Department of Cell Biology and Anatomy, Augusta University, Augusta, GA, USA. .,Department of Ophthalmology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
77
|
Molecular Communication of a Dying Neuron in Stroke. Int J Mol Sci 2018; 19:ijms19092834. [PMID: 30235837 PMCID: PMC6164443 DOI: 10.3390/ijms19092834] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 02/06/2023] Open
Abstract
When a main artery of the brain occludes, a cellular response involving multiple cell types follows. Cells directly affected by the lack of glucose and oxygen in the neuronal core die by necrosis. In the periphery surrounding the ischemic core (the so-called penumbra) neurons, astrocytes, microglia, oligodendrocytes, pericytes, and endothelial cells react to detrimental factors such as excitotoxicity, oxidative stress, and inflammation in different ways. The fate of the neurons in this area is multifactorial, and communication between all the players is important for survival. This review focuses on the latest research relating to synaptic loss and the release of apoptotic bodies and other extracellular vesicles for cellular communication in stroke. We also point out possible treatment options related to increasing neuronal survival and regeneration in the penumbra.
Collapse
|
78
|
Amantea D, Greco R, Micieli G, Bagetta G. Paradigm Shift to Neuroimmunomodulation for Translational Neuroprotection in Stroke. Front Neurosci 2018; 12:241. [PMID: 29692708 PMCID: PMC5903066 DOI: 10.3389/fnins.2018.00241] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022] Open
Abstract
The treatment of acute ischemic stroke is still an unresolved clinical problem since the only approved therapeutic intervention relies on early blood flow restoration through pharmacological thrombolysis, mechanical thrombus removal, or a combination of both strategies. Due to their numerous complications and to the narrow time-window for the intervention, only a minority of stroke patients can actually benefit from revascularization procedures, highlighting the urgent need of identifying novel strategies to prevent the progression of an irreversible damage in the ischemic penumbra. During the past three decades, the attempts to target the pathways implicated in the ischemic cascade (e.g., excitotoxicity, calcium channels overactivation, reactive oxygen species (ROS) production) have failed in the clinical setting. Based on a better understanding of the pathobiological mechanisms and on a critical reappraisal of most failed trials, numerous findings from animal studies have demonstrated that targeting the immune system may represent a promising approach to achieve neuroprotection in stroke. In particular, given the dualistic role of distinct components of both the innate and adaptive arms of the immune system, a strategic intervention should be aimed at establishing the right equilibrium between inflammatory and reparative mechanisms, taking into consideration their spatio-temporal recruitment after the ischemic insult. Thus, the application of immunomodulatory drugs and their ability to ameliorate outcomes deserve validation in patients with acute ischemic stroke.
Collapse
Affiliation(s)
- Diana Amantea
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| | - Rosaria Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, IRCCS Mondino Foundation, Pavia, Italy
| | - Giuseppe Micieli
- Department of Emergency Neurology, IRCCS Mondino Foundation, Pavia, Italy
| | - Giacinto Bagetta
- Section of Preclinical and Translational Pharmacology, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| |
Collapse
|