1
|
Abbaspour S, Fahanik-Babaei J, Adeli S, Hermann DM, Sardari M. Acute nicotine exposure attenuates neurological deficits, ischemic injury and brain inflammatory responses and restores hippocampal long-term potentiation in ischemic stroke followed by lipopolysaccharide-induced sepsis-like state. Exp Neurol 2024; 382:114946. [PMID: 39278587 DOI: 10.1016/j.expneurol.2024.114946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/11/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
Ischemic stroke is followed by an increased susceptibility to bacterial infections, which exacerbate histological stroke outcome, neurological deficits and memory impairment due to increased neuroinflammation and neurotransmitter dysfunction. Pharmacological activation of nicotinic acetylcholine receptors was suggested to mitigate brain inflammatory responses in ischemic stroke. The functional responses associated with nicotinic acetylcholine receptor activation were unknown. In this study, male NMRI mice subjected to transient intraluminal middle cerebral artery occlusion (MCAO) were intraperitoneally exposed to vehicle treatment or Escherichia coli lipopolysaccharide (LPS; 4 mg/kg)-induced sepsis-like state 24 h post-MCAO, followed by intraperitoneal administration of vehicle or nicotine (0.5 mg/kg) 30 min later. Over 96 h, rectal temperature, neurological deficits, spontaneous locomotor activity, working memory, ischemic injury, synaptic plasticity, and brain inflammatory responses were evaluated by temperature measurement, behavioral analysis, infarct volumetry, electrophysiological recordings, and polymerase-chain reaction analysis. LPS-induced sepsis induced hypothermia, increased general and focal neurological deficits, reduced spontaneous exploration behavior, reduced working memory, and increased infarct volume post-MCAO. Additional treatment with nicotine attenuated LPS-induced hypothermia, reduced neurological deficits, restored exploration behavior, restored working memory, and reduced infarct volume. Local field potential recordings revealed that LPS-induced sepsis decreased long-term potentiation (LTP) in the dentate gyrus post-MCAO, whereas concomitant nicotine exposure restored LTP in the contralateral dentate gyrus. LPS-induced sepsis increased microglial/ macrophage Iba-1 mRNA and astrocytic GFAP mRNA levels post-MCAO, whereas add-on nicotine treatment reduced astrocytic GFAP mRNA. Taken together, these findings indicate that acute nicotine exposure enhances functional stroke recovery. Future studies will have to evaluate the effects of (1) chronic nicotine exposure, a clinically relevant vascular risk factor, and (2) the cessation of nicotine exposure, which is widely recommended post-stroke, but might have detrimental effects in the early stroke recovery phase.
Collapse
Affiliation(s)
- Sonia Abbaspour
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Javad Fahanik-Babaei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Adeli
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany.
| | - Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
2
|
Błaż M, Natorska J, Bembenek JP, Członkowska A, Ząbczyk M, Polak M, Undas A. Elevated lipopolysaccharide level is largely driven by time since symptom onset in acute ischemic stroke: the impact on clinical outcomes. J Thromb Haemost 2024; 22:3161-3171. [PMID: 39122194 DOI: 10.1016/j.jtha.2024.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/06/2024] [Accepted: 06/24/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Gut dysbiosis leading to increased intestinal barrier permeability and translocation of lipopolysaccharide (LPS) in the circulation has been demonstrated in patients with acute myocardial infarction and pulmonary embolism. OBJECTIVES We investigated changes in circulating LPS concentrations in acute ischemic stroke (AIS) and their consequences, including prognosis. METHODS We studied 98 AIS patients, aged 74 ± 12 years, including 74 (75.5%) thrombolysed individuals. We determined serum LPS and zonulin, a marker of gut permeability, along with protein carbonyl (PC), fibrin clot properties, and thrombin generation on admission, at 24 hours and 3 months. Stroke severity was assessed using the National Institutes of Health Stroke Scale. Stroke functional outcome using modified Rankin scale and stroke-related mortality were evaluated at 3 months. RESULTS Serum LPS and zonulin levels on admission were associated with time since symptom onset (r = 0.57; P < .0001; and r = 0.40; P < .0001). Baseline LPS levels correlated with PC (r = 0.51; P < .0001) but not with coagulation and fibrinolysis markers. LPS levels increased at 24 hours in thrombolysed patients (P < .001) and correlated with the National Institutes of Health Stroke Scale score (r = 0.31; P = .002) and PC (r = 0.32; P = .0057). Both LPS and zonulin levels measured at 24 hours increased the odds of having unfavorable modified Rankin scale scores (odds ratio [OR], 1.22; 95% CI, 1.04-1.42; and OR, 2.36; 95% CI, 1.24-4.49 per unit). Elevated LPS level, but not zonulin, was associated with stroke-related mortality (OR, 1.26; 95% CI, 1.02-1.55 per unit). CONCLUSION In AIS patients intestinal permeability is mainly driven by increasing time since the symptom onset. Our findings suggest that LPS, with a trend toward its further rise following thrombolysis, adversely affects neurologic functional outcomes and 3-month mortality.
Collapse
Affiliation(s)
- Michał Błaż
- Department of Neurology, St. John Paul II Hospital, Krakow, Poland
| | - Joanna Natorska
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland; Department of Thromboembolic Diseases, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Jan P Bembenek
- Department of Clinical Neurophysiology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Anna Członkowska
- 2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland
| | - Michał Ząbczyk
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland; Department of Thromboembolic Diseases, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Polak
- Department of Epidemiology and Population Studies, Institute of Public Health, Jagiellonian University Medical College, Krakow, Poland
| | - Anetta Undas
- Krakow Centre for Medical Research and Technologies, St. John Paul II Hospital, Krakow, Poland; Department of Thromboembolic Diseases, Institute of Cardiology, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
3
|
Jiang X, Hu Y, Wang J, Ma M, Bao J, Fang J, He L. Outcomes and risk factors for infection after endovascular treatment in patients with acute ischemic stroke. CNS Neurosci Ther 2024; 30:e14753. [PMID: 38727582 PMCID: PMC11086021 DOI: 10.1111/cns.14753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
AIMS Infection is a common complication following acute ischemic stroke (AIS) and significantly contributes to poor functional outcomes after stroke. This study aimed to investigate the effects of infection after endovascular treatment (post-EVT infection) on clinical outcomes and risk factors in patients with AIS. METHODS We retrospectively analyzed AIS patients treated with endovascular treatment (EVT) between January 2016 and December 2022. A post-EVT infection was defined as any infection diagnosed within 7 days after EVT. The primary outcome was functional independence, defined as a modified Rankin scale (mRS) score of 0-2 at 90 days. A multivariable logistic regression analysis was conducted to determine independent predictors of post-EVT infection and the associations between post-EVT infection and clinical outcomes. RESULTS A total of 675 patients were included in the analysis; 306 (45.3%) of them had post-EVT infections. Patients with post-EVT infection had a lower rate of functional independence than patients without infection (31% vs 65%, p = 0.006). In addition, patients with post-EVT infection achieved less early neurological improvement (ENI) after EVT (25.8% vs 47.4%, p < 0.001). For safety outcomes, the infection group had a higher incidence of any intracranial hemorrhage (23.9% vs 15.7%, p = 0.01) and symptomatic intracranial hemorrhage (10.1% vs 5.1%, p = 0.01). Unsuccessful recanalization (aOR 1.87, 95% CI 1.11-3.13; p = 0.02) and general anesthesia (aOR 2.22, 95% CI 1.25-3.95; p = 0.01) were identified as independent predictors for post-EVT infection in logistic regression analysis. CONCLUSION AIS patients who develop post-EVT infections are more likely to experience poor clinical outcomes. Unsuccessful recanalization and general anesthesia were independent risk factors for the development of post-EVT infection.
Collapse
Affiliation(s)
- Xin Jiang
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Yaowen Hu
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Jian Wang
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Mengmeng Ma
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Jiajia Bao
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Jinghuan Fang
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| | - Li He
- Department of Neurology, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
4
|
Wang T, Pan C, Xie C, Chen L, Song Z, Liao H, Xin C. Microbiota Metabolites and Immune Regulation Affect Ischemic Stroke Occurrence, Development, and Prognosis. Mol Neurobiol 2023; 60:6176-6187. [PMID: 37432592 DOI: 10.1007/s12035-023-03473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
The gut microbiota are not only related to the development and occurrence of digestive system disease, but also have a bidirectional relationship with nervous system diseases via the microbiota-gut-brain axis. At present, correlations between the gut microbiota and neurological diseases, including stroke, are one of the focuses of investigation and attention in the medical community. Ischemic stroke (IS) is a cerebrovascular disease accompanied by focal neurological deficit or central nervous system injury or death. In this review, we summarize the contemporary latest research on correlations between the gut microbiota and IS. Additionally, we discuss the mechanisms of gut microbiota implicated in IS and related to metabolite production and immune regulation. Moreover, the factors of gut microbiota that affecting IS occurrence, and research implicating the gut microbiota as potential therapeutic targets for IS, are highlighted. Our review highlights the evidential relationships and connections between the gut microbiota and IS pathogenesis and prognosis.
Collapse
Affiliation(s)
- Tao Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Chuanling Pan
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Cheng Xie
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Liying Chen
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Zhangyong Song
- Southwest Medical University, 646000, Luzhou, People's Republic of China
| | - Huiling Liao
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China.
| | - Caiyan Xin
- Southwest Medical University, 646000, Luzhou, People's Republic of China.
| |
Collapse
|
5
|
Dzirkale Z, Pilipenko V, Pijet B, Klimaviciusa L, Upite J, Protokowicz K, Kaczmarek L, Jansone B. Long-term behavioural alterations in mice following transient cerebral ischemia. Behav Brain Res 2023; 452:114589. [PMID: 37481076 DOI: 10.1016/j.bbr.2023.114589] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Ischemic stroke is one of the leading causes of disability and mortality worldwide. Acute and chronic post-stroke changes have variable effects on the functional outcomes of the disease. Therefore, it is imperative to identify what daily activities are altered after stroke and to what extent, keeping in mind that ischemic stroke patients often have long-term post-stroke complications. Translational studies in stroke have also been challenging due to inconsistent study design of animal experiments. The objective of this study was to clarify whether and to what extent mouse behaviour was altered during a 6 months period after cerebral stroke. Experimental stroke was induced in mice by intraluminal filament insertion into the middle cerebral artery (fMCAo). Neurological deficits, recovery rate, motor performance, and circadian activity were evaluated following ischemia. We observed severe neurological deficits, motor impairments, and delay in the recovery rate of mice during the first 14 days after fMCAo. Aberrant circadian activity and distorted space map were seen in fMCAo mice starting one month after ischemia, similarly to altered new and familiar cage activity and sucrose preference using the IntelliCage, and was still evident 60- and 180- days following stroke in the voluntary running wheel using the PhenoMaster system. A preference towards ipsilateral side turns was observed in fMCAo mice both acutely and chronically after the stroke induction. Overall, our study shows the importance of determining time-dependent differences in the long-term post-stroke recovery (over 180 days after fMCAo) using multiple behavioural assessments.
Collapse
Affiliation(s)
- Zane Dzirkale
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia.
| | - Vladimirs Pilipenko
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Barbara Pijet
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Linda Klimaviciusa
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Jolanta Upite
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia
| | - Karolina Protokowicz
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, BRAINCITY - Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Baiba Jansone
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 3 Jelgavas Street, LV-1004 Riga, Latvia.
| |
Collapse
|
6
|
Boboc IKS, Rotaru-Zavaleanu AD, Calina D, Albu CV, Catalin B, Turcu-Stiolica A. A Preclinical Systematic Review and Meta-Analysis of Behavior Testing in Mice Models of Ischemic Stroke. Life (Basel) 2023; 13:life13020567. [PMID: 36836924 PMCID: PMC9964520 DOI: 10.3390/life13020567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Stroke remains one of the most important causes of death and disability. Preclinical research is a powerful tool for understanding the molecular and cellular response to stroke. However, a lack of standardization in animal evaluation does not always ensure reproducible results. In the present study, we wanted to identify the best strategy for evaluating animal behavior post-experimental stroke. As such, a meta-analysis was made, evaluating behavioral tests done on male C57BL/6 mice subjected to stroke or sham surgery. Overall, fifty-six studies were included. Our results suggest that different types of tests should be used depending on the post-stroke period one needs to analyze. In the hyper-acute, post-stroke period, the best quantifier will be animal examination scoring, as it is a fast and inexpensive way to identify differences between groups. When evaluating stoke mice in the acute phase, a mix of animal examination and motor tests that focus on movement asymmetry (foot-fault and cylinder testing) seem to have the best chance of picking up differences between groups. Complex tasks (the rotarod test and Morris water maze) should be used within the chronic phase to evaluate differences between the late-subacute and chronic phases.
Collapse
Affiliation(s)
- Ianis Kevyn Stefan Boboc
- Department of Pharmacology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- U.M.F. Doctoral School Craiova, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandra Daniela Rotaru-Zavaleanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Carmen Valeria Albu
- Department of Neurology, Clinical Hospital of Neuropsychiatry, 200473 Craiova, Romania
| | - Bogdan Catalin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence:
| | - Adina Turcu-Stiolica
- Department of Pharmaceutical Management and Marketing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
7
|
Engler-Chiurazzi EB, Russell AE, Povroznik JM, McDonald KO, Porter KN, Wang DS, Hammock J, Billig BK, Felton CC, Yilmaz A, Schreurs BG, O'Callaghan JD, Zwezdaryk KJ, Simpkins JW. Intermittent systemic exposure to lipopolysaccharide-induced inflammation disrupts hippocampal long-term potentiation and impairs cognition in aging male mice. Brain Behav Immun 2023; 108:279-291. [PMID: 36549577 PMCID: PMC10019559 DOI: 10.1016/j.bbi.2022.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Age-related cognitive decline, a common component of the brain aging process, is associated with significant impairment in daily functioning and quality of life among geriatric adults. While the complexity of mechanisms underlying cognitive aging are still being elucidated, microbial exposure and the multifactorial inflammatory cascades associated with systemic infections are emerging as potential drivers of neurological senescence. The negative cognitive and neurobiological consequences of a single pathogen-associated inflammatory experience, such as that modeled through treatment with lipopolysaccharide (LPS), are well documented. Yet, the brain aging impacts of repeated, intermittent inflammatory challenges are less well studied. To extend the emerging literature assessing the impact of infection burden on cognitive function among normally aging mice, here, we repeatedly exposed adult mice to intermittent LPS challenges during the aging period. Male 10-month-old C57BL6 mice were systemically administered escalating doses of LPS once every two weeks for 2.5 months. We evaluated cognitive consequences using the non-spatial step-through inhibitory avoidance task, and both spatial working and reference memory versions of the Morris water maze. We also probed several potential mechanisms, including cortical and hippocampal cytokine/chemokine gene expression, as well as hippocampal neuronal function via extracellular field potential recordings. Though there was limited evidence for an ongoing inflammatory state in cortex and hippocampus, we observed impaired learning and memory and a disruption of hippocampal long-term potentiation. These data suggest that a history of intermittent exposure to LPS-induced inflammation is associated with subtle but significantly impaired cognition among normally aging mice. The broader impact of these findings may have important implications for standard of care involving infections in aging individuals or populations at-risk for dementia.
Collapse
Affiliation(s)
- E B Engler-Chiurazzi
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane Brain Institute, Tulane University, New Orleans, LA 70114, USA; Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA.
| | - A E Russell
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; Department of Biology, School of Science, Penn State Erie, The Behrend College, Erie, PA 16563, USA; Magee Women's Research Institute, Allied Member, Pittsburgh, PA 15213, USA
| | - J M Povroznik
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - K O McDonald
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane Brain Institute, Tulane University, New Orleans, LA 70114, USA
| | - K N Porter
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - D S Wang
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - J Hammock
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - B K Billig
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - C C Felton
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - A Yilmaz
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - B G Schreurs
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| | - J D O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - K J Zwezdaryk
- Department of Microbiology and Immunology, Tulane Brain Institute, Tulane University, New Orleans, LA 70114, USA
| | - J W Simpkins
- Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA; Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26505, USA
| |
Collapse
|
8
|
Kalyan M, Tousif AH, Sonali S, Vichitra C, Sunanda T, Praveenraj SS, Ray B, Gorantla VR, Rungratanawanich W, Mahalakshmi AM, Qoronfleh MW, Monaghan TM, Song BJ, Essa MM, Chidambaram SB. Role of Endogenous Lipopolysaccharides in Neurological Disorders. Cells 2022; 11:cells11244038. [PMID: 36552802 PMCID: PMC9777235 DOI: 10.3390/cells11244038] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Lipopolysaccharide (LPS) is a cell-wall immunostimulatory endotoxin component of Gram-negative bacteria. A growing body of evidence reveals that alterations in the bacterial composition of the intestinal microbiota (gut dysbiosis) disrupt host immune homeostasis and the intestinal barrier function. Microbial dysbiosis leads to a proinflammatory milieu and systemic endotoxemia, which contribute to the development of neurodegenerative diseases and metabolic disorders. Two important pathophysiological hallmarks of neurodegenerative diseases (NDDs) are oxidative/nitrative stress and inflammation, which can be initiated by elevated intestinal permeability, with increased abundance of pathobionts. These changes lead to excessive release of LPS and other bacterial products into blood, which in turn induce chronic systemic inflammation, which damages the blood-brain barrier (BBB). An impaired BBB allows the translocation of potentially harmful bacterial products, including LPS, and activated neutrophils/leucocytes into the brain, which results in neuroinflammation and apoptosis. Chronic neuroinflammation causes neuronal damage and synaptic loss, leading to memory impairment. LPS-induced inflammation causes inappropriate activation of microglia, astrocytes, and dendritic cells. Consequently, these alterations negatively affect mitochondrial function and lead to increases in oxidative/nitrative stress and neuronal senescence. These cellular changes in the brain give rise to specific clinical symptoms, such as impairment of locomotor function, muscle weakness, paralysis, learning deficits, and dementia. This review summarizes the contributing role of LPS in the development of neuroinflammation and neuronal cell death in various neurodegenerative diseases.
Collapse
Affiliation(s)
- Manjunath Kalyan
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Ahmed Hediyal Tousif
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Sharma Sonali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Tuladhar Sunanda
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Sankar Simla Praveenraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Bipul Ray
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
| | - Vasavi Rakesh Gorantla
- Department of Anatomical sciences, School of Medicine, St. George’s University Grenada, West Indies FZ818, Grenada
| | - Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - M. Walid Qoronfleh
- Q3CG Research Institute (QRI), Research & Policy Division, 7227 Rachel Drive, Ypsilanti, MI 48917, USA
- 21 Health Street, Consulting Services, 1 Christian Fields, London SW16 3JY, UK
| | - Tanya M. Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, MD 20892, USA
- Correspondence: (B.-J.S.); (M.M.E.); (S.B.C.)
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman
- Aging and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman
- Correspondence: (B.-J.S.); (M.M.E.); (S.B.C.)
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Correspondence: (B.-J.S.); (M.M.E.); (S.B.C.)
| |
Collapse
|
9
|
Long J, Wang J, Li Y, Chen S. Gut microbiota in ischemic stroke: Where we stand and challenges ahead. Front Nutr 2022; 9:1008514. [DOI: 10.3389/fnut.2022.1008514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Gut microbiota is increasingly recognized to affect host health and disease, including ischemic stroke (IS). Here, we systematically review the current understanding linking gut microbiota as well as the associated metabolites to the pathogenesis of IS (e.g., oxidative stress, apoptosis, and neuroinflammation). Of relevance, we highlight that the implications of gut microbiota-dependent intervention could be harnessed in orchestrating IS.
Collapse
|
10
|
Kim S, Park ES, Chen PR, Kim E. Dysregulated Hypothalamic–Pituitary–Adrenal Axis Is Associated With Increased Inflammation and Worse Outcomes After Ischemic Stroke in Diabetic Mice. Front Immunol 2022; 13:864858. [PMID: 35784349 PMCID: PMC9243263 DOI: 10.3389/fimmu.2022.864858] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/09/2022] [Indexed: 01/08/2023] Open
Abstract
Diabetic patients have larger infarcts, worse neurological deficits, and higher mortality rate after an ischemic stroke. Evidence shows that in diabetes, the hypothalamic–pituitary–adrenal (HPA) axis was dysregulated and levels of cortisol increased. Based on the role of the HPA axis in immunity, we hypothesized that diabetes-dysregulated stress response exacerbates stroke outcomes via regulation of inflammation. To test this hypothesis, we assessed the regulation of the HPA axis in diabetic mice before and after stroke and determined its relevance in the regulation of post-stroke injury and inflammation. Diabetes was induced in C57BL/6 mice by feeding a high-fat diet and intraperitoneal injection of streptozotocin (STZ), and then the mice were subjected to 30 min of middle cerebral artery occlusion (MCAO). Infarct volume and neurological scores were measured in the ischemic mice. The inflammatory cytokine and chemokine levels were also determined in the ischemic brain. To assess the effect of diabetes on the stroke-modulated HPA axis, we measured the expression of components in the HPA axis including corticotropin-releasing hormone (CRH) in the hypothalamus, proopiomelanocortin (POMC) in the pituitary, and plasma adrenocorticotropic hormone (ACTH) and corticosterone. Diabetic mice had larger infarcts and worse neurological scores after stroke. The exacerbated stroke outcomes in diabetic mice were accompanied by the upregulated expression of inflammatory factors (including IL-1β, TNF-α, IL-6, CCR2, and MCP-1) in the ischemic brain. We also confirmed increased levels of hypothalamic CRH, pituitary POMC, and plasma corticosterone in diabetic mice before and after stroke, suggesting the hyper-activated HPA axis in diabetic conditions. Finally, we confirmed that post-stroke treatment of metyrapone (an inhibitor of glucocorticoid synthesis) reduced IL-6 expression and the infarct size in the ischemic brain of diabetic mice. These results elucidate the mechanisms in which the HPA axis in diabetes exacerbates ischemic stroke. Maintaining an optimal level of the stress response by regulating the HPA axis may be an effective approach to improving stroke outcomes in patients with diabetes.
Collapse
|
11
|
Shi J, Li W, Zhang F, Park JH, An H, Guo S, Duan Y, Wu D, Hayakawa K, Lo EH, Ji X. CCL2 (C-C Motif Chemokine Ligand 2) Biomarker Responses in Central Versus Peripheral Compartments After Focal Cerebral Ischemia. Stroke 2021; 52:3670-3679. [PMID: 34587791 PMCID: PMC8545911 DOI: 10.1161/strokeaha.120.032782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background and Purpose Inflammatory mediators in blood have been proposed as potential biomarkers in stroke. However, a direct relationship between these circulating factors and brain-specific ischemic injury remains to be fully defined. Methods An unbiased screen in a nonhuman primate model of stroke was used to find out the most responsive circulating biomarker flowing ischemic stroke. Then this phenomenon was checked in human beings and mice. Finally, we observed the temporospatial responsive characteristics of this biomarker after ischemic brain injury in mice to evaluate the direct relationship between this circulating factor and central nervous system–specific ischemic injury. Results In a nonhuman primate model, an unbiased screen revealed CCL2 (C-C motif chemokine ligand 2) as a major response factor in plasma after stroke. In mouse models of focal cerebral ischemia, plasma levels of CCL2 showed a transient response, that is, rapidly elevated by 2 to 3 hours postischemia but then renormalized back to baseline levels by 24 hours. However, a different CCL2 temporal profile was observed in whole brain homogenate, cerebrospinal fluid, and isolated brain microvessels, with a progressive increase over 24 hours, demonstrating a mismatch between brain versus plasma responses. In contrast to the lack of correlation with central nervous system responses, 2 peripheral compartments showed transient profiles that matched circulating plasma signatures. CCL2 protein in lymph nodes and adipose tissue was significantly increased at 2 hours and renormalized by 24 hours. Conclusions These findings may provide a cautionary tale for biomarker pursuits in plasma. Besides a direct central nervous system response, peripheral organs may also contribute to blood signatures in complex and indirect ways.
Collapse
Affiliation(s)
- Jingfei Shi
- Cerebrovascular and Neuroscience Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wenlu Li
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Zhang
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ji Hyun Park
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hong An
- Cerebrovascular and Neuroscience Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shuzhen Guo
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yunxia Duan
- Cerebrovascular and Neuroscience Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Di Wu
- Cerebrovascular and Neuroscience Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H. Lo
- Neuroprotection Research Laboratories, Departments of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xunming Ji
- Cerebrovascular and Neuroscience Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China.,Departments of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
12
|
Quintana D, Ren X, Hu H, Corbin D, Engler-Chiurazzi E, Alvi M, Simpkins J. IL-1β Antibody Protects Brain from Neuropathology of Hypoperfusion. Cells 2021; 10:855. [PMID: 33918659 PMCID: PMC8069995 DOI: 10.3390/cells10040855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 11/16/2022] Open
Abstract
Chronic brain hypoperfusion is the primary cause of vascular dementia and has been implicated in the development of white matter disease and lacunar infarcts. Cerebral hypoperfusion leads to a chronic state of brain inflammation with immune cell activation and production of pro-inflammatory cytokines, including IL-1β. In the present study, we induced chronic, progressive brain hypoperfusion in mice using ameroid constrictor, arterial stenosis (ACAS) surgery and tested the efficacy of an IL-1β antibody on the resulting brain damage. We observed that ACAS surgery causes a reduction in cerebral blood flow (CBF) of about 30% and grey and white matter damage in and around the hippocampus. The IL-1β antibody treatment did not significantly affect CBF but largely eliminated grey matter damage and reduced white matter damage caused by ACAS surgery. Over the course of hypoperfusion/injury, grip strength, coordination, and memory-related behavior were not significantly affected by ACAS surgery or antibody treatment. We conclude that antibody neutralization of IL-1β is protective from the brain damage caused by chronic, progressive brain hypoperfusion.
Collapse
Affiliation(s)
- Dominic Quintana
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
| | - Xuefang Ren
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
| | - Heng Hu
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
| | - Deborah Corbin
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
| | - Elizabeth Engler-Chiurazzi
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
| | - Muhammad Alvi
- Center for Basic and Translational Stroke Research, Department of Neurology, Rockefeller Neuroscience Institute, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| | - James Simpkins
- Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, USA; (D.Q.); (X.R.); (H.H.); (D.C.); (E.E.-C.)
- Center for Basic and Translational Stroke Research, Department of Neurology, Rockefeller Neuroscience Institute, School of Medicine, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
13
|
Balkaya M, Kim ID, Shakil F, Cho S. CD36 deficiency reduces chronic BBB dysfunction and scar formation and improves activity, hedonic and memory deficits in ischemic stroke. J Cereb Blood Flow Metab 2021; 41:486-501. [PMID: 32404022 PMCID: PMC7922745 DOI: 10.1177/0271678x20924099] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/05/2020] [Accepted: 04/05/2020] [Indexed: 01/06/2023]
Abstract
Ameliorating blood-brain barrier disruption and altering scar formation dynamics are potential strategies that may improve post-stroke recovery. CD36 is a class B scavenger receptor that plays a role in innate immunity, inflammation and vascular dysfunction and regulates post-stroke injury, neovascularization, reactive astrogliosis and scar formation. By subjecting WT and CD36KO mice to different MCAo occlusion durations to generate comparable acute lesion sizes, we addressed the role of CD36 in BBB dysfunction, scar formation and recovery. The majority of stroke recovery studies primarily focus on motor function. Here, we employed an extensive behavioral test arsenal to evaluate psychological and cognitive endpoints. While not evident during the acute phase, CD36 deficient mice displayed significantly attenuated BBB leakage and scar formation at three months after stroke compared to wild-type littermates. Assessment of motor (open field, rotarod), anxiety (plus maze, light-dark box), depression (forced swim, sucrose preference) and memory tests (water maze) revealed that CD36 deficiency ameliorated stroke-induced behavioral impairments in activity, hedonic responses and spatial learning and strategy switching. Our findings indicate that CD36 contributes to stroke-induced BBB dysfunction and scar formation in an injury-independent manner, as well as to the chronic motor and neurophysiological deficits in chronic stroke.
Collapse
Affiliation(s)
- Mustafa Balkaya
- Burke Neurological Research Institute, White Plains, NY,
USA
| | - Il-doo Kim
- Burke Neurological Research Institute, White Plains, NY,
USA
| | - Faariah Shakil
- Burke Neurological Research Institute, White Plains, NY,
USA
| | - Sunghee Cho
- Burke Neurological Research Institute, White Plains, NY,
USA
- Feil Family Brain and Mind Research Institute, Weill Cornell
Medicine at Burke Neurological Research Institute, White Plains, NY USA
| |
Collapse
|
14
|
Russell AE, Cavendish JZ, Rai A, Vannoy M, Dakhlallah AH, Hu H, Ren X, Amer A, Brown CM, Marsh CB, Simpkins JW, Dakhlallah D. Intermittent Lipopolysaccharide Exposure Significantly Increases Cortical Infarct Size and Impairs Autophagy. ASN Neuro 2021; 13:1759091421991769. [PMID: 33626880 PMCID: PMC8020222 DOI: 10.1177/1759091421991769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Globally, stroke is a leading cause of death and disability. Traditional risk factors like hypertension, diabetes, and obesity do not fully account for all stroke cases. Recent infection is regarded as changes in systemic immune signaling, which can increase thrombosis formation and other stroke risk factors. We have previously shown that administration of lipopolysaccharide (LPS) 30-minutes prior to stroke increases in infarct volume. In the current study, we found that animals intermittently exposed to LPS have larger cortical infarcts when compared to saline controls. To elucidate the mechanism behind this phenomenon, several avenues were investigated. We observed significant upregulation of tumor necrosis factor-alpha (TNF-α) mRNA, especially in the ipsilateral hemisphere of both saline and LPS exposed groups compared to sham surgery animals. We also observed significant reductions in expression of genes involved in autophagy in the ipsilateral hemisphere of LPS stroke animals. In addition, we assessed DNA methylation of autophagy genes and observed a significant increase in the ipsilateral hemisphere of LPS stroke animals. Intermittent exposure to LPS increases cortical infarct volume, downregulates autophagy genes, and induces hypermethylation of the corresponding CpG islands. These data suggest that intermittent immune activation may deregulate epigenetic mechanisms and promote neuropathological outcomes after stroke.
Collapse
Affiliation(s)
- Ashley E Russell
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Department of Biology, School of Science, Penn State Erie, The Behrend College, Erie, Pennsylvania, United States
| | - John Z Cavendish
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States
| | - Ali Rai
- Department of Biomedical Engineering, West Virginia University School of Medicine, Morgantown, United States
| | - Mya Vannoy
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, United States
| | - Ahmad H Dakhlallah
- Department of Biology, West Virginia University School of Medicine, Morgantown, United States
| | - Heng Hu
- Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, United States
| | - Xuefang Ren
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States
| | - Amal Amer
- Department of Microbial Infection and Immunity, Infectious Diseases Institute, Ohio State University, Columbus, United States
| | - Candice M Brown
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States
| | - Clay B Marsh
- Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, Morgantown, United States
| | - James W Simpkins
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, United States.,Rockerfeller Center for Neuroscience, West Virginia University School of Medicine, Morgantown, United States
| | - Duaa Dakhlallah
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, United States.,School of Sciences and Engineering, American University in Cairo, New Cairo, Egypt
| |
Collapse
|
15
|
Abstract
Background Ischemic stroke is one of the non-communicable diseases that contribute to the significant number of deaths worldwide. However, the relationship between microbiome and ischemic stroke remained unknown. Hence, the objective of this study was to perform systematic review on the relationship between human microbiome and ischemic stroke. Methods A systematic review on ischemic stroke was carried out for all articles obtained from databases until 22nd October 2020. Main findings were extracted from all the eligible studies. Results Eighteen eligible studies were included in the systematic review. These studies suggested that aging, inflammation, and different microbial compositions could contribute to ischemic stroke. Phyla Firmicutes and Bacteroidetes also appeared to manipulate post-stroke outcome. The important role of microbiota-derived short-chain fatty acids and trimethylamine N-oxide in ischemic stroke were also highlighted. Conclusions This is the first systematic review that investigates the relationship between microbiome and ischemic stroke. Aging and inflammation contribute to differential microbial compositions and predispose individuals to ischemic stroke.
Collapse
|
16
|
Zhang P, Zhang X, Huang Y, Chen J, Shang W, Shi G, Zhang L, Zhang C, Chen R. Atorvastatin alleviates microglia-mediated neuroinflammation via modulating the microbial composition and the intestinal barrier function in ischemic stroke mice. Free Radic Biol Med 2021; 162:104-117. [PMID: 33279615 DOI: 10.1016/j.freeradbiomed.2020.11.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/03/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022]
Abstract
Our previous work has shown that atorvastatin exerts anti-inflammatory properties in ischemic stroke, and recent studies have revealed that intestinal microbiota plays a vital role in the pathogenesis of stroke. However, it is not clear whether the anti-inflammatory effects of atorvastatin against ischemic stroke is related to gut function and microbiota. We report herein that atorvastatin significantly ameliorated the defects in sensorimotor behaviors and reduced microglia-mediated neuroinflammation by inhibiting proinflammatory polarization of microglia in the peri-infarct cortex of the mice with permanent middle cerebral artery occlusion (pMCAO). Moreover, atorvastatin reversed microbial composition (characterized by increased abundance of Firmicutes and Lactobacillus and decreased Bacteroidetes abundance), increased fecal butyrate level, promoted intestinal barrier function (elevated protein levels of claudin-1, occludin and mucoprotein 2), as well as regulated intestinal immune function (decreased MCP-1, TNF-α and increased IL-10). Atorvastatin also significantly reduced the level of circulating endotoxin (lipopolysaccharide-binding protein), which is a biomarker of leaky gut. Transplantation of fecal microbiota collected from atorvastatin treated mice potently attenuated neuroinflammation in pMCAO mice, and the anti-inflammatory effects of fecal microbiota transplantation were similar to those of oral atorvastatin administration. These results suggested that the atorvastatin-mediated restoration of gut microbiota, improvement of intestinal barrier function and regulation of intestinal immunity were involved in the anti-inflammatory function in stroke mice.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Xiangjian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China.
| | - Yuanxiang Huang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Junmin Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Wenyan Shang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Guang Shi
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Lan Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Cong Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| | - Rong Chen
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, 050000, Hebei, China
| |
Collapse
|
17
|
Loppi S, Korhonen P, Bouvy‐Liivrand M, Caligola S, Turunen TA, Turunen MP, Hernandez de Sande A, Kołosowska N, Scoyni F, Rosell A, García‐Berrocoso T, Lemarchant S, Dhungana H, Montaner J, Koistinaho J, Kanninen KM, Kaikkonen MU, Giugno R, Heinäniemi M, Malm T. Peripheral inflammation preceeding ischemia impairs neuronal survival through mechanisms involving miR-127 in aged animals. Aging Cell 2021; 20:e13287. [PMID: 33369048 PMCID: PMC7811844 DOI: 10.1111/acel.13287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/06/2020] [Accepted: 11/27/2020] [Indexed: 01/02/2023] Open
Abstract
Ischemic stroke, the third leading cause of death in the Western world, affects mainly the elderly and is strongly associated with comorbid conditions such as atherosclerosis or diabetes, which are pathologically characterized by increased inflammation and are known to influence the outcome of stroke. Stroke incidence peaks during influenza seasons, and patients suffering from infections such as pneumonia prior to stroke exhibit a worse stroke outcome. Earlier studies have shown that comorbidities aggravate the outcome of stroke, yet the mediators of this phenomenon remain obscure. Here, we show that acute peripheral inflammation aggravates stroke‐induced neuronal damage and motor deficits specifically in aged mice. This is associated with increased levels of plasma proinflammatory cytokines, rather than with an increase of inflammatory mediators in the affected brain parenchyma. Nascent transcriptomics data with mature microRNA sequencing were used to identify the neuron‐specific miRNome, in order to decipher dysregulated miRNAs in the brains of aged animals with stroke and co‐existing inflammation. We pinpoint a previously uninvestigated miRNA in the brain, miR‐127, that is highly neuronal, to be associated with increased cell death in the aged, LPS‐injected ischemic mice. Target prediction tools indicate that miR‐127 interacts with several basally expressed neuronal genes, and of these we verify miR‐127 binding to Psmd3. Finally, we report reduced expression of miR‐127 in human stroke brains. Our results underline the impact of peripheral inflammation on the outcome of stroke in aged subjects and pinpoint molecular targets for restoring endogenous neuronal capacity to combat ischemic stroke.
Collapse
Affiliation(s)
- Sanna Loppi
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Department of Immunobiology University of Arizona Tucson Arizona USA
| | - Paula Korhonen
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | | | - Simone Caligola
- Department of Computer Science University of Verona Verona Italy
| | - Tiia A. Turunen
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Mikko P. Turunen
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | | | - Natalia Kołosowska
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Flavia Scoyni
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Anna Rosell
- Neurovascular Research Laboratory Vall d’Hebron Institute of Research (VHIR) Universitat Autònoma de Barcelona Barcelona Spain
| | - Teresa García‐Berrocoso
- Neurovascular Research Laboratory Vall d’Hebron Institute of Research (VHIR) Universitat Autònoma de Barcelona Barcelona Spain
| | - Sighild Lemarchant
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Hiramani Dhungana
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Neuroscience Center University of Helsinki Helsinki Finland
| | - Joan Montaner
- Neurovascular Research Laboratory Vall d’Hebron Institute of Research (VHIR) Universitat Autònoma de Barcelona Barcelona Spain
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
- Neuroscience Center University of Helsinki Helsinki Finland
| | - Katja M. Kanninen
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Minna U. Kaikkonen
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| | - Rosalba Giugno
- Department of Computer Science University of Verona Verona Italy
| | | | - Tarja Malm
- A.I. Virtanen Institute for Molecular Sciences University of Eastern Finland Kuopio Finland
| |
Collapse
|
18
|
Atif F, Yousuf S, Espinosa-Garcia C, Harris WAC, Stein DG. Post-ischemic stroke systemic inflammation: Immunomodulation by progesterone and vitamin D hormone. Neuropharmacology 2020; 181:108327. [PMID: 32950558 DOI: 10.1016/j.neuropharm.2020.108327] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022]
Abstract
Post-stroke systemic inflammation, due to the injury itself and exacerbated by in-hospital infections, can increase morbidity and mortality in stroke patients. In this study, we examined the immunomodulatory effects of progesterone (P4) alone and in combination with vitamin D hormone (VDH) on acute phase post-stroke peripheral immune dysfunction and functional/behavioral deficits. Adult rats underwent transient middle cerebral artery occlusion/reperfusion (tMCAO) and delayed systemic inflammation was induced by injections of lipopolysaccharide (LPS) beginning 24 h post-stroke. Animals were tested for behavioral outcomes and immune function at day 4 post-stroke. We also measured infarction volume and markers of neuronal inflammation (GFAP, IL-6) and apoptosis (cleaved caspase-3) in brain post-stroke. We observed the worst stroke outcomes in the stroke + systemic inflammation group compared to the stroke-alone group. Flow cytometric analysis of different subsets of immune cells in blood, spleen and thymus revealed peripheral immune dysfunction which was restored by both P4 and VDH monotherapy. P4 monotherapy reduced infarction volume, behavioral/functional deficits, peripheral immune dysfunction, neuronal inflammation, and apoptosis induced by post-stroke systemic inflammation. Combination treatment with P4+VDH improved outcomes better than monotherapy. Our findings can be taken to suggest that the current standard of care for stroke and post-stroke infection can be substantially improved by P4 and VDH combination therapy.
Collapse
Affiliation(s)
- Fahim Atif
- Brain Research Laboratory, Department of Emergency Medicine, USA.
| | - Seema Yousuf
- Brain Research Laboratory, Department of Emergency Medicine, USA
| | | | - Wayne A C Harris
- Emory Integrated Computing Core, School of Medicine, Emory University, Atlanta, GA, 30322,, USA
| | - Donald G Stein
- Brain Research Laboratory, Department of Emergency Medicine, USA
| |
Collapse
|
19
|
Engler-Chiurazzi EB, Monaghan KL, Wan ECK, Ren X. Role of B cells and the aging brain in stroke recovery and treatment. GeroScience 2020; 42:1199-1216. [PMID: 32767220 PMCID: PMC7525651 DOI: 10.1007/s11357-020-00242-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023] Open
Abstract
As mitigation of brain aging continues to be a key public health priority, a wholistic and comprehensive consideration of the aging body has identified immunosenescence as a potential contributor to age-related brain injury and disease. Importantly, the nervous and immune systems engage in bidirectional communication and can exert profound influence on each other. Emerging evidence supports numerous impacts of innate, inflammatory immune responses and adaptive T cell-mediated immunity in neurological function and diseased or injured brain states, such as stroke. Indeed, a growing body of evidence supports key impacts of brain-resident immune cell activation and peripheral immune infiltration in both the post-stroke acute injury phase and the long-term recovery period. As such, modulation of the immune system is an attractive strategy for novel therapeutic interventions for a devastating age-related brain injury for which there are few readily available neuroprotective treatments or neurorestorative approaches. However, the role of B cells in the context of brain function, and specifically in response to stroke, has not been thoroughly elucidated and remains controversial, leaving our understanding of neuroimmune interactions incomplete. Importantly, emerging evidence suggests that B cells are not pathogenic contributors to stroke injury, and in fact may facilitate functional recovery, supporting their potential value as novel therapeutic targets. By summarizing the current knowledge of the role of B cells in stroke pathology and recovery and interpreting their role in the context of their interactions with other immune cells as well as the immunosenescence cascades that alter their function in aged populations, this review supports an increased understanding of the complex interplay between the nervous and immune systems in the context of brain aging, injury, and disease.
Collapse
Affiliation(s)
- E. B. Engler-Chiurazzi
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
| | - K. L. Monaghan
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| | - E. C. K. Wan
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| | - X. Ren
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Center for Basic & Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV 26506 USA
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV 26506 USA
| |
Collapse
|
20
|
Castellanos DM, Sun J, Yang J, Ou W, Zambon AC, Pardridge WM, Sumbria RK. Acute and Chronic Dosing of a High-Affinity Rat/Mouse Chimeric Transferrin Receptor Antibody in Mice. Pharmaceutics 2020; 12:pharmaceutics12090852. [PMID: 32911688 PMCID: PMC7558337 DOI: 10.3390/pharmaceutics12090852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/17/2022] Open
Abstract
Non-invasive brain delivery of neurotherapeutics is challenging due to the blood-brain barrier. The revived interest in transferrin receptor antibodies (TfRMAbs) as brain drug-delivery vectors has revealed the effect of dosing regimen, valency, and affinity on brain uptake, TfR expression, and Fc-effector function side effects. These studies have primarily used monovalent TfRMAbs with a human constant region following acute intravenous dosing in mice. The effects of a high-affinity bivalent TfRMAb with a murine constant region, without a fusion partner, following extravascular dosing in mice are, however, not well characterized. Here we elucidate the plasma pharmacokinetics and safety of a high-affinity bivalent TfRMAb with a murine constant region following acute and chronic subcutaneous dosing in adult C57BL/6J male mice. Mice received a single (acute dosing) 3 mg/kg dose, or were treated for four weeks (chronic dosing). TfRMAb and control IgG1 significantly altered reticulocyte counts following acute and chronic dosing, while other hematologic parameters showed minimal change. Chronic TfRMAb dosing did not alter plasma- and brain-iron measurements, nor brain TfR levels, however, it significantly increased splenic-TfR and -iron. Plasma concentrations of TfRMAb were significantly lower in mice chronically treated with IgG1 or TfRMAb. Overall, no injection related reactions were observed in mice.
Collapse
Affiliation(s)
- Demi M. Castellanos
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (D.M.C.); (J.Y.)
| | - Jiahong Sun
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (J.S.); (W.O.); (A.C.Z.)
| | - Joshua Yang
- Henry E. Riggs School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (D.M.C.); (J.Y.)
| | - Weijun Ou
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (J.S.); (W.O.); (A.C.Z.)
| | - Alexander C. Zambon
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (J.S.); (W.O.); (A.C.Z.)
| | | | - Rachita K. Sumbria
- Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA 91711, USA; (J.S.); (W.O.); (A.C.Z.)
- Department of Neurology, University of California, Irvine, CA 92868, USA
- Correspondence: ; Tel.: +1-(909)-607-0319; Fax: +1-(909)-607-9826
| |
Collapse
|
21
|
Ramsey AM, Stowie A, Castanon-Cervantes O, Davidson AJ. Environmental Circadian Disruption Increases Stroke Severity and Dysregulates Immune Response. J Biol Rhythms 2020; 35:368-376. [PMID: 32508262 DOI: 10.1177/0748730420929450.environmental] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Understanding the health consequences of chronic disruption of circadian rhythms can contribute to improving prevention strategies for shift workers. Chronic circadian disruption in shift work has been linked to a higher risk of stroke. Dysregulated immune responses are also linked to circadian disruption and may be a factor in stroke outcomes in shift workers. In this study, we test the hypotheses that specific schedules of circadian disruption exacerbate inflammatory responses in the brain, causing an increase in infarct size after experimentally induced ischemic stroke. Mice were exposed to 1 of 5 different lighting schedules followed by a 30-min middle cerebral artery occlusion, then reperfusion and 3-day recovery. A history of weekly phase advances resulted in an increased infarct volume versus the control lighting schedule. These effects were shift-direction specific, nonpermanent, and required multiple shifts to occur. In a separate cohort, stereotaxic injections of lipopolysaccharide were given bilaterally after exposure to 1 of 3 different lighting schedules. Ratios of pro- to anti-inflammatory cytokine expression show dysregulated responses after a history of phase advances. We conclude that chronic circadian disruption leads to worsened stroke outcome in a direction- and schedule-specific manner likely because of priming of the inflammatory response in the brain. These pieces of evidence suggest that the health impacts of shift work may be improved by targeting shift work scheduling, inflammatory mediators, or both.
Collapse
Affiliation(s)
- Anne M Ramsey
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Adam Stowie
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | | | - Alec J Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
22
|
Ramsey AM, Stowie A, Castanon-Cervantes O, Davidson AJ. Environmental Circadian Disruption Increases Stroke Severity and Dysregulates Immune Response. J Biol Rhythms 2020; 35:368-376. [PMID: 32508262 DOI: 10.1177/0748730420929450] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Understanding the health consequences of chronic disruption of circadian rhythms can contribute to improving prevention strategies for shift workers. Chronic circadian disruption in shift work has been linked to a higher risk of stroke. Dysregulated immune responses are also linked to circadian disruption and may be a factor in stroke outcomes in shift workers. In this study, we test the hypotheses that specific schedules of circadian disruption exacerbate inflammatory responses in the brain, causing an increase in infarct size after experimentally induced ischemic stroke. Mice were exposed to 1 of 5 different lighting schedules followed by a 30-min middle cerebral artery occlusion, then reperfusion and 3-day recovery. A history of weekly phase advances resulted in an increased infarct volume versus the control lighting schedule. These effects were shift-direction specific, nonpermanent, and required multiple shifts to occur. In a separate cohort, stereotaxic injections of lipopolysaccharide were given bilaterally after exposure to 1 of 3 different lighting schedules. Ratios of pro- to anti-inflammatory cytokine expression show dysregulated responses after a history of phase advances. We conclude that chronic circadian disruption leads to worsened stroke outcome in a direction- and schedule-specific manner likely because of priming of the inflammatory response in the brain. These pieces of evidence suggest that the health impacts of shift work may be improved by targeting shift work scheduling, inflammatory mediators, or both.
Collapse
Affiliation(s)
- Anne M Ramsey
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | - Adam Stowie
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| | | | - Alec J Davidson
- Neuroscience Institute, Morehouse School of Medicine, Atlanta, Georgia
| |
Collapse
|
23
|
Nwafor DC, Chakraborty S, Brichacek AL, Jun S, Gambill CA, Wang W, Engler-Chiurazzi EB, Dakhlallah D, Pinkerton AB, Millán JL, Benkovic SA, Brown CM. Loss of tissue-nonspecific alkaline phosphatase (TNAP) enzyme activity in cerebral microvessels is coupled to persistent neuroinflammation and behavioral deficits in late sepsis. Brain Behav Immun 2020; 84:115-131. [PMID: 31778743 PMCID: PMC7010562 DOI: 10.1016/j.bbi.2019.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/12/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a host response to systemic inflammation and infection that may lead to multi-organ dysfunction and eventual death. While acute brain dysfunction is common among all sepsis patients, chronic neurological impairment is prevalent among sepsis survivors. The brain microvasculature has emerged as a major determinant of sepsis-associated brain dysfunction, yet the mechanisms that underlie its associated neuroimmune perturbations and behavioral deficits are not well understood. An emerging body of data suggests that inhibition of tissue-nonspecific alkaline phosphatase (TNAP) enzyme activity in cerebral microvessels may be associated with changes in endothelial cell barrier integrity. The objective of this study was to elucidate the connection between alterations in cerebrovascular TNAP enzyme activity and brain microvascular dysfunction in late sepsis. We hypothesized that the disruption of TNAP enzymatic activity in cerebral microvessels would be coupled to the sustained loss of brain microvascular integrity, elevated neuroinflammatory responses, and behavioral deficits. Male mice were subjected to cecal ligation and puncture (CLP), a model of experimental sepsis, and assessed up to seven days post-sepsis. All mice were observed daily for sickness behavior and underwent behavioral testing. Our results showed a significant decrease in brain microvascular TNAP enzyme activity in the somatosensory cortex and spinal cord of septic mice but not in the CA1 and CA3 hippocampal regions. Furthermore, we showed that loss of cerebrovascular TNAP enzyme activity was coupled to a loss of claudin-5 and increased perivascular IgG infiltration in the somatosensory cortex. Analyses of whole brain myeloid and T-lymphoid cell populations also revealed a persistent elevation of infiltrating leukocytes, which included both neutrophil and monocyte myeloid derived suppressor cells (MDSCs). Regional analyses of the somatosensory cortex, hippocampus, and spinal cord revealed significant astrogliosis and microgliosis in the cortex and spinal cord of septic mice that was accompanied by significant microgliosis in the CA1 and CA3 hippocampal regions. Assessment of behavioral deficits revealed no changes in learning and memory or evoked locomotion. However, the hot plate test uncovered a novel anti-nociceptive phenotype in our septic mice, and we speculate that this phenotype may be a consequence of sustained GFAP astrogliosis and loss of TNAP activity in the somatosensory cortex and spinal cord of septic mice. Taken together, these results demonstrate that the loss of TNAP enzyme activity in cerebral microvessels during late sepsis is coupled to sustained neuroimmune dysfunction which may underlie, in part, the chronic neurological impairments observed in sepsis survivors.
Collapse
Affiliation(s)
- Divine C. Nwafor
- Department of Neuroscience, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| | - Sreeparna Chakraborty
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| | - Allison L. Brichacek
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| | - Sujung Jun
- Wilmer Eye Institute, John Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Catheryne A. Gambill
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| | - Wei Wang
- Department of Neuroscience, West Virginia University Health Science Center, Morgantown, WV 26506, USA.
| | | | - Duaa Dakhlallah
- Department of Neuroscience, West Virginia University Health Science Center, Morgantown, WV 26506, USA; Cancer Institute, West Virginia University Health Science Center, Morgantown, WV 26506, USA.
| | | | - José Luis Millán
- Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| | - Stanley A. Benkovic
- Department of Neuroscience, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| | - Candice M. Brown
- Department of Neuroscience, West Virginia University Health Science Center, Morgantown, WV 26506, USA,Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA,Corresponding Author: Candice M. Brown, Ph.D., Assistant Professor, Neuroscience, 108 Biomedical Road, Box 9303, Center for Basic and Translational Stroke Research, Rockefeller Neuroscience Institute, West Virginia University Health Sciences, Morgantown, WV 26506, Phone: 304-293-0589,
| |
Collapse
|
24
|
Over-expression of miR-34a induces rapid cognitive impairment and Alzheimer's disease-like pathology. Brain Res 2019; 1721:146327. [PMID: 31295467 DOI: 10.1016/j.brainres.2019.146327] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/28/2019] [Accepted: 07/07/2019] [Indexed: 01/24/2023]
Abstract
Autosomal dominant Alzheimer disease (AD) is caused by rare mutations in one of three specific genes. This is in contrast to idiopathic, late-onset AD (LOAD), which has a more polygenetic risk profile and represents more than 95% of cases. Previously, we have demonstrated that increased expression of microRNA (miRNA)-34a (miR-34a) in AD brain targets genes linked to synaptic plasticity, energy metabolism, and resting state network activity. Here we report the generation of a heterozygous, conditional miR-34a overexpression mouse (miR-34a+/-(TetR-TetO-miR-34a) Transgenic Mice). Doxycycline-treated mice of either sex exhibited profound behavioral impairment compared to untreated groups with only 1-2 months of over-expression of miR-34a. Cognitive impairment of individual mice in T- and Y-maze tasks correlated with elevated miR-34a expression in many parts of the brain including the hippocampus and prefrontal cortex, regions which are known to be involved in this task and implicated in LOAD dysfunction. Immunocytochemistry of brain sections from mice show high amyloid β and phosphorylated tau-specific staining in the hippocampus and cortex. Analysis of protein samples from these mice revealed that miR-34a targets specific genes involved in memory formation, amyloid precursor protein (APP) metabolism and phosphorylation-dephosphorylation of tau. Thus, our results suggest that the polygenetic dysfunction caused by miR-34a may occur in LOAD and disclose miR-34a as a potential therapeutic target. SIGNIFICANCE STATEMENT: Late-onset Alzheimer disease (LOAD) is associated with multiple gene alleles, a polygenetic profile of risk factors that is difficult to model in animals. Our approach to modeling LOAD was to produce a conditional over-expressing, miR-34a mouse using doxycycline-induction to activate expression. We observed that miR-34a over-expression results in a rapid cognitive impairment, associated with accumulation of intracellular Aβ and tau hyperphosphorylation in multiple brain regions. Targets for miR-34a, including ADAM10, NMDAR 2B, and SIRT1 RNAs, were profoundly reduced by miR-34a over-expression. Collectively, these results indicate that a rapid, profound cognitive decline and Alzheimer's disease neuropathology can be induced with miR-34a over-expression, suggesting that this animal model may represent a polygenetic risk factor model for LOAD.
Collapse
|
25
|
Nadareishvili Z, Kelley D, Luby M, Simpkins AN, Leigh R, Lynch JK, Hsia AW, Benson RT, Johnson KR, Hallenbeck JM, Latour LL. Molecular signature of penumbra in acute ischemic stroke: a pilot transcriptomics study. Ann Clin Transl Neurol 2019; 6:817-820. [PMID: 31020007 PMCID: PMC6469246 DOI: 10.1002/acn3.757] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/19/2019] [Accepted: 02/20/2019] [Indexed: 12/29/2022] Open
Abstract
We aimed to characterize peripheral blood gene expression profile of penumbra defined as MRI perfusion–diffusion mismatch (PD MM) in peripheral blood of patients with acute ischemic stroke. We studied 23 patients. Perfusion–diffusion mismatch volume was observed to be associated and significantly correlated with the expression of 34 genes including those related to inflammation, SUMOylation, and coagulation; while lipopolysaccharide inhibition was identified to be a candidate upstream regulator of these processes (z‐score −2.38, P = 0.04). Penumbral volume is correlated with a specific gene expression profile in the peripheral blood characterized by overlap of inflammatory and neuroprotective pathways that are regulated by lipopolysaccharide inhibition.
Collapse
Affiliation(s)
- Zurab Nadareishvili
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland.,Department of Neurology George Washington University Washington District of Columbia
| | - Devon Kelley
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - Marie Luby
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - Alexis N Simpkins
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - Richard Leigh
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - John K Lynch
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - Amie W Hsia
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland.,Medstar Washington Hospital Center Stroke Center Washington District of Columbia
| | - Richard T Benson
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland.,Medstar Washington Hospital Center Stroke Center Washington District of Columbia
| | - Kory R Johnson
- Bioinformatics Section Information Technology Program Division of Intramural Research NINDS NIH Bethesda Maryland
| | - John M Hallenbeck
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| | - Lawrence L Latour
- Section on Stroke Diagnostics and Therapeutics NINDS NIH Bethesda Maryland
| |
Collapse
|
26
|
Chen F, Weng Z, Xia Q, Cao C, Leak RK, Han L, Xiao J, Graham SH, Cao G. Intracerebroventricular Delivery of Recombinant NAMPT Deters Inflammation and Protects Against Cerebral Ischemia. Transl Stroke Res 2019; 10:719-728. [PMID: 30820847 DOI: 10.1007/s12975-019-00692-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/07/2019] [Accepted: 02/12/2019] [Indexed: 01/18/2023]
Abstract
Our previous study indicated that nicotinamide phosphoribosyltransferase (NAMPT) is released from cells and might be an important extracellular neuroprotective factor in brain ischemia. Here, we tested whether NAMPT protects against ischemic brain injury when administered directly into the intracerebroventricular (ICV) compartment of the cranium. Recombinant NAMPT protein (2 μg) was delivered ICV in mice subjected to 45-min middle cerebral artery occlusion (MCAO), and the effects on infarct volume, sensorimotor function, microglia/macrophage polarization, neutrophil infiltration, and BBB integrity were analyzed. The results indicate that ICV administration of NAMPT significantly reduced infarct volume, retained its beneficial properties even when ICV administration was delayed by 6 h after MCAO, and improved neurological outcomes. NAMPT treatment inhibited pro-inflammatory microglia/macrophages, promoted microglia/macrophage polarization toward the anti-inflammatory phenotype, and reduced the infiltration of neutrophils into the perilesional area after brain ischemia. In vitro studies indicated that multiple pro-inflammatory microglial markers/cytokines were downregulated while multiple anti-inflammatory microglial markers/cytokines were induced in primary microglial cultures treated with NAMPT protein. NAMPT treatment also fortified the blood-brain barrier (BBB), as shown by reduced extravascular leakage of the small-molecule tracer Alexa Fluor 555 Cadaverine and larger-sized endogenous IgGs into brain parenchyma. Thus, NAMPT may protect against ischemic brain injury partly through a novel anti-inflammatory mechanism, which in turn maintains BBB integrity and reduces the infiltration of peripheral inflammatory cells. Taken together, these results provide validation of recombinant NAMPT delivery into the extracellular space as a potential neuroprotective strategy for stroke.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Neurology, BST S520, University of Pittsburgh School of Medicine, 206 Lothrop Street, Pittsburgh, PA, 15260, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Zhongfang Weng
- Department of Neurology, BST S520, University of Pittsburgh School of Medicine, 206 Lothrop Street, Pittsburgh, PA, 15260, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Qinghai Xia
- Department of Neurology, BST S520, University of Pittsburgh School of Medicine, 206 Lothrop Street, Pittsburgh, PA, 15260, USA
| | - Catherine Cao
- North Allegheny Senior High School, Pittsburgh, PA, 15237, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Duquesne University, Pittsburgh, PA, USA
| | - Lihong Han
- Department of Biochemistry, Baotou Medical College, Baotou, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, Wenzhou Medical University, Zhejian, China
| | - Steven H Graham
- Department of Neurology, BST S520, University of Pittsburgh School of Medicine, 206 Lothrop Street, Pittsburgh, PA, 15260, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA
| | - Guodong Cao
- Department of Neurology, BST S520, University of Pittsburgh School of Medicine, 206 Lothrop Street, Pittsburgh, PA, 15260, USA. .,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
27
|
Zhang J, Wu Y, Gao Z. Correlations of C-Reactive Protein (CRP), Interleukin-6 (IL-6), and Insulin Resistance with Cerebral Infarction in Hypertensive Patients. Med Sci Monit 2019; 25:1506-1511. [PMID: 30804318 PMCID: PMC6400023 DOI: 10.12659/msm.912898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The aim of this study was to investigate the correlations of C-reactive protein (CRP), interleukin-6 (IL-6), and insulin resistance (IR) with cerebral infarction in hypertensive patients. Material/Methods A total of 80 patients with cerebral infarction admitted to our hospital from March 2016 to November 2017 were selected and divided into 2 groups according to the diameter of cerebral infarction, namely, lacunar cerebral infarction group (n=40) and cerebral infarction group (n=40). The levels of high-sensitivity CRP (hs-CRP) and IL-6, homeostasis model assessment of IR (HOMA-IR) index and blood pressure level were compared between the 2 groups. The correlations of hs-CRP level, IL-6 level, and IR with the diameter of cerebral infarction, as well as the relationships of hs-CRP level and IR with the neurological function score after cerebral infarction were analyzed. Results The levels of hs-CRP and IL-6 in the cerebral infarction group were significantly higher than those in the lacunar cerebral infarction group (P<0.05). The cerebral infarction group had a markedly higher HOMA-IR index than the lacunar cerebral infarction group (P<0.05), but it had remarkably decreased systolic blood pressure and diastolic blood pressure compared with those in the lacunar cerebral infarction group (P<0.05). There were positive correlations of hs-CRP level, IL-6 level, and IR with the diameter of cerebral infarction (P<0.05). The hs-CRP level and IR had positive correlations with the neurological function score after cerebral infarction (P<0.05). Conclusions In hypertensive patients complicated with cerebral infarction, the body’s inflammatory factors, and IR are positively correlated with the diameter of cerebral infarction, as well as the neurological prognosis of the patients.
Collapse
Affiliation(s)
- Jiang Zhang
- Department of Neurology, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Yunxia Wu
- Department of Neurology, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| | - Zhiqiang Gao
- Department of Neurology, Linyi Central Hospital, Linyi, Shandong, China (mainland)
| |
Collapse
|
28
|
Hersh J, Yang SH. Glia-immune interactions post-ischemic stroke and potential therapies. Exp Biol Med (Maywood) 2018; 243:1302-1312. [PMID: 30537868 DOI: 10.1177/1535370218818172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
IMPACT STATEMENT This article reviews glial cell interactions with the immune system post-ischemic stroke. Research has shown that glial cells in the brain play a role in altering phenotypes of other glial cells and have downstream immune cell targets ultimately regulating a neuroinflammatory response. These interactions may play a deleterious as well as beneficial role in stroke recovery. Furthermore, they may provide a novel way to approach potential therapies, since current stroke drug therapy is limited to only one Food and Drug Administration-approved drug complicated by a narrow therapeutic window. Until this point, most research has emphasized neuroimmune interactions, but little focus has been on bidirectional communication of glial-immune interactions in the ischemic brain. By expanding our understanding of these interactions through a compilation of glial cell effects, we may be able to pinpoint major modulating factors in brain homeostasis to maintain or discover ways to suppress irreversible ischemic damage and improve brain repair.
Collapse
Affiliation(s)
- Jessica Hersh
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Shao-Hua Yang
- Department of Neuroscience and Pharmacology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
29
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
30
|
Shukla V, Fuchs P, Liu A, Cohan CH, Dong C, Wright CB, Perez-Pinzon MA, Dave KR. Recurrent Hypoglycemia Exacerbates Cerebral Ischemic Damage in Diabetic Rats via Enhanced Post-Ischemic Mitochondrial Dysfunction. Transl Stroke Res 2018; 10:78-90. [PMID: 29569040 DOI: 10.1007/s12975-018-0622-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/06/2018] [Accepted: 03/08/2018] [Indexed: 12/17/2022]
Abstract
Diabetes significantly increases the risk of stroke and post-stroke mortality. Recurrent hypoglycemia (RH) is common among diabetes patients owing to glucose-lowering therapies. Earlier, we showed that RH in a rat model of insulin-dependent diabetes exacerbates cerebral ischemic damage. Impaired mitochondrial function has been implicated as a central player in the development of cerebral ischemic damage. Hypoglycemia is also known to affect mitochondrial functioning. The present study tested the hypothesis that prior exposure of insulin-treated diabetic (ITD) rats to RH exacerbates brain damage via enhanced post-ischemic mitochondrial dysfunction. In a rat model of streptozotocin-induced diabetes, we evaluated post-ischemic mitochondrial function in RH-exposed ITD rats. Rats were exposed to five episodes of moderate hypoglycemia prior to the induction of cerebral ischemia. We also evaluated the impact of RH, both alone and in combination with cerebral ischemia, on cognitive function using the Barnes circular platform maze test. We observed that RH exposure to ITD rats leads to increased cerebral ischemic damage and decreased mitochondrial complex I activity. Exposure of ITD rats to RH impaired spatial learning and memory. Our results demonstrate that RH exposure to ITD rats potentially increases post-ischemic damage via enhanced post-ischemic mitochondrial dysfunction.
Collapse
Affiliation(s)
- Vibha Shukla
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Perry Fuchs
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Allen Liu
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Charles H Cohan
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Chuanhui Dong
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Clinton B Wright
- Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA.,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA.,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA.,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami School of Medicine, 1420 NW 9th Ave, NRB/203E, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Evelyn F. McKnight Brain Institute, University of Miami School of Medicine, Miami, FL, 33136, USA. .,Neuroscience Program, University of Miami School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
31
|
The association between plasma endotoxin, endotoxin pathway proteins and outcome after ischemic stroke. Atherosclerosis 2018; 269:138-143. [DOI: 10.1016/j.atherosclerosis.2017.12.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/14/2017] [Accepted: 12/22/2017] [Indexed: 11/21/2022]
|
32
|
Stroke-associated infection independently predicts 3-month poor functional outcome and mortality. J Neurol 2017; 265:370-375. [DOI: 10.1007/s00415-017-8714-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 12/13/2017] [Accepted: 12/13/2017] [Indexed: 10/18/2022]
|
33
|
Schönfeld LM, Dooley D, Jahanshahi A, Temel Y, Hendrix S. Evaluating rodent motor functions: Which tests to choose? Neurosci Biobehav Rev 2017; 83:298-312. [PMID: 29107829 DOI: 10.1016/j.neubiorev.2017.10.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/18/2017] [Accepted: 10/23/2017] [Indexed: 01/11/2023]
Abstract
Damage to the motor cortex induced by stroke or traumatic brain injury (TBI) can result in chronic motor deficits. For the development and improvement of therapies, animal models which possess symptoms comparable to the clinical population are used. However, the use of experimental animals raises valid ethical and methodological concerns. To decrease discomfort by experimental procedures and to increase the quality of results, non-invasive and sensitive rodent motor tests are needed. A broad variety of rodent motor tests are available to determine deficits after stroke or TBI. The current review describes and evaluates motor tests that fall into three categories: Tests to evaluate fine motor skills and grip strength, tests for gait and inter-limb coordination and neurological deficit scores. In this review, we share our thoughts on standardized data presentation to increase data comparability between studies. We also critically evaluate current methods and provide recommendations for choosing the best behavioral test for a new research line.
Collapse
Affiliation(s)
- Lisa-Maria Schönfeld
- Comparative Psychology, Institute of Experimental Psychology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Ali Jahanshahi
- Department of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Yasin Temel
- Department of Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Sven Hendrix
- Department of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium.
| |
Collapse
|
34
|
Mardiguian S, Ladds E, Turner R, Shepherd H, Campbell SJ, Anthony DC. The contribution of the acute phase response to the pathogenesis of relapse in chronic-relapsing experimental autoimmune encephalitis models of multiple sclerosis. J Neuroinflammation 2017; 14:196. [PMID: 28964257 PMCID: PMC5622564 DOI: 10.1186/s12974-017-0969-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 09/21/2017] [Indexed: 12/29/2022] Open
Abstract
Background Increased relapse rates in multiple sclerosis (MS) as a consequence of peripheral immune system activation, owing to infection for example, have been widely reported, but the mechanism remains unclear. Acute brain injury models can be exacerbated by augmenting the hepatic acute phase response (APR). Here, we explored the contribution of the hepatic APR to relapse in two rodent models of MS. Methods Mice with MOG-CFA-induced chronic relapsing experimental autoimmune encephalitis (CR-EAE) were killed before, during and after the first phase of disease, and the brain and liver chemokine, cytokine and acute phase protein (APP) mRNA expression profile was determined. During remission, the APR was reactivated with an intraperitoneal lipopolysaccharide (LPS) and clinical score was monitored throughout. To explore the downstream mediators, CXCL-1, which is induced as part of the APR, was injected into animals with a focal, cytokine/MOG-induced EAE lesion (fEAE) and the cellularity of the lesions was assessed. Results Compared to CFA control, in a rodent CR-EAE model, an hepatic APR preceded clinical signs and central cytokine production in the initial phase of disease. Compared to administration in naïve animals, an LPS challenge during the asymptomatic remission phase of CR-EAE rodents provoked relapse and resulted in the increased and extended expression of specific peripheral hepatic chemokines. CXCL-1 and several other APPs were markedly elevated. A single intravenous administration of the highly induced chemokine, CXCL-1, was found to be sufficient to reactivate the lesions by increasing microglial activation and the recruitment of T cells in fEAE lesions. Conclusions The APR plays a contributing role to the pathology seen in models of chronic brain injury and in translating the effects of peripheral immune system stimulation secondary to trauma or infection into central pathology and behavioural signs. Further elucidation of the exact mechanisms in this process will inform development of more effective, selective therapies in MS that, by suppressing the hepatic chemokine response, may prevent relapse.
Collapse
Affiliation(s)
- Silvy Mardiguian
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK
| | - Emma Ladds
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK.,Department of Primary Care Health Sciences, University of Oxford, Oxford, OX2 6GG, UK
| | - Roberta Turner
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK
| | - Hazel Shepherd
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK
| | - Sandra J Campbell
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford, OX1 4QT, UK.
| |
Collapse
|
35
|
Abstract
Each year, millions of persons worldwide are disabled by stroke. The burden of stroke is expected to increase as a consequence of growth in our elderly population. Outcome is dependent upon limitation of secondary medical processes in the acute setting that lead to deterioration and increased long-term disability. The prevalence of infection after stroke is greater that seen in other medical conditions with similar acuity and its impact upon morbidity and mortality is substantial. Physical impairment and immune modulation are chief determinants in rate of infection after stroke. Each of these factors has been a target for therapeutic intervention. Current best practices for acute stroke management implement strategies for prevention, prompt identification, and treatment of infection. Novel therapies are currently being explored which have the opportunity to greatly minimize infectious complications following stroke. Fever commonly accompanies infection and independently influences stroke outcome. Targeted temperature management provides an additional chance to improve stroke recovery.
Collapse
Affiliation(s)
- Chad M Miller
- Riverside Methodist Hospital, OhioHealth, Columbus, OH, USA.
| | - Réza Behrouz
- Division of Cerebrovascular Diseases, Department of Neurology, School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
36
|
Carson MJ. Recruiting pro-inflammatory monocytes to ischemic injuries: On the job training required! Brain Behav Immun 2016; 53:16-17. [PMID: 26685803 DOI: 10.1016/j.bbi.2015.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 10/22/2022] Open
Affiliation(s)
- Monica J Carson
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, University of California Riverside School of Medicine, United States.
| |
Collapse
|