51
|
Liu W, Fan M, Lu W, Zhu W, Meng L, Lu S. Emerging Roles of T Helper Cells in Non-Infectious Neuroinflammation: Savior or Sinner. Front Immunol 2022; 13:872167. [PMID: 35844577 PMCID: PMC9280647 DOI: 10.3389/fimmu.2022.872167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/30/2022] [Indexed: 12/03/2022] Open
Abstract
CD4+ T cells, also known as T helper (Th) cells, contribute to the adaptive immunity both in the periphery and in the central nervous system (CNS). At least seven subsets of Th cells along with their signature cytokines have been identified nowadays. Neuroinflammation denotes the brain’s immune response to inflammatory conditions. In recent years, various CNS disorders have been related to the dysregulation of adaptive immunity, especially the process concerning Th cells and their cytokines. However, as the functions of Th cells are being discovered, it’s also found that their roles in different neuroinflammatory conditions, or even the participation of a specific Th subset in one CNS disorder may differ, and sometimes contrast. Based on those recent and contradictory evidence, the conflicting roles of Th cells in multiple sclerosis, Alzheimer’s disease, Parkinson’s disease, epilepsy, traumatic brain injury as well as some typical mental disorders will be reviewed herein. Research progress, limitations and novel approaches concerning different neuroinflammatory conditions will also be mentioned and compared.
Collapse
Affiliation(s)
- Wenbin Liu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Department of Neurosurgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Meiyang Fan
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wen Lu
- Department of Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Wenhua Zhu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Liesu Meng
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
- *Correspondence: Wenhua Zhu, ; Liesu Meng,
| | - Shemin Lu
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- National Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi’an Jiaotong University), Ministry of Education, Xi’an, China
| |
Collapse
|
52
|
Johnson NH, Hadad R, Taylor RR, Rodríguez Pilar J, Salazar O, Llompart-Pou JA, Dietrich WD, Keane RW, Pérez-Bárcena J, de Rivero Vaccari JP. Inflammatory Biomarkers of Traumatic Brain Injury. Pharmaceuticals (Basel) 2022; 15:ph15060660. [PMID: 35745576 PMCID: PMC9227014 DOI: 10.3390/ph15060660] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 12/26/2022] Open
Abstract
Traumatic brain injury (TBI) has a complex pathology in which the initial injury releases damage associated proteins that exacerbate the neuroinflammatory response during the chronic secondary injury period. One of the major pathological players in the inflammatory response after TBI is the inflammasome. Increased levels of inflammasome proteins during the acute phase after TBI are associated with worse functional outcomes. Previous studies reveal that the level of inflammasome proteins in biological fluids may be used as promising new biomarkers for the determination of TBI functional outcomes. In this study, we provide further evidence that inflammatory cytokines and inflammasome proteins in serum may be used to determine injury severity and predict pathological outcomes. In this study, we analyzed blood serum from TBI patients and respective controls utilizing Simple Plex inflammasome and V-PLEX inflammatory cytokine assays. We performed statistical analyses to determine which proteins were significantly elevated in TBI individuals. The receiver operating characteristics (ROC) were determined to obtain the area under the curve (AUC) to establish the potential fit as a biomarker. Potential biomarkers were then compared to documented patient Glasgow coma scale scores via a correlation matrix and a multivariate linear regression to determine how respective biomarkers are related to the injury severity and pathological outcome. Inflammasome proteins and inflammatory cytokines were elevated after TBI, and the apoptosis-associated speck like protein containing a caspase recruitment domain (ASC), interleukin (IL)-18, tumor necrosis factor (TNF)-α, IL-4 and IL-6 were the most reliable biomarkers. Additionally, levels of these proteins were correlated with known clinical indicators of pathological outcome, such as the Glasgow coma scale (GCS). Our results show that inflammatory cytokines and inflammasome proteins are promising biomarkers for determining pathological outcomes after TBI. Additionally, levels of biomarkers could potentially be utilized to determine a patient’s injury severity and subsequent pathological outcome. These findings show that inflammation-associated proteins in the blood are reliable biomarkers of injury severity that can also be used to assess the functional outcomes of TBI patients.
Collapse
Affiliation(s)
- Nathan H. Johnson
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
| | - Roey Hadad
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
| | - Ruby Rose Taylor
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Javier Rodríguez Pilar
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Osman Salazar
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Juan Antonio Llompart-Pou
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - W. Dalton Dietrich
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Robert W. Keane
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (N.H.J.); (R.H.); (R.W.K.)
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
| | - Jon Pérez-Bárcena
- Intensive Care Department, Son Espases University Hospital, 07120 Palma de Mallorca, Spain; (J.R.P.); (O.S.); (J.A.L.-P.); (J.P.-B.)
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (R.R.T.); (W.D.D.)
- Correspondence:
| |
Collapse
|
53
|
Zhu G, Wang X, Chen L, Lenahan C, Fu Z, Fang Y, Yu W. Crosstalk Between the Oxidative Stress and Glia Cells After Stroke: From Mechanism to Therapies. Front Immunol 2022; 13:852416. [PMID: 35281064 PMCID: PMC8913707 DOI: 10.3389/fimmu.2022.852416] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Stroke is the second leading cause of global death and is characterized by high rates of mortality and disability. Oxidative stress is accompanied by other pathological processes that together lead to secondary brain damage in stroke. As the major component of the brain, glial cells play an important role in normal brain development and pathological injury processes. Multiple connections exist in the pathophysiological changes of reactive oxygen species (ROS) metabolism and glia cell activation. Astrocytes and microglia are rapidly activated after stroke, generating large amounts of ROS via mitochondrial and NADPH oxidase pathways, causing oxidative damage to the glial cells themselves and neurons. Meanwhile, ROS cause alterations in glial cell morphology and function, and mediate their role in pathological processes, such as neuroinflammation, excitotoxicity, and blood-brain barrier damage. In contrast, glial cells protect the Central Nervous System (CNS) from oxidative damage by synthesizing antioxidants and regulating the Nuclear factor E2-related factor 2 (Nrf2) pathway, among others. Although numerous previous studies have focused on the immune function of glial cells, little attention has been paid to the role of glial cells in oxidative stress. In this paper, we discuss the adverse consequences of ROS production and oxidative-antioxidant imbalance after stroke. In addition, we further describe the biological role of glial cells in oxidative stress after stroke, and we describe potential therapeutic tools based on glia cells.
Collapse
Affiliation(s)
- Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luxi Chen
- Department of Medical Genetics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Biomedical Science, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhua Yu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
54
|
Arora P, Singh K, Kumari M, Trivedi R. Temporal profile of serum metabolites and inflammation following closed head injury in rats is associated with HPA axis hyperactivity. Metabolomics 2022; 18:28. [PMID: 35486220 DOI: 10.1007/s11306-022-01886-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/06/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Closed head injury (CHI) causes neurological disability along with systemic alterations that can activate neuro-endocrine response through hypothalamic-pituitary-adrenal (HPA) axis activation. A dysregulated HPA axis function can lead to relocation of energy substrates and alteration in metabolic pathways and inflammation at the systemic level. OBJECTIVES Assessment of time-dependent changes in serum metabolites and inflammation after both mild and moderate CHI. Along with this, serum corticosterone levels and hypothalamic microglial response were observed. METHODS Rats underwent mild and moderate weight-drop injury and their serum and hypothalamus were assessed at acute, sub-acute and chronic timepoints. Changes in serum metabolomics were determined using high resolution NMR spectroscopy. Serum inflammatory cytokine, corticosterone levels and hypothalamic microglia were assessed at all timepoints. RESULTS Metabolites including lactate, choline and branched chain amino acids were found as the classifiers that helped distinguish between control and injured rats during acute, sub-acute and chronic timepoints. While, increased αglucose: βglucose and TMAO: choline ratios after acute and sub-acute timepoints of mild injury differentiated from moderate injured rats. The injured rats also showed distinct inflammatory profile where IL-1β and TNF-α levels were upregulated in moderate injured rats while IL-10 levels were downregulated in mild injured rats. Furthermore, injury specific alterations in serum metabolic and immunologic profile were found to be associated with hyperactive HPA axis, with consistent increase in serum corticosterone concentration post injury. The hypothalamic microglia showed a characteristic activated de-ramified cellular morphology in both mild and moderate injured rats. CONCLUSION The study suggests that HPA axis hyperactivity along with hypothalamic microglial activation led to temporal changes in the systemic metabolism and inflammation. These time dependent changes in the metabolite profile of rats can further strengthen the knowledge of diagnostic markers and help distinguish injury related outcomes after TBI.
Collapse
Affiliation(s)
- Palkin Arora
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Department of Biochemistry, Panjab University, Chandigarh, 160014, India
| | - Kavita Singh
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Megha Kumari
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India
- Department of Biotechnology, Delhi Technological University (DTU), Delhi, 110042, India
| | - Richa Trivedi
- Radiological, Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences (INMAS), DRDO, Delhi, 110054, India.
| |
Collapse
|
55
|
Mostajeran M, Edvinsson L, Ahnstedt H, Arkelius K, Ansar S. Repair-related molecular changes during recovery phase of ischemic stroke in female rats. BMC Neurosci 2022; 23:23. [PMID: 35413803 PMCID: PMC9004052 DOI: 10.1186/s12868-022-00696-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 02/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Some degree of spontaneous recovery is usually observed after stroke. Experimental studies have provided information about molecular mechanisms underlying this recovery. However, the majority of pre-clinical stroke studies are performed in male rodents, and females are not well studied. This is a clear discrepancy when considering the clinical situation. Thus, it is important to include females in the evaluation of recovery mechanisms for future therapeutic strategies. This study aimed to evaluate spontaneous recovery and molecular mechanisms involved in the recovery phase two weeks after stroke in female rats. METHODS Transient middle cerebral artery occlusion was induced in female Wistar rats using a filament model. Neurological functions were assessed up to day 14 after stroke. Protein expression of interleukin 10 (IL-10), transforming growth factor (TGF)-β, neuronal specific nuclei protein (NeuN), nestin, tyrosine-protein kinase receptor Tie-2, extracellular signal-regulated kinase (ERK) 1/2, and Akt were evaluated in the peri-infarct and ischemic core compared to contralateral side of the brain at day 14 by western blot. Expression of TGF-β in middle cerebral arteries was evaluated by immunohistochemistry. RESULTS Spontaneous recovery after stroke was observed from day 2 to day 14 and was accompanied by a significantly higher expression of nestin, p-Akt, p-ERK1/2 and TGF-β in ischemic regions compared to contralateral side at day 14. In addition, a significantly higher expression of TGF-β was observed in occluded versus non-occluded middle cerebral arteries. The expression of Tie-2 and IL-10 did not differ between the ischemic and contralateral sides. CONCLUSION Spontaneous recovery after ischemic stroke in female rats was coincided by a difference observed in the expression of molecular markers. The alteration of these markers might be of importance to address future therapeutic strategies.
Collapse
Affiliation(s)
- Maryam Mostajeran
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lars Edvinsson
- Division of Experimental Vascular Research, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hilda Ahnstedt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kajsa Arkelius
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Saema Ansar
- Applied Neurovascular Research, Neurosurgery, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
56
|
Rutin protects hemorrhagic stroke development via supressing oxidative stress and inflammatory events in a zebrafish model. Eur J Pharmacol 2022; 925:174973. [DOI: 10.1016/j.ejphar.2022.174973] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/11/2022] [Accepted: 04/19/2022] [Indexed: 01/06/2023]
|
57
|
Luo C, Yao J, Bi H, Li Z, Li J, Xue G, Li K, Zhang S, Zan K, Meng W, Zhang Z, Chen H. Clinical Value of Inflammatory Cytokines in Patients with Aneurysmal Subarachnoid Hemorrhage. Clin Interv Aging 2022; 17:615-626. [PMID: 35502188 PMCID: PMC9056097 DOI: 10.2147/cia.s362854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/20/2022] [Indexed: 12/16/2022] Open
Abstract
Background Inflammation is closely associated with prognosis in patients with aneurysmal subarachnoid hemorrhage (aSAH), which is orchestrated by inflammatory cytokines. Therefore, this study aimed to investigate the levels of inflammatory cytokines in the early stage of aSAH and their predictive value for prognosis. Methods In this retrospective study, 206 patients with aSAH were recruited and assigned to a severe group (WFNS grade ≥ 4) and a mild group (WFNS grade < 4) according to the severity of patients on admission. Flow cytometry was performed to detect the levels of 12 inflammatory cytokines in the serum of patients. Then, patients were grouped into a poor prognosis group (mRS score ≥ 4) and a good prognosis group (mRS score < 4) based on their prognosis after 3 months of discharge to compare the relationship between cytokines and prognosis. Propensity score matching (PSM) was utilized to control confounding factors. The correlation between inflammatory factors and prognosis was determined using Spearman correlation, and the predictive efficacy of inflammatory factors was tested by a receiver operating characteristic curve. Results Serum IL-1β, IL-5, IL-6, IL-8, IL-10, IFN-γ, and TNF-α levels were significantly higher in the mild group than in the severe group and in the poor prognosis group than in the good prognosis group. After PSM, the differences in IL-1β, IL-5, IFN-α, and IFN-γ levels disappeared between the two groups, whereas IL-2, IL-6, IL-8, IL-10, and TNF-α levels remained higher in the poor prognosis group than in the good prognosis group. Additionally, IL-2, IL-6, IL-8, and IL-10 levels were positively correlated with mRS scores. Moreover, the predictive value was found to be the highest for IL-6 and the lowest for TNF-α. Conclusion Inflammation degree was related to the severity of aSAH. Inflammatory markers, including IL-6, IL-10, IL-8, IL-2, and TNF-α, might predict the poor prognosis of aSAH.
Collapse
Affiliation(s)
- Cong Luo
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Jiaxin Yao
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Haoran Bi
- Department of Biostatistics, Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Zhen Li
- Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Ju Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Guosong Xue
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Ke Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Shenyang Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Kun Zan
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Wenqing Meng
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| | - Zunsheng Zhang
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
- Correspondence: Zunsheng Zhang; Hao Chen, Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai West Road, Quanshan District, Xuzhou, Jiangsu, People’s Republic of China, Tel +86-13913473179; +86-15252006510, Email ;
| | - Hao Chen
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
58
|
Salas-Venegas V, Flores-Torres RP, Rodríguez-Cortés YM, Rodríguez-Retana D, Ramírez-Carreto RJ, Concepción-Carrillo LE, Pérez-Flores LJ, Alarcón-Aguilar A, López-Díazguerrero NE, Gómez-González B, Chavarría A, Konigsberg M. The Obese Brain: Mechanisms of Systemic and Local Inflammation, and Interventions to Reverse the Cognitive Deficit. Front Integr Neurosci 2022; 16:798995. [PMID: 35422689 PMCID: PMC9002268 DOI: 10.3389/fnint.2022.798995] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Overweight and obesity are now considered a worldwide pandemic and a growing public health problem with severe economic and social consequences. Adipose tissue is an organ with neuroimmune-endocrine functions, which participates in homeostasis. So, adipocyte hypertrophy and hyperplasia induce a state of chronic inflammation that causes changes in the brain and induce neuroinflammation. Studies with obese animal models and obese patients have shown a relationship between diet and cognitive decline, especially working memory and learning deficiencies. Here we analyze how obesity-related peripheral inflammation can affect central nervous system physiology, generating neuroinflammation. Given that the blood-brain barrier is an interface between the periphery and the central nervous system, its altered physiology in obesity may mediate the consequences on various cognitive processes. Finally, several interventions, and the use of natural compounds and exercise to prevent the adverse effects of obesity in the brain are also discussed.
Collapse
Affiliation(s)
- Verónica Salas-Venegas
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Rosa Pamela Flores-Torres
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana - Unidad Iztapalapa, Mexico City, Mexico
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Yesica María Rodríguez-Cortés
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Diego Rodríguez-Retana
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Ricardo Jair Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Luis Edgar Concepción-Carrillo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Laura Josefina Pérez-Flores
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Adriana Alarcón-Aguilar
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Norma Edith López-Díazguerrero
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
| | - Beatriz Gómez-González
- Departamento de Biología de la Reproducción, DCBS, Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México (CDMX), Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, CDMX, Mexico City, Mexico
| | - Mina Konigsberg
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, CDMX, Mexico City, Mexico
- *Correspondence: Mina Konigsberg,
| |
Collapse
|
59
|
Gao T, Huang F, Wang W, Xie Y, Wang B. Interleukin-10 genetically modified clinical-grade mesenchymal stromal cells markedly reinforced functional recovery after spinal cord injury via directing alternative activation of macrophages. Cell Mol Biol Lett 2022; 27:27. [PMID: 35300585 PMCID: PMC8931978 DOI: 10.1186/s11658-022-00325-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Background After spinal cord injury (SCI), dysregulated or nonresolving inflammatory processes can severely disturb neuronal homeostasis and drive neurodegeneration. Although mesenchymal stromal cell (MSC)-based therapies have showed certain therapeutic efficacy, no MSC therapy has reached its full clinical goal. In this study, we examine interleukin-10 (IL10) genetically modified clinical-grade MSCs (IL10-MSCs) and evaluate their clinical safety, effectiveness, and therapeutic mechanism in a completely transected SCI mouse model. Methods We established stable IL10-overexpressing human umbilical-cord-derived MSCs through electric transduction and screened out clinical-grade IL10-MSCs according to the criteria of cell-based therapeutic products, which were applied to mice with completely transected SCI by repeated tail intravenous injections. Then we comprehensively investigated the motor function, histological structure, and nerve regeneration in SCI mice, and further explored the potential therapeutic mechanism after IL10-MSC treatment. Results IL10-MSC treatment markedly reinforced locomotor improvement, accompanied with decreased lesion volume, regeneration of axons, and preservation of neurons, compared with naïve unmodified MSCs. Further, IL10-MSC transplantation increased the ratio of microglia to infiltrated alternatively activated macrophages (M2), and reduced the ratio of classically activated macrophages (M1) at the injured spinal cord, meanwhile increasing the percentage of Treg and Th2 cells, and reducing the percentage of Th1 cells in the peripheral circulatory system. In addition, IL10-MSC administration could prevent apoptosis and promote neuron differentiation of neural stem cells (NSCs) under inflammatory conditions in vitro. Conclusions IL10-MSCs exhibited a reliable safety profile and demonstrated promising therapeutic efficacy in SCI compared with naïve MSCs, providing solid support for future clinical application of genetically engineered MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00325-9.
Collapse
Affiliation(s)
- Tianyun Gao
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Feifei Huang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wenqing Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
60
|
Amantea D, La Russa D, Frisina M, Giordano F, Di Santo C, Panno ML, Pignataro G, Bagetta G. Ischemic Preconditioning Modulates the Peripheral Innate Immune System to Promote Anti-Inflammatory and Protective Responses in Mice Subjected to Focal Cerebral Ischemia. Front Immunol 2022; 13:825834. [PMID: 35359933 PMCID: PMC8962743 DOI: 10.3389/fimmu.2022.825834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/21/2022] [Indexed: 11/26/2022] Open
Abstract
The development of tolerance triggered by a sublethal ischemic episode (preconditioning, PC) involves a complex crosstalk between neurons, astrocytes and microglia, although the role of the peripheral immune system in this context is largely unexplored. Here, we report that severe cerebral ischemia caused by transient middle cerebral artery occlusion (MCAo) in adult male mice elevates blood counts of inflammatory neutrophils and monocytes, and plasma levels of miRNA-329-5p. These inflammatory responses are prevented by ischemic PC induced by 15 min MCAo, 72h before the severe insult (1h MCAo). As compared with sham-operated animals, mice subjected to either ischemic PC, MCAo or a combination of both (PC+MCAo) display spleen contraction. However, protein levels of Ym1 (a marker of polarization of myeloid cells towards M2/N2 protective phenotypes) are elevated only in spleen from the experimental groups PC and PC+MCAo, but not MCAo. Conversely, Ym1 protein levels only increase in circulating leukocytes from mice subjected to 1h MCAo, but not in preconditioned animals, which is coincident with a dramatic elevation of Ym1 expression in the ipsilateral cortex. By immunofluorescence analysis, we observe that expression of Ym1 occurs in amoeboid-shaped myeloid cells, mainly representing inflammatory monocytes/macrophages and neutrophils. As a result of its immune-regulatory functions, ischemic PC prevents elevation of mRNA levels of the pro-inflammatory cytokine interleukin (IL)-1β in the ipsilateral cortex, while not affecting IL-10 mRNA increase induced by MCAo. Overall, the elevated anti-inflammatory/pro-inflammatory ratio observed in the brain of mice pre-exposed to PC is associated with reduced brain infarct volume and ischemic edema, and with amelioration of functional outcome. These findings reaffirm the crucial and dualistic role of the innate immune system in ischemic stroke pathobiology, extending these concepts to the context of ischemic tolerance and underscoring their relevance for the identification of novel therapeutic targets for effective stroke treatment.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Daniele La Russa
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Marialaura Frisina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Chiara Di Santo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Maria Luisa Panno
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Giuseppe Pignataro
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, “Federico II” University, Naples, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| |
Collapse
|
61
|
Hosein W, Henkin RI. Therapeutic diminution of Interleukin-10 with intranasal theophylline administration in hyposmic patients. Am J Otolaryngol 2022; 43:103375. [PMID: 35124402 DOI: 10.1016/j.amjoto.2022.103375] [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: 12/21/2021] [Accepted: 01/22/2022] [Indexed: 11/01/2022]
Abstract
OBJECTIVE To determine changes in nasal mucus Interleukin-10 (IL-10) before and after intranasal theophylline treatment in hyposmic patients, and the relationship of these changes to orally administered theophylline treatment. DESIGN IL-10 was measured in nasal mucus samples of 17 normal subjects and 39 patients with hyposmia of multiple etiologies by use of a sensitive spectrophotometric ELISA assay. Hyposmia is defined clinically by standardized evaluation of impaired olfactometry, as well as subjectively self reported by the patient. RESULTS Prior to treatment, IL-10 levels in nasal mucus were increased in hyposmic patients compared to controls. Following intranasal theophylline administration, over half of treated patients experienced a decrease of nasal mucus IL-10 toward control levels, correlated with a significant improvement in taste and smell function. Patients who increased with intranasal administration, similar to previously reported oral administration, showed no significant improvement in taste or smell function. CONCLUSIONS Nasal mucus IL-10 decreased in patients treated with intranasal theophylline. Mechanisms of action include phosphodiesterase and nuclear factor kappa-B inhibition, correlated with an improvement in both olfaction and gustation. IL-10 is induced by therapeutic and systemic administration of theophylline as with oral dosage, but reduced by lower and localized treatment.
Collapse
|
62
|
Goit RK, Taylor AW, Lo ACY. Anti-inflammatory α-Melanocyte-Stimulating Hormone Protects Retina After Ischemia/Reperfusion Injury in Type I Diabetes. Front Neurosci 2022; 16:799739. [PMID: 35281489 PMCID: PMC8914517 DOI: 10.3389/fnins.2022.799739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Retinal ischemia/reperfusion (I/R) injury is a major cause of vision loss in many ocular diseases. Retinal I/R injury is common in diabetic retinopathy, which as a result of hyperglycemia damages the retina and can cause blindness if left untreated. Inflammation is a major contributing factor in the pathogenesis of I/R injury. α-Melanocyte-stimulating hormone (α-MSH) is an anti-inflammatory peptide hormone that has displayed protective effects against I/R-induced organ damages. Here, we aimed to investigate the protective role of α-MSH on I/R-induced diabetic retinal damage using hyperglycemic C57BL/6J Ins2Akita/+ mice. Experimental I/R injury was induced by blocking the right middle cerebral artery (MCA) for 2 h followed by 2 h or 22 h of reperfusion using the intraluminal method. Since ophthalmic artery originates proximal to the origin of the MCA, the filament also blocked blood supply to the retina. Upon treatment with α-MSH at 1 h after ischemia and 1 h after reperfusion, animals displayed significant improvement in amplitudes of b-wave and oscillatory potentials during electroretinography. α-MSH also prevented I/R-induced histological alterations and inhibited the development of retinal swelling. Loss of retinal ganglion cells as well as oxidative stress were significantly attenuated in the α-MSH-treated retinae. Level of interleukin 10 was significantly increased after α-MSH treatment. Moreover, gene expression of glutamate aspartate transporter 1, monocarboxylate transporter (MCT) 1 and MCT-2 were significantly higher after α-MSH administration. In conclusion, α-MSH mitigates the severity of I/R-induced retinal damage under hyperglycemic condition. These beneficial effects of α-MSH may have important therapeutic implications against retinal I/R injury under hyperglycemic condition.
Collapse
Affiliation(s)
- Rajesh Kumar Goit
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Andrew W. Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- *Correspondence: Amy C. Y. Lo, , orcid.org/0000-0003-4239-6851
| |
Collapse
|
63
|
Ávila-Gómez P, Pérez-Mato M, Hervella P, Dopico-López A, da Silva-Candal A, Bugallo-Casal A, López-Amoedo S, Candamo-Lourido M, Sobrino T, Iglesias-Rey R, Castillo J, Campos F. Associations between RNA-Binding Motif Protein 3, Fibroblast Growth Factor 21, and Clinical Outcome in Patients with Stroke. J Clin Med 2022; 11:jcm11040949. [PMID: 35207221 PMCID: PMC8875775 DOI: 10.3390/jcm11040949] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/27/2023] Open
Abstract
Background: RNA-binding motif protein 3 (RBM3) is a cold-induced marker of good functional outcome of ischemic stroke that is promising as a protective target. Fibroblast growth factor 21 (FGF21) is an obesity- and temperature-related hormone that upregulates the expression of RBM3, which is beneficial as a recombinant treatment and has been tested under different experimental pathological conditions, including stroke. However, the interaction between RBM3 and FGF21 has not yet been tested for clinical stroke conditions. Methods: In a sample of 66 stroke patients, we analyzed the associations between the FGF21 and RBM3 serum concentrations on admission and at 72 h, body weight, maximum temperature during the first 24 h, and the outcome of patients at 3 months. We also analyzed their association with biomarkers of obesity (adiponectin and leptin) and inflammation (interleukin-6 (IL-6) and interleukin (IL-10)). Results: Higher concentrations of FGF21 on admission and RBM3 at 72 h were associated with good outcomes. Serum FGF21 and RBM3 were directly related to body mass index and inversely related to the maximum temperature during the first 24 h. We found a positive association between the FGF21 concentrations in obese patients with leptin and a negative correlation with adiponectin in non-obese participants. Conclusions: This clinical study demonstrates the association between RBM3 and FGF21 levels and the outcome of stroke patients. Although further investigations are required, these data support the pharmacological induction of RBM3 as a promising protective therapy.
Collapse
Affiliation(s)
- Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - María Pérez-Mato
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Paseo de la Castellana 261, 28046 Madrid, Spain;
| | - Pablo Hervella
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Andrés da Silva-Candal
- Neurovascular Diseases Laboratory, Neurology Service, Biomedical Research Institute (INIBIC), University Hospital Complex of A Coruña, 15006 A Coruña, Spain;
| | - Ana Bugallo-Casal
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Sonia López-Amoedo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - María Candamo-Lourido
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.Á.-G.); (P.H.); (A.D.-L.); (A.B.-C.); (S.L.-A.); (M.C.-L.); (T.S.); (R.I.-R.); (J.C.)
- Correspondence: ; Tel./Fax: +34-981951097
| |
Collapse
|
64
|
Vaughn MN, Winston CN, Levin N, Rissman RA, Risbrough VB. Developing Biomarkers of Mild Traumatic Brain Injury: Promise and Progress of CNS-Derived Exosomes. Front Neurol 2022; 12:698206. [PMID: 35222223 PMCID: PMC8866179 DOI: 10.3389/fneur.2021.698206] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 12/30/2021] [Indexed: 01/18/2023] Open
Abstract
Mild traumatic brain injuries (mTBI) are common injuries across civilian and military populations. Although most individuals recover after mTBI, some individuals continue to show long-term symptoms as well as increased risk for neurodegenerative and neuropsychiatric disorders. Currently, diagnosing TBI severity relies primarily on self-report and subjective symptoms, with limited tools for diagnosis or prognosis. Brain-derived exosomes, a form of extracellular vesicle, may offer a solution for interpreting injury states by aiding in diagnosis as well as outcome prediction with relatively low patient burden. Exosomes, which are released into circulation, contain both protein and RNA cargo that can be isolated and quantified, providing a molecular window into molecular status of the exosome source. Here we examined the current literature studying the utility of exosomes, in particular neuronal- and astrocyte-derived exosomes, to identify protein and miRNA biomarkers of injury severity, trajectory, and functional outcome. Current evidence supports the potential for these emerging new tools to capture an accessible molecular window into the brain as it responds to a traumatic injury, however a number of limitations must be addressed in future studies. Most current studies are relatively small and cross sectional; prospective, longitudinal studies across injury severity, and populations are needed to track exosome cargo changes after injury. Standardized exosome isolation as well as advancement in identifying/isolating exosomes from CNS-specific tissue sources will improve mechanistic understanding of cargo changes as well as reliability of findings. Exosomes are also just beginning to be used in model systems to understand functional effects of TBI-associated cargo such as toxicity. Finally linking exosome cargo changes to objective markers of neuronal pathology and cognitive changes will be critical in validating these tools to provide insights into injury and recovery states after TBI.
Collapse
Affiliation(s)
- Melonie N. Vaughn
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Charisse N. Winston
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Natalie Levin
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
- Veterans Affairs San Diego Health System, University of California, San Diego, San Diego, CA, United States
| | - Victoria B. Risbrough
- Veterans Affairs San Diego Health System, University of California, San Diego, San Diego, CA, United States
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
- VA Center of Excellence for Stress and Mental Health, La Jolla, CA, United States
| |
Collapse
|
65
|
Jash S, Sharma S. Pathogenic Infections during Pregnancy and the Consequences for Fetal Brain Development. Pathogens 2022; 11:pathogens11020193. [PMID: 35215136 PMCID: PMC8877441 DOI: 10.3390/pathogens11020193] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Pathogens comprised of viruses, bacteria, gut microbiome, and parasites are a leading cause of ever-emerging diseases in humans. Studying pathogens for their ability to cause diseases is a topic of critical discussion among scientists and pharmaceutical centers for effective drug development that diagnose, treat, and prevent infection-associated disorders. Pathogens impact health either directly by invading the host or by eliciting an acute inflammatory immune response. This paradigm of inflammatory immune responses is even more consequential in people who may be immunocompromised. In this regard, pregnancy offers an altered immunity scenario, which may allow the onset of severe diseases. Viruses, such as Influenza, HIV, and now SARS-CoV-2, associated with the COVID-19 pandemic, raise new concerns for maternal and fetal/neonatal health. Intrauterine bacterial and parasitic infections are also known to impact pregnancy outcomes and neonatal health. More importantly, viral and bacterial infections during pregnancy have been identified as a common contributor to fetal brain development defects. Infection-mediated inflammatory uterine immune milieu is thought to be the main trigger for causing poor fetal brain development, resulting in long-term cognitive impairments. The concept of in utero programming of childhood and adult disorders has revolutionized the field of neurodevelopment and its associated complications. Recent findings in mice and humans clearly support the idea that uterine immunity during pregnancy controls the health trajectory of the child and considerably influences the cognitive function and mental health. In this review, we focus on the in utero programming of autism spectrum disorders (ASD) and assess the effects of pathogens on the onset of ASD-like symptoms.
Collapse
|
66
|
Kristinsson S, Fridriksson J. Genetics in aphasia recovery. HANDBOOK OF CLINICAL NEUROLOGY 2022; 185:283-296. [PMID: 35078606 DOI: 10.1016/b978-0-12-823384-9.00015-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Considerable research efforts have been exerted toward understanding the mechanisms underlying recovery in aphasia. However, predictive models of spontaneous and treatment-induced recovery remain imprecise. Some of the hitherto unexplained variability in recovery may be accounted for with genetic data. A few studies have examined the effects of the BDNF val66met polymorphism on aphasia recovery, yielding mixed results. Advances in the study of stroke genetics and genetics of stroke recovery, including identification of several susceptibility genes through candidate-gene or genome-wide association studies, may have implications for the recovery of language function. The current chapter discusses both the direct and indirect evidence for a genetic basis of aphasia recovery, the implications of recent findings within the field, and potential future directions to advance understanding of the genetics-recovery associations.
Collapse
Affiliation(s)
- Sigfus Kristinsson
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, SC, United States
| | - Julius Fridriksson
- Department of Communication Sciences and Disorders, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
67
|
The Hypoxia-Related Gene COL5A1 Is a Prognostic and Immunological Biomarker for Multiple Human Tumors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6419695. [PMID: 35082969 PMCID: PMC8786464 DOI: 10.1155/2022/6419695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/15/2021] [Accepted: 12/14/2021] [Indexed: 01/19/2023]
Abstract
Background Collagen type V alpha 1 chain (COL5A1) is a hypoxia-related gene (a collagen family protein) and participates in the formation of the extracellular matrix. Although some evidence supports a significant role for COL5A1 in the progression of several cancers, a pan-cancer analysis of COL5A1 is not currently available. Herein, we aimed to assess the prognostic value of COL5A1 in 33 human cancers and to investigate its underlying immunological function. Methods Through multiple bioinformatics methods, we analyzed the data from Oncomine, TCGA, CCLE, HPA, DNMIVD, and cBioPortal database to explore the potential underlying carcinogenic effect of COL5A1, including the relevance of COL5A1 to the outcome, DNA methylation, tumor microenvironment, immune cells infiltration, and drug sensitivity in 33 human cancers. The effects of COL5A1 on glioma cell proliferation, migration, and invasion were verified in cellular experiments. Results Our findings indicated that COL5A1 was expressed at high levels in 13 cancers and was negatively related to the prognosis of 11 cancers. Additionally, COL5A1 was coexpressed with genes encoding the major histocompatibility complex, immune activators, immune suppressors, chemokines, chemokine receptors, mismatch repair genes, and immune checkpoints. We also identified different roles for COL5A1 in the immunocyte infiltration in different cancers. The correlation between COL5A1 and drug sensitivity was found in several cancers. COL5A1 potentially influenced the tumor progression through immune-related pathways, negative regulation of immune system processes, chemokine signaling pathways, JAK-STAT pathways, T cell receptor pathways, lymphocyte migration, and antigen processing and presentation, among other processes. Conclusions Based on our study, COL5A1 may be employed as a prognostic marker in different malignancies because of its impact on tumorigenesis and immune cell infiltration and have implications for cancer immune checkpoint inhibitors and chemotherapy.
Collapse
|
68
|
Huang Y, Wang X, Guan S, Lin H, Mei Z, Huang Z. Syringin protects against cerebral ischemia and reperfusion injury via suppression of inflammatory mediators and toll-like receptor/MyD88 signaling pathway in rats. Pharmacogn Mag 2022. [DOI: 10.4103/pm.pm_98_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
69
|
Chronic Ketosis Modulates HIF1α-Mediated Inflammatory Response in Rat Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1395:75-79. [PMID: 36527617 DOI: 10.1007/978-3-031-14190-4_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Hypoxia inducible factor alpha (HIF1α) is associated with neuroprotection conferred by diet-induced ketosis but the underlying mechanism remains unclear. In this study we use a ketogenic diet in rodents to induce a metabolic state of chronic ketosis, as measured by elevated blood ketone bodies. Chronic ketosis correlates with neuroprotection in both aged and following focal cerebral ischaemia and reperfusion (via middle cerebral artery occlusion, MCAO) in mouse and rat models. Ketone bodies are known to be used efficiently by the brain and metabolism of ketone bodies is associated with increased cytosolic succinate levels that inhibits prolyl hydroxylases allowing HIF1α to accumulate. Ketosis also regulates inflammatory pathways, and HIF1α is reported to be essential for gene expression of interleukin10 (IL10). Therefore we hypothesised that ketosis-stabilised HIF1α modulates the expression of inflammatory cytokines orchestrating neuroprotection. To test changes in cytokine levels in rodent brain, eight-week-old rats were fed either the standard chow diet (SD) or the ketogenic (KG) diet for 4 weeks before ischaemia experiments (MCAO) were performed and the brain tissues were collected. Consistent with our hypothesis, immunoblotting analysis shows IL10 levels were significantly higher in KG diet rat brain compared to SD, whereas the TNFα and IL6 levels were significantly lower in the brains of KG diet fed group.
Collapse
|
70
|
Koivikko P, Posti JP, Mohammadian M, Lagerstedt L, Azurmendi L, Hossain I, Katila AJ, Menon D, Newcombe VFJ, Hutchinson PJ, Maanpää HR, Tallus J, Zetterberg H, Blennow K, Tenovuo O, Sanchez JC, Takala RSK. Potential of heart fatty-acid binding protein, neurofilament light, interleukin-10 and S100 calcium-binding protein B in the acute diagnostics and severity assessment of traumatic brain injury. Emerg Med J 2021; 39:206-212. [PMID: 34916280 DOI: 10.1136/emermed-2020-209471] [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: 01/23/2020] [Accepted: 11/29/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND There is substantial interest in blood biomarkers as fast and objective diagnostic tools for traumatic brain injury (TBI) in the acute setting. METHODS Adult patients (≥18) with TBI of any severity and indications for CT scanning and orthopaedic injury controls were prospectively recruited during 2011-2013 at Turku University Hospital, Finland. The severity of TBI was classified with GCS: GCS 13-15 was classified as mild (mTBI); GCS 9-12 as moderate (moTBI) and GCS 3-8 as severe (sTBI). Serum samples were collected within 24 hours of admission and biomarker levels analysed with high-performance kits. The ability of biomarkers to distinguish between severity of TBI and CT-positive and CT-negative patients was assessed. RESULTS Among 189 patients recruited, neurofilament light (NF-L) was obtained from 175 patients with TBI and 40 controls. S100 calcium-binding protein B (S100B), heart fatty-acid binding protein (H-FABP) and interleukin-10 (IL-10) were analysed for 184 patients with TBI and 39 controls. There were statistically significant differences between levels of all biomarkers between the severity classes, but none of the biomarkers distinguished patients with moTBI from patients with sTBI. Patients with mTBI discharged from the ED had lower levels of IL-10 (0.26, IQR=0.21, 0.39 pg/mL), H-FABP (4.15, IQR=2.72, 5.83 ng/mL) and NF-L (8.6, IQR=6.35, 15.98 pg/mL) compared with those admitted to the neurosurgical ward, IL-10 (0.55, IQR=0.31, 1.42 pg/mL), H-FABP (6.022, IQR=4.19, 20.72 ng/mL) and NF-L (13.95, IQR=8.33, 19.93 pg/mL). We observed higher levels of H-FABP and NF-L in older patients with mTBI. None of the biomarkers or their combinations was able to distinguish CT-positive (n=36) or CT-negative (n=58) patients with mTBI from controls. CONCLUSIONS S100B, H-FABP, NF-L and IL-10 levels in patients with mTBI were significantly lower than in patients with moTBI and sTBI but alone or in combination, were unable to distinguish patients with mTBI from orthopaedic controls. This suggests these biomarkers cannot be used alone to diagnose mTBI in trauma patients in the acute setting.
Collapse
Affiliation(s)
- Pia Koivikko
- Perioperative Services, Intensive Care Medicine, and Pain Management, Turku University Hospital, Turku, Finland .,Anaesthesiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - Jussi P Posti
- Neurocenter, Department of Neurosurgery and Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,Department of Clinical Neurosciences, University of Turku, Turku, Finland
| | - Mehrbod Mohammadian
- Department of Clinical Neurosciences, University of Turku, Turku, Finland.,Neurocenter, Turku Brain Injury Center, Turku University Hospital, Turku, Finland
| | - Linnea Lagerstedt
- Department of Specialities of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Leire Azurmendi
- Department of Specialities of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Iftakher Hossain
- Neurocenter, Department of Neurosurgery and Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Cambridge, UK
| | - Ari J Katila
- Perioperative Services, Intensive Care Medicine, and Pain Management, Turku University Hospital, Turku, Finland.,Anaesthesiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| | - David Menon
- Department of Anaesthesia, University of Cambridge, Cambridge, UK
| | | | - Peter John Hutchinson
- Department of Clinical Neurosciences, Neurosurgery Unit, University of Cambridge, Cambridge, UK
| | - Henna-Riikka Maanpää
- Neurocenter, Department of Neurosurgery and Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,Department of Clinical Neurosciences, University of Turku, Turku, Finland
| | - Jussi Tallus
- Neurocenter, Turku Brain Injury Center, Turku University Hospital, Turku, Finland.,Department of Radiology, University of Turku, Turku, Finland
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg Sahlgrenska Academy, Mölndal, Sweden.,UK Dementia Research Institute, UCL, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, University of Gothenburg Sahlgrenska Academy, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Olli Tenovuo
- Department of Clinical Neurosciences, University of Turku, Turku, Finland.,Neurocenter, Turku Brain Injury Center, Turku University Hospital, Turku, Finland
| | - Jean-Charles Sanchez
- Department of Specialities of Internal Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Riikka S K Takala
- Perioperative Services, Intensive Care Medicine, and Pain Management, Turku University Hospital, Turku, Finland.,Anaesthesiology, Intensive Care, Emergency Care and Pain Medicine, University of Turku, Turku, Finland
| |
Collapse
|
71
|
Ahmadi F, Salmasi Z, Mojarad M, Eslahi A, Tayarani-Najaran Z. G-CSF augments the neuroprotective effect of conditioned medium of dental pulp stem cells against hypoxic neural injury in SH-SY5Y cells. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1743-1752. [PMID: 35432810 PMCID: PMC8976909 DOI: 10.22038/ijbms.2021.60217.13344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022]
Abstract
Objective(s): Dental pulp stem cells (DPSCs) can differentiate into functional neurons and have the potential for cell therapy in neurological diseases. Granulocyte colony-stimulating factor (G-CSF) is a glycoprotein family shown neuroprotective effect in models of nerve damage. we evaluated the protective effects of G-CSF, conditioned media from DPSCs (DPSCs-CM) and conditioned media from transfected DPSCs with plasmid encoding G-CSF (DPSC-CMT) on SH-SY5Y exposed to CoCl2 as a model of hypoxia-induced neural damage. Materials and Methods: SH-SY5Y exposed to CoCl2 were treated with DPSCs-CM, G-CSF, simultaneous combination of DPSCs-CM and G-CSF and finally DPSC-CMT. Cell viability and apoptosis were determined by resazurin (or lactate dehydrogenase (LDH) assay alternatively) and propidium iodide (PI) staining. Western blot analysis was performed to detect changes in apoptotic protein levels. The interleukin-6 and interleukin-10 IL6/IL10 levels were measured with Enzyme-Linked Immunosorbent Assay (ELISA). Results: DPSCs-CM and G-CSF were able to significantly protect SH-SY5Y against neural cell damage caused by CoCl2 according to resazurin and LDH analysis. Also, the percentage of apoptotic cells decreased when SH-SY5Y were treated with DPSCs-CM and G-CSF simultaneously. After transfection of DPSCs with G-CSF plasmid, DPSC-CMT could significantly improve the protection. The amount of β-catenin, cleaved PARP and caspase-3 were significantly decreased and the expression of survivin was considerably increased when hypoxic SH-SY5Y treated with DPSCs-CM plus G-CSF according to Western blot. Decreased level of IL-6/IL-10, which exposed to CoCl2, after treatment with DPSCs-CM indicated the suppression of inflammatory mediators. Conclusion: Combination therapy of G-CSF and DPSCs-CM improved the protective activity.
Collapse
Affiliation(s)
- Farahnaz Ahmadi
- Medical Toxicology Research Center, 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
| | - Majid Mojarad
- Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atieh Eslahi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran,Corresponding author: Zahra Tayarani-Najaran. Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Mashhad, Iran. Tel: +98-51-31801178;
| |
Collapse
|
72
|
Visser K, Koggel M, Blaauw J, van der Horn HJ, Jacobs B, van der Naalt J. Blood-based biomarkers of inflammation in mild traumatic brain injury: A systematic review. Neurosci Biobehav Rev 2021; 132:154-168. [PMID: 34826510 DOI: 10.1016/j.neubiorev.2021.11.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/17/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022]
Abstract
VISSER, K., M. Koggel, J. Blaauw, H.J.v.d. Horn, B. Jacobs, and J.v.d. Naalt. Blood based biomarkers of inflammation in mild traumatic brain injury: A systematic review. NEUROSCI BIOBEHAV REV XX(X) XXX-XXX, 2021. - Inflammation is an important secondary physiological response to traumatic brain injury (TBI). Most of the current knowledge on this response is derived from research in moderate and severe TBI. In this systematic review we summarize the literature on clinical studies measuring blood based inflammatory markers following mild traumatic brain injury (mTBI) and identify the value of inflammatory markers as biomarkers. Twenty-three studies were included. This review suggests a distinct systemic inflammatory response following mTBI, quantifiable within 6 h up to 12 months post-injury. Interleukin-6 is the most promising biomarker for the clinical diagnosis of brain injury while interleukin-10 is a potential candidate for triaging CT scans. The diagnostic and prognostic utility of inflammatory markers may be more fully appreciated as a component of a panel of biomarkers. However, discrepancies in study design, analysis and reporting make it difficult to draw any definite conclusions. For the same reasons, a meta-analysis was not possible. We provide recommendations to follow standardized methodologies to allow for reproducibility of results in future studies.
Collapse
Affiliation(s)
- Koen Visser
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.
| | - Milou Koggel
- Faculty of Science, Department of Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands
| | - Jurre Blaauw
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Harm Jan van der Horn
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Bram Jacobs
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Joukje van der Naalt
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
73
|
Zhao J, Liu S, Yan J, Zhu X. The Impact of Gut Microbiota on Post-Stroke Management. Front Cell Infect Microbiol 2021; 11:724376. [PMID: 34712621 PMCID: PMC8546011 DOI: 10.3389/fcimb.2021.724376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 09/17/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Junyi Zhao
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Siyu Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Jingyi Yan
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Xinzhou Zhu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| |
Collapse
|
74
|
Levin SG, Pershina EV, Bugaev-Makarovskiy NA, Chernomorets IY, Konakov MV, Arkhipov VI. Why Do Levels Of Anti-inflammatory Cytokines Increase During Memory Acquisition? Neuroscience 2021; 473:159-169. [PMID: 34418518 DOI: 10.1016/j.neuroscience.2021.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/14/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022]
Abstract
The role of anti-inflammatory cytokines in the mechanisms of learning and memory, modulation of synaptic plasticity in the mammalian brain has not received sufficient attention. These issues are discussed in this review, and among the many cytokines, attention is paid to the most studied in this respect IL-10, IL-4, IL-13 and TGF-β. The level of anti-inflammatory cytokines in the brain tends to increase during memory acquisition, but the significance of such an increase is unclear. We hypothesize that anti-inflammatory cytokines primarily protect and optimize the functioning of neuronal circuits involved in information processing. The increased local activity of neurons during memory acquisition activates many signaling molecules, and some of them can trigger unwanted processes (including neuroinflammation), but increased levels of anti-inflammatory cytokines prevent this triggering. Each of the anti-inflammatory cytokines plays a specific role in supporting information processing. For example, the role of IL-4 and IL-13 in recruiting T cells to the meninges during training in healthy animals has been most studied. It has also been shown that TGF-β is able to optimize late stage LTP in the hippocampus and support the consolidation of memory traces in behavioral studies. Cytokines have an effect on learning and memory through their influence on neuroplasticity, neurogenesis in the hippocampus and regulation of the neurovascular unit. Experiments have shown such an effect, and the data obtained create the prerequisites for new therapeutic approaches to the correction of cognitive impairments.
Collapse
Affiliation(s)
- Sergey G Levin
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Ekaterina V Pershina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia.
| | - Nickolay A Bugaev-Makarovskiy
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Irina Yu Chernomorets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Maxim V Konakov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| | - Vladimir I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region 142290, Russia
| |
Collapse
|
75
|
Chi CH, Huang YY, Ye SZ, Shao MM, Jiang MX, Yang MY, Wu Q, Shao B, Li XM. Interleukin-10 level is associated with post-stroke depression in acute ischaemic stroke patients. J Affect Disord 2021; 293:254-260. [PMID: 34217963 DOI: 10.1016/j.jad.2021.06.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/16/2021] [Accepted: 06/19/2021] [Indexed: 01/24/2023]
Abstract
BACKGROUND Interleukin-10 (IL-10) is a pathophysiological factor in acute ischaemic stroke (AIS) and is relevant to mood disorders after stroke. We evaluated the predictive value of IL-10 in patients with post-stroke depression (PSD). METHODS A total of 350 stroke patients were recruited at baseline, and 151 AIS patients were screened and completed a 1-month follow-up. Serum IL-10 levels were measured within 24 h of admission. We used the 17-item Hamilton Depression Scale (HAMD-17) to evaluate depression symptoms; PSD was defined as an HAMD score ≥ 7. RESULTS Fifty-one (33.8%) patients showed a more serious stroke degree, larger infarction volume, and poorer daily life activities and prognosis (P < 0.05) and were diagnosed with PSD at the 1-month follow-up. Their IL-10 level decreased significantly compared to the non-PSD group (P < 0.001). After adjusting for confounders, IL-10 could be used as an independent predictor for PSD with an adjusted odds ratio (OR) of 0.615 (95% CI 0.410-0.923, P = 0.019). In addition, the optimal cut-off value of IL-10 was 0.615 pg/mL based on an area under the receiver operating characteristic curve of 0.692 (95% CI 0.604-0.781, P < 0.001), demonstrating that IL-10 could predict the occurrence of PSD. Moreover, IL-10 was an indicator of stroke severity, living ability, and functional outcomes (P < 0.05). LIMITATIONS IL-10 was only measured upon admission; dynamic changes need to be further monitored. This was also a single-centre study with a relatively small sample. CONCLUSIONS Lower IL-10 levels may be used to predict PSD.
Collapse
Affiliation(s)
- Chu-Huai Chi
- Department of Hepatobiliary Surgery, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Yuan-Yuan Huang
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Su-Zhen Ye
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Meng-Meng Shao
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ming-Xia Jiang
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Meng-Yao Yang
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qian Wu
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bei Shao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xian-Mei Li
- Department of Rehabilitation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
76
|
Bao W, Lin Y, Chen Z. The Peripheral Immune System and Traumatic Brain Injury: Insight into the role of T-helper cells. Int J Med Sci 2021; 18:3644-3651. [PMID: 34790036 PMCID: PMC8579286 DOI: 10.7150/ijms.46834] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/17/2021] [Indexed: 12/15/2022] Open
Abstract
Emerging evidence suggests that immune-inflammatory processes are key elements in the physiopathological events associated with traumatic brain injury (TBI). TBI is followed by T-cell-specific immunological changes involving several subsets of T-helper cells and the cytokines they produce; these processes can have opposite effects depending on the disease course and cytokine concentrations. Efforts are underway to identify the T-helper cells and cytokine profiles associated with prognosis. These predictors may eventually serve as effective treatment targets to decrease morbidity and mortality and to improve the management of TBI patients. Here, we review the immunological response to TBI, the possible molecular mechanisms of this response, and therapeutic strategies to address it.
Collapse
Affiliation(s)
| | | | - Zuobing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
77
|
Perinatal blood biomarkers for the identification of brain injury in very low birth weight growth-restricted infants. J Perinatol 2021; 41:2252-2260. [PMID: 34083761 PMCID: PMC8496988 DOI: 10.1038/s41372-021-01112-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/29/2021] [Accepted: 05/18/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To determine if blood biomarkers measured at delivery and shortly after birth can identify growth-restricted infants at risk for developing severe brain injury. STUDY DESIGN In a cohort of very low birth weight neonates, fetal growth restricted (FGR) (birth weight <10%) were compared to non-FGR neonates, and within the FGR group those with brain injury were compared to those without. Biomarkers were measured in cord blood at delivery, and daily for the 1st 5 days of life. RESULT FGR was associated with significantly higher levels of interleukin (IL)-6, IL-8, IL-10, and lower levels of vascular endothelial growth factor (VEGF). FGR and brain injury were associated with significantly higher levels of IL-6, IL-8, IL-10, and glial fibrillary acidic protein (GFAP). CONCLUSION Interleukins may be involved in a common pathway contributing to both the development of growth restriction and brain injury, and GFAP may help identify brain injury within this growth-restricted group.
Collapse
|
78
|
Salivary cytokine profile in patients with ischemic stroke. Sci Rep 2021; 11:17185. [PMID: 34433866 PMCID: PMC8387378 DOI: 10.1038/s41598-021-96739-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/17/2021] [Indexed: 11/23/2022] Open
Abstract
Inflammation plays a crucial role in stroke pathogenesis. Thus, it is not surprising that cytokines, chemokines, and growth factors have been advocated in stroke diagnostics. Our study is the first to evaluate the salivary cytokine profile in patients with ischemic stroke. Twenty-five patients with subacute ischemic stroke and an age-, sex-, and oral hygiene status-matched control group were enrolled in the study. The number of patients was set a priori based on our previous experiment (α = 0.05, test power = 0.9). Salivary concentrations of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), and interleukin 10 (IL-10) were assessed using an ELISA method. We showed that salivary TNF-α and IL-6 were significantly higher, whereas IL-10 content was statistically lower in both non-stimulated (NWS) and stimulated (SWS) whole saliva of ischemic stroke patients. However, evaluation of cytokines in NWS rather than in SWS may be of greater diagnostic value. Of particular note is salivary TNF-α, which may indicate cognitive/physical impairment in post-stroke individuals. This parameter distinguishes stroke patients from healthy controls and correlates with cognitive decline and severity of functional impairment. It also differentiates (with high sensitivity and specificity) stroke patients with normal cognition from mild to moderate cognitive impairment. Saliva may be an alternative to blood for assessing cytokines in stroke patients, although further studies on a larger patient population are needed.
Collapse
|
79
|
Buchmann Godinho D, da Silva Fiorin F, Schneider Oliveira M, Furian AF, Rechia Fighera M, Freire Royes LF. The immunological influence of physical exercise on TBI-induced pathophysiology: Crosstalk between the spleen, gut, and brain. Neurosci Biobehav Rev 2021; 130:15-30. [PMID: 34400178 DOI: 10.1016/j.neubiorev.2021.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a non-degenerative and non-congenital insult to the brain and is recognized as a global public health problem, with a high incidence of neurological disorders. Despite the causal relationship not being entirely known, it has been suggested that multiorgan inflammatory response involving the autonomic nervous system and the spleen-gut brain axis dysfunction exacerbate the TBI pathogenesis in the brain. Thus, applying new therapeutic tools, such as physical exercise, have been described in the literature to act on the immune modulation induced by brain injuries. However, there are caveats to consider when interpreting the effects of physical exercise on this neurological injury. Given the above, this review will highlight the main findings of the literature involving peripheral immune responses in TBI-induced neurological damage and how changes in the cellular metabolism of the spleen-gut brain axis elicited by different protocols of physical exercise alter the pathophysiology induced by this neurological injury.
Collapse
Affiliation(s)
- Douglas Buchmann Godinho
- Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil; Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Fernando da Silva Fiorin
- Programa de Pós-Graduação em Neuroengenharia, Instituto Internacional de Neurociências Edmond e Lily Safra, Instituto Santos Dumont, Macaíba, RN, Brazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Ana Flavia Furian
- Centro de Ciências da Saúde, Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Michele Rechia Fighera
- Laboratório de Bioquímica do Exercício, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil; Centro de Ciências da Saúde, Departamento de Clínica Médica e Pediatria, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | |
Collapse
|
80
|
Akamatsu Y, Chaitin HJ, Hanafy KA. Post-stroke recrudescence-a possible connection to autoimmunity? Rev Neurosci 2021; 33:207-212. [PMID: 34363383 DOI: 10.1515/revneuro-2021-0062] [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: 04/23/2021] [Accepted: 06/27/2021] [Indexed: 11/15/2022]
Abstract
Early recanalization of the occluded vessel is the only efficient intervention that improves outcome after ischemic stroke. In contrast, interventions for chronic issues facing stroke patients are limited. Recent clinical and preclinical studies have shown a correlation between upregulated immune responses to brain antigens and post-stroke recrudescence (PSR), post-stroke fatigue (PSF), and dementia (PSD); all of which are associated with poor long-term stroke outcome. Recent retrospective studies have demonstrated a strong correlation between the onset of PSR and acute infection during acute stroke, suggesting some adaptive immune system mediated pathology. This review will discuss the mechanisms and epidemiology of PSR based on the current clinical and pre-clinical evidence. Accordingly, PSR does appear correlated with populations that are prone to autoimmunity, infection, and subsequent triggers, which corroborate autoimmune responses to self-brain antigens as an underlying mechanism. Moreover, PSR as well as PSF and PSD seem to be partly explained by the development of a neuro-inflammatory response to brain antigens. Therefore, the future of improving long-term stroke outcome could be bright with more accurate pre-clinical models focusing on the role of adaptive immune-mediated post stroke neuroinflammation and more clinical studies of PSR.
Collapse
Affiliation(s)
- Yosuke Akamatsu
- Department of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA02215, USA.,Department of Neurosurgery, Iwate Medical University, 19-1 Uchimaru, Morioka, Iwate020-0023, Japan
| | - Hersh J Chaitin
- College of Medicine, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL33431, USA
| | - Khalid A Hanafy
- College of Medicine, Florida Atlantic University, 777 Glades Rd., Boca Raton, FL33431, USA.,Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA02215, USA.,Division of Neurocritical Care, Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL, USA
| |
Collapse
|
81
|
The Prognostic Determinant of Interleukin-10 in Patients with Acute Ischemic Stroke: An Analysis from the Perspective of Disease Management. DISEASE MARKERS 2021; 2021:6423244. [PMID: 34336007 PMCID: PMC8313368 DOI: 10.1155/2021/6423244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/02/2021] [Accepted: 07/10/2021] [Indexed: 11/18/2022]
Abstract
Background In patients with ischemic stroke, the role of anti-inflammatory cytokine Interleukin-10 (IL-10) in predicting risk and outcomes is not very clear. This study is aimed at prospectively assessing the prognostic determinant value of IL-10 in patients with acute ischemic stroke in a cohort of Chinese people. Methods In a prospective cohort study, consecutive first-ever patients with acute ischemic stroke admitted to our hospital were included from October 2019 to October 2020. The serum level of IL-10 was measured at baseline. A structured follow-up telephone interview was performed on day 90 after admission. Logistic regression analyses were used to assess the prognostic value of IL-10 to predict the poor functional outcome (defined as a modified Rankin Scale score of 3 to 6) and mortality. Results The median age of the 236 enrolled patients was 65 years (interquartile range (IQR), 56-76), and 57.6% were male. There was a negative correlation between the National Institutes of Health Stroke Scale (NIHSS) score and IL-10 serum levels (r (Spearman) = −0.221, P = 0.001). Patients with elevated IL-10 levels (> the highest quartile = 5.24 pg/mL; n = 79) were at significantly lower risk of poor functional outcomes (odds ratio (OR), 0.35; 95% confidence interval (CI), 0.19 to 0.63; P < 0.001) and mortality (OR = 0.24; 95% CI = 0.11–0.52; P < 0.001) compared with patients with IL-10 levels in the lowest three quartiles. Conclusions Reduced serum levels of IL-10 were independently associated with both the clinical severity at admission and a poor functional prognosis in ischemic stroke patients, suggesting that the anti-inflammatory cytokine IL-10 was an important prognostic determinant.
Collapse
|
82
|
Blood biomarkers for neonatal hypoxic-ischemic encephalopathy in the presence and absence of sentinel events. J Perinatol 2021; 41:1322-1330. [PMID: 33024259 PMCID: PMC8021592 DOI: 10.1038/s41372-020-00850-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine if neonatal serum biomarkers representing different pathways of injury differ for cases of HIE of unknown cause to gain insight into timing and mechanism of injury. STUDY DESIGN In this cohort of all neonates with HIE admitted to our NICU, newborns with sentinel events were compared to those without during the 1st 3 days of life. Discard neonatal blood during the 1st 3 days of life was used for analysis. RESULTS Of 277 babies with HIE treated with whole-body hypothermia, 190 (68.6%) had blood available for biomarker analysis. In total, 71 (37.4%) were born within our system, and 119 (62.6%) were transferred in from outside hospitals. Of these babies, 77 (40.5%) had a sentinel event and 113 (59.6%) had no sentinel event. Although the degree of metabolic acidosis was similar, repeated measures analysis showed that during the initial 3 days of life neonates born with HIE in the absence of sentinel events had 41.4% decreased VEGF (p = 0.027) and 62.5% increased IL-10 serum concentrations (p = 0.005). CONCLUSION These changes indicate that neonatal HIE in the absence of sentinel events is not related to an unrecognized acute intrapartum event and is possibly related to chronic hypoxia of lower severity or recovery from a remote event.
Collapse
|
83
|
Jiang Q, Stone CR, Elkin K, Geng X, Ding Y. Immunosuppression and Neuroinflammation in Stroke Pathobiology. Exp Neurobiol 2021; 30:101-112. [PMID: 33972464 PMCID: PMC8118752 DOI: 10.5607/en20033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Over the preceding decades, there have been substantial advances in our knowledge of the pathophysiology of stroke. One such advance has been an increased understanding of the multifarious crosstalk in which the nervous and immune systems engage in order to maintain homeostasis. By interrupting the immune-nervous nexus, it is thought that stroke induces change in both systems. Additionally, it has been found that both innate and adaptive immunosuppression play protective roles against the effects of stroke. The release of danger-/damage-associated molecular patterns (DAMPs) activates Toll-like receptors (TLRs), contributing to the harmful inflammatory effects of ischemia/reperfusion injury after stroke; the Tyro3, Axl, and MerTK (TAM)/Gas6 system, however, has been shown to suppress inflammation via downstream signaling molecules that inhibit TLR signaling. Anti-inflammatory cytokines have also been found to promote neuroprotection following stroke. Additionally, adaptive immunosuppression merits further consideration as a potential endogenous protective mechanism. In this review, we highlight recent studies regarding the effects and mechanism of immunosuppression on the pathophysiology of stroke, with the hope that a better understanding of the function of both of innate and adaptive immunity in this setting will facilitate the development of effective therapies for post-stroke inflammation.
Collapse
Affiliation(s)
- Qian Jiang
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Christopher R Stone
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Kenneth Elkin
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Xiaokun Geng
- China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit 48201, MI, USA.,Department of Research & Development Center, John D. Dingell VA Medical Center, Detroit 48201, MI, USA
| |
Collapse
|
84
|
Katsuki M, Kakizawa Y, Nishikawa A, Yamamoto Y, Uchiyama T. Temporal muscle thickness and area are an independent prognostic factors in patients aged 75 or younger with aneurysmal subarachnoid hemorrhage treated by clipping. Surg Neurol Int 2021; 12:151. [PMID: 33948321 PMCID: PMC8088495 DOI: 10.25259/sni_814_2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/16/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Skeletal muscle mass is an important factor for various diseases' outcomes. As for its indicators, temporal muscle thickness (TMT) and temporal muscle area (TMA) on the head computed tomography are useful, and TMT and TMA were reported as potential prognostic factors for aneurysmal subarachnoid hemorrhage (SAH). We examined the clinical characteristics, including TMT and TMA, of SAH patients aged 75 or younger. METHODS We retrospectively investigated 127 SAH patients with all World Federation of Neurosurgical Societies (WFNS) grades and treated by clipping between 2009 and 2019. Clinical outcome was measured with the modified Rankin Scale (mRS) at 6 months, with favorable outcome defined as mRS 0-2. The associations between the clinical variables and the outcomes were analyzed. RESULTS The mean age was 60.6 (32-74) years, and 65% were women. The mean ± standard deviation of WFNS grade was 2.8 ± 1.4. TMT and TMA were larger in the favorable outcome group than the poor one. Multivariate analysis revealed that age, smoking, WFNS grade, and TMT or TMA were associated with favorable outcome. Receiver operating characteristic analysis found that the threshold of TMT was 4.9 mm in female and 6.7 mm in male, and that of TMA was 193 mm2 in female and 333 mm2 in male. CONCLUSION The odds ratios for TMT and TMA related to clinical outcome were lower than for smoking and WFNS grade; however, on multivariate analysis they remained independent prognostic factors in SAH patients aged 75 or younger treated by clipping. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
| | - Yukinari Kakizawa
- Department of Neurosurgery, Suwa Red Cross Hospital, Suwa, Nagano, Japan
| | | | | | | |
Collapse
|
85
|
Tommy T, Islam AA, Hatta M, Bukhari A, Adhimarta W, Zainuddin AA. Effect of folinic acid on serum homocysteine, TNFα, IL-10, and HMGB1 gene expression in head injury model. Ann Med Surg (Lond) 2021; 65:102273. [PMID: 33996045 PMCID: PMC8100092 DOI: 10.1016/j.amsu.2021.102273] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Background Head injury or traumatic brain injury is the leading cause of mortality and morbidity. Many modalities of neuroprotection had been developed in brain injury but there was no much information regarding folinic acid's effect on neuroinflammation associated with homocysteine, TNFα, IL-10, and HMGB1. Objective This study aimed to investigate whether folinic acid has improving effect on head injury model. Method This study was done in the rat's head injury model using modified Marmarou weight drop model. Fifteen rats were randomized and grouped into 3 groups: Group A: Folinic acid (+), head injury (−); Group B: Folinic acid (−), head injury (+); Group C: Folinic acid (+), head injury (+). Folinic acid was administered intraperitoneally with a dose of 60 mg/m2. Blood samples were taken immediately after head injury (H0), 12 h (H12), and 24 h (H24) after head injury from the lateral vein of tail. Serum level of homocysteine, TNFα, and IL-10 were measured using ELISA, and HMGB1 gene expression was measured with Real-Time RT-PCR. Results This study found serum level of homocysteine, TNFα, IL-10 and HMGB1 gene expression were markedly increased at all time points after head injury. Significantly lower level of serum homocysteine, TNFα, IL-10 and HMGB1 gene expression were found after 24 h treatment with folinic acid in group C compared to those in group B. Conclusion Folinic acid may have anti-inflammatory properties in traumatic brain injury by inhibition of serum level of homocysteine, TNFα, IL-10 and HMGB1 gene expression. An animal study of head injury model using modified Marmarou weight drop model. This three groups study with folinic acid treatment on head injury rats. Folinic acid has anti-inflammatory properties in traumatic brain injury model. Folinic acid inhibits serum level of homocysteine, TNFα, IL-10 and HMGB1expression.
Collapse
Affiliation(s)
- Thomas Tommy
- Department of Neurosurgery, Faculty of Medicine, Universitas Pelita Harapan, Tangerang, Indonesia
| | - Andi A Islam
- Department of Neurosurgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mochammad Hatta
- Molecular Biology and Immunology Laboratory, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Agussalim Bukhari
- Department of Nutritional Sciences, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Willy Adhimarta
- Department of Neurosurgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Andi Alfian Zainuddin
- Department of Public Health and Community Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
86
|
Yu H, Xu Z, Qu G, Wang H, Lin L, Li X, Xie X, Lei Y, He X, Chen Y, Li Y. Hypoxic Preconditioning Enhances the Efficacy of Mesenchymal Stem Cells-Derived Conditioned Medium in Switching Microglia toward Anti-inflammatory Polarization in Ischemia/Reperfusion. Cell Mol Neurobiol 2021; 41:505-524. [PMID: 32424775 DOI: 10.1007/s10571-020-00868-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023]
Abstract
Activation of pro-inflammatory microglia is an important mechanism of the cerebral ischemia-reperfusion (I/R)-induced neuronal injury and dysfunction. Mesenchymal stem cells (MSCs) together with their paracrine factors demonstrated curative potential in immune disorders and inflammatory diseases, as well as in ischemic diseases. However, it remains unclear whether conditioned medium from MSCs could effectively regulate the activation and polarization of microglia exposed to I/R stimulation. In this study, we investigated the effects of conditioned medium from bone marrow MSCs (BMSCs-CM) on I/R-stimulated microglia and the potential mechanism involved, as well as the way to obtain more effective BMSCs-CM. First, cell model of oxygen-glucose deprivation/reoxygenation (OGD/R) was established in microglia to mimic the I/R. BMSCs-CM from different culture conditions (normoxic: 21% O2; hypoxic: 1% O2; hypoxia preconditioning: preconditioning with 1% O2 for 24 h) was used to treat the microglia. Our results showed that BMSCs-CM effectively promoted the survival and alleviated the injury of microglia. Moreover, in microglia exposed to OGD/R, BMSCs-CM inhibited significantly the expression of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6), CD86 and inducible nitric oxide synthase, whereas upregulated the levels of anti-inflammatory cytokine (IL-10), CD206 and Arginase-1. These results suggested that BMSCs-CM promoted the polarization of anti-inflammatory microglia. In particular, BMSCs-CM from cultures with hypoxia preconditioning was more effective in alleviating cell injury and promoting anti-inflammatory microglia polarization than BMSCs-CM from normoxic cultures and from hypoxic cultures. Furthermore, inhibition of exosomes secretion could largely mitigate these effects of BMSCs-CM. In conclusion, our results suggested that hypoxia preconditioning of BMSCs could enhance the efficacy of BMSCs-CM in alleviating OGD/R-induced injury and in promoting the anti-inflammatory polarization of microglia, and these beneficial effects of BMSCs-CM owed substantially to exosomes.
Collapse
Affiliation(s)
- Han Yu
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Pathology, The Affiliated Hospital of Hubei University of Medicine, The First People's Hospital of Xiangyang, Xiangyang, 441000, China
| | - Zhihong Xu
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Gaojing Qu
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huimin Wang
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lulu Lin
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xianyu Li
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaolin Xie
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yifeng Lei
- The Institute of Technological Sciences & School of Power and Mechanical Engineering, Wuhan University, Wuhan, 430072, China
| | - Xiaohua He
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yun Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yinping Li
- Department of Pathophysiology & Hubei Province Key Laboratory of Allergy and Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
87
|
Li C, Liu Y, Zhou X, Wang Y. A paper-based SERS assay for sensitive duplex cytokine detection towards the atherosclerosis-associated disease diagnosis. J Mater Chem B 2021; 8:3582-3589. [PMID: 31872850 DOI: 10.1039/c9tb02469g] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Atherosclerosis (AS) is the most common factor causing many cardiovascular and cerebrovascular diseases and has received considerable attention. The occurrence mechanism of AS is uncertain because it is a choronically pathological process that is influenced by multi-aspects, among which cytokines play the key roles in regulating the processes of the immune system. For example, two key cytokines, namely, IL-10 and MCP-1 (chemokine), which are involved in AS progression with varied levels, can be used for AS status monitoring and early diagnosis of AS-associated diseases. Hence, a new paper-based, surface-enhanced Raman spectroscopy (SERS) sensing platform was established for the detection of these two key cytokines. By combining a nanoporous networking membrane as the substrate and SERS nanotags as the probe for signal reading, together with a sandwich design, sensitive and specific identification and quantification of cytokine targets in human serum were achieved with excellent sensing characteristics. The lowest detectable concentration was determined to be 0.1 pg mL-1 for both IL-10 and MCP-1 in human serum. The assay also exhibits high specificity towards target cytokine detection, with low-nonspecific binding and acceptable cross-reactivity in the presence of other structurally similar targets. Finally, the practicability was validated by performing duplex detection in human serum, which further demonstrates the high specificity of the assay for the detection of target cytokines. Taken together, these promising results illustrate that this developed sensing assay is a candidate for clinical multi-target analysis in real environments.
Collapse
Affiliation(s)
- Chunxia Li
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | |
Collapse
|
88
|
Toledo RS, Stein DJ, Sanches PRS, da Silva LS, Medeiros HR, Fregni F, Caumo W, Torres ILS. rTMS induces analgesia and modulates neuroinflammation and neuroplasticity in neuropathic pain model rats. Brain Res 2021; 1762:147427. [PMID: 33737061 DOI: 10.1016/j.brainres.2021.147427] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/28/2021] [Accepted: 03/07/2021] [Indexed: 12/30/2022]
Abstract
Neuropathic pain (NP) is related to the presence of hyperalgesia, allodynia, and spontaneous pain, affecting 7%-10% of the general population. Repetitive transcranial magnetic stimulation (rTMS) is applied for NP relief, especially in patients with refractory pain. As NP response to existing treatments is often insufficient, we aimed to evaluate rTMS treatment on the nociceptive response of rats submitted to an NP model and its effect on pro-and anti-neuroinflammatory cytokine and neurotrophin levels. A total of 106 adult male Wistar rats (60 days old) were divided into nine experimental groups: control, control + sham rTMS, control + rTMS, sham NP, sham neuropathic pain + sham rTMS, sham neuropathic pain + rTMS, NP, neuropathic pain + sham rTMS, and neuropathic pain + rTMS. NP establishment was achieved 14 days after the surgery to establish chronic constriction injury (CCI) of the sciatic nerve. Rats were treated with 5 min daily sessions of rTMS for eight consecutive days. Nociceptive behavior was assessed using von Frey and hot plate tests at baseline, after NP establishment, and post-treatment. Biochemical assays to assess the levels of brain-derived neurotrophic factor (BDNF), tumor necrosis factor-alpha (TNF-α), and interleukin (IL)-10, were performed in the prefrontal cortex (PFC) and spinal cord tissue homogenates. rTMS treatment promoted a partial reversal of mechanical allodynia and total reversal of thermal hyperalgesia induced by CCI. Moreover, rTMS increased the levels of BDNF, TNF-α, and IL-10 in the PFC. rTMS may be a promising tool for the treatment of NP. The alterations induced by rTMS on neurochemical parameters may have contributed to the analgesic effect presented.
Collapse
Affiliation(s)
- Roberta Ströher Toledo
- Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica - Instituto de Ciências Básicas da Saúde - Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Dirson João Stein
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Paulo Roberto Stefani Sanches
- Serviço de Pesquisa e Desenvolvimento em Engenharia Biomédica, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lisiane Santos da Silva
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Helouise Richardt Medeiros
- Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Fregni
- Laboratory of Neuromodulation, Department of Physical Medicine & Rehabilitation, Spaulding Rehabilitation Hospital and Massachusetts General Hospital, Harvard University, Boston, MA, United States
| | - Wolnei Caumo
- Programa de Pós-Graduação em Medicina: Ciências Médicas - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Iraci L S Torres
- Programa de Pós-Graduação em Ciências Biológicas: Farmacologia e Terapêutica - Instituto de Ciências Básicas da Saúde - Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Farmacologia da Dor e Neuromodulação: Investigações Pré-clínicas - Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Programa de Pós-Graduação em Medicina: Ciências Médicas - Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
89
|
Vincenti I, Merkler D. New advances in immune components mediating viral control in the CNS. Curr Opin Virol 2021; 47:68-78. [PMID: 33636592 DOI: 10.1016/j.coviro.2021.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 11/26/2022]
Abstract
Protective immune responses in the central nervous system (CNS) must act efficiently but need to be tightly controlled to avoid excessive damage to this vital organ. Under homeostatic conditions, the immune surveillance of the CNS is mediated by innate immune cells together with subsets of memory lymphocytes accumulating over lifetime. Accordingly, a wide range of immune responses can be triggered upon pathogen infection that can be associated with devastating clinical outcomes, and which most frequently are due to neurotropic viruses. Here, we discuss recent advances about our understanding of anti-viral immune responses with special emphasis on mechanisms operating in the various anatomical compartments of the CNS.
Collapse
Affiliation(s)
- Ilena Vincenti
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland
| | - Doron Merkler
- University of Geneva, Department of Pathology and Immunology, Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1211 Geneva, Switzerland.
| |
Collapse
|
90
|
Lackner I, Weber B, Haffner-Luntzer M, Hristova S, Gebhard F, Lam C, Morioka K, Marcucio RS, Miclau T, Kalbitz M. Systemic and local cardiac inflammation after experimental long bone fracture, traumatic brain injury and combined trauma in mice. J Orthop Translat 2021; 28:39-46. [PMID: 33717980 PMCID: PMC7906881 DOI: 10.1016/j.jot.2020.12.003] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 11/28/2022] Open
Abstract
Background Trauma is the leading cause of death and disability worldwide, especially in the young population. Cardiac injuries are an independent predictor for a poor overall outcome after trauma. The aim of the present study was to analyze systemic inflammation as well as local cardiac inflammation after experimental limb-, neuro- and combined trauma in mice. Methods Male C57BL/6 mice received either a closed tibia fracture (Fx), isolated traumatic brain injury (TBI) or a combination of both (Fx + TBI). Control animals underwent sham procedure. After 6 and 24 h, systemic levels of inflammatory mediators were analyzed, respectively. Locally, cardiac inflammation and cardiac structural alterations were investigated in left ventricular tissue of mice 6 and 24 h after trauma. Results Mice showed enhanced systemic inflammation after combined trauma, which was manifested by increased levels of KC, MCP-1 and G-CSF. Locally, mice exhibited increased expression of inflammatory cytokines (IL-1β, TNF) in heart tissue, which was probably mediated via toll-like receptor (TLR) signaling. Furthermore, mice demonstrated a redistribution of connexin 43 in cardiac tissue, which appeared predominantly after combined trauma. Besides inflammation and structural cardiac alterations, expression of glucose transporter 4 (GLUT4) mRNA was increased in the heart early after TBI and after combination of TBI and limb fracture, indicating a modification of energy metabolism. Early after combination of TBI and tibia fracture, nitrosative stress was increased, manifested by elevation of nitrotyrosine in cardiac tissue. Finally, mice showed a trend of increased systemic levels of cardiac troponin I and heart-fatty acid binding protein (HFABP) after combined trauma, which was associated with a significant decrease of troponin I and HFABP mRNA expression in cardiac tissue after TBI and combination of TBI and limb fracture. Conclusion Mice exhibited early cardiac alterations as well as alterations in cardiac glucose transporter expression, indicating a modification of energy metabolism, which might be linked to increased systemic- and local cardiac inflammation after limb-, neuro- and combined trauma. These cardiac alterations might predispose individuals for secondary cardiac damage after trauma that might compromise cardiac function after TBI and long bone fracture. Translational potential statement Injuries to the head and extremities frequently occur after severe trauma. In our study, we analyzed the effects of closed tibia fracture, isolated TBI, and the combination of both injuries with regard to the development of post-traumatic secondary cardiac injuries.
Collapse
Affiliation(s)
- Ina Lackner
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Birte Weber
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany.,Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Simona Hristova
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany
| | - Charles Lam
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Kazuhito Morioka
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Ralph S Marcucio
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Theodore Miclau
- Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Miriam Kalbitz
- Department of Traumatology, Hand-, Plastic- and Reconstructive Surgery, University Medical Center Ulm, Ulm, Germany.,Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
91
|
Dong G, Li C, Hu Q, Wang Y, Sun J, Gao F, Yang M, Sun B, Mao L. Low-Dose IL-2 Treatment Affords Protection against Subarachnoid Hemorrhage Injury by Expanding Peripheral Regulatory T Cells. ACS Chem Neurosci 2021; 12:430-440. [PMID: 33476129 DOI: 10.1021/acschemneuro.0c00611] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Subarachnoid hemorrhage (SAH) is considered a devastating disease, leaving survivors with lifelong neurological impairment. With increased knowledge that regulatory T cells (Tregs) provide protection against stroke, novel agents which could expand Treg populations have been assessed in terms of the potential clinical neuroprotection effect. Using a rat SAH model, we investigated the number variation of Tregs induced by SAH and the protective effect of low-dose interleukin-2 (IL-2) treatment on the SAH model. We observed that the number of peripheral Tregs significantly decreased soon after SAH, accompanying with reactivity recovery after 3 days. Our results also revealed that low-dose IL-2 treatment not only elevated Tregs numbers but significantly reduced neuronal injury and improved neurological functions up to 21 days (d) after SAH. Furthermore, compared with PBS-treatment group, cerebral proinflammatory factors and peripheral neutrophils were significantly suppressed by low-dose IL-2 after SAH. Therefore, the results suggest that low-dose IL-2 treatment is a novel and clinically feasible immunotherapy to improve long-term outcomes after SAH, perhaps via up-regulating Treg population to suppress neuroinflammation induced by SAH.
Collapse
Affiliation(s)
- Guoping Dong
- Department of Neurology, Second Affiliated Hospital; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
- Department of Neurology, Caoxian People’s Hospital, Heze, Shandong 061000, China
| | - Cong Li
- College of Medical Information Enginerring, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Quan Hu
- Department of Neurosurgery, Taian City Central Hospital, Taian, Shandong 271000, China
| | - Yuan Wang
- Department of Neurology, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China
| | - Jingyi Sun
- Department of Neurology, Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250021, China
| | - Feng Gao
- Department of Neurology, Second Affiliated Hospital; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Mingfeng Yang
- Department of Neurology, Second Affiliated Hospital; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Baoliang Sun
- Department of Neurology, Second Affiliated Hospital; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| | - Leilei Mao
- Department of Neurology, Second Affiliated Hospital; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong 271000, China
| |
Collapse
|
92
|
Lingam I, Avdic-Belltheus A, Meehan C, Martinello K, Ragab S, Peebles D, Barkhuizen M, Tann CJ, Tachtsidis I, Wolfs TGAM, Hagberg H, Klein N, Fleiss B, Gressens P, Golay X, Kramer BW, Robertson NJ. Serial blood cytokine and chemokine mRNA and microRNA over 48 h are insult specific in a piglet model of inflammation-sensitized hypoxia-ischaemia. Pediatr Res 2021; 89:464-475. [PMID: 32521540 DOI: 10.1038/s41390-020-0986-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/06/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Exposure to inflammation exacerbates injury in neonatal encephalopathy (NE). We hypothesized that brain biomarker mRNA, cytokine mRNA and microRNA differentiate inflammation (E. coli LPS), hypoxia (Hypoxia), and inflammation-sensitized hypoxia (LPS+Hypoxia) in an NE piglet model. METHODS Sixteen piglets were randomized: (i) LPS 2 μg/kg bolus; 1 μg/kg infusion (LPS; n = 5), (ii) Saline with hypoxia (Hypoxia; n = 6), (iii) LPS commencing 4 h pre-hypoxia (LPS+Hypoxia; n = 5). Total RNA was acquired at baseline, 4 h after LPS and 1, 3, 6, 12, 24, 48 h post-insult (animals euthanized at 48 h). Quantitative PCR was performed for cytokines (IL1A, IL6, CXCL8, IL10, TNFA) and brain biomarkers (ENO2, UCHL1, S100B, GFAP, CRP, BDNF, MAPT). MicroRNA was detected using GeneChip (Affymetrix) microarrays. Fold changes from baseline were compared between groups and correlated with cell death (TUNEL) at 48 h. RESULTS Within 6 h post-insult, we observed increased IL1A, CXCL8, CCL2 and ENO2 mRNA in LPS+Hypoxia and LPS compared to Hypoxia. IL10 mRNA differentiated all groups. Four microRNAs differentiated LPS+Hypoxia and Hypoxia: hsa-miR-23a, 27a, 31-5p, 193-5p. Cell death correlated with TNFA (R = 0.69; p < 0.01) at 1-3 h and ENO2 (R = -0.69; p = 0.01) at 48 h. CONCLUSIONS mRNA and miRNA differentiated hypoxia from inflammation-sensitized hypoxia within 6 h in a piglet model. This information may inform human studies to enable triage for tailored neuroprotection in NE. IMPACT Early stratification of infants with neonatal encephalopathy is key to providing tailored neuroprotection. IL1A, CXCL8, IL10, CCL2 and NSE mRNA are promising biomarkers of inflammation-sensitized hypoxia. IL10 mRNA levels differentiated all three pathological states; fold changes from baseline was the highest in LPS+Hypoxia animals, followed by LPS and Hypoxia at 6 h. miR-23, -27, -31-5p and -193-5p were significantly upregulated within 6 h of a hypoxia insult. Functional analysis highlighted the diverse roles of miRNA in cellular processes.
Collapse
Affiliation(s)
- Ingran Lingam
- Neonatology, Institute for Women's Health, University College London, London, UK
| | | | - Christopher Meehan
- Neonatology, Institute for Women's Health, University College London, London, UK
| | - Kathryn Martinello
- Neonatology, Institute for Women's Health, University College London, London, UK.,Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Sara Ragab
- Neonatology, Institute for Women's Health, University College London, London, UK
| | - Donald Peebles
- Maternal Fetal Medicine, Institute for Women's Health, University College London, London, UK
| | - Melinda Barkhuizen
- Department of Pediatrics, University of Maastricht, Maastricht, The Netherlands
| | - Cally J Tann
- Neonatology, Institute for Women's Health, University College London, London, UK.,Maternal Adolescent, Reproductive and Child Health (MARCH) Centre, Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Ilias Tachtsidis
- Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Tim G A M Wolfs
- Department of Pediatrics, University of Maastricht, Maastricht, The Netherlands
| | - Henrik Hagberg
- Centre of Perinatal Medicine & Health, Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
| | - Nigel Klein
- Paediatric Infectious Diseases & Immunology, Institute of Child Health, University College London, London, UK
| | - Bobbi Fleiss
- Centre for the Developing Brain, Kings College London, London, UK
| | - Pierre Gressens
- Centre for the Developing Brain, Kings College London, London, UK.,PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, 75019, Paris, France
| | - Xavier Golay
- Department of Brain Repair & Rehabilitation, Institute of Neurology, University College London, London, UK
| | - Boris W Kramer
- Maternal Fetal Medicine, Institute for Women's Health, University College London, London, UK
| | - Nicola J Robertson
- Neonatology, Institute for Women's Health, University College London, London, UK.
| |
Collapse
|
93
|
Rahman Z, Dandekar MP. Crosstalk between gut microbiome and immunology in the management of ischemic brain injury. J Neuroimmunol 2021; 353:577498. [PMID: 33607506 DOI: 10.1016/j.jneuroim.2021.577498] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/30/2020] [Accepted: 01/21/2021] [Indexed: 02/06/2023]
Abstract
Ischemic brain injury is a serious neurological complication, which accrues an immense activation of neuroinflammatory responses. Several lines of research suggested the interconnection of gut microbiota perturbation with the activation of proinflammatory mediators. Intestinal microbial communities also interchange information with the brain through various afferent and efferent channels and microbial by-products. Herein, we discuss the different microelements of gut microbiota and its connection with the host immune system and how change in immune-microbial signatures correlates with the stroke incidence and post-injury neurological sequelae. The activated inflammatory cells increase the production of proinflammatory cytokines, chemokines, proteases and adhesive proteins that are involved in the systemic inflammation, blood brain barrier disruption, gut dysbiosis and aggravation of ischemic brain injury. We suggest that fine-tuning of commensal gut microbiota (eubiosis) may regulate the activation of CNS resident cells like microglial, astrocytes, mast cells and natural killer cells.
Collapse
Affiliation(s)
- Ziaur Rahman
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
94
|
Katsuki M, Ozaki D, Narita N, Ishida N, Watanabe O, Cai S, Shimabukuro S, Tominaga T. Unilateral posterior reversible encephalopathy syndrome characterized with a long and gradually exacerbating course over 3 years and that presented propofol infusion syndrome - A case report. Surg Neurol Int 2021; 12:19. [PMID: 33500834 PMCID: PMC7827507 DOI: 10.25259/sni_853_2020] [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: 11/29/2020] [Accepted: 12/25/2020] [Indexed: 12/13/2022] Open
Abstract
Background: Posterior reversible encephalopathy syndrome (PRES) is characterized by acute neurological symptoms and vasogenic edema, and most patients wholly recover. We report a unilateral PRES patient characterized by a gradual onset followed by propofol infusion syndrome (PRIS) due to general anesthesia therapy. Case Description: A 32-year-old woman had ovarian dysfunction treated by Kaufmann’s treatment for 17 years. Three years ago, she developed seizures, and photophobia and myoclonus sometimes occurred. This time, she had strong photophobia and nausea for 3 months and then developed tonic-clonic seizures for 3 min. Her blood pressure and laboratory test on admission were all within normal limits. She presented no neurological deficits at admission, but the T2-weighted image (T2WI) showed a high-intensity area (HIA), and arterial spin labeling (ASL) image described cerebral blood flow (CBF) increase in the left parieto-occipital region. We diagnosed PRES and started anticonvulsants, antihypertensive, and steroid pulse therapy. However, her aphasia and neuroimaging findings worsened, so we started general anesthesia treatment with propofol on day 29. On day 32, she suddenly developed multiple organ dysfunctions due to PRIS. After intensive care with other sedatives over 2 months, the systemic status and neurological symptoms gradually improved almost as before the onset. On day 90, HIA in the T2WI in the lesion became small, and CBF was severely downregulated in the ASL image. Conclusion: Unilateral PRES’s pathophysiology and the association with the female hormone remain unknown. Some patients undergo gradual onset and long-term courses, and we should care for PRIS during PRES treatment.
Collapse
Affiliation(s)
- Masahito Katsuki
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Dan Ozaki
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Norio Narita
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Naoya Ishida
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Ohmi Watanabe
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Siqi Cai
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Shinya Shimabukuro
- Department of Neurosurgery, Kesennuma City Hospital, Kesennuma, Miyagi, Japan
| | - Teiji Tominaga
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Aobaku, Sendai, Miyagi, Japan
| |
Collapse
|
95
|
Kirsten K, Pompermaier A, Koakoski G, Mendonça-Soares S, da Costa RA, Maffi VC, Kreutz LC, Barcellos LJG. Acute and chronic stress differently alter the expression of cytokine and neuronal markers genes in zebrafish brain. Stress 2021; 24:107-112. [PMID: 32013653 DOI: 10.1080/10253890.2020.1724947] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We report the effects of acute and chronic stress on the expression of selective immune-related genes and markers of neuronal function in the brain of the zebrafish (Danio rerio). Fish were distributed into three groups: the non-stressed control group; the acute stress (AS) group, submitted to a single stressing episode; and the unpredictable chronic stress (UCS) group, submitted to two daily stressing episodes of alternating times and types of stress. The stressing protocols were applied for a period of 14 days. The UCS protocol triggered the expression of the pro-inflammatory cytokine genes IL-1β and TNF-α, the anti-inflammatory cytokine IL-10 (negative feedback from the immune system), reduction in cFOS gene expression, and caused neuro-inflammation. The AS protocol had no effect on gene expression. Altered expression of cytokine genes, as observed in our study, correlates with several pathologies associated with neuro-inflammation, and the reduction of cFOS gene expression may indicate the occurrence of reduced neuronal plasticity. Our study further extends our knowledge about the interaction of the immune system and the different forms of stress.
Collapse
Affiliation(s)
- Karina Kirsten
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brasil
| | - Aline Pompermaier
- Programa de Pós-Graduação em Ciências Ambientais, Instituto de Ciências Biológicas, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
- Programa de Pós-Graduação em Bioexperimentação, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), Passo Fundo, Brasil
| | - Gessi Koakoski
- Programa de Pós-Graduação em Bioexperimentação, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), Passo Fundo, Brasil
| | - Suelen Mendonça-Soares
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brasil
| | - Roberta Angnes da Costa
- Curso de Medicina Veterinária, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
| | - Victória Costa Maffi
- Curso de Medicina Veterinária, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
| | - Luiz Carlos Kreutz
- Programa de Pós-Graduação em Bioexperimentação, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), Passo Fundo, Brasil
- Curso de Medicina Veterinária, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
| | - Leonardo José Gil Barcellos
- Programa de Pós-Graduação em Farmacologia, Universidade Federal de Santa Maria (UFSM), Santa Maria, Brasil
- Programa de Pós-Graduação em Ciências Ambientais, Instituto de Ciências Biológicas, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
- Programa de Pós-Graduação em Bioexperimentação, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo (UPF), Passo Fundo, Brasil
- Curso de Medicina Veterinária, Faculdade de Agronomia e Medicina Veterinária, Universidade de Passo Fundo, (UPF), Passo Fundo, Brasil
| |
Collapse
|
96
|
Sethuraman A, Rao P, Pranay A, Xu K, LaManna JC, Puchowicz MA. Chronic Ketosis Modulates HIF1α-Mediated Inflammatory Response in Rat Brain. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1269:3-7. [PMID: 33966187 DOI: 10.1007/978-3-030-48238-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hypoxia inducible factor alpha (HIF1α) is associated with neuroprotection conferred by diet-induced ketosis, but the underlying mechanism remains unclear. In this study, we use a ketogenic diet in rodents to induce a metabolic state of chronic ketosis, as measured by elevated blood ketone bodies. Chronic ketosis correlates with neuroprotection in both aged and following focal cerebral ischemia and reperfusion (via middle cerebral artery occlusion, MCAO) in mouse and rat models. Ketone bodies are known to be used efficiently by the brain, and metabolism of ketone bodies is associated with increased cytosolic succinate levels that inhibits prolyl hydroxylases allowing HIF1α to accumulate. Ketosis also regulates inflammatory pathways, and HIF1α is reported to be essential for gene expression of interleukin 10 (IL10). Therefore, we hypothesized that ketosis-stabilized HIF1α modulates the expression of inflammatory cytokines orchestrating neuroprotection. To test changes in cytokine levels in rodent brain, 8-week-rats were fed either the standard chow diet (SD) or the KG diet for 4 weeks before ischemia experiments (MCAO) were performed and the brain tissues were collected. Consistent with our hypothesis, immunoblotting analysis shows IL10 levels were significantly higher in KG diet rat brain compared to SD, whereas the TNFα and IL6 levels were significantly lower in the brains of KG diet-fed group.
Collapse
Affiliation(s)
- Aarti Sethuraman
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Prahlad Rao
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Atul Pranay
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kui Xu
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Joseph C LaManna
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Michelle A Puchowicz
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA.
- Department of Nutrition, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
97
|
Rong T, He M, Hua Y, Chen D, Chen M. Associations of Interleukin 10, Matrix Metallopeptidase 9, and Legumain with Blood Pressure Variability and Neurologic Outcomes in Patients with Ischemic Stroke. Int J Gen Med 2020; 13:1595-1602. [PMID: 33364822 PMCID: PMC7751781 DOI: 10.2147/ijgm.s285003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/30/2020] [Indexed: 01/01/2023] Open
Abstract
Background Timely diagnosis and treatment are crucial to improve prognosis of ischemic stroke, making exploring factors associated with prognosis essential. Blood pressure variability (BPV) was reported to be associated with neurologic outcome, and basic researches on cardiovascular diseases found abnormal expression patterns of several factors including interleukin 10 (IL-10), matrix metallopeptidase 9 (MMP-9), and legumain which might be related to abnormal BPV but yet to prove in ischemic stroke. The study aimed to investigate whether IL-10, MMP-9, and legumain are associated with BPV and neurologic outcome of patients with ischemic stroke. Patients and Methods Newly diagnosed ischemic stroke patients admitted to the department of neurology, Shidong Hospital of Yangpu District in Shanghai between July 2017 and January 2019 were enrolled. IL-10, MMP-9, and legumain were detected and BPV was assessed within 72 hours after admission. All the patients were followed for neurologic outcomes at discharge and 6 months after admission based on the Modified Rankin Scale (MRS). Correlations of IL-10, MMP-9, and legumain with BPV were examined by Spearman correlation coefficient, and their associations with neurologic outcomes were evaluated by multivariable linear regression. Results A total of 349 patients with ischemic stroke were enrolled with an average age of 72.97±11.47 years. Compared with non-progressive ischemic stroke, patients with progressive ischemic stroke had significantly higher IL-10, MMP-9, and legumain on admission. MMP-9 was found to be positively correlated with BPV while no significant correlation was found for IL-10 and legumain with BPV. MMP-9 was associated with progressive ischemic stroke [β=0.23 (95% CI 0.11-0.35) per SD increase for MRS at discharge, and β=0.32 (95% CI 0.20-0.43) per SD increase for MRS at 6 months after admission]. Conclusion Increased MMP-9 was associated with increased BPV and progressive ischemic stroke for patients with ischemic stroke, which might partially explain the effect of BPV on neurologic outcomes.
Collapse
Affiliation(s)
- Tianyi Rong
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| | - Min He
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| | - Yun Hua
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| | - Deyan Chen
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| | - Miao Chen
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200438, People's Republic of China
| |
Collapse
|
98
|
Kang R, Gamdzyk M, Lenahan C, Tang J, Tan S, Zhang JH. The Dual Role of Microglia in Blood-Brain Barrier Dysfunction after Stroke. Curr Neuropharmacol 2020; 18:1237-1249. [PMID: 32469699 PMCID: PMC7770642 DOI: 10.2174/1570159x18666200529150907] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/26/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
It is well-known that stroke is one of the leading causes of death and disability all over the world. After a stroke, the blood-brain barrier subsequently breaks down. The BBB consists of endothelial cells surrounded by astrocytes. Microglia, considered the long-living resident immune cells of the brain, play a vital role in BBB function. M1 microglia worsen BBB disruption, while M2 microglia assist in repairing BBB damage. Microglia can also directly interact with endothelial cells and affect BBB permeability. In this review, we are going to discuss the mechanisms responsible for the dual role of microglia in BBB dysfunction after stroke.
Collapse
Affiliation(s)
- Ruiqing Kang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA,Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Sheng Tan
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
99
|
Farr SA, Cuzzocrea S, Esposito E, Campolo M, Niehoff ML, Doyle TM, Salvemini D. Adenosine A 3 receptor as a novel therapeutic target to reduce secondary events and improve neurocognitive functions following traumatic brain injury. J Neuroinflammation 2020; 17:339. [PMID: 33183330 PMCID: PMC7659122 DOI: 10.1186/s12974-020-02009-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a common pathological condition that presently lacks a specific pharmacological treatment. Adenosine levels rise following TBI, which is thought to be neuroprotective against secondary brain injury. Evidence from stroke and inflammatory disease models suggests that adenosine signaling through the G protein-coupled A3 adenosine receptor (A3AR) can provide antiinflammatory and neuroprotective effects. However, the role of A3AR in TBI has not been investigated. Methods Using the selective A3AR agonist, MRS5980, we evaluated the effects of A3AR activation on the pathological outcomes and cognitive function in CD1 male mouse models of TBI. Results When measured 24 h after controlled cortical impact (CCI) TBI, male mice treated with intraperitoneal injections of MRS5980 (1 mg/kg) had reduced secondary tissue injury and brain infarction than vehicle-treated mice with TBI. These effects were associated with attenuated neuroinflammation marked by reduced activation of nuclear factor of kappa light polypeptide gene enhancer in B cells (NFκB) and MAPK (p38 and extracellular signal-regulated kinase (ERK)) pathways and downstream NOD-like receptor pyrin domain-containing 3 inflammasome activation. MRS5980 also attenuated TBI-induced CD4+ and CD8+ T cell influx. Moreover, when measured 4–5 weeks after closed head weight-drop TBI, male mice treated with MRS5980 (1 mg/kg) performed significantly better in novel object-placement retention tests (NOPRT) and T maze trials than untreated mice with TBI without altered locomotor activity or increased anxiety. Conclusion Our results provide support for the beneficial effects of small molecule A3AR agonists to mitigate secondary tissue injury and cognitive impairment following TBI.
Collapse
Affiliation(s)
- Susan A Farr
- Veterans Affairs Medical Center, 915 N Grand Blvd, St. Louis, MO, 63106, USA.,Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Salvatore Cuzzocrea
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Emanuela Esposito
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Michela Campolo
- Department of Clinical and Experimental Medicine and Pharmacology, University of Messina, 98122, Messina, Italy
| | - Michael L Niehoff
- Department of Internal Medicine, Division of Geriatric Medicine, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Timothy M Doyle
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA
| | - Daniela Salvemini
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA. .,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 S. Grand Blvd, St. Louis, MO, 63104, USA.
| |
Collapse
|
100
|
Vazifehkhah S, Khanizadeh AM, Mojarad TB, Nikbakht F. The possible role of progranulin on anti-inflammatory effects of metformin in temporal lobe epilepsy. J Chem Neuroanat 2020; 109:101849. [DOI: 10.1016/j.jchemneu.2020.101849] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 12/21/2022]
|