1
|
Lu W, Wen J. The relationship among H 2S, neuroinflammation and MMP-9 in BBB injury following ischemic stroke. Int Immunopharmacol 2025; 146:113902. [PMID: 39724730 DOI: 10.1016/j.intimp.2024.113902] [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: 09/25/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Blood-brain barrier (BBB) is located at the interface between the central nervous system (CNS) and the circulatory system, which maintains the microenvironmental homeostasis of the CNS. BBB damage is a result of CNS diseases, including ischemic stroke, and is a cause of CNS deterioration. Cerebral ischemia unleashes a profound inflammatory response to remove the damaged tissue in the CNS and prepare the brain for repair. However, the excessive neuroinflammation following stroke onset is associated with BBB breakdown, resulting in neuronal injury and worse neurological outcomes. Additionally, matrix metalloproteinases (MMPs) are likewise responsible for the BBB injury and participate in the pathological processes of neuroinflammation following ischemic stroke. Hydrogen sulfide (H2S) is one of gaseous signaling and freely diffusing molecules. Low concentration of H2S yields the neuroprotection against BBB damage following stroke. This review discussed the current knowledge about the detrimental roles of neuroinflammation and MMPs in BBB injury following ischemic stroke. Specifically, we provided an updated overview of H2S in protecting against BBB injury following ischemic stroke via anti-neuroinflammation and inhibiting MMP-9.
Collapse
Affiliation(s)
- Weizhuo Lu
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Medical Branch, Hefei Technology College, Hefei, China.
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
2
|
Lapin D, Sharma A, Wang P. Extracellular cold-inducible RNA-binding protein in CNS injury: molecular insights and therapeutic approaches. J Neuroinflammation 2025; 22:12. [PMID: 39838468 PMCID: PMC11752631 DOI: 10.1186/s12974-025-03340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Central nervous system (CNS) injuries, such as ischemic stroke (IS), intracerebral hemorrhage (ICH) and traumatic brain injury (TBI), are a significant global burden. The complex pathophysiology of CNS injury is comprised of primary and secondary injury. Inflammatory secondary injury is incited by damage-associated molecular patterns (DAMPs) which signal a variety of resident CNS cells and infiltrating immune cells. Extracellular cold-inducible RNA-binding protein (eCIRP) is a DAMP which acts through multiple immune and non-immune cells to promote inflammation. Despite the well-established role of eCIRP in systemic and sterile inflammation, its role in CNS injury is less elucidated. Recent literature suggests that eCIRP is a pleiotropic inflammatory mediator in CNS injury. eCIRP is also being evaluated as a clinical biomarker to indicate prognosis in CNS injuries. This review provides a broad overview of CNS injury, with a focus on immune-mediated secondary injury and neuroinflammation. We then review what is known about eCIRP in CNS injury, and its known mechanisms in both CNS and non-CNS cells, identifying opportunities for further study. We also explore eCIRP's potential as a prognostic marker of CNS injury severity and outcome. Next, we provide an overview of eCIRP-targeting therapeutics and suggest strategies to develop these agents to ameliorate CNS injury. Finally, we emphasize exploring novel molecular mechanisms, aside from neuroinflammation, by which eCIRP acts as a critical mediator with significant potential as a therapeutic target and prognostic biomarker in CNS injury.
Collapse
Affiliation(s)
- Dmitriy Lapin
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA
| | - Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
- Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, 11030, USA.
| |
Collapse
|
3
|
Owjfard M, Rahimian Z, Karimi F, Borhani-Haghighi A, Mallahzadeh A. A comprehensive review on the neuroprotective potential of resveratrol in ischemic stroke. Heliyon 2024; 10:e34121. [PMID: 39082038 PMCID: PMC11284444 DOI: 10.1016/j.heliyon.2024.e34121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 06/07/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Stroke is the second leading cause of death and the third leading cause of disability worldwide. Globally, 68 % of all strokes are ischemic, with 32 % being hemorrhagic. Ischemic stroke (IS) poses significant challenges globally, necessitating the development of effective therapeutic strategies. IS is among the deadliest illnesses. Major functions are played by neuroimmunity, inflammation, and oxidative stress in the multiple intricate pathways of IS. Secondary brain damage is specifically caused by the early pro-inflammatory activity that follows cerebral ischemia, which is brought on by excessive activation of local microglia and the infiltration of circulating monocytes and macrophages. Resveratrol, a natural polyphenol found in grapes and berries, has shown promise as a neuroprotective agent in IS. This review offers a comprehensive overview of resveratrol's neuroprotective role in IS, focusing on its mechanisms of action and therapeutic potential. Resveratrol exerts neuroprotective effects by activating nuclear factor erythroid 2-related factor 2 (NRF2) and sirtuin 1 (SIRT1) pathways. SIRT1 activation by resveratrol triggers the deacetylation and activation of downstream targets like peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) and forkhead box protein O (FOXO), regulating mitochondrial biogenesis, antioxidant defense, and cellular stress response. Consequently, resveratrol promotes cellular survival and inhibits apoptosis in IS. Moreover, resveratrol activates the NRF2 pathway, a key mediator of the cellular antioxidant response. Activation of NRF2 through resveratrol enhances the expression of antioxidant enzymes, like heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1), which neutralize reactive oxygen species and mitigate oxidative stress in the ischemic brain. Combined, the activation of SIRT1 and NRF2 pathways contributes to resveratrol's neuroprotective effects by reducing oxidative stress, inflammation, and apoptosis in IS. Preclinical studies demonstrate that resveratrol improves functional outcomes, reduces infarct size, regulates cerebral blood flow and preserves neuronal integrity. Gaining a comprehensive understanding of these mechanisms holds promise for the development of targeted therapeutic interventions aimed at promoting neuronal survival and facilitating functional recovery in IS patients and to aid future studies in this matter.
Collapse
Affiliation(s)
- Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Rahimian
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Arashk Mallahzadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Van Roy Z, Kielian T. Tumor necrosis factor regulates leukocyte recruitment but not bacterial persistence during Staphylococcus aureus craniotomy infection. J Neuroinflammation 2024; 21:179. [PMID: 39044282 PMCID: PMC11264501 DOI: 10.1186/s12974-024-03174-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/14/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Craniotomy is a common neurosurgery used to treat intracranial pathologies. Nearly 5% of the 14 million craniotomies performed worldwide each year become infected, most often with Staphylococcus aureus (S. aureus), which forms a biofilm on the surface of the resected bone segment to establish a chronic infection that is recalcitrant to antibiotics and immune-mediated clearance. Tumor necrosis factor (TNF), a prototypical proinflammatory cytokine, has been implicated in generating protective immunity to various infections. Although TNF is elevated during S. aureus craniotomy infection, its functional importance in regulating disease pathogenesis has not been explored. METHODS A mouse model of S. aureus craniotomy infection was used to investigate the functional importance of TNF signaling using TNF, TNFR1, and TNFR2 knockout (KO) mice by quantifying bacterial burden, immune infiltrates, inflammatory mediators, and transcriptional changes by RNA-seq. Complementary experiments examined neutrophil extracellular trap formation, leukocyte apoptosis, phagocytosis, and bactericidal activity. RESULTS TNF transiently regulated neutrophil and granulocytic myeloid-derived suppressor cell recruitment to the brain, subcutaneous galea, and bone flap as evident by significant reductions in both cell types between days 7 to 14 post-infection coinciding with significant decreases in several chemokines, which recovered to wild type levels by day 28. Despite these defects, bacterial burdens were similar in TNF KO and WT mice. RNA-seq revealed enhanced lymphotoxin-α (Lta) expression in TNF KO granulocytes. Since both TNF and LTα signal through TNFR1 and TNFR2, KO mice for each receptor were examined to assess potential redundancy; however, neither strain had any impact on S. aureus burden. In vitro studies revealed that TNF loss selectively altered macrophage responses to S. aureus since TNF KO macrophages displayed significant reductions in phagocytosis, apoptosis, IL-6 production, and bactericidal activity in response to live S. aureus, whereas granulocytes were not affected. CONCLUSION These findings implicate TNF in modulating granulocyte recruitment during acute craniotomy infection via secondary effects on chemokine production and identify macrophages as a key cellular target of TNF action. However, the lack of changes in bacterial burden in TNF KO animals suggests the involvement of additional signals that dictate S. aureus pathogenesis during craniotomy infection.
Collapse
Affiliation(s)
- Zachary Van Roy
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Tammy Kielian
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE, 68198-5900, USA.
| |
Collapse
|
5
|
Owjfard M, Rahmani N, Mallahzadeh A, Bayat M, Borhani-Haghighi A, Karimi F, Namavar MR. Mechanism of action and neuroprotective role of nicorandil in ischemic stroke. Heliyon 2024; 10:e26640. [PMID: 38434007 PMCID: PMC10906150 DOI: 10.1016/j.heliyon.2024.e26640] [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: 01/28/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Nicorandil is a dual mechanism anti-anginal agent that acts as a nitric oxide (NO) donor and a potassium (K+) channel opener. Recent studies have evaluated the effect of nicorandil on ischemic stroke. Neurons have a low tolerance to hypoxia and therefore the brain tissue is significantly vulnerable to ischemia. Current approved treatments for ischemic stroke are tissue plasminogen activators and clot retrieval methods. The narrow therapeutic time window and lack of efficacy in restoring the dying neurons urge researchers to develop an alternative approach. In the terminal stages of anoxia, K+ channels induce hyperpolarization in various types of neuronal cells, leading to decreased neuronal activity and the preservation of the brain's energy. Nicorandil can open these K+ channels and sustain the hyperpolarization phase, which may have a neuroprotective effect during hypoxia. Additionally, we review how nicorandil can improve overall stroke outcomes through its anti-inflammatory, anti-oxidative, and edema-reducing effects. One of the major components evaluated in stroke patients is blood pressure. Studies have demonstrated that the effect of nicorandil on blood pressure is related to both its K+ channel opening and NO donating mechanisms. Since both hypertension and hypotension need correction before stroke intervention, it's crucial to consider the role of nicorandil and its impact on blood pressure. Previously published studies indicate that the right dosage of nicorandil can improve cerebral blood flow without significant changes in hemodynamic profiles. In this review, we discuss how nicorandil may contribute to better stroke outcomes based on previously published literature and laboratory findings.
Collapse
Affiliation(s)
- Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negin Rahmani
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Arashk Mallahzadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Mohammad Reza Namavar
- Histomorphometry & Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
6
|
Gao Y, Fang C, Wang J, Ye Y, Li Y, Xu Q, Kang X, Gu L. Neuroinflammatory Biomarkers in the Brain, Cerebrospinal Fluid, and Blood After Ischemic Stroke. Mol Neurobiol 2023; 60:5117-5136. [PMID: 37258724 DOI: 10.1007/s12035-023-03399-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
The most frequent type of stroke, known as ischemic stroke (IS), is a significant global public health issue. The pathological process of IS and post-IS episodes has not yet been fully explored, but neuroinflammation has been identified as one of the key processes. Biomarkers are objective indicators used to assess normal or pathological processes, evaluate responses to treatment, and predict outcomes, and some biomarkers can also be used as therapeutic targets. After IS, various molecules are produced by different cell types, such as microglia, astrocytes, infiltrating leukocytes, endothelial cells, and damaged neurons, that participate in the neuroinflammatory response within the ischemic brain region. These molecules may either promote or inhibit neuroinflammation and may be released into extracellular spaces, including cerebrospinal fluid (CSF) and blood, due to reasons such as BBB damage. These neuroinflammatory molecules should be valued as biomarkers to monitor whether their expression levels in the blood, CSF, and brain correlate with the diagnosis and prognosis of IS patients or whether they have potential as therapeutic targets. In addition, although some molecules do not directly participate in the process of neuroinflammation, they have been reported to have potential diagnostic or therapeutic value against post-IS neuroinflammation, and these molecules will also be listed. In this review, we summarize the neuroinflammatory biomarkers in the brain, CSF, and blood after an IS episode and the potential value of these biomarkers for the diagnosis, treatment, and prognosis of IS patients.
Collapse
Affiliation(s)
- Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Congcong Fang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xianhui Kang
- Department of Anesthesia, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310006, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
7
|
Xie Q, Lu D, Yuan J, Ren M, Li Y, Wang J, Ma R, Wang J. l-borneol promotes neurovascular unit protection in the subacute phase of transient middle cerebral artery occlusion rats: p38-MAPK pathway activation, anti-inflammatory, and anti-apoptotic effect. Phytother Res 2023; 37:4166-4184. [PMID: 37310024 DOI: 10.1002/ptr.7878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/14/2023]
Abstract
Our previous study showed l-borneol reduced cerebral infarction in the acute stage after cerebral ischemia, but there is little about the study of subacute phase. We herein investigated the cerebral protective effects of l-borneol on neurovascular units (NVU) in the subacute phase after transient middle cerebral artery occlusion (t-MCAO). The t-MCAO model was prepared by the line embolus method. Zea Longa, mNss, HE, and TTC staining were used to evaluate the effect of l-borneol. We evaluated the mechanisms of l-borneol on inflammation, p38 MAPK pathway, and apoptosis, etc. through various technologies. l-borneol 0.2, 0.1, 0.05 g·kg-1 could significantly reduce cerebral infarction rate, alleviate the pathological injury, and inhibit inflammation reaction. l-borneol could also significantly increase brain blood supply, Nissl bodies, and the expression of GFAP. Additionally, l-borneol activated the p38 MAPK signaling pathway, inhibited cell apoptosis, and maintained BBB integrity. l-borneol had a neuroprotective effect, which was related to activating the p38 MAPK signaling pathway, inhibiting inflammatory response and apoptosis, and improving cerebral blood supply to protect BBB and stabilize and remodel NVU. The study will provide a reference for the use of l-borneol in the treatment of ischemic stroke in the subacute phase.
Collapse
Affiliation(s)
- Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Foshan University, Foshan, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Foshan University, Foshan, China
- South China University of Technology, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Rejdak K, Sienkiewicz-Jarosz H, Bienkowski P, Alvarez A. Modulation of neurotrophic factors in the treatment of dementia, stroke and TBI: Effects of Cerebrolysin. Med Res Rev 2023; 43:1668-1700. [PMID: 37052231 DOI: 10.1002/med.21960] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023]
Abstract
Neurotrophic factors (NTFs) are involved in the pathophysiology of neurological disorders such as dementia, stroke and traumatic brain injury (TBI), and constitute molecular targets of high interest for the therapy of these pathologies. In this review we provide an overview of current knowledge of the definition, discovery and mode of action of five NTFs, nerve growth factor, insulin-like growth factor 1, brain derived NTF, vascular endothelial growth factor and tumor necrosis factor alpha; as well as on their contribution to brain pathology and potential therapeutic use in dementia, stroke and TBI. Within the concept of NTFs in the treatment of these pathologies, we also review the neuropeptide preparation Cerebrolysin, which has been shown to resemble the activities of NTFs and to modulate the expression level of endogenous NTFs. Cerebrolysin has demonstrated beneficial treatment capabilities in vitro and in clinical studies, which are discussed within the context of the biochemistry of NTFs. The review focuses on the interactions of different NTFs, rather than addressing a single NTF, by outlining their signaling network and by reviewing their effect on clinical outcome in prevalent brain pathologies. The effects of the interactions of these NTFs and Cerebrolysin on neuroplasticity, neurogenesis, angiogenesis and inflammation, and their relevance for the treatment of dementia, stroke and TBI are summarized.
Collapse
Affiliation(s)
- Konrad Rejdak
- Department of Neurology, Medical University of Lublin, Lublin, Poland
| | | | | | - Anton Alvarez
- Medinova Institute of Neurosciences, Clinica RehaSalud, Coruña, Spain
| |
Collapse
|
9
|
Che X, Roy A, Bresnahan M, Mjaaland S, Reichborn-Kjennerud T, Magnus P, Stoltenberg C, Shang Y, Zhang K, Susser E, Fiehn O, Lipkin WI. Metabolomic analysis of maternal mid-gestation plasma and cord blood in autism spectrum disorders. Mol Psychiatry 2023; 28:2355-2369. [PMID: 37037873 DOI: 10.1038/s41380-023-02051-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/12/2023]
Abstract
The discovery of prenatal and neonatal molecular biomarkers has the potential to yield insights into autism spectrum disorder (ASD) and facilitate early diagnosis. We characterized metabolomic profiles in ASD using plasma samples collected in the Norwegian Autism Birth Cohort from mothers at weeks 17-21 gestation (maternal mid-gestation, MMG, n = 408) and from children on the day of birth (cord blood, CB, n = 418). We analyzed associations using sex-stratified adjusted logistic regression models with Bayesian analyses. Chemical enrichment analyses (ChemRICH) were performed to determine altered chemical clusters. We also employed machine learning algorithms to assess the utility of metabolomics as ASD biomarkers. We identified ASD associations with a variety of chemical compounds including arachidonic acid, glutamate, and glutamine, and metabolite clusters including hydroxy eicospentaenoic acids, phosphatidylcholines, and ceramides in MMG and CB plasma that are consistent with inflammation, disruption of membrane integrity, and impaired neurotransmission and neurotoxicity. Girls with ASD have disruption of ether/non-ether phospholipid balance in the MMG plasma that is similar to that found in other neurodevelopmental disorders. ASD boys in the CB analyses had the highest number of dysregulated chemical clusters. Machine learning classifiers distinguished ASD cases from controls with area under the receiver operating characteristic (AUROC) values ranging from 0.710 to 0.853. Predictive performance was better in CB analyses than in MMG. These findings may provide new insights into the sex-specific differences in ASD and have implications for discovery of biomarkers that may enable early detection and intervention.
Collapse
Affiliation(s)
- Xiaoyu Che
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ayan Roy
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Michaeline Bresnahan
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | | | - Ted Reichborn-Kjennerud
- Norwegian Institute of Public Health, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Per Magnus
- Norwegian Institute of Public Health, Oslo, Norway
| | - Camilla Stoltenberg
- Norwegian Institute of Public Health, Oslo, Norway
- Department of Global Public Health, University of Bergen, Bergen, Norway
| | - Yimeng Shang
- Department of Public Health Sciences, College of Medicine, Penn State University, State College, PA, 16801, USA
| | - Keming Zhang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Ezra Susser
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- New York State Psychiatric Institute, New York, NY, USA
| | - Oliver Fiehn
- UC Davis Genome Center-Metabolomics, University of California, Davis, CA, USA
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA.
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA.
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
10
|
Owjfard M, Karimi F, Mallahzadeh A, Nabavizadeh SA, Namavar MR, Saadi MI, Hooshmandi E, Salehi MS, Zafarmand SS, Bayat M, Karimlou S, Borhani-Haghighi A. Mechanism of action and therapeutic potential of dimethyl fumarate in ischemic stroke. J Neurosci Res 2023. [PMID: 37183360 DOI: 10.1002/jnr.25202] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/16/2023]
Abstract
Dimethyl fumarate (DMF) is an immunomodulatory drug currently approved for the treatment of multiple sclerosis and psoriasis. Its benefits on ischemic stroke outcomes have recently come to attention. To date, only tissue plasminogen activators (tPAs) and clot retrieval methods have been approved by the FDA for the treatment of ischemic stroke. Ischemic conditions lead to inflammation through diverse mechanisms, and recanalization can worsen the state. DMF and the nuclear factor erythroid-derived 2-related factor 2 (Nrf2) pathway it regulates seem to be important in postischemic inflammation, and animal studies have demonstrated that the drug improves overall stroke outcomes. Although the exact mechanism is still unknown, studies indicate that these beneficial impacts are due to the modulation of immune responses, blood-brain barrier permeability, and hemodynamic adjustments. One major component evaluated before, during, and after tPA therapy in stroke patients is blood pressure (BP). Recent studies have found that DMF may impact BP. Both hypotension and hypertension need correction before treatment, which may delay the appropriate intervention. Since BP management is crucial in managing stroke patients, it is important to consider DMF's role in this matter. That being said, it seems further investigations on DMF may lead to an alternative approach for stroke patients. In this article, we discuss the mechanistic roles of DMF and its potential role in stroke based on previously published literature and laboratory findings.
Collapse
Affiliation(s)
- Maryam Owjfard
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz University of Applied Science and Technology (UAST), Shiraz, Iran
| | | | - Arashk Mallahzadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Nabavizadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namavar
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Karimlou
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
11
|
Blood brain barrier-on-a-chip to model neurological diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
12
|
Li C, Zheng H, Xiong J, Huang Y, Li H, Jin H, Ai S, Wang Y, Su T, Sun G, Xiao X, Fu T, Wang Y, Gao X, Liang P. miR-596-3p suppresses brain metastasis of non-small cell lung cancer by modulating YAP1 and IL-8. Cell Death Dis 2022; 13:699. [PMID: 35961957 PMCID: PMC9374706 DOI: 10.1038/s41419-022-05062-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 03/22/2022] [Accepted: 07/04/2022] [Indexed: 01/21/2023]
Abstract
Brain metastasis (BM) frequently occurs in advanced non-small cell lung cancer (NSCLC) and is associated with poor clinical prognosis. Due to the location of metastatic lesions, the surgical resection is limited and the chemotherapy is ineffective because of the existence of the blood brain barrier (BBB). Therefore, it is essential to enhance our understanding about the underlying mechanisms associated with brain metastasis in NSCLC. In the present study, we explored the RNA-Seq data of brain metastasis cells from the GEO database, and extracted RNA collected from primary NSCLC tumors as well as paired brain metastatic lesions followed by microRNA PCR array. Meanwhile, we improved the in vivo model and constructed a cancer stem cell-derived transplantation model of brain metastasis in mice. Our data indicated that the level of miR-596-3p is high in primary NSCLC tumors, but significantly downregulated in the brain metastatic lesion. The prediction target of microRNA suggested that miR-596-3p was considered to modulate two genes essential in the brain invasion process, YAP1 and IL-8 that restrain the invasion of cancer cells and permeability of BBB, respectively. Moreover, in vivo experiments suggested that our model mimics the clinical aspect of NSCLC and improves the success ratio of brain metastasis model. The results demonstrated that miR-596-3p significantly inhibited the capacity of NSCLC cells to metastasize to the brain. Furthermore, these finding elucidated that miR-596-3p exerts a critical role in brain metastasis of NSCLC by modulating the YAP1-IL8 network, and this miRNA axis may provide a potential therapeutic strategy for brain metastasis.
Collapse
Affiliation(s)
- Chenlong Li
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Hongshan Zheng
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Jinsheng Xiong
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Yuxin Huang
- Heilongjiang Tuomeng Technology Co.Ltd, Harbin, 150040, Heilongjiang, China
| | - Haoyang Li
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
- Cancer Systems Biology Center, the China-Japan Union Hospital, Jilin University, Changchun, 130001, Jilin, China
- College of Computer Science and Technology, Jilin University, Changchun, 130001, Jilin, China
| | - Hua Jin
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Siqi Ai
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Yingjie Wang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Tianqi Su
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Guiyin Sun
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Xu Xiao
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Tianjiao Fu
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Yujie Wang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China
| | - Xin Gao
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Peng Liang
- Department of Neurosurgery, Harbin Medical University Cancer Hospital, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
13
|
Ryan E, Kelly L, Stacey C, Huggard D, Duff E, McCollum D, Leonard A, Boran G, Doherty DR, Bolger T, Molloy EJ. Mild-to-severe traumatic brain injury in children: altered cytokines reflect severity. J Neuroinflammation 2022; 19:36. [PMID: 35130911 PMCID: PMC8822689 DOI: 10.1186/s12974-022-02390-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/14/2022] [Indexed: 11/10/2022] Open
Abstract
Background Paediatric traumatic brain injury (TBI) is recognised to have significant longer-term neurocognitive effects. Childhood is a time of high risk for head injury. Functional recovery is variable with a combination of any or all of physical, cognitive and emotional impairment. Immune activation and alteration in cytokine levels are present following TBI which may differ from adults. Methods Pro- and anti-inflammatory cytokines including Interleukin (IL)-2, IL-4, IL-6, IL-8, IL-10, IL-17A, Tumor Necrosis Factor (TNF)-α and Interferon (IFN)-γ were examined at baseline and following in vitro treatment with endotoxin of whole blood, in the following children: severe TBI (sTBI: initial Glasgow coma scale(GCS) ≤ 8), mild TBI (mTBI; GCS 14/15) at 0-4d and at 10-14d post-TBI and compared to healthy age-matched controls. Results The study enrolled 208 children, including 110 with TBI cohort (n = 104 mild; 6 severe) and controls (n = 98). At baseline all children with TBI had increased IL-6. The mTBI group had significantly increased IFN-γ versus controls. In sTBI at baseline, IFN-γ was decreased compared to controls. At baseline IL-8, IL-10, IL-17A, and TNF-α were decreased in mTBI compared to controls. This persisted at 2 week post-mTBI. The AUC for detecting mTBI was 0.801 CI (0.73–086) using IL6/IL10 ratio. mTBI showed a greater fold change in IL-8 and TNF-α in response to endotoxin stimulation, a response that persisted at 2 weeks. Children with sTBI did not have a significant IL-6 response to endotoxin, but did show an increase in IL-17A. Conclusion Children with all TBI including mTBI show altered cytokine profiles and altered endotoxin responses. Although cytokines increased in sTBI especially in response to endotoxin, suppressed responses were found in mTBI coupled with persistent immune dysfunction post-injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02390-5.
Collapse
Affiliation(s)
- Emer Ryan
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland
| | - Lynne Kelly
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Catherine Stacey
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Dean Huggard
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Department of Paediatrics, Waterford Regional Hospital, Waterford, Ireland
| | - Eimear Duff
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland
| | - Danielle McCollum
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland.,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland.,Department of Paediatric Emergency Medicine, Royal Children's Hospital, Melbourne, Australia
| | - Ann Leonard
- Department of Biochemistry, Tallaght University Hospital, Dublin 24, Ireland
| | - Gerard Boran
- Department of Biochemistry, Tallaght University Hospital, Dublin 24, Ireland
| | - Dermot R Doherty
- Critical Care Medicine, Children's Health Ireland (CHI) at Temple Street, Dublin 1, Ireland
| | - Turlough Bolger
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland.,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland
| | - Eleanor J Molloy
- Department of Paediatrics, Trinity College, The University of Dublin, Dublin, Ireland. .,Trinity Translational Medicine Institute (TTMI), Trinity Centre for Health Sciences, Tallaght University Hospital, Dublin 24, Ireland. .,Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Trinity Translational Medicine Institute, St James Hospital, Dublin 8, Ireland. .,Paediatric Emergency Medicine, Children's Hospital Ireland (CHI) at Tallaght, Dublin 24, Ireland. .,Department of Neonatology, CHI at Crumlin, Dublin, Ireland. .,Department of Neonatology, Coombe Women and Infants University Hospital Dublin, Dublin, Ireland.
| |
Collapse
|
14
|
Vedam-Mai V. Harnessing the immune system for the treatment of Parkinson's disease. Brain Res 2021; 1758:147308. [PMID: 33524380 DOI: 10.1016/j.brainres.2021.147308] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/11/2020] [Accepted: 01/16/2021] [Indexed: 01/03/2023]
Abstract
Current treatment options for Parkinson's disease (PD) typically aim to replace dopamine, and hence only provide symptomatic relief. However, in the long run, this approach alone loses its efficacy as it is associated with debilitating side effects. Hence there is an unmet clinical need for addressing levodopa resistant symptoms, and an urgency to develop therapies that can halt or prevent the course of PD. The premise that α-syn can transmit from cell-to-cell in a prion like manner has opened up the possibility for the use of immunotherapy in PD. There is evidence for inflammation in PD as is evidenced by microglial activation, as well as the involvement of the peripheral immune system in PD, and peripheral inflammation can exacerbate dopaminergic degeneration as seen in animal models of the disease. However, mechanisms that link the immune system with PD are not clear, and the sequence of immune responses with respect to PD are still unknown. Nevertheless, our present knowledge offers avenues for the development of immune-based therapies for PD. In order to successfully employ such strategies, we must comprehend the state of the peripheral immune system during the course of PD. This review describes the developments in the field of both active and passive immunotherapies in the treatment of PD, and highlights the crucial need for future research for clarifying the role of inflammation and immunity in this debilitating disease.
Collapse
|
15
|
From "Leaky Gut" to Impaired Glia-Neuron Communication in Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:129-155. [PMID: 33834399 DOI: 10.1007/978-981-33-6044-0_9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the last three decades, the robust scientific data emerged, demonstrating that the immune-inflammatory response is a fundamental component of the pathophysiology of major depressive disorder (MDD). Psychological stress and various inflammatory comorbidities contribute to such immune activation. Still, this is not uncommon that patients with depression do not have defined inflammatory comorbidities, and alternative mechanisms of immune activation need to take place. The gastrointestinal (GI) tract, along with gut-associated lymphoid tissue (GALT), constitutes the largest lymphatic organ in the human body and forms the biggest surface of contact with the external environment. It is also the most significant source of bacterial and food-derived antigenic material. There is a broad range of reciprocal interactions between the GI tract, intestinal microbiota, increased intestinal permeability, activation of immune-inflammatory response, and the CNS that has crucial implications in brain function and mental health. This intercommunication takes place within the microbiota-gut-immune-glia (MGIG) axis, and glial cells are the main orchestrator of this communication. A broad range of factors, including psychological stress, inflammation, dysbiosis, may compromise the permeability of this barrier. This leads to excessive bacterial translocation and the excessive influx of food-derived antigenic material that contributes to activation of the immune-inflammatory response and depressive psychopathology. This chapter summarizes the role of increased intestinal permeability in MDD and mechanisms of how the "leaky gut" may contribute to immune-inflammatory response in this disorder.
Collapse
|
16
|
Nian K, Harding IC, Herman IM, Ebong EE. Blood-Brain Barrier Damage in Ischemic Stroke and Its Regulation by Endothelial Mechanotransduction. Front Physiol 2020; 11:605398. [PMID: 33424628 PMCID: PMC7793645 DOI: 10.3389/fphys.2020.605398] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
Ischemic stroke, a major cause of mortality in the United States, often contributes to disruption of the blood-brain barrier (BBB). The BBB along with its supportive cells, collectively referred to as the “neurovascular unit,” is the brain’s multicellular microvasculature that bi-directionally regulates the transport of blood, ions, oxygen, and cells from the circulation into the brain. It is thus vital for the maintenance of central nervous system homeostasis. BBB disruption, which is associated with the altered expression of tight junction proteins and BBB transporters, is believed to exacerbate brain injury caused by ischemic stroke and limits the therapeutic potential of current clinical therapies, such as recombinant tissue plasminogen activator. Accumulating evidence suggests that endothelial mechanobiology, the conversion of mechanical forces into biochemical signals, helps regulate function of the peripheral vasculature and may similarly maintain BBB integrity. For example, the endothelial glycocalyx (GCX), a glycoprotein-proteoglycan layer extending into the lumen of bloods vessel, is abundantly expressed on endothelial cells of the BBB and has been shown to regulate BBB permeability. In this review, we will focus on our understanding of the mechanisms underlying BBB damage after ischemic stroke, highlighting current and potential future novel pharmacological strategies for BBB protection and recovery. Finally, we will address the current knowledge of endothelial mechanotransduction in BBB maintenance, specifically focusing on a potential role of the endothelial GCX.
Collapse
Affiliation(s)
- Keqing Nian
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Ian C Harding
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Ira M Herman
- Department of Development, Molecular, and Chemical Biology, Tufts Sackler School of Graduate Biomedical Sciences, Boston, MA, United States.,Center for Innovations in Wound Healing Research, Tufts University School of Medicine, Boston, MA, United States
| | - Eno E Ebong
- Department of Bioengineering, Northeastern University, Boston, MA, United States.,Department of Chemical Engineering, Northeastern University, Boston, MA, United States.,Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
17
|
Sato K, Tobo A, Mogi C, Tobo M, Yamane N, Tosaka M, Tomura H, Im DS, Okajima F. The protective role of proton-sensing TDAG8 in the brain injury in a mouse ischemia reperfusion model. Sci Rep 2020; 10:17193. [PMID: 33057165 PMCID: PMC7566628 DOI: 10.1038/s41598-020-74372-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
Extracellular acidification in the brain has been observed in ischemia; however, the physiological and pathophysiological implications of the pH reduction remain largely unknown. Here, we analyzed the roles of proton-sensing G protein-coupled receptors, including T-cell death-associated gene 8 (TDAG8), ovarian cancer G protein-coupled receptor 1 (OGR1), and G protein-coupled receptor 4 (GPR4) in a mouse ischemia reperfusion model. Cerebral infarction and dysfunctional behavior with transient middle cerebral artery occlusion (tMCAO) and subsequent reperfusion were exacerbated by the deficiency of TDAG8, whereas no significant effect was observed with the deficiency of OGR1 or GPR4. We confirmed that the pH of the predicted infarction region was 6.5. TDAG8 mRNA was observed in Iba1-positive microglia in the mouse brain. The tMCAO increased the mRNA expression of tumor necrosis factor-α in the ipsilateral cerebral hemisphere and evoked morphological changes in microglia in an evolving cerebral injury. These tMCAO-induced actions were significantly enhanced by the TDAG8 deficiency. Administration of minocycline, which is known to inhibit microglial activation, improved the cerebral infarction and dysfunctional behavior induced by tMCAO in the TDAG8-deficient mouse. Thus, acidic pH/TDAG8 protects against cerebral infarction caused by tMCAO, at least due to the mechanism involving the inhibition of microglial functions.
Collapse
Affiliation(s)
- Koichi Sato
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan.
| | - Ayaka Tobo
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Chihiro Mogi
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Masayuki Tobo
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, 371-8512, Japan
| | - Nobuhiro Yamane
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Masahiko Tosaka
- Department of Neurosurgery, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Hideaki Tomura
- Laboratory of Cell Signaling Regulation, Department of Life Sciences, School of Agriculture, Meiji University, Kawasaki, 214-8571, Japan
| | - Dong-Soon Im
- College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Fumikazu Okajima
- Laboratory of Signal Transduction, Faculty of Pharmaceutical Sciences, Aomori University, Aomori, 030-0943, Japan
| |
Collapse
|
18
|
Zhang G, Lu Y, Yang L, Dong Y, Wen J, Xu J, Zhang Q. Methylene blue post-treatment improves hypoxia-ischemic recovery in a neonatal rat model. Neurochem Int 2020; 139:104782. [PMID: 32628986 DOI: 10.1016/j.neuint.2020.104782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/11/2020] [Accepted: 06/03/2020] [Indexed: 02/05/2023]
Abstract
Recent work suggested that methylene blue (MB) has beneficial effects in a variety of neurological disorders, while its role in neonatal hypoxic-ischemic (HI) encephalopathy is still unclear. The current study was designed to investigate the effects of MB on HI-induced brain damage and its underlying mechanisms. The results showed that MB treatment can strongly attenuate HI-induced brain loss and neuronal damage in the cortex and hippocampus of neonatal rats. Further mechanistic analysis suggested that MB treatment was able to significantly reduce blood-brain barrier disruption after HI insult. In addition, MB profoundly inhibited microglia and astrocyte activation and the pro-inflammatory cytokines production in neonatal cortex and hippocampus after HI. Further, MB treatment resulted in dramatic suppression of oxidative damage, as evidenced by robustly decreased DHE and protein carbonyls levels in HI brain. Moreover, MB strongly preserved mitochondrial function by repressing HI-induced mitochondrial fragmentation, and the following neuronal death in cortex and hippocampus. Finally, behavioral tests revealed that MB significantly improved the spatial reference memory and motor coordination of neonatal HI rats. Taken together, these findings demonstrate that the mechanisms behind neuroprotective actions of methylene blue are multifactorial, including suppression of oxidative stress and neuroinflammation, restoration of mitochondrial function, as well as attenuation of blood-brain barrier disruption. Our study might provide further directions for MB as a promising option in neonatal HI encephalopathy therapy.
Collapse
Affiliation(s)
- Guangwei Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu, Sichuan, 610041, PR China; Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, No.439 Xuanhua Rd., Yongchuan, Chongqing, 646000, PR China; Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Luodan Yang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Jin Wen
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu, Sichuan, 610041, PR China; Department of Neurosurgery, Yongchuan Hospital of Chongqing Medical University, No.439 Xuanhua Rd., Yongchuan, Chongqing, 646000, PR China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, No. 37 Guoxue Street, Chengdu, Sichuan, 610041, PR China.
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
19
|
Rudzki L, Maes M. The Microbiota-Gut-Immune-Glia (MGIG) Axis in Major Depression. Mol Neurobiol 2020; 57:4269-4295. [DOI: 10.1007/s12035-020-01961-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
|
20
|
Benmakhlouf Y, Zian Z, Nourouti NG, Barakat A, Mechita MB. Potential Cytokine Biomarkers in Intellectual Disability. Endocr Metab Immune Disord Drug Targets 2020; 21:569-576. [PMID: 32600239 DOI: 10.2174/1871530320666200628024944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/22/2022]
Abstract
Intellectual disability (ID), previously called mental retardation, is the most common neurodevelopmental disorder characterized by life-long intellectual and adaptive functioning impairments that have an impact on individuals, families, and society. Its prevalence is estimated to 3% of the general population and its etiology is still insufficiently understood. Besides the involvement of genetic and environmental factors, immunological dysfunctions have been also suggested to contribute to the pathophysiology of ID. Over the years, immune biomarkers related to ID have gained significant attention and researchers have begun to look at possible cytokine profiles in individuals suffered from this disorder. In fact, in addition to playing crucial physiological roles in the majority of normal neurodevelopmental processes, cytokines exert an important role in neuroinflammation under pathological conditions, and interactions between the immune system and central nervous system have long been under investigation. Cytokine levels imbalance has been reported associated with some behavioral characteristics and the onset of some syndromic forms of ID. In this review, we will focus on immunological biomarkers, especially the cytokine profiles that have been identified in people with ID. Thus, data reported and discussed in the present paper may provide additional information to start further studies and to plan strategies for early identification and managing of ID.
Collapse
Affiliation(s)
- Yousra Benmakhlouf
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Zeineb Zian
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Naima G Nourouti
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Amina Barakat
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| | - Mohcine B Mechita
- Biomedical Genomics and Oncogenetics Research Laboratory, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University, Tetouan, Morocco
| |
Collapse
|
21
|
Löscher W, Potschka H, Sisodiya SM, Vezzani A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Pharmacol Rev 2020; 72:606-638. [PMID: 32540959 PMCID: PMC7300324 DOI: 10.1124/pr.120.019539] [Citation(s) in RCA: 446] [Impact Index Per Article: 89.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Epilepsy is a chronic neurologic disorder that affects over 70 million people worldwide. Despite the availability of over 20 antiseizure drugs (ASDs) for symptomatic treatment of epileptic seizures, about one-third of patients with epilepsy have seizures refractory to pharmacotherapy. Patients with such drug-resistant epilepsy (DRE) have increased risks of premature death, injuries, psychosocial dysfunction, and a reduced quality of life, so development of more effective therapies is an urgent clinical need. However, the various types of epilepsy and seizures and the complex temporal patterns of refractoriness complicate the issue. Furthermore, the underlying mechanisms of DRE are not fully understood, though recent work has begun to shape our understanding more clearly. Experimental models of DRE offer opportunities to discover, characterize, and challenge putative mechanisms of drug resistance. Furthermore, such preclinical models are important in developing therapies that may overcome drug resistance. Here, we will review the current understanding of the molecular, genetic, and structural mechanisms of ASD resistance and discuss how to overcome this problem. Encouragingly, better elucidation of the pathophysiological mechanisms underpinning epilepsies and drug resistance by concerted preclinical and clinical efforts have recently enabled a revised approach to the development of more promising therapies, including numerous potential etiology-specific drugs ("precision medicine") for severe pediatric (monogenetic) epilepsies and novel multitargeted ASDs for acquired partial epilepsies, suggesting that the long hoped-for breakthrough in therapy for as-yet ASD-resistant patients is a feasible goal. SIGNIFICANCE STATEMENT: Drug resistance provides a major challenge in epilepsy management. Here, we will review the current understanding of the molecular, genetic, and structural mechanisms of drug resistance in epilepsy and discuss how the problem might be overcome.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Heidrun Potschka
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Sanjay M Sisodiya
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| | - Annamaria Vezzani
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany (W.L.); Center for Systems Neuroscience, Hannover, Germany (W.L.); Institute of Pharmacology, Toxicology and Pharmacy, Ludwig-Maximilians-University, Munich, Germany (H.P.); Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, United Kingdom (S.S); and Department of Neuroscience, Mario Negri Institute for Pharmacological Research Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy (A.V.)
| |
Collapse
|
22
|
Cuprizone Affects Hypothermia-Induced Neuroprotection and Enhanced Neuroblast Differentiation in the Gerbil Hippocampus after Ischemia. Cells 2020; 9:cells9061438. [PMID: 32531881 PMCID: PMC7349804 DOI: 10.3390/cells9061438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022] Open
Abstract
In the present study, we investigated the effects of cuprizone on cell death, glial activation, and neuronal plasticity induced by hypothermia after ischemia in gerbils. Food was supplemented with cuprizone at 0.2% ad libitum for eight weeks. At six weeks after diet feeing, gerbils received transient forebrain ischemia with or without hypothermic preconditioning. Cuprizone treatment for 8 weeks increased the number of astrocytes, microglia, and pro-inflammatory cytokine levels in the hippocampus. In addition, cuprizone treatment significantly decreased the number of proliferating cells and neuroblasts in the dentate gyrus. Brain ischemia caused cell death, disruption of myelin basic proteins, and reactive gliosis in CA1. In addition, ischemia significantly increased pro-inflammatory cytokines and the number of proliferating cells and differentiating neuroblasts in the dentate gyrus. In contrast, hypothermic conditioning attenuated these changes in CA1 and the dentate gyrus. However, cuprizone treatment decreased cell survival induced by hypothermic preconditioning after ischemia and increased the number of reactive microglia and astrocytes in CA1 as well as that of macrophages in the subcallosal zone. These changes occurred because the protective effect of hypothermia in ischemic damage was disrupted by cuprizone administration. Furthermore, cuprizone decreased ischemia-induced proliferating cells and neuroblasts in the dentate gyrus.
Collapse
|
23
|
Goasdoue K, Chand KK, Miller SM, Lee KM, Colditz PB, Wixey JA, Bjorkman ST. Seizures Are Associated with Blood-Brain Barrier Disruption in a Piglet Model of Neonatal Hypoxic-Ischaemic Encephalopathy. Dev Neurosci 2019; 40:1-16. [PMID: 31048585 DOI: 10.1159/000499365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/04/2019] [Indexed: 11/19/2022] Open
Abstract
Seizures in the neonatal period are most often symptomatic of central nervous system (CNS) dysfunction and the most common cause is hypoxic-ischaemic encephalopathy (HIE). Seizures are associated with poor long-term outcomes and increased neuropathology. Blood-brain barrier (BBB) disruption and inflammation may contribute to seizures and increased neuropathology but are incompletely understood in neonatal HIE. The aim of this study was to investigate the impact of seizures on BBB integrity in a preclinical model of neonatal hypoxic-ischaemic (HI) injury. Piglets (age: <24 h) were subjected to a 30-min HI insult followed by recovery to 72 h post-insult. Amplitude-integrated electroencephalography (aEEG) was performed and seizure burden and background aEEG pattern were analysed. BBB disruption was evaluated in the parietal cortex and hippocampus by means of immunohistochemistry and Western blot. mRNA and protein expression of tight-junction proteins (zonula-occludens 1 [ZO1], occludin [OCLN], and claudin-5 [CLDN5]) was assessed using quantitative polymerase chain reaction (qPCR) and Western blot. In addition, mRNA from genes associated with BBB disruption vascular endothelial growth factor (VEGF) and matrix metalloproteinase 2 (MMP2) as well as inflammatory cytokines and chemokines was assessed with qPCR. Piglets that developed seizures following HI (HI-Sz) had significantly greater injury, as demonstrated by poorer aEEG background pattern scores, lower neurobehavioural scores, and greater histopathology. HI-Sz animals had severe IgG extravasation into brain tissue and uptake into neurons as well as significantly greater levels of IgG in both brain regions as assessed by Western blot. IgG protein in both brain regions was significantly associated with seizure burden, aEEG pattern scores, and neurobehavioural scores. There was no difference in mRNA expression of the tight junctions, however a significant loss of ZO1 and OCLN protein was observed in the parietal cortex. The inflammatory genes TGFβ, IL1β, IL8, IL6, and TNFα were significantly upregulated in HI-Sz animals. MMP2 was significantly increased in animals with seizures compared with animals without seizures. Increasing our understanding of neuropathology associated with seizure is vital because of the association between seizure and poor outcomes. Investigating the BBB is a major untapped area of research and a potential avenue for novel treatments.
Collapse
Affiliation(s)
- Kate Goasdoue
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kirat Kishore Chand
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stephanie Melita Miller
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Kah Meng Lee
- Institute of Health Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul Bernard Colditz
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Julie Anne Wixey
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia
| | - Stella Tracey Bjorkman
- The University of Queensland Perinatal Research Centre, UQ Centre for Clinical Research, Herston, Queensland, Australia,
| |
Collapse
|
24
|
Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cell Physiol 2019; 316:C135-C153. [PMID: 30379577 PMCID: PMC6397344 DOI: 10.1152/ajpcell.00136.2018] [Citation(s) in RCA: 546] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022]
Abstract
As part of the neurovascular unit, the blood-brain barrier (BBB) is a unique, dynamic regulatory boundary that limits and regulates the exchange of molecules, ions, and cells between the blood and the central nervous system. Disruption of the BBB plays an important role in the development of neurological dysfunction in ischemic stroke. Blood-borne substances and cells have restricted access to the brain due to the presence of tight junctions between the endothelial cells of the BBB. Following stroke, there is loss of BBB tight junction integrity, leading to increased paracellular permeability, which results in vasogenic edema, hemorrhagic transformation, and increased mortality. Thus, understanding principal mediators and molecular mechanisms involved in BBB disruption is critical for the development of novel therapeutics to treat ischemic stroke. This review discusses the current knowledge of how neuroinflammation contributes to BBB damage in ischemic stroke. Specifically, we provide an updated overview of the role of cytokines, chemokines, oxidative and nitrosative stress, adhesion molecules, matrix metalloproteinases, and vascular endothelial growth factor as well as the role of different cell types in the regulation of BBB permeability in ischemic stroke.
Collapse
Affiliation(s)
- Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Kimberly E Hawkins
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Sylvain Doré
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
- Departments of Anesthesiology, Neurology, Psychiatry, Psychology, and Pharmaceutics, McKnight Brain Institute, University of Florida , Gainesville, Florida
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida , Gainesville, Florida
| |
Collapse
|
25
|
Jank L, Pinto-Espinoza C, Duan Y, Koch-Nolte F, Magnus T, Rissiek B. Current Approaches and Future Perspectives for Nanobodies in Stroke Diagnostic and Therapy. Antibodies (Basel) 2019; 8:antib8010005. [PMID: 31544811 PMCID: PMC6640704 DOI: 10.3390/antib8010005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/20/2018] [Accepted: 12/27/2018] [Indexed: 12/15/2022] Open
Abstract
Antibody-based biologics are the corner stone of modern immunomodulatory therapy. Though highly effective in dampening systemic inflammatory processes, their large size and Fc-fragment mediated effects hamper crossing of the blood brain barrier (BBB). Nanobodies (Nbs) are single domain antibodies derived from llama or shark heavy-chain antibodies and represent a new generation of biologics. Due to their small size, they display excellent tissue penetration capacities and can be easily modified to adjust their vivo half-life for short-term diagnostic or long-term therapeutic purposes or to facilitate crossing of the BBB. Furthermore, owing to their characteristic binding mode, they are capable of antagonizing receptors involved in immune signaling and of neutralizing proinflammatory mediators, such as cytokines. These qualities combined make Nbs well-suited for down-modulating neuroinflammatory processes that occur in the context of brain ischemia. In this review, we summarize recent findings on Nbs in preclinical stroke models and how they can be used as diagnostic and therapeutic reagents. We further provide a perspective on the design of innovative Nb-based treatment protocols to complement and improve stroke therapy.
Collapse
Affiliation(s)
- Larissa Jank
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Carolina Pinto-Espinoza
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Yinghui Duan
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany.
| |
Collapse
|
26
|
Sun L, Zhang Y, Liu E, Ma Q, Anatol M, Han H, Yan J. The roles of astrocyte in the brain pathologies following ischemic stroke. Brain Inj 2018; 33:712-716. [PMID: 30335519 DOI: 10.1080/02699052.2018.1531311] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aim: In this work, we systematically explored the physiological functions of astrocytes and their roles following ischemic stroke, additionally, the potential therapy strategy targeting the astrocytes was also discussed. Methods: This work searched the PubMed database (including MEDLINE) until 14 Feb 2018, and furthermore, the studies were identified through cross-referencing and by consulting the experts in this field. Results: This study indicated that the astrocytes can not only play harmful roles following ischemic stroke through release of inflammatory factors and formation of glial scar but also have protective effects through quenching glutamate excitotoxicity and maintaining the clearance function of glymphatic system in brain. Conclusion: Owing to their important roles in physiological functions of brain and in the pathological conditions following ischemic stroke, the astrocytes might be a potential but promising therapeutic target for treating the ischemic stroke in the future.
Collapse
Affiliation(s)
- Linlin Sun
- a Department of Anatomy and Histology, School of Basic Medical Sciences , Peking University , Beijing , China
| | - Yixuan Zhang
- a Department of Anatomy and Histology, School of Basic Medical Sciences , Peking University , Beijing , China
| | - E Liu
- a Department of Anatomy and Histology, School of Basic Medical Sciences , Peking University , Beijing , China
| | - Qingyi Ma
- b Center for Perinatal Biology, School of Medicine , Loma Linda University , Loma Linda , USA
| | - Manaenko Anatol
- c Departments of Neurology , University of Erlangen-Nuremberg , Erlangen , Germany
| | - Hongbin Han
- d Beijing Key Lab of Magnetic Resonance Imaging Technology , Beijing , China
| | - Junhao Yan
- a Department of Anatomy and Histology, School of Basic Medical Sciences , Peking University , Beijing , China.,d Beijing Key Lab of Magnetic Resonance Imaging Technology , Beijing , China
| |
Collapse
|
27
|
Behrouzifar S, Vakili A, Barati M. The Effects of Mouse Recombinant Resistin on mRNA Expression of Proinflammatory and Anti-Inflammatory Cytokines and Heat Shock Protein-70 in Experimental Stroke Model. J Stroke Cerebrovasc Dis 2018; 27:3272-3279. [PMID: 30120034 DOI: 10.1016/j.jstrokecerebrovasdis.2018.07.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 07/04/2018] [Accepted: 07/16/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Our recent research showed that resistin has a neuroprotective effect against stroke-induced injury through suppressing apoptosis and oxidative stress. However, the molecular mechanism of neuroprotection of resistin is unclear. This work was designed to examine the effect of mouse recombinant resistin on mRNA expression of Tumor necrosis factor-α (TNF-α), Interleukin-1β (IL-1β), Interleukin-10 (IL-10), Transforming growth factor-β1 (TGF- β1), and Heat shock protein-70 (HSP-70) in mouse model of stroke. MATERIALS AND METHODS Transient focal cerebral ischemia was induced by the middle cerebral artery occlusion (MCAO) in mice. TNF-α, IL-1β, IL-10, TGF-β1, and HSP-70 mRNA were detected at sham (0 hour), 3 hours, 6 hours, 12 hours, and 24 hours after MCAO using real-time QRT-PCR method. Moreover, animals were treated with resistin at the dose of 400ng/mouse at the commencement of MCAO, and mRNA expression of the cytokines and HSP-70 was measured 24 hours after MCAO. RESULTS Tumor necrosis factor-α and IL-1β mRNA expression markedly increased at 12-hour time point and then returned to the basal level at 24 hours after MCAO; but HSP-70 mRNA expression increased at 24-hour time point. Furthermore, resistin (400 ng/mouse) significantly increased TGF-β1 and IL-10 and decreased HSP-70 gene expression at 24 hours after MCAO. CONCLUSIONS Our findings revealed that a molecular mechanism of attenuating ischemic damage by resistin administration probably is increased mRNA expression of anti-inflammatory cytokines. However, applying resistin in the clinical settings for the treatment of stroke deserves further researches in the future.
Collapse
Affiliation(s)
- Sedigheh Behrouzifar
- Research Center and Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Abedin Vakili
- Research Center and Department of Physiology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | - Mehdi Barati
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
28
|
Flurbiprofen axetil attenuates cerebral ischemia/reperfusion injury by reducing inflammation in a rat model of transient global cerebral ischemia/reperfusion. Biosci Rep 2018. [PMID: 29540536 PMCID: PMC6435563 DOI: 10.1042/bsr20171562] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke has been ranked as the second cause of death in patients worldwide. Inflammation which is activated during cerebral ischemia/reperfusion (I/R) is an important mechanism leading to brain injury. The present study aimed to investigate the effect of flurbiprofen axetil on cerebral I/R injury and the role of inflammation in this process. Rats were subjected to sham operation or global cerebral I/R with or without flurbiprofen axetil (5 or 10 mg/kg). Global cerebral ischemia was achieved by occlusion of bilateral common carotid arteries combined with hypotension for 20 min followed by reperfusion for 72 h. Then the neurological deficit score, hippocampal cell apoptosis, levels of aquaporin (AQP) 4, AQP9, intercellular cell adhesion molecule-1 (ICAM-1), nuclear factor-κB (NF-κB), tumor necrosis factor (TNF-α), interleukin-1 β (IL-1β), thromboxane B2 (TXB2), and 6-keto-PGI1α were assessed. After reperfusion, neurological deficit score was significantly increased accompanied by severe neuronal damage (exacerbated morphological deficit, increased terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay (TUNEL)-positive cells and cleaved caspase-3 protein expression in hippocampal CA1 region). Cerebral I/R injury also enhanced expressions of TNF-α, IL-1β, NF-κB, AQP4 and AQP9 as well as TXB2 and TXB2/6-keto-PGI1α. All these changes were reversed by pretreatment with flurbiprofen axetil. Flurbiprofen axetil protects the brain from cerebral I/R injury through reducing inflammation and brain edema.
Collapse
|
29
|
Toyama K, Spin JM, Deng AC, Huang TT, Wei K, Wagenhäuser MU, Yoshino T, Nguyen H, Mulorz J, Kundu S, Raaz U, Adam M, Schellinger IN, Jagger A, Tsao PS. MicroRNA-Mediated Therapy Modulating Blood–Brain Barrier Disruption Improves Vascular Cognitive Impairment. Arterioscler Thromb Vasc Biol 2018; 38:1392-1406. [DOI: 10.1161/atvbaha.118.310822] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Kensuke Toyama
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Joshua M. Spin
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Alicia C. Deng
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences (T.-T.H., H.N.), Stanford University School of Medicine, CA
| | - Ke Wei
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Markus U. Wagenhäuser
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Takuya Yoshino
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Huy Nguyen
- Department of Neurology and Neurological Sciences (T.-T.H., H.N.), Stanford University School of Medicine, CA
| | - Joscha Mulorz
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Soumajit Kundu
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Uwe Raaz
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Matti Adam
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Isabel N. Schellinger
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Ann Jagger
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| | - Philip S. Tsao
- From the Division of Cardiovascular Medicine (K.T., J.M.S., A.C.D., M.U.W., T.Y., J.M., U.R., M.A., I.N.S., A.J., P.S.T.)
- VA Palo Alto Health Care System, CA (K.T., J.M.S., A.C.D., K.W., M.U.W., T.Y., J.M., S.K., U.R., M.A., I.N.S., A.J., P.S.T.)
| |
Collapse
|
30
|
Rudzki L, Szulc A. "Immune Gate" of Psychopathology-The Role of Gut Derived Immune Activation in Major Psychiatric Disorders. Front Psychiatry 2018; 9:205. [PMID: 29896124 PMCID: PMC5987016 DOI: 10.3389/fpsyt.2018.00205] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022] Open
Abstract
Interaction between the gastrointestinal tract (GI) and brain functions has recently become a topic of growing interest in psychiatric research. These multidirectional interactions take place in the so-called gut-brain axis or more precisely, the microbiota-gut-brain axis. The GI tract is the largest immune organ in the human body and is also the largest surface of contact with the external environment. Its functions and permeability are highly influenced by psychological stress, which are often a precipitating factor in the first episode, reoccurrence and/or deterioration of symptoms of psychiatric disorders. In recent literature there is growing evidence that increased intestinal permeability with subsequent immune activation has a major role in the pathophysiology of various psychiatric disorders. Numerous parameters measured in this context seem to be aftermaths of those mechanisms, yet at the same time they may be contributing factors for immune mediated psychopathology. For example, immune activation related to gut-derived bacterial lipopolysaccharides (LPS) or various food antigens and exorphins were reported in major depression, schizophrenia, bipolar disorder, alcoholism and autism. In this review the authors will summarize the evidence and roles of such parameters and their assessment in major psychiatric disorders.
Collapse
Affiliation(s)
- Leszek Rudzki
- Department of Psychiatry, Medical University of BialystokBialystok, Poland
- Three Towns Resource Centre, Saltcoats, United Kingdom
| | - Agata Szulc
- Department of Psychiatry, Medical University of WarsawWarsaw, Poland
| |
Collapse
|
31
|
Proinflammatory Factors Mediate Paclitaxel-Induced Impairment of Learning and Memory. Mediators Inflamm 2018; 2018:3941840. [PMID: 29681766 PMCID: PMC5842689 DOI: 10.1155/2018/3941840] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/17/2017] [Accepted: 01/08/2018] [Indexed: 11/17/2022] Open
Abstract
The chemotherapeutic agent paclitaxel is widely used for cancer treatment. Paclitaxel treatment impairs learning and memory function, a side effect that reduces the quality of life of cancer survivors. However, the neural mechanisms underlying paclitaxel-induced impairment of learning and memory remain unclear. Paclitaxel treatment leads to proinflammatory factor release and neuronal apoptosis. Thus, we hypothesized that paclitaxel impairs learning and memory function through proinflammatory factor-induced neuronal apoptosis. Neuronal apoptosis was assessed by TUNEL assay in the hippocampus. Protein expression levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in the hippocampus tissue were analyzed by Western blot assay. Spatial learning and memory function were determined by using the Morris water maze (MWM) test. Paclitaxel treatment significantly increased the escape latencies and decreased the number of crossing in the MWM test. Furthermore, paclitaxel significantly increased the number of TUNEL-positive neurons in the hippocampus. Also, paclitaxel treatment increased the expression levels of TNF-α and IL-1β in the hippocampus tissue. In addition, the TNF-α synthesis inhibitor thalidomide significantly attenuated the number of paclitaxel-induced TUNEL-positive neurons in the hippocampus and restored the impaired spatial learning and memory function in paclitaxel-treated rats. These data suggest that TNF-α is critically involved in the paclitaxel-induced impairment of learning and memory function.
Collapse
|
32
|
Child neurodevelopmental outcomes following preterm and term birth: What can the placenta tell us? Placenta 2017; 57:79-86. [DOI: 10.1016/j.placenta.2017.06.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/17/2017] [Accepted: 06/12/2017] [Indexed: 11/21/2022]
|
33
|
Omrani H, Alipour MR, Farajdokht F, Ebrahimi H, Mesgari Abbasi M, Mohaddes G. Effects of Chronic Ghrelin Treatment on Hypoxia-Induced Brain Oxidative Stress and Inflammation in a Rat Normobaric Chronic Hypoxia Model. High Alt Med Biol 2017; 18:145-151. [DOI: 10.1089/ham.2016.0132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- Hasan Omrani
- Drug Applied Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alipour
- Neurosciences Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fereshteh Farajdokht
- Neurosciences Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Ebrahimi
- Drug Applied Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Gisou Mohaddes
- Drug Applied Research Center of Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Hu J, Ferchmin PA, Hemmerle AM, Seroogy KB, Eterovic VA, Hao J. 4R-Cembranoid Improves Outcomes after 6-Hydroxydopamine Challenge in Both In vitro and In vivo Models of Parkinson's Disease. Front Neurosci 2017; 11:272. [PMID: 28611572 PMCID: PMC5447022 DOI: 10.3389/fnins.2017.00272] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/28/2017] [Indexed: 11/13/2022] Open
Abstract
(1S, 2E, 4R, 6R,-7E, 11E)-2, 7, 11-cembratriene-4, 6-diol (4R) is one of the cembranoids found in tobacco leaves. Previous studies have found that 4R protected acute rat hippocampal slices against neurotoxicity induced by N-methyl-D-aspartate (NMDA) and against the toxic organophosphorus compounds paraoxon and diisopropylfluorophosphate (DFP). Furthermore, in vivo, 4R reduced the infarct size in a rodent ischemic stroke model and neurodegeneration caused by DFP. The present study expanded our previous study by focusing on the effect of 4R in Parkinson's disease (PD) and elucidating its underlying mechanisms using 6-hydroxydopamine (6-OHDA)-induced injury models. We found that 4R exhibited significant neuroprotective activity in the rat unilateral 6-OHDA-induced PD model in vivo. The therapeutic effect was evident both at morphological and behavioral levels. 4R (6 and 12 mg/kg) treatments significantly improved outcomes of 6-OHDA-induced PD in vivo as indicated by reducing forelimb asymmetry scores and corner test scores 4 weeks after injection of 6-OHDA (p < 0.05). The therapeutic effect of 4R was also reflected by decreased depletion of tyrosine hydroxylase (TH) in the striatum and substantia nigra (SN) on the side injected with 6-OHDA. TH expression was 70.3 and 62.8% of the contralateral side in striatum and SN, respectively, after 6 mg/kg 4R treatment; furthermore, it was 80.1 and 79.3% after treatment with 12 mg/kg of 4R. In the control group, it was 51.9 and 23.6% of the contralateral striatum and SN (p < 0.05). Moreover, 4R also protected differentiated neuro-2a cells from 6-OHDA-induced cytotoxicity in vitro. The activation of p-AKT and HAX-1, and inhibition of caspase-3 and endothelial inflammation, were involved in 4R-mediated protection against 6-OHDA-induced injury. In conclusion, the present study indicates that 4R shows a therapeutic effect in the rat 6-OHDA-induced PD model in vivo and in 6-OHDA-challenged neuro-2a cells in vitro.
Collapse
Affiliation(s)
- Jing Hu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of CincinnatiCincinnati, OH, United States
| | - P A Ferchmin
- Department of Neurosciences, School of Medicine, Universidad Central del CaribeBayamón, Puerto Rico
| | - Ann M Hemmerle
- Department of Neurology and Rehabilitation Medicine, University of CincinnatiCincinnati, OH, United States
| | - Kim B Seroogy
- Department of Neurology and Rehabilitation Medicine, University of CincinnatiCincinnati, OH, United States
| | - Vesna A Eterovic
- Department of Neurosciences, School of Medicine, Universidad Central del CaribeBayamón, Puerto Rico
| | - Jiukuan Hao
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of CincinnatiCincinnati, OH, United States
| |
Collapse
|
35
|
The Effect of Stereotactic Injections on Demyelination and Remyelination: a Study in the Cuprizone Model. J Mol Neurosci 2017; 61:479-488. [PMID: 28124770 DOI: 10.1007/s12031-017-0888-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/13/2017] [Indexed: 12/11/2022]
Abstract
Remyelination is the natural repair mechanism in demyelinating disorders of the central nervous system (CNS) such as multiple sclerosis. Several animal models have been used to study demyelination and remyelination. Among toxic animal models, oral administration of the toxin cuprizone leads to white and gray matter demyelination. In contrast, focal demyelination models include the stereotactic application of a toxin such as lysolecithin or ethidium bromide. The injection procedure generates a local disruption of the blood-brain barrier (BBB) and might thus trigger a local inflammatory reaction and consequently may influence demyelination and remyelination. In order to study such consequences, we applied stereotactic injections in the cuprizone model where demyelination and remyelination are mediated independent of this procedure. Immunohistochemistry was performed to detect the presence of lymphocytes and activated glial cells in the injection area. Blood protein stainings were used to assess the integrity of the BBB and myelin staining to evaluate demyelination and remyelination processes. Stereotactic injection led to a local disruption of the BBB as shown by local extravasation of blood proteins. Along the injection canal, T and B lymphocytes could be detected and there was a tendency of a higher microgliosis and astrocytosis. However, these changes did not influence demyelination and remyelination processes at the site of injection, in the corpus callosum, or in the cerebral cortex. Our results suggest that a local stereotactic injection has no major impact on CNS demyelination and remyelination.
Collapse
|
36
|
Goksu E, Dogan O, Ulker P, Tanrıover G, Konuk E, Dilmac S, Kirac E, Demır N, Aslan M. Pentoxifylline Alleviates Early Brain Injury in a Rat Model of Subarachnoid Hemorrhage. Acta Neurochir (Wien) 2016; 158:1721-30. [PMID: 27311763 DOI: 10.1007/s00701-016-2866-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/02/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) is a severe cerebrovascular disease frequently caused by ruptured aneurysms. Early brain injury (EBI) is the primary cause of morbidity and mortality in patients diagnosed with SAH and is associated with increased intracranial pressure, decreased cerebral blood flow and cerebral ischemia. Pentoxifylline (PTX) is a methylxanthine derivative clinically proven to improve perfusion in the peripheral microcirculation and has been shown to have neuroprotective effects in brain trauma and global cerebral ischemia in experimental animal models. This study aimed to determine the effect of PTX in experimental SAH, which has not been investigated yet. METHODS An experimental SAH model was induced in male Wistar rats by autologous blood injection into the prechiasmatic cistern, and PTX was injected intraperitoneally immediately after SAH. The effects of PTX were evaluated 24 h after SAH via assessing the cerebral ultrastructure via transmission electron microscopy (TEM). Brain edema, blood-brain barrier (BBB) permeability, red blood cell deformability, tumor necrosis factor-alpha (TNF-alpha), nitrite-nitrate levels and apoptotic neuron death were also determined 24 h after SAH. The BBB permeability was measured by Evans blue (EB) extravasation, erythrocyte deformability was determined by filtration technique, and TNF-alpha and reactive nitrogen metobolites were analyzed in brain tissue by ELISA and spectral analysis, respectively. Apoptotic neurons were determined in brain sections by cleaved caspase-3 immunohistochemical analysis, and expression intensity was quantified using image J software. RESULTS Cerebral ultrastructure in SAH group animals revealed intense perivascular edema and distortion in the astrocyte foot processes. PTX treatment attenuated structural deterioration due to SAH. Brain water content, BBB permeability, TNF-alpha, nitrite-nitrate levels and apoptotic neuronal death were significantly increased 24 h after SAH and were significantly alleviated by PTX treatment. There was no significant change in red cell deformability after SAH. CONCLUSIONS Our results show that PTX reduces brain edema, BBB permeability, TNF-alpha expression, reactive nitrogen metobolites and apopotosis in experimental SAH. Based on our findings we suggest that PTX exerts neuroprotection against SAH-induced EBI, which might be associated with the inhibition of inflammation and apoptotic neuronal cell death.
Collapse
Affiliation(s)
- Ethem Goksu
- Department of Neurosurgery, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Ozgur Dogan
- Division of Neurosurgery, Denizli State Hospital, Denizli, 20125, Turkey
| | - Pınar Ulker
- Department of Physiology, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Gamze Tanrıover
- Department of Histology, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Esma Konuk
- Department of Histology, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Sayra Dilmac
- Department of Histology, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Ebru Kirac
- Medical Biochemistry, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Necdet Demır
- Department of Histology, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey
| | - Mutay Aslan
- Medical Biochemistry, Akdeniz University Faculty of Medicine, Antalya, 07070, Turkey.
| |
Collapse
|
37
|
Nitsos I, Rees SM, Duncan J, Kramer BW, Harding R, Newnham JP, Moss TJM. Chronic Exposure to Intra-Amniotic Lipopolysaccharide Affects the Ovine Fetal Brain. ACTA ACUST UNITED AC 2016; 13:239-47. [PMID: 16697939 DOI: 10.1016/j.jsgi.2006.02.011] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2005] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Fetal brain injury is associated with chorioamnionitis, which is often present without signs of overt infection or fetal compromise. We aimed to determine if prolonged exposure to intrauterine inflammation caused by intra-amniotic infusion of lipopolysaccharide (LPS) would affect the fetal brain. METHODS At 80 days of pregnancy ewes bearing singletons had osmotic pumps implanted intra-amniotically to infuse Escherichia coli LPS (055:B5; n = 8) or saline (n = 7) for 28 days. At delivery (110 days), umbilical arterial blood and chorioamnion were assessed for inflammation; cytokine concentrations (interleukin [IL]-6 and IL-8) in amniotic fluid and fetal and maternal plasma were measured. The fetal cerebral hemispheres were examined for gross anatomical changes and the number of activated microglia/macrophages, astrocytes, and oligodendrocytes estimated after immunohistochemical staining. RESULTS Intra-amniotic administration of LPS caused chorioamnionitis, fetal leucocytosis, and a moderate to extensive infiltration of activated microglia/macrophages in the subcortical white matter in six of eight fetuses; the remaining two fetuses were less affected. Within these focal regions of damage there was an attenuation of astrocytic processes, axonal injury, and a reduction in the number of 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) immunoreactive oligodendrocytes in areas of extensive focal damage. In control fetuses there was mild (3/7) or no infiltration of activated microglia/macrophages in the subcortical white matter. Overall the infiltration of activated microglia/macrophages in the white matter was significantly greater in LPS-exposed fetuses compared to controls. In regions devoid of injury, the number of oligodendrocytes and astrocytes was not different between groups, nor was there a difference in the volume of cerebral white matter or density of blood vessels within the white matter. Amniotic fluid IL-6 and IL-8, and maternal plasma IL-8 concentrations were significantly increased by LPS infusion. CONCLUSIONS An increase in inflammatory cells and axonal disruption in the subcortical white matter of the fetal brain can accompany chorioamnionitis induced by intra-amniotic administration of LPS, but cystic lesions do not occur. Thus, the effect on the fetal brain is milder than that reported from animal models of acute fetal/intrauterine infection.
Collapse
Affiliation(s)
- Ilias Nitsos
- School of Women's and Infants' Health, The University of Western Australia, Crawley, Western Australia, Australia.
| | | | | | | | | | | | | |
Collapse
|
38
|
Hu J, Al-Waili D, Hassan A, Fan GC, Xin M, Hao J. Inhibition of cerebral vascular inflammation by brain endothelium-targeted oligodeoxynucleotide complex. Neuroscience 2016; 329:30-42. [PMID: 27132231 DOI: 10.1016/j.neuroscience.2016.04.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 11/24/2022]
Abstract
The present study generated a novel DNA complex to specifically target endothelial NF-κB to inhibit cerebral vascular inflammation. This DNA complex (GS24-NFκB) contains a DNA decoy which inhibits NF-κB activity, and a DNA aptamer (GS-24), a ligand of transferrin receptor (TfR), which allows for targeted delivery of the DNA decoy into cells. The results indicate that GS24-NFκB was successfully delivered into a murine brain-derived endothelial cell line, bEND5, and inhibited inflammatory responses induced by tumor necrosis factor α (TNF-α) or oxygen-glucose deprivation/re-oxygenation (OGD/R) via down-regulation of the nuclear NF-κB subunit, p65, as well as its downstream inflammatory cytokines, inter-cellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM-1). The inhibitory effect of the GS24-NFκB was demonstrated by a significant reduction in TNF-α or OGD/R induced monocyte adhesion to the bEND5 cells after GS24-NFκB treatment. Intravenous (i.v.) injection of GS24-'NFκB (15mg/kg) was able to inhibit the levels of phoseph-p65 and VCAM-1 in brain endothelial cells in a mouse lipopolysaccharide (LPS)-induced inflammatory model in vivo. In conclusion, our approach using DNA nanotechnology for DNA decoy delivery could potentially be utilized for inhibition of inflammation in ischemic stroke and other neuro-inflammatory diseases affecting cerebral vasculature.
Collapse
Affiliation(s)
- Jing Hu
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Daniah Al-Waili
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Aishlin Hassan
- Department of Pediatrics, University of Cincinnati & Cincinnati Children's Hospital Medical Center, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Mei Xin
- Department of Pediatrics, University of Cincinnati & Cincinnati Children's Hospital Medical Center, USA
| | - Jiukuan Hao
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
39
|
|
40
|
Abdullah Z, Rakkar K, Bath PMW, Bayraktutan U. Inhibition of TNF-α protects in vitro brain barrier from ischaemic damage. Mol Cell Neurosci 2015; 69:65-79. [PMID: 26546149 DOI: 10.1016/j.mcn.2015.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/11/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
Cerebral ischaemia, associated with neuroinflammation and oxidative stress, is known to perturb blood-brain barrier (BBB) integrity and promote brain oedema formation. Using an in vitro model of human BBB composed of brain microvascular endothelial cells and astrocytes, this study examined whether suppression of TNF-α, a potent pro-inflammatory cytokine, might attenuate ischaemia-mediated cerebral barrier damage. Radical decreases in transendothelial electrical resistance and concomitant increases in paracellular flux across co-cultures exposed to increasing periods of oxygen-glucose deprivation alone (0.5-20 h) or followed by 20 h of reperfusion (OGD ± R) confirmed the deleterious effects of ischaemic injury on cerebral barrier integrity and function which concurred with reductions in tight junction protein (claudin-5 and occludin) expressions. OGD ± R elevated TNF-α secretion, NADPH oxidase activity, O2(-) production, actin stress fibre formation, MMP-2/9 activities and apoptosis in both endothelial cells and astrocytes. Increases in MMP-2 activity were confined to its extracellular isoform and treatments with OGD+R in astrocytes where MMP-9 could not be detected at all. Co-exposure of individual cell lines or co-cultures to an anti-TNF-α antibody dramatically diminished the extent of OGD ± R-evoked oxidative stress, morphological changes, apoptosis, MMP-2/9 activities while improving the barrier function through upregulation of tight junction protein expressions. In conclusion, vitiation of the exaggerated release of TNF-α may be an important therapeutic strategy in preserving cerebral integrity and function during and following a cerebral ischaemic attack.
Collapse
Affiliation(s)
- Zuraidah Abdullah
- Stroke, Division of Clinical Neuroscience, University of Nottingham, UK
| | - Kamini Rakkar
- Stroke, Division of Clinical Neuroscience, University of Nottingham, UK
| | - Philip M W Bath
- Stroke, Division of Clinical Neuroscience, University of Nottingham, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, University of Nottingham, UK.
| |
Collapse
|
41
|
Liu Z, Chopp M. Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Prog Neurobiol 2015; 144:103-20. [PMID: 26455456 DOI: 10.1016/j.pneurobio.2015.09.008] [Citation(s) in RCA: 432] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 08/06/2015] [Accepted: 09/05/2015] [Indexed: 01/04/2023]
Abstract
Astrocytes are the most abundant cell type within the central nervous system. They play essential roles in maintaining normal brain function, as they are a critical structural and functional part of the tripartite synapses and the neurovascular unit, and communicate with neurons, oligodendrocytes and endothelial cells. After an ischemic stroke, astrocytes perform multiple functions both detrimental and beneficial, for neuronal survival during the acute phase. Aspects of the astrocytic inflammatory response to stroke may aggravate the ischemic lesion, but astrocytes also provide benefit for neuroprotection, by limiting lesion extension via anti-excitotoxicity effects and releasing neurotrophins. Similarly, during the late recovery phase after stroke, the glial scar may obstruct axonal regeneration and subsequently reduce the functional outcome; however, astrocytes also contribute to angiogenesis, neurogenesis, synaptogenesis, and axonal remodeling, and thereby promote neurological recovery. Thus, the pivotal involvement of astrocytes in normal brain function and responses to an ischemic lesion designates them as excellent therapeutic targets to improve functional outcome following stroke. In this review, we will focus on functions of astrocytes and astrocyte-mediated events during stroke and recovery. We will provide an overview of approaches on how to reduce the detrimental effects and amplify the beneficial effects of astrocytes on neuroprotection and on neurorestoration post stroke, which may lead to novel and clinically relevant therapies for stroke.
Collapse
Affiliation(s)
- Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA.
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA; Department of Physics, Oakland University, Rochester, MI, USA
| |
Collapse
|
42
|
Xiong X, Sun Y, Sattiraju A, Jung Y, Mintz A, Hayasaka S, Li KCP. Remote spatiotemporally controlled and biologically selective permeabilization of blood-brain barrier. J Control Release 2015; 217:113-20. [PMID: 26334482 DOI: 10.1016/j.jconrel.2015.08.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 12/13/2022]
Abstract
The blood-brain barrier (BBB), comprised of brain endothelial cells with tight junctions (TJ) between them, regulates the extravasation of molecules and cells into and out of the central nervous system (CNS). Overcoming the difficulty of delivering therapeutic agents to specific regions of the brain presents a major challenge to treatment of a broad range of brain disorders. Current strategies for BBB opening are invasive, not specific, and lack precise control over the site and timing of BBB opening, which may limit their clinical translation. In the present report, we describe a novel approach based on a combination of stem cell delivery, heat-inducible gene expression and mild heating with high-intensity focused ultrasound (HIFU) under MRI guidance to remotely permeabilize BBB. The permeabilization of the BBB will be controlled with, and limited to where selected pro-inflammatory factors will be secreted secondary to HIFU activation, which is in the vicinity of the engineered stem cells and consequently both the primary and secondary disease foci. This therapeutic platform thus represents a non-invasive way for BBB opening with unprecedented spatiotemporal precision, and if properly and specifically modified, can be clinically translated to facilitate delivery of different diagnostic and therapeutic agents which can have great impact in treatment of various disease processes in the central nervous system.
Collapse
Affiliation(s)
- Xiaobing Xiong
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - Yao Sun
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - Anirudh Sattiraju
- Comprehensive Cancer Center, Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - Youngkyoo Jung
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA; Comprehensive Cancer Center, Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem 27157, USA; Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - Akiva Mintz
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA; Comprehensive Cancer Center, Brain Tumor Center of Excellence, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - Satoru Hayasaka
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA; Department of Biostatistics Sciences, Wake Forest School of Medicine, Winston-Salem 27157, USA
| | - King C P Li
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem 27157, USA.
| |
Collapse
|
43
|
Abstract
Astrocytes were historically classified as supporting cells; however, it is becoming increasingly clear that they actively contribute to neuronal functioning under normal and pathological conditions. As interest in the contribution of neuroinflammation to Alzheimer's disease (AD) progression has grown, manipulating glial cells has become an attractive target for future therapies. Astrocytes have largely been under-represented in studies that assess the role of glia in these processes, despite substantial evidence of astrogliosis in AD. The actual role of astrocytes in AD remains elusive, as they seem to adopt different functions dependent on disease progression and the extent of accompanying parenchymal inflammation. Astrocytes may contribute to the clearance of amyloid β-peptide (Aβ) and restrict the spread of inflammation in the brain. Conversely, they may contribute to neurodegeneration in AD by releasing neurotoxins and neglecting crucial metabolic roles. The present review summarizes current evidence on the multi-faceted functions of astrocytes in AD, highlighting the significant scope available for future therapeutic targets.
Collapse
|
44
|
Ji R, Meng L, Li Q, Lu Q. TAM receptor deficiency affects adult hippocampal neurogenesis. Metab Brain Dis 2015; 30:633-44. [PMID: 25487541 PMCID: PMC4414696 DOI: 10.1007/s11011-014-9636-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
The Tyro3, Axl and Mertk (TAM) subfamily of receptor protein tyrosine kinases functions in cell growth, differentiation, survival, and most recently found, in the regulation of immune responses and phagocytosis. All three receptors and their ligands, Gas6 (growth arrest-specific gene 6) and protein S, are expressed in the central nervous system (CNS). TAM receptors play pivotal roles in adult hippocampal neurogenesis. Loss of these receptors causes a comprised neurogenesis in the dentate gyrus of adult hippocampus. TAM receptors have a negative regulatory effect on microglia and peripheral antigen-presenting cells, and play a critical role in preventing overproduction of pro-inflammatory cytokines detrimental to the proliferation, differentiation, and survival of adult neuronal stem cells (NSCs). Besides, these receptors also play an intrinsic trophic function in supporting NSC survival, proliferation, and differentiation into immature neurons. All these events collectively ensure a sustained neurogenesis in adult hippocampus.
Collapse
Affiliation(s)
- Rui Ji
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Lingbin Meng
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Qiutang Li
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Qingxian Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
45
|
TAK-242, an antagonist for Toll-like receptor 4, protects against acute cerebral ischemia/reperfusion injury in mice. J Cereb Blood Flow Metab 2015; 35:536-42. [PMID: 25586141 PMCID: PMC4420883 DOI: 10.1038/jcbfm.2014.240] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/29/2014] [Accepted: 12/03/2014] [Indexed: 02/07/2023]
Abstract
Toll-like receptor 4 (TLR4) contributes to cerebral ischemia/reperfusion (I/R) injury and is a potential target for the treatment of ischemic stroke. This experiment is to evaluate the effect of an exogenous TLR4 antagonist, TAK-242, against acute cerebral I/R injury. A mouse model of cerebral I/R was induced by transient middle cerebral artery occlusion. TAK-242 (3 mg/kg body weight) was injected intraperitoneally 1 hour after ischemia. Our results showed that the concentration of TAK-242 in plasma increased to 52.0 ng/mL 3 hours after injection, was maintained at 54.1 ng/mL 8 hours after injection, and decreased to 22.6 ng/mL 24 hours after injection. The concentration of TAK-242 in brain tissue increased to 26.1 ng/mL in ischemic hemisphere and 14.2 ng/mL in nonischemic hemisphere 3 hours after injection, and was maintained at the similar levels 24 hours after injection. We found that TAK-242 significantly reduced cerebral infarction compared with vehicle control, improved neurologic function, inhibited the phosphorylation of downstream protein kinases in TLR4 signaling pathway, and downregulated the expression of inflammatory cytokines. We conclude that TAK-242 is able to cross blood-brain barrier, blocks TLR4 signaling, mediates the expression of inflammatory cytokines, and protects the brain from acute damage induced by I/R.
Collapse
|
46
|
Northrop NA, Yamamoto BK. Methamphetamine effects on blood-brain barrier structure and function. Front Neurosci 2015; 9:69. [PMID: 25788874 PMCID: PMC4349189 DOI: 10.3389/fnins.2015.00069] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/17/2015] [Indexed: 01/28/2023] Open
Abstract
Methamphetamine (Meth) is a widely abuse psychostimulant. Traditionally, studies have focused on the neurotoxic effects of Meth on monoaminergic neurotransmitter terminals. Recently, both in vitro and in vivo studies have investigated the effects of Meth on the BBB and found that Meth produces a decrease in BBB structural proteins and an increase in BBB permeability to various molecules. Moreover, preclinical studies are validated by clinical studies in which human Meth users have increased concentrations of toxins in the brain. Therefore, this review will focus on the structural and functional disruption of the BBB caused by Meth and the mechanisms that contribute to Meth-induced BBB disruption. The review will reveal that the mechanisms by which Meth damages dopamine and serotonin terminals are similar to the mechanisms by which the blood-brain barrier (BBB) is damaged. Furthermore, this review will cover the factors that are known to potentiate the effects of Meth (McCann et al., 1998) on the BBB, such as stress and HIV, both of which are co-morbid conditions associated with Meth abuse. Overall, the goal of this review is to demonstrate that the scope of damage produced by Meth goes beyond damage to monoaminergic neurotransmitter systems to include BBB disruption as well as provide a rationale for investigating therapeutics to treat Meth-induced BBB disruption. Since a breach of the BBB can have a multitude of consequences, therapies directed toward the treatment of BBB disruption may help to ameliorate the long-term neurodegeneration and cognitive deficits produced by Meth and possibly even Meth addiction.
Collapse
Affiliation(s)
- Nicole A Northrop
- Department of Neurosciences, University of Toledo College of Medicine Toledo, OH, USA
| | - Bryan K Yamamoto
- Department of Neurosciences, University of Toledo College of Medicine Toledo, OH, USA
| |
Collapse
|
47
|
Xing F, Sharma S, Liu Y, Mo YY, Wu K, Zhang YY, Pochampally R, Martinez LA, Lo HW, Watabe K. miR-509 suppresses brain metastasis of breast cancer cells by modulating RhoC and TNF-α. Oncogene 2015; 34:4890-900. [PMID: 25659578 PMCID: PMC4530094 DOI: 10.1038/onc.2014.412] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 11/04/2014] [Accepted: 11/08/2014] [Indexed: 12/18/2022]
Abstract
The median survival time of breast cancer patients with brain metastasis is less than 6 months, and even a small metastatic lesion often causes severe neurological disabilities. Because of the location of metastatic lesions, a surgical approach is limited and most chemotherapeutic drugs are ineffective due to the blood brain barrier (BBB). Despite this clinical importance, the molecular basis of the brain metastasis is poorly understood. In this study, we have isolated RNA from samples obtained from primary breast tumors and also from brain metastatic lesions followed by microRNA profiling analysis. Our results revealed that the miR-509 is highly expressed in the primary tumors, while the expression of this microRNA is significantly decreased in the brain metastatic lesions. MicroRNA target prediction and the analysis of cytokine array for the cells ectopically expressed with miR-509 demonstrated that this microRNA was capable of modulating two genes essential for brain invasion, RhoC and TNFα that affect the invasion of cancer cells and permeability of BBB, respectively. Importantly, high levels of TNFα and RhoC-induced MMP9 were significantly correlated with brain metastasis-free survival of breast cancer patients. Furthermore, the results of our in vivo experiments indicate that miR-509 significantly suppressed the ability of cancer cells to metastasize to the brain. These findings suggest that miR-509 plays a critical role in brain metastasis of breast cancer by modulating the RhoC-TNFα network and that this miR-509 axis may represent a potential therapeutic target or serve as a prognostic tool for brain metastasis.
Collapse
Affiliation(s)
- F Xing
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - S Sharma
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Y Liu
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Y-Y Mo
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - K Wu
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Y-Y Zhang
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - R Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - L A Martinez
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - H-W Lo
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - K Watabe
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
48
|
Chen X, Sadowska GB, Zhang J, Kim JE, Cummings EE, Bodge CA, Lim YP, Makeyev O, Besio WG, Gaitanis J, Threlkeld SW, Banks WA, Stonestreet BS. Neutralizing anti-interleukin-1β antibodies modulate fetal blood-brain barrier function after ischemia. Neurobiol Dis 2015; 73:118-29. [PMID: 25258170 PMCID: PMC4252260 DOI: 10.1016/j.nbd.2014.09.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 08/15/2014] [Accepted: 09/12/2014] [Indexed: 02/08/2023] Open
Abstract
We have previously shown that increases in blood-brain barrier permeability represent an important component of ischemia-reperfusion related brain injury in the fetus. Pro-inflammatory cytokines could contribute to these abnormalities in blood-brain barrier function. We have generated pharmacological quantities of mouse anti-ovine interleukin-1β monoclonal antibody and shown that this antibody has very high sensitivity and specificity for interleukin-1β protein. This antibody also neutralizes the effects of interleukin-1β protein in vitro. In the current study, we hypothesized that the neutralizing anti-interleukin-1β monoclonal antibody attenuates ischemia-reperfusion related fetal blood-brain barrier dysfunction. Instrumented ovine fetuses at 127 days of gestation were studied after 30 min of carotid occlusion and 24h of reperfusion. Groups were sham operated placebo-control- (n=5), ischemia-placebo- (n=6), ischemia-anti-IL-1β antibody- (n=7), and sham-control antibody- (n=2) treated animals. Systemic infusions of placebo (0.154M NaCl) or anti-interleukin-1β monoclonal antibody (5.1±0.6 mg/kg) were given intravenously to the same sham or ischemic group of fetuses at 15 min and 4h after ischemia. Concentrations of interleukin-1β protein and anti-interleukin-1β monoclonal antibody were measured by ELISA in fetal plasma, cerebrospinal fluid, and parietal cerebral cortex. Blood-brain barrier permeability was quantified using the blood-to-brain transfer constant (Ki) with α-aminoisobutyric acid in multiple brain regions. Interleukin-1β protein was also measured in parietal cerebral cortices and tight junction proteins in multiple brain regions by Western immunoblot. Cerebral cortical interleukin-1β protein increased (P<0.001) after ischemia-reperfusion. After anti-interleukin-1β monoclonal antibody infusions, plasma anti-interleukin-1β monoclonal antibody was elevated (P<0.001), brain anti-interleukin-1β monoclonal antibody levels were higher (P<0.03), and interleukin-1β protein concentrations (P<0.03) and protein expressions (P<0.001) were lower in the monoclonal antibody-treated group than in placebo-treated-ischemia-reperfusion group. Monoclonal antibody infusions attenuated ischemia-reperfusion-related increases in Ki across the brain regions (P<0.04), and Ki showed an inverse linear correlation (r= -0.65, P<0.02) with anti-interleukin-1β monoclonal antibody concentrations in the parietal cortex, but had little effect on tight junction protein expression. We conclude that systemic anti-interleukin-1β monoclonal antibody infusions after ischemia result in brain anti-interleukin-1β antibody uptake, and attenuate ischemia-reperfusion-related interleukin-1β protein up-regulation and increases in blood-brain barrier permeability across brain regions in the fetus. The pro-inflammatory cytokine, interleukin-1β, contributes to impaired blood-brain barrier function after ischemia in the fetus.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Grazyna B Sadowska
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Jiyong Zhang
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Jeong-Eun Kim
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Erin E Cummings
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Courtney A Bodge
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, USA
| | - Oleksandr Makeyev
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, RI, USA
| | - Walter G Besio
- Department of Electrical, Computer, and Biomedical Engineering, University of Rhode Island, Kingston, RI, USA
| | - John Gaitanis
- Department of Neurology, The Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Steven W Threlkeld
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA
| | - William A Banks
- Geriatric Research Educational, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, The Alpert Medical School of Brown University, Women & Infants Hospital of Rhode Island, Providence, RI, USA.
| |
Collapse
|
49
|
Doll D, Barr TL, Simpkins JW. Cytokines: their role in stroke and potential use as biomarkers and therapeutic targets. Aging Dis 2014; 5:294-306. [PMID: 25276489 DOI: 10.14336/ad.2014.0500294] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 05/16/2014] [Accepted: 05/27/2014] [Indexed: 12/30/2022] Open
Abstract
Inflammatory mechanisms both in the periphery and in the CNS are important in the pathophysiologic processes occurring after the onset of ischemic stroke (IS). Cytokines are key players in the inflammatory mechanism and contribute to the progression of ischemic damage. This literature review focuses on the effects of inflammation on ischemic stroke, and the role pro-inflammatory and anti-inflammatory cytokines play on deleterious or beneficial stroke outcome. The discovery of biomarkers and novel therapeutics for stroke has been the focus of extensive research recently; thus, understanding the roles of pro-inflammatory and anti-inflammatory cytokines that are up-regulated during stroke will help us further understand how inflammation contributes to the progression of ischemic damage and provide potential targets for novel therapeutics and biomarkers for diagnosis and prognosis of stroke.
Collapse
Affiliation(s)
| | - Taura L Barr
- School of Nursing, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA ; Center for Basic and Translational Stroke Research, West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| |
Collapse
|
50
|
Pires PW, Girgla SS, Moreno G, McClain JL, Dorrance AM. Tumor necrosis factor-α inhibition attenuates middle cerebral artery remodeling but increases cerebral ischemic damage in hypertensive rats. Am J Physiol Heart Circ Physiol 2014; 307:H658-69. [PMID: 25015967 DOI: 10.1152/ajpheart.00018.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypertension causes vascular inflammation evidenced by an increase in perivascular macrophages and proinflammatory cytokines in the arterial wall. Perivascular macrophage depletion reduced tumor necrosis factor (TNF)-α expression in cerebral arteries of hypertensive rats and attenuated inward remodeling, suggesting that TNF-α might play a role in the remodeling process. We hypothesized that TNF-α inhibition would improve middle cerebral artery (MCA) structure and reduce damage after cerebral ischemia in hypertensive rats. Six-week-old male stroke-prone spontaneously hypertensive rats (SHRSP) were treated with the TNF-α inhibitor etanercept (ETN; 1.25 mg·kg(-1)·day(-1) ip daily) or PBS (equivolume) for 6 wk. The myogenic tone generation, postischemic dilation, and passive structure of MCAs were assessed by pressure myography. Cerebral ischemia was induced by MCA occlusion (MCAO). Myogenic tone was unchanged, but MCAs from SHRSP + ETN had larger passive lumen diameter and reduced wall thickness and wall-to-lumen ratio. Cerebral infarct size was increased in SHRSP + ETN after transient MCAO, despite an improvement in dilation of nonischemic MCA. The increase in infarct size was linked to a reduction in the number of microglia in the infarct core and upregulation of markers of classical macrophage/microglia polarization. There was no difference in infarct size after permanent MCAO or when untreated SHRSP subjected to transient MCAO were given ETN at reperfusion. Our data suggests that TNF-α inhibition attenuates hypertensive MCA remodeling but exacerbates cerebral damage following ischemia/reperfusion injury likely due to inhibition of the innate immune response of the brain.
Collapse
Affiliation(s)
- Paulo W Pires
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Saavia S Girgla
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Guillermo Moreno
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Jonathon L McClain
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Anne M Dorrance
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|