1
|
Graves SI, Meyer CF, Jeganathan KB, Baker DJ. p16-expressing microglia and endothelial cells promote tauopathy and neurovascular abnormalities in PS19 mice. Neuron 2025:S0896-6273(25)00303-4. [PMID: 40381614 DOI: 10.1016/j.neuron.2025.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/15/2024] [Accepted: 04/21/2025] [Indexed: 05/20/2025]
Abstract
Cellular senescence is characterized by irreversible cell-cycle exit, a pro-inflammatory secretory phenotype, macromolecular damage, and deregulated metabolism. Senescent cells are highly associated with age-related diseases. We previously demonstrated that targeted elimination of senescent cells prevents neurodegenerative disease in tau (MAPTP301S;PS19) mutant mice. Here, we show that genetic ablation of the senescence mediator p16Ink4a is sufficient to attenuate senescence signatures in PS19 mice. Disease phenotypes-including neuroinflammation, phosphorylated tau, neurodegeneration, and cognitive impairment-were blunted in the absence of p16Ink4a. Additionally, we found that PS19 mouse brains display p16Ink4-dependent neurovascular alterations such as vessel dilation, increased vessel density, deregulated endothelial cell extracellular matrix, and astrocytic endfoot depolarization. Finally, we show that p16Ink4a deletion in endothelial cells and microglia alone attenuates many of the same phenotypes. Altogether, these results indicate that neurodegenerative disease in PS19 mice is driven, at least in part, by p16Ink4a-expressing endothelial cells and microglia.
Collapse
Affiliation(s)
- Sara I Graves
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA
| | - Charlton F Meyer
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA
| | - Karthik B Jeganathan
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA; Paul F. Glenn Center for Biology of Aging Research, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA; Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 1st ST., Rochester, MN 55905, USA.
| |
Collapse
|
2
|
Zhang R, Liu Z, Cai Q, Xie Y, Liu Y, Peng L. Association between albumin-to-alkaline phosphatase ratio and a 3-month unfavorable outcome in patients with acute ischemic stroke. Front Nutr 2025; 12:1537954. [PMID: 40248032 PMCID: PMC12003142 DOI: 10.3389/fnut.2025.1537954] [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: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Background The albumin-to-alkaline phosphatase ratio (AAPR) is a predictor of several disease outcomes. However, there is no study about AAPR and acute ischemic stroke outcomes. This study aims to investigate the relationship between AAPR and a 3-month unfavorable outcome in patients with acute ischemic stroke. Methods This prospective cohort study included 2084 patients with acute ischemic stroke in South Korea. After applying strict exclusion criteria, 1,886 patients were included in our analysis and divided into three groups based on AAPR tertiles. An unfavorable outcome was defined as a 3-month modified Rankin scale (mRS) score > 2. Logistic regression analysis and smooth curve fitting analysis were applied to investigate the relationship between AAPR and unfavorable outcomes. Subgroup analysis was also performed to assess whether influencing factors changed the association between AAPR and unfavorable outcomes. Results After adjusting for potential confounders, multivariate analysis showed that AAPR was significantly associated with a 3-month unfavorable outcome (OR 0.18, 95% CI 0.09-0.35, p < 0.001). The smooth curve fitting analysis showed a nonlinear relationship between AAPR and a 3-month unfavorable outcome. The infection point was 0.588 according to the recursive method, and the threshold analysis showed when AAPR was ≤0.588, with the per unit increase of AAPR, the 3-month unfavorable outcome risk decreased by 96% (OR 0.04, 95% CI 0.01-0.2, p < 0.001). However, when AAPR was >0.588, there was no negative correlation between AAPR and a 3-month unfavorable outcome (OR 0.33, 95% CI 0.08-1.3, p = 0.112). Conclusion This study is the first to suggest a non-linear relationship between AAPR and a 3-month unfavorable outcome of acute ischemic stroke. AAPR was negatively correlated with a 3-month unfavorable outcome when AAPR was <0.588.
Collapse
Affiliation(s)
- Renwei Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenxing Liu
- Department of Neurology, Yiling Hospital of Yichang, Yichang, China
| | - Qi Cai
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu Xie
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Liu
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Li Peng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Emrani S, Koutures A, Tripodis Y, Uretsky M, Abdolmohammadi B, Nowinski C, Daneshvar DH, Dwyer B, Katz DI, Goldstein LE, Cantu RC, Martin BM, Palmisano JN, Dams-O'Connor K, Crary JF, Stern RA, Mez J, Alvarez VE, Huber BR, McKee AC, Stein TD, Alosco ML. Characterizing white matter and vascular pathologies in brain donors exposed to repetitive head impacts. Acta Neuropathol 2025; 149:24. [PMID: 40047953 PMCID: PMC11885321 DOI: 10.1007/s00401-025-02860-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/29/2025] [Accepted: 02/14/2025] [Indexed: 03/09/2025]
Abstract
Chronic traumatic encephalopathy (CTE) is a progressive brain disease linked to repetitive head impacts (RHI), often incurred from contact sports, and can lead to dementia. Here, we investigated the association between RHI and white matter/vascular neuropathologies and their relative contribution to dementia status in deceased men 50 + years old with and without exposure to RHI from various types of contact and collision sports. Our sample included two RHI groups from the UNITE brain bank: (1) American Football players (RHI-AF, n = 79), and (2) non-AF contact and collision sport athletes (e.g., boxing, rugby; RHI-CCS, n = 49). Controls included similarly aged (± 5 years) male brain donors without RHI. A modified ischemic injury scale (mIIS) served as a global measure of white matter and vascular neuropathologies, encompassing nine subcomponents. Dementia was determined through diagnostic consensus conference based on interviews with families. Using linear regression models controlling for age at death, mIIS was different in RHI-AF versus non-RHI only (p = 0.036). Subsequent logistic regression of each mIIS subcomponent, controlling for age at death, demonstrated that worse white matter rarefaction (RHI-AF; Beta = 1.42, [95% CI 2.03-8.43]; RHI-CCS; Beta = 1.93, [95% CI 2.35-20.17]) and hippocampal sclerosis (RHI-AF; Beta = 2.01, [95% CI 2.69-20.81]; RHI-CCS; Beta = 2.19, [95% CI 2.49-32.10]) was more common in RHI groups from their controls. Further, logistic regressions found that higher global mIIS correlated with increased odds of dementia in only the RHI-AF group (p = 0.02), driven by white matter rarefaction (β = 0.94, [95% CI 1.66-4.00]) and hippocampal sclerosis (β = 1.08, [95% CI 1.35-6.42]). There were similar findings in RHI-CCS group for odds of dementia (p = 0.048), including white matter rarefaction (β = 0.68, [95% CI 1.22-3.21], p = 0.05). Overall, these results demonstrate that white matter rarefaction and hippocampal sclerosis are linked to RHI exposure across all types of contact sports. Further, these pathologies contribute to dementia independent of p-tau pathology in American football players.
Collapse
Affiliation(s)
- Sheina Emrani
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Yorghos Tripodis
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Madeline Uretsky
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
| | - Bobak Abdolmohammadi
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
| | | | - Daniel H Daneshvar
- Department of Rehabilitation Medicine, Harvard Medical School, Boston, MA, USA
| | - Brigid Dwyer
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Douglas I Katz
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Braintree Rehabilitation Hospital, Braintree, MA, USA
| | - Lee E Goldstein
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Radiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Departments of Biomedical, Electrical & Computer Engineering, Boston University College of Engineering, Boston, MA, USA
| | - Robert C Cantu
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Concussion Legacy Foundation, Boston, MA, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Emerson Hospital, Concord, MA, USA
| | - Brett M Martin
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Joseph N Palmisano
- Biostatistics and Epidemiology Data Analytics Center, Boston University School of Public Health, Boston, MA, USA
| | - Kristen Dams-O'Connor
- Department of Rehabilitation and Human Performance, Brain Injury Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Crary
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert A Stern
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurosurgery, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Anatomy & Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jesse Mez
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
| | - Victor E Alvarez
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - Bertrand R Huber
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - Ann C McKee
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - Thor D Stein
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA
- Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- U.S. Department of Veteran Affairs, VA Boston Healthcare System, Boston, MA, USA
- Department of Veterans Affairs Medical Center, Bedford, MA, USA
| | - Michael L Alosco
- Alzheimer's Disease Research Center and Chronic Traumatic Encephalopathy Center, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA.
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, 72 E Concord St, Boston, MA, 02118, USA.
| |
Collapse
|
4
|
Pan X, Tan Z, Meng F, Zhang L, Chen Z, Mao J. Uvaol alleviates oxidative stress induced human umbilical vein endothelial cell injury by suppressing mitogen-activated protein kinase signaling pathway. Blood Coagul Fibrinolysis 2024; 35:248-255. [PMID: 38700418 DOI: 10.1097/mbc.0000000000001302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Deep venous thrombosis (DVT) is a potentially life-threatening disorder with high morbidity. Uvaol is a natural pentacyclic triterpene possessing multiple pharmacological activities. Nevertheless, the role of uvaol in DVT is unclarified. Human umbilical vein endothelial cells (HUVECs) were treated with hydrogen peroxide (H 2 O 2 ) to mimic DVT in vitro . CCK-8 assay and flow cytometry were utilized for measuring cell viability and apoptosis, respectively. Levels of the cell injury marker, thrombosis-associated factors, inflammatory cytokines, and oxidative stress-related markers were examined by commercial assay kits. Western blotting was used for evaluating the expression of mitogen-activated protein kinase (MAPK) signaling-associated proteins. Uvaol treatment attenuated H 2 O 2 -induced HUVEC apoptosis and injury. Uvaol reduced the expression of pro-thrombotic factors and inflammatory cytokines and attenuated oxidative stress in H 2 O 2 -stimulated HUVECs. Uvaol inhibited MAPK signaling pathway in H 2 O 2 -stimulated HUVECs. Activating MAPK signaling reversed uvaol-mediated protective effects on H 2 O 2 -treated HUVECs. Uvaol treatment alleviates H 2 O 2 -induced HUVEC injury, apoptosis, and oxidative stress by inactivating MAPK signaling.
Collapse
Affiliation(s)
| | | | | | - Ling Zhang
- Department of Medical Imaging, The People's Hospital of Dan Yang, Dan Yang, Jiangsu Province, P.R. China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | | |
Collapse
|
5
|
Zhou LQ, Chu YH, Dong MH, Yang S, Chen M, Tang Y, Pang XW, You YF, Wu LJ, Wang W, Qin C, Tian DS. Ldl-stimulated microglial activation exacerbates ischemic white matter damage. Brain Behav Immun 2024; 119:416-430. [PMID: 38636563 DOI: 10.1016/j.bbi.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/16/2024] [Indexed: 04/20/2024] Open
Abstract
The role of microglia in triggering the blood-brain barrier (BBB) impairment and white matter damage after chronic cerebral hypoperfusion is unclear. Here we demonstrated that the vessel-adjacent microglia were specifically activated by the leakage of plasma low-density lipoprotein (LDL), which led to BBB breakdown and ischemic demyelination. Interestingly, we found that LDL stimulation enhanced microglial phagocytosis, causing excessive engulfment of myelin debris and resulting in an overwhelming lipid burden in microglia. Surprisingly, these lipid-laden microglia exhibited a suppressed profile of inflammatory response and compromised pro-regenerative properties. Microglia-specific knockdown of LDLR or systematic medication lowering circulating LDL-C showed protective effects against ischemic demyelination. Overall, our findings demonstrated that LDL-stimulated vessel-adjacent microglia possess a disease-specific molecular signature, characterized by suppressed regenerative properties, which is associated with the propagation of demyelination during ischemic white matter damage.
Collapse
Affiliation(s)
- Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yue Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
6
|
van Hameren G, Aboghazleh R, Parker E, Dreier JP, Kaufer D, Friedman A. From spreading depolarization to blood-brain barrier dysfunction: navigating traumatic brain injury for novel diagnosis and therapy. Nat Rev Neurol 2024; 20:408-425. [PMID: 38886512 DOI: 10.1038/s41582-024-00973-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/08/2024] [Indexed: 06/20/2024]
Abstract
Considerable strides in medical interventions during the acute phase of traumatic brain injury (TBI) have brought improved overall survival rates. However, following TBI, people often face ongoing, persistent and debilitating long-term complications. Here, we review the recent literature to propose possible mechanisms that lead from TBI to long-term complications, focusing particularly on the involvement of a compromised blood-brain barrier (BBB). We discuss evidence for the role of spreading depolarization as a key pathological mechanism associated with microvascular dysfunction and the transformation of astrocytes to an inflammatory phenotype. Finally, we summarize new predictive and diagnostic biomarkers and explore potential therapeutic targets for treating long-term complications of TBI.
Collapse
Affiliation(s)
- Gerben van Hameren
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Refat Aboghazleh
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Basic Medical Sciences, Faculty of Medicine, Al-Balqa Applied University, Al-Salt, Jordan
| | - Ellen Parker
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
- Division of Neurosurgery, Dalhousie University QEII Health Sciences Centre, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Jens P Dreier
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine and Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada.
- Department of Cell Biology, Cognitive and Brain Sciences, Zelman Inter-Disciplinary Center of Brain Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
7
|
Ravizza T, Scheper M, Di Sapia R, Gorter J, Aronica E, Vezzani A. mTOR and neuroinflammation in epilepsy: implications for disease progression and treatment. Nat Rev Neurosci 2024; 25:334-350. [PMID: 38531962 DOI: 10.1038/s41583-024-00805-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2024] [Indexed: 03/28/2024]
Abstract
Epilepsy remains a major health concern as anti-seizure medications frequently fail, and there is currently no treatment to stop or prevent epileptogenesis, the process underlying the onset and progression of epilepsy. The identification of the pathological processes underlying epileptogenesis is instrumental to the development of drugs that may prevent the generation of seizures or control pharmaco-resistant seizures, which affect about 30% of patients. mTOR signalling and neuroinflammation have been recognized as critical pathways that are activated in brain cells in epilepsy. They represent a potential node of biological convergence in structural epilepsies with either a genetic or an acquired aetiology. Interventional studies in animal models and clinical studies give strong support to the involvement of each pathway in epilepsy. In this Review, we focus on available knowledge about the pathophysiological features of mTOR signalling and the neuroinflammatory brain response, and their interactions, in epilepsy. We discuss mitigation strategies for each pathway that display therapeutic effects in experimental and clinical epilepsy. A deeper understanding of these interconnected molecular cascades could enhance our strategies for managing epilepsy. This could pave the way for new treatments to fill the gaps in the development of preventative or disease-modifying drugs, thus overcoming the limitations of current symptomatic medications.
Collapse
Affiliation(s)
- Teresa Ravizza
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Mirte Scheper
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Rossella Di Sapia
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy
| | - Jan Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands.
| | - Annamaria Vezzani
- Department of Acute Brain and Cardiovascular Injury, Mario Negri Institute for Pharmacological Research IRCCS, Milano, Italy.
| |
Collapse
|
8
|
Zapata-Acevedo JF, Mantilla-Galindo A, Vargas-Sánchez K, González-Reyes RE. Blood-brain barrier biomarkers. Adv Clin Chem 2024; 121:1-88. [PMID: 38797540 DOI: 10.1016/bs.acc.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The blood-brain barrier (BBB) is a dynamic interface that regulates the exchange of molecules and cells between the brain parenchyma and the peripheral blood. The BBB is mainly composed of endothelial cells, astrocytes and pericytes. The integrity of this structure is essential for maintaining brain and spinal cord homeostasis and protection from injury or disease. However, in various neurological disorders, such as traumatic brain injury, Alzheimer's disease, and multiple sclerosis, the BBB can become compromised thus allowing passage of molecules and cells in and out of the central nervous system parenchyma. These agents, however, can serve as biomarkers of BBB permeability and neuronal damage, and provide valuable information for diagnosis, prognosis and treatment. Herein, we provide an overview of the BBB and changes due to aging, and summarize current knowledge on biomarkers of BBB disruption and neurodegeneration, including permeability, cellular, molecular and imaging biomarkers. We also discuss the challenges and opportunities for developing a biomarker toolkit that can reliably assess the BBB in physiologic and pathophysiologic states.
Collapse
Affiliation(s)
- Juan F Zapata-Acevedo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Mantilla-Galindo
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias, Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
9
|
He M, Lian T, Liu Z, Li J, Qi J, Li J, Guo P, Zhang Y, Luo D, Guan H, Zhang W, Zheng Z, Yue H, Zhang W, Wang R, Zhang F, Zhang W. An investigation into the potential association between nutrition and Alzheimer's disease. Front Nutr 2024; 11:1306226. [PMID: 38515521 PMCID: PMC10955128 DOI: 10.3389/fnut.2024.1306226] [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: 10/06/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024] Open
Abstract
Background Malnutrition is the most common nutritional issue in Alzheimer's disease (AD) patients, but there is still a lack of a comprehensive evaluation of the nutritional status in AD patients. This study aimed to determine the potential association of various nutritional indices with AD at different stages. Methods Subjects, including individuals with normal cognition (NC) and patients diagnosed with AD, were consecutively enrolled in this cross-sectional study. Demographics, body composition, dietary patterns, nutritional assessment scales and nutrition-related laboratory variables were collected. Binary logistics regression analyses and receiver operating characteristic (ROC) curves were used to indicate the association between nutrition-related variables and AD at different stages. Results Totals of 266 subjects, including 73 subjects with NC, 72 subjects with mild cognitive impairment due to AD (AD-MCI) and 121 subjects with dementia due to AD (AD-D) were included. There was no significant difference in dietary patterns, including Mediterranean diet and Mediterranean-DASH diet intervention for neurodegenerative delay (MIND) diet between the three groups. Lower BMI value, smaller hip and calf circumferences, lower Mini Nutritional Assessment (MNA) and Geriatric Nutritional Risk Index (GNRI) scores, and lower levels of total protein, albumin, globulin, and apolipoprotein A1 were associated with AD (all p < 0.05). Total protein and albumin levels had the greatest ability to distinguish AD from non-AD (AUC 0.80, 95% CI 0.74-0.84, p < 0.001), increased by combining calf circumference, MNA score and albumin level (AUC 0.83, 95% CI 0.77-0.88, p < 0.001). Albumin level had the greatest ability to distinguish NC from AD-MCI (AUC 0.75, 95% CI 0.67-0.82, p < 0.001), and MNA score greatest ability to distinguish AD-MCI from AD-D (AUC 0.72, 95% CI 0.65-0.78, p < 0.001). Conclusion Nutritional status of AD patients is significantly compromised compared with normal controls, and tends to be worsened with AD progresses. Early identification and intervention of individuals with nutritional risk or malnutrition may be significantly beneficial for reducing the risk, development, and progression of AD.
Collapse
Affiliation(s)
- Mingyue He
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tenghong Lian
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhan Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinghui Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Qi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jing Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peng Guo
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yanan Zhang
- Department of Blood Transfusion, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dongmei Luo
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huiying Guan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Weijia Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zijing Zheng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Yue
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wenjing Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruidan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wei Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Center of Parkinson’s Disease Institute for Brain Disorders, Beijing, China
- Beijing Key Laboratory on Parkinson Disease, Beijing, China
| |
Collapse
|
10
|
Amlerova Z, Chmelova M, Anderova M, Vargova L. Reactive gliosis in traumatic brain injury: a comprehensive review. Front Cell Neurosci 2024; 18:1335849. [PMID: 38481632 PMCID: PMC10933082 DOI: 10.3389/fncel.2024.1335849] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 01/03/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the most common pathological conditions impacting the central nervous system (CNS). A neurological deficit associated with TBI results from a complex of pathogenetic mechanisms including glutamate excitotoxicity, inflammation, demyelination, programmed cell death, or the development of edema. The critical components contributing to CNS response, damage control, and regeneration after TBI are glial cells-in reaction to tissue damage, their activation, hypertrophy, and proliferation occur, followed by the formation of a glial scar. The glial scar creates a barrier in damaged tissue and helps protect the CNS in the acute phase post-injury. However, this process prevents complete tissue recovery in the late/chronic phase by producing permanent scarring, which significantly impacts brain function. Various glial cell types participate in the scar formation, but this process is mostly attributed to reactive astrocytes and microglia, which play important roles in several brain pathologies. Novel technologies including whole-genome transcriptomic and epigenomic analyses, and unbiased proteomics, show that both astrocytes and microglia represent groups of heterogenic cell subpopulations with different genomic and functional characteristics, that are responsible for their role in neurodegeneration, neuroprotection and regeneration. Depending on the representation of distinct glia subpopulations, the tissue damage as well as the regenerative processes or delayed neurodegeneration after TBI may thus differ in nearby or remote areas or in different brain structures. This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS injury and the distance of the explored area from the lesion site. Here, we also discuss findings concerning intercellular signaling, long-term impacts of TBI and the possibilities of novel therapeutical approaches. We believe that a comprehensive study with an emphasis on glial cells, involved in tissue post-injury processes, may be helpful for further research of TBI and be the decisive factor when choosing a TBI model.
Collapse
Affiliation(s)
- Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Miroslava Anderova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
11
|
Grewal S, Gonçalves de Andrade E, Kofoed RH, Matthews PM, Aubert I, Tremblay MÈ, Morse SV. Using focused ultrasound to modulate microglial structure and function. Front Cell Neurosci 2023; 17:1290628. [PMID: 38164436 PMCID: PMC10757935 DOI: 10.3389/fncel.2023.1290628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Transcranial focused ultrasound (FUS) has the unique ability to target regions of the brain with high spatial precision, in a minimally invasive manner. Neuromodulation studies have shown that FUS can excite or inhibit neuronal activity, demonstrating its tremendous potential to improve the outcome of neurological diseases. Recent evidence has also shed light on the emerging promise that FUS has, with and without the use of intravenously injected microbubbles, in modulating the blood-brain barrier and the immune cells of the brain. As the resident immune cells of the central nervous system, microglia are at the forefront of the brain's maintenance and immune defense. Notably, microglia are highly dynamic and continuously survey the brain parenchyma by extending and retracting their processes. This surveillance activity aids microglia in performing key physiological functions required for brain activity and plasticity. In response to stressors, microglia rapidly alter their cellular and molecular profile to help facilitate a return to homeostasis. While the underlying mechanisms by which both FUS and FUS + microbubbles modify microglial structure and function remain largely unknown, several studies in adult mice have reported changes in the expression of the microglia/macrophage marker ionized calcium binding adaptor molecule 1, and in their phagocytosis, notably of protein aggregates, such as amyloid beta. In this review, we discuss the demonstrated and putative biological effects of FUS and FUS + microbubbles in modulating microglial activities, with an emphasis on the key cellular and molecular changes observed in vitro and in vivo across models of brain health and disease. Understanding how this innovative technology can modulate microglia paves the way for future therapeutic strategies aimed to promote beneficial physiological microglial roles, and prevent or treat maladaptive responses.
Collapse
Affiliation(s)
- Sarina Grewal
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Center for Experimental Neuroscience-CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Paul M. Matthews
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sophie V. Morse
- Department of Bioengineering, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
12
|
Kang YJ, Diep YN, Tran M, Tran VTA, Ambrin G, Ngo H, Cho H. Three-dimensional human neural culture on a chip recapitulating neuroinflammation and neurodegeneration. Nat Protoc 2023; 18:2838-2867. [PMID: 37542184 DOI: 10.1038/s41596-023-00861-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 05/30/2023] [Indexed: 08/06/2023]
Abstract
Neuroinflammation has either beneficial or detrimental effects, depending on risk factors and neuron-glia interactions in neurological disorders. However, studying neuroinflammation has been challenging due to the complexity of cell-cell interactions and lack of physio-pathologically relevant neuroinflammatory models. Here, we describe our three-dimensional microfluidic multicellular human neural culture model, referred to as a 'brain-on-a-chip' (BoC). This elucidates neuron-glia interactions in a controlled manner and recapitulates pathological signatures of the major neurological disorders: dementia, brain tumor and brain edema. This platform includes a chemotaxis module offering a week-long, stable chemo-gradient compared with the few hours in other chemotaxis models. Additionally, compared with conventional brain models cultured with mixed phenotypes of microglia, our BoC can separate the disease-associated microglia out of heterogeneous population and allow selective neuro-glial engagement in three dimensions. This provides benefits of interpreting the neuro-glia interactions while revealing that the prominent activation of innate immune cells is the risk factor leading to synaptic impairment and neuronal loss, validated in our BoC models of disorders. This protocol describes how to fabricate and implement our human BoC, manipulate in real time and perform end-point analyses. It takes 2 d to set up the device and cell preparations, 1-9 weeks to develop brain models under disease conditions and 2-3 d to carry out analyses. This protocol requires at least 1 month training for researchers with basic molecular biology techniques. Taken together, our human BoCs serve as reliable and valuable platforms to investigate pathological mechanisms involving neuroinflammation and to assess therapeutic strategies modulating neuroinflammation in neurological disorders.
Collapse
Affiliation(s)
- You Jung Kang
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yen N Diep
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Van Thi Ai Tran
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ghuncha Ambrin
- Department of Psychiatry, School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Huyen Ngo
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
13
|
Altendorfer-Kroath T, Hummer J, Birngruber T. In vivo monitoring of brain pharmacokinetics and pharmacodynamics with cerebral open flow microperfusion. Biopharm Drug Dispos 2023; 44:84-93. [PMID: 36650922 DOI: 10.1002/bdd.2343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/16/2022] [Accepted: 12/25/2022] [Indexed: 01/19/2023]
Abstract
In vivo investigation of brain pharmacokinetics and pharmacodynamics (PK/PD) is an integral part of neurological drug development. However, drugs intended to act in the brain may reach it at very low concentrations due to the protective effect of the blood-brain barrier (BBB). Consequently, very sensitive measurement methods are required to investigate PK/PD of drugs in the brain. Also, these methods must be capable of continuously assessing cerebral drug concentrations with verifiable intact BBB, as disrupted BBB may lead to compound efflux from blood into brain and to biased results. To date, only a few techniques are available that can sensitively measure drug concentrations in the brain over time; one of which is cerebral open flow microperfusion (cOFM). cOFM's key features are that it enables measurement of cerebral compound concentrations with intact BBB, induces only minor tissue reactions, and that no scar formation occurs around the probe. The membrane-free cOFM probes collect diluted cerebral interstitial fluid (ISF) samples that are containing the whole molecule spectrum of the ISF. Further, combining cOFM with an in vivo calibration protocol (e.g. Zero Flow Rate) enables absolute quantification of compounds in cerebral ISF. In general, three critical aspects have to be considered when measuring cerebral drug concentrations and recording PK/PD profiles with cOFM: (a) the BBB integrity during sampling, (b) the status of the brain tissue next to the cOFM probe during sampling, and (c) the strategy to absolutely quantify drugs in cerebral ISF. This work aims to review recent applications of cOFM for PK/PD assessment with a special focus on these critical aspects.
Collapse
Affiliation(s)
| | - Joanna Hummer
- Institute for Biomedical Research and Technologies, JOANNEUM RESEARCH - HEALTH, Graz, Austria
| | - Thomas Birngruber
- Institute for Biomedical Research and Technologies, JOANNEUM RESEARCH - HEALTH, Graz, Austria
| |
Collapse
|
14
|
Kirsch D, Shah A, Dixon E, Kelley H, Cherry JD, Xia W, Daley S, Aytan N, Cormier K, Kubilus C, Mathias R, Alvarez VE, Huber BR, McKee AC, Stein TD. Vascular injury is associated with repetitive head impacts and tau pathology in chronic traumatic encephalopathy. J Neuropathol Exp Neurol 2023; 82:127-139. [PMID: 36617181 PMCID: PMC9852946 DOI: 10.1093/jnen/nlac122] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease linked to repetitive head impacts (RHI) and characterized by perivascular hyperphosphorylated tau (p-tau) deposits. The role of vascular injury, blood-brain barrier leakage, and neuroinflammation in CTE pathogenesis is not well understood. We performed quantitative immunoassays for intercellular adhesion molecule 1 (ICAM1), vascular cellular adhesion molecule 1 (VCAM1), and C-reactive protein (CRP) within the postmortem dorsolateral frontal cortex of participants with and without a history of RHI and CTE (n = 156), and tested for associations with RHI, microgliosis, and tau pathology measures. Levels of vascular injury-associated markers ICAM1, VCAM1, and CRP were increased in CTE compared to RHI-exposed and -naïve controls. ICAM1 and CRP increased with RHI exposure duration (p < 0.01) and were associated with increased microglial density (p < 0.001) and tau pathology (AT8, p-tau396, p-tau202; p < 0.05). Histologically, there was significantly increased ICAM1 staining of the microvasculature, extracellular space, and astrocytes at the sulcal depths in high stage CTE compared to both low stage CTE and controls. Multifocal perivascular immunoreactivity for serum albumin was present in all RHI-exposed individuals. These findings demonstrate that vascular injury markers are associated with RHI exposure, duration, and microgliosis, are elevated in CTE, and increase with disease severity.
Collapse
Affiliation(s)
- Daniel Kirsch
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Arsal Shah
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Erin Dixon
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Hunter Kelley
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Jonathan D Cherry
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Weiming Xia
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Sarah Daley
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Nurgul Aytan
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Kerry Cormier
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Carol Kubilus
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| | - Rebecca Mathias
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Victor E Alvarez
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Bertrand R Huber
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Ann C McKee
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Thor D Stein
- Boston University Alzheimer’s Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- VA Bedford Healthcare System, Bedford, Massachusetts, USA
| |
Collapse
|
15
|
Vezzani A, Ravizza T, Bedner P, Aronica E, Steinhäuser C, Boison D. Astrocytes in the initiation and progression of epilepsy. Nat Rev Neurol 2022; 18:707-722. [PMID: 36280704 PMCID: PMC10368155 DOI: 10.1038/s41582-022-00727-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/09/2022]
Abstract
Epilepsy affects ~65 million people worldwide. First-line treatment options include >20 antiseizure medications, but seizure control is not achieved in approximately one-third of patients. Antiseizure medications act primarily on neurons and can provide symptomatic control of seizures, but do not alter the onset and progression of epilepsy and can cause serious adverse effects. Therefore, medications with new cellular and molecular targets and mechanisms of action are needed. Accumulating evidence indicates that astrocytes are crucial to the pathophysiological mechanisms of epilepsy, raising the possibility that these cells could be novel therapeutic targets. In this Review, we discuss how dysregulation of key astrocyte functions - gliotransmission, cell metabolism and immune function - contribute to the development and progression of hyperexcitability in epilepsy. We consider strategies to mitigate astrocyte dysfunction in each of these areas, and provide an overview of how astrocyte activation states can be monitored in vivo not only to assess their contribution to disease but also to identify markers of disease processes and treatment effects. Improved understanding of the roles of astrocytes in epilepsy has the potential to lead to novel therapies to prevent the initiation and progression of epilepsy.
Collapse
Affiliation(s)
- Annamaria Vezzani
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy.
| | - Teresa Ravizza
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, Italy
| | - Peter Bedner
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro)Pathology, Amsterdam Neuroscience, Amsterdam, Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany
| | - Detlev Boison
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
16
|
Williams NP, Kushwah N, Dhawan V, Zheng XS, Cui XT. Effects of central nervous system electrical stimulation on non-neuronal cells. Front Neurosci 2022; 16:967491. [PMID: 36188481 PMCID: PMC9521315 DOI: 10.3389/fnins.2022.967491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past few decades, much progress has been made in the clinical use of electrical stimulation of the central nervous system (CNS) to treat an ever-growing number of conditions from Parkinson's disease (PD) to epilepsy as well as for sensory restoration and many other applications. However, little is known about the effects of microstimulation at the cellular level. Most of the existing research focuses on the effects of electrical stimulation on neurons. Other cells of the CNS such as microglia, astrocytes, oligodendrocytes, and vascular endothelial cells have been understudied in terms of their response to stimulation. The varied and critical functions of these cell types are now beginning to be better understood, and their vital roles in brain function in both health and disease are becoming better appreciated. To shed light on the importance of the way electrical stimulation as distinct from device implantation impacts non-neuronal cell types, this review will first summarize common stimulation modalities from the perspective of device design and stimulation parameters and how these different parameters have an impact on the physiological response. Following this, what is known about the responses of different cell types to different stimulation modalities will be summarized, drawing on findings from both clinical studies as well as clinically relevant animal models and in vitro systems.
Collapse
Affiliation(s)
- Nathaniel P. Williams
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Neetu Kushwah
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Vaishnavi Dhawan
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
| | - Xin Sally Zheng
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| |
Collapse
|
17
|
Mahmoudi A, Heydari S, Markina YV, Barreto GE, Sahebkar A. Role of statins in regulating molecular pathways following traumatic brain injury: A system pharmacology study. Biomed Pharmacother 2022; 153:113304. [PMID: 35724514 DOI: 10.1016/j.biopha.2022.113304] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022] Open
Abstract
Traumatic brain injury (TBI) is a serious disorder with debilitating physical and psychological complications. Previous studies have indicated that genetic factors have a critical role in modulating the secondary phase of injury in TBI. Statins have interesting pleiotropic properties such as antiapoptotic, antioxidative, and anti-inflammatory effects, which make them a suitable class of drugs for repurposing in TBI. In this study, we aimed to explore how statins modulate proteins and pathways involved in TBI using system pharmacology. We first explored the target associations with statins in two databases to discover critical clustering groups, candidate hub and critical hub genes in the network of TBI, and the possible connections of statins with TBI-related genes. Our results showed 1763 genes associated with TBI. Subsequently, the analysis of centralities in the PPI network displayed 55 candidate hub genes and 15 hub genes. Besides, MCODE analysis based on threshold score:10 determined four modular clusters. Intersection analysis of genes related to TBI and statins demonstrated 204 shared proteins, which suggested that statins influence 31 candidate hub and 9 hub genes. Moreover, statins had the highest interaction with MCODE1. The biological processes of the 31 shared proteins are related to gene expression, inflammation, antioxidant activity, and cell proliferation. Biological enriched pathways showed Programmed Cell Death proteins, AGE-RAGE signaling pathway, C-type lectin receptor signalling pathway, and MAPK signaling pathway as top clusters. In conclusion, statins could target several critical post-TBI genes mainly involved in inflammation and apoptosis, supporting the previous research results as a potential therapeutic agent.
Collapse
Affiliation(s)
- Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177899191, the Islamic Republic of Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran
| | - Sahar Heydari
- Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, the Islamic Republic of Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran
| | - Yuliya V Markina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology of FSBI "Petrovsky National Research Center of Surgery", 3 Tsyurupy Str., 117418, Moscow, the Russian Federation
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, the Islamic Republic of Iran.
| |
Collapse
|
18
|
Laminin as a Biomarker of Blood-Brain Barrier Disruption under Neuroinflammation: A Systematic Review. Int J Mol Sci 2022; 23:ijms23126788. [PMID: 35743229 PMCID: PMC9224176 DOI: 10.3390/ijms23126788] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/03/2022] [Accepted: 06/10/2022] [Indexed: 01/01/2023] Open
Abstract
Laminin, a non-collagenous glycoprotein present in the brain extracellular matrix, helps to maintain blood–brain barrier (BBB) integrity and regulation. Neuroinflammation can compromise laminin structure and function, increasing BBB permeability. The aim of this paper is to determine if neuroinflammation-induced laminin functional changes may serve as a potential biomarker of alterations in the BBB. The 38 publications included evaluated neuroinflammation, BBB disruption, and laminin, and were assessed for quality and risk of bias (protocol registered in PROSPERO; CRD42020212547). We found that laminin may be a good indicator of BBB overall structural integrity, although changes in expression are dependent on the pathologic or experimental model used. In ischemic stroke, permanent vascular damage correlates with increased laminin expression (β and γ subunits), while transient damage correlates with reduced laminin expression (α subunits). Laminin was reduced in traumatic brain injury and cerebral hemorrhage studies but increased in multiple sclerosis and status epilepticus studies. Despite these observations, there is limited knowledge about the role played by different subunits or isoforms (such as 411 or 511) of laminin in maintaining structural architecture of the BBB under neuroinflammation. Further studies may clarify this aspect and the possibility of using laminin as a biomarker in different pathologies, which have alterations in BBB function in common.
Collapse
|
19
|
Preininger MK, Kaufer D. Blood-Brain Barrier Dysfunction and Astrocyte Senescence as Reciprocal Drivers of Neuropathology in Aging. Int J Mol Sci 2022; 23:6217. [PMID: 35682895 PMCID: PMC9180977 DOI: 10.3390/ijms23116217] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 01/27/2023] Open
Abstract
As the most abundant cell types in the brain, astrocytes form a tissue-wide signaling network that is responsible for maintaining brain homeostasis and regulating various brain activities. Here, we review some of the essential functions that astrocytes perform in supporting neurons, modulating the immune response, and regulating and maintaining the blood-brain barrier (BBB). Given their importance in brain health, it follows that astrocyte dysfunction has detrimental effects. Indeed, dysfunctional astrocytes are implicated in age-related neuropathology and participate in the onset and progression of neurodegenerative diseases. Here, we review two mechanisms by which astrocytes mediate neuropathology in the aging brain. First, age-associated blood-brain barrier dysfunction (BBBD) causes the hyperactivation of TGFβ signaling in astrocytes, which elicits a pro-inflammatory and epileptogenic phenotype. Over time, BBBD-associated astrocyte dysfunction results in hippocampal and cortical neural hyperexcitability and cognitive deficits. Second, senescent astrocytes accumulate in the brain with age and exhibit a decreased functional capacity and the secretion of senescent-associated secretory phenotype (SASP) factors, which contribute to neuroinflammation and neurotoxicity. Both BBBD and senescence progressively increase during aging and are associated with increased risk of neurodegenerative disease, but the relationship between the two has not yet been established. Thus, we discuss the potential relationship between BBBD, TGFβ hyperactivation, and senescence with respect to astrocytes in the context of aging and disease and identify future areas of investigation in the field.
Collapse
Affiliation(s)
- Marcela K. Preininger
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA;
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, Berkeley, CA 94720, USA;
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
20
|
Shojai S, Haeri Rohani SA, Moosavi-Movahedi AA, Habibi-Rezaei M. Human serum albumin in neurodegeneration. Rev Neurosci 2022; 33:803-817. [PMID: 35363449 DOI: 10.1515/revneuro-2021-0165] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/02/2022] [Indexed: 11/15/2022]
Abstract
Serum albumin (SA) exists in relatively high concentrations, in close contact with most cells. However, in the adult brain, except for cerebrospinal fluid (CSF), SA concentration is relatively low. It is mainly produced in the liver to serve as the main protein of the blood plasma. In the plasma, it functions as a carrier, chaperon, antioxidant, source of amino acids, osmoregulator, etc. As a carrier, it facilitates the stable presence and transport of the hydrophobic and hydrophilic molecules, including free fatty acids, steroid hormones, medicines, and metal ions. As a chaperon, SA binds to and protects other proteins. As an antioxidant, thanks to a free sulfhydryl group (-SH), albumin is responsible for most antioxidant properties of plasma. These functions qualify SA as a major player in, and a mirror of, overall health status, aging, and neurodegeneration. The low concentration of SA is associated with cognitive deterioration in the elderly and negative prognosis in multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). SA has been shown to be structurally modified in neurological conditions such as Alzheimer's disease (AD). During blood-brain barrier damage albumin enters the brain tissue and could trigger epilepsy and neurodegeneration. SA is able to bind to the precursor agent of the AD, amyloid-beta (Aβ), preventing its toxic effects in the periphery, and is being tested for treating this disease. SA therapy may also be effective in brain rejuvenation. In the current review, we will bring forward the prominent properties and roles of SA in neurodegeneration.
Collapse
Affiliation(s)
- Sajjad Shojai
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | | | | | - Mehran Habibi-Rezaei
- School of Biology, College of Science, University of Tehran, Tehran, Iran
- Nano-Biomedicine Center of Excellence, Nanoscience and Nanotechnology Research Center, University of Tehran, Tehran, Iran
| |
Collapse
|
21
|
Chen S, Nazeri A, Baek H, Ye D, Yang Y, Yuan J, Rubin JB, Chen H. A review of bioeffects induced by focused ultrasound combined with microbubbles on the neurovascular unit. J Cereb Blood Flow Metab 2022; 42:3-26. [PMID: 34551608 PMCID: PMC8721781 DOI: 10.1177/0271678x211046129] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 01/29/2023]
Abstract
Focused ultrasound combined with circulating microbubbles (FUS+MB) can transiently enhance blood-brain barrier (BBB) permeability at targeted brain locations. Its great promise in improving drug delivery to the brain is reflected by a rapidly growing number of clinical trials using FUS+MB to treat various brain diseases. As the clinical applications of FUS+MB continue to expand, it is critical to have a better understanding of the molecular and cellular effects induced by FUS+MB to enhance the efficacy of current treatment and enable the discovery of new therapeutic strategies. Existing studies primarily focus on FUS+MB-induced effects on brain endothelial cells, the major cellular component of BBB. However, bioeffects induced by FUS+MB expand beyond the BBB to cells surrounding blood vessels, including astrocytes, microglia, and neurons. Together these cell types comprise the neurovascular unit (NVU). In this review, we examine cell-type-specific bioeffects of FUS+MB on different NVU components, including enhanced permeability in endothelial cells, activation of astrocytes and microglia, as well as increased intraneuron protein metabolism and neuronal activity. Finally, we discuss knowledge gaps that must be addressed to further advance clinical applications of FUS+MB.
Collapse
Affiliation(s)
- Si Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hongchae Baek
- Imaging Institute and Neurological Institute, Cleveland Clinic, Cleveland Clinic, Cleveland, OH, USA
| | - Dezhuang Ye
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinyun Yuan
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
| | - Joshua B Rubin
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, USA
- Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
22
|
Angelopoulou E, Paudel YN, Piperi C. Role of Liver Growth Factor (LGF) in Parkinson's Disease: Molecular Insights and Therapeutic Opportunities. Mol Neurobiol 2021; 58:3031-3042. [PMID: 33608826 DOI: 10.1007/s12035-021-02326-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 02/09/2021] [Indexed: 11/26/2022]
Abstract
Parkinson's disease is the most common neurodegenerative movement disorder with unclear etiology and only symptomatic treatment to date. Toward the development of novel disease-modifying agents, neurotrophic factors represent a reasonable and promising therapeutic approach. However, despite the robust preclinical evidence, clinical trials using glial-derived neurotrophic factor (GDNF) and neurturin have been unsuccessful. In this direction, the therapeutic potential of other trophic factors in PD and the elucidation of the underlying molecular mechanisms are of paramount importance. The liver growth factor (LGF) is an albumin-bilirubin complex acting as a hepatic mitogen, which also exerts regenerative effects on several extrahepatic tissues including the brain. Accumulating evidence suggests that intracerebral and peripheral administration of LGF can enhance the outgrowth of nigrostriatal dopaminergic axonal terminals; promote the survival, migration, and differentiation of neuronal stem cells; and partially protect against dopaminergic neuronal loss in the substantia nigra of PD animal models. In most studies, these effects are accompanied by improved motor behavior of the animals. Potential underlying mechanisms involve transient microglial activation, TNF-α upregulation, and activation of the extracellular signal-regulated kinases 1/2 (ERK1/2) and of the transcription factor cyclic AMP response-element binding protein (CREB), along with anti-inflammatory and antioxidant pathways. Herein, we summarize recent preclinical evidence on the potential role of LGF in PD pathogenesis, aiming to shed more light on the underlying molecular mechanisms and reveal novel therapeutic opportunities for this debilitating disease.
Collapse
Affiliation(s)
- Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
23
|
Dubaniewicz M, Eles JR, Lam S, Song S, Cambi F, Sun D, Wellman SM, Kozai TDY. Inhibition of Na +/H +exchanger modulates microglial activation and scar formation following microelectrode implantation. J Neural Eng 2021; 18. [PMID: 33621208 DOI: 10.1088/1741-2552/abe8f1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/23/2021] [Indexed: 11/12/2022]
Abstract
Objective.Intracortical microelectrodes are an important tool for neuroscience research and have great potential for clinical use. However, the use of microelectrode arrays to treat neurological disorders and control prosthetics is limited by biological challenges such as glial scarring, which can impair chronic recording performance. Microglia activation is an early and prominent contributor to glial scarring. After insertion of an intracortical microelectrode, nearby microglia transition into a state of activation, migrate, and encapsulate the device. Na+/H+exchanger isoform-1 (NHE-1) is involved in various microglial functions, including their polarity and motility, and has been implicated in pro-inflammatory responses to tissue injury. HOE-642 (cariporide) is an inhibitor of NHE-1 and has been shown to depress microglial activation and inflammatory response in brain injury models.Approach.In this study, the effects of HOE-642 treatment on microglial interactions to intracortical microelectrodes was evaluated using two-photon microscopyin vivo.Main results.The rate at which microglia processes and soma migrate in response to electrode implantation was unaffected by HOE-642 administration. However, HOE-642 administration effectively reduced the radius of microglia activation at 72 h post-implantation from 222.2µm to 177.9µm. Furthermore, treatment with HOE-642 significantly reduced microglial encapsulation of implanted devices at 5 h post-insertion from 50.7 ± 6.0% to 8.9 ± 6.1%, which suggests an NHE-1-specific mechanism mediating microglia reactivity and gliosis during implantation injury.Significance.This study implicates NHE-1 as a potential target of interest in microglial reactivity and HOE-642 as a potential treatment to attenuate the glial response and scar formation around implanted intracortical microelectrodes.
Collapse
Affiliation(s)
- Mitchell Dubaniewicz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James R Eles
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Stephanie Lam
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Shanshan Song
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Dandan Sun
- Veterans Administration Pittsburgh, Pittsburgh, PA, United States of America.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America.,McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America.,NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
24
|
Liu KJ, Lv YX, Niu YM, Bu Y. Prognostic value of γ-glutamyl transpeptidase to albumin ratio combined with aspartate aminotransferase to lymphocyte ratio in patients with hepatocellular carcinoma after hepatectomy. Medicine (Baltimore) 2020; 99:e23339. [PMID: 33235099 PMCID: PMC7710195 DOI: 10.1097/md.0000000000023339] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor associated with a high recurrence rate after hepatectomy. Recently, preoperative inflammatory and liver function reserve indices were found to predict increased risk of recurrence and decreased survival in HCC patients. This study aims to evaluate the ability of the γ-glutamyl transpeptidase-to-albumin ratio (GAR) and aspartate aminotransferase-to-lymphocyte ratio (ALRI), individually and in combination, to predict the prognosis of HCC patients after hepatectomy.We retrospectively reviewed 206 HCC patients who underwent radical resection at the General Hospital of Ningxia Medical University from January 2011 to November 2016. Receiver operating characteristic (ROC) curve analysis was performed to determine the optimal cut-off value for GAR and ALRI. The Pearson Chi-Squared test was used to analyze the correlations between GAR, ALRI and clinicopathological characteristics. Univariate and multivariate analyses were used to determine the predictive value of these factors for disease-free survival (DFS) and overall survival (OS). Survival rates were drawn according to the Kaplan-Meier method and differences between subgroups were compared by the log-rank statistics.GAR and ALRI were significantly correlated with gender, history of smoking, prothrombin time, tumor diameter, T stage and early intrahepatic recurrence by the Pearson Chi-Squared test (all P < .05). Univariate analysis indicated that T stage, GAR and ALRI were significantly correlated with DFS and OS in HCC patients after hepatectomy. Multivariate analysis illustrated that GAR and ALRI were independently related to DFS and OS in HCC patients. Preoperative GAR > 0.946 or ALRI > 18.734 predicted poor prognosis in HCC patients after hepatectomy. Additionally, the predictive scope of GAR combined with ALRI was more sensitive than that of either individual measurement alone.Our data indicate that there is a close association between the clinicopathological characteristics in HCC patients and increased GAR or ALRI. Higher levels of GAR and ALRI could sensitively and specifically predict a poor prognosis in HCC patients after hepatectomy. Furthermore, combined usage of GAR and ALRI could improve the accuracy of this prediction.
Collapse
Affiliation(s)
- Ke-jun Liu
- School of Clinical Medicine, Ningxia Medical University
| | - Yong-xue Lv
- School of Basic Medicine, Ningxia Medical University
| | - Yi-ming Niu
- School of Clinical Medicine, Ningxia Medical University
| | - Yang Bu
- School of Clinical Medicine, Ningxia Medical University
- Department of Hepatobiliary Surgery, People‘s Hospital of Ningxia Hui Autonomous Region, Ningxia, China
| |
Collapse
|
25
|
Epigallocatechin-3-Gallate and PEDF 335 Peptide, 67LR Activators, Attenuate Vasogenic Edema, and Astroglial Degeneration Following Status Epilepticus. Antioxidants (Basel) 2020; 9:antiox9090854. [PMID: 32933011 PMCID: PMC7555521 DOI: 10.3390/antiox9090854] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Non-integrin 67-kDa laminin receptor (67LR) is involved in cell adherence to the basement membrane, and it regulates the interactions between laminin and other receptors. The dysfunction of 67LR leads to serum extravasation via blood-brain barrier (BBB) disruption. Polyphenol (–)-epigallocatechin-3-O-gallate (EGCG) and pigment epithelium-derived factor (PEDF) bind to 67LR and inhibit neovascularization. Therefore, in the present study, we investigated the effects of EGCG and NU335, a PEDF-derive peptide, on BBB integrity and their possible underlying mechanisms against vasogenic edema formation induced by status epilepticus (SE, a prolonged seizure activity). Following SE, both EGCG and NU335 attenuated serum extravasation and astroglial degeneration in the rat piriform cortex (PC). Both EGCG and NU335 reversely regulated phosphatidylinositol 3 kinase (PI3K)/AKT–eNOS (endothelial nitric oxide synthase) mediated BBB permeability and aquaporin 4 (AQP4) expression in endothelial cells and astrocytes through the p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways, respectively. Furthermore, EGCG and NU335 decreased p47Phox (a nicotinamide adenine dinucleotide phosphate oxidase subunit) expression in astrocytes under physiological and post-SE conditions. Therefore, we suggest that EGCG and PEDF derivatives may activate 67LR and its downstream effectors, and they may be considerable anti-vasogenic edema agents.
Collapse
|
26
|
Kim JE, Park H, Lee JE, Kang TC. Blockade of 67-kDa Laminin Receptor Facilitates AQP4 Down-Regulation and BBB Disruption via ERK1/2-and p38 MAPK-Mediated PI3K/AKT Activations. Cells 2020; 9:cells9071670. [PMID: 32664509 PMCID: PMC7407797 DOI: 10.3390/cells9071670] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
Recently, we have reported that dysfunctions of 67-kDa laminin receptor (67LR) induced by status epilepticus (SE, a prolonged seizure activity) and 67LR neutralization are involved in vasogenic edema formation, accompanied by the reduced aquaporin 4 (AQP4, an astroglial specific water channel) expression in the rat piriform cortex (PC). In the present study, we found that the blockade of 67LR activated p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways, which enhanced phosphatidylinositol 3 kinase (PI3K)/AKT phosphorylations in endothelial cells and astrocytes, respectively. 67LR-p38 MAPK-PI3K-AKT activation in endothelial cells increased vascular permeability. In contrast, 67LR-ERK1/2-PI3K-AKT signaling pathways in astrocytes regulated astroglial viability and AQP4 expression. These findings indicate that PI3K/AKT may integrate p38 MAPK and ERK1/2 signaling pathways to regulate AQP4 expression when 67LR functionality is reduced. Thus, we suggest that 67LR-p38 MAPK/ERK1/2-PI3K-AKT-AQP4 signaling cascades may mediate serum extravasation and AQP4 expression in astroglio-vascular systems, which is one of the considerable therapeutic targets for vasogenic edema in various neurological diseases.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (H.P.); (J.-E.L.)
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Hana Park
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (H.P.); (J.-E.L.)
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Ji-Eun Lee
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (H.P.); (J.-E.L.)
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon 24252, Korea; (J.-E.K.); (H.P.); (J.-E.L.)
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon 24252, Korea
- Correspondence: ; Tel.: +82-33-248-2524; Fax: +82-33-248-2525
| |
Collapse
|
27
|
Yang Q, Wu B, Eles JR, Vazquez AL, Kozai TDY, Cui XT. Zwitterionic Polymer Coating Suppresses Microglial Encapsulation to Neural Implants In Vitro and In Vivo. ADVANCED BIOSYSTEMS 2020; 4:e1900287. [PMID: 32363792 PMCID: PMC7686959 DOI: 10.1002/adbi.201900287] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
For brain computer interfaces (BCI), the immune response to implanted electrodes is a major biological cause of device failure. Bioactive coatings such as neural adhesion molecule L1 have been shown to improve the biocompatibility, but are difficult to handle or produce in batches. Here, a synthetic zwitterionic polymer coating, poly(sulfobetaine methacrylate) (PSBMA) is developed for neural implants with the goal of reducing the inflammatory host response. In tests in vitro, the zwitterionic coating inhibits protein adsorption and the attachment of fibroblasts and microglia, and remains stable for at least 4 weeks. In vivo two-photon microscopy on CX3CR1-GFP mice shows that the zwitterionic coating significantly suppresses the microglial encapsulation of neural microelectrodes over a 6 h observation period. Furthermore, the lower microglial encapsulation on zwitterionic polymer-coated microelectrodes is revealed to originate from a reduction in the size but not the number of microglial end feet. This work provides a facile method for coating neural implants with zwitterionic polymers and illustrates the initial interaction between microglia and coated surface at high temporal and spatial resolution.
Collapse
Affiliation(s)
- Qianru Yang
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Bingchen Wu
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - James R Eles
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| | - Alberto L Vazquez
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 East Carson Street, Pittsburgh, PA, 15219, USA
| | - Takashi D Y Kozai
- Center for Biotechnology and Bioengineering, University of Pittsburgh, 300 Technology Dr, Pittsburgh, PA, 15213, USA
| | - X Tracy Cui
- Biomedical Science Tower 3, University of Pittsburgh, 3501 Fifth Ave, Pittsburgh, PA, 15232, USA
| |
Collapse
|
28
|
Gonzalo-Gobernado R, Reimers D, Casarejos MJ, Calatrava Ferreras L, Vallejo-Muñoz M, Jiménez-Escrig A, Diaz-Gil JJ, Ulzurrun de Asanza GM, Bazán E. Liver Growth Factor Induces Glia-Associated Neuroprotection in an In Vitro Model of Parkinson´s Disease. Brain Sci 2020; 10:brainsci10050315. [PMID: 32455921 PMCID: PMC7287666 DOI: 10.3390/brainsci10050315] [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: 05/06/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 11/16/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by the progressive death of dopaminergic (DA) neurons in the substantia nigra (SN), which leads to a loss of the neurotransmitter dopamine in the basal ganglia. Current treatments relieve the symptoms of the disease, but none stop or delay neuronal degeneration. Liver growth factor (LGF) is an albumin-bilirubin complex that stimulates axonal growth in the striatum and protects DA neurons in the SN of 6-hydroxydopamine-lesioned rats. Our previous results suggested that these effects observed in vivo are mediated by microglia and/or astrocytes. To determine if these cells are LGF targets, E14 (embryos from Sprague Dawley rats of 14 days) rat mesencephalic glial cultures were used. Treatment with 100 pg/mL of LGF up-regulated the mitogen-activated protein kinases (MAPKs) extracellular signal-regulated kinases 1/2 (ERK1/2) and the cyclic AMP response element binding protein (CREB) phosphorylation in glial cultures, and it increased the microglia marker Iba1 and tumor necrosis factor alpha (TNF-alpha) protein levels. The treatment of E14 midbrain neurons with a glial-conditioned medium from LGF-treated glial cultures (GCM-LGF) prevented the loss of DA neurons caused by 6-hydroxy-dopamine. This neuroprotective effect was not observed when GCM-LGF was applied in the presence of a blocking antibody of TNF-alpha activity. Altogether, our findings strongly suggest the involvement of microglia and TNF-alpha in the neuroprotective action of LGF on DA neurons observed in vitro.
Collapse
Affiliation(s)
- Rafael Gonzalo-Gobernado
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
- National Centre for Biotechnology (CNB), CSIC, 28049 Madrid, Spain
- Correspondence: (R.G.-G.); (E.B.); Tel.: +34-913-368-168 (R.G.-G. & E.B.)
| | - Diana Reimers
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | - María José Casarejos
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | - Lucía Calatrava Ferreras
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | - Manuela Vallejo-Muñoz
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | | | - Juan José Diaz-Gil
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | - Gonzalo M. Ulzurrun de Asanza
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
| | - Eulalia Bazán
- Servicio de Neurobiología, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain; (D.R.); (M.J.C.); (L.C.F.); (M.V.-M.); (J.J.D.-G.); (G.M.U.d.A.)
- Correspondence: (R.G.-G.); (E.B.); Tel.: +34-913-368-168 (R.G.-G. & E.B.)
| |
Collapse
|
29
|
Perez-Ramirez MB, Gu F, Prince DA. Prolonged prophylactic effects of gabapentin on status epilepticus-induced neocortical injury. Neurobiol Dis 2020; 142:104949. [PMID: 32442680 PMCID: PMC8083016 DOI: 10.1016/j.nbd.2020.104949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/17/2020] [Accepted: 05/14/2020] [Indexed: 01/21/2023] Open
Abstract
Long-term consequences of status epilepticus (SE) occur in a significant proportion of those who survive the acute episode. We developed an in vivo model of acute focal neocortical SE (FSE) to study long-term effects on local cortical structure and function and potential strategies to mitigate adverse consequences of SE. An acute 2 h episode of FSE was induced in anesthetized mice by epidural application of gabazine +4-aminopyridine over sensorimotor neocortex. Ten and 30 days later, the morphological and functional consequences of this single episode of FSE were studied using immunocytochemical and electrophysiological techniques. Results, focused on cortical layer V, showed astrogliosis, microgliosis, decreased neuronal density, and increased excitatory synapses, along with increased immunoreactivity for thrombospondin 2 (TSP2) and α2δ-1 proteins. In addition, neocortical slices, obtained from the area of prior focal seizure activity, showed abnormal epileptiform burst discharges along with increases in the frequency of miniature and spontaneous excitatory postsynaptic currents in layer V pyramidal cells, together with decreases in both parvalbumin immunoreactivity (PV-IR) and the frequency of miniature inhibitory postsynaptic currents in layer V pyramidal cells. Treatment with an approved drug, gabapentin (GBP) (ip 100 mg/kg/day 3 × /day for 7 days following the FSE episode), prevented the gliosis, the enhanced TSP2- and α2δ-1- IR and the increased excitatory synaptic density in the affected neocortex. This model provides an approach for assessing adverse effects of FSE on neocortical structure and function and potential prophylactic treatments.
Collapse
Affiliation(s)
- Maria-Belen Perez-Ramirez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Feng Gu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David A Prince
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
30
|
Veersema TJ, Neef A, Scheppingen J, Ferrier CH, Eijsden P, Gosselaar PH, Rijen PC, Spliet WG, Braun KP, Mühlebner A, Aronica E. Changes in vascular density in resected tissue of 97 patients with mild malformation of cortical development, focal cortical dysplasia or TSC‐related cortical tubers. Int J Dev Neurosci 2019; 79:96-104. [DOI: 10.1016/j.ijdevneu.2019.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/13/2019] [Accepted: 11/22/2019] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tim J. Veersema
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Andrew Neef
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Jackelien Scheppingen
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Cyrille H. Ferrier
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Pieter Eijsden
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Peter H. Gosselaar
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Peter C. Rijen
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Wim G.M. Spliet
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Kees P.J. Braun
- Department of Neurology and NeurosurgeryUMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Angelika Mühlebner
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Eleonora Aronica
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN)The Netherlands
| |
Collapse
|
31
|
CDDO-Me Attenuates Vasogenic Edema and Astroglial Death by Regulating NF-κB p65 Phosphorylations and Nrf2 Expression Following Status Epilepticus. Int J Mol Sci 2019; 20:ijms20194862. [PMID: 31574956 PMCID: PMC6801369 DOI: 10.3390/ijms20194862] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022] Open
Abstract
2-Cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) is a triterpenoid analogue of oleanolic acid that has anti-inflammatory, antioxidant, and neuroprotective activities. In the present study, we evaluate the effects of CDDO-Me on serum extravasation and astroglial death in the rat piriform cortex (PC) induced by status epilepticus (a prolonged seizure activity, SE) in order to propose an underlying pharmacological mechanism of CDDO-Me and its availability for treatment of vasogenic edema. CDDO-Me effectively mitigated serum extravasation and a massive astroglial loss in the PC following SE. CDDO-Me abrogated tumor necrosis factor-α (TNF-α) synthesis in activated microglia by inhibiting nuclear factor-κB (NF-κB) p65 serine 276 phosphorylation. CDDO-Me also abolished NF-κB threonine 435 phosphorylation in endothelial cells and TNF-α-mediated-phosphatidylinositol-3-kinase (PI3K)/AKT/endothelial nitric oxide synthase (eNOS) signaling cascades, which trigger vasogenic edema following SE. Furthermore, CDDO-Me increased astroglial viability via the up-regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) expression. Therefore, our findings suggest that CDDO-Me may ameliorate SE-induced vasogenic edema formation by regulating NF-κB p65 phosphorylations in microglia as well as endothelial cells and enhancing Nrf2 expression in astrocytes, respectively.
Collapse
|
32
|
Chen Y, Zhu G, Shi L, Liu D, Zhang X, Liu Y, Yuan T, Du T, Zhang J. Establishment of a novel mesial temporal lobe epilepsy rhesus monkey model via intra-hippocampal and intra-amygdala kainic acid injection assisted by neurosurgical robot system. Brain Res Bull 2019; 149:32-41. [DOI: 10.1016/j.brainresbull.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/26/2019] [Accepted: 04/02/2019] [Indexed: 01/11/2023]
|
33
|
Park H, Choi SH, Kong MJ, Kang TC. Dysfunction of 67-kDa Laminin Receptor Disrupts BBB Integrity via Impaired Dystrophin/AQP4 Complex and p38 MAPK/VEGF Activation Following Status Epilepticus. Front Cell Neurosci 2019; 13:236. [PMID: 31178701 PMCID: PMC6542995 DOI: 10.3389/fncel.2019.00236] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 05/09/2019] [Indexed: 12/21/2022] Open
Abstract
Status epilepticus (SE, a prolonged seizure activity) impairs brain-blood barrier (BBB) integrity, which results in secondary complications following SE. The non-integrin 67-kDa laminin receptor (67-kDa LR) plays a role in cell adherence to laminin (a major glycoprotein component in basement membrane), and participates laminin-mediated signaling pathways including p38 mitogen-activated protein kinase (p38 MAPK). Thus, we investigated the role of 67-kDa LR in SE-induced vasogenic edema formation in the rat piriform cortex (PC). SE diminished 67-kDa LR expression, but increased laminin expression, in endothelial cells accompanied by the reduced SMI-71 (a rat BBB barrier marker) expression. Astroglial 67-kDa LR expression was also reduced in the PC due to massive astroglial loss. 67-kDa LR neutralization led to serum extravasation in the PC concomitant with the reduced SMI-71 expression. 67-kDa LR neutralization also decreased expressions of dystrophin and aquaporin-4 (AQP4). In addition, it increased p38 MAPK phosphorylation and expressions of vascular endothelial growth factor (VEGF), laminin and endothelial nitric oxide synthase (eNOS), which were abrogated by SB202190, a p38 MAPK inhibitor. Therefore, our findings indicate that 67-kDa LR dysfunction may disrupt dystrophin-AQP4 complex, which would evoke vasogenic edema formation and subsequent laminin over-expression via activating p38 MAPK/VEGF axis.
Collapse
Affiliation(s)
- Hana Park
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Seo-Hyeon Choi
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Min-Jeong Kong
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, South Korea.,Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
34
|
Lan YL, Wang X, Zou YJ, Xing JS, Lou JC, Zou S, Ma BB, Ding Y, Zhang B. Bazedoxifene protects cerebral autoregulation after traumatic brain injury and attenuates impairments in blood-brain barrier damage: involvement of anti-inflammatory pathways by blocking MAPK signaling. Inflamm Res 2019; 68:311-323. [PMID: 30706110 DOI: 10.1007/s00011-019-01217-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/04/2019] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Traumatic brain injury (TBI) is a significant cause of death and long-term deficits in motor and cognitive functions for which there are currently no effective chemotherapeutic drugs. Bazedoxifene (BZA) is a third-generation selective estrogen receptor modulator (SERM) and has been investigated as a treatment for postmenopausal osteoporosis. It is generally safe and well tolerated, with favorable endometrial and breast safety profiles. Recent findings have shown that SERMs may have therapeutic benefits; however, the role of BZA in the treatment of TBI and its molecular and cellular mechanisms remain poorly understood. The aim of the present study was to examine the neuroprotective effects of BZA on early TBI in rats and to explore the underlying mechanisms of these effects. MATERIALS AND METHODS TBI was induced using a modified weight-drop method. Neurological deficits were evaluated according to the neurological severity score (NSS). Morris water maze and open-field behavioral tests were used to test cognitive functions. Brain edema was measured by brain water content, and impairments in the blood-brain barrier (BBB) were evaluated by expression analysis of tight junction-associated proteins, such as occludin and zonula occludens-1 (ZO-1). Neuronal injury was assessed by hematoxylin and eosin (H&E) staining. LC-MS/MS analysis was performed to determine the ability of BZA to cross the BBB. RESULTS Our results indicated that BZA attenuated the impaired cognitive functions and the increased BBB permeability of rats subjected to TBI through activation of inflammatory cascades. In vivo experiments further revealed that BZA provided this neuroprotection by suppressing TBI-induced activation of the MAPK/NF-κB signaling pathway. Thus, mechanically, the anti-inflammatory effects of BZA in TBI may be partially mediated by blocking the MAPK signaling pathway. CONCLUSIONS These findings suggest that BZA might attenuate neurological deficits and BBB damage to protect against TBI by blocking the MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China.,Department of Pharmacy, Dalian Medical University, Dalian, 116044, China.,Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Xun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China
| | - Yu-Jie Zou
- Department of Nursing, The First Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Jin-Shan Xing
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China
| | - Jia-Cheng Lou
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China
| | - Shuang Zou
- Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Bin-Bin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China
| | - Yan Ding
- Department of Pediatrics, Children's Hospital of Boston, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China. .,Department of Neurosurgery, Shenzhen People's Hospital, Shenzhen, China.
| |
Collapse
|
35
|
Li TZ, Deng H, Liu Q, Xia YZ, Darwazeh R, Yan Y. Protease-activated receptor-2 regulates glial scar formation via JNK signaling. Physiol Res 2019; 68:305-316. [PMID: 30628825 DOI: 10.33549/physiolres.933908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The study aimed to determine the effects of protease-activated receptor-2 (PAR-2) on glial scar formation after spinal cord injury (SCI) in Sprague-Dawley (SD) rats and the underlying mechanisms. Rivlin and Tator's acute extradural clip compression injury (CCI) model of severe SCI was established in this study. Animals were divided into four groups: 1) sham group (laminectomy only); 2) model group, treated with normal saline; 3) PAR-2 inhibitor group; 4) PAR-2 activator group. Enhanced GFAP and vimentin expression were the markers of glial scar formation. To determine whether JNK was involved in the effects of PAR-2 on GFAP and vimentin expression, we administered anisomycin (a JNK activator) in the presence of PAR-2 inhibitor and SP600125 (a JNK inhibitor) in the presence of PAR-2 activator. At 1, 7, 14 and 28 day after SCI, Basso, Beattie, and Bresnahan (BBB) locomotor score test was used to assess the locomotor functional recovery; immunofluorescence and western blot analysis were used to assess the expression level of GFAP, vimentin and p-JNK. Double immunofluorescence staining with GFAP and tubulin beta was used to assess the glial scar formation and the remaining neurons. Results suggested that PAR-2 is involved in glial scar formation and reduces neurons residues which can cause a further worsening in the functional outcomes after SCI via JNK signaling. Therefore, it may be effective to target PAR-2 in the treatment of SCI.
Collapse
Affiliation(s)
- Tian-Zun Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | | | | | | | | | | |
Collapse
|
36
|
Liang J, Wu Y, Yuan H, Yang Y, Xiong Q, Liang C, Li Z, Li C, Zhang G, Lai X, Hu Y, Hou S. Dendrobium officinale polysaccharides attenuate learning and memory disabilities via anti-oxidant and anti-inflammatory actions. Int J Biol Macromol 2018; 126:414-426. [PMID: 30593810 DOI: 10.1016/j.ijbiomac.2018.12.230] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/14/2018] [Accepted: 12/22/2018] [Indexed: 12/29/2022]
Abstract
The aim of this study was to explore the therapeutic effect and underling mechanism of Dendrobium officinale polysaccharides (DOPS) on two well-established animal models of learning and memory disabilities. Model of estrogen deficiency caused learning and memory disability can be induced by ovariectomy in mice, and mice were injected subcutaneously with d-galactose, which can also cause cognitive decline. H&E staining and Nissl staining were employed to confirm the protective effect of DOPS on hippocampal neuron. Morris water maze test, biochemical analysis, immunohistochemistry and immunofluorescence assay were used to study the effect and underlying mechanism of DOPS on two different learning and memory impairment models. Administration of DOPS significantly improved learning and memory disability in both models. Further studies showed that DOPS could attenuate oxidative stress and reduce neuro-inflammation via up-regulating expressions of Nrf2/HO-1 pathway and inhibiting activation of astrocytes and microglia in ovariectomy- and d-galactose-induced cognitive decline. These findings suggest that DOPS have an appreciable therapeutic effect on learning and memory disabilities and its mechanism may be related to activate Nrf2/HO-1 pathway to reduce oxidative stress and neuro-inflammation.
Collapse
Affiliation(s)
- Jian Liang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yanfang Wu
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Han Yuan
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Yiqi Yang
- The second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Qingping Xiong
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Chuyan Liang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510080, Guangdong, China
| | - Zhimeng Li
- The Fifth People's Hospital of Tangshan, Tangshan 063004, Hebei, PR China
| | - Cantao Li
- School of pharmaceutical sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Guifang Zhang
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China
| | - Xiaoping Lai
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China.
| | - Youdong Hu
- Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223002, Jiangsu, PR China.
| | - Shaozhen Hou
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, and Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China; School of pharmaceutical sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
37
|
Wellman SM, Cambi F, Kozai TD. The role of oligodendrocytes and their progenitors on neural interface technology: A novel perspective on tissue regeneration and repair. Biomaterials 2018; 183:200-217. [PMID: 30172245 PMCID: PMC6469877 DOI: 10.1016/j.biomaterials.2018.08.046] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
Oligodendrocytes and their precursors are critical glial facilitators of neurophysiology, which is responsible for cognition and behavior. Devices that are used to interface with the brain allow for a more in-depth analysis of how neurons and these glia synergistically modulate brain activity. As projected by the BRAIN Initiative, technologies that acquire a high resolution and robust sampling of neural signals can provide a greater insight in both the healthy and diseased brain and support novel discoveries previously unobtainable with the current state of the art. However, a complex series of inflammatory events triggered during device insertion impede the potential applications of implanted biosensors. Characterizing the biological mechanisms responsible for the degradation of intracortical device performance will guide novel biomaterial and tissue regenerative approaches to rehabilitate the brain following injury. Glial subtypes which assist with neuronal survival and exchange of electrical signals, mainly oligodendrocytes, their precursors, and the insulating myelin membranes they produce, are sensitive to inflammation commonly induced from insults to the brain. This review explores essential physiological roles facilitated by oligodendroglia and their precursors and provides insight into their pathology following neurodegenerative injury and disease. From this knowledge, inferences can be made about the impact of device implantation on these supportive glia in order to engineer effective strategies that can attenuate their responses, enhance the efficacy of neural interfacing technology, and provide a greater understanding of the challenges that impede wound healing and tissue regeneration during pathology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, USA
| | - Takashi Dy Kozai
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, USA; NeuroTech Center, University of Pittsburgh Brain Institute, USA.
| |
Collapse
|
38
|
Amengual-Gual M, Sánchez Fernández I, Wainwright MS. Novel drugs and early polypharmacotherapy in status epilepticus. Seizure 2018; 68:79-88. [PMID: 30473267 DOI: 10.1016/j.seizure.2018.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/05/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Rescue medications for status epilepticus (SE) have a relatively high rate of failure. The purpose of this review is to summarize the evidence for the efficacy of novel drugs and early polypharmacotherapy for SE. METHOD Literature review. RESULTS New drugs and treatment strategies aim to target the pathophysiology of SE in order to improve seizure control and outcomes. Changes at the synapse level during SE include a progressive decrease in synaptic GABAA receptors and increase in synaptic NMDA receptors. These changes tend to promote self-sustaining seizures. Current SE guidelines recommend a rapid stepwise treatment using benzodiazepines in monotherapy as the first-line treatment, targeting GABAA synaptic receptors. Novel treatment approaches target GABAA synaptic and extrasynaptic receptors with allopregnanolone, and NMDA receptors with ketamine. Novel rescue treatments used for SE include topiramate, brivaracetam, and perampanel, which are already marketed in epilepsy. Some available drugs not marketed for use in epilepsy have been used in the treatment of SE, and other agents are being studied for this purpose. Early polytherapy, most frequently combining a benzodiazepine with a second-line drug or an NMDA receptor antagonist, might potentially increase seizure control with relatively minor increase in side effects. Although many preclinical studies support novel drugs and early polytherapy in SE, human studies are scarce and inconclusive. Currently, evidence is lacking to recommend specific combinations of these new agents. CONCLUSIONS Novel drugs and strategies target the underlying pathophysiology of SE with the intent to improve seizure control and outcomes.
Collapse
Affiliation(s)
- Marta Amengual-Gual
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Son Espases, Universitat de les Illes Balears, Palma, Spain.
| | - Iván Sánchez Fernández
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Child Neurology, Hospital Sant Joan de Déu, Universidad de Barcelona, Spain
| | - Mark S Wainwright
- Department of Neurology, Division of Pediatric Neurology. University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
39
|
Shijo T, Warita H, Suzuki N, Ikeda K, Mitsuzawa S, Akiyama T, Ono H, Nishiyama A, Izumi R, Kitajima Y, Aoki M. Antagonizing bone morphogenetic protein 4 attenuates disease progression in a rat model of amyotrophic lateral sclerosis. Exp Neurol 2018; 307:164-179. [PMID: 29932880 DOI: 10.1016/j.expneurol.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 05/25/2018] [Accepted: 06/15/2018] [Indexed: 12/20/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset, fatal neurodegenerative syndrome characterized by the systemic loss of motor neurons with prominent astrocytosis and microgliosis in the spinal cord and brain. Astrocytes play an essential role in maintaining extracellular microenvironments that surround motor neurons, and are activated by various insults. Growing evidence points to a non-cell autonomous neurotoxicity caused by chronic and sustained astrocytic activation in patients with neurodegenerative diseases, including ALS. However, the mechanisms that underlie the harmful effects of astrocytosis in patients with ALS remain unresolved. We focused on bone morphogenetic proteins as a major soluble factor that promotes astrocytogenesis and its activation in the adult spinal cord. In a transgenic rat model with ALS-linked mutant Cu/Zn superoxide dismutase gene, BMP4 was progressively up-regulated in reactive astrocytes of the spinal ventral horns, whereas the BMP-antagonist noggin was decreased in association with neuronal degeneration. Continuous intrathecal noggin supplementation after disease onset significantly ameliorated motor dysfunction symptoms, neurogenic muscle atrophy, and extended survival of symptomatic ALS model rats, despite lack of deterrence against neuronal death itself. The exogenous noggin inhibited astrocytic hypertrophy, astrocytogenesis, and neuroinflammation by inactivating both Smad1/5/8 and p38 mitogen-activated protein kinase pathways. Moreover, intrathecal infusion of a Bmp4-targeted antisense oligonucleotides and provided selective Bmp4 knockdown in vivo, which suppressed astrocyte and microglia activation, reproducing the aforementioned results by noggin treatment. Collectively, we clarified the involvement of BMP4 in the processes of excessive gliosis that exacerbate the disease progression of the ALS model rats. Our study demonstrated that BMP4, with its downstream signaling, might be a novel therapeutic target for disease-modifying therapies in ALS.
Collapse
Affiliation(s)
- Tomomi Shijo
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Hitoshi Warita
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Naoki Suzuki
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Kensuke Ikeda
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Shio Mitsuzawa
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Tetsuya Akiyama
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Hiroya Ono
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Ayumi Nishiyama
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Rumiko Izumi
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Yasuo Kitajima
- Department of Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Masashi Aoki
- Department of Neurology, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| |
Collapse
|
40
|
Hikmat O, Naess K, Engvall M, Klingenberg C, Rasmussen M, Tallaksen CME, Brodtkorb E, Fiskerstrand T, Isohanni P, Uusimaa J, Darin N, Rahman S, Bindoff LA. Elevated cerebrospinal fluid protein inPOLG-related epilepsy: Diagnostic and prognostic implications. Epilepsia 2018; 59:1595-1602. [DOI: 10.1111/epi.14459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2018] [Indexed: 12/01/2022]
Affiliation(s)
- Omar Hikmat
- Department of Pediatrics; Haukeland University Hospital; Bergen Norway
- Department of Clinical Medicine (K1); University of Bergen; Bergen Norway
| | - Karin Naess
- Center for Inherited Metabolic Diseases; Karolinska University Hospital; Stockholm Sweden
- Department of Medical Biochemistry and Biophysics; Karolinska Institute; Stockholm Sweden
| | - Martin Engvall
- Center for Inherited Metabolic Diseases; Karolinska University Hospital; Stockholm Sweden
- Department of Molecular Medicine and Surgery; Karolinska Institute; Stockholm Sweden
| | - Claus Klingenberg
- Department of Pediatric and Adolescent Medicine; University Hospital of North Norway; Tromso Norway
- Pediatric Research Group; Department of Clinical Medicine; UiT-Arctic University of Norway; Tromso Norway
| | - Magnhild Rasmussen
- Women and Children's Division; Department of Clinical Neurosciences for Children; Oslo University Hospital; Oslo Norway
- Unit for Congenital and Hereditary Neuromuscular Disorders; Department of Neurology; Oslo University Hospital; Oslo Norway
| | - Chantal M. E. Tallaksen
- Department of Neurology; Oslo University Hospital; Oslo Norway
- Institute of Clinical Medicine; Faculty of Medicine; University of Oslo; Oslo Norway
| | - Eylert Brodtkorb
- Department of Neuroscience; Norwegian University of Science and Technology; Trondheim Norway
- Department of Neurology and Clinical Neurophysiology; St. Olav's University Hospital; Trondheim Norway
| | - Torunn Fiskerstrand
- Department of Medical Genetics and Molecular Medicine; Haukeland University Hospital; Bergen Norway
- Department of Clinical Science (K2); University of Bergen; Bergen Norway
| | - Pirjo Isohanni
- Department of Pediatric Neurology; Children's Hospital; University of Helsinki and Helsinki University Hospital; Helsinki Finland
- Research Programs Unit; Molecular Neurology; Biomedicum Helsinki; University of Helsinki; Helsinki Finland
| | - Johanna Uusimaa
- PEDEGO Research Unit and Biocenter Oulu; University of Oulu; Oulu Finland
- Department of Children and Adolescents; Medical Research Center; Oulu University Hospital; Oulu Finland
| | - Niklas Darin
- Department of Pediatrics; Queen Silvia Children's Hospital; University of Gothenburg; Gothenburg Sweden
| | - Shamima Rahman
- Mitochondrial Research Group; University College London Great Ormond Street Institute of Child Health; London UK
- Metabolic Unit; Great Ormond Street Hospital for Children; National Health Service Foundation Trust; London UK
| | - Laurence A. Bindoff
- Department of Clinical Medicine (K1); University of Bergen; Bergen Norway
- Department of Neurology; Haukeland University Hospital; Bergen Norway
| |
Collapse
|
41
|
Affiliation(s)
- Yongli He
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyou Cai
- Department of Neurology, Chongqing General Hospital, Chongqing, China
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Wellman SM, Kozai TDY. In vivo spatiotemporal dynamics of NG2 glia activity caused by neural electrode implantation. Biomaterials 2018; 164:121-133. [PMID: 29501892 PMCID: PMC5951685 DOI: 10.1016/j.biomaterials.2018.02.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023]
Abstract
Neural interface technology provides direct sampling and analysis of electrical and chemical events in the brain in order to better understand neuronal function and treat neurodegenerative disease. However, intracortical electrodes experience inflammatory reactions that reduce long-term stability and functionality and are understood to be facilitated by activated microglia and astrocytes. Emerging studies have identified another cell type that participates in the formation of a high-impedance glial scar following brain injury; the oligodendrocyte precursor cell (OPC). These cells maintain functional synapses with neurons and are a crucial source of neurotrophic support. Following injury, OPCs migrate toward areas of tissue injury over the course of days, similar to activated microglia. The delayed time course implicates these OPCs as key components in the formation of the outer layers of the glial scar around the implant. In vivo two-photon laser scanning microscopy (TPLSM) was employed to observe fluorescently-labeled OPC and microglia reactivity up to 72 h following probe insertion. OPCs initiated extension of cellular processes (2.5 ± 0.4 μm h-1) and cell body migration (1.6 ± 0.3 μm h-1) toward the probe beginning 12 h after insertion. By 72 h, OPCs became activated at a radius of about 190.3 μm away from the probe surface. This study characterized the early spatiotemporal dynamics of OPCs involved in the inflammatory response induced by microelectrode insertion. OPCs are key mediators of tissue health and are understood to have multiple fate potentials. Detailed spatiotemporal characterization of glial behavior under pathological conditions may allow identification of alternative intervention targets for mitigating the formation of a glial scar and subsequent neurodegeneration that debilitates chronic neural interfaces.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, United States; Center for the Basis of Neural Cognition, United States
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, United States; Center for the Basis of Neural Cognition, United States; Center for Neuroscience, University of Pittsburgh, United States; McGowan Institute of Regenerative Medicine, University of Pittsburgh, United States; NeuroTech Center, University of Pittsburgh Brain Institute, United States.
| |
Collapse
|
43
|
Abstract
The success of naturalistic or therapeutic neuroregeneration likely depends on an internal milieu that facilitates the survival, proliferation, migration, and differentiation of stem cells and their assimilation into neural networks. Migraine attacks are an integrated sequence of physiological processes that may protect the brain from oxidative stress by releasing growth factors, suppressing apoptosis, stimulating neurogenesis, encouraging mitochondrial biogenesis, reducing the production of oxidants, and upregulating antioxidant defenses. Thus, the migraine attack may constitute a physiologic environment conducive to stem cells. In this paper, key components of migraine are reviewed – neurogenic inflammation with release of calcitonin gene-related peptide (CGRP) and substance P, plasma protein extravasation, platelet activation, release of serotonin by platelets and likely by the dorsal raphe nucleus, activation of endothelial nitric oxide synthase (eNOS), production of brain-derived neurotrophic factor (BDNF) and, in migraine aura, cortical spreading depression – along with their potential neurorestorative aspects. The possibility is considered of using these components to facilitate successful stem cell transplantation. Potential methods for doing so are discussed, including chemical stimulation of the TRPA1 ion channel, conjoint activation of a subset of migraine components, invasive and noninvasive deep brain stimulation of the dorsal raphe nucleus, transcranial focused ultrasound, and stimulation of the Zusanli (ST36) acupuncture point.
Collapse
Affiliation(s)
- Jonathan M Borkum
- Department of Psychology, University of Maine, Orono; Health Psych Maine, Waterville, ME, USA
| |
Collapse
|
44
|
Chen YC, Zhu GY, Wang X, Shi L, Du TT, Liu DF, Liu YY, Jiang Y, Zhang X, Zhang JG. Anterior thalamic nuclei deep brain stimulation reduces disruption of the blood-brain barrier, albumin extravasation, inflammation and apoptosis in kainic acid-induced epileptic rats. Neurol Res 2017; 39:1103-1113. [PMID: 28918702 DOI: 10.1080/01616412.2017.1379241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 09/08/2017] [Indexed: 12/15/2022]
Abstract
Objective The therapeutic efficacy of anterior thalamic nuclei deep brain stimulation (ATN-DBS) against seizures has been largely accepted; however, the effects of ATN-DBS on disruption of the blood-brain barrier (BBB), albumin extravasation, inflammation and apoptosis still remain unclear. Methods Rats were distributed into four treatment groups: physiological saline (PS, N = 12), kainic acid (KA, N = 12), KA-sham-DBS (N = 12) and KA-DBS (N = 12). Seizures were monitored using video-electroencephalogram (EEG). One day after surgery, all rats were sacrificed. Then, samples were prepared for quantitative real-time PCR (qPCR), western blot, immunofluorescence (IF) staining, and transmission electron microscopy to evaluate the disruption of the BBB, albumin extravasation, inflammation, and apoptosis. Result Because of the KA injection, the disruption of the BBB, albumin extravasation, inflammation and apoptosis were more severe in the KA and the KA-sham-DBS groups compared to the PS group (all Ps < 0.05 or < 0.01). The ideal outcomes were observed in the KA-DBS group. ATN-DBS produced a 46.3% reduction in seizure frequency and alleviated the disruption of the BBB, albumin extravasation, inflammatory reaction and apoptosis in comparison to the KA-sham-DBS group (all Ps < 0.05 or < 0.01). Conclusion (1) Seizures can be reduced using ATN-DBS in the epileptogenic stage. (2) ATN-DBS can reduce the disruption of the BBB and albumin extravasation. (3) ATN-DBS has an anti-inflammatory effect in epileptic models.
Collapse
Affiliation(s)
- Ying-Chuan Chen
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Guan-Yu Zhu
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Xiu Wang
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Lin Shi
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Ting-Ting Du
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - De-Feng Liu
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Yu-Ye Liu
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
| | - Yin Jiang
- b Department of Functional Neurosurgery , Beijing Neurosurgical Institute, Capital Medical University , Beijing , China
| | - Xin Zhang
- b Department of Functional Neurosurgery , Beijing Neurosurgical Institute, Capital Medical University , Beijing , China
| | - Jian-Guo Zhang
- a Department of Neurosurgery , Beijing Tiantan Hospital, Capital Medical University , Beijing , China
- b Department of Functional Neurosurgery , Beijing Neurosurgical Institute, Capital Medical University , Beijing , China
- c Beijing Key Laboratory of Neurostimulation , Beijing , China
| |
Collapse
|
45
|
Scheiblich H, Schlütter A, Golenbock DT, Latz E, Martinez-Martinez P, Heneka MT. Activation of the NLRP3 inflammasome in microglia: the role of ceramide. J Neurochem 2017; 143:534-550. [DOI: 10.1111/jnc.14225] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Hannah Scheiblich
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology; University of Bonn - Medical Center; Bonn Germany
| | - Anna Schlütter
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology; University of Bonn - Medical Center; Bonn Germany
- Department of Neuroscience; Maastricht University; Maastricht The Netherlands
| | - Douglas T. Golenbock
- Department of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester Massachusetts USA
| | - Eicke Latz
- Department of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester Massachusetts USA
- Institute of Innate Immunity; University of Bonn; Bonn Germany
- German Center for Neurodegenerative Diseases (DZNE); Bonn Germany
| | - Pilar Martinez-Martinez
- Department of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester Massachusetts USA
| | - Michael T. Heneka
- Department of Neurodegenerative Disease and Gerontopsychiatry/Neurology; University of Bonn - Medical Center; Bonn Germany
- Department of Infectious Diseases and Immunology; University of Massachusetts Medical School; Worcester Massachusetts USA
- German Center for Neurodegenerative Diseases (DZNE); Bonn Germany
| |
Collapse
|
46
|
Borkum JM. The Migraine Attack as a Homeostatic, Neuroprotective Response to Brain Oxidative Stress: Preliminary Evidence for a Theory. Headache 2017; 58:118-135. [DOI: 10.1111/head.13214] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Jonathan M. Borkum
- Department of Psychology; University of Maine; Orono ME USA
- Health Psych Maine; Waterville ME USA
| |
Collapse
|
47
|
Cold Environment Exacerbates Brain Pathology and Oxidative Stress Following Traumatic Brain Injuries: Potential Therapeutic Effects of Nanowired Antioxidant Compound H-290/51. Mol Neurobiol 2017; 55:276-285. [DOI: 10.1007/s12035-017-0740-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
48
|
Stokum JA, Keledjian K, Hayman E, Karimy JK, Pampori A, Imran Z, Woo SK, Gerzanich V, Simard JM. Glibenclamide pretreatment protects against chronic memory dysfunction and glial activation in rat cranial blast traumatic brain injury. Behav Brain Res 2017; 333:43-53. [PMID: 28662892 DOI: 10.1016/j.bbr.2017.06.038] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/20/2017] [Accepted: 06/24/2017] [Indexed: 02/03/2023]
Abstract
Blast traumatic brain injury (bTBI) affects both military and civilian populations, and often results in chronic deficits in cognition and memory. Chronic glial activation after bTBI has been linked with cognitive decline. Pharmacological inhibition of sulfonylurea receptor 1 (SUR1) with glibenclamide was shown previously to reduce glial activation and improve cognition in contusive models of CNS trauma, but has not been examined in bTBI. We postulated that glibenclamide would reduce chronic glial activation and improve long-term memory function after bTBI. Using a rat direct cranial model of bTBI (dc-bTBI), we evaluated the efficacy of two glibenclamide treatment paradigms: glibenclamide prophylaxis (pre-treatment), and treatment with glibenclamide starting after dc-bTBI (post-treatment). Our results show that dc-bTBI caused hippocampal astrocyte and microglial/macrophage activation that was associated with hippocampal memory dysfunction (rapid place learning paradigm) at 28days, and that glibenclamide pre-treatment, but not post-treatment, effectively protected against glial activation and memory dysfunction. We also report that a brief transient time-window of blood-brain barrier (BBB) disruption occurs after dc-bTBI, and we speculate that glibenclamide, which is mostly protein bound and does not normally traverse the intact BBB, can undergo CNS delivery only during this brief transient opening of the BBB. Together, our findings indicate that prophylactic glibenclamide treatment may help to protect against chronic cognitive sequelae of bTBI in warfighters and other at-risk populations.
Collapse
Affiliation(s)
- Jesse A Stokum
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA.
| | - Kaspar Keledjian
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Erik Hayman
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Jason K Karimy
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Adam Pampori
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Ziyan Imran
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Seung Kyoon Woo
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - Volodymyr Gerzanich
- Departments of Neurosurgery, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| | - J Marc Simard
- Departments of Pathology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA; Departments of Physiology, University of Maryland School of Medicine, 10 S Pine St, MSTF, Room 634B, Baltimore, MD 21201, USA
| |
Collapse
|
49
|
Abstract
Access to the CNS and delivery of therapeutics across the blood-brain barrier remains a challenge for most treatments of major neurological diseases such as AD or PD. Focused ultrasound represents a potential approach for overcoming these barriers to treating AD and PD and perhaps other neurological diseases. Ultrasound (US) is best known for its imaging capabilities of organs in the periphery, but various arrangements of the transducers producing the acoustic signal allow the energy to be precisely focused (F) within the skull. Using FUS in combination with MRI and contrast agents further enhances accuracy by providing clear information on location. Varying the acoustic power allows FUS to be used in applications ranging from imaging, stimulation of brain circuits, to ablation of tissue. In several transgenic mouse models of AD, the use of FUS with microbubbles reduces plaque load and improves cognition and suggests the need to investigate this technology for plaque removal in AD. In PD, FUS is being explored as a way to non-invasively ablate the brain areas responsible for the tremor and dyskinesia associated with the disease, but has yet to be utilized for non-invasive delivery of putative therapeutics. The FUS approach also greatly increases the range of possible CNS therapeutics as it overcomes the issues of BBB penetration. In this review we discuss how the characteristics and various applications of FUS may advance the therapeutics available for treating or preventing neurodegenerative disorders with an emphasis on treating AD and PD.
Collapse
Affiliation(s)
- Diane B Miller
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505.
| | - James P O'Callaghan
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health, Morgantown, WV 26505.
| |
Collapse
|
50
|
Corrigan F, Mander KA, Leonard AV, Vink R. Neurogenic inflammation after traumatic brain injury and its potentiation of classical inflammation. J Neuroinflammation 2016; 13:264. [PMID: 27724914 PMCID: PMC5057243 DOI: 10.1186/s12974-016-0738-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/28/2016] [Indexed: 01/05/2023] Open
Abstract
Background The neuroinflammatory response following traumatic brain injury (TBI) is known to be a key secondary injury factor that can drive ongoing neuronal injury. Despite this, treatments that have targeted aspects of the inflammatory pathway have not shown significant efficacy in clinical trials. Main body We suggest that this may be because classical inflammation only represents part of the story, with activation of neurogenic inflammation potentially one of the key initiating inflammatory events following TBI. Indeed, evidence suggests that the transient receptor potential cation channels (TRP channels), TRPV1 and TRPA1, are polymodal receptors that are activated by a variety of stimuli associated with TBI, including mechanical shear stress, leading to the release of neuropeptides such as substance P (SP). SP augments many aspects of the classical inflammatory response via activation of microglia and astrocytes, degranulation of mast cells, and promoting leukocyte migration. Furthermore, SP may initiate the earliest changes seen in blood-brain barrier (BBB) permeability, namely the increased transcellular transport of plasma proteins via activation of caveolae. This is in line with reports that alterations in transcellular transport are seen first following TBI, prior to decreases in expression of tight-junction proteins such as claudin-5 and occludin. Indeed, the receptor for SP, the tachykinin NK1 receptor, is found in caveolae and its activation following TBI may allow influx of albumin and other plasma proteins which directly augment the inflammatory response by activating astrocytes and microglia. Conclusions As such, the neurogenic inflammatory response can exacerbate classical inflammation via a positive feedback loop, with classical inflammatory mediators such as bradykinin and prostaglandins then further stimulating TRP receptors. Accordingly, complete inhibition of neuroinflammation following TBI may require the inhibition of both classical and neurogenic inflammatory pathways.
Collapse
Affiliation(s)
- Frances Corrigan
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Kimberley A Mander
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anna V Leonard
- Adelaide Centre for Neuroscience Research, School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert Vink
- Sansom Institute for Health Research, The University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|