1
|
Badaut J, Blochet C, Obenaus A, Hirt L. Physiological and pathological roles of caveolins in the central nervous system. Trends Neurosci 2024; 47:651-664. [PMID: 38972795 PMCID: PMC11324375 DOI: 10.1016/j.tins.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 05/14/2024] [Accepted: 06/12/2024] [Indexed: 07/09/2024]
Abstract
Caveolins are a family of transmembrane proteins located in caveolae, small lipid raft invaginations of the plasma membrane. The roles of caveolin-enriched lipid rafts are diverse, and include mechano-protection, lipid homeostasis, metabolism, transport, and cell signaling. Caveolin-1 (Cav-1) and other caveolins were described in endothelial cells and later in other cell types of the central nervous system (CNS), including neurons, astrocytes, oligodendrocytes, microglia, and pericytes. This pancellular presence of caveolins demands a better understanding of their functional roles in each cell type. In this review we describe the various functions of Cav-1 in the cells of normal and pathological brains. Several emerging preclinical findings suggest that Cav-1 could represent a potential therapeutic target in brain disorders.
Collapse
Affiliation(s)
- Jérôme Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| | - Camille Blochet
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| | - André Obenaus
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA; Division of Biomedical Sciences, University of California Riverside, Riverside, CA, USA
| | - Lorenz Hirt
- Department of Clinical Neurosciences, CHUV, Lausanne, Switzerland; Department of Fundamental Neuroscience, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Clark IH, Natera D, Grande AW, Low WC. Ex vivo method for rapid quantification of post traumatic brain injury lesion volumes using ultrasound. J Neurosci Methods 2024; 407:110140. [PMID: 38663553 DOI: 10.1016/j.jneumeth.2024.110140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/02/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Studies of traumatic brain injury often involve the quantification of the lesion volume as a major outcome measure. The determination of lesion volume typically employs the cutting and mounting of brain tissue, and the calculation of the cross-sectional area of the lesion within each section of brain after histological staining. This is a time consuming and laborious task often requiring many weeks to determine the lesion volume for an individual brain. METHODS In this report we present a method for determining the lesion volume within the brain following traumatic brain injury that involves the use of ultrasound imaging. With this process the lesion volume can be determined within a time period of 90 min per brain rather than weeks and months. Moreover, we have developed a pipeline that will combine the cross-sectional ultrasound images of the brain with the Allen Mouse Brain Atlas to provide the precise anatomical structures that are affected by traumatic injury to the brain. The anatomical detail was lastly paired with behavioral data showing neurological deficits correlated with specific areas of brain injury. RESULTS The accuracy and precision of this method was shown to be highly consistent with the traditional histological approach. Additionally, the mapping process and behavioral data show that neurological recovery from 1 to 3 weeks post injury is not correlated with gross anatomical recovery of the TBI lesion in our TBI model. CONCLUSION Together these approaches will enhance the pipeline for processing brain tissue in experimental conditions where the lesion volume is an important outcome parameter and provide more high resolution information about the identity of the damaged regions of the brain.
Collapse
Affiliation(s)
- Isaac H Clark
- Biomedical Engineering Graduate Program, United States.
| | | | - Andrew W Grande
- Biomedical Engineering Graduate Program, United States; Department of Neurosurgery, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Walter C Low
- Biomedical Engineering Graduate Program, United States; Department of Neurosurgery, United States; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
3
|
Ahmed ME, Suhail H, Nematullah M, Hoda MN, Giri S, Ahmad AS. Loss of AMPK potentiates inflammation by activating the inflammasome after traumatic brain injury in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600422. [PMID: 38979231 PMCID: PMC11230198 DOI: 10.1101/2024.06.25.600422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Traumatic brain injury (TBI) is a significant public health concern characterized by a complex cascade of cellular events. TBI induces adenosine monophosphate-activated protein kinase (AMPK) dysfunction impairs energy balance activates inflammatory cytokines and leads to neuronal damage. AMPK is a key regulator of cellular energy homeostasis during inflammatory responses. Recent research has revealed its key role in modulating the inflammatory process in TBI. Following TBI the activation of AMPK can influence various important pathways and mechanisms including metabolic pathways and inflammatory signaling. Our study investigated the effects of post-TBI loss of AMPK function on functional outcomes inflammasome activation, and inflammatory cytokine production. Male C57BL/6 adult wild-type (WT) and AMPK knockout (AMPK-KO) mice were subjected to a controlled cortical impact (CCI) model of TBI or sham surgery. The mice were tested for behavioral impairment at 24 h post-TBI thereafter, mice were anesthetized, and their brains were quickly removed for histological and biochemical evaluation. In vitro we investigated inflammasome activation in mixed glial cells stimulated with lipopolysaccharides+ Interferon-gamma (LI) (0.1 μg/20 ng/ml LPS/IFNg) for 6 h to induce an inflammatory response. Estimating the nucleotide-binding domain, leucine-rich-containing family pyrin domain containing western blotting ELISA and qRT-PCR performed 3 (NLRP3) inflammasome activation and cytokine production. Our findings suggest that TBI leads to reduced AMPK phosphorylation in WT mice and that the loss of AMPK correlates with worsened behavioral deficits at 24 h post-TBI in AMPK-KO mice as compared to WT mice. Moreover compared with the WT mice AMPK-KO mice exhibit exacerbated NLRP3 inflammasome activation and increased expression of proinflammatory mediators such as IL-1b IL-6 TNF-a iNOS and Cox 2. These results align with the in vitro studies using brain glial cells under inflammatory conditions, demonstrating greater activation of inflammasome components in AMPK-KO mice than in WT mice. Our results highlighted the critical role of AMPK in TBI outcomes. We found that the absence of AMPK worsens behavioral deficits and heightens inflammasome-mediated inflammation thereby exacerbating brain injury after TBI. Restoring AMPK activity after TBI could be a promising therapeutic approach for alleviating TBI-related damage.
Collapse
Affiliation(s)
| | - Hamid Suhail
- Department of Neurology, Henry Ford Health, Detroit, MI 48202
| | | | - Md Nasrul Hoda
- Department of Neurology, Henry Ford Health, Detroit, MI 48202
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health, Detroit, MI 48202
| | | |
Collapse
|
4
|
Magdy O, Eshra M, Rashed L, Maher M, Hosny SA, ShamsEldeen AM. Amelioration of cisplatin-induced neurodegenerative changes in rats and restoration of mitochondrial biogenesis by 6-bromoindirubin-3'-oxime: The implication of the GSK-3β/PGC1-α axis. Tissue Cell 2024; 88:102393. [PMID: 38705086 DOI: 10.1016/j.tice.2024.102393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND The cognitive deficits observed after treatment with chemotherapeutic drugs are obvious clinical problems. For treating chemotherapy-induced cognitive deficits (CICD), the treatment modalities must target its underlying mechanisms. Specifically, cisplatin may activate glycogen synthase kinase-3β (GSK-3β), thereby enhancing neuronal apoptosis. 6-bromoindirubin-3'-oxime (6BIO) was not investigated previously in a model of CICD. Therefore, this investigation aimed to address the impacts of GSK3 inhibition on regulating cell signaling, which contributes to neurodegeneration and cognitive impairment. METHODS Thirty adult male Wistar rats were randomly allocated into control groups, while two experimental groups were exposed to repeated cisplatin injections (2 mg/kg intraperitoneally (ip), twice weekly, nine injections), termed chemobrain groups. The rats in the two experimental groups were equally divided into the chemobrain group (untreated) and the chemobrain-6BIO group (treated with 6BIO at a dose of 8.5 μg/kg ip every two days, started after the last dose of cisplatin and continued for two weeks). RESULTS Repeated exposure to cisplatin led to a marked decline in cognitive functions. GSK3 inhibition exerted neuroprotection by decreasing the expression of p-tau and amyloid β, thereby improving cognition. 6BIO, the GSK-3β inhibitor, restored mitochondrial biogenesis by augmenting the protein levels of PGC1-α and increasing the number of mitochondria in the cerebral cortex and hippocampus. CONCLUSION 6BIO provided neuroprotection and exhibited anti-apoptotic and anti-oxidative effects in a rat model of chemobrain.
Collapse
Affiliation(s)
- Ola Magdy
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Mohammed Eshra
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Laila Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Egypt
| | - Muhammed Maher
- Department of Physiology, Faculty of Medicine, Cairo University, Egypt
| | - Sara Adel Hosny
- Department of Histology, Faculty of Medicine, Cairo University, Egypt
| | | |
Collapse
|
5
|
Velezmoro Jauregui G, Vukić D, Onyango IG, Arias C, Novotný JS, Texlová K, Wang S, Kovačovicova KL, Polakova N, Zelinkova J, Čarna M, Lacovich V, Head BP, Havas D, Mistrik M, Zorec R, Verkhratsky A, Keegan L, O'Connell MA, Rissman R, Stokin GB. Amyloid precursor protein induces reactive astrogliosis. Acta Physiol (Oxf) 2024; 240:e14142. [PMID: 38584589 DOI: 10.1111/apha.14142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024]
Abstract
AIM Astrocytes respond to stressors by acquiring a reactive state characterized by changes in their morphology and function. Molecules underlying reactive astrogliosis, however, remain largely unknown. Given that several studies observed increase in the Amyloid Precursor Protein (APP) in reactive astrocytes, we here test whether APP plays a role in reactive astrogliosis. METHODS We investigated whether APP instigates reactive astroglios by examining in vitro and in vivo the morphology and function of naive and APP-deficient astrocytes in response to APP and well-established stressors. RESULTS Overexpression of APP in cultured astrocytes led to remodeling of the intermediate filament network, enhancement of cytokine production, and activation of cellular programs centered around the interferon (IFN) pathway, all signs of reactive astrogliosis. Conversely, APP deletion abrogated remodeling of the intermediate filament network and blunted expression of IFN-stimulated gene products in response to lipopolysaccharide. Following traumatic brain injury (TBI), mouse reactive astrocytes also exhibited an association between APP and IFN, while APP deletion curbed the increase in glial fibrillary acidic protein observed canonically in astrocytes in response to TBI. CONCLUSIONS The APP thus represents a candidate molecular inducer and regulator of reactive astrogliosis. This finding has implications for understanding pathophysiology of neurodegenerative and other diseases of the nervous system characterized by reactive astrogliosis and opens potential new therapeutic avenues targeting APP and its pathways to modulate reactive astrogliosis.
Collapse
Affiliation(s)
- Gretsen Velezmoro Jauregui
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dragana Vukić
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Faculty of Science, National Centre for Biomedical Research, Masaryk University, Brno, Czech Republic
| | - Isaac G Onyango
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Carlos Arias
- Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Jan S Novotný
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Kateřina Texlová
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesia, University of California San Diego, La Jolla, California, USA
| | | | - Natalie Polakova
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jana Zelinkova
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Maria Čarna
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Valentina Lacovich
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesia, University of California San Diego, La Jolla, California, USA
| | | | - Martin Mistrik
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Robert Zorec
- Laboratory of Neuroendocrinology, Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
- Celica Biomedical, Technology Park, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Achucarro Centre for Neuroscience, IIKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
| | - Liam Keegan
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Mary A O'Connell
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Robert Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Gorazd B Stokin
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Neurology, Gloucestershire Royal Hospital, Gloucestershire NHS Foundation Trust, Gloucester, UK
| |
Collapse
|
6
|
Chen AC, Lai SC, Lu CY, Chen KM. Exploration of the Molecular Mechanism by Which Caveolin-1 Regulates Changes in Blood-Brain Barrier Permeability Leading to Eosinophilic Meningoencephalitis. Trop Med Infect Dis 2024; 9:124. [PMID: 38922036 PMCID: PMC11209224 DOI: 10.3390/tropicalmed9060124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Angiostrongylus cantonensis, a zoonotic parasite, can invade the human central nervous system (CNS) and cause acute eosinophilic meningitis or eosinophilic meningoencephalitis. Mice infected with A. cantonensis show elevated levels of pro-inflammatory cytokines, plasminogen activators, and matrix metalloproteinase-9, resulting in disruption of the blood-brain barrier (BBB) and immune cell infiltration into the CNS. Caveolin-1 (Cav-1) regulates the permeability of the BBB, which affects immune cells and cerebrospinal fluid. This intricate interaction ultimately fuels the progression of brain damage and edema. This study aims to investigate the regulatory role of Cav-1 in the pathogenesis of meningoencephalitis induced by A. cantonensis infection. We investigated pathological alterations by triphenyl-tetrazolium chloride, brain water content, BBB permeability, Western blot analysis, and gelatin zymography in BALB/c mice after A. cantonensis. The study evaluates the critical role of Cav-1 regulation through the TLR4/MyD88 signaling pathway, modulates tight junction proteins, influences BBB permeability, and contributes to brain damage in A. cantonensis-induced meningoencephalitis.
Collapse
Affiliation(s)
- An-Chih Chen
- Department of Neurology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
| | - Shih-Chan Lai
- Department of Parasitology, Chung Shan Medical University, Taichung 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Cheng-You Lu
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 40227, Taiwan;
| | - Ke-Min Chen
- Department of Parasitology, Chung Shan Medical University, Taichung 40201, Taiwan;
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| |
Collapse
|
7
|
Clarke GJB, Follestad T, Skandsen T, Zetterberg H, Vik A, Blennow K, Olsen A, Håberg AK. Chronic immunosuppression across 12 months and high ability of acute and subacute CNS-injury biomarker concentrations to identify individuals with complicated mTBI on acute CT and MRI. J Neuroinflammation 2024; 21:109. [PMID: 38678300 PMCID: PMC11056044 DOI: 10.1186/s12974-024-03094-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/05/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Identifying individuals with intracranial injuries following mild traumatic brain injury (mTBI), i.e. complicated mTBI cases, is important for follow-up and prognostication. The main aims of our study were (1) to assess the temporal evolution of blood biomarkers of CNS injury and inflammation in individuals with complicated mTBI determined on computer tomography (CT) and magnetic resonance imaging (MRI); (2) to assess the corresponding discriminability of both single- and multi-biomarker panels, from acute to chronic phases after injury. METHODS Patients with mTBI (n = 207), defined as Glasgow Coma Scale score between 13 and 15, loss of consciousness < 30 min and post-traumatic amnesia < 24 h, were included. Complicated mTBI - i.e., presence of any traumatic intracranial injury on neuroimaging - was present in 8% (n = 16) on CT (CT+) and 12% (n = 25) on MRI (MRI+). Blood biomarkers were sampled at four timepoints following injury: admission (within 72 h), 2 weeks (± 3 days), 3 months (± 2 weeks) and 12 months (± 1 month). CNS biomarkers included were glial fibrillary acidic protein (GFAP), neurofilament light (NFL) and tau, along with 12 inflammation markers. RESULTS The most discriminative single biomarkers of traumatic intracranial injury were GFAP at admission (CT+: AUC = 0.78; MRI+: AUC = 0.82), and NFL at 2 weeks (CT+: AUC = 0.81; MRI+: AUC = 0.89) and 3 months (MRI+: AUC = 0.86). MIP-1β and IP-10 concentrations were significantly lower across follow-up period in individuals who were CT+ and MRI+. Eotaxin and IL-9 were significantly lower in individuals who were MRI+ only. FGF-basic concentrations increased over time in MRI- individuals and were significantly higher than MRI+ individuals at 3 and 12 months. Multi-biomarker panels improved discriminability over single biomarkers at all timepoints (AUCs > 0.85 for admission and 2-week models classifying CT+ and AUC ≈ 0.90 for admission, 2-week and 3-month models classifying MRI+). CONCLUSIONS The CNS biomarkers GFAP and NFL were useful single diagnostic biomarkers of complicated mTBI, especially in acute and subacute phases after mTBI. Several inflammation markers were suppressed in patients with complicated versus uncomplicated mTBI and remained so even after 12 months. Multi-biomarker panels improved diagnostic accuracy at all timepoints, though at acute and 2-week timepoints, the single biomarkers GFAP and NFL, respectively, displayed similar accuracy compared to multi-biomarker panels.
Collapse
Affiliation(s)
- Gerard Janez Brett Clarke
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
| | - Turid Follestad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, N-7491, Norway
| | - Toril Skandsen
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Sha Tin, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anne Vik
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway
- Department of Neurosurgery, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Alexander Olsen
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Psychology, Norwegian University of Science and Technology, Trondheim, Norway
- NorHEAD - Norwegian Centre for Headache Research, Trondheim, Norway
| | - Asta Kristine Håberg
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.
- Department of Neuromedicine and Movement Sciences, NTNU, Trondheim, Norway.
| |
Collapse
|
8
|
Clarke GJB, Skandsen T, Zetterberg H, Follestad T, Einarsen CE, Vik A, Mollnes TE, Pischke SE, Blennow K, Håberg AK. Longitudinal Associations Between Persistent Post-Concussion Symptoms and Blood Biomarkers of Inflammation and CNS-Injury After Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:862-878. [PMID: 38117157 DOI: 10.1089/neu.2023.0419] [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: 12/21/2023] Open
Abstract
The aim of our study was to investigate the biological underpinnings of persistent post-concussion symptoms (PPCS) at 3 months following mild traumatic brain injury (mTBI). Patients (n = 192, age 16-60 years) with mTBI, defined as Glasgow Coma Scale (GCS) score between 13 and 15, loss of consciousness (LOC) <30 min, and post-traumatic amnesia (PTA) <24 h were included. Blood samples were collected at admission (within 72 h), 2 weeks, and 3 months. Concentrations of blood biomarkers associated with central nervous system (CNS) damage (glial fibrillary acidic protein [GFAP], neurofilament light [NFL], and tau) and inflammation (interferon gamma [IFNγ], interleukin [IL]-8, eotaxin, macrophage inflammatory protein-1-beta [MIP]-1β, monocyte chemoattractant protein [MCP]-1, interferon-gamma-inducible protein [IP]-10, IL-17A, IL-9, tumor necrosis factor [TNF], basic fibroblast growth factor [FGF]-basic platelet-derived growth factor [PDGF], and IL-1 receptor antagonist [IL-1ra]) were obtained. Demographic and injury-related factors investigated were age, sex, GCS score, LOC, PTA duration, traumatic intracranial finding on magnetic resonance imaging (MRI; within 72 h), and extracranial injuries. Delta values, that is, time-point differences in biomarker concentrations between 2 weeks minus admission and 3 months minus admission, were also calculated. PPCS was assessed with the British Columbia Post-Concussion Symptom Inventory (BC-PSI). In single variable analyses, longer PTA duration and a higher proportion of intracranial findings on MRI were found in the PPCS group, but no single biomarker differentiated those with PPCS from those without. In multi-variable models, female sex, longer PTA duration, MRI findings, and lower GCS scores were associated with increased risk of PPCS. Inflammation markers, but not GFAP, NFL, or tau, were associated with PPCS. At admission, higher concentrations of IL-8 and IL-9 and lower concentrations of TNF, IL-17a, and MCP-1 were associated with greater likelihood of PPCS; at 2 weeks, higher IL-8 and lower IFNγ were associated with PPCS; at 3 months, higher PDGF was associated with PPCS. Higher delta values of PDGF, IL-17A, and FGF-basic at 2 weeks compared with admission, MCP-1 at 3 months compared with admission, and TNF at 2 weeks and 3 months compared with admission were associated with greater likelihood of PPCS. Higher IL-9 delta values at both time-point comparisons were negatively associated with PPCS. Discriminability of individual CNS-injury and inflammation biomarkers for PPCS was around chance level, whereas the optimal combination of biomarkers yielded areas under the curve (AUCs) between 0.62 and 0.73. We demonstrate a role of biological factors on PPCS, including both positive and negative effects of inflammation biomarkers that differed based on sampling time-point after mTBI. PPCS was associated more with acute inflammatory processes, rather than ongoing inflammation or CNS-injury biomarkers. However, the modest discriminative ability of the models suggests other factors are more important in the development of PPCS.
Collapse
Affiliation(s)
- Gerard Janez Brett Clarke
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Toril Skandsen
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Turid Follestad
- Department of Clinical and Molecular Medicine, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinical Research Unit Central Norway, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Cathrine Elisabeth Einarsen
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Anne Vik
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurosurgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Søren Erik Pischke
- Department of Immunology, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
- Clinic for Emergencies and Critical Care, Department of Anesthesiology and Intensive Care Medicine, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Asta Kristine Håberg
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Department of Neuromedicine and Movement Sciences, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
9
|
da Silva Fiorin F, do Espírito Santo CC, Da Silva JT, Chung MK. Inflammation, brain connectivity, and neuromodulation in post-traumatic headache. Brain Behav Immun Health 2024; 35:100723. [PMID: 38292321 PMCID: PMC10827408 DOI: 10.1016/j.bbih.2024.100723] [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: 08/25/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Post-traumatic headache (PTH) is a debilitating condition that affects individuals with different levels of traumatic brain injury (TBI) severity. The difficulties in developing an effective treatment are related to a lack of understanding the complicated mechanisms and neurobiological changes in brain function after a brain injury. Preclinical studies have indicated that peripheral and central sensitization of the trigeminal nociceptive pathways contributes to PTH. While recent brain imaging studies have uncovered widespread changes in brain functional connectivity following trauma, understanding exactly how these networks contribute to PTH after injury remains unknown. Stimulation of peripheral (trigeminal or vagus) nerves show promising efficacies in PTH experimental animals, likely mediated by influencing TBI-induced pathological plasticity by decreasing neuroinflammation and neuronal apoptosis. Non-invasive brain stimulations, such as transcranial magnetic or direct current stimulations, show analgesia for multiple chronic pain conditions, including PTH. Better mechanistic understanding of analgesia achieved by neuromodulations can define peripheral and central mechanisms involved in the development, the resolution, and the management of PTH.
Collapse
Affiliation(s)
- Fernando da Silva Fiorin
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Caroline Cunha do Espírito Santo
- Graduate Program in Neuroengineering, Edmond and Lily Safra International Institute of Neuroscience, Santos Dumont Institute, Brazil
| | - Joyce T. Da Silva
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland Baltimore, Program in Neuroscience, Center to Advance Chronic Pain Research, Baltimore, MD, USA
| |
Collapse
|
10
|
Jauregui GV, Vukić D, Onyango IG, Arias C, Novotný JS, Texlová K, Wang S, Kovačovicova KL, Polakova N, Zelinkova J, Čarna M, Strašil VL, Head BP, Havas D, Mistrik M, Zorec R, Verkhratsky A, Keegan L, O'Connel M, Rissman R, Stokin GB. Amyloid precursor protein induces reactive astrogliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.571817. [PMID: 38187544 PMCID: PMC10769227 DOI: 10.1101/2023.12.18.571817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
We present in vitro and in vivo evidence demonstrating that Amyloid Precursor Protein (APP) acts as an essential instigator of reactive astrogliosis. Cell-specific overexpression of APP in cultured astrocytes led to remodelling of the intermediate filament network, enhancement of cytokine production and activation of cellular programs centred around the interferon (IFN) pathway, all signs of reactive astrogliosis. Conversely, APP deletion in cultured astrocytes abrogated remodelling of the intermediate filament network and blunted expression of IFN stimulated gene (ISG) products in response to lipopolysaccharide (LPS). Following traumatic brain injury (TBI), mouse reactive astrocytes also exhibited an association between APP and IFN, while APP deletion curbed the increase in glial fibrillary acidic protein (GFAP) observed canonically in astrocytes in response to TBI. Thus, APP represents a molecular inducer and regulator of reactive astrogliosis.
Collapse
Affiliation(s)
- Gretsen Velezmoro Jauregui
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Dragana Vukić
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- National Centre for Biomedical Research, Faculty of Science, Masaryk University, Brno Czech Republic
| | - Isaac G Onyango
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Carlos Arias
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Jan S Novotný
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Kateřina Texlová
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
| | - Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesia, University of California San Diego, San Diego, USA
| | | | - Natalie Polakova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jana Zelinkova
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Maria Čarna
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | | | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesia, University of California San Diego, San Diego, USA
| | | | - Martin Mistrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Robert Zorec
- Laboratory of Neuroendocrinology - Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Celica Biomedical, Technology Park, Ljubljana, Slovenia
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Achucarro Centre for Neuroscience, IIKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Stem Cell Biology, State Research Institute Centre for innovative Medicine, Vilnius, Lithuania
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning Province, China
| | - Liam Keegan
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Mary O'Connel
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Robert Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Gorazd B Stokin
- Translational Ageing and Neuroscience Program, Centre for Translational Medicine, International Clinical Research Centre, St. Anne's University Hospital, Brno, Czech Republic
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Neurology, Gloucestershire Royal Hospital, Gloucestershire NHS Foundation Trust, Gloucester, UK
| |
Collapse
|
11
|
Chen KS, Noureldein MH, McGinley LM, Hayes JM, Rigan DM, Kwentus JF, Mason SN, Mendelson FE, Savelieff MG, Feldman EL. Human neural stem cells restore spatial memory in a transgenic Alzheimer's disease mouse model by an immunomodulating mechanism. Front Aging Neurosci 2023; 15:1306004. [PMID: 38155736 PMCID: PMC10753006 DOI: 10.3389/fnagi.2023.1306004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023] Open
Abstract
Introduction Stem cells are a promising therapeutic in Alzheimer's disease (AD) given the complex pathophysiologic pathways involved. However, the therapeutic mechanisms of stem cells remain unclear. Here, we used spatial transcriptomics to elucidate therapeutic mechanisms of human neural stem cells (hNSCs) in an animal model of AD. Methods hNSCs were transplanted into the fimbria fornix of the hippocampus using the 5XFAD mouse model. Spatial memory was assessed by Morris water maze. Amyloid plaque burden was quantified. Spatial transcriptomics was performed and differentially expressed genes (DEGs) identified both globally and within the hippocampus. Subsequent pathway enrichment and ligand-receptor network analysis was performed. Results hNSC transplantation restored learning curves of 5XFAD mice. However, there were no changes in amyloid plaque burden. Spatial transcriptomics showed 1,061 DEGs normalized in hippocampal subregions. Plaque induced genes in microglia, along with populations of stage 1 and stage 2 disease associated microglia (DAM), were normalized upon hNSC transplantation. Pathologic signaling between hippocampus and DAM was also restored. Discussion hNSCs normalized many dysregulated genes, although this was not mediated by a change in amyloid plaque levels. Rather, hNSCs appear to exert beneficial effects in part by modulating microglia-mediated neuroinflammation and signaling in AD.
Collapse
Affiliation(s)
- Kevin S. Chen
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Mohamed H. Noureldein
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Lisa M. McGinley
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Diana M. Rigan
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Jacquelin F. Kwentus
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Shayna N. Mason
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| | - Masha G. Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, United States
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
12
|
Cao D, Li B, Cao C, Zhang J, Li X, Li H, Yu Z, Shen H, Ye M. Caveolin-1 aggravates neurological deficits by activating neuroinflammation following experimental intracerebral hemorrhage in rats. Exp Neurol 2023; 368:114508. [PMID: 37598879 DOI: 10.1016/j.expneurol.2023.114508] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/30/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is one of the stroke subtypes with the highest mortality. Secondary brain injury is associated with neurological dysfunction and poor prognosis after ICH. Caveolin-1 (CAV1) is the key protein of Caveolae. Previous studies have shown that CAV1 plays an important role in central nervous system diseases, and pointed out that in a collagenase-induced ICH model in vivo, CAV1 is associated with neuroinflammatory activation and poor neurological prognosis. In this study, we explore the role and the molecular mechanism of CAV1 in brain injury via a rat autologous whole blood injection model and an in vitro model of ICH. METHODS Adult male Sprague-Dawley rats ICH model was induced through autologous whole blood injecting into the right basal ganglia. The changes in protein levels of CAV1 in brain tissues of ICH rats were detected by western blot analysis. The immunofluorescent staining was used to explore the changes of CAV1 in microglia/macrophages (Iba1+ cells). Lentivirus vectors were administered by intracerebroventricular injection to induce CAV1 overexpression and knockdown respectively. The western blot analysis, immunofluorescence staining, enzyme-linked immunosorbent assay, terminal deoxynucleotidyl transferase dUTP nick end labeling and Nissl staining were performed to explore the role of CAV1 in secondary brain injury after ICH. Meanwhile, the rotarod test, foot fault test, adhesive-removal test, and Modified Garcia Test, as well as Morris Water Maze test, were performed to evaluate the behavioral cognitive impairment of ICH rats after genetic intervention. Additionally, BV-2 cells treated with oxygen hemoglobin for 24 h, were used as an in vitro model of ICH in this study to explore the molecular mechanism of CAV1 in brain injury; we performed western blot analysis after precise regulation of CAV1 in BV2 cells to observe changes in protein levels and phosphorylated levels of C-Src, IKK-β, and NF-κB. RESULTS The expression of CAV1 in microglia/macrophages (Iba1+ cells) was elevated and reached the peak at 24 h after ICH. CAV1 knockdown ameliorated ICH-induced neurological deficits, while CAV1 overexpression significantly worsened neurological dysfunction of ICH rats. CAV1 knockdown attenuated cellular apoptosis and promoted neuronal survival in brain tissues of ICH rats, while the ICH rats with CAV1 overexpression presented more cellular apoptosis and neuronal loss. Meanwhile, CAV1 knockdown inhibited the microglia activation and neuroinflammatory response, while CAV1 overexpression abolished these effects and aggravated neuroinflammation in brain tissues of ICH rats. Additionally, by inducing to CAV1 knockdown in BV2 cells in an in vitro model of ICH, the levels of p-C-Src, CAV-1, p-CAV-1, and p-IKK-β in cytoplasm and the level of NF-κB p65 in nucleus of BV2 cells were significantly decreased, while they were increased by inducing to CAV1 overexpression. CONCLUSIONS Our research revealed CAV1 aggravated neurological dysfunction in a rat ICH model. CAV1 knockdown exerted neuroprotective effect by suppressing microglia activation and neuroinflammation after ICH might via the C-Src/CAV1/IKK-β/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Demao Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of neurosurgery, The Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Bing Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Neurosurgery, Yancheng City No.1 People's Hospital, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224006, Jiangsu Province, China
| | - Cheng Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China; Department of Neurocritical Intensive Care Unit, Jiangyin Clinical College of Xuzhou Medical College, Jiangyin, Jiangsu Province, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| | - Ming Ye
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu Province, China.
| |
Collapse
|
13
|
The prognostic value of caveolin-1 levels in ischemic stroke patients after mechanical thrombectomy. Neurol Sci 2023; 44:2081-2086. [PMID: 36746844 DOI: 10.1007/s10072-023-06606-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND PURPOSE The impact of serum caveolin-1 (Cav-1) on clinical outcomes in patients after mechanical thrombectomy (MT) is unclear. We aimed to investigate the association between serum cav-1 levels and the 3-month functional outcome. METHODS We prospectively enrolled and analyzed patients with an anterior circulation large vessel occlusion who underwent MT. Serum cav-1 concentrations were tested after admission. The primary outcome was a 90-day modified Rankin Scale score of 3-6. RESULTS Of the 237 recruited patients (mean age, 69.7 ± 12.1 years; 152 male), 131 (55.3%) experienced a 90-day poor outcome. After adjustment for demographic characteristics and other covariates, patients with higher serum Cav-1 levels had a reduced risk of poor outcome at 3 months (Per 1-standard deviation increase; odd ratios [OR], 0.59; 95% confidence interval [CI], 0.39 - 0.89, P = 0.013). Elevated Cav-1 concentrations (Per 1-standard deviation increase; OR, 0.59; 95% CI, 0.40 - 0.88, P = 0.011) were significantly associated with a favorable shift in modified Rankin Scale score distribution. Similar results were confirmed when the Cav-1 levels were analyzed as a categorical variable. Furthermore, the restricted cubic spline showed a linear association between Cav-1 levels and 90-day poor outcome (P = 0.032 for linearity). CONCLUSIONS Increased serum Cav-1 levels were associated with improved prognosis at 3 months in ischemic stroke patients after MT, suggesting that Cav-1 may be a potential prognostic biomarker for ischemic stroke after reperfusion therapy.
Collapse
|
14
|
Aiyede M, Lim XY, Russell AAM, Patel RP, Gueven N, Howells DW, Bye N. A Systematic Review and Meta-Analysis on the Therapeutic Efficacy of Heparin and Low Molecular Weight Heparins in Animal Studies of Traumatic Brain Injury. J Neurotrauma 2023; 40:4-21. [PMID: 35880422 DOI: 10.1089/neu.2022.0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The identification of effective pharmacotherapies for traumatic brain injury (TBI) remains a major challenge. Treatment with heparin and its derivatives is associated with neuroprotective effects after experimental TBI; however, the optimal dosage and method of administration, modes of action, and effects on hemorrhage remain unclear. Therefore, this review aimed to systematically evaluate, analyze, and summarize the available literature on the use of heparin and low molecular weight heparins (LMWHs) as treatment options for experimental TBI. We searched two online databases (PubMed and ISI Web of Science) to identify relevant studies. Data pertaining to TBI paradigm, animal subjects, drug administration, and all pathological and behavior outcomes were extracted. Eleven studies met our pre-specified inclusion criteria, and for outcomes with sufficient numbers, data from seven publications were analyzed in a weighted mean difference meta-analysis using a random-effects model. Study quality and risk of bias were also determined. Meta-analysis revealed that heparin and its derivatives decreased brain edema, leukocyte rolling, and vascular permeability, and improved neurological function. Further, treatment did not aggravate hemorrhage. These findings must be interpreted with caution, however, because they were determined from a limited number of studies with substantial heterogeneity. Also, overall study quality was low based on absences of data reporting, and potential publication bias was identified. Importantly, we found that there are insufficient data to evaluate the variables we had hoped to investigate. The beneficial effects of heparin and LMWHs, however, suggest that further pre-clinical studies are warranted.
Collapse
Affiliation(s)
- Mimieveshiofuo Aiyede
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia
| | - Xin Yi Lim
- Herbal Medicine Research Centre, Institute for Medical Research, Ministry of Health, Kuala Lumpur, Malaysia
| | - Ash A M Russell
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia
| | - David W Howells
- School of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Nicole Bye
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
15
|
Rawat V, Eastman CL, Amaradhi R, Banik A, Fender JS, Dingledine RJ, D’Ambrosio R, Ganesh T. Temporal Expression of Neuroinflammatory and Oxidative Stress Markers and Prostaglandin E2 Receptor EP2 Antagonist Effect in a Rat Model of Epileptogenesis. ACS Pharmacol Transl Sci 2022; 6:128-138. [PMID: 36654746 PMCID: PMC9841781 DOI: 10.1021/acsptsci.2c00189] [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: 09/28/2022] [Indexed: 12/13/2022]
Abstract
Traumatic brain injury (TBI) in patients results in a massive inflammatory reaction, disruption of blood-brain barrier, and oxidative stress in the brain, and these inciting features may culminate in the emergence of post-traumatic epilepsy (PTE). We hypothesize that targeting these pathways with pharmacological agents could be an effective therapeutic strategy to prevent epileptogenesis. To design therapeutic strategies targeting neuroinflammation and oxidative stress, we utilized a fluid percussion injury (FPI) rat model to study the temporal expression of neuroinflammatory and oxidative stress markers from 3 to 24 h following FPI. FPI results in increased mRNA expression of inflammatory mediators including cyclooxygenase-2 (COX-2) and prostanoid receptor EP2, marker of oxidative stress (NOX2), astrogliosis (GFAP), and microgliosis (CD11b) in ipsilateral cortex and hippocampus. The analysis of protein levels indicated a significant increase in the expression of COX-2 in ipsilateral hippocampus and cortex post-FPI. We tested FPI rats with an EP2 antagonist TG8-260 which produced a statistically significant reduction in the distribution of seizure duration post-FPI and trends toward a reduction in seizure incidence, seizure frequency, and duration, hinting a proof of concept that EP2 antagonism must be further optimized for therapeutic applications to prevent epileptogenesis.
Collapse
Affiliation(s)
- Varun Rawat
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Clifford L. Eastman
- Department
of Neurological Surgery, University of Washington, 325 Ninth Avenue, Seattle, Washington 98104, United States
| | - Radhika Amaradhi
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Avijit Banik
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jason S. Fender
- Department
of Neurological Surgery, University of Washington, 325 Ninth Avenue, Seattle, Washington 98104, United States
| | - Raymond J. Dingledine
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States
| | - Raimondo D’Ambrosio
- Department
of Neurological Surgery, University of Washington, 325 Ninth Avenue, Seattle, Washington 98104, United States,Regional
Epilepsy Center, University of Washington, 325 Ninth Avenue, Seattle, Washington 98104, United States
| | - Thota Ganesh
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, Atlanta, Georgia 30322, United States,. Phone: 404-727-7393. Fax: 404-727-0365
| |
Collapse
|
16
|
Xia W, Li X, Wu Q, Xu A, Zhang L, Xia Z. The importance of caveolin as a target in the prevention and treatment of diabetic cardiomyopathy. Front Immunol 2022; 13:951381. [PMID: 36405687 PMCID: PMC9666770 DOI: 10.3389/fimmu.2022.951381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
The diabetic population has been increasing in the past decades and diabetic cardiomyopathy (DCM), a pathology that is defined by the presence of cardiac remodeling and dysfunction without conventional cardiac risk factors such as hypertension and coronary heart diseases, would eventually lead to fatal heart failure in the absence of effective treatment. Impaired insulin signaling, commonly known as insulin resistance, plays an important role in the development of DCM. A family of integral membrane proteins named caveolins (mainly caveolin-1 and caveolin-3 in the myocardium) and a protein hormone adiponectin (APN) have all been shown to be important for maintaining normal insulin signaling. Abnormalities in caveolins and APN have respectively been demonstrated to cause DCM. This review aims to summarize recent research findings of the roles and mechanisms of caveolins and APN in the development of DCM, and also explore the possible interplay between caveolins and APN.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xia Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Liangqing Zhang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Simon DW, Raphael I, Johnson KM, Dixon CE, Vagni V, Janesko-Feldman K, Kochanek PM, Bayir H, Clark RS, McGeachy MJ. Endogenous Interleukin-17a Contributes to Normal Spatial Memory Retention but Does Not Affect Early Behavioral or Neuropathological Outcomes after Experimental Traumatic Brain Injury. Neurotrauma Rep 2022; 3:340-351. [PMID: 36204388 PMCID: PMC9531893 DOI: 10.1089/neur.2022.0017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interleukin-17 (IL-17) is a proinflammatory cytokine primarily secreted in the brain by inflammatory T lymphocytes and glial cells. IL-17+ T-helper (Th17) cells are increased in the ipsilateral hemisphere after experimental traumatic brain injury (TBI), and IL-17 levels are increased in serum and brain tissue. We hypothesized that il17a and related gene expression would be increased in brain tissue after TBI in mice and il17a-/- mice would demonstrate neuroprotection versus wild type. The controlled cortical impact (CCI) model of TBI in adult male C57BL6/J mice was used for all experiments. Data were analyzed by analysis of variance (ANOVA) or repeated-measures two-way ANOVA with the Bonferroni correction. A value of p < 0.05 determined significance. Expression of il17a was significantly reduced in the ipsilateral cortex and hippocampus by day 3 after TBI, and expression remained low at 28 days. There were no differences between il17a-/- and il17a+/+ mice in beam balance, Morris water maze performance, or lesion volume after CCI. Surprisingly, naïve il17a -/- mice performed significantly (p = 0.02) worse than naïve il17a+/+ mice on the probe trial. In conclusion, sustained depression of il17a gene expression was observed in brains after TBI in adult mice. Genetic knockout of IL-17 was not neuroprotective after TBI. IL-17a may be important for memory retention in naïve mice.
Collapse
Affiliation(s)
- Dennis W. Simon
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Itay Raphael
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kendall M. Johnson
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - C. Edward Dixon
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vincent Vagni
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Keri Janesko-Feldman
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Patrick M. Kochanek
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hülya Bayir
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert S.B. Clark
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Clinical and Translational Science Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mandy J. McGeachy
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
18
|
Regulation of Neuroinflammatory Signaling by PPARγ Agonist in Mouse Model of Diabetes. Int J Mol Sci 2022; 23:ijms23105502. [PMID: 35628311 PMCID: PMC9141386 DOI: 10.3390/ijms23105502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/09/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
Many relevant studies, as well as clinical practice, confirm that untreated diabetes predisposes the development of neuroinflammation and cognitive impairment. Having regard for the fact that PPARγ are widely distributed in the brain and PPARγ ligands may regulate the inflammatory process, the anti-inflammatory potential of the PPARγ agonist, pioglitazone, was assessed in a mouse model of neuroinflammation related with diabetes. In this regard, the biochemical and molecular indicators of neuroinflammation were determined in the hippocampus and prefrontal cortex of diabetes mice. The levels of cytokines (IL-1β, IL-6, and TNF) and the expression of genes (Tnfrsf1a and Cav1) were measured. In addition, behavioral tests such as the open field test, the hole-board test, and the novel object recognition test were conducted. A 14-day treatment with pioglitazone significantly decreased IL-6 and TNFα levels in the prefrontal cortex and led to the downregulation of Tnfrsf1a expression and the upregulation of Cav1 expression in both brain regions of diabetic mice. Pioglitazone, by targeting neuroinflammatory signaling, improved memory and exploratory activity in behavioral tests. The present study provided a potential theoretical basis and therapeutic target for the treatment of neuroinflammation associated with diabetes. Pioglitazone may provide a promising therapeutic strategy in diabetes patients with muffled of behavioral activity.
Collapse
|
19
|
Amaradhi R, Mohammed S, Banik A, Franklin R, Dingledine R, Ganesh T. Second-Generation Prostaglandin Receptor EP2 Antagonist, TG8-260, with High Potency, Selectivity, Oral Bioavailability, and Anti-Inflammatory Properties. ACS Pharmacol Transl Sci 2022; 5:118-133. [PMID: 35187419 PMCID: PMC8844972 DOI: 10.1021/acsptsci.1c00255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Indexed: 02/08/2023]
Abstract
EP2, a G-protein-coupled prostaglandin-E2 receptor, has emerged as a seminal biological target for drug discovery. EP2 receptor activation is typically proinflammatory; therefore, the development of EP2 antagonists to mitigate the severity and disease pathology in a variety of inflammation-driven central nervous system and peripheral disorders would be a novel strategy. We have recently developed a second-generation EP2 antagonist TG8-260 and shown that it reduces hippocampal neuroinflammation and gliosis after pilocarpine-induced status epilepticus in rats. Here, we present details of synthesis, lead optimization on earlier leads that resulted in TG8-260, potency and selectivity evaluations using cAMP-driven time-resolved fluorescence resonance energy-transfer (TR-FRET) assays and [H3]-PGE2-binding assays, absorption, distribution, metabolism, and excretion (ADME), and pharmacokinetics. TG8-260 (2f) showed Schild K B = 13.2 nM (3.6-fold more potent than the previous lead TG8-69 (1c)) and 500-fold selectivity to EP2 against other prostanoid receptors. Pharmacokinetic data indicated that TG8-260 has a plasma half-life of 2.14 h (PO) and excellent oral bioavailability (77.3%). Extensive ADME tests indicated that TG8-260 is a potent inhibitor of CYP450 enzymes. Further, we show that TG8-260 displays antagonistic activity on the induction of EP2 receptor-mediated inflammatory gene expression in microglia BV2-hEP2 cells; therefore, it can serve as a tool for investigating anti-inflammatory pathways in peripheral inflammatory disease animal models.
Collapse
Affiliation(s)
- Radhika Amaradhi
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, 1510 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Shabber Mohammed
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, 1510 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Avijit Banik
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, 1510 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Ronald Franklin
- Franklin
ADME Consult, LLC, Boulder, Colorado 80303, United States
| | - Raymond Dingledine
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, 1510 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Thota Ganesh
- Department
of Pharmacology and Chemical Biology, Emory
University School of Medicine, 1510 Clifton Road NE, Atlanta, Georgia 30322, United States,. Tel.: 404-727-7393. Fax: 404-727-0365
| |
Collapse
|
20
|
Li W, Cao F, Takase H, Arai K, Lo EH, Lok J. Blood-Brain Barrier Mechanisms in Stroke and Trauma. Handb Exp Pharmacol 2022; 273:267-293. [PMID: 33580391 DOI: 10.1007/164_2020_426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The brain microenvironment is tightly regulated. The blood-brain barrier (BBB), which is composed of cerebral endothelial cells, astrocytes, and pericytes, plays an important role in maintaining the brain homeostasis by regulating the transport of both beneficial and detrimental substances between circulating blood and brain parenchyma. After brain injury and disease, BBB tightness becomes dysregulated, thus leading to inflammation and secondary brain damage. In this chapter, we overview the fundamental mechanisms of BBB damage and repair after stroke and traumatic brain injury (TBI). Understanding these mechanisms may lead to therapeutic opportunities for brain injury.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Cao
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Josephine Lok
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
21
|
Firdaus R, Theresia S, Austin R, Tiara R. Propofol effects in rodent models of traumatic brain injury: a systematic review. ASIAN BIOMED 2021; 15:253-265. [PMID: 37551361 PMCID: PMC10321222 DOI: 10.2478/abm-2021-0032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Traumatic brain injury (TBI) causes high mortality and disability worldwide. Animal models have been developed to explore the complex processes in TBI. Propofol is used to manage head injuries during surgical intervention and mechanical ventilation in patients with TBI. Many studies have investigated the neuroprotective effect of propofol on TBI. However, other studies have shown neurotoxic effects. Objectives To review systematically the literature regarding the neuroprotective and neurotoxic effects of propofol in rodent models of TBI. Methods Data from rodents as models of TBI with propofol as one of the intervention agents, and/or comparing the neuroprotective effects of propofol with the other substances in rodent models of TBI, were obtained from PubMed, EBSCO Host, and ProQuest databases. The PRISMA 2020 statement recommendations were followed and research questions were developed based on PICOS guidelines. Data was extracted from the literature using a standardized Cochrane method. Results We analyzed data from 12 articles on physiological changes of experimental animals before and after trauma, the effects of propofol administration, and the observed neurotoxic effects. The effects of propofol administration were observed in terms of changes in traumatic lesion volume, the release of antioxidants and inflammatory factors, and the neurological function of rodent models of TBI. Conclusion Propofol has neuroprotective and neurotoxic effects via several mechanisms, and various doses have been used in research to determine its effects. The timing of administration, the dose administered, and the duration of administration contribute to determine the effect of propofol in rodent models of TBI. However, the doses that produce neuroprotective and neurotoxic effects are not yet clear and further research is needed to determine them.
Collapse
Affiliation(s)
- Riyadh Firdaus
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Sandy Theresia
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Ryan Austin
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| | - Rani Tiara
- Department of Anesthesiology and Intensive Therapy, Faculty of Medicine, Universitas Indonesia, Dr. Cipto Mangunkusumo Hospital, Jakarta10430, Indonesia
| |
Collapse
|
22
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
23
|
Lucas-Ruiz F, Galindo-Romero C, Albaladejo-García V, Vidal-Sanz M, Agudo-Barriuso M. Mechanisms implicated in the contralateral effect in the central nervous system after unilateral injury: focus on the visual system. Neural Regen Res 2021; 16:2125-2131. [PMID: 33818483 PMCID: PMC8354113 DOI: 10.4103/1673-5374.310670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/21/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022] Open
Abstract
The retina, as part of the central nervous system is an ideal model to study the response of neurons to injury and disease and to test new treatments. During the last decade is becoming clear that unilateral lesions in bilateral areas of the central nervous system trigger an inflammatory response in the contralateral uninjured site. This effect has been better studied in the visual system where, as a rule, one retina is used as experimental and the other as control. Contralateral retinas in unilateral models of retinal injury show neuronal degeneration and glial activation. The mechanisms by which this adverse response in the central nervous system occurs are discussed in this review, focusing primarily on the visual system.
Collapse
Affiliation(s)
- Fernando Lucas-Ruiz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Caridad Galindo-Romero
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Virginia Albaladejo-García
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| |
Collapse
|
24
|
Fernández-Albarral JA, Martínez-López MA, Marco EM, de Hoz R, Martín-Sánchez B, San Felipe D, Salobrar-García E, López-Cuenca I, Pinazo-Durán MD, Salazar JJ, Ramírez JM, López-Gallardo M, Ramírez AI. Is Saffron Able to Prevent the Dysregulation of Retinal Cytokines Induced by Ocular Hypertension in Mice? J Clin Med 2021; 10:jcm10214801. [PMID: 34768320 PMCID: PMC8584889 DOI: 10.3390/jcm10214801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
Cytokine- and chemokine-mediated signalling is involved in the neuroinflammatory process that leads to retinal ganglion cell (RGC) damage in glaucoma. Substances with anti-inflammatory properties could decrease these cytokines and chemokines and thus prevent RGC death. The authors of this study analysed the anti-inflammatory effect of a hydrophilic saffron extract standardized to 3% crocin content, focusing on the regulation of cytokine and chemokine production, in a mouse model of unilateral laser-induced ocular hypertension (OHT). We demonstrated that following saffron treatment, most of the concentration of proinflammatory cytokines (IL-1β, IFN-γ, TNF-α, and IL-17), anti-inflammatory cytokines (IL-4 and IL-10), Brain-derived Neurotrophic Factor (BDNF), Vascular Endothelial Growth Factor (VEGF), and fractalkine were unaffected in response to laser-induced OHT in both the OHT eye and its contralateral eye. Only IL-6 levels were significantly increased in the OHT eye one day after laser induction compared with the control group. These results differed from those observed in animals subjected to unilateral OHT and not treated with saffron, where changes in cytokine levels occurred in both eyes. Therefore, saffron extract regulates the production of proinflammatory cytokines, VEGF, and fractalkine induced by increasing intraocular pressure (IOP), protecting the retina from inflammation. These results indicate that saffron could be beneficial in glaucoma by helping to reduce the inflammatory process.
Collapse
Affiliation(s)
- José A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
| | - Miguel A. Martínez-López
- Departamento de Fisiología, Facultad de Medicina, Grupo UCM 951579, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.A.M.-L.); (B.M.-S.); (D.S.F.)
| | - Eva M. Marco
- Departamento de Genética, Facultad de CC. Biológicas, Fisiología y Microbiología, Grupo UCM 951579, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Beatriz Martín-Sánchez
- Departamento de Fisiología, Facultad de Medicina, Grupo UCM 951579, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.A.M.-L.); (B.M.-S.); (D.S.F.)
| | - Diego San Felipe
- Departamento de Fisiología, Facultad de Medicina, Grupo UCM 951579, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.A.M.-L.); (B.M.-S.); (D.S.F.)
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
| | - María D. Pinazo-Durán
- Ophthalmic Research Unit “Santiago Grisolía”—FISABIO and Cellular and Molecular Ophthalmobiology Unit, University of Valencia, 46017 Valencia, Spain;
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
- Departamento de Inmunología, Facultad de Medicina, Oftalmología y ORL, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Meritxell López-Gallardo
- Departamento de Fisiología, Facultad de Medicina, Grupo UCM 951579, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.A.M.-L.); (B.M.-S.); (D.S.F.)
- Correspondence: (M.L.-G.); (A.I.R.)
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (E.S.-G.); (I.L.-C.); (J.J.S.); (J.M.R.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence: (M.L.-G.); (A.I.R.)
| |
Collapse
|
25
|
Barnes SE, Zera KA, Ivison GT, Buckwalter MS, Engleman EG. Brain profiling in murine colitis and human epilepsy reveals neutrophils and TNFα as mediators of neuronal hyperexcitability. J Neuroinflammation 2021; 18:199. [PMID: 34511110 PMCID: PMC8436533 DOI: 10.1186/s12974-021-02262-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Patients with chronic inflammatory disorders such as inflammatory bowel disease frequently experience neurological complications including epilepsy, depression, attention deficit disorders, migraines, and dementia. However, the mechanistic basis for these associations is unknown. Given that many patients are unresponsive to existing medications or experience debilitating side effects, novel therapeutics that target the underlying pathophysiology of these conditions are urgently needed. METHODS Because intestinal disorders such as inflammatory bowel disease are robustly associated with neurological symptoms, we used three different mouse models of colitis to investigate the impact of peripheral inflammatory disease on the brain. We assessed neuronal hyperexcitability, which is associated with many neurological symptoms, by measuring seizure threshold in healthy and colitic mice. We profiled the neuroinflammatory phenotype of colitic mice and used depletion and neutralization assays to identify the specific mediators responsible for colitis-induced neuronal hyperexcitability. To determine whether our findings in murine models overlapped with a human phenotype, we performed gene expression profiling, pathway analysis, and deconvolution on microarray data from hyperexcitable human brain tissue from patients with epilepsy. RESULTS We observed that murine colitis induces neuroinflammation characterized by increased pro-inflammatory cytokine production, decreased tight junction protein expression, and infiltration of monocytes and neutrophils into the brain. We also observed sustained neuronal hyperexcitability in colitic mice. Colitis-induced neuronal hyperexcitability was ameliorated by neutrophil depletion or TNFα blockade. Gene expression profiling of hyperexcitable brain tissue resected from patients with epilepsy also revealed a remarkably similar pathology to that seen in the brains of colitic mice, including neutrophil infiltration and high TNFα expression. CONCLUSIONS Our results reveal neutrophils and TNFα as central regulators of neuronal hyperexcitability of diverse etiology. Thus, there is a strong rationale for evaluating anti-inflammatory agents, including clinically approved TNFα inhibitors, for the treatment of neurological and psychiatric symptoms present in, and potentially independent of, a diagnosed inflammatory disorder.
Collapse
Affiliation(s)
- Sarah E Barnes
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Kristy A Zera
- Department of Neurology, Stanford University, Stanford, CA, USA
| | - Geoffrey T Ivison
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Infectious Diseases, Stanford University, Stanford, CA, USA
| | | | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
26
|
Caveolin-1, a novel player in cognitive decline. Neurosci Biobehav Rev 2021; 129:95-106. [PMID: 34237390 DOI: 10.1016/j.neubiorev.2021.06.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 06/29/2021] [Indexed: 12/12/2022]
Abstract
Cognitive decline (CD), which related to vascular dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and diabetes mellitus, is a growing health concern that has a great impact on the patients' quality of life. Although extensive efforts, the mechanisms of CD are still far from being clarified, not to mention the effective treatment and prevention strategies. Caveolin-1 (Cav-1), a trans-membrane protein, is a major component of the caveolae structure and scaffolding proteins. Recently, ample evidence depicts a strong correlation between Cav-1 and CD, however, the specific role of Cav-1 in CD has not been clearly examined and how they might be connected have yet to be identified. This review seeks to provide a comprehensive overview about how Cav-1 modulates pathogeneses of CD-associated diseases. In summary, Cav-1 can promote structural and functional plasticity of neurons, improve neurogenesis, relieve mitochondrial dysfunction, inhibit inflammation and suppress oxidative stress, which have shed light on the idea that Cav-1 may be an efficacious therapeutic target to treat CD.
Collapse
|
27
|
Wang S, Leem JS, Podvin S, Hook V, Kleschevnikov N, Savchenko P, Dhanani M, Zhou K, Kelly IC, Zhang T, Miyanohara A, Nguyen P, Kleschevnikov A, Wagner SL, Trojanowski JQ, Roth DM, Patel HH, Patel PM, Head BP. Synapsin-caveolin-1 gene therapy preserves neuronal and synaptic morphology and prevents neurodegeneration in a mouse model of AD. Mol Ther Methods Clin Dev 2021; 21:434-450. [PMID: 33981778 PMCID: PMC8065227 DOI: 10.1016/j.omtm.2021.03.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/24/2021] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most common form of neurodegeneration and cognitive dysfunction in the elderly. Identifying molecular signals that mitigate and reverse neurodegeneration in AD may be exploited therapeutically. Transgenic AD mice (PSAPP) exhibit learning and memory deficits at 9 and 11 months, respectively, with associated decreased expression of caveolin-1 (Cav-1), a membrane/lipid raft (MLR) scaffolding protein necessary for synaptic and neuroplasticity. Neuronal-targeted gene therapy using synapsin-Cav-1 cDNA (SynCav1) was delivered to the hippocampus of PSAPP mice at 3 months using adeno-associated virus serotype 9 (AAV9). Bilateral SynCav1 gene therapy was able to preserve MLRs profile, learning and memory, hippocampal dendritic arbor, synaptic ultrastructure, and axonal myelin content in 9- and 11-month PSAPP mice, independent of reducing toxic amyloid deposits and astrogliosis. Our data indicate that SynCav1 gene therapy may be an option for AD and potentially in other forms of neurodegeneration of unknown etiology.
Collapse
Affiliation(s)
- Shanshan Wang
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Joseph S. Leem
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Sonia Podvin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Vivian Hook
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Natalia Kleschevnikov
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Paul Savchenko
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Mehul Dhanani
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Kimberly Zhou
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Isabella C. Kelly
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Tong Zhang
- Campus Microscopy & Imaging Facility (CMIF)/Microscopy Shared Resource (MSR), The Ohio State University, OH, USA
| | - Atsushi Miyanohara
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Phuong Nguyen
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | | | - Steve L. Wagner
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - John Q. Trojanowski
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104-4283, USA
| | - David M. Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Hemal H. Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Piyush M. Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| | - Brian P. Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
- Department of Anesthesiology, University of California San Diego, San Diego, CA 92161, USA
| |
Collapse
|
28
|
Amoo M, O'Halloran PJ, Henry J, Husien MB, Brennan P, Campbell M, Caird J, Curley GF. Permeability of the Blood-Brain Barrier after Traumatic Brain Injury; Radiological Considerations. J Neurotrauma 2021; 39:20-34. [PMID: 33632026 DOI: 10.1089/neu.2020.7545] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability, especially in young persons, and constitutes a major socioeconomic burden worldwide. It is regarded as the leading cause of mortality and morbidity in previously healthy young persons. Most of the mechanisms underpinning the development of secondary brain injury are consequences of disruption of the complex relationship between the cells and proteins constituting the neurovascular unit or a direct result of loss of integrity of the tight junctions (TJ) in the blood-brain barrier (BBB). A number of changes have been described in the BBB after TBI, including loss of TJ proteins, pericyte loss and migration, and altered expressions of water channel proteins at astrocyte end-feet processes. There is a growing research interest in identifying optimal biological and radiological biomarkers of severity of BBB dysfunction and its effects on outcomes after TBI. This review explores the microscopic changes occurring at the neurovascular unit, after TBI, and current radiological adjuncts for its evaluation in pre-clinical and clinical practice.
Collapse
Affiliation(s)
- Michael Amoo
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Royal College of Surgeons in Ireland, Dublin, Ireland.,Beacon Academy, Beacon Hospital, Sandyford, Dublin, Ireland
| | - Philip J O'Halloran
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Neurosurgery, Royal London Hospital, Whitechapel, London, United Kingdom
| | - Jack Henry
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Mohammed Ben Husien
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland.,Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paul Brennan
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Radiology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | | | - John Caird
- National Centre for Neurosurgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Gerard F Curley
- Royal College of Surgeons in Ireland, Dublin, Ireland.,Department of Anaesthesia and Critical Care, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
29
|
Retinal Molecular Changes Are Associated with Neuroinflammation and Loss of RGCs in an Experimental Model of Glaucoma. Int J Mol Sci 2021; 22:ijms22042066. [PMID: 33669765 PMCID: PMC7922243 DOI: 10.3390/ijms22042066] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/14/2021] [Accepted: 02/17/2021] [Indexed: 12/19/2022] Open
Abstract
Signaling mediated by cytokines and chemokines is involved in glaucoma-associated neuroinflammation and in the damage of retinal ganglion cells (RGCs). Using multiplexed immunoassay and immunohistochemical techniques in a glaucoma mouse model at different time points after ocular hypertension (OHT), we analyzed (i) the expression of pro-inflammatory cytokines, anti-inflammatory cytokines, BDNF, VEGF, and fractalkine; and (ii) the number of Brn3a+ RGCs. In OHT eyes, there was an upregulation of (i) IFN-γ at days 3, 5, and 15; (ii) IL-4 at days 1, 3, 5, and 7 and IL-10 at days 3 and 5 (coinciding with downregulation of IL1-β at days 1, 5, and 7); (iii) IL-6 at days 1, 3, and 5; (iv) fractalkine and VEGF at day 1; and (v) BDNF at days 1, 3, 7, and 15. In contralateral eyes, there were (i) an upregulation of IL-1β at days 1 and 3 and a downregulation at day 7, coinciding with the downregulation of IL4 at days 3 and 5 and the upregulation at day 7; (ii) an upregulation of IL-6 at days 1, 5, and 7 and a downregulation at 15 days; (iii) an upregulation of IL-10 at days 3 and 7; and (iv) an upregulation of IL-17 at day 15. In OHT eyes, there was a reduction in the Brn3a+ RGCs number at days 3, 5, 7, and 15. OHT changes cytokine levels in both OHT and contralateral eyes at different time points after OHT induction, confirming the immune system involvement in glaucomatous neurodegeneration.
Collapse
|
30
|
Abstract
Due to the growing number of chronic traumatic encephalopathy (CTE) cases in the military and contact sports, defining the cellular and molecular substrate of this disorder is crucial. Most classic neuropathological investigations describe cortical tau and, to a lesser extent, amyloid lesions, which may underlie the clinical sequela associated with CTE. The application of modern molecular biologic technology to postmortem human brain tissue has made it possible to evaluate the genetic signature of specific neuronal phenotypes at different stages of CTE pathology. Most recently, molecular pathobiology has been used in the field of CTE, with an emphasis on the cholinergic neurons located within the nucleus basalis of Meynert, which develop tau pathology and are associated with cognitive dysfunction similar to that found in Alzheimer's disease (AD). Quantitative findings derived from single-cell transcript investigations provide clues to our understanding of the selective vulnerability of neurons containing AD-like tau pathology at different stages of CTE. Since human tissue-based studies provide a gold standard for the field of CTE, continued molecular pathological studies are needed to reveal novel drug targets for the treatment of this disorder.
Collapse
|
31
|
Fang H, Li HF, Yan JY, Yang M, Zhang JP. Dexmedetomidine-up-regulated microRNA-381 exerts anti-inflammatory effects in rats with cerebral ischaemic injury via the transcriptional factor IRF4. J Cell Mol Med 2020; 25:2098-2109. [PMID: 33314611 PMCID: PMC7882963 DOI: 10.1111/jcmm.16153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/29/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Dexmedetomidine (Dex) possesses analgesic and anaesthetic values and reported being used in cerebral ischaemic injury therapeutics. Accumulating studies have determined the effect of microRNAs (miRNAs) on the cerebral ischaemic injury. Thus, the present study aimed to unravel the molecular mechanism of miR-381 and Dex in cerebral ischaemic injury. For this purpose, the cerebral ischaemic injury rat model was established by induction of middle cerebral artery occlusion (MCAO) and expression of miR-381 and IRF4 was determined. Thereafter, MCAO rats were treated with Dex, miR-381 mimic, miR-381 inhibitor and oe-IRF4 respectively, followed by evaluation of neurological function. Furthermore, neuron cells were isolated from the hippocampus of rats and subjected to oxygen-glucose deprivation (OGD). Then, OGD-treated neuron cells and primary neuron cells were examined by gain- and loss-of-function assay. Neuron cell apoptosis was detected using TUNEL staining and flow cytometry. The correlation between interferon regulatory factor 4 (IRF4) and interleukin (IL)-9 was detected. Our results showed down-regulated miR-38 and up-regulated IRF4 in MCAO rats. Besides, IRF4 was targeted by miR-381 in neuron cells. Dex and overexpressed miR-381, or silenced IRF4 improved the neurological function and inhibited neuron cell apoptosis in MCAO rats. Additionally, in MCAO rats, Dex was found to increase the miR-381 expression and reduced IRF4 expression to decrease the IL-9 expression, which suppressed the inflammatory response and cell apoptosis both in vivo and in vitro. Importantly, our study demonstrated that Dex elevated the expression of miR-381, which ultimately results in the inhibition of inflammation response in rats with cerebral ischaemic injury.
Collapse
Affiliation(s)
- Hua Fang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Hua-Feng Li
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jian-Yong Yan
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Miao Yang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| | - Jian-Ping Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China.,Department of Anesthesiology, Guizhou University People's Hospital, Guiyang, China.,Laboratory of Anesthesiology & Perioperative Medicine, Guizhou University School of Medicine, Guiyang, China
| |
Collapse
|
32
|
Shultz SR, McDonald SJ, Corrigan F, Semple BD, Salberg S, Zamani A, Jones NC, Mychasiuk R. Clinical Relevance of Behavior Testing in Animal Models of Traumatic Brain Injury. J Neurotrauma 2020; 37:2381-2400. [DOI: 10.1089/neu.2018.6149] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sandy R. Shultz
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J. McDonald
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Melbourne, Victoria, Australia
| | - Frances Corrigan
- Department of Anatomy, University of South Australia, Adelaide, South Australia, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Sabrina Salberg
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
| | - Nigel C. Jones
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, Victoria, Australia
- Department of Psychology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
33
|
Chaban V, Clarke GJ, Skandsen T, Islam R, Einarsen CE, Vik A, Damås JK, Mollnes TE, Håberg AK, Pischke SE. Systemic Inflammation Persists the First Year after Mild Traumatic Brain Injury: Results from the Prospective Trondheim Mild Traumatic Brain Injury Study. J Neurotrauma 2020; 37:2120-2130. [PMID: 32326805 PMCID: PMC7502683 DOI: 10.1089/neu.2019.6963] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Innate immune activation has been attributed a key role in traumatic brain injury (TBI) and successive morbidity. In mild TBI (mTBI), however, the extent and persistence of innate immune activation are unknown. We determined plasma cytokine level changes over 12 months after an mTBI in hospitalized and non-hospitalized patients compared with community controls; and examined their associations to injury-related and demographic variables at admission. Prospectively, 207 patients presenting to the emergency department (ED) or general practitioner with clinically confirmed mTBI and 82 matched community controls were included. Plasma samples were obtained at admission, after 2 weeks, 3 months, and 12 months. Cytokine levels were analysed with a 27-plex beads-based immunoassay. Brain magnetic resonance imaging (MRI) was performed on all participants. Twelve cytokines were reliably detected. Plasma levels of interferon gamma (IFN-γ), interleukin 8 (IL-8), eotaxin, macrophage inflammatory protein-1-beta (MIP-1β), monocyte chemoattractant protein 1 (MCP-1), IL-17A, IL-9, tumor necrosis factor (TNF), and basic fibroblast growth factor (FGF-basic) were significantly increased at all time-points in patients compared with controls, whereas IFN-γ-inducing protein 10 (IP-10), platelet-derived growth factor (PDGF), and IL-1ra were not. IL-17A and FGF-basic showed significant increases in patients from admission to follow-up at 3 months, and remained increased at 12 months compared with admission. Interestingly, MRI findings were negatively associated with four cytokines: eotaxin, MIP-1β, IL-9, and IP-10, whereas age was positively associated with nine cytokines: IL-8, eotaxin, MIP-1β, MCP-1, IL-17A, IL-9, TNF, FGF-basic, and IL-1ra. TNF was also increased in those with presence of other injuries. In conclusion, mTBI activated the innate immune system consistently and this is the first study to show that several inflammatory cytokines remain increased for up to 1 year post-injury.
Collapse
Affiliation(s)
- Viktoriia Chaban
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Gerard J.B. Clarke
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Toril Skandsen
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Physical Medicine and Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Rakibul Islam
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Cathrine E. Einarsen
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Physical Medicine and Rehabilitation, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Anne Vik
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Neurosurgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Jan K. Damås
- Center of Molecular Inflammation Research, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Infectious Diseases, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tom E. Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Department of Clinical and Molecular Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Research Laboratory, Nordland Hospital Bodø, and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway
| | - Asta K. Håberg
- Department of Neuromedicine and Movement Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Soeren E. Pischke
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway
- Clinic for Emergencies and Critical Care, Oslo University Hospital and University of Oslo, Oslo, Norway
| |
Collapse
|
34
|
León-Moreno LC, Castañeda-Arellano R, Aguilar-García IG, Desentis-Desentis MF, Torres-Anguiano E, Gutiérrez-Almeida CE, Najar-Acosta LJ, Mendizabal-Ruiz G, Ascencio-Piña CR, Dueñas-Jiménez JM, Rivas-Carrillo JD, Dueñas-Jiménez SH. Kinematic Changes in a Mouse Model of Penetrating Hippocampal Injury and Their Recovery After Intranasal Administration of Endometrial Mesenchymal Stem Cell-Derived Extracellular Vesicles. Front Cell Neurosci 2020; 14:579162. [PMID: 33192324 PMCID: PMC7533596 DOI: 10.3389/fncel.2020.579162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Locomotion speed changes appear following hippocampal injury. We used a hippocampal penetrating brain injury mouse model to analyze other kinematic changes. We found a significant decrease in locomotion speed in both open-field and tunnel walk tests. We described a new quantitative method that allows us to analyze and compare the displacement curves between mice steps. In the tunnel walk, we marked mice with indelible ink on the knee, ankle, and metatarsus of the left and right hindlimbs to evaluate both in every step. Animals with hippocampal damage exhibit slower locomotion speed in both hindlimbs. In contrast, in the cortical injured group, we observed significant speed decrease only in the right hindlimb. We found changes in the displacement patterns after hippocampal injury. Mesenchymal stem cell-derived extracellular vesicles had been used for the treatment of several diseases in animal models. Here, we evaluated the effects of intranasal administration of endometrial mesenchymal stem cell-derived extracellular vesicles on the outcome after the hippocampal injury. We report the presence of vascular endothelial growth factor, granulocyte–macrophage colony-stimulating factor, and interleukin 6 in these vesicles. We observed locomotion speed and displacement pattern preservation in mice after vesicle treatment. These mice had lower pyknotic cells percentage and a smaller damaged area in comparison with the nontreated group, probably due to angiogenesis, wound repair, and inflammation decrease. Our results build up on the evidence of the hippocampal role in walk control and suggest that the extracellular vesicles could confer neuroprotection to the damaged hippocampus.
Collapse
Affiliation(s)
- Lilia Carolina León-Moreno
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico.,Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Rolando Castañeda-Arellano
- Laboratory of Tissue Engineering and Transplant, Department of Physiology, cGMP Cell Processing Facility, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Irene Guadalupe Aguilar-García
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | | | - Elizabeth Torres-Anguiano
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Coral Estefanía Gutiérrez-Almeida
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Luis Jesús Najar-Acosta
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Gerardo Mendizabal-Ruiz
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - César Rodolfo Ascencio-Piña
- Department of Computer Sciences, University Center of Exact Sciences and Engineering, University of Guadalajara, Guadalajara, Mexico
| | - Judith Marcela Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Jorge David Rivas-Carrillo
- Department of Biomedical Sciences, University Center of Tonala, University of Guadalajara, Guadalajara, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Laboratory of Neurophysiology, Department of Neuroscience, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
35
|
Diaz-Chávez A, Lajud N, Roque A, Cheng JP, Meléndez-Herrera E, Valdéz-Alarcón JJ, Bondi CO, Kline AE. Early life stress increases vulnerability to the sequelae of pediatric mild traumatic brain injury. Exp Neurol 2020; 329:113318. [PMID: 32305419 DOI: 10.1016/j.expneurol.2020.113318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
Early life stress (ELS) is a risk factor for many psychopathologies that happen later in life. Although stress can occur in cases of child abuse, studies on non-accidental brain injuries in pediatric populations do not consider the possible increase in vulnerability caused by ELS. Hence, we sought to determine whether ELS increases the effects of pediatric mild traumatic brain injury (mTBI) on cognition, hippocampal inflammation, and plasticity. Male rats were subjected to maternal separation for 180 min per day (MS180) or used as controls (CONT) during the first 21 post-natal (P) days. At P21 the rats were anesthetized with isoflurane and subjected to a mild controlled cortical impact or sham injury. At P32 the rats were injected with the cell proliferation marker bromodeoxyuridine (BrdU, 500 mg/kg), then evaluated for spatial learning and memory in a water maze (P35-40) and sacrificed for quantification of Ki67+, BrdU+ and Iba1+ (P42). Neither MS180 nor mTBI impacted cognitive outcome when provided alone but their combination (MS180 + mTBI) decreased spatial learning and memory relative to Sham controls (p < .01). mTBI increased microglial activation and affected BrdU+ cell survival in the ipsilateral hippocampus without affecting proliferation rates. However, only MS180 + mTBI increased microglial activation in the area adjacent to the injury and the contralateral CA1 hippocampal subfield, and decreased cell proliferation in the ipsilateral neurogenic niche. Overall, the data show that ELS increases the vulnerability to the sequelae of pediatric mTBI and may be mediated by increased neuroinflammation.
Collapse
Affiliation(s)
- Arturo Diaz-Chávez
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Jeffrey P Cheng
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Juan José Valdéz-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Corina O Bondi
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Neurobiology, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Anthony E Kline
- Physical Medicine & Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America; Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America; Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States of America; Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America; Psychology, University of Pittsburgh, Pittsburgh, PA, United States of America.
| |
Collapse
|
36
|
Tian J, Popal MS, Huang R, Zhang M, Zhao X, Zhang M, Song X. Caveolin as a Novel Potential Therapeutic Target in Cardiac and Vascular Diseases: A Mini Review. Aging Dis 2020; 11:378-389. [PMID: 32257548 PMCID: PMC7069461 DOI: 10.14336/ad.2019.09603] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 06/03/2019] [Indexed: 12/27/2022] Open
Abstract
Caveolin, a structural protein of caveolae, play roles in the regulation of endothelial function, cellular lipid homeostasis, and cardiac function by affecting the activity and biogenesis of nitric oxide, and by modulating signal transduction pathways that mediate inflammatory responses and oxidative stress. In this review, we present the role of caveolin in cardiac and vascular diseases and the relevant signaling pathways involved. Furthermore, we discuss a novel therapeutic perspective comprising crosstalk between caveolin and autophagy.
Collapse
Affiliation(s)
- Jinfan Tian
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mohammad Sharif Popal
- 2 Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - RongChong Huang
- 3 Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100010, China
| | - Min Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xin Zhao
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mingduo Zhang
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Xiantao Song
- 1 Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
37
|
Neuroinflammation in CNS diseases: Molecular mechanisms and the therapeutic potential of plant derived bioactive molecules. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100176] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
38
|
Amaradhi R, Banik A, Mohammed S, Patro V, Rojas A, Wang W, Motati DR, Dingledine R, Ganesh T. Potent, Selective, Water Soluble, Brain-Permeable EP2 Receptor Antagonist for Use in Central Nervous System Disease Models. J Med Chem 2020; 63:1032-1050. [PMID: 31904232 PMCID: PMC7394479 DOI: 10.1021/acs.jmedchem.9b01218] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activation of prostanoid EP2 receptor exacerbates neuroinflammatory and neurodegenerative pathology in central nervous system diseases such as epilepsy, Alzheimer's disease, and cerebral aneurysms. A selective and brain-permeable EP2 antagonist will be useful to attenuate the inflammatory consequences of EP2 activation and to reduce the severity of these chronic diseases. We recently developed a brain-permeable EP2 antagonist 1 (TG6-10-1), which displayed anti-inflammatory and neuroprotective actions in rodent models of status epilepticus. However, this compound exhibited moderate selectivity to EP2, a short plasma half-life in rodents (1.7 h) and low aqueous solubility (27 μM), limiting its use in animal models of chronic disease. With lead-optimization studies, we have developed several novel EP2 antagonists with improved water solubility, brain penetration, high EP2 potency, and selectivity. These novel inhibitors suppress inflammatory gene expression induced by EP2 receptor activation in a microglial cell line, reinforcing the use of EP2 antagonists as anti-inflammatory agents.
Collapse
Affiliation(s)
- Radhika Amaradhi
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Avijit Banik
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Shabber Mohammed
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Vidyavathi Patro
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Asheebo Rojas
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Wenyi Wang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Damoder Reddy Motati
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Ray Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| | - Thota Ganesh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Rd; Atlanta, GA, 30322, United States of America
| |
Collapse
|
39
|
Haddad D, Al Madhoun A, Nizam R, Al-Mulla F. Role of Caveolin-1 in Diabetes and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9761539. [PMID: 32082483 PMCID: PMC7007939 DOI: 10.1155/2020/9761539] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/10/2019] [Accepted: 12/26/2019] [Indexed: 12/25/2022]
Abstract
It is estimated that in 2017 there were 451 million people with diabetes worldwide. These figures are expected to increase to 693 million by 2045; thus, innovative preventative programs and treatments are a necessity to fight this escalating pandemic disorder. Caveolin-1 (CAV1), an integral membrane protein, is the principal component of caveolae in membranes and is involved in multiple cellular functions such as endocytosis, cholesterol homeostasis, signal transduction, and mechanoprotection. Previous studies demonstrated that CAV1 is critical for insulin receptor-mediated signaling, insulin secretion, and potentially the development of insulin resistance. Here, we summarize the recent progress on the role of CAV1 in diabetes and diabetic complications.
Collapse
Affiliation(s)
- Dania Haddad
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Ashraf Al Madhoun
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait City, Kuwait
| |
Collapse
|
40
|
Janatpour ZC, Korotcov A, Bosomtwi A, Dardzinski BJ, Symes AJ. Subcutaneous Administration of Angiotensin-(1-7) Improves Recovery after Traumatic Brain Injury in Mice. J Neurotrauma 2019; 36:3115-3131. [DOI: 10.1089/neu.2019.6376] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Zachary C. Janatpour
- Department of Pharmacology and Molecular Therapeutics, Program in Molecular and Cell Biology, Uniformed Services University, Bethesda, Maryland
| | - Alexandru Korotcov
- Translational Imaging Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, Maryland
| | - Asamoah Bosomtwi
- Translational Imaging Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, Maryland
| | - Bernard J. Dardzinski
- Translational Imaging Core, Center for Neuroscience and Regenerative Medicine, Uniformed Services University, Bethesda, Maryland
- Department of Radiology and Radiological Sciences, Uniformed Services University, Bethesda, Maryland
| | - Aviva J. Symes
- Department of Pharmacology and Molecular Therapeutics, Program in Molecular and Cell Biology, Uniformed Services University, Bethesda, Maryland
| |
Collapse
|
41
|
Egawa J, Zemljic-Harpf A, Mandyam CD, Niesman IR, Lysenko LV, Kleschevnikov AM, Roth DM, Patel HH, Patel PM, Head BP. Neuron-Targeted Caveolin-1 Promotes Ultrastructural and Functional Hippocampal Synaptic Plasticity. Cereb Cortex 2019; 28:3255-3266. [PMID: 28981594 DOI: 10.1093/cercor/bhx196] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Indexed: 12/15/2022] Open
Abstract
A delicate interneuronal communication between pre- and postsynaptic membranes is critical for synaptic plasticity and the formation of memory. Evidence shows that membrane/lipid rafts (MLRs), plasma membrane microdomains enriched in cholesterol and sphingolipids, organize presynaptic proteins and postsynaptic receptors necessary for synaptic formation and signaling. MLRs establish a cell polarity that facilitates transduction of extracellular cues to the intracellular environment. Here we show that neuron-targeted overexpression of an MLR protein, caveolin-1 (SynCav1), in the adult mouse hippocampus increased the number of presynaptic vesicles per bouton, total excitatory type I glutamatergic synapses, number of same-dendrite multiple-synapse boutons, increased myelination, increased long-term potentiation, and increased MLR-localized N-methyl-d-aspartate receptor subunits (GluN1, GluN2A, and GluN2B). Immunogold electron microscopy revealed that Cav-1 localizes to both the pre- and postsynaptic membrane regions as well as in the synaptic cleft. These findings, which are consistent with a significant increase in ultrastructural and functional synaptic plasticity, provide a fundamental framework that underlies previously demonstrated improvements in learning and memory in adult and aged mice by SynCav1. Such observations suggest that Cav-1 and MLRs alter basic aspects of synapse biology that could serve as potential therapeutic targets to promote neuroplasticity and combat neurodegeneration in a number of neurological disorders.
Collapse
Affiliation(s)
- Junji Egawa
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Alice Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chitra D Mandyam
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | | | - Larisa V Lysenko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | | | - David M Roth
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hemal H Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Piyush M Patel
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brian P Head
- Veterans Affairs San Diego Healthcare System, San Diego, CA, USA.,Department of Anesthesiology, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
42
|
Dissemination of brain inflammation in traumatic brain injury. Cell Mol Immunol 2019; 16:523-530. [PMID: 30846842 DOI: 10.1038/s41423-019-0213-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) is recognized as a global health problem due to its increasing occurrence, challenging treatment, and persistent impacts on brain pathophysiology. Neural cell death in patients with TBI swiftly causes inflammation in the injured brain areas, which is recognized as focal brain inflammation. Focal brain inflammation causes secondary brain injury by exacerbating brain edema and neuronal death, while also exerting divergent beneficial effects, such as sealing the damaged limitans and removing cellular debris. Recent evidence from patients with TBI and studies on animal models suggest that brain inflammation after TBI is not only restricted to the focal lesion but also disseminates to remote areas of the brain. The dissemination of inflammation has been detected within days after the primary injury and persists chronically. This state of inflammation may be related to remote complications of TBI in patients, such as hyperthermia and hypopituitarism, and may lead to progressive neurodegeneration, such as chronic traumatic encephalopathy. Future studies should focus on understanding the mechanisms that govern the initiation and propagation of brain inflammation after TBI and its impacts on post-trauma brain pathology.
Collapse
|
43
|
Huang Q, Zhong W, Hu Z, Tang X. A review of the role of cav-1 in neuropathology and neural recovery after ischemic stroke. J Neuroinflammation 2018; 15:348. [PMID: 30572925 PMCID: PMC6302517 DOI: 10.1186/s12974-018-1387-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke starts a series of pathophysiological processes that cause brain injury. Caveolin-1 (cav-1) is an integrated protein and locates at the caveolar membrane. It has been demonstrated that cav-1 can protect blood–brain barrier (BBB) integrity by inhibiting matrix metalloproteases (MMPs) which degrade tight junction proteins. This article reviews recent developments in understanding the mechanisms underlying BBB dysfunction, neuroinflammation, and oxidative stress after ischemic stroke, and focuses on how cav-1 modulates a series of activities after ischemic stroke. In general, cav-1 reduces BBB permeability mainly by downregulating MMP9, reduces neuroinflammation through influencing cytokines and inflammatory cells, promotes nerve regeneration and angiogenesis via cav-1/VEGF pathway, reduces apoptosis, and reduces the damage mediated by oxidative stress. In addition, we also summarize some experimental results that are contrary to the above and explore possible reasons for these differences.
Collapse
Affiliation(s)
- Qianyi Huang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Wei Zhong
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Zhiping Hu
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China
| | - Xiangqi Tang
- Department of Neurology, The Second Xiangya Hospital, Central South University, Renmin Road 139#, Changsha, 410011, Hunan, China.
| |
Collapse
|
44
|
Zhang XY, Xu ZP, Wang W, Cao JB, Fu Q, Zhao WX, Li Y, Huo XL, Zhang LM, Li YF, Mi WD. Vitamin C alleviates LPS-induced cognitive impairment in mice by suppressing neuroinflammation and oxidative stress. Int Immunopharmacol 2018; 65:438-447. [DOI: 10.1016/j.intimp.2018.10.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 02/08/2023]
|
45
|
Chen Z, Nie SD, Qu ML, Zhou D, Wu LY, Shi XJ, Ma LR, Li X, Zhou SL, Wang S, Wu J. The autophagic degradation of Cav-1 contributes to PA-induced apoptosis and inflammation of astrocytes. Cell Death Dis 2018; 9:771. [PMID: 29991726 PMCID: PMC6039485 DOI: 10.1038/s41419-018-0795-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 12/23/2022]
Abstract
The accumulation of palmitic acid (PA), implicated in obesity, can induce apoptotic cell death and inflammation of astrocytes. Caveolin-1 (Cav-1), an essential protein for astrocytes survival, can be degraded by autophagy, which is a double-edge sword that can either promote cell survival or cell death. The aim of this study was to delineate whether the autophagic degradation of Cav-1 is involved in PA-induced apoptosis and inflammation in hippocampal astrocytes. In this study we found that: (1) PA caused apoptotic death and inflammation by autophagic induction; (2) Cav-1 was degraded by PA-induced autophagy and PA induced autophagy in a Cav-1-independent manner; (3) the degradation of Cav-1 was responsible for PA-induced autophagy-dependent apoptotic cell death and inflammation; (4) chronic high-fat diet (HFD) induced Cav-1 degradation, apoptosis, autophagy, and inflammation in the hippocampal astrocytes of rats. Our results suggest that the autophagic degradation of Cav-1 contributes to PA-induced apoptosis and inflammation of astrocytes. Therefore, Cav-1 may be a potential therapeutic target for central nervous system injuries caused by PA accumulation.
Collapse
Affiliation(s)
- Zi Chen
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Sheng-Dan Nie
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Min-Li Qu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Di Zhou
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Liang-Yan Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xia-Jie Shi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Ling-Ran Ma
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Xin Li
- Institute of Clinical Medicine, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Shan-Lei Zhou
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Shan Wang
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| | - Jing Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.
| |
Collapse
|
46
|
The PKC-β selective inhibitor, Enzastaurin, impairs memory in middle-aged rats. PLoS One 2018; 13:e0198256. [PMID: 29870545 PMCID: PMC5988320 DOI: 10.1371/journal.pone.0198256] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/16/2018] [Indexed: 01/14/2023] Open
Abstract
Enzastaurin is a Protein Kinase C-β selective inhibitor that was developed to treat cancers. Protein Kinase C-β is an important enzyme for a variety of neuronal functions; in particular, previous rodent studies have reported deficits in spatial and fear-conditioned learning and memory with lower levels of Protein Kinase C-β. Due to Enzastaurin's mechanism of action, the present study investigated the consequences of Enzastaurin exposure on learning and memory in 12-month-old Fischer-344 male rats. Rats were treated daily with subcutaneous injections of either vehicle or Enzastaurin, and behaviorally tested using the spatial reference memory Morris Water Maze. Rats treated with Enzastaurin exhibited decreased overnight retention and poorer performance on the latter testing day, indicating a mild, but significant, memory impairment. There were no differences during the probe trial indicating that all animals were able to spatially localize the platform to the proper quadrant by the end of testing. RNA isolated from the hippocampus was analyzed using Next Generation Sequencing (Illumina). No statistically significant transcriptional differences were noted. Our findings suggest that acute Enzastaurin treatment can impair hippocampal-based learning and memory performance, with no effects on transcription in the hippocampus. We propose that care should be taken in future clinical trials that utilize Protein Kinase C-ß inhibitors, to monitor for possible cognitive effects, future research should examine if these effects are fully reversible.
Collapse
|
47
|
Ma MW, Wang J, Dhandapani KM, Wang R, Brann DW. NADPH oxidases in traumatic brain injury - Promising therapeutic targets? Redox Biol 2018; 16:285-293. [PMID: 29571125 PMCID: PMC5952873 DOI: 10.1016/j.redox.2018.03.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/22/2022] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Despite intense investigation, no neuroprotective agents for TBI have yet translated to the clinic. Recent efforts have focused on identifying potential therapeutic targets that underlie the secondary TBI pathology that evolves minutes to years following the initial injury. Oxidative stress is a key player in this complex cascade of secondary injury mechanisms and prominently contributes to neurodegeneration and neuroinflammation. NADPH oxidase (NOX) is a family of enzymes whose unique function is to produce reactive oxygen species (ROS). Human post-mortem and animal studies have identified elevated NOX2 and NOX4 levels in the injured brain, suggesting that these two NOXs are involved in the pathogenesis of TBI. In support of this, NOX2 and NOX4 deletion studies have collectively revealed that targeting NOX enzymes can reduce oxidative stress, attenuate neuroinflammation, promote neuronal survival, and improve functional outcomes following TBI. In addition, NOX inhibitor studies have confirmed these findings and demonstrated an extended critical window of efficacious TBI treatment. Finally, the translational potential, caveats, and future directions of the field are highlighted and discussed throughout the review.
Collapse
Affiliation(s)
- Merry W Ma
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ruimin Wang
- Department of Neurobiology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
48
|
Mufson EJ, He B, Ginsberg SD, Carper BA, Bieler GS, Crawford F, Alvarez VE, Huber BR, Stein TD, McKee AC, Perez SE. Gene Profiling of Nucleus Basalis Tau Containing Neurons in Chronic Traumatic Encephalopathy: A Chronic Effects of Neurotrauma Consortium Study. J Neurotrauma 2018; 35:1260-1271. [PMID: 29338612 PMCID: PMC5962931 DOI: 10.1089/neu.2017.5368] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Military personnel and athletes exposed to traumatic brain injury may develop chronic traumatic encephalopathy (CTE). Brain pathology in CTE includes intracellular accumulation of abnormally phosphorylated tau proteins (p-tau), the main constituent of neurofibrillary tangles (NFTs). Recently, we found that cholinergic basal forebrain (CBF) neurons within the nucleus basalis of Meynert (nbM), which provide the major cholinergic innervation to the cortex, display an increased number of NFTs across the pathological stages of CTE. However, molecular mechanisms underlying nbM neurodegeneration in the context of CTE pathology remain unknown. Here, we assessed the genetic signature of nbM neurons containing the p-tau pretangle maker pS422 from CTE subjects who came to autopsy and received a neuropathological CTE staging assessment (Stages II, III, and IV) using laser capture microdissection and custom-designed microarray analysis. Quantitative analysis revealed dysregulation of key genes in several gene ontology groups between CTE stages. Specifically, downregulation of the nicotinic cholinergic receptor subunit β-2 gene (CHRNB2), monoaminergic enzymes catechol-O-methyltransferase (COMT) and dopa decarboxylase (DDC), chloride channels CLCN4 and CLCN5, scaffolding protein caveolin 1 (CAV1), cortical development/cytoskeleton element lissencephaly 1 (LIS1), and intracellular signaling cascade member adenylate cyclase 3 (ADCY3) was observed in pS422-immunreactive nbM neurons in CTE patients. By contrast, upregulation of calpain 2 (CAPN2) and microtubule-associated protein 2 (MAP2) transcript levels was found in Stage IV CTE patients. These single-population data in vulnerable neurons indicate alterations in gene expression associated with neurotransmission, signal transduction, the cytoskeleton, cell survival/death signaling, and microtubule dynamics, suggesting novel molecular pathways to target for drug discovery in CTE.
Collapse
Affiliation(s)
- Elliott J. Mufson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| | - Bin He
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| | - Stephen D. Ginsberg
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, New York and NYU Medical Center, New York, New York
| | | | | | | | - Victor E. Alvarez
- VA Boston HealthCare System, Boston University School of Medicine, Boston, Massachusetts
- Alzheimer Disease Center and CTE Center Program, Boston University School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Bertrand R. Huber
- VA Boston HealthCare System, Boston University School of Medicine, Boston, Massachusetts
- Alzheimer Disease Center and CTE Center Program, Boston University School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
| | - Thor D. Stein
- VA Boston HealthCare System, Boston University School of Medicine, Boston, Massachusetts
- Alzheimer Disease Center and CTE Center Program, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts
| | - Ann C. McKee
- VA Boston HealthCare System, Boston University School of Medicine, Boston, Massachusetts
- Alzheimer Disease Center and CTE Center Program, Boston University School of Medicine, Boston, Massachusetts
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts
| | - Sylvia E. Perez
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona
| |
Collapse
|
49
|
Wu J, Zhou SL, Pi LH, Shi XJ, Ma LR, Chen Z, Qu ML, Li X, Nie SD, Liao DF, Pei JJ, Wang S. High glucose induces formation of tau hyperphosphorylation via Cav-1-mTOR pathway: A potential molecular mechanism for diabetes-induced cognitive dysfunction. Oncotarget 2018; 8:40843-40856. [PMID: 28489581 PMCID: PMC5522306 DOI: 10.18632/oncotarget.17257] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/07/2017] [Indexed: 12/31/2022] Open
Abstract
The abnormally hyperphosphorylated tau is thought to be implicated in diabetes-associated cognitive deficits. The role of mammalian target of rapamycin (mTOR) / S6 kinase (S6K) signalling in the formation of tau hyperphosphorylation has been previously studied. Caveolin-1 (Cav-1), the essential structure protein of caveolae, promotes neuronal survival and growth, and inhibits glucose metabolism. In this study, we aimed to investigate the role of Cav-1 in the formation of tau hyperphosphorylation under chronic hyperglycemic condition (HGC). Diabetic rats were induced by streptozotocin (STZ). Primary hippocampal neurons with or without molecular intervention such as the transient over-expression or knock-down were subjected to HGC. The obtained experimental samples were analyzed by real time quantitative RT-PCR, Western blot, immunofluorescence or immunohistochemisty. We found: 1) that a chronic HGC directly decreases Cav-1 expression, increases tau phosphorylation and activates mTOR/S6K signalling in the brain neurons of diabetic rats, 2) that overexpression of Cav-1 attenuates tau hyperphosphorylation induced by chronic HGC in primary hippocampal neurons, whereas down-regulation of Cav-1 using Cav-1 siRNA dramatically worsens tau hyperphosphorylation via mTOR/S6K signalling pathway, and 3) that the down-regulation of Cav-1 induced by HGC is independent of mTOR signalling. Our results suggest that tau hyperphosphorylation and the sustained over-activated mTOR signalling under hyperglycemia may be due to the suppression of Cav-1. Therefore, Cav-1 is a potential therapeutic target for diabetes-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Jing Wu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Shan-Lei Zhou
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Lin-Hua Pi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xia-Jie Shi
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Ling-Ran Ma
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Zi Chen
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Min-Li Qu
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xin Li
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Sheng-Dan Nie
- Institute of Clinical Medicine, People's Hospital of Hunan province, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, School of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Jin-Jing Pei
- KI-Alzheimer's Disease Research Center, Karolinska Institutet, Novum, Stockholm, Sweden.,Department of Neurology, Xuan Wu Hospital, Capital Medical University, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - Shan Wang
- Department of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.,Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| |
Collapse
|
50
|
Li L, Saiyin H, Xie J, Ma L, Xue L, Wang W, Liang W, Yu Q. Sevoflurane preconditioning induced endogenous neurogenesis against ischemic brain injury by promoting microglial activation. Oncotarget 2018; 8:28544-28557. [PMID: 28212538 PMCID: PMC5438671 DOI: 10.18632/oncotarget.15325] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/10/2017] [Indexed: 01/15/2023] Open
Abstract
Brain ischemia causes irreversible damage to functional neurons in cases of infarct. Promoting endogenous neurogenesis to replace necrotic neurons is a promising therapeutic strategy for ischemia patients. The neuroprotective role of sevoflurane preconditioning implies that it might also enhance endogenous neurogenesis and functional restoration in the infarct region. By using a transient middle cerebral artery occlusion (tMCAO) model, we discovered that endogenous neurogenesis was enhanced by sevoflurane preconditioning. This enhancement process is characterized by the promotion of neuroblast proliferation within the subventricular zone (SVZ), migration and differentiation into neurons, and the presence of astrocytes and oligodendrocytes at the site of infarct. The newborn neurons in the sevoflurane preconditioning group showed miniature excitatory postsynaptic currents (mEPSCs), increased synaptophysin and PSD95 staining density, indicating normal neuronal function. Furthermore, long-term behavioral improvement was observed in the sevoflurane preconditioning group consistent with endogenous neurogenesis. Further histological analyses showed that sevoflurane preconditioning accelerated microglial activation, including migration, phagocytosis and secretion of brain-derived neurotrophic factor (BDNF). Intraperitoneal injection of minocycline, a microglial inhibitor, suppressed microglial activation and reversed neurogenesis. Our data showed that sevoflurane preconditioning promoted microglial activities, created a favorable microenvironment for endogenous neurogenesis and accelerated functional reconstruction in the infarct region.
Collapse
Affiliation(s)
- Li Li
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Hexige Saiyin
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Jingmo Xie
- Department of Anatomy, Histology & Embryology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lixiang Ma
- Department of Anatomy, Histology & Embryology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Lei Xue
- Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Wei Wang
- Department of Physiology and Biophysics, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Weimin Liang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qiong Yu
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|