1
|
Gonzalez CE, Vaidya RS, Clayton SW, Tang SY. Secreted chemokines and transcriptomic analyses reveal diverse inflammatory and degenerative processes in the intervertebral disc of the STZ-HFD mouse model of Type 2 diabetes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.31.605332. [PMID: 39131361 PMCID: PMC11312574 DOI: 10.1101/2024.07.31.605332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The chronic inflammation resultant from type 2 diabetes (T2D) is also associated with spinal pathologies, including intervertebral disc (IVD) degeneration and chronic neck and back pain. Although confounding factors, such as increased weight gain in obesity, studies have shown that even after adjusting age, body mass index, and genetics (e.g. twins), patients with T2D suffer from disproportionately more IVD degeneration and back pain. We hypothesize that chronic T2D fosters a proinflammatory microenvironment within the IVD that promotes degeneration and disrupts disc homeostasis. To test this hypothesis, we evaluated two commonly used mouse models of T2D - the leptin-receptor deficient mouse (db/db) and the chronic high-fat diet in mice with impaired beta-cell function (STZ-HFD). STZ-HFD IVDs were more degenerated and showed differential expression of chemokines from the db/db models. Moreover, the RNAseq analysis revealed vast transcriptional dysregulation of many pathways in the STZ-HFD but not in the db/db tissues. Leptin signaling may be essential to mediating the inflammation in T2D. Taken together, the STZ-HFD may better recapitulates the complexities of the chronic inflammatory processes in the IVD during T2D.
Collapse
|
2
|
Nikolova D, Todorova Y, Hammoudeh Z, Rukova B, Emilova R, Aleksova M, Koleva V, Nikolova M. Gene Expression Changes as Biomarkers of Immunosenescence in Bulgarian Individuals of Active Age. Biomedicines 2025; 13:721. [PMID: 40149697 PMCID: PMC11940667 DOI: 10.3390/biomedicines13030721] [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: 02/10/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Immunosenescence implies innate and adaptive immunity dysfunction, which naturally occurs with aging. It is a complex multifactorial process which can be triggered by either genetic changes, immune changes or both. Numerous research studies have shown that the process of senescence goes alongside chronic immune activation. The purpose of this study is to analyze the changes in the expression of genes associated with adaptive and innate immune responses in order to identify reliable biomarkers for immune aging. Methods: For that aim, 55 clinically healthy individuals of active age (21-65 years) were distributed based on immunophenotyping in two groups, with and without signs of premature senescence. A gene expression analysis was subsequently made on those two groups, and the differentially expressed genes were presented and interpreted. Results: Altogether, forty-eight (48) genes exhibited differential expression between the two groups, most of which showed up-regulation (45) (fold change more than 2), and only three were down-regulated (fold change less than -2). The highest positive fold change showed IL-1β (10.76), BCL6 (13.25) and CCL4 (15.91), while the highest negative fold changes were documented for IL23R (-3.10), IL5 (-2.66) and PTGS2 (COX-2) (-2.15). Conclusions: Our results reveal that immunosenescence is positively associated with chronic inflammation, which is typical for the aging process. On the other hand, we identified markers of possible protective effects against oxidative stress and tumorigenesis. These findings can aid the early diagnosis of chronic degenerative diseases in subclinical phase, as well as the development of strategies to prevent the processes of premature immune aging.
Collapse
Affiliation(s)
- Dragomira Nikolova
- Department of Medical Genetics, Medical Faculty, Medical University, 1431 Sofia, Bulgaria;
| | - Yana Todorova
- National Reference Laboratory of Immunology, National Center for Infectious and Parasitic Diseases, 1504 Sofia, Bulgaria; (Y.T.); (R.E.); (M.A.); (M.N.)
| | - Zora Hammoudeh
- Department of Clinical Laboratory, Acibadem City Clinic Tokuda Hospital, 1407 Sofia, Bulgaria; (Z.H.); (V.K.)
| | - Blaga Rukova
- Department of Medical Genetics, Medical Faculty, Medical University, 1431 Sofia, Bulgaria;
| | - Radoslava Emilova
- National Reference Laboratory of Immunology, National Center for Infectious and Parasitic Diseases, 1504 Sofia, Bulgaria; (Y.T.); (R.E.); (M.A.); (M.N.)
| | - Milena Aleksova
- National Reference Laboratory of Immunology, National Center for Infectious and Parasitic Diseases, 1504 Sofia, Bulgaria; (Y.T.); (R.E.); (M.A.); (M.N.)
| | - Vesselina Koleva
- Department of Clinical Laboratory, Acibadem City Clinic Tokuda Hospital, 1407 Sofia, Bulgaria; (Z.H.); (V.K.)
| | - Maria Nikolova
- National Reference Laboratory of Immunology, National Center for Infectious and Parasitic Diseases, 1504 Sofia, Bulgaria; (Y.T.); (R.E.); (M.A.); (M.N.)
| |
Collapse
|
3
|
Sun R, Yu J, Zou Z, Yang S, Tuo Y, Tan L, Zhang H, Sun L, Bai H. FcγRI plays a pro-inflammatory role in the immune response to Chlamydia respiratory infection by upregulating dendritic cell-related genes. Int Immunopharmacol 2025; 147:113943. [PMID: 39752758 DOI: 10.1016/j.intimp.2024.113943] [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: 12/09/2024] [Revised: 12/15/2024] [Accepted: 12/22/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND FcγRI, a pivotal cell surface receptor, is implicated in diverse immune responses and is ubiquitously expressed on numerous immune cells. However, its role in intracellular bacterial infections remains understudied. METHODS Wild-type (WT) and FcγRI knockout (FcγRI-KO) mice were inoculated intranasally with a specific dose of C. muridarum. Lung tissues were harvested for transcriptome sequencing, and flow cytometry was employed to validate bioinformatics immune infiltration analysis. Differentially expressed DC-associated genes were subjected to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses to elucidate their functions during infection. A PPI network was constructed to pinpoint crucial genes, and qPCR was utilized to confirm their expression changes. Additionally, we compared body weight, lung Chlamydia load, and pathological alterations between WT and FcγRI-KO mice post-infection to assess the effect of FcγRI on inflammation via gene regulation. Lastly, an mRNA-miRNA-lncRNA network was formulated to further probe the molecular mechanisms of FcγRI in C. muridarum infection. RESULTS Post-C. muridarum infection, FcγRI-KO mice exhibited a notable decrease in DC infiltration and maturation, along with downregulated co-stimulatory molecules (CD40, CD80, CD86) in lung tissues. Differential gene analysis identified 26 differentially expressed DC-related genes implicated in DC proliferation, migration, and inflammatory responses. KEGG analysis revealed their close association with key immune pathways. The PPI network delineated two modules, with the top six genes in the pivotal cluster 1 (Ccl4, Il6, Ccl3, Ptgs2, Il 1α, Il7) being significantly downregulated in FcγRI-KO mice. A ceRNA network encompassing 12 miRNAs and 37 lncRNAs regulating four key genes (Ptgs2, Il1α, Il6, Il7) was also constructed. CONCLUSIONS In C. muridarum respiratory infections, FcγRI facilitates DC infiltration and maturation, upregulates six pro-inflammatory genes (Ccl4, Il6, Ccl3, Ptgs2, Il1α, Il7), and exhibits a pro-inflammatory role. A key ceRNA network was formulated to unravel the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Ruoyuan Sun
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Jinxi Yu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Zeyang Zou
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Shuaini Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Yuqing Tuo
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Lu Tan
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Hong Zhang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China
| | - Longhao Sun
- Department of General Surgery, Tianjin Medial University General Hospital, Tianjin 300052, China.
| | - Hong Bai
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
4
|
Peterson IL, Scholpa NE, Bachtle KJ, Frye JB, Loppi SH, Thompson AD, Doyle K, Largent-Milnes TM, Schnellmann RG. Formoterol alters chemokine expression and ameliorates pain behaviors after moderate spinal cord injury in female mice. J Pharmacol Exp Ther 2025; 392:100015. [PMID: 40023611 DOI: 10.1124/jpet.124.002171] [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: 02/07/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Secondary spinal cord injury (SCI) is characterized by increased cytokines and chemokines at the site of injury that have been associated with the development of neuropathic pain. Nearly 80% of SCI patients report suffering from chronic pain, which is poorly managed with available analgesics. While treatment with the US Food and Drug Administration-approved β2-adrenergic receptor agonist formoterol improves various aspects of recovery post-SCI in vivo, its effects on cytokines, chemokines, and neuropathic pain remain unknown. Female mice were subjected to moderate (60 kilodynes [kdyn]) or severe (80 kdyn) SCI followed by daily treatment with vehicle or formoterol (0.3 mg/kg, i.p.) beginning 8 hours after injury. The expression of proinflammatory cytokines/chemokines, such as interferon gamma-induced protein 10, macrophage inflammatory protein 1a, monocyte chemoattractant protein 1, B-cell attracting chemokine 1, and nuclear factor kappa-light-chain-enhancer of activated B-cells, was increased in the injury site of vehicle-treated mice 24 hours post-SCI, which was ameliorated with formoterol treatment, regardless of injury severity. Thermal hyperalgesia and mechanical allodynia, as measured by Hargreaves infrared apparatus and von Frey filaments, respectively, were assessed prior to SCI and then weekly beginning 21 days post-injury (DPI). While all injured mice exhibited decreased withdrawal latency following thermal stimulation compared with baseline, formoterol treatment reduced this response ∼15% by 35 DPI. Vehicle-treated mice displayed significant mechanical allodynia, as evidenced by a 55% decrease in withdrawal threshold from baseline. In contrast, mice treated with formoterol maintained a consistent withdrawal time at all times tested. These data indicate that formoterol reduces inflammation post-SCI, likely contributing to mitigation of neuropathic pain and further supporting the therapeutic potential of this treatment strategy. SIGNIFICANCE STATEMENT: Chronic pain is a detrimental consequence of spinal cord injury (SCI). We show that treatment with the US Food and Drug Administration-approved drug formoterol after SCI decreases injury site proinflammatory chemo-/cytokines and alters markers of glial cell activation and infiltration. Additionally, formoterol treatment improves locomotor function and body composition, and decreases lesion volume. Finally, formoterol treatment decreased mechanical allodynia and thermal hyperalgesia post-SCI. These data are suggestive of the mechanism of formoterol-induced recovery, and further indicate its potential as a therapeutic strategy for SCI.
Collapse
Affiliation(s)
- Ingrid L Peterson
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona; Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Natalie E Scholpa
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona.
| | - Kiara J Bachtle
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Jennifer B Frye
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Sanna H Loppi
- Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | - Austin D Thompson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona
| | - Kristian Doyle
- Southern Arizona VA Health Care System, Tucson, Arizona; Department of Immunobiology, College of Medicine, University of Arizona, Tucson, Arizona
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona; Southern Arizona VA Health Care System, Tucson, Arizona; Department of Neuroscience, College of Medicine, University of Arizona, Tucson, Arizona; Southwest Environmental Health Science Center, University of Arizona, Tucson, Arizona; Center for Innovation in Brain Science, University of Arizona, Tucson, Arizona.
| |
Collapse
|
5
|
Hasan A, Ardizzone A, Giosa D, Scuderi SA, Calcaterra E, Esposito E, Capra AP. The Therapeutic Potential of MicroRNA-21 in the Treatment of Spinal Cord Injury. Curr Issues Mol Biol 2025; 47:70. [PMID: 39996791 PMCID: PMC11854632 DOI: 10.3390/cimb47020070] [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: 12/18/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Spinal cord injury (SCI) involves complex pathological processes that often result in significant and long-term neurological deficits. Increasingly, research has identified microRNA-21 (miR-21) as a pivotal regulator in SCI, with studies focusing on its roles in inflammation, apoptosis, and tissue repair. This review synthesizes current findings on miR-21's involvement in post-injury molecular events, emphasizing its interactions with regulatory targets such as Phosphatase and Tensin Homolog (PTEN) and Programmed Cell Death Protein 4 (PDCD4), as well as its broader effects on inflammatory and apoptotic signaling pathways. Evidence from both in vitro and in vivo studies suggests that modulating miR-21 influences lesion size, cellular dynamics, and functional recovery, highlighting its potential as a therapeutic target for SCI. Nonetheless, the clinical translation of miR-21-based therapies poses significant challenges, including the need to optimize dosages, delivery mechanisms, and long-term safety profiles. Further research is crucial to fully delineate miR-21's therapeutic potential and determine its feasibility for integration into SCI treatment protocols. This review aims to provide a comprehensive overview of miR-21's roles in SCI pathology, offering insights into the molecular mechanisms underlying recovery and the emerging potential of miR-21 in SCI management to enhance outcomes and quality of life for affected patients.
Collapse
Affiliation(s)
- Ahmed Hasan
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
- Center of Neuroscience, School of Advanced Studies, University of Camerino, 62032 Camerino, Italy
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| | - Domenico Giosa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| | - Elsa Calcaterra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31, 98166 Messina, Italy; (A.H.); (A.A.); (D.G.); (S.A.S.); (E.C.); (A.P.C.)
| |
Collapse
|
6
|
Liu Z, Xiang C, Zhao X, Aizawa T, Niu R, Zhao J, Guo F, Li Y, Luo W, Liu W, Gu R. Regulation of dynamic spatiotemporal inflammation by nanomaterials in spinal cord injury. J Nanobiotechnology 2024; 22:767. [PMID: 39696584 DOI: 10.1186/s12951-024-03037-8] [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: 09/13/2024] [Accepted: 11/24/2024] [Indexed: 12/20/2024] Open
Abstract
Spinal cord injury (SCI) is a common clinical condition of the central nervous system that can lead to sensory and motor impairment below the injury level or permanent loss of function in severe cases. Dynamic spatiotemporal neuroinflammation is vital to neurological recovery, which is collectively constituted by the dynamic changes in a series of inflammatory cells, including microglia, neutrophils, and astrocytes, among others. Immunomodulatory nanomaterials can readily improve the therapeutic effects and simultaneously overcome various drawbacks associated with treatment, such as the off-target side effects and loss of bioactivity of immune agents during circulation. In this review, we discuss the role of dynamic spatiotemporal inflammation in secondary injuries after SCI, elaborate on the mechanism of action and effect of existing nanomaterials in treating SCI, and summarize the mechanism(s) whereby they regulate inflammation. Finally, the challenges and prospects associated with using nanotechnology to modulate immunotherapy are discussed to provide new insights for future treatment. Deciphering the intricate spatiotemporal mechanisms of neuroinflammation in SCI requires further in-depth studies. Therefore, SCI continues to represent a formidable challenge.
Collapse
Affiliation(s)
- Zeping Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Chunyu Xiang
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Xu Zhao
- Department of Orthopedics, Third Military Medical University, Xinqiao Hosp, 83 Xinqiao Main St, Chongqing, 400037, PR China
| | - Toshimi Aizawa
- Department of Orthopedic Surgery, Tohoku University Graduate School of Medicine, Sendai, 980-8574, Japan
| | - Renrui Niu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Jianhui Zhao
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Fengshuo Guo
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Yueying Li
- Department of Hand & Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China
| | - Wenqi Luo
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| | - Wanguo Liu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| | - Rui Gu
- Department of Orthopedic Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, PR China.
| |
Collapse
|
7
|
Zhang S, Yuan M, He D, Dang W, Zhang W. Long-term follow-up of brain regional changes and the association with cognitive impairment in quarantined COVID-19 survivors. Eur Arch Psychiatry Clin Neurosci 2024; 274:1911-1922. [PMID: 38319396 DOI: 10.1007/s00406-023-01741-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 12/11/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVE This study aimed to evaluate the neuropsychiatric symptoms of quarantined COVID-19 survivors 15 months after discharge and explore its potential association with structural and functional brain changes and inflammation. METHODS A total of 51 quarantined COVID-19 survivors and 74 healthy controls were included in this study. Cognitive function was assessed using the THINC-integrated tool. Structural brain changes were examined through both surface- and volume-based analyses, and functional changes were assessed using resting-state amplitude low-frequency fluctuation (ALFF). Serum inflammatory markers were measured by a multiplexed flow cytometric assay. RESULTS COVID-19 survivors exhibited subjective cognitive decline compared to healthy controls, despite no significant differences in objective cognitive tasks. Structural analysis revealed significantly increased gray matter volume and cortical surface area in the left transverse temporal gyrus (Heschl's gyrus) in quarantined COVID-19 survivors. This enlargement was negatively correlated with cognitive impairment. The ALFF analysis showed decreased neural activity in multiple brain regions. Elevated levels of serum inflammatory markers were also found in COVID-19 survivors, including MIP-1a, MIP-1b, TNF-a, and IL-8, which correlated with functional abnormalities. CONCLUSIONS Our findings indicate a subjective cognitive decline in quarantined COVID-19 survivors 15 months after discharge, which is associated with brain structural alterations in the left Heschl's gyrus. The observed elevation of inflammatory markers suggests a potential mechanism involving inflammation-induced neurogenesis. These results contribute to our understanding of the possible mechanisms underlying long-term neuropsychiatric consequences of COVID-19 and highlight the need for further research to develop targeted interventions.
Collapse
Affiliation(s)
- Simai Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu, 610041, China
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China
| | - Minlan Yuan
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Danmei He
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wen Dang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu, 610041, China.
- Med-X Center for Informatics, Sichuan University, Chengdu, 610041, China.
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Yu X, Chen Z, Bao W, Jiang Y, Ruan F, Wu D, Le K. The neutrophil extracellular traps in neurological diseases: an update. Clin Exp Immunol 2024; 218:264-274. [PMID: 38975702 PMCID: PMC11557138 DOI: 10.1093/cei/uxae057] [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: 01/16/2024] [Revised: 05/08/2024] [Accepted: 07/06/2024] [Indexed: 07/09/2024] Open
Abstract
Neutrophil extracellular traps (NETs) released by neutrophils are web-like DNA structures adhered to granulin proteins with bactericidal activity and can be an important mechanism for preventing pathogen dissemination or eliminating microorganisms. However, they also play important roles in diseases of other systems, such as the central nervous system. We tracked the latest advances and performed a review based on published original and review articles related to NETs and neurological diseases. Generally, neutrophils barely penetrate the blood-brain barrier into the brain parenchyma, but when pathological changes such as infection, trauma, or neurodegeneration occur, neutrophils rapidly infiltrate the central nervous system to exert their defensive effects. However, neutrophils may adversely affect the host when they uncontrollably release NETs upon persistent neuroinflammation. This review focused on recent advances in understanding the mechanisms and effects of NETs release in neurological diseases, and we also discuss the role of molecules that regulate NETs release in anticipation of clinical applications in neurological diseases.
Collapse
Affiliation(s)
- Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong S.A.R., China
| |
Collapse
|
9
|
Wang C, Wang J, Zhu Z, Hu J, Lin Y. Spotlight on pro-inflammatory chemokines: regulators of cellular communication in cognitive impairment. Front Immunol 2024; 15:1421076. [PMID: 39011039 PMCID: PMC11247373 DOI: 10.3389/fimmu.2024.1421076] [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: 04/21/2024] [Accepted: 06/12/2024] [Indexed: 07/17/2024] Open
Abstract
Cognitive impairment is a decline in people's ability to think, learn, and remember, and so forth. Cognitive impairment is a global health challenge that affects the quality of life of thousands of people. The condition covers a wide range from mild cognitive impairment to severe dementia, which includes Alzheimer's disease (AD) and Parkinson's disease (PD), among others. While the etiology of cognitive impairment is diverse, the role of chemokines is increasingly evident, especially in the presence of chronic inflammation and neuroinflammation. Although inflammatory chemokines have been linked to cognitive impairment, cognitive impairment is usually multifactorial. Researchers are exploring the role of chemokines and other inflammatory mediators in cognitive dysfunction and trying to develop therapeutic strategies to mitigate their effects. The pathogenesis of cognitive disorders is very complex, their underlying causative mechanisms have not been clarified, and their treatment is always one of the challenges in the field of medicine. Therefore, exploring its pathogenesis and treatment has important socioeconomic value. Chemokines are a growing family of structurally and functionally related small (8-10 kDa) proteins, and there is growing evidence that pro-inflammatory chemokines are associated with many neurobiological processes that may be relevant to neurological disorders beyond their classical chemotactic function and play a crucial role in the pathogenesis and progression of cognitive disorders. In this paper, we review the roles and regulatory mechanisms of pro-inflammatory chemokines (CCL2, CCL3, CCL4, CCL5, CCL11, CCL20, and CXCL8) in cognitive impairment. We also discuss the intrinsic relationship between the two, hoping to provide some valuable references for the treatment of cognitive impairment.
Collapse
Affiliation(s)
- Chenxu Wang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Endocrinology and Metabolism, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jiayi Wang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Endocrinology and Metabolism, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Zhichao Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Endocrinology and Metabolism, The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang, China
| | - Yong Lin
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesia, The First Affiliated Hospital of GanNan Medical University, Ganzhou, China
| |
Collapse
|
10
|
Hellenbrand DJ, Quinn CM, Piper ZJ, Elder RT, Mishra RR, Marti TL, Omuro PM, Roddick RM, Lee JS, Murphy WL, Hanna AS. The secondary injury cascade after spinal cord injury: an analysis of local cytokine/chemokine regulation. Neural Regen Res 2024; 19:1308-1317. [PMID: 37905880 PMCID: PMC11467934 DOI: 10.4103/1673-5374.385849] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/24/2023] [Accepted: 09/04/2023] [Indexed: 11/02/2023] Open
Abstract
After spinal cord injury, there is an extensive infiltration of immune cells, which exacerbates the injury and leads to further neural degeneration. Therefore, a major aim of current research involves targeting the immune response as a treatment for spinal cord injury. Although much research has been performed analyzing the complex inflammatory process following spinal cord injury, there remain major discrepancies within previous literature regarding the timeline of local cytokine regulation. The objectives of this study were to establish an overview of the timeline of cytokine regulation for 2 weeks after spinal cord injury, identify sexual dimorphisms in terms of cytokine levels, and determine local cytokines that significantly change based on the severity of spinal cord injury. Rats were inflicted with either a mild contusion, moderate contusion, severe contusion, or complete transection, 7 mm of spinal cord centered on the injury was harvested at varying times post-injury, and tissue homogenates were analyzed with a Cytokine/Chemokine 27-Plex assay. Results demonstrated pro-inflammatory cytokines including tumor necrosis factor α, interleukin-1β, and interleukin-6 were all upregulated after spinal cord injury, but returned to uninjured levels within approximately 24 hours post-injury, while chemokines including monocyte chemoattractant protein-1 remained upregulated for days post-injury. In contrast, several anti-inflammatory cytokines and growth factors including interleukin-10 and vascular endothelial growth factor were downregulated by 7 days post-injury. After spinal cord injury, tissue inhibitor of metalloproteinase-1, which specifically affects astrocytes involved in glial scar development, increased more than all other cytokines tested, reaching 26.9-fold higher than uninjured rats. After a mild injury, 11 cytokines demonstrated sexual dimorphisms; however, after a severe contusion only leptin levels were different between female and male rats. In conclusion, pro-inflammatory cytokines initiate the inflammatory process and return to baseline within hours post-injury, chemokines continue to recruit immune cells for days post-injury, while anti-inflammatory cytokines are downregulated by a week post-injury, and sexual dimorphisms observed after mild injury subsided with more severe injuries. Results from this work define critical chemokines that influence immune cell infiltration and important cytokines involved in glial scar development after spinal cord injury, which are essential for researchers developing treatments targeting secondary damage after spinal cord injury.
Collapse
Affiliation(s)
- Daniel J. Hellenbrand
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Charles M. Quinn
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Zachariah J. Piper
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Ryan T. Elder
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Raveena R. Mishra
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Taylor L. Marti
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Phoebe M. Omuro
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Rylie M. Roddick
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Forward BIO Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Amgad S. Hanna
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
deKay JT, Chepurko E, Chepurko V, Knudsen L, Lord C, Searight M, Tsibulnikov S, Robich MP, Sawyer DB, Gagnon DJ, May T, Riker R, Seder DB, Ryzhov S. Delayed CCL23 response is associated with poor outcomes after cardiac arrest. Cytokine 2024; 176:156536. [PMID: 38325139 PMCID: PMC10915974 DOI: 10.1016/j.cyto.2024.156536] [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: 09/13/2023] [Revised: 12/10/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
Chemokines, a family of chemotactic cytokines, mediate leukocyte migration to and entrance into inflamed tissue, contributing to the intensity of local inflammation. We performed an analysis of chemokine and immune cell responses to cardiac arrest (CA). Forty-two patients resuscitated from cardiac arrest were analyzed, and twenty-two patients who underwent coronary artery bypass grafting (CABG) surgery were enrolled. Quantitative antibody array, chemokines, and endotoxin quantification were performed using the patients blood. Analysis of CCL23 production in neutrophils obtained from CA patients and injected into immunodeficient mice after CA and cardiopulmonary resuscitation (CPR) were done using flow cytometry. The levels of CCL2, CCL4, and CCL23 are increased in CA patients. Temporal dynamics were different for each chemokine, with early increases in CCL2 and CCL4, followed by a delayed elevation in CCL23 at forty-eight hours after CA. A high level of CCL23 was associated with an increased number of neutrophils, neuron-specific enolase (NSE), worse cerebral performance category (CPC) score, and higher mortality. To investigate the role of neutrophil activation locally in injured brain tissue, we used a mouse model of CA/CPR. CCL23 production was increased in human neutrophils that infiltrated mouse brains compared to those in the peripheral circulation. It is known that an early intense inflammatory response (within hours) is associated with poor outcomes after CA. Our data indicate that late activation of neutrophils in brain tissue may also promote ongoing injury via the production of CCL23 and impair recovery after cardiac arrest.
Collapse
Affiliation(s)
| | | | | | - Lacey Knudsen
- MaineHealth Institue for Research, Scarborough, ME USA
| | - Christine Lord
- Maine Medical Center Department of Critical Care Services, Portland, ME, USA
| | - Meghan Searight
- Maine Medical Center Department of Critical Care Services, Portland, ME, USA
| | | | | | | | - David J Gagnon
- MaineHealth Institue for Research, Scarborough, ME USA; MaineHealth Department of Pharmacy, Portland, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Teresa May
- MaineHealth Institue for Research, Scarborough, ME USA; Maine Medical Center Department of Critical Care Services, Portland, ME, USA
| | - Richard Riker
- MaineHealth Institue for Research, Scarborough, ME USA; Maine Medical Center Department of Critical Care Services, Portland, ME, USA
| | - David B Seder
- MaineHealth Institue for Research, Scarborough, ME USA; Maine Medical Center Department of Critical Care Services, Portland, ME, USA.
| | - Sergey Ryzhov
- MaineHealth Institue for Research, Scarborough, ME USA.
| |
Collapse
|
12
|
Fang S, Tang H, Li HL, Han TC, Li ZJ, Yin ZS, Chu JJ. CCL2 Knockdown Attenuates Inflammatory Response After Spinal Cord Injury Through the PI3K/Akt Signaling Pathway: Bioinformatics Analysis and Experimental Validation. Mol Neurobiol 2024; 61:1433-1447. [PMID: 37721689 DOI: 10.1007/s12035-023-03641-z] [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: 07/28/2022] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
Spinal cord injury (SCI) is a common clinical problem in orthopedics with a lack of effective treatments and drug targets. In the present study, we performed bioinformatic analysis of SCI datasets GSE464 and GSE45006 in the Gene Expression Omnibus (GEO) public database and experimentally validated CCL2 expression in an animal model of SCI. This was followed by stimulation of PC-12 cells using hydrogen peroxide to construct a cellular model of SCI. CCL2 expression was knocked down using small interfering RNA (si-CCL2), and PI3K signaling pathway inhibitors and activators were used to validate and observe the changes in downstream inflammation. Through data mining, we found that the inflammatory chemokine CCL2 and PI3K/Akt signaling pathways after SCI expression were significantly increased, and after peroxide stimulation of PC-12 cells with CCL2 knockdown, their downstream cellular inflammatory factor levels were decreased. The PI3K/Akt signaling pathway was blocked by PI3K inhibitors, and the downstream inflammatory response was suppressed. In contrast, when PI3K activators were used, the inflammatory response was enhanced, indicating that the CCL2-PI3K/Akt signaling pathway plays a key role in the regulation of the inflammatory response. This study revealed that the inflammatory chemokine CCL2 can regulate the inflammatory response of PC-12 cells through the PI3K/Akt signaling pathway, and blocking the expression of the inflammatory chemokine CCL2 may be a promising strategy for the treatment of secondary injury after SCI.
Collapse
Affiliation(s)
- Sheng Fang
- School of Medicine, Lishui University, Lishui, 323000, China
| | - Hao Tang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China
| | - Hai-Long Li
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China
| | - Ti-Chao Han
- Department of Orthopedics, The Linquan County People's Hospital, 109 Tong Yang Road, Fuyang, Anhui Province, 236400, People's Republic of China
| | - Zi-Jie Li
- Department of Anesthesiology, The Linquan County People's Hospital, 109 Tong Yang Road, Fuyang, Anhui Province, 236400, People's Republic of China
| | - Zong-Sheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, China.
| | - Jian-Jun Chu
- Department of Orthopedics, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, China.
| |
Collapse
|
13
|
Ryan F, Francos-Quijorna I, Hernández-Mir G, Aquino C, Schlapbach R, Bradbury EJ, David S. Tlr4 Deletion Modulates Cytokine and Extracellular Matrix Expression in Chronic Spinal Cord Injury, Leading to Improved Secondary Damage and Functional Recovery. J Neurosci 2024; 44:e0778232023. [PMID: 38326029 PMCID: PMC10860514 DOI: 10.1523/jneurosci.0778-23.2023] [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: 04/30/2023] [Revised: 11/03/2023] [Accepted: 11/12/2023] [Indexed: 02/09/2024] Open
Abstract
Toll-like receptors (TLRs) play an important role in the innate immune response after CNS injury. Although TLR4 is one of the best characterized, its role in chronic stages after spinal cord injury (SCI) is not well understood. We examined the role of TLR4 signaling in injury-induced responses at 1 d, 7 d, and 8 weeks after spinal cord contusion injury in adult female TLR4 null and wild-type mice. Analyses include secondary damage, a range of transcriptome and protein analyses of inflammatory, cell death, and extracellular matrix (ECM) molecules, as well as immune cell infiltration and changes in axonal sprouting and locomotor recovery. Lack of TLR4 signaling results in reduced neuronal and myelin loss, reduced activation of NFκB, and decreased expression of inflammatory cytokines and necroptotic cell death pathway at a late time point (8 weeks) after injury. TLR4 null mice also showed reduction of scar-related ECM molecules at 8 weeks after SCI, accompanied by increase in ECM molecules associated with perineuronal nets, increased sprouting of serotonergic fibers, and improved locomotor recovery. These findings reveal novel effects of TLR4 signaling in chronic SCI. We show that TLR4 influences inflammation, cell death, and ECM deposition at late-stage post-injury when secondary injury processes are normally considered to be over. This highlights the potential for late-stage targeting of TLR4 as a potential therapy for chronic SCI.
Collapse
Affiliation(s)
- Fari Ryan
- Centre for Research in Neuroscience and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec H3G 1A4, Canada
| | - Isaac Francos-Quijorna
- The Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - Gerard Hernández-Mir
- Centre for Immunobiology, Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London E1 2AT, United Kingdom
| | - Catharine Aquino
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich 8057, Switzerland
| | - Ralph Schlapbach
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich 8057, Switzerland
| | - Elizabeth J Bradbury
- The Wolfson Centre for Age-Related Diseases, King's College London, London SE1 1UL, United Kingdom
| | - Samuel David
- Centre for Research in Neuroscience and BRaIN Program, Research Institute of the McGill University Health Centre, Montreal, Quebec H3G 1A4, Canada
| |
Collapse
|
14
|
Rademeyer KM, R Nass S, Jones AM, Ohene-Nyako M, Hauser KF, McRae M. Fentanyl dysregulates neuroinflammation and disrupts blood-brain barrier integrity in HIV-1 Tat transgenic mice. J Neurovirol 2024; 30:1-21. [PMID: 38280928 PMCID: PMC11232468 DOI: 10.1007/s13365-023-01186-4] [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: 06/17/2023] [Revised: 10/29/2023] [Accepted: 11/16/2023] [Indexed: 01/29/2024]
Abstract
Opioid overdose deaths have dramatically increased by 781% from 1999 to 2021. In the setting of HIV, opioid drug abuse exacerbates neurotoxic effects of HIV in the brain, as opioids enhance viral replication, promote neuronal dysfunction and injury, and dysregulate an already compromised inflammatory response. Despite the rise in fentanyl abuse and the close association between opioid abuse and HIV infection, the interactive comorbidity between fentanyl abuse and HIV has yet to be examined in vivo. The HIV-1 Tat-transgenic mouse model was used to understand the interactive effects between fentanyl and HIV. Tat is an essential protein produced during HIV that drives the transcription of new virions and exerts neurotoxic effects within the brain. The Tat-transgenic mouse model uses a glial fibrillary acidic protein (GFAP)-driven tetracycline promoter which limits Tat production to the brain and this model is well used for examining mechanisms related to neuroHIV. After 7 days of fentanyl exposure, brains were harvested. Tight junction proteins, the vascular cell adhesion molecule, and platelet-derived growth factor receptor-β were measured to examine the integrity of the blood brain barrier. The immune response was assessed using a mouse-specific multiplex chemokine assay. For the first time in vivo, we demonstrate that fentanyl by itself can severely disrupt the blood-brain barrier and dysregulate the immune response. In addition, we reveal associations between inflammatory markers and tight junction proteins at the blood-brain barrier.
Collapse
Affiliation(s)
- Kara M Rademeyer
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Sara R Nass
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Austin M Jones
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University, Richmond, VA, 23298, U.S.A
| | - Michael Ohene-Nyako
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - Kurt F Hauser
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, U.S.A
| | - MaryPeace McRae
- Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA, 22908, U.S.A..
| |
Collapse
|
15
|
Yari D, Saberi A, Salmasi Z, Ghoreishi SA, Etemad L, Movaffagh J, Ganjeifar B. Recent Advances in the Treatment of Spinal Cord Injury. THE ARCHIVES OF BONE AND JOINT SURGERY 2024; 12:380-399. [PMID: 38919744 PMCID: PMC11195032 DOI: 10.22038/abjs.2023.73944.3424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/07/2023] [Indexed: 06/27/2024]
Abstract
Spinal cord injury (SCI) is a complex, multifaceted, progressive, and yet incurable complication that can cause irreversible damage to the individual, family, and society. In recent years strategies for the management and rehabilitation of SCI besides axonal regeneration, remyelination, and neuronal plasticity of the injured spinal cord have significantly improved. Although most of the current research and therapeutic advances have been made in animal models, so far, no specific and complete treatment has been reported for SCI in humans. The failure to treat this complication has been due to the inherent neurological complexity and the structural, cellular, molecular, and biochemical characteristics of spinal cord injury. In this review, in addition to elucidating the causes of spinal cord injury from a molecular and pathophysiological perspective, the complexity and drawbacks of neural regeneration that lead to the failure in SCI treatment are described. Also, recent advances and cutting-edge strategies in most areas of SCI treatment are presented.
Collapse
Affiliation(s)
- Davood Yari
- Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Salmasi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Alireza Ghoreishi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Orthopedic Surgery, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jebrail Movaffagh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Babak Ganjeifar
- Department of Neurosurgery, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
16
|
Haro S, Gomez-Lahoz AM, Monserrat J, Atienza-Pérez M, Fraile-Martinez O, Ortega MA, García-Montero C, Díaz D, Lopez-Dolado E, Álvarez-Mon M. Patients with Chronic Spinal Cord Injury Display a Progressive Alteration over the Years of the Activation Stages of the T Lymphocyte Compartment. Int J Mol Sci 2023; 24:17596. [PMID: 38139422 PMCID: PMC10744286 DOI: 10.3390/ijms242417596] [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: 11/13/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023] Open
Abstract
Spinal cord injury (SCI) is a serious medical condition associated with severe morbidities and disability. Chronic SCI patients present an enhanced susceptibility to infections and comorbidities with inflammatory pathogenesis. Chronic SCI appears to be associated with a systemic dysfunction of the immune system. We investigated the alteration of the pivotal CD4+ and CD8+ T lymphocytes in patients with chronic SCI at different years of evolution. A clinically homogenous population of 105 patients with chronic SCI (31 with time of evolution less than 5 years (SCI SP); 32 early chronic (SCI ECP) with time of evolution between 5 and 15 years; and 42 late chronic (SCI LCP) with time of evolution more than 15 years) and 38 healthy controls were enrolled. SCI ECP and SCI LCP patients showed significant CD4+ and CD8+ T lymphopenia, ascribed to a reduction in naïve and CM subsets. Furthermore, SCI ECP and SCI LCP patients showed a significant reduction in the expression of CD28 on CD8+ T lymphocytes. The expression of CCR6 by CD4+ T lymphocytes was decreased during the evolution of chronic SCI, but on CD8+ T lymphocytes, it was observed during the first 15 years of evolution. In conclusion, the chronic SCI course with severe damage to T lymphocytes mainly worsens over the years of disease evolution.
Collapse
Affiliation(s)
- Sergio Haro
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Ana M. Gomez-Lahoz
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Mar Atienza-Pérez
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain;
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - David Díaz
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Elisa Lopez-Dolado
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Service of Rehabilitation, National Hospital for Paraplegic Patients, Carr. de la Peraleda, S/N, 45004 Toledo, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Crta N-II km 33.6, 28871 Alcala de Henares, Spain; (S.H.); (A.M.G.-L.); (J.M.); (O.F.-M.); (M.A.O.); (C.G.-M.); (D.D.); (E.L.-D.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
- Immune System Diseases-Rheumatology Service and Internal Medicine, Prince of Asturias University Hospital (CIBEREHD), 28806 Alcala de Henares, Spain
| |
Collapse
|
17
|
Kitade K, Kobayakawa K, Saiwai H, Matsumoto Y, Kawaguchi K, Iida K, Kijima K, Iura H, Tamaru T, Haruta Y, Ono G, Konno D, Maeda T, Okada S, Nakashima K, Nakashima Y. Reduced Neuroinflammation Via Astrocytes and Neutrophils Promotes Regeneration After Spinal Cord Injury in Neonatal Mice. J Neurotrauma 2023; 40:2566-2579. [PMID: 37503626 DOI: 10.1089/neu.2023.0044] [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: 07/29/2023] Open
Abstract
Neonatal spinal cord injury (SCI) shows better functional outcomes than adult SCI. Although the regenerative capability in the neonatal spinal cord may have cues in the treatment of adult SCI, the mechanism underlying neonatal spinal cord regeneration after SCI is unclear. We previously reported age-dependent variation in the pathogenesis of inflammation after SCI. Therefore, we explored differences in the pathogenesis of inflammation after SCI between neonatal and adult mice and their effect on axon regeneration and functional outcome. We established two-day-old spinal cord crush mice as a model of neonatal SCI. Immunohistochemistry of the spinal cord revealed that the nuclear translocation of NF-κB, which promotes the expression of chemokines, was significantly lower in the astrocytes of neonates than in those of adults. Flow cytometry revealed that neonatal astrocytes secrete low levels of chemokines to recruit circulating neutrophils (e.g., Cxcl1 and Cxcl2) after SCI in comparison with adults. We also found that the expression of a chemokine receptor (CXCR2) and an adhesion molecule (β2 integrin) quantified by flow cytometry was lower in neonatal circulating neutrophils than in adult neutrophils. Strikingly, these neonate-specific cellular properties seemed to be associated with no neutrophil infiltration into the injured spinal cord, followed by significantly lower expression of inflammatory cytokines (Il-1β, Il-6 and TNF-α) after SCI in the spinal cords of neonates than in those of adults. At the same time, significantly fewer apoptotic neurons and greater axonal regeneration were observed in neonates in comparison with adults, which led to a marked recovery of locomotor function. This neonate-specific mechanism of inflammation regulation may have potential therapeutic applications in controlling inflammation after adult SCI.
Collapse
Affiliation(s)
- Kazuki Kitade
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirokazu Saiwai
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keiichiro Iida
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kijima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Iura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Tamaru
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yohei Haruta
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gentaro Ono
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daijiro Konno
- Department of Energy and Materials, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Takeshi Maeda
- Department of Orthopedic Surgery, Spinal Injuries Center, Iizuka, Japan
| | - Seiji Okada
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Osaka University, Osaka, Japan
| | - Kinichi Nakashima
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
18
|
Balog BM, Sonti A, Zigmond RE. Neutrophil biology in injuries and diseases of the central and peripheral nervous systems. Prog Neurobiol 2023; 228:102488. [PMID: 37355220 PMCID: PMC10528432 DOI: 10.1016/j.pneurobio.2023.102488] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/24/2023] [Accepted: 06/16/2023] [Indexed: 06/26/2023]
Abstract
The role of inflammation in nervous system injury and disease is attracting increased attention. Much of that research has focused on microglia in the central nervous system (CNS) and macrophages in the peripheral nervous system (PNS). Much less attention has been paid to the roles played by neutrophils. Neutrophils are part of the granulocyte subtype of myeloid cells. These cells, like macrophages, originate and differentiate in the bone marrow from which they enter the circulation. After tissue damage or infection, neutrophils are the first immune cells to infiltrate into tissues and are directed there by specific chemokines, which act on chemokine receptors on neutrophils. We have reviewed here the basic biology of these cells, including their differentiation, the types of granules they contain, the chemokines that act on them, the subpopulations of neutrophils that exist, and their functions. We also discuss tools available for identification and further study of neutrophils. We then turn to a review of what is known about the role of neutrophils in CNS and PNS diseases and injury, including stroke, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, spinal cord and traumatic brain injuries, CNS and PNS axon regeneration, and neuropathic pain. While in the past studies have focused on neutrophils deleterious effects, we will highlight new findings about their benefits. Studies on their actions should lead to identification of ways to modify neutrophil effects to improve health.
Collapse
Affiliation(s)
- Brian M Balog
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Anisha Sonti
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
19
|
Asveda T, Priti T, Ravanan P. Exploring microglia and their phenomenal concatenation of stress responses in neurodegenerative disorders. Life Sci 2023:121920. [PMID: 37429415 DOI: 10.1016/j.lfs.2023.121920] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023]
Abstract
Neuronal cells are highly functioning but also extremely stress-sensitive cells. By defending the neuronal cells against pathogenic insults, microglial cells, a unique cell type, act as the frontline cavalry in the central nervous system (CNS). Their remarkable and unique ability to self-renew independently after their creation is crucial for maintaining normal brain function and neuroprotection. They have a wide range of molecular sensors that help maintain CNS homeostasis during development and adulthood. Despite being the protector of the CNS, studies have revealed that persistent microglial activation may be the root cause of innumerable neurodegenerative illnesses, including Alzheimer's disease (AD), Parkinson's disease (PD), and Amyloid Lateral Sclerosis (ALS). From our vigorous review, we state that there is a possible interlinking between pathways of Endoplasmic reticulum (ER) stress response, inflammation, and oxidative stress resulting in dysregulation of the microglial population, directly influencing the accumulation of pro-inflammatory cytokines, complement factors, free radicals, and nitric oxides leading to cell death via apoptosis. Recent research uses the suppression of these three pathways as a therapeutic approach to prevent neuronal death. Hence, in this review, we have spotlighted the advancement in microglial studies, which focus on their molecular defenses against multiple stresses, and current therapeutic strategies indirectly targeting glial cells for neurodevelopmental diseases.
Collapse
Affiliation(s)
- Thankavelu Asveda
- Functional Genomics Laboratory, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, Tamil Nadu, India
| | - Talwar Priti
- Apoptosis and Cell Survival Research Laboratory, 412G Pearl Research Park, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| | - Palaniyandi Ravanan
- Functional Genomics Laboratory, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, Tamil Nadu, India.
| |
Collapse
|
20
|
Kalhori MR, Soleimani M, Alibakhshi R, Kalhori AA, Mohamadi P, Azreh R, Farzaei MH. The Potential of miR-21 in Stem Cell Differentiation and its Application in Tissue Engineering and Regenerative Medicine. Stem Cell Rev Rep 2023; 19:1232-1251. [PMID: 36899116 DOI: 10.1007/s12015-023-10510-8] [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] [Accepted: 01/18/2023] [Indexed: 03/12/2023]
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are two important types of non-coding RNAs that are not translated into protein. These molecules can regulate various biological processes, including stem cell differentiation and self-renewal. One of the first known miRNAs in mammals is miR-21. Cancer-related studies have shown that this miRNA has proto-oncogene activity and is elevated in cancers. However, it is confirmed that miR-21 inhibits stem cell pluripotency and self-renewal and induces differentiation by targeting various genes. Regenerative medicine is a field of medical science that tries to regenerate and repair damaged tissues. Various studies have shown that miR-21 plays an essential role in regenerative medicine by affecting stem cell proliferation and differentiation. In this review, we will discuss the function of miR-21 in regenerative medicine of the liver, nerve, spinal cord, wound, bone, and dental tissues. In addition, the function of natural compounds and lncRNAs will be analyzed as potential regulators of miR-21 expression in regenerative medicine.
Collapse
Affiliation(s)
- Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Alibakhshi
- Department of Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Ali Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mohamadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical, Sciences, Tehran, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rasoul Azreh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hosien Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
21
|
Shafqat A, Albalkhi I, Magableh HM, Saleh T, Alkattan K, Yaqinuddin A. Tackling the glial scar in spinal cord regeneration: new discoveries and future directions. Front Cell Neurosci 2023; 17:1180825. [PMID: 37293626 PMCID: PMC10244598 DOI: 10.3389/fncel.2023.1180825] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Axonal regeneration and functional recovery are poor after spinal cord injury (SCI), typified by the formation of an injury scar. While this scar was traditionally believed to be primarily responsible for axonal regeneration failure, current knowledge takes a more holistic approach that considers the intrinsic growth capacity of axons. Targeting the SCI scar has also not reproducibly yielded nearly the same efficacy in animal models compared to these neuron-directed approaches. These results suggest that the major reason behind central nervous system (CNS) regeneration failure is not the injury scar but a failure to stimulate axon growth adequately. These findings raise questions about whether targeting neuroinflammation and glial scarring still constitute viable translational avenues. We provide a comprehensive review of the dual role of neuroinflammation and scarring after SCI and how future research can produce therapeutic strategies targeting the hurdles to axonal regeneration posed by these processes without compromising neuroprotection.
Collapse
|
22
|
Oprişoreanu AM, Ryan F, Richmond C, Dzekhtsiarova Y, Carragher NO, Becker T, David S, Becker CG. Drug screening in zebrafish larvae reveals inflammation-related modulators of secondary damage after spinal cord injury in mice. Theranostics 2023; 13:2531-2551. [PMID: 37215570 PMCID: PMC10196818 DOI: 10.7150/thno.81332] [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: 11/29/2022] [Accepted: 04/11/2023] [Indexed: 05/24/2023] Open
Abstract
Prolonged inflammation after spinal cord injury is detrimental to recovery. To find pharmacological modulators of the inflammation response, we designed a rapid drug screening paradigm in larval zebrafish followed by testing of hit compounds in a mouse spinal cord injury model. Methods: We used reduced il-1β linked green fluorescent protein (GFP) reporter gene expression as a read-out for reduced inflammation in a screen of 1081 compounds in larval zebrafish. Hit drugs were tested in a moderate contusion model in mice for cytokine regulation, and improved tissue preservation and locomotor recovery. Results: Three compounds robustly reduced il-1β expression in zebrafish. Cimetidine, an over-the-counter H2 receptor antagonist, also reduced the number of pro-inflammatory neutrophils and rescued recovery after injury in a zebrafish mutant with prolonged inflammation. Cimetidine action on il-1β expression levels was abolished by somatic mutation of H2 receptor hrh2b, suggesting specific action. In mice, systemic treatment with Cimetidine led to significantly improved recovery of locomotor behavior as compared to controls, accompanied by decreased neuronal tissue loss and a shift towards a pro-regenerative profile of cytokine gene expression. Conclusion: Our screen revealed H2 receptor signaling as a promising target for future therapeutic interventions in spinal cord injury. This work highlights the usefulness of the zebrafish model for rapid screening of drug libraries to identify therapeutics to treat mammalian spinal cord injury.
Collapse
Affiliation(s)
- Ana-Maria Oprişoreanu
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Fari Ryan
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec, H3G 1A4
| | - Claire Richmond
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Yuliya Dzekhtsiarova
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Neil O. Carragher
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Samuel David
- Centre for Research in Neuroscience, Research Institute of the McGill University Health Centre, 1650 Cedar Ave., Montreal, Quebec, H3G 1A4
| | - Catherina G. Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, The Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
- Center for Regenerative Therapies Dresden, TU Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
23
|
Lee CY, Chooi WH, Ng S, Chew SY. Modulating neuroinflammation through molecular, cellular and biomaterial-based approaches to treat spinal cord injury. Bioeng Transl Med 2023; 8:e10389. [PMID: 36925680 PMCID: PMC10013833 DOI: 10.1002/btm2.10389] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/02/2022] [Accepted: 07/16/2022] [Indexed: 11/09/2022] Open
Abstract
The neuroinflammatory response that is elicited after spinal cord injury contributes to both tissue damage and reparative processes. The complex and dynamic cellular and molecular changes within the spinal cord microenvironment result in a functional imbalance of immune cells and their modulatory factors. To facilitate wound healing and repair, it is necessary to manipulate the immunological pathways during neuroinflammation to achieve successful therapeutic interventions. In this review, recent advancements and fresh perspectives on the consequences of neuroinflammation after SCI and modulation of the inflammatory responses through the use of molecular-, cellular-, and biomaterial-based therapies to promote tissue regeneration and functional recovery will be discussed.
Collapse
Affiliation(s)
- Cheryl Yi‐Pin Lee
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Shi‐Yan Ng
- Institute of Molecular and Cell BiologyA*STAR Research EntitiesSingaporeSingapore
| | - Sing Yian Chew
- School of Chemical and Biomedical EngineeringNanyang Technological UniversitySingaporeSingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
- School of Materials Science and EngineeringNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
24
|
Drake SS, Zaman A, Simas T, Fournier AE. Comparing RNA-sequencing datasets from astrocytes, oligodendrocytes, and microglia in multiple sclerosis identifies novel dysregulated genes relevant to inflammation and myelination. WIREs Mech Dis 2023; 15:e1594. [PMID: 36600404 DOI: 10.1002/wsbm.1594] [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: 06/20/2022] [Revised: 09/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
Central nervous system (CNS) inflammation is a key factor in multiple sclerosis (MS). Invasion of peripheral immune cells into the CNS resulting from an unknown signal or combination of signals results in activation of resident immune cells and the hallmark feature of the disease: demyelinating lesions. These lesion sites are an amalgam of reactive peripheral and central immune cells, astrocytes, damaged and dying oligodendrocytes, and injured neurons and axons. Sustained inflammation affects cells directly located within the lesion site and further abnormalities are apparent diffusely throughout normal-appearing white matter and grey matter. It is only relatively recently, using animal models, new tissue sampling techniques, and next-generation sequencing, that molecular changes occurring in CNS resident cells have been broadly captured. Advances in cell isolation through Fluorescence Activated Cell Sorting (FACS) and laser-capture microdissection together with the emergence of single-cell sequencing have enabled researchers to investigate changes in gene expression in astrocytes, microglia, and oligodendrocytes derived from animal models of MS as well as from primary patient tissue. The contribution of some dysregulated pathways has been followed up in individual studies; however, corroborating results often go unreported between sequencing studies. To this end, we have consolidated results from numerous RNA-sequencing studies to identify and review novel patterns of differentially regulated genes and pathways occurring within CNS glial cells in MS. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Sienna S Drake
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Aliyah Zaman
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Tristan Simas
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Alyson E Fournier
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Li S, Wu C, Lin S, Wen Z, Luo W, Li C, Wang X, Li X, Gao L, Ding Y. HUCMSC-derived Exosomes Suppress the Titanium Particles-induced Osteolysis in Mice through Inhibiting CCL2 and CCL3. Orthop Surg 2023; 15:888-898. [PMID: 36720704 PMCID: PMC9977603 DOI: 10.1111/os.13608] [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: 01/19/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 02/02/2023] Open
Abstract
OBJECTIVE Wear particles induce inflammation and the further osteolysis around the prosthesis, has been proven to be the main cause of aseptic hip joint loosening. In this research, we aimed to clarify whether human umbilical cord mesenchymal stem cells (HUCMSCs) could inhibit the titanium particles-induced osteolysis and shed light upon its mechanism. METHODS The expression of chemokine (C-C motif) ligand 2 (CCL2), chemokine (C-C motif) ligand 3 (CCL3) and chemokine (C-C motif) ligand 5 (CCL5) were examinjed in clinical specimens of aseptic hip prosthesis loosening patients. Local injection of lentivirus that knocked down CCL2 or CCL3 in a cranial osteolysis mice model were used to exam the effect of CCL2 and CCL3 on titanium particles-induced osteolysis in vivo. Transwell assay was used to examine the effect of CCL2 and CCL3 on titanium particles-induced activation of macrophage in vitro. Furthermore, the therapeutic effect of HUCMSCs, and exosomes from HUCMSCs were also examed in vivo and vitro. Immunohistochemical and real-time PCR were used to examine the expression of relative pathways. Analysis of variance (ANOVA) and Student-Newman-Keuls post hoc t test were used to analyze the results and determine the statistical significance of the differences. RESULTS Results showed that titanium particles caused the osteolysis at the mice cranial in vivo and a large number of macrophages that migrated, while local injection of HUCMSCs and exosomes did inhibit the cranial osteolysis and migration. An exosome inhibitor GW4869 significantly increased the osteolysis area in the mice cranium osteolysis model, and increased the number of migrated macrophages. Immunohistochemical results suggested that the expression of CCL2, CCL3 and CD68 in the cranial in Titanium particles mice increased significantly, but was significantly reduced by HUCMSCs or exosomes. HUCMSC and exosomes down-regulate the expression of CCL3 in vitro and in vivo. CONCLUSION HUCMSCs and HUCMSC-derived exosomes could suppress the titanium particles-induced osteolysis in mice through inhibiting chemokine (C-C motif) ligand 2, chemokine (C-C motif) ligand 3.
Collapse
Affiliation(s)
- Shixun Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Chuangran Wu
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Sipeng Lin
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Zhenkang Wen
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Wenqiang Luo
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Changchuan Li
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Xiaoyan Wang
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Xuejia Li
- Guangzhou Saliai Stem Cell Science and Technology Co., LTDGuangzhouChina
| | - Liangbin Gao
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Yue Ding
- Department of Orthopaedic SurgerySun Yat‐Sen Memorial Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
26
|
Expression Profiles of Long Noncoding RNAs and Messenger RNAs in a Rat Model of Spinal Cord Injury. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2023; 2023:6033020. [PMID: 36714328 PMCID: PMC9879695 DOI: 10.1155/2023/6033020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/20/2023]
Abstract
Spinal cord injury (SCI) is a serious disorder of the central nervous system with a high disability rate. Long noncoding RNAs (lncRNAs) are reported to mediate many biological processes. The aim of this study was to explore lncRNA and mRNA expression profiles and functional networks after SCI. Differentially expressed genes between SCI model rats and sham controls were identified by microarray assays and analyzed by functional enrichment. Key lncRNAs were identified using a support vector machine- (SVM-) recursive feature elimination (RFE) algorithm. A trans and cis regulation model was used to analyze the regulatory relationships between lncRNAs and their targets. An lncRNA-related ceRNA network was established. We identified 5465 differentially expressed lncRNAs (DE lncRNAs) and 8366 differentially expressed mRNAs (DE mRNAs) in the SCI group compared with the sham group (fold change > 2.0, p < 0.05). Four genes were confirmed by qRT-PCR which were consistent with the microarray data. GSEA analysis showed that most marked changes occurred in pathways related to immune inflammation and nerve cell function, including cytokine-cytokine receptor interaction, neuroactive ligand-receptor interaction, and GABAergic synapse. Enrichment analysis identified 30 signaling pathways, including those associated with immune inflammation response. A total of 40 key lncRNAs were identified using the SVM-RFE algorithm. A key lncRNA-mRNAs coexpression network was generated for 230 951 lncRNA-mRNA pairs with half showing positive correlations. Several key DE lncRNAs were predicted to have "cis"- or "trans"-regulated target genes. The transcription factors, Sp1, JUN, and SOX10, may regulate the interaction between XR_001837123.1 and ETS 1. In addition, five pairs of ceRNA regulatory sequences were constructed. Many mRNAs and lncRNAs were found to be dysregulated after SCI. Bioinformatic analysis showed that DE lncRNAs may play crucial roles in SCI. It is anticipated that these findings will provide new insights into the underlying mechanisms and potential therapeutic targets for SCI.
Collapse
|
27
|
Guan B, Li H, Yao J, Guo J, Yu F, Li G, Wan B, Ma J, Huang D, Sun L, Chen Y. CCL3-CCR5 axis promotes cell migration and invasion of colon adenocarcinoma via Akt signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:172-184. [PMID: 36346222 DOI: 10.1002/tox.23675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/19/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Infiltration of tumor-associated macrophages (TAMs) can promote tumorigenesis and development. C-C motif chemokine ligand 3 (CCL3) was reported to be derived from TAMs and tumor cells and facilitate the progression of several cancers. Nevertheless, whether CCL3 can be derived from TAMs and tumor cells of colon adenocarcinoma (COAD) is unclarified. METHODS Peripheral blood monocytes-derived macrophages were polarized by the conditioned medium from COAD cells to establish TAM-like macrophages (TAM1/2). RT-qPCR and western blotting were used for detection of expression levels of CCL3 and its receptors C-C motif chemokine receptor 1 (CCR1) and CCR5 in TAM1/2 and COAD cells. Immunofluorescence staining was utilized for evaluating CCL3, CD163 and CCR5 expression. The Akt signaling pathway-associated protein levels were measured by western blotting. Transwell assays were used for assessing cell migration and invasiveness. RESULTS CCL3 displayed a high level in TAMs and cancer cells of COAD. CCL3 activated the Akt signaling pathway by binding to CCR5. CCL3-CCR5 axis facilitated COAD cell migration and invasiveness by activating the Akt signaling. CCL3 derived from both TAMs and cancer cells contributed to the malignant behaviors of COAD cells. High expression of CCL3/CCR5 was closely associated with poor prognoses of COAD patients. CONCLUSION CCL3-CCR5 interaction promotes cell migration and invasiveness, and functions as a prognostic biomarker for COAD.
Collapse
Affiliation(s)
- Bugao Guan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Hongbo Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jian Yao
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jinbao Guo
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Fei Yu
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Guangrun Li
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Benhai Wan
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Jun Ma
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Desong Huang
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Lu Sun
- Department of General Surgery, Jinhu People's Hospital, Huai'an, China
| | - Yan Chen
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| |
Collapse
|
28
|
CCL3 aggravates intestinal damage in NEC by promoting macrophage chemotaxis and M1 macrophage polarization. Pediatr Res 2022:10.1038/s41390-022-02409-w. [PMID: 36550354 DOI: 10.1038/s41390-022-02409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 11/04/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND NEC is a life-threatening gastrointestinal disease in neonates, the pathogenesis of which remains poorly understood. METHODS CCL3 levels in intestinal tissue of mice were measured and analyzed. HE staining was used to assess pathological changes in intestinal tissue. FCM was used to detect the proportion and phenotype of macrophages. RNA-seq and RT-PCR were used to evaluate the effect of CCL3 on macrophages. RESULTS CCL3 was highly expressed in the intestinal tissues of mice with NEC and induced macrophage infiltration. Transcriptome data showed that CCL3 strongly induced a transition in the phenotype of macrophages into a proinflammatory one. Mechanistically, in vivo experiments confirmed that CCL3 induced M1 macrophage polarization in NEC intestinal tissue, thereby aggravating inflammatory injury of intestinal tissue, which was alleviated by anti-CCL3 treatment. In addition, in vitro experiments showed that CCL3 significantly enhances the expression of M1-related genes in both PMφ and BMDM while inhibiting the expression of M2-related genes, which was also alleviated by anti-CCl3 treatment. CONCLUSIONS Our data elucidated the involvement of CCL3 in the pathogenesis of NEC, in which upregulated CCL3 expression exacerbated inflammatory intestinal damage by regulating macrophage chemotaxis and M1 phenotype polarization, suggesting that blocking CCL3 may be a potential strategy for effective intervention in NEC. IMPACT Our study represents an important conceptual advancement that CCL3 may be one of the key culprits of intestinal tissue damage in patients with NEC. CCL3 aggravates inflammatory intestinal injury and intestinal mucosal barrier imbalance by regulating the chemotaxis, polarization, and function of macrophages. Blocking CCL3 significantly reduced NEC-mediated intestinal injury, suggesting a new potential therapeutic strategy.
Collapse
|
29
|
Transplantation of olfactory ensheathing cells decreases local and serological monocyte chemoattractant protein 1 level during the acute phase of rat spinal cord injury. Neuroreport 2022; 33:729-741. [PMID: 36250430 DOI: 10.1097/wnr.0000000000001839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Monocyte chemoattractant protein 1 (MCP1) is one of the most upregulated cytokines in the spinal cord and serum throughout acute spinal cord injury (SCI). Olfactory ensheathing cells (OECs) transplantation improves SCI through multiple mechanisms, including immunomodulation. Our study aimed to investigate whether OECs ameliorate acute inflammation after SCI by modulating MCP1 expression. METHODS We established a standardized clinically relevant contusion model using the NYU impactor. OECs were administered to the injured spinal cord via microinjection 30 minutes after injury. Rat locomotor functions were assessed by the Basso-Beattie-Bresnahan scale score. Time-course histopathological (H&E and IHC) analyses were performed to record rapid changes in acute inflammation at lesion epicenters. Serum MCP1 level was detected by ELISA assay. RESULTS BBB scores showed improved locomotor functional recoveries in the OECs transplantation group after SCI ( P < 0.05). Staining of H&E and CD68 illustrated that OECs transplantation attenuated inflammatory response by reducing lesion areas and infiltrating myeloid cell numbers. We further revealed significantly decreased MCP1 levels in the spinal cord and serum after OECs transplantation ( P < 0.05). Noteworthily, distinct expression levels of MCP1 were found in rats undergoing a mild injury (cord impacted from a 10-mm height) compared to the moderate injury (25-mm) group. CONCLUSION Our study reports that transplantation of OECs promotes locomotor functional recovery after SCI and alleviates acute inflammation by decreasing local and serological MCP1 levels. We provide preliminary evidence that MCP1 might serve as a potential biomarker to reflect the severity of SCI, which is of great interest in future studies.
Collapse
|
30
|
Xia M, Li X, Ye S, Zhang Q, Zhao T, Li R, Zhang Y, Xian M, Li T, Li H, Hong X, Zheng S, Qian Z, Yang L. FANCC deficiency mediates microglial pyroptosis and secondary neuronal apoptosis in spinal cord contusion. Cell Biosci 2022; 12:82. [PMID: 35659106 PMCID: PMC9164466 DOI: 10.1186/s13578-022-00816-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/15/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Traumatic spinal cord injury (SCI)-induced neuroinflammation results in secondary neurological destruction and functional disorder. Previous findings showed that microglial pyroptosis plays a crucial role in neuroinflammation. Thus, it is necessary to conduct a comprehensive investigation of the mechanisms associated with post-SCI microglial pyroptosis. The Fanconi Anemia Group C complementation group gene (FANCC) has been previously reported to have an anti-inflammation effect; however, whether it can regulate microglial pyroptosis remains unknown. Therefore, we probed the mechanism associated with FANCC-mediated microglial pyroptosis and neuroinflammation in vitro and in vivo in SCI mice.
Methods
Microglial pyroptosis was assessed by western blotting (WB) and immunofluorescence (IF), whereas microglial-induced neuroinflammation was evaluated by WB, Enzyme-linked immunosorbent assays and IF. Besides, flow cytometry, TdT-mediated dUTP Nick-End Labeling staining and WB were employed to examine the level of neuronal apoptosis. Morphological changes in neurons were assessed by hematoxylin–eosin and Luxol Fast Blue staining. Finally, locomotor function rehabilitation was analyzed using the Basso Mouse Scale and Louisville Swim Scale.
Results
Overexpression of FANCC suppressed microglial pyroptosis via inhibiting p38/NLRP3 expression, which in turn reduced neuronal apoptosis. By contrast, knockdown of FANCC increased the degree of neuronal apoptosis by aggravating microglial pyroptosis. Besides, increased glial scar formation, severe myelin sheath destruction and poor axon outgrowth were observed in the mice transfected with short hairpin RNA of FANCC post SCI, which caused reduced locomotor function recovery.
Conclusions
Taken together, a previously unknown role of FANCC was identified in SCI, where its deficiency led to microglia pyroptosis, neuronal apoptosis and neurological damage. Mechanistically, FANCC mediated microglia pyroptosis and the inflammatory response via regulating the p38/NLRP3 pathway.
Collapse
|
31
|
Single-Cell Sequencing Reveals the Regulatory Role of Maresin1 on Neutrophils during Septic Lung Injury. Cells 2022; 11:cells11233733. [PMID: 36496993 PMCID: PMC9739442 DOI: 10.3390/cells11233733] [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: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Acute lung injury (ALI) is the most common type of organ injury in sepsis, with high morbidity and mortality. Sepsis is characterized by an inappropriate inflammatory response while neutrophils exert an important role in the excessive inflammatory response. The discovery of specialized pro-resolving mediators (SPMs) provides a new direction for the treatment of a series of inflammatory-related diseases including sepsis. Among them, the regulation of Maresin1 on immune cells was widely demonstrated. However, current research on the regulatory effects of Maresin1 on immune cells has remained at the level of certain cell types. Under inflammatory conditions, the immune environment is complex and immune cells exhibit obvious heterogeneity. Neutrophils play a key role in the occurrence and development of septic lung injury. Whether there is a subpopulation bias in the regulation of neutrophils by Maresin1 has not been elucidated. Therefore, with the well-established cecal ligation and puncture (CLP) model and single-cell sequencing technology, our study reveals for the first time the regulatory mechanism of Maresin1 on neutrophils at the single-cell level. Our study suggested that Maresin1 can significantly reduce neutrophil infiltration in septic lung injury and that this regulatory effect is more concentrated in the Neutrophil-Cxcl3 subpopulation. Maresin1 can significantly reduce the infiltration of the Neutrophil-Cxcl3 subpopulation and inhibit the expression of related inflammatory genes and key transcription factors in the Neutrophil-Cxcl3 subpopulation. Our study provided new possibilities for specific modulation of neutrophil function in septic lung injury.
Collapse
|
32
|
Fisher ES, Amarante MA, Lowry N, Lotz S, Farjood F, Temple S, Hill CE, Kiehl TR. Single cell profiling of CD45+ spinal cord cells reveals microglial and B cell heterogeneity and crosstalk following spinal cord injury. J Neuroinflammation 2022; 19:266. [PMID: 36333772 PMCID: PMC9635187 DOI: 10.1186/s12974-022-02627-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Abstract
Background
Immune cells play crucial roles after spinal cord injury (SCI). However, incomplete knowledge of immune contributions to injury and repair hinders development of SCI therapies. We leveraged single-cell observations to describe key populations of immune cells present in the spinal cord and changes in their transcriptional profiles from uninjured to subacute and chronic stages of SCI.
Methods
Deep-read single-cell sequencing was performed on CD45+ cells from spinal cords of uninjured and injured Swiss-webster mice. After T9 thoracic contusion, cells were collected 3-, 7-, and 60-day post-injury (dpi). Subpopulations of CD45+ immune cells were identified informatically, and their transcriptional responses characterized with time. We compared gene expression in spinal cord microglia and B cell subpopulations with those in published models of disease and injury. Microglia were compared with Disease Associated Microglia (DAM) and Injury Responsive Microglia (IRM). B cells were compared to developmental lineage states and to an Amyotrophic Lateral Sclerosis (ALS) model.
Results
In uninjured and 7 dpi spinal cord, most CD45+ cells isolated were microglia while chronically B cells predominated. B cells accumulating in the spinal cord following injury included immature B to mature stages and were predominantly found in the injury zone. We defined diverse subtypes of microglia and B cells with altered gene expression with time after SCI. Spinal cord microglia gene expression indicates differences from brain microglia at rest and in inflammatory states. Expression analysis of signaling ligand–receptor partners identified microglia–B cell interactions at acute and chronic stages that may be involved in B cell recruitment, retention, and formation of ectopic lymphoid follicles.
Conclusions
Immune cell responses to SCI have region-specific aspects and evolve with time. Developmentally diverse populations of B cells accumulate in the spinal cord following injury. Microglia at subacute stages express B cell recruitment factors, while chronically, they express factors predicted to reduce B cell inflammatory state. In the injured spinal cord, B cells create ectopic lymphoid structures, and express secreted factors potentially acting on microglia. Our study predicts previously unidentified crosstalk between microglia and B cells post-injury at acute and chronic stages, revealing new potential targets of inflammatory responses for SCI repair warranting future functional analyses.
Collapse
|
33
|
Brianna, Ling APK, Wong YP. Applying stem cell therapy in intractable diseases: a narrative review of decades of progress and challenges. Stem Cell Investig 2022; 9:4. [PMID: 36238449 PMCID: PMC9552054 DOI: 10.21037/sci-2022-021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/09/2022] [Indexed: 08/10/2023]
Abstract
Background and Objective Stem cell therapy (SCT) is one of the vastly researched branches of regenerative medicine as a therapeutic tool to treat incurable diseases. With the use of human stem cells such as embryonic stem cells (ESCs), adult stem cells (ASCs) and induced pluripotent stem cells (iPSCs), stem cell therapy aims to regenerate or repair damaged tissues and congenital defects. As stem cells are able to undergo infinite self-renewal, differentiate into various types of cells and secrete protective paracrine factors, many researchers have investigated the potential of SCT in regenerative medicine. Therefore, this review aims to provide a comprehensive review on the recent application of SCT in various intractable diseases, namely, haematological diseases, neurological diseases, diabetes mellitus, retinal degenerative disorders and COVID-19 infections along with the challenges faced in the clinical translation of SCT. Methods An extensive search was conducted on Google scholar, PubMed and Clinicaltrials.gov using related keywords. Latest articles on stem cell therapy application in selected diseases along with their challenges in clinical applications were selected. Key content and findings In vitro and in vivo studies involving SCT are shown to be safe and efficacious in treating various diseases covered in this review. There are also a number of small-scale clinical trials that validated the positive therapeutic outcomes of SCT. Nevertheless, the effectiveness of SCT are highly variable as some SCT works best in patients with early-stage diseases while in other diseases, SCT is more likely to work in patients in late stages of illnesses. Among the challenges identified in SCT translation are uncertainty in the underlying stem cell mechanism, ethical issues, genetic instability and immune rejection. Conclusions SCT will be a revolutionary treatment in the future that will provide hope to patients with intractable diseases. Therefore, studies ought to be done to ascertain the long-term effects of SCT while addressing the challenges faced in validating SCT for clinical use. Moreover, as there are many studies investigating the safety and efficacy of SCT, future studies should look into elucidating the regenerative and reparative capabilities of stem cells which largely remains unknown.
Collapse
Affiliation(s)
- Brianna
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Ying Pei Wong
- Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Malvandi AM, Rastegar-Moghaddam SH, Ebrahimzadeh-Bideskan S, Lombardi G, Ebrahimzadeh-Bideskan A, Mohammadipour A. Targeting miR-21 in spinal cord injuries: a game-changer? Mol Med 2022; 28:118. [PMID: 36138359 PMCID: PMC9502625 DOI: 10.1186/s10020-022-00546-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/12/2022] [Indexed: 12/03/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating neurological state causing physical disability, psychological stress and financial burden. SCI global rate is estimated between 250,000 and 500,000 individuals every year, of which 60% of victims are young, healthy males between 15 and 35 years. A variety of pathological conditions such as neuroinflammation, mitochondrial dysfunction, apoptosis, glial scar formation, blood-spinal cord barrier disruption, and angiogenesis disruption occur after SCI leading to a limitation in recovery. MicroRNAs (miRs) are endogenous and non-coding RNAs consisting of 22 nucleotides that regulate 60% of all human genes and involve several normal physiological processes and pathological conditions. miR-21 is among the most highly expressed miRs and its expression has been shown to increase one day after SCI and this elevation is sustained up to 28 days after injury. Overexpression of miR-21 exerts many protective effects against SCI by inhibiting neuroinflammation, improving blood-spinal cord barrier function, regulating angiogenesis, and controlling glial scar formation. It also exhibits anti-apoptotic effects in SCI by down-regulating the expression of PTEN, Spry2, and PDCD4. This review provides a novel therapeutic perspective for miR-21 in SCI.
Collapse
Affiliation(s)
- Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy.
| | - Seyed Hamidreza Rastegar-Moghaddam
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Anatomy and Cell Biology, Faculty of Medicine, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq, Vakilabad Blvd, Mashhad, Iran
| | | | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, Faculty of Medicine, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq, Vakilabad Blvd, Mashhad, Iran.,Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Mohammadipour
- Department of Anatomy and Cell Biology, Faculty of Medicine, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq, Vakilabad Blvd, Mashhad, Iran.,Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
35
|
Xiao S, Zhong N, Yang Q, Li A, Tong W, Zhang Y, Yao G, Wang S, Liu J, Liu Z. Aucubin promoted neuron functional recovery by suppressing inflammation and neuronal apoptosis in a spinal cord injury model. Int Immunopharmacol 2022; 111:109163. [PMID: 35994851 DOI: 10.1016/j.intimp.2022.109163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) can cause severe motor impairment. Post-SCI treatment has focused primarily on secondary injury, with neuroinflammation and neuronal apoptosis as the primary therapeutic targets. Aucubin (Au), a Chinese herbal medicine, exerts anti-inflammatory and neuroprotective effects. The therapeutic effects of Aucubin in SCI have not been reported. METHODS In this study, we carried out an in vivo SCI model and a series of in vitro experiments to explore the therapeutic effect of Aucubin. Western Blotting and immunofluorescence were used to study the effect of Aucubin on microglial polarization and neuronal apoptosis and its underlying mechanism. RESULTS We found that Aucubin can promote axonal regeneration by reducing neuroinflammation and neuronal apoptosis, which is beneficial to motor recovery after spinal cord injury in rats. Our further in vitro experiments showed that Aucubin can activate the toll-like receptor 4 (TLR4)/myeloid differentiation protein-88 (MyD88)/IκBα/nuclear factor kappa B (NF-κB) signaling pathway to reduce neuroinflammation and reverse mitochondrial dysfunction to reduce neuronal apoptosis. CONCLUSIONS In summary, these results suggest that Aucubin may ameliorate secondary injury after SCI by reducing neuroinflammation and neuronal apoptosis. Therefore, Au may be a promising post-SCI therapeutic drug.
Collapse
Affiliation(s)
- Shining Xiao
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Nanshan Zhong
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Quanming Yang
- Department of Orthopedics, Ningbo First Hospital, Ningbo 315000, China
| | - Anan Li
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Weilai Tong
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Yu Zhang
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Geliang Yao
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Shijiang Wang
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China; The First Clinical Medical College of Nanchang University, Nanchang 330006, China
| | - Jiaming Liu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China.
| | - Zhili Liu
- Medical Innovation Center, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Institute of Spine and Spinal Cord, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
36
|
Lolansen SD, Rostgaard N, Barbuskaite D, Capion T, Olsen MH, Norager NH, Vilhardt F, Andreassen SN, Toft-Bertelsen TL, Ye F, Juhler M, Keep RF, MacAulay N. Posthemorrhagic hydrocephalus associates with elevated inflammation and CSF hypersecretion via activation of choroidal transporters. Fluids Barriers CNS 2022; 19:62. [PMID: 35948938 PMCID: PMC9367104 DOI: 10.1186/s12987-022-00360-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/14/2022] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Posthemorrhagic hydrocephalus (PHH) often develops following hemorrhagic events such as intraventricular hemorrhage (IVH) and subarachnoid hemorrhage (SAH). Treatment is limited to surgical diversion of the cerebrospinal fluid (CSF) since no efficient pharmacological therapies are available. This limitation follows from our incomplete knowledge of the molecular mechanisms underlying the ventriculomegaly characteristic of PHH. Here, we aimed to elucidate the molecular coupling between a hemorrhagic event and the subsequent PHH development, and reveal the inflammatory profile of the PHH pathogenesis. METHODS CSF obtained from patients with SAH was analyzed for inflammatory markers using the proximity extension assay (PEA) technique. We employed an in vivo rat model of IVH to determine ventricular size, brain water content, intracranial pressure, and CSF secretion rate, as well as for transcriptomic analysis. Ex vivo radio-isotope assays of choroid plexus transport were employed to determine the direct effect of choroidal exposure to blood and inflammatory markers, both with acutely isolated choroid plexus and after prolonged exposure obtained with viable choroid plexus kept in tissue culture conditions. RESULTS The rat model of IVH demonstrated PHH and associated CSF hypersecretion. The Na+/K+-ATPase activity was enhanced in choroid plexus isolated from IVH rats, but not directly stimulated by blood components. Inflammatory markers that were elevated in SAH patient CSF acted on immune receptors upregulated in IVH rat choroid plexus and caused Na+/K+/2Cl- cotransporter 1 (NKCC1) hyperactivity in ex vivo experimental conditions. CONCLUSIONS CSF hypersecretion may contribute to PHH development, likely due to hyperactivity of choroid plexus transporters. The hemorrhage-induced inflammation detected in CSF and in the choroid plexus tissue may represent the underlying pathology. Therapeutic targeting of such pathways may be employed in future treatment strategies towards PHH patients.
Collapse
Affiliation(s)
- Sara Diana Lolansen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Nina Rostgaard
- Department of Neurosurgery, The Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Dagne Barbuskaite
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Tenna Capion
- Department of Neurosurgery, The Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Markus Harboe Olsen
- Department of Neuroanaesthesiology, The Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Nicolas H Norager
- Department of Neurosurgery, The Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Søren Norge Andreassen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Trine L Toft-Bertelsen
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Fenghui Ye
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Marianne Juhler
- Department of Neurosurgery, The Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Nanna MacAulay
- Department of Neuroscience, University of Copenhagen, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark.
| |
Collapse
|
37
|
Takamiya S, Kawabori M, Yamazaki K, Yamaguchi S, Tanimori A, Yamamoto K, Ohnishi S, Seki T, Konno K, Tha KK, Hashimoto D, Watanabe M, Houkin K, Fujimura M. Intravenous transplantation of amnion-derived mesenchymal stem cells promotes functional recovery and alleviates intestinal dysfunction after spinal cord injury. PLoS One 2022; 17:e0270606. [PMID: 35802703 PMCID: PMC9269969 DOI: 10.1371/journal.pone.0270606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/13/2022] [Indexed: 11/19/2022] Open
Abstract
Spinal cord injury (SCI) is often accompanied by gastrointestinal dysfunction due to the disconnection of the spinal autonomic nervous system. Gastrointestinal dysfunction reportedly upregulates intestinal permeability, leading to bacterial translocation of the gut microbiome to the systemic circulation, which further activates systemic inflammation, exacerbating neuronal damage. Mesenchymal stem cells (MSC) reportedly ameliorate SCI. Here, we aimed to investigate their effect on the associated gastrointestinal dysfunction. Human amnion-derived MSC (AMSCs) were intravenously transplanted one day after a rat model of midthoracic SCI. Biodistribution of transplanted cells, behavioral assessment, and histological evaluations of the spinal cord and intestine were conducted to elucidate the therapeutic effect of AMSCs. Bacterial translocation of the gut microbiome was examined by in situ hybridization and bacterial culture of the liver. Systemic inflammations were examined by blood cytokines, infiltrating immune cells in the spinal cord, and the size of the peripheral immune tissue. AMSCs released various neurotrophic factors and were mainly distributed in the liver and lung after transplantation. AMSC-transplanted animals showed smaller spinal damage and better neurological recovery with preserved neuronal tract. AMSCs transplantation ameliorated intestinal dysfunction both morphologically and functionally, which prevented translocation of the gut microbiome to the systemic circulation. Systemic inflammations were decreased in animals receiving AMSCs in the chronic phase. Intravenous AMSC administration during the acute phase of SCI rescues both spinal damage and intestinal dysfunction. Reducing bacterial translocation may contribute to decreasing systemic inflammation.
Collapse
Affiliation(s)
- Soichiro Takamiya
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| | - Kazuyoshi Yamazaki
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Hyogo, Japan
| | - Aki Tanimori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Yamamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shunsuke Ohnishi
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Toshitaka Seki
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kotaro Konno
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Khin Khin Tha
- Global Center for Biomedical Science and Engineering, Hokkaido University Faculty of Medicine, Sapporo, Hokkaido, Japan
| | - Daigo Hashimoto
- Department of Hematology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masahiko Watanabe
- Department of Anatomy and Embryology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
38
|
Pang QM, Qian NN, Zou WH, Yang YC, Chen H, Zhang M, Zhang Q, Ao J, Zhang T. PBMSCs transplantation facilitates functional recovery after spinal cord injury by regulating microglia/macrophages plasticity. Transpl Immunol 2022; 72:101592. [DOI: 10.1016/j.trim.2022.101592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/26/2022] [Indexed: 10/18/2022]
|
39
|
Xu S, Wang C, Mao R, Liang X, Wang H, Lin Z, Li J, Li S, Jiang J, Zhang T, Ma Y, Liu Y, Han CC, Liu Y. Surface structure change properties: Auto-soft bionic fibrous membrane in reducing postoperative adhesion. Bioact Mater 2022; 12:16-29. [PMID: 35087960 PMCID: PMC8777249 DOI: 10.1016/j.bioactmat.2021.10.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/19/2022] Open
Abstract
Peritoneal adhesion is the most common adverse effect following abdominal surgery or inflammation. The occurrence in clinical trials has been successfully reduced using barriers. However, the shortcomings of frequently used adhesion barriers, such as rapid degradation rate of gel barrier and inadequate operation ability of solid barrier, cannot be ignored. In this study, a fibrous membrane with an ECM-like structure was prepared. The adhesion properties were reduced significantly by changing the surface structure. The fibrous membrane caused less inflammatory response and much less peripheral adhesion and intestinal obstruction compared to the casting film and the commercial film with smooth surface, though with the same components. Because of the auto-soft bionic structure and similarity in the mechanical modulus of the tissues, the fibrous membrane was more flexible when it adhered to the tissues, showed excellent effectiveness and biocompatibility. In addition to the rat and miniature pig models, a randomized, placebo-controlled, and multicenter clinical pilot study with 150 patients confirmed that because of its flexibility, biodegradability, and similarity to mechanical modulus and structure with tissues involved, the fibrous membrane served as a favorable implant for preventing post-operation adhesion. The fibrous membrane with an ECM-like structure can be used the adhesion barrier. The fibrous membrane, with auto-soft bionic structure and similar mechanical modulus to the tissues, was flexible. The fibrous membrane caused less inflammatory response and intestinal obstruction.
Collapse
|
40
|
Van Broeckhoven J, Erens C, Sommer D, Scheijen E, Sanchez S, Vidal PM, Dooley D, Van Breedam E, Quarta A, Ponsaerts P, Hendrix S, Lemmens S. Macrophage-based delivery of interleukin-13 improves functional and histopathological outcomes following spinal cord injury. J Neuroinflammation 2022; 19:102. [PMID: 35488301 PMCID: PMC9052547 DOI: 10.1186/s12974-022-02458-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) elicits a robust neuroinflammatory reaction which, in turn, exacerbates the initial mechanical damage. Pivotal players orchestrating this response are macrophages (Mφs) and microglia. After SCI, the inflammatory environment is dominated by pro-inflammatory Mφs/microglia, which contribute to secondary cell death and prevent regeneration. Therefore, reprogramming Mφ/microglia towards a more anti-inflammatory and potentially neuroprotective phenotype has gained substantial therapeutic interest in recent years. Interleukin-13 (IL-13) is a potent inducer of such an anti-inflammatory phenotype. In this study, we used genetically modified Mφs as carriers to continuously secrete IL-13 (IL-13 Mφs) at the lesion site. METHODS Mφs were genetically modified to secrete IL-13 (IL-13 Mφs) and were phenotypically characterized using qPCR, western blot, and ELISA. To analyze the therapeutic potential, the IL-13 Mφs were intraspinally injected at the perilesional area after hemisection SCI in female mice. Functional recovery and histopathological improvements were evaluated using the Basso Mouse Scale score and immunohistochemistry. Neuroprotective effects of IL-13 were investigated using different cell viability assays in murine and human neuroblastoma cell lines, human neurospheroids, as well as murine organotypic brain slice cultures. RESULTS In contrast to Mφs prestimulated with recombinant IL-13, perilesional transplantation of IL-13 Mφs promoted functional recovery following SCI in mice. This improvement was accompanied by reduced lesion size and demyelinated area. The local anti-inflammatory shift induced by IL-13 Mφs resulted in reduced neuronal death and fewer contacts between dystrophic axons and Mφs/microglia, suggesting suppression of axonal dieback. Using IL-4Rα-deficient mice, we show that IL-13 signaling is required for these beneficial effects. Whereas direct neuroprotective effects of IL-13 on murine and human neuroblastoma cell lines or human neurospheroid cultures were absent, IL-13 rescued murine organotypic brain slices from cell death, probably by indirectly modulating the Mφ/microglia responses. CONCLUSIONS Collectively, our data suggest that the IL-13-induced anti-inflammatory Mφ/microglia phenotype can preserve neuronal tissue and ameliorate axonal dieback, thereby promoting recovery after SCI.
Collapse
Affiliation(s)
- Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Céline Erens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Daniela Sommer
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Elle Scheijen
- Department of Neurosciences, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Selien Sanchez
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Pia M Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, 4090541, Concepción, Chile
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular & Biomedical Research University College Dublin, Belfield, Dublin 4, Ireland
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium. .,Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| |
Collapse
|
41
|
The cytokines interleukin-6 and interferon-α induce distinct microglia phenotypes. J Neuroinflammation 2022; 19:96. [PMID: 35429976 PMCID: PMC9013466 DOI: 10.1186/s12974-022-02441-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background Elevated production of the cytokines interleukin (IL)-6 or interferon (IFN)-α in the central nervous system (CNS) is implicated in the pathogenesis of neurological diseases such as neuromyelitis optica spectrum disorders or cerebral interferonopathies, respectively. Transgenic mice with CNS-targeted chronic production of IL-6 (GFAP-IL6) or IFN-α (GFAP-IFN) recapitulate important clinical and pathological features of these human diseases. The activation of microglia is a prominent manifestation found both in the human diseases and in the transgenic mice, yet little is known about how this contributes to disease pathology. Methods Here, we used a combination of ex vivo and in situ techniques to characterize the molecular, cellular and transcriptomic phenotypes of microglia in GFAP-IL6 versus GFAP-IFN mice. In addition, a transcriptomic meta-analysis was performed to compare the microglia response from GFAP-IL6 and GFAP-IFN mice to the response of microglia in a range of neurodegenerative and neuroinflammatory disorders. Results We demonstrated that microglia show stimulus-specific responses to IL-6 versus IFN-α in the brain resulting in unique and extensive molecular and cellular adaptations. In GFAP-IL6 mice, microglia proliferated, had shortened, less branched processes and elicited transcriptomic and molecular changes associated with phagocytosis and lipid processing. In comparison, microglia in the brain of GFAP-IFN mice exhibited increased proliferation and apoptosis, had larger, hyper-ramified processes and showed transcriptomic and surface marker changes associated with antigen presentation and antiviral response. Further, a transcriptomic meta-analysis revealed that IL-6 and IFN-α both contribute to the formation of a core microglia response in animal models of neurodegenerative and neuroinflammatory disorders, such as Alzheimer’s disease, tauopathy, multiple sclerosis and lipopolysaccharide-induced endotoxemia. Conclusions Our findings demonstrate that microglia responses to IL-6 and IFN-α are highly stimulus-specific, wide-ranging and give rise to divergent phenotypes that modulate microglia responses in neuroinflammatory and neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02441-x.
Collapse
|
42
|
Menden A, Hall D, Hahn-Townsend C, Broedlow CA, Joshi U, Pearson A, Crawford F, Evans JE, Klatt N, Crynen S, Mullan M, Ait-Ghezala G. Exogenous lipase administration alters gut microbiota composition and ameliorates Alzheimer's disease-like pathology in APP/PS1 mice. Sci Rep 2022; 12:4797. [PMID: 35314754 PMCID: PMC8938460 DOI: 10.1038/s41598-022-08840-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 03/03/2022] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) represents the most common form of dementia in the elderly with no available disease modifying treatments. Altered gut microbial composition has been widely acknowledged as a common feature of AD, which potentially contributes to progression or onset of AD. To assess the hypothesis that Candida rugosa lipase (CRL), which has been shown to enhance gut microbiome and metabolite composition, can rebalance the gut microbiome composition and reduce AD pathology, the treatment effects in APPswe/PS1de9 (APP/PS1) mice were investigated. The analysis revealed an increased abundance of Acetatifactor and Clostridiales vadin BB60 genera in the gut; increased lipid hydrolysis in the gut lumen, normalization of peripheral unsaturated fatty acids, and reduction of neuroinflammation and memory deficits post treatment. Finally, we demonstrated that the evoked benefits on memory could be transferred via fecal matter transplant (FMT) into antibiotic-induced microbiome-depleted (AIMD) wildtype mice, ameliorating their memory deficits. The findings herein contributed to improve our understanding of the role of the gut microbiome in AD's complex networks and suggested that targeted modification of the gut could contribute to amelioration of AD neuropathology.
Collapse
Affiliation(s)
- Ariane Menden
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA.
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK.
| | - Davane Hall
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | | | - Courtney A Broedlow
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Utsav Joshi
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Andrew Pearson
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Fiona Crawford
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
- James A. Haley Veterans' Hospital, 13000 Bruce B. Downs Boulevard, Tampa, FL, 33612, USA
| | - James E Evans
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
| | - Nichole Klatt
- Division of Surgical Outcomes and Precision Medicine Research, Department of Surgery, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Stefan Crynen
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Michael Mullan
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| | - Ghania Ait-Ghezala
- Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA
- Open University, Walton Hall, Kents Hill, Milton-Keynes, MK7 6AA, UK
| |
Collapse
|
43
|
Phosphoinositide 3 Kinase γ Plays a Critical Role in Acute Kidney Injury. Cells 2022; 11:cells11050772. [PMID: 35269396 PMCID: PMC8909888 DOI: 10.3390/cells11050772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/05/2022] [Accepted: 02/17/2022] [Indexed: 11/17/2022] Open
Abstract
Inflammatory cells contribute to the pathogenesis of renal ischemia-reperfusion injury (IRI). However, the signaling mechanisms underlying the infiltration of inflammatory cells into the kidney are not well understood. In this study, we examined the effects of phosphoinositide 3 kinase γ (PI3Kγ) on inflammatory cells infiltration into the kidney in response to ischemia-reperfusion injury. Compared with wild-type mice, PI3Kγ knockout mice displayed less IRI in the kidney with fewer tubular apoptotic cell. Furthermore, PI3Kγ deficiency decreased the number of infiltrated neutrophils, macrophages, and T cells in the kidney, which was accompanied by a decrease in the expression of pro-inflammatory cytokines in the kidney. Moreover, wild-type mice treated with AS-605240, a selective PI3Kγ inhibitor, displayed less tubular damage, accumulated fewer inflammatory cells, and expressed less proinflammatory molecules in the kidney following IRI. These results demonstrate that PI3Kγ has a critical role in the pathogenesis of kidney damage in IRI, indicating that PI3Kγ inhibition may serve as a potential therapeutic strategy for the prevention of ischemia-reperfusion-induced kidney injury.
Collapse
|
44
|
Lan YX, Yang P, Zeng Z, Yadav N, Zhang LJ, Wang LB, Xia HC. Gene and protein expression profiles of olfactory ensheathing cells from olfactory bulb versus olfactory mucosa. Neural Regen Res 2022; 17:440-449. [PMID: 34269221 PMCID: PMC8463967 DOI: 10.4103/1673-5374.317986] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Olfactory ensheathing cells (OECs) from the olfactory bulb (OB) and the olfactory mucosa (OM) have the capacity to repair nerve injury. However, the difference in the therapeutic effect between OB-derived OECs and OM-derived OECs remains unclear. In this study, we extracted OECs from OB and OM and compared the gene and protein expression profiles of the cells using transcriptomics and non-quantitative proteomics techniques. The results revealed that both OB-derived OECs and OM-derived OECs highly expressed genes and proteins that regulate cell growth, proliferation, apoptosis and vascular endothelial cell regeneration. The differentially expressed genes and proteins of OB-derived OECs play a key role in regulation of nerve regeneration and axon regeneration and extension, transmission of nerve impulses and response to axon injury. The differentially expressed genes and proteins of OM-derived OECs mainly participate in the positive regulation of inflammatory response, defense response, cytokine binding, cell migration and wound healing. These findings suggest that differentially expressed genes and proteins may explain why OB-derived OECs and OM-derived OECs exhibit different therapeutic roles. This study was approved by the Animal Ethics Committee of the General Hospital of Ningxia Medical University (approval No. 2017-073) on February 13, 2017.
Collapse
Affiliation(s)
- Yuan-Xiang Lan
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Ping Yang
- Clinical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Zhong Zeng
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Neeraj Yadav
- Department of Neurosurgery, General Hospital of Ningxia Medical University; School of International Education, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li-Jian Zhang
- School of Clinical Medicine, Ningxia Medical University; Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li-Bin Wang
- Biochip Research Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - He-Chun Xia
- Ningxia Human Stem Cell Institute; Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| |
Collapse
|
45
|
Hellenbrand DJ, Quinn CM, Piper ZJ, Morehouse CN, Fixel JA, Hanna AS. Inflammation after spinal cord injury: a review of the critical timeline of signaling cues and cellular infiltration. J Neuroinflammation 2021; 18:284. [PMID: 34876174 PMCID: PMC8653609 DOI: 10.1186/s12974-021-02337-2] [Citation(s) in RCA: 276] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/30/2021] [Indexed: 03/02/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating neurological condition that results in a loss of motor and sensory function. Although extensive research to develop treatments for SCI has been performed, to date, none of these treatments have produced a meaningful amount of functional recovery after injury. The primary injury is caused by the initial trauma to the spinal cord and results in ischemia, oxidative damage, edema, and glutamate excitotoxicity. This process initiates a secondary injury cascade, which starts just a few hours post-injury and may continue for more than 6 months, leading to additional cell death and spinal cord damage. Inflammation after SCI is complex and driven by a diverse set of cells and signaling molecules. In this review, we utilize an extensive literature survey to develop the timeline of local immune cell and cytokine behavior after SCI in rodent models. We discuss the precise functional roles of several key cytokines and their effects on a variety of cell types involved in the secondary injury cascade. Furthermore, variations in the inflammatory response between rats and mice are highlighted. Since current SCI treatment options do not successfully initiate functional recovery or axonal regeneration, identifying the specific mechanisms attributed to secondary injury is critical. With a more thorough understanding of the complex SCI pathophysiology, effective therapeutic targets with realistic timelines for intervention may be established to successfully attenuate secondary damage.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Charles M Quinn
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Zachariah J Piper
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Carolyn N Morehouse
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Jordyn A Fixel
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA
| | - Amgad S Hanna
- Department of Neurological Surgery, School of Medicine and Public Health (UWSMPH), University of Wisconsin, 600 Highland Ave, Madison, WI, 53792, USA.
| |
Collapse
|
46
|
Jia Y, Lu T, Chen Q, Pu X, Ji L, Yang J, Luo C. Exosomes secreted from sonic hedgehog-modified bone mesenchymal stem cells facilitate the repair of rat spinal cord injuries. Acta Neurochir (Wien) 2021; 163:2297-2306. [PMID: 33821317 PMCID: PMC8270837 DOI: 10.1007/s00701-021-04829-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/23/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Spinal cord injuries (SCIs) can cause a loss of neurons and associated sensory and motor functionality below the injured site. No approaches to treating SCIs in humans have been developed to date. Exosomes are extracellular vesicles that hold promise as a potential therapeutic modality when treating such injuries. The present study was thus designed to determine whether sonic hedgehog (Shh)-overexpressing bone mesenchymal stem cell (BMSC)-derived exosomes were protective in the context of SCIs. METHODS Exosomes were extracted from control or Shh lentivirus-transduced BMSCs, yielding respective BMSC-Exo and BMSC-Shh-Exo preparations which were intravenously injected into SCI model rats. Shh expression in spinal cord tissues in these animals was then assessed via immunohistochemical staining, while Basso-Beattie-Bresnahan (BBB) scores were utilized to measure high limb motor function. Neuronal damage and regeneration within the spinal cord were additionally evaluated via terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), Nissl, hematoxylin and eosin, and immunofluorescent staining. RESULTS Both BMSC-Exo and BMSC-Shh-Exo preparations significantly increased Shh expression in the spinal cord of SCI model rats and improved BBB scores in these treated animals, while also increasing the frequencies of Nissl- and NeuN-positive neurons are reducing the numbers of apoptotic and GFAP-positive neurons. While both treatments yielded some degree of benefit to treated animals relative to untreated controls, BMSC-Shh-Exos were more beneficial than were control BMSC-Exos. CONCLUSIONS Shh-overexpressing BMSC-derived exosomes represent an effective treatment that can facilitate SCI repair in rats.
Collapse
Affiliation(s)
- Yijia Jia
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Tingsheng Lu
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Qiling Chen
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Xingwei Pu
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Linsong Ji
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Jianwen Yang
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China
| | - Chunshan Luo
- Department of Spine Surgery, Guizhou Province Osteological Hospital, 123 Shachong South Street, Nanming District, Guiyang, 550002, Guizhou Province, China.
| |
Collapse
|
47
|
Wei P, Wang K, Luo C, Huang Y, Misilimu D, Wen H, Jin P, Li C, Gong Y, Gao Y. Cordycepin confers long-term neuroprotection via inhibiting neutrophil infiltration and neuroinflammation after traumatic brain injury. J Neuroinflammation 2021; 18:137. [PMID: 34130727 PMCID: PMC8207641 DOI: 10.1186/s12974-021-02188-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/30/2021] [Indexed: 02/06/2023] Open
Abstract
Background The secondary injury caused by traumatic brain injury (TBI), especially white matter injury (WMI), is highly sensitive to neuroinflammation, which further leads to unfavored long-term outcomes. Although the cross-talk between the three active events, immune cell infiltration, BBB breakdown, and proinflammatory microglial/macrophage polarization, plays a role in the vicious cycle, its mechanisms are not fully understood. It has been reported that cordycepin, an extract from Cordyceps militaris, can inhibit TBI-induced neuroinflammation although the long-term effects of cordycepin remain unknown. Here, we report our investigation of cordycepin’s long-term neuroprotective function and its underlying immunological mechanism. Methods TBI mice model was established with a controlled cortical impact (CCI) method. Cordycepin was intraperitoneally administered twice daily for a week. Neurological outcomes were assessed by behavioral tests, including grid walking test, cylinder test, wire hang test, and rotarod test. Immunofluorescence staining, transmission electron microscopy, and electrophysiology recording were employed to assess histological and functional lesions. Quantitative-PCR and flow cytometry were used to detect neuroinflammation. The tracers of Sulfo-NHS-biotin and Evans blue were assessed for the blood-brain barrier (BBB) leakage. Western blot and gelatin zymography were used to analyze protein activity or expression. Neutrophil depletion in vivo was performed via using Ly6G antibody intraperitoneal injection. Results Cordycepin administration ameliorated long-term neurological deficits and reduced neuronal tissue loss in TBI mice. Meanwhile, the long-term integrity of white matter was also preserved, which was revealed in multiple dimensions, such as morphology, histology, ultrastructure, and electrical conductivity. Cordycepin administration inhibited microglia/macrophage pro-inflammatory polarization and promoted anti-inflammatory polarization after TBI. BBB breach was attenuated by cordycepin administration at 3 days after TBI. Cordycepin suppressed the activities of MMP-2 and MMP-9 and the neutrophil infiltration at 3 days after TBI. Moreover, neutrophil depletion provided a cordycepin-like effect, and cordycepin administration united with neutrophil depletion did not show a benefit of superposition. Conclusions The long-term neuroprotective function of cordycepin via suppressing neutrophil infiltration after TBI, thereby preserving BBB integrity and changing microglia/macrophage polarization. These findings provide significant clinical potentials to improve the quality of life for TBI patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02188-x.
Collapse
Affiliation(s)
- Pengju Wei
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ke Wang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chen Luo
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yichen Huang
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Dilidaer Misilimu
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Huimei Wen
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Peng Jin
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Chuhua Li
- School of Life Science, South China Normal University, Guangzhou, 510631, China
| | - Ye Gong
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Yanqin Gao
- Department of Critical Care Medicine and Neurosurgery of Huashan Hospital, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
48
|
Zou HJ, Guo SW, Zhu L, Xu X, Liu JB. Methylprednisolone Induces Neuro-Protective Effects via the Inhibition of A1 Astrocyte Activation in Traumatic Spinal Cord Injury Mouse Models. Front Neurosci 2021; 15:628917. [PMID: 34135725 PMCID: PMC8200570 DOI: 10.3389/fnins.2021.628917] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/24/2021] [Indexed: 11/13/2022] Open
Abstract
Traumatic spinal cord injury (TSCI) leads to pathological changes such as inflammation, edema, and neuronal apoptosis. Methylprednisolone (MP) is a glucocorticoid that has a variety of beneficial effects, including decreasing inflammation and ischemic reaction, as well as inhibiting lipid peroxidation. However, the efficacy and mechanism of MP in TSCI therapy is yet to be deciphered. In the present study, MP significantly attenuated the apoptotic effects of H2O2 in neuronal cells. Western blot analysis demonstrated that the levels of apoptotic related proteins, Bax and cleaved caspase-3, were reduced while levels of anti-apoptotic Bcl-2 were increased. In vivo TUNEL assays further demonstrated that MP effectively protected neuronal cells from apoptosis after TSCI, and was consistent with in vitro studies. Furthermore, we demonstrated that MP could decrease expression levels of IBA1, Il-1α, TNFα, and C3 and suppress A1 neurotoxic reactive astrocyte activation in TSCI mouse models. Neurological function was evaluated using the Basso Mouse Scale (BMS) and Footprint Test. Results demonstrated that the neurological function of MP-treated injured mice was significantly increased. In conclusion, our study demonstrated that MP could attenuate astrocyte cell death, decrease microglia activation, suppress A1 astrocytes activation, and promote functional recovery after acute TSCI in mouse models.
Collapse
Affiliation(s)
- Hong-Jun Zou
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Shi-Wu Guo
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Lin Zhu
- Department of Cardiology, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Xu Xu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| | - Jin-Bo Liu
- Department of Spinal Surgery, The Third Affiliated Hospital of Soochow University (The First People's Hospital of Changzhou), Changzhou, China
| |
Collapse
|
49
|
Vargova I, Machova Urdzikova L, Karova K, Smejkalova B, Sursal T, Cimermanova V, Turnovcova K, Gandhi CD, Jhanwar-Uniyal M, Jendelova P. Involvement of mTOR Pathways in Recovery from Spinal Cord Injury by Modulation of Autophagy and Immune Response. Biomedicines 2021; 9:biomedicines9060593. [PMID: 34073791 PMCID: PMC8225190 DOI: 10.3390/biomedicines9060593] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is untreatable and remains the leading cause of disability. Neuroprotection and recovery after SCI can be partially achieved by rapamycin (RAPA) treatment, an inhibitor of mTORC1, complex 1 of the mammalian target of rapamycin (mTOR) pathway. However, mechanisms regulated by the mTOR pathway are not only controlled by mTORC1, but also by a second mTOR complex (mTORC2). Second-generation inhibitor, pp242, inhibits both mTORC1 and mtORC2, which led us to explore its therapeutic potential after SCI and compare it to RAPA treatment. In a rat balloon-compression model of SCI, the effect of daily RAPA (5 mg/kg; IP) and pp242 (5 mg/kg; IP) treatment on inflammatory responses and autophagy was observed. We demonstrated inhibition of the mTOR pathway after SCI through analysis of p-S6, p-Akt, and p-4E-BP1 levels. Several proinflammatory cytokines were elevated in pp242-treated rats, while RAPA treatment led to a decrease in proinflammatory cytokines. Both RAPA and pp242 treatments caused an upregulation of LC3B and led to improved functional and structural recovery in acute SCI compared to the controls, however, a greater axonal sprouting was seen following RAPA treatment. These results suggest that dual mTOR inhibition by pp242 after SCI induces distinct mechanisms and leads to recovery somewhat inferior to that following RAPA treatment.
Collapse
Affiliation(s)
- Ingrid Vargova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
- 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague, Czech Republic
| | - Lucia Machova Urdzikova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
| | - Kristyna Karova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
| | - Barbora Smejkalova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
- 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague, Czech Republic
| | - Tolga Sursal
- Department of Neurosurgery, Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA; (T.S.); (C.D.G.)
| | - Veronika Cimermanova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
| | - Karolina Turnovcova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
| | - Chirag D. Gandhi
- Department of Neurosurgery, Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA; (T.S.); (C.D.G.)
| | - Meena Jhanwar-Uniyal
- Department of Neurosurgery, Westchester Medical Center, New York Medical College, Valhalla, NY 10595, USA; (T.S.); (C.D.G.)
- Correspondence: (M.J.-U.); (P.J.); Tel.: +420-2-4106-2828 (P.J.)
| | - Pavla Jendelova
- Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska, 1083, 142 20 Prague, Czech Republic; (I.V.); (L.M.U.); (K.K.); (B.S.); (V.C.); (K.T.)
- 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague, Czech Republic
- Correspondence: (M.J.-U.); (P.J.); Tel.: +420-2-4106-2828 (P.J.)
| |
Collapse
|
50
|
Zivkovic S, Ayazi M, Hammel G, Ren Y. For Better or for Worse: A Look Into Neutrophils in Traumatic Spinal Cord Injury. Front Cell Neurosci 2021; 15:648076. [PMID: 33967695 PMCID: PMC8100532 DOI: 10.3389/fncel.2021.648076] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are short-lived cells of the innate immune system and the first line of defense at the site of an infection and tissue injury. Pattern recognition receptors on neutrophils recognize pathogen-associated molecular patterns or danger-associated molecular patterns, which recruit them to the destined site. Neutrophils are professional phagocytes with efficient granular constituents that aid in the neutralization of pathogens. In addition to phagocytosis and degranulation, neutrophils are proficient in creating neutrophil extracellular traps (NETs) that immobilize pathogens to prevent their spread. Because of the cytotoxicity of the associated granular proteins within NETs, the microbes can be directly killed once immobilized by the NETs. The role of neutrophils in infection is well studied; however, there is less emphasis placed on the role of neutrophils in tissue injury, such as traumatic spinal cord injury. Upon the initial mechanical injury, the innate immune system is activated in response to the molecules produced by the resident cells of the injured spinal cord initiating the inflammatory cascade. This review provides an overview of the essential role of neutrophils and explores the contribution of neutrophils to the pathologic changes in the injured spinal cord.
Collapse
Affiliation(s)
- Sandra Zivkovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Grace Hammel
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|