1
|
Zhao B, Wang C, Sun M, Ma X, Zeng Q, Xi J, Zhou J, Pei X, Jia Y, Yue W. UC-MSCs based on biomimetic microniche exert excellent regulatory effects on acute brain inflammation through advantageous properties. Biomaterials 2025; 315:122945. [PMID: 39522143 DOI: 10.1016/j.biomaterials.2024.122945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 10/13/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
Neuroinflammation triggered by activated microglia leads to neuronal damage and, to a certain extent, neurodegeneration. Human umbilical cord mesenchymal stem cells (UC-MSCs) have good immunomodulatory and neuroprotective effects as well as therapeutic potential for neuroinflammation-related diseases. However, the complex microenvironment created by neuroinflammation poses a challenge to transplanted UC-MSCs. The emerging biomimetic microniche (BN)-based culture technology provides new opportunities to optimize the preparation of UC-MSCs; but the fundamental changes in the characteristics of UC-MSCs based on BN remain unclear, and more reliable preclinical data are needed to support their ability to regulate inflammation. Here, we systematically studied the cellular properties and inflammation regulatory capacity of UC-MSCs in conventional static planar culture (SP-UCMSCs) and suspension culture based on BN (BN-UCMSCs). In vitro, compared with SP-UCMSCs, BN-UCMSCs not only maintained the fundamental characteristics of MSCs, but also significantly enhanced cell proliferation, adhesion, and migration capabilities, etc; notably, the paracrine function and anti-inflammatory capacity of BN-UCMSCs were also enhanced. We further established a murine model of acute brain inflammation and demonstrated that the expression level of pro-inflammatory cytokines in hippocampal and cortical tissues of the BN-UCMSCs group was significantly decreased compared with that in the SP-UCMSCs group. Subsequent transcriptomic analysis of hippocampal and cortical tissues revealed that BN-UCMSCs had the advantage of significantly reducing the expression of pro-inflammatory cytokines through the TLR4-Myd88-NF-κB axis, which was further validated at the gene and protein levels. Taken together, these data strongly indicated that BN-UCMSCs exerts excellent regulatory effects on acute brain inflammation through advantageous properties.
Collapse
Affiliation(s)
- Bichun Zhao
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chao Wang
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Manqiang Sun
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiaocao Ma
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, Chinese PLA General Hospital, Beijing, 100853, China
| | - Quan Zeng
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Jiafei Xi
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Junnian Zhou
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yali Jia
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
2
|
Fu Z, Ganesana M, Hwang P, Tan X, Kinkaid MM, Sun YY, Bian E, Weybright A, Chen HR, Sol-Church K, Eyo UB, Pridans C, Quintana FJ, Robson SC, Kumar P, Venton BJ, Schaefer A, Kuan CY. Microglia modulate the cerebrovascular reactivity through ectonucleotidase CD39. Nat Commun 2025; 16:956. [PMID: 39843911 PMCID: PMC11754601 DOI: 10.1038/s41467-025-56093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
Microglia and the border-associated macrophages contribute to the modulation of cerebral blood flow, but the mechanisms have remained uncertain. Here, we show that microglia regulate the cerebral blood flow baseline and the responses to whisker stimulation or intra-cisternal magna injection of adenosine triphosphate, but not intra-cisternal magna injection of adenosine in mice model. Notably, microglia repopulation corrects these cerebral blood flow anomalies. The microglial-dependent regulation of cerebral blood flow requires the adenosine triphosphate-sensing P2RY12 receptor and ectonucleotidase CD39 that initiates the dephosphorylation of extracellular adenosine triphosphate into adenosine in both male and female mice. Pharmacological inhibition or CX3CR1-CreER-mediated deletion of CD39 mimics the cerebral blood flow anomalies in microglia-deficient mice and reduces the upsurges of extracellular adenosine following whisker stimulation. Together, these results suggest that the microglial CD39-initiated breakdown of extracellular adenosine triphosphate co-transmitter is an important step in neurovascular coupling and the regulation of cerebrovascular reactivity.
Collapse
Affiliation(s)
- Zhongxiao Fu
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | | | - Philip Hwang
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xiao Tan
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Melissa Marie Kinkaid
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yu-Yo Sun
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Emily Bian
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Aden Weybright
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Hong-Ru Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Katia Sol-Church
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Clare Pridans
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Pankaj Kumar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, USA
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - B Jill Venton
- Department of Chemistry, University of Virginia, Charlottesville, VA, USA
| | - Anne Schaefer
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- MPI Biology of Ageing, Cologne, Germany
| | - Chia-Yi Kuan
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
3
|
Nunes RR, Durán-Carabali LE, Ribeiro NH, Sirena DH, Tassinari ID, Netto CA, Paz AH, de Fraga LS. Impact of peripheral immune cells in experimental neonatal hypoxia-ischemia: A systematic review and meta-analysis. Int Immunopharmacol 2025; 145:113682. [PMID: 39637576 DOI: 10.1016/j.intimp.2024.113682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Infiltration of peripheral immune cells into the brain following neonatal hypoxia-ischemia (HI) contributes to increased neuroinflammation and brain injury. However, the specific roles of different immune cell types in neonatal brain injury remain poorly understood. Although existing evidence suggests a potential role for sexual dimorphism in HI outcomes, this aspect has been insufficiently investigated. In this systematic review and meta-analysis, we examined the brain infiltration of peripheral immune cells in rodents of both sexes following neonatal HI. A total of 25 studies were included. Our analysis revealed significant increases in the infiltration of various subtypes of leukocytes after HI, along with increased brain injury, cell death, and neuroinflammation, and reduced neuronal survival. Notably, males exhibited a greater degree of immune cell infiltration and more pronounced neuroinflammation compared to females. These findings suggest that infiltrating leukocytes contribute significantly to the pathophysiology of neonatal HI, with sexually dimorphic responses further influencing the outcomes. It is crucial that future research focuses on elucidating the specific roles of immune cell subtypes to better understand the mechanisms underlying brain damage after HI and identify novel therapeutic targets. Moreover, the observed sex differences highlight the need to consider sex as a key factor when developing strategies for the treatment of neonatal HI.
Collapse
Affiliation(s)
- Ricardo Ribeiro Nunes
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Luz Elena Durán-Carabali
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Ciências Fisiológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Nícolas Heller Ribeiro
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Dienifer Hermann Sirena
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Helena Paz
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Ciências Morfológicas, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil; Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil.
| |
Collapse
|
4
|
Cai Q, Shen L, Zhang X, Zhang Z, Wang T. The IRE1-XBP1 Axis Regulates NLRP3 Inflammasome-Mediated Microglia Activation in Hypoxic Ischemic Encephalopathy. Crit Rev Immunol 2025; 45:55-64. [PMID: 39612277 DOI: 10.1615/critrevimmunol.2024053554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a perinatal injury caused by cerebral hypoxia and reduced blood perfusion. Microglia activation-induced neuroinflammatory injury is a leading cause of neuron loss and brain injury. Efficient treatment strategies are still required further investigation. Our study is aimed to investigate the role of IRE1-XBP1 inhibitor 4μ8С in HIE. Rat pups (7 d) were used to establish HIE model using unilateral carotid artery ligation and hypoxia. A series of experiments including Western blot, Morris water maze test, TTC staining, RT-qPCR, TUNEL staining, and immunofluorescence staining were operated to evaluate the role of 4μ8С in HIE. 4μ8С treatment effectively reduced phosphorylated IRElα and XBP1 protein levels. 4μ8С treatment improves cognition and learning abilities of HIE rats. 4μ8С treatment alleviated brain infarction and cell apoptosis in HIE rats. 4μ8С treatment inhibited NLRP3 inflammasome activation-mediated microglia activation and inflammatory response. In conclusion, 4μ8С suppressed microglia and NLRP3 inflammasome activation by inactivating IRE1/XBP1 axis during HIE development, which revealed IRE1α inhibition as a novel mechanism for neuron protection.
Collapse
Affiliation(s)
- Qun Cai
- Department of Pediatric, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Liyuan Shen
- Department of Pediatric, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Xiaoqun Zhang
- Department of Pediatric, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Zhijun Zhang
- Department of Human Anatomy, School of Medicine, Nantong University, Nantong 226001, China
| | - Ting Wang
- Affiliated Hospital of Nantong University
| |
Collapse
|
5
|
Chen W, Zhang L, Zhong G, Liu S, Sun Y, Zhang J, Liu Z, Wang L. Regulation of microglia inflammation and oligodendrocyte demyelination by Engeletin via the TLR4/RRP9/NF-κB pathway after spinal cord injury. Pharmacol Res 2024; 209:107448. [PMID: 39395773 DOI: 10.1016/j.phrs.2024.107448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024]
Abstract
Microglia polarization is crucial for neuroinflammatory response after spinal cord injury (SCI). Small molecule compounds and hub genes play an important role in regulating microglia polarization, reducing neuroinflammatory response and oligodendrocyte demyelination after SCI. In this study, suitable data sets were used to screen hub genes, and Western blot and Immunofluorescence (IF) experiments were used to confirm the expressions of proteins related to SDAD1, RRP9 and NF-κB pathways under LPS/SCI conditions. Engeletin (ENG) reduced microglia polarization and inflammation in vivo and in vitro via the SDAD1, RRP9 or NF-κB signaling pathways. In addition, ENG binds to the membrane receptor Toll-like receptor 4 (TLR4) through small molecule-protein docking. COIP experiment and protein-protein docking revealed protein-protein interaction (PPI) between RRP9 and SDAD1. By gene knock-down (KD) / overexpression (OE) and Western blot experiments, RRP9 and SDAD1 can regulate inflammatory response through NF-κB signaling and ribosome biogenesis pathway. Western blot analysis showed that CU increased the expression of SDAD1, RRP9 and NF-κB pathway related proteins through TLR1/2, while C34 decreased the expression of SDAD1 and RRP9 proteins through TLR4. These results suggest that ENG can reduce inflammation through TLR4/RRP9(SDAD1)/NF-κB signaling pathway. In addition, we demonstrated that oligodendrocyte apoptosis and demyelination could be influenced by the regulation of microglia and tissue inflammation. In conclusion, this study found the gene Rrp9/Sdad1 and the small molecule compound ENG, which control the inflammatory response of microglia, and further explored the related mechanism of oligodendrocyte demyelination, which has important theoretical significance.
Collapse
Affiliation(s)
- Wang Chen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Leshu Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Guangdi Zhong
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Shuang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Yuxuan Sun
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Jiayun Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China; Harbin Medical University, Nangang District, Harbin, Heilongjiang, China
| | - Zehan Liu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China.
| | - Lichun Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, Heilongjiang, China.
| |
Collapse
|
6
|
He L, Yin R, Hang W, Han J, Chen J, Wen B, Chen L. Oxygen Glucose Deprivation-Induced Lactylation of H3K9 Contributes to M1 Polarization and Inflammation of Microglia Through TNF Pathway. Biomedicines 2024; 12:2371. [PMID: 39457683 PMCID: PMC11504212 DOI: 10.3390/biomedicines12102371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/29/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Hypoxia-induced M1 polarization of microglia and resultant inflammation take part in the damage caused by hypoxic-ischemic encephalopathy (HIE). Histone lactylation, a novel epigenetic modification where lactate is added to lysine residues, may play a role in HIE pathogenesis. This study investigates the role of histone lactylation in hypoxia-induced M1 microglial polarization and inflammation, aiming to provide insights for HIE treatment. METHODS In this study, we assessed the effects of hypoxia on microglial polarization using both an HIE animal model and an oxygen-glucose deprivation cell model. Histone lactylation at various lysine residues was detected by Western blotting. Microglial polarization and inflammatory cytokines were analyzed by immunofluorescence, qPCR, and Western blotting. RNA sequencing, ChIP-qPCR, and siRNA were used to elucidate mechanisms of H3K9 lactylation. RESULTS H3K9 lactylation increased due to cytoplasmic lactate during M1 polarization. Inhibiting P300 or reducing lactate dehydrogenase A expression decreased H3K9 lactylation, suppressing M1 polarization. Transcriptomic analysis indicated that H3K9 lactylation regulated M1 polarization via the TNF signaling pathway. ChIP-qPCR confirmed H3K9 lactylation enrichment at the TNFα locus, promoting OGD-induced M1 polarization and inflammation. CONCLUSIONS H3K9 lactylation promotes M1 polarization and inflammation via the TNF pathway, identifying it as a potential therapeutic target for neonatal HIE.
Collapse
Affiliation(s)
- Lu He
- Division of Neonatology, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Rui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (R.Y.); (J.C.)
| | - Weijian Hang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Jinli Han
- Department of Pediatrics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China;
| | - Juan Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (R.Y.); (J.C.)
| | - Bin Wen
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Ling Chen
- Division of Neonatology, Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
- Department of Pediatrics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China;
| |
Collapse
|
7
|
Jiang Y, Li R, Ban Y, Zhang W, Kong N, Tang J, Ma B, Shao Y, Jin R, Sun L, Yue H, Zhang H. EPO modified MSCs protects SH-SY5Y cells against ischemia/hypoxia-induced apoptosis via REST-dependent epigenetic remodeling. Sci Rep 2024; 14:23252. [PMID: 39370424 PMCID: PMC11456618 DOI: 10.1038/s41598-024-74261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a diffuse brain tissue injury caused by acute ischemia and hypoxia, and it is most commonly found in newborn infants but can also occur in adults. Mesenchymal stem cell (MSC) therapies have showed improved outcomes for treating HIE-induced neuronal defects. However, many key issues associated with poor cell viability and tolerance of grafted MSCs after HIE remain to be resolved. Genetic engineering could endow MSCs with more robust regenerative capacities. Our research, along with that of other scientists, has found that the expression of intracellular erythropoietin (EPO) in human umbilical cord MSCs (hUC-MSCs) increases proportionally with the duration of hypoxia exposure. Furthermore, we observed that EPO, when introduced into the EPO gene-modified hUC-MSCs, can be secreted into the extracellular space. However, the underlying mechanisms that support the neuroprotective effects of EPO-MSCs remain unclear. EPO-MSCs, hUC-MSCs, and NC-MSCs were identified by flow cytometry, osteogenic, and adipogenic differentiation assays. The oxygen-glucose deprivation (OGD)-induced SH-SY5Y cell-line was established, and five groups were set up: control, 24-h ischemia-hypoxia, co-cultured with hUC-MSCs, NC-MSCs, and EPO-MSCs after hypoxia. LEGENDplex™ multi-factor flow cytometry was used to detect the secretion of inflammatory factors in cell supernatants and cerebrospinal fluid. Chromosome-targeted excision and tagging (CUT&Tag) sequencing was applied to detect genomic H3K4me2 modifications, and conjoint analysis with transcriptome sequencing (RNA-seq) was performed. Lentiviral vector infection was used to construct SH-SY5Y cells with stable knockdown of RE1-silencing transcription factor (REST), and flow cytometry was used to detect alterations in apoptosis. Finally, the molecular mechanism underlying the neuroprotective and anti-apoptotic effects of EPO-MSCs was investigated using RNA sequencing, qRT-PCR, and western blot assays. Our results suggest that EPO-MSCs are genetically engineered to secrete significantly more EPO. EPO-MSCs treatment has anti-apoptotic properties and offers neuronal protection during ischemic-hypoxic injury. Furthermore, RNA-seq results suggest that multiple inflammation-related genes were down-regulated after EPO-MSCs treatment. Application of RNA-seq and CUT&Tag combined analysis found that the expressions of REST were significantly up-regulated. Lentiviral vector infection to construct REST knockdown SH-SY5Y failed to rescue apoptosis after hypoxia and co-culture with EPO-MSCs, and SETD2-mediated H3K36me3 protein level expression was reduced. EPO-MSCs may promote neuronal survival by affecting H3K4me2 and thus activating the expression of REST and TET3. EPO-MSCs also upregulated the modification level of SETD2-mediated H3K36me3 and regulated the expression of inflammation-related genes such as PLCG2, as well as apoptosis genes BCL2A1. To investigate the neuroprotective effects of EPO-modified hUC-MSCs and the underlying epigenetic regulatory mechanisms, this study aims to provide a theoretical foundation for the potential application of EPO gene-modified hUC-MSCs in the treatment of HIE.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ruibo Li
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Yueyao Ban
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Wenjin Zhang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ning Kong
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Jixiang Tang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Baodong Ma
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Yiming Shao
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Ranran Jin
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China
| | - Lei Sun
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China.
| | - Han Yue
- Stem Cell Research Center, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.
| | - Hui Zhang
- Department of Neurosurgery, Zhengzhou Central Hospital Affiliated Zhengzhou University, Zhengzhou, 450000, China.
| |
Collapse
|
8
|
Zhao Y, Song C, Wang H, Gai C, Li T, Cheng Y, Liu J, Song Y, Luo Q, Gu B, Liu W, Chai L, Liu D, Wang Z. Polydopamine-Cloaked Nanoarchitectonics of Prussian Blue Nanoparticles Promote Functional Recovery in Neonatal and Adult Ischemic Stroke Models. Biomater Res 2024; 28:0079. [PMID: 39296854 PMCID: PMC11409202 DOI: 10.34133/bmr.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 09/21/2024] Open
Abstract
Ischemic stroke is a devastating disease and one of the leading causes of mortality worldwide. Overproduction of reactive oxygen species and inflammatory response contribute to secondary damage following ischemic insult. Nanozymes with robust anti-oxidative stress properties possess therapeutic possibility for ischemic insult. However, insufficiency of nanozyme accumulation in the neuronal mitochondria hindered their application. Herein, we constructed polydopamine-coated Prussian blue nanoparticles (PB@PDA NPs) to realize the targeting neuronal mitochondria for ischemic stroke, with the properties of antioxidant and anti-inflammation. After administration, much higher accumulation of PB@PDA NPs in the brain was observed compared to that in the PB NP group. Moreover, PB@PDA NPs effectively attenuated brain infarct than that of PB NPs in neonatal mice following hypoxia-ischemia (HI) insult. PB@PDA NPs mainly colocated with neuronal mitochondria in vivo and in vitro. Apart from attenuating oxidative stress, PB@PDA NPs also suppressed neuronal apoptosis and counteracted inflammation, which effectively promote a short- and long-term functional recovery in HI mice. Further, the therapeutic efficacy of PB@PDA NPs was also found in adult ischemic mice via tail vein injection. Collectively, these findings illustrate that PB@PDA NPs via system injection accumulate in neuronal mitochondria and are beneficial for ischemic stroke.
Collapse
Affiliation(s)
- Yijing Zhao
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Cong Song
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Haijun Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Chengcheng Gai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Tingting Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yahong Cheng
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Junjie Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, Shandong 250117, P.R. China
| | - Yan Song
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qian Luo
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bing Gu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Weiyang Liu
- Jinan Xicheng Experimental High School, Dezhou Road1999, Jinan, Shandong, P.R. China
| | - Liwei Chai
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhen Wang
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
9
|
Pan J, Wang K, Qu J, Chen D, Chen A, You Y, Tang J, Zhang H. Activated tissue-resident macrophages contribute to hair cell insults in noise-induced hearing loss in mice. Commun Biol 2024; 7:1078. [PMID: 39223249 PMCID: PMC11368919 DOI: 10.1038/s42003-024-06768-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophages serve as the primary immune cell population and assume a pivotal role in the immune response within the damaged cochleae. Yet, the origin and role of macrophages in response to noise exposure remain controversial. Here, we take advantage of Ccr2RFP/+ Cx3cr1GFP/+ dual-reporter mice to identify the infiltrated and tissue-resident macrophages. After noise exposure, we reveal that activated resident macrophages change in morphology, increase in abundance, and migrate to the region of hair cells, leading to the loss of outer hair cells and the damage of ribbon synapses. Meanwhile, peripheral monocytes are not implicated in the noise-induced hair cell insults. These noise-induced activities of macrophages are abolished by inhibiting TLR4 signaling, resulting in alleviated insults of hair cells and partial recovery of hearing. Our findings indicate cochlear resident macrophages are pro-inflammatory and detrimental players in acoustic trauma and introduce a potential therapeutic target in noise-induced hearing loss.
Collapse
Affiliation(s)
- Jing Pan
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Kaiye Wang
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jiaxi Qu
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Dongxiu Chen
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Anning Chen
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Yunyou You
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China
| | - Jie Tang
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
- Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, 510515, China.
- Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Southern Medical University, Guangzhou, 510515, China.
| | - Hongzheng Zhang
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Ear Research Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
10
|
Dhillon SK, Gressens P, Barks J, Gunn AJ. Uncovering the Role of Inflammation with Asphyxia in the Newborn. Clin Perinatol 2024; 51:551-564. [PMID: 39095095 DOI: 10.1016/j.clp.2024.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The etiology of perinatal brain injury is multifactorial, but exposure to perinatal hypoxiaischemia (HI) is a major underlying factor. This review discusses the role of exposure to infection/inflammation in the evolution of HI brain injury, changes in immune responsiveness to subsequent inflammatory challenges after HI and modulation of neural outcomes with interaction between perinatal HI and inflammatory insults. The authors critically assess the clinical and preclinical evidence for the neuroprotective efficacy of therapeutic hypothermia and other anti-inflammatory treatments for inflammation-sensitized HI injury.
Collapse
Affiliation(s)
- Simerdeep K Dhillon
- Department of Physiology, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand
| | - Pierre Gressens
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France
| | - John Barks
- Department of Pediatrics and Communicable Diseases, The University of Michigan, 2018 MLB, Ann Arbor, MI 48109, USA
| | - Alistair J Gunn
- Department of Physiology, The University of Auckland, 85 Park Road, Grafton, Auckland 1023, New Zealand.
| |
Collapse
|
11
|
Xue L, Du R, Bi N, Xiao Q, Sun Y, Niu R, Tan Y, Chen L, Liu J, Wang T, Xiong L. Transplantation of human placental chorionic plate-derived mesenchymal stem cells for repair of neurological damage in neonatal hypoxic-ischemic encephalopathy. Neural Regen Res 2024; 19:2027-2035. [PMID: 38227532 DOI: 10.4103/1673-5374.390952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 09/18/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00035/figure1/v/2024-01-16T170235Z/r/image-tiff Neonatal hypoxic-ischemic encephalopathy is often associated with permanent cerebral palsy, neurosensory impairments, and cognitive deficits, and there is no effective treatment for complications related to hypoxic-ischemic encephalopathy. The therapeutic potential of human placental chorionic plate-derived mesenchymal stem cells for various diseases has been explored. However, the potential use of human placental chorionic plate-derived mesenchymal stem cells for the treatment of neonatal hypoxic-ischemic encephalopathy has not yet been investigated. In this study, we injected human placental chorionic plate-derived mesenchymal stem cells into the lateral ventricle of a neonatal hypoxic-ischemic encephalopathy rat model and observed significant improvements in both cognitive and motor function. Protein chip analysis showed that interleukin-3 expression was significantly elevated in neonatal hypoxic-ischemic encephalopathy model rats. Following transplantation of human placental chorionic plate-derived mesenchymal stem cells, interleukin-3 expression was downregulated. To further investigate the role of interleukin-3 in neonatal hypoxic-ischemic encephalopathy, we established an in vitro SH-SY5Y cell model of hypoxic-ischemic injury through oxygen-glucose deprivation and silenced interleukin-3 expression using small interfering RNA. We found that the activity and proliferation of SH-SY5Y cells subjected to oxygen-glucose deprivation were further suppressed by interleukin-3 knockdown. Furthermore, interleukin-3 knockout exacerbated neuronal damage and cognitive and motor function impairment in rat models of hypoxic-ischemic encephalopathy. The findings suggest that transplantation of hpcMSCs ameliorated behavioral impairments in a rat model of hypoxic-ischemic encephalopathy, and this effect was mediated by interleukin-3-dependent neurological function.
Collapse
Affiliation(s)
- Lulu Xue
- Transformation Research Laboratory, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruolan Du
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ning Bi
- Department of Animal Zoology, Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Qiuxia Xiao
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yifei Sun
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruize Niu
- Department of Animal Zoology, Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Yaxin Tan
- Department of Pediatrics, the People's Liberation Army Rocket Force Characteristic Medical Center, Beijing, China
| | - Li Chen
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jia Liu
- Department of Animal Zoology, Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Tinghua Wang
- Transformation Research Laboratory, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, Sichuan Province, China
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Animal Zoology, Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan Province, China
| | - Liulin Xiong
- Transformation Research Laboratory, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
12
|
Zhang Z, Mao Y, Huang S, Xu R, Huang Y, Li S, Sun Y, Gu X, Ma Z. Microglia Promote Inhibitory Synapse Phagocytosis in the Spinal Cord Dorsal Horn and Modulate Pain-Like Behaviors in a Murine Cancer-Induced Bone Pain Model. Anesth Analg 2024; 139:411-419. [PMID: 38241681 DOI: 10.1213/ane.0000000000006824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
BACKGROUND The microglial activation has been implicated in cancer-induced bone pain. Recent studies have revealed that microglia mediate synaptic pruning in the central nervous system, where the cluster of differentiation 47-signal regulatory protein α (CD47-SIRPα) axis creates a "don't eat me" signal and elicits an antiphagocytic effect to protect synapses against elimination. To date, the synaptic phagocytosis in microglia has never been investigated in the murine cancer-induced bone pain model. The present experiments sought to explore whether microglia phagocytize synapses in mice with bone cancer pain as well as the possible mechanisms. METHODS Male C3H/HeN mice were used to induce bone cancer pain. Minocycline and S-ketamine were injected into D14. The number of spontaneous flinches (NSF) and paw withdrawal mechanical thresholds (PWMT) were measured on D0, D4, D7, D10, D14, D21, and D28. Hematoxylin and eosin staining presented bone lesions. Western blotting examined the Gephyrin, CD47, and SIRPα expression. Flow cytometry evaluated the proportion of SIRPα + cells in the spine. Immunofluorescence and 3-dimensional reconstruction showed the Gephyrin puncta inside microglial lysosomes. RESULTS Mice embedded with tumor cells induced persistent spontaneous pain and mechanical hyperalgesia. Hematoxylin and eosin staining revealed bone destruction and tumor infiltration in marrow cavities. Microglia underwent a responsive and proliferative burst (t = -16.831, P < .001). Western blotting manifested lowered Gephyrin expression in the tumor group (D4, D7, D10, D14, D21, and D28: P < .001). Immunofluorescence and 3-dimensional reconstruction showed larger volumes of Gephyrin puncta inside microglial lysosomes (t = -23.273, P < .001; t = -27.997, P < .001). Treatment with minocycline or S-ketamine exhibited pain relief and antiphagocytic effects (t = -6.191, P < .001, t = -7.083, P < .001; t = -20.767, P < .001, t = -17.080, P < .001; t = 11.789, P < .001, t = 16.777, P < .001; t = 8.868, P < .001, t = 21.319, P < .001). Last but not least, the levels of CD47 and SIRPα proteins were downregulated (D10: P = .004, D14, D21, and D28: P < .001; D10, D14, D21, and D28: P < .001). Flow cytometry and immunofluorescence substantiated reduced microglial SIRPα (t = 11.311, P < .001; t = 12.189, P < .001). CONCLUSIONS Microglia-mediated GABAergic synapse pruning in the spinal cord dorsal horn in bone cancer pain mice, which might be associated with the declined CD47-SIRPα signal. Our research uncovered an innovative mechanism that highlighted microglia-mediated synaptic phagocytosis in a murine cancer-induced bone pain model.
Collapse
Affiliation(s)
- Zuoxia Zhang
- From the Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yanting Mao
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Simin Huang
- From the Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Shuming Li
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yu'e Sun
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaoping Gu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhengliang Ma
- From the Department of Anesthesiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
13
|
Farris T, González-Ochoa S, Mohammed M, Rajakaruna H, Tonello J, Kanagasabai T, Korolkova O, Shimamoto A, Ivanova A, Shanker A. Loss of Mitochondrial Tusc2/Fus1 Triggers a Brain Pro-Inflammatory Microenvironment and Early Spatial Memory Impairment. Int J Mol Sci 2024; 25:7406. [PMID: 39000512 PMCID: PMC11242373 DOI: 10.3390/ijms25137406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Brain pathological changes impair cognition early in disease etiology. There is an urgent need to understand aging-linked mechanisms of early memory loss to develop therapeutic strategies and prevent the development of cognitive impairment. Tusc2 is a mitochondrial-resident protein regulating Ca2+ fluxes to and from mitochondria impacting overall health. We previously reported that Tusc2-/- female mice develop chronic inflammation and age prematurely, causing age- and sex-dependent spatial memory deficits at 5 months old. Therefore, we investigated Tusc2-dependent mechanisms of memory impairment in 4-month-old mice, comparing changes in resident and brain-infiltrating immune cells. Interestingly, Tusc2-/- female mice demonstrated a pro-inflammatory increase in astrocytes, expression of IFN-γ in CD4+ T cells and Granzyme-B in CD8+T cells. We also found fewer FOXP3+ T-regulatory cells and Ly49G+ NK and Ly49G+ NKT cells in female Tusc2-/- brains, suggesting a dampened anti-inflammatory response. Moreover, Tusc2-/- hippocampi exhibited Tusc2- and sex-specific protein changes associated with brain plasticity, including mTOR activation, and Calbindin and CamKII dysregulation affecting intracellular Ca2+ dynamics. Overall, the data suggest that dysregulation of Ca2+-dependent processes and a heightened pro-inflammatory brain microenvironment in Tusc2-/- mice could underlie cognitive impairment. Thus, strategies to modulate the mitochondrial Tusc2- and Ca2+- signaling pathways in the brain should be explored to improve cognitive health.
Collapse
Affiliation(s)
- Tonie Farris
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Salvador González-Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Muna Mohammed
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Harshana Rajakaruna
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| | - Jane Tonello
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Thanigaivelan Kanagasabai
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Olga Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Akiko Shimamoto
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Alla Ivanova
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208, USA; (T.F.); (M.M.); (T.K.)
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience & Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA; (S.G.-O.); (J.T.); (O.K.); (A.S.)
- The Office for Research and Innovation, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
14
|
Boles J, Uriarte Huarte O, Tansey MG. Peripheral endotoxin exposure in mice activates crosstalk between phagocytes in the brain and periphery. RESEARCH SQUARE 2024:rs.3.rs-4478250. [PMID: 38883776 PMCID: PMC11177977 DOI: 10.21203/rs.3.rs-4478250/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Background Inflammation is a central process of many neurological diseases, and a growing number of studies suggest that non-brain-resident immune cells may contribute to this neuroinflammation. However, the unique contributions of specific immune cell subsets to neuroinflammation are presently unknown, and it is unclear how communication between brain-resident and non-resident immune cells underlies peripheral immune cell involvement in neuroinflammation. Methods In this study, we employed the well-established model of lipopolysaccharide (LPS)-induced neuroinflammation and captured brain-resident and non-resident immune cells from the brain and its vasculature by magnetically enriching cell suspensions from the non-perfused brain for CD45 + cells. Then, we identified immune subtype-specific neuroinflammatory processes using single-cell genomics and predicted the crosstalk between immune cell subtypes by analyzing the simultaneous expression of ligands and receptors. Results We observed a greater abundance of peripheral phagocytes associated with the brain in this model of neuroinflammation, and report that these professional phagocytes activated similar transcriptional profiles to microglia during LPS-induced neuroinflammation. And, we observed that the probable crosstalk between microglia and peripheral phagocytes was activated in this model while homotypic microglial communication was likely to be decreased. Conclusions Our novel findings reveal that microglia signaling to non-brain-resident peripheral phagocytes is preferentially triggered by peripheral inflammation, which is associated with brain infiltration of peripheral cells. Overall, our study supports the involvement of peripheral immune cells in neuroinflammation and suggests several possible molecular signaling pathways between microglia and peripheral cells that may facilitate central-peripheral crosstalk during inflammation. Examining these molecular mediators in human disease and other rodent models may reveal novel targets that modify brain health, especially in comorbidities characterized by peripheral inflammation.
Collapse
|
15
|
Lin HB, Hong P, Yin MY, Yao ZJ, Zhang JY, Jiang YP, Huang XX, Xu SY, Li FX, Zhang HF. Monocyte-Derived Macrophages Aggravate Cardiac Dysfunction After Ischemic Stroke in Mice. J Am Heart Assoc 2024; 13:e034731. [PMID: 38700011 PMCID: PMC11179859 DOI: 10.1161/jaha.123.034731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/25/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.
Collapse
MESH Headings
- Animals
- Macrophages/metabolism
- Disease Models, Animal
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- Ischemic Stroke/physiopathology
- Ischemic Stroke/metabolism
- Ischemic Stroke/pathology
- Mice, Inbred C57BL
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Male
- Mice, Knockout
- Mice
- Infarction, Middle Cerebral Artery/physiopathology
- Infarction, Middle Cerebral Artery/pathology
- Sympathetic Nervous System/physiopathology
- Myocardium/pathology
- Myocardium/metabolism
- Heart Diseases/etiology
- Heart Diseases/physiopathology
- Heart Diseases/pathology
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- CX3C Chemokine Receptor 1/deficiency
Collapse
Affiliation(s)
- Hong-Bin Lin
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Pu Hong
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Meng-Yu Yin
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Zhi-Jun Yao
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Jin-Yu Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science Guangzhou Guangdong China
| | - Yan-Pin Jiang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Xuan-Xuan Huang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Shi-Yuan Xu
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Feng-Xian Li
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| | - Hong-Fei Zhang
- Department of Anesthesiology, Zhujiang Hospital Southern Medical University Guangzhou Guangdong China
| |
Collapse
|
16
|
Di Martino E, Ambikan A, Ramsköld D, Umekawa T, Giatrellis S, Vacondio D, Romero AL, Galán MG, Sandberg R, Ådén U, Lauschke VM, Neogi U, Blomgren K, Kele J. Inflammatory, metabolic, and sex-dependent gene-regulatory dynamics of microglia and macrophages in neonatal hippocampus after hypoxia-ischemia. iScience 2024; 27:109346. [PMID: 38500830 PMCID: PMC10945260 DOI: 10.1016/j.isci.2024.109346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/02/2024] [Accepted: 02/22/2024] [Indexed: 03/20/2024] Open
Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of perinatal death and long-term disabilities worldwide. Post-ischemic neuroinflammation plays a pivotal role in HI pathophysiology. In the present study, we investigated the temporal dynamics of microglia (CX3CR1GFP/+) and infiltrating macrophages (CCR2RFP/+) in the hippocampi of mice subjected to HI at postnatal day 9. Using inflammatory pathway and transcription factor (TF) analyses, we identified a distinct post-ischemic response in CCR2RFP/+ cells characterized by differential gene expression in sensome, homeostatic, matrisome, lipid metabolic, and inflammatory molecular signatures. Three days after injury, transcriptomic signatures of CX3CR1GFP/+ and CCR2RFP/+ cells isolated from hippocampi showed a partial convergence. Interestingly, microglia-specific genes in CX3CR1GFP/+ cells showed a sexual dimorphism, where expression returned to control levels in males but not in females during the experimental time frame. These results highlight the importance of further investigations on metabolic rewiring to pave the way for future interventions in asphyxiated neonates.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
| | - Anoop Ambikan
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| | - Daniel Ramsköld
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Takashi Umekawa
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sarantis Giatrellis
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Davide Vacondio
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
| | | | - Marta Gómez Galán
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Rickard Sandberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Ulrika Ådén
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, 58183 Linköping, Sweden
- Neonatology, Karolinska University Hospital, Stockholm, Sweden
| | - Volker M. Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
- University of Tuebingen, 72074 Tuebingen, Germany
| | - Ujjwal Neogi
- The Systems Virology Lab, Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| | - Klas Blomgren
- Department of Women’s and Children’s Health, Karolinska Institutet, 17177 Stockholm, Sweden
- Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Julianna Kele
- Department of Physiology and Pharmacology, Karolinska Institutet, 17165 Stockholm, Sweden
- Team Neurovascular Biology and Health, Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet, 14152 Huddinge, Sweden
| |
Collapse
|
17
|
Bolini L, Campos RMP, Spiess DA, Lima-Rosa FL, Dantas DP, Conde L, Mendez-Otero R, Vale AM, Pimentel-Coelho PM. Long-term recruitment of peripheral immune cells to brain scars after a neonatal insult. Glia 2024; 72:546-567. [PMID: 37987116 DOI: 10.1002/glia.24490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/23/2023] [Accepted: 10/31/2023] [Indexed: 11/22/2023]
Abstract
Although brain scars in adults have been extensively studied, there is less data available regarding scar formation during the neonatal period, and the involvement of peripheral immune cells in this process remains unexplored in neonates. Using a murine model of neonatal hypoxic-ischemic encephalopathy (HIE) and confocal microscopy, we characterized the scarring process and examined the recruitment of peripheral immune cells to cortical and hippocampal scars for up to 1 year post-insult. Regional differences in scar formation were observed, including the presence of reticular fibrotic networks in the cortex and perivascular fibrosis in the hippocampus. We identified chemokines with chronically elevated levels in both regions and demonstrated, through a parabiosis-based strategy, the recruitment of lymphocytes, neutrophils, and monocyte-derived macrophages to the scars several weeks after the neonatal insult. After 1 year, however, neutrophils and lymphocytes were absent from the scars. Our data indicate that peripheral immune cells are transient components of HIE-induced brain scars, opening up new possibilities for late therapeutic interventions.
Collapse
Affiliation(s)
- Lukas Bolini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Daiane Aparecida Spiess
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Frederico Luis Lima-Rosa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danillo Pereira Dantas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andre M Vale
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
18
|
Zhang X, Qiu L, Sultan DH, Luehmann HP, Yu Y, Zhang X, Heo GS, Li A, Lahad D, Rho S, Tu Z, Liu Y. Development of a CCR2 targeted 18F-labeled radiotracer for atherosclerosis imaging with PET. Nucl Med Biol 2024; 130-131:108893. [PMID: 38422918 PMCID: PMC10964492 DOI: 10.1016/j.nucmedbio.2024.108893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease and the leading cause of morbidity and mortality worldwide. CC motif chemokine ligand 2 and its corresponding cognate receptor 2 (CCL2/CCR2) signaling has been implicated in regulating monocyte recruitment and macrophage polarization during inflammatory responses that plays a pivotal role in atherosclerosis initiation and progression. In this study, we report the design and synthesis of a novel 18F radiolabeled small molecule radiotracer for CCR2-targeted positron emission tomography (PET) imaging in atherosclerosis. The binding affinity of this radiotracer to CCR2 was evaluated via in vitro binding assay using CCR2+ membrane and cells. Ex vivo biodistribution was carried out in wild type mice to assess radiotracer pharmacokinetics. CCR2 targeted PET imaging of plaques was performed in two murine atherosclerotic models. The sensitive detection of atherosclerotic lesions highlighted the potential of this radiotracer for CCR2 targeted PET and warranted further optimization.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Lin Qiu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Debbie H Sultan
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Hannah P Luehmann
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yanbo Yu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Xiuli Zhang
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Alexandria Li
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Divangana Lahad
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Shinji Rho
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Zhude Tu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
19
|
Tian S, Zheng H, Wu W, Wu L. Predicting Diagnostic Biomarkers Associated with Pyroptosis in Neuropathic Pain Based on Machine Learning and Experimental Validation. J Inflamm Res 2024; 17:1121-1145. [PMID: 38406324 PMCID: PMC10893895 DOI: 10.2147/jir.s445382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Purpose Previous studies have shown that pyroptosis plays a vital role in the progress of neuropathic pain (NP), but the molecular mechanisms have not been fully elucidated. The aim of this study was to identify crucial pyroptosis-related genes (PRGs) in NP. Methods We identified pyroptosis-related differentially expressed genes (PRDEGs) in NP by machine learning analysis of the GSE24982 and GSE60670 datasets. Furthermore, these PRDEGs were subjected to Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, Gene Set Enrichment Analysis (GSEA) and Friends analysis, respectively. Meanwhile, receiver operator characteristic (ROC) analysis was performed to assess the diagnostic value of PRDEGs in NP. Finally, we performed immune infiltration analysis of key PRDEGs using CIBERSORTR R package. Results We found that 5 PRDEGs by least absolute shrinkage and selection operator (LASSO) regression and random forest and verified by RT-qPCR. GO, KEGG and GSEA revealed that these PRDEGs were mainly enriched in regulation of neuron death, IL-4 signaling, IL-23 pathway, and NF-κB pathway. ROC analysis revealed that most of the PRDEGs performed well in diagnosing NP. We also revealed transcription factors, miRNA regulatory networks and drug interaction networks of PRDEGs. For immune infiltration analysis, PRDEGs were mainly correlated with dendritic cells, monocytes and follicular T helper cells, suggested that it might be involved in the regulation of neuroimmune-related signaling. Conclusion A total of five PRDEGs were can be employed as NP biomarkers, particularly Tlr4, Il1b and Casp8, and provide additional evidence for a vital role of pyroptosis in NP.
Collapse
Affiliation(s)
- Sheng Tian
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Heqing Zheng
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Wei Wu
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Lanxiang Wu
- Department of Neurology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, People’s Republic of China
- Institute of Neuroscience, Nanchang University, Nanchang, 330006, People’s Republic of China
| |
Collapse
|
20
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
21
|
Yu Z, Pang H, Yang Y, Luo D, Zheng H, Huang Z, Zhang M, Ren K. Microglia dysfunction drives disrupted hippocampal amplitude of low frequency after acute kidney injury. CNS Neurosci Ther 2024; 30:e14363. [PMID: 37469216 PMCID: PMC10848109 DOI: 10.1111/cns.14363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/21/2023] Open
Abstract
AIMS Acute kidney injury (AKI) has been associated with a variety of neurological problems, while the neurobiological mechanism remains unclear. In the present study, we utilized resting-state functional magnetic resonance imaging (rs-fMRI) to detect brain injury at an early stage and investigated the impact of microglia on the neuropathological mechanism of AKI. METHODS Rs-fMRI data were collected from AKI rats and the control group with a 9.4-Tesla scanner at 24, 48, and 72 h post administration of contrast medium or saline. The amplitude of low-frequency fluctuations (ALFF) was then compared across the groups at each time course. Additionally, flow cytometry and SMART-seq2 were employed to evaluate microglia. Furthermore, pathological staining and Western blot were used to analyze the samples. RESULTS MRI results revealed that AKI led to a decreased ALFF in the hippocampus, particularly in the 48 h and 72 h groups. Additionally, western blot suggested that AKI-induced the neuronal apoptosis at 48 h and 72 h. Flow cytometry and confocal microscopy images demonstrated that AKI activated the aggregation of microglia into neurons at 24 h, with a strong upregulation of M1 polarization at 48 h and peaking at 72 h, accompanying with the release of proinflammatory cytokines. The ALFF value was strongly correlated with the proportion of microglia (|r| > 0.80, p < 0.001). CONCLUSIONS Our study demonstrated that microglia aggregation and inflammatory factor upregulation are significant mechanisms of AKI-induced neuronal apoptosis. We used fMRI to detect the alterations in hippocampal function, which may provide a noninvasive method for the early detection of brain injury after AKI.
Collapse
Affiliation(s)
- Ziyang Yu
- School of MedicineXiamen UniversityXiamenChina
| | - Huize Pang
- Department of RadiologyThe First Hospital of China Medical UniversityShenyangChina
| | - Yifan Yang
- School of MedicineXiamen UniversityXiamenChina
| | - Doudou Luo
- School of MedicineXiamen UniversityXiamenChina
| | - Haiping Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Zicheng Huang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public HealthXiamen UniversityXiamenChina
| | - Mingxia Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life SciencesXiamen UniversityXiamenChina
| | - Ke Ren
- School of MedicineXiamen UniversityXiamenChina
- Department of RadiologyThe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
22
|
Lu H, Shen H, Mao L, Mussap M, Song L. A ferroptosis-related ceRNA network for investigating the molecular mechanisms and the treatment of neonatal hypoxic-ischemic encephalopathy. Transl Pediatr 2024; 13:119-136. [PMID: 38323182 PMCID: PMC10839276 DOI: 10.21037/tp-23-596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/16/2024] [Indexed: 02/08/2024] Open
Abstract
Background Neonatal hypoxic-ischemic brain damage (HIBD) is a clinical syndrome causing brain injury in newborns with obscure etiology. Increasing evidence suggests that ferroptosis plays a role in HIBD. This study aimed to clarify the key ferroptosis-related genes (FRGs) of HIBD, construct a long non-coding RNA-microRNA-messenger RNA (lncRNA-miRNA-mRNA) network, and further investigate the pathogenesis of HIBD. Methods Gene expression data were downloaded from the Gene Expression Omnibus and FerrDb databases. The differentially expressed lncRNAs and FRGs were screened, and the related miRNAs and mRNAs were predicted. The obtained mRNA was intersected with the differentially expressed FRGs (DE-FRGs) to identify the key DE-FRGs. Cell-type Identification by Estimating Relative Subsets of RNA Transcripts method was applied to analyze the immune cell infiltration level and the relationship between key genes and immune cells. Results Gene differential expression analysis revealed that 1,178 lncRNAs, 207 miRNAs, and 647 mRNAs were differentially expressed in the blood of HIBD patients in comparison to healthy controls. The correlations of the lncRNAs, miRNAs, and mRNAs lead to the establishment of a competing endogenous RNA (ceRNA) network associated with ferroptosis in HIBD. Further validation using an external dataset and quantitative real-time polymerase chain reaction (PCR) analysis of brain tissues from hypoxic-ischemic encephalopathy rats confirmed the expression patterns of three key genes, including HMOX1, MYCN, and QSOX1. Meanwhile, the three key genes were closely correlated with the infiltration of multiple immune cells and might affect the function of HIBD regulatory genes such as CPT2 and GCK. In addition, drug prediction suggested that four drugs, including cephaeline, emetine, mestranol, and sulmazole, might alleviate HIBD. Conclusions Our study established a ceRNA network, identified three key genes, and predicted four drugs that are associated with ferroptosis in HIBD, which provides new ideas for the investigation of the disease mechanisms and might facilitate the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Hongyi Lu
- Department of Pediatrics, Nantong First People’s Hospital (The Second Affiliated Hospital of Nantong University), Nantong, China
| | - Haiyan Shen
- Department of Pediatrics, Nantong First People’s Hospital (The Second Affiliated Hospital of Nantong University), Nantong, China
| | - Liming Mao
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China
| | - Michele Mussap
- Laboratory Unit, Department of Surgical Sciences, University of Cagliari, Monserrato, Italy
| | - Lei Song
- Department of Pediatrics, Nantong First People’s Hospital (The Second Affiliated Hospital of Nantong University), Nantong, China
| |
Collapse
|
23
|
June A, Matysik W, Marlicz M, Zucker E, Wagley PK, Kuan CY, Burnsed J. Acute seizure activity in neonatal inflammation-sensitized hypoxia-ischemia in mice. PLoS One 2024; 19:e0295860. [PMID: 38206902 PMCID: PMC10783742 DOI: 10.1371/journal.pone.0295860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/30/2023] [Indexed: 01/13/2024] Open
Abstract
OBJECTIVE To examine acute seizure activity and neuronal damage in a neonatal mouse model of inflammation-sensitized hypoxic-ischemic (IS-HI) brain injury utilizing continuous electroencephalography (cEEG) and neurohistology. METHODS Neonatal mice were exposed to either IS-HI with Escherichia coli lipopolysaccharide (LPS) or HI alone on postnatal (p) day 10 using unilateral carotid artery ligation followed by global hypoxia (n = 10 [5 female, 5 male] for IS-HI, n = 12 [5 female, 7 male] for HI alone). Video cEEG was recorded for the duration of the experiment and analyzed for acute seizure activity and behavior. Brain tissue was stained and scored based on the degree of neuronal injury in the hippocampus, cortex, and thalamus. RESULTS There was no significant difference in acute seizure activity among mice exposed to IS-HI compared to HI with regards to seizure duration (mean = 63 ± 6 seconds for HI vs mean 62 ± 5 seconds for IS-HI, p = 0.57) nor EEG background activity. Mice exposed to IS-HI had significantly more severe neural tissue damage at p30 as measured by neuropathologic scores (mean = 8 ± 1 vs 23 ± 3, p < 0.0001). INTERPRETATION In a neonatal mouse model of IS-HI, there was no significant difference in acute seizure activity among mice exposed to IS-HI compared to HI. Mice exposed to IS-HI did show more severe neuropathologic damage at a later age, which may indicate the presence of chronic inflammatory mechanisms of brain injury distinct from acute seizure activity.
Collapse
Affiliation(s)
- Angelina June
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Weronika Matysik
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Maria Marlicz
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Emily Zucker
- College of Arts and Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Pravin K. Wagley
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Chia-Yi Kuan
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jennifer Burnsed
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Neurology, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
24
|
Jin L, Xiao L, Manley BJ, Oh EG, Huang W, Zhang Y, Chi J, Shi W, Kerrigan JR, Sung SSJ, Kuan CY, Li X. CCR2 monocytes as therapeutic targets for acute disc herniation and radiculopathy in mouse models. Osteoarthritis Cartilage 2024; 32:52-65. [PMID: 37802464 PMCID: PMC10873076 DOI: 10.1016/j.joca.2023.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/27/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023]
Abstract
OBJECTIVE Back pain and radiculopathy caused by disc herniation are major health issues worldwide. While macrophages are key players in disc herniation induced inflammation, their roles and origins in disease progression remain unclear. We aim to study the roles of monocytes and derivatives in a mouse model of disc herniation. METHODS Using a CCR2-CreER; R26R-EGFP (Ai6) transgenic mouse strain, we fate-mapped C-C chemokine receptor type 2 (CCR2) expressing monocytes and derivatives at disc herniation sites, and employed a CCR2RFP/RFP mouse strain and a CCR2-specific antagonist to study the effects of CCR2+ monocytes on local inflammatory responses, pain level, and disc degeneration by immunostaining, flow cytometry, and histology. RESULTS CCR2+ monocytes (GFP+) increased at the sites of disc hernia over postoperative day 4, 6, and 9 in CCR2-CreER; Ai6 mice. F4/80+ cells increased, and meanwhile, CD11b+ cells trended downward. Co-localization analysis revealed that both GFP+CD11b+ and GFP+F4/80+ constituted the majority of CD11b+ and F4/80+ cells at disc hernia sites. Fluorescence activated cell sorter purified GFP+ cells exhibited higher cytokine expressions than GFP- cells. Inhibition of CCR2 signaling reduced infiltration of monocytes and macrophages, alleviated pain, maintained disc height, and reduced osteoclast activity in adjacent cortical bone for up to 1 month. CONCLUSION Our findings suggest that circulating CCR2+ monocytes play important roles in initiating and promoting the local inflammatory responses, pain sensitization, and degenerative changes after disc herniation, and thus may serve as therapeutic targets for disc herniation induced back and leg pain.
Collapse
Affiliation(s)
- Li Jin
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Li Xiao
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Brock J Manley
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Eunha G Oh
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Wendy Huang
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Yi Zhang
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Jialun Chi
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA
| | - Weibin Shi
- Department of Radiology and Medical Imaging, Charlottesville, VA 22908, USA; Department of Biochemistry and Molecular Genetics, Charlottesville, VA 22908, USA
| | - Jason R Kerrigan
- Department of Mechanical and Aerospace Engineering, Center of Applied Biomechanics, University of Virginia, Charlottesville, VA 22904, USA
| | - Sun-Sang J Sung
- Department of Medicine, Center for Immunity, Inflammation, and Regenerative Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Chia-Yi Kuan
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA 22908, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22904, USA.
| |
Collapse
|
25
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
26
|
Wang R, Li T, Diao S, Chen C. Inhibition of the proteoglycan receptor PTPσ promotes functional recovery on a rodent model of preterm hypoxic-ischemic brain injury. Exp Neurol 2023; 370:114564. [PMID: 37806512 DOI: 10.1016/j.expneurol.2023.114564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Preterm white matter injury (WMI) is the most common brain injury in preterm infants and is associated with long-term adverse neurodevelopmental outcomes. Protein tyrosine phosphatase sigma (PTPσ) was discovered as chondroitin sulfate proteoglycan (CSPG) receptor that played roles in inhibiting myelin regeneration in spinal injury, experimental autoimmune encephalomyelitis, and stroke models. However, the role of PTPσ in perinatal WMI is not well understood. AIMS This study examines the effect of PTPσ inhibition on neurodevelopmental outcomes, myelination, and neuroinflammation in a mouse model of preterm WMI. MATERIALS AND METHODS Modified Rice-Vannucci model was performed on postnatal day 3 (P3) C57BL/6 mice. Intracellular Sigma Peptide (ISP) or vehicle was administrated subcutaneously one hour after injury for an additional 14 consecutive days. A battery of behavioral tests was performed to evaluate the short- and long-term effects of ISP on neurobehavioral deficit. Real time qPCR, western blot, immunofluorescence, and transmission electron microscopy were performed to assess white matter development. qPCR and flow cytometry were performed to evaluate neuroinflammation and microglia/macrophage phenotype. RESULTS The expression of PTPσ was increased after preterm WMI. ISP improved short-term neurological outcomes and ameliorated long-term motor and cognitive function of mice after preterm WMI. ISP promoted oligodendrocyte differentiation, maturation, myelination, and improved microstructure of myelin after preterm WMI. Furthermore, ISP administration fostered a beneficial inflammatory response in the acute phase after preterm WMI, inhibited the infiltration of peripheral macrophages, and promoted anti-inflammatory phenotype of microglia/macrophages. CONCLUSION PTPσ inhibition can ameliorate neurofunctional deficit, promote white matter development, modulate neuroinflammation and microglia/macrophage phenotype after preterm WMI. Thus, ISP administration may be a potential therapeutic strategy to improve neurodevelopmental outcomes of perinatal WMI.
Collapse
Affiliation(s)
- Ran Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China; Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tiantian Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Sihao Diao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China.
| |
Collapse
|
27
|
Quan H, Zhang R. Microglia dynamic response and phenotype heterogeneity in neural regeneration following hypoxic-ischemic brain injury. Front Immunol 2023; 14:1320271. [PMID: 38094292 PMCID: PMC10716326 DOI: 10.3389/fimmu.2023.1320271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Hypoxic-ischemic brain injury poses a significant threat to the neural niche within the central nervous system. In response to this pathological process, microglia, as innate immune cells in the central nervous system, undergo rapid morphological, molecular and functional changes. Here, we comprehensively review these dynamic changes in microglial response to hypoxic-ischemic brain injury under pathological conditions, including stroke, chronic intermittent hypoxia and neonatal hypoxic-ischemic brain injury. We focus on the regulation of signaling pathways under hypoxic-ischemic brain injury and further describe the process of microenvironment remodeling and neural tissue regeneration mediated by microglia after hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Hongxin Quan
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Runrui Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
28
|
Tansey M, Boles J, Uriarte Huarte O. Microfluidics-free single-cell genomics reveals complex central-peripheral immune crosstalk in the mouse brain during peripheral inflammation. RESEARCH SQUARE 2023:rs.3.rs-3428910. [PMID: 37886510 PMCID: PMC10602178 DOI: 10.21203/rs.3.rs-3428910/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Inflammation is a realized detriment to brain health in a growing number of neurological diseases, but querying neuroinflammation in its cellular complexity remains a challenge. This manuscript aims to provide a reliable and accessible strategy for examining the brain's immune system. We compare the efficacy of cell isolation methods in producing ample and pure immune samples from mouse brains. Then, with the high-input single-cell genomics platform PIPseq, we generate a rich neuroimmune dataset containing microglia and many peripheral immune populations. To demonstrate this strategy's utility, we interrogate the well-established model of LPS-induced neuroinflammation with single-cell resolution. We demonstrate the activation of crosstalk between microglia and peripheral phagocytes and highlight the unique contributions of microglia and peripheral immune cells to neuroinflammation. Our approach enables the high-depth evaluation of inflammation in longstanding rodent models of neurological disease to reveal novel insight into the contributions of the immune system to brain health.
Collapse
|
29
|
Zhang A, Liu Y, Wang X, Xu H, Fang C, Yuan L, Wang K, Zheng J, Qi Y, Chen S, Zhang J, Shao A. Clinical Potential of Immunotherapies in Subarachnoid Hemorrhage Treatment: Mechanistic Dissection of Innate and Adaptive Immune Responses. Aging Dis 2023; 14:1533-1554. [PMID: 37196120 PMCID: PMC10529760 DOI: 10.14336/ad.2023.0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/26/2023] [Indexed: 05/19/2023] Open
Abstract
Subarachnoid hemorrhage (SAH), classified as a medical emergency, is a devastating and severe subtype of stroke. SAH induces an immune response, which further triggers brain injury; however, the underlying mechanisms need to be further elucidated. The current research is predominantly focused on the production of specific subtypes of immune cells, especially innate immune cells, post-SAH onset. Increasing evidence suggests the critical role of immune responses in SAH pathophysiology; however, studies on the role and clinical significance of adaptive immunity post-SAH are limited. In this present study, we briefly review the mechanistic dissection of innate and adaptive immune responses post-SAH. Additionally, we summarized the experimental studies and clinical trials of immunotherapies for SAH treatment, which may form the basis for the development of improved therapeutic approaches for the clinical management of SAH in the future.
Collapse
Affiliation(s)
- Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - KaiKai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yangjian Qi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
30
|
Murata S, Baig N, Decker K, Halaris A. Systemic Inflammatory Response Index (SIRI) at Baseline Predicts Clinical Response for a Subset of Treatment-Resistant Bipolar Depressed Patients. J Pers Med 2023; 13:1408. [PMID: 37763175 PMCID: PMC10533150 DOI: 10.3390/jpm13091408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Background: in a recent double-blind, placebo controlled RCT we demonstrated that selective inhibition of cyclo-oxygenase 2 (COX2) is an effective adjunctive strategy in treatment-resistant bipolar depression (TRBDD). To better clarify the mechanisms underlying TRBDD and treatment response, we conducted a retrospective exploratory analysis of the systemic inflammatory response index (SIRI = absolute neutrophils × absolute monocytes/absolute lymphocytes) in relation to other biomarkers and clinical outcomes after escitalopram (ESC), combined with the COX-2 inhibitor, celecoxib (CBX), versus placebo. Methods: Baseline measures of SIRI were compared between TRBDD and healthy controls (HC), and correlated with blood-based inflammatory cytokines, kynurenines, and growth factors. Post-treatment Hamilton Depression Rating Scale 17 (HAMD-17) total scores (clinical outcome) were modelled according to SIRI adjusting for demographics (including relevant interactions with SIRI), baseline depression, treatment arm, and treatment timepoint using multiple linear regression and robust linear mixed effects models. Results: Baseline SIRI did not distinguish TRBDD from HC groups. Baseline SIRI was significantly correlated with lower baseline MCP-1. The relationship between SIRI and HAMD-17 was significant at treatment week 8, in contrast to baseline. Finally, baseline SIRI predicted elevated post-treatment HAMD-17 scores, amongst patients with elevated depression scores at baseline. Significance: High pre-treatment SIRI may predict poorer depressive outcomes amongst TRBDD patients with baseline elevated depression.
Collapse
Affiliation(s)
- Stephen Murata
- Pine Rest Christian Mental Health Services, Michigan State University, 300 68th Street SE, Grand Rapids, MI 49548, USA
| | - Nausheen Baig
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA; (N.B.); (K.D.); (A.H.)
- Stritch School of Medicine, Loyola University, Maywood, IL 60153, USA
| | - Kyle Decker
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA; (N.B.); (K.D.); (A.H.)
- Stritch School of Medicine, Loyola University, Maywood, IL 60153, USA
| | - Angelos Halaris
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago, Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA; (N.B.); (K.D.); (A.H.)
| |
Collapse
|
31
|
Pimentel‐Coelho PM. Monocytes in neonatal stroke and hypoxic‐ischemic encephalopathy: Pathophysiological mechanisms and therapeutic possibilities. NEUROPROTECTION 2023; 1:66-79. [DOI: 10.1002/nep3.22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/13/2023] [Indexed: 01/03/2025]
Abstract
AbstractNeonatal arterial ischemic stroke (NAIS) and neonatal hypoxic‐ischemic encephalopathy (HIE) are common causes of neurological impairments in infants, for which treatment options are very limited. NAIS and HIE induce an innate immune response that involves the recruitment of peripheral immune cells, including monocytes, into the brain. Monocytes and monocyte‐derived cells have the potential to contribute to both harmful and beneficial pathophysiological processes, such as neuroinflammation and brain repair, but their roles in NAIS and HIE remain poorly understood. Furthermore, recent evidence indicates that monocyte‐derived macrophages can persist in the brain for several months following NAIS and HIE in mice, with possible long‐lasting consequences that are still unknown. This review provides a comprehensive overview of the mechanisms of monocyte infiltration and their potential functions in the ischemic brain, focusing on HIE and NAIS. Therapeutic strategies targeting monocytes and the possibility of using monocytes for cell‐based therapies are also discussed.
Collapse
Affiliation(s)
- Pedro M. Pimentel‐Coelho
- Carlos Chagas Filho Biophysics Institute Federal University of Rio de Janeiro Rio de Janeiro Brazil
| |
Collapse
|
32
|
Monet MC, Quan N. Complex Neuroimmune Involvement in Neurodevelopment: A Mini-Review. J Inflamm Res 2023; 16:2979-2991. [PMID: 37489149 PMCID: PMC10363380 DOI: 10.2147/jir.s410562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023] Open
Abstract
It is increasingly evident that cells and molecules of the immune system play significant roles in neurodevelopment. As perinatal infection is associated with the development of neurodevelopmental disorders, previous research has focused on demonstrating that the induction of neuroinflammation in the developing brain is capable of causing neuropathology and behavioral changes. Recent studies, however, have revealed that immune cells and molecules in the brain can influence neurodevelopment without the induction of overt inflammation, identifying neuroimmune activities as integral parts of normal neurodevelopment. This mini-review describes the shift in literature that has moved from emphasizing the intrusion of inflammatory events as a main culprit of neurodevelopmental disorders to evaluating the deviation of the normal neuroimmune activities in neurodevelopment as a potential pathogenic mechanism.
Collapse
Affiliation(s)
- Marianne C Monet
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Jupiter, FL, USA
| |
Collapse
|
33
|
Hu H, Yang X, He Y, Duan C, Sun N. Psychological stress induces depressive-like behavior associated with bone marrow-derived monocyte infiltration into the hippocampus independent of blood-brain barrier disruption. J Neuroinflammation 2022; 19:208. [PMID: 36002834 PMCID: PMC9400267 DOI: 10.1186/s12974-022-02569-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
Abstract
Background Psychological stress is one of the most important factors that trigger emotional disorders, such as depression and anxiety. Emerging evidence suggests that neuroinflammation exacerbated by bidirectional communication between the peripheral immune system and the central nervous system facilitates abnormal psychiatric symptoms. This study aimed to investigate the hippocampal migration of bone marrow (BM)-derived monocytes and its role in regulating depressive-like behaviors using the chronic psychological stress (CPS) mouse model. More importantly, whether the central migration of these peripheral BM-derived cells depend on the disruption of the blood–brain barrier (BBB) was also investigated. Methods and findings Green fluorescent protein-positive (GFP+) BM chimeric mice were used to distinguish BM-derived monocytes within the brain. A CPS mouse model was established to explore the effect of CPS on hippocampal migration of BM-derived monocytes and its role in the regulation of depressive-like behaviors. The results revealed that BM-derived GFP+ cells accumulated in the hippocampus and differentiated into microglia-like cells after exposure to CPS. Interestingly, this migration was not associated with BBB disruption. Furthermore, treatment with C–C chemokine receptor 2 (CCR2) antagonist (RS102895) suppressed the recruitment of BM-derived monocytes to the hippocampus and alleviated depressive-like symptoms. Conclusion These findings indicate that monocyte recruitment to the hippocampus in response to psychological stress may represent a novel cellular mechanism that contributes to the development of depression. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02569-w.
Collapse
Affiliation(s)
- Huiling Hu
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xue Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Yuqing He
- Translational Medicine Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chaohui Duan
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Nannan Sun
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
34
|
Cai Q, Zhang X, Shen L, Song H, Wang T. The protective effect of MiR-27a on the neonatal hypoxic-ischemic encephalopathy by targeting FOXO1 in rats. Transl Pediatr 2022; 11:1199-1208. [PMID: 35958013 PMCID: PMC9360825 DOI: 10.21037/tp-22-259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/04/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic encephalopathy (HIE), a kind of hypoxic-ischemic brain damage caused by perinatal asphyxia, is the most crucial cause of neonatal death and long-term neurological dysfunction in children. We aimed to investigate the protective effects of micro (mi)R-27a on HIE in neonatal rats. METHODS A rat model of neonatal HIE was constructed by modification of the Rice-Vannucci model. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to test the expressions of miR-27a, FOXO1 messenger RNA (mRNA), interleukin-1β (IL-1β) mRNA, and tumor necrosis factor-α (TNF-α) mRNA, and western blot was applied to test the expression of FOXO1. In order to overexpress miR-27a, an intracerebroventricular injection (i.c.v) of miR-27a mimic was administered. We adopted 2,3,5-triphenytetrazolium chloride (TTC) staining and brain water content measurement to test the effects of miR-27a on the infarcted volume and edema in brain after HIE. Flow cytometry (FCM) analysis was applied to test the effects of miR-27a on the infiltrated peripheral immune cells in the rat brains after HIE. RESULTS We successfully established a rat model of neonatal HIE. It was revealed that the expressions of miR-27a decreased gradually after HIE, however, the expressions of FOXO1 mRNA increased. After injection of the miR-27a mimic, the expression of miR-27a in the rat HIE model brains was significantly upregulated, however, the expression of FOXO1 was robustly downregulated. Both TTC staining and brain water content showed that the infarcted volume and brain edema was markedly increased after HIE. Interestingly, the overexpression of miR-27a reduced the infarcted volume and edema induced by HIE. Additionally, RT-qPCR and FCM analysis showed that HIE lead to increases of IL-1β, TNF-α, and infiltrated immune cells. Overexpression of miR-27a could reduce the expressions of IL-1β mRNA and TNF-α mRNA, and the cell numbers of infiltrated peripheral macrophages and neutrophils in the brain. CONCLUSIONS MiR-27a plays protective roles by reducing infarct volume and brain edema, and inhibiting inflammatory factors and infiltrated peripheral immune cells by targeting FOXO1 in neonatal HIE rats.
Collapse
Affiliation(s)
- Qun Cai
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoqun Zhang
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Liyuan Shen
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Honghua Song
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Ting Wang
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|