1
|
Viegas A, Araújo R, Ramalhete L, Von Rekowski C, Fonseca TAH, Bento L, Calado CRC. Discovery of Delirium Biomarkers through Minimally Invasive Serum Molecular Fingerprinting. Metabolites 2024; 14:301. [PMID: 38921436 PMCID: PMC11205956 DOI: 10.3390/metabo14060301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Delirium presents a significant clinical challenge, primarily due to its profound impact on patient outcomes and the limitations of the current diagnostic methods, which are largely subjective. During the COVID-19 pandemic, this challenge was intensified as the frequency of delirium assessments decreased in Intensive Care Units (ICUs), even as the prevalence of delirium among critically ill patients increased. The present study evaluated how the serum molecular fingerprint, as acquired by Fourier-Transform InfraRed (FTIR) spectroscopy, can enable the development of predictive models for delirium. A preliminary univariate analysis of serum FTIR spectra indicated significantly different bands between 26 ICU patients with delirium and 26 patients without, all of whom were admitted with COVID-19. However, these bands resulted in a poorly performing Naïve-Bayes predictive model. Considering the use of a Fast-Correlation-Based Filter for feature selection, it was possible to define a new set of spectral bands with a wider coverage of molecular functional groups. These bands ensured an excellent Naïve-Bayes predictive model, with an AUC, a sensitivity, and a specificity all exceeding 0.92. These spectral bands, acquired through a minimally invasive analysis and obtained rapidly, economically, and in a high-throughput mode, therefore offer significant potential for managing delirium in critically ill patients.
Collapse
Affiliation(s)
- Ana Viegas
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Avenida D. João II, Lote 4.58.01, 1990-096 Lisbon, Portugal;
- Neurosciences Area, Clinical Neurophysiology Unit, ULSSJ—Unidade Local de Saúde São José, Rua José António Serrano, 1150-199 Lisbon, Portugal
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (R.A.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Rúben Araújo
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (R.A.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Luís Ramalhete
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Blood and Transplantation Center of Lisbon, Instituto Português do Sangue e da Transplantação, Alameda das Linhas de Torres, n° 117, 1769-001 Lisboa, Portugal
- iNOVA4Health—Advancing Precision Medicine, RG11: Reno-Vascular Diseases Group, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
| | - Cristiana Von Rekowski
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (R.A.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Tiago A. H. Fonseca
- CHRC—Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082 Lisbon, Portugal; (R.A.)
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
| | - Luís Bento
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1169-056 Lisbon, Portugal
- Intensive Care Department, ULSSJ—Unidade Local de Saúde São José, Rua José António Serrano, 1150-199 Lisbon, Portugal
- Integrated Pathophysiological Mechanisms, CHRC—Comprehensive Health Research Centre, NMS—NOVA Medical School, FCM—Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal
| | - Cecília R. C. Calado
- ISEL—Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, 1959-007 Lisbon, Portugal
- iBB—Institute for Bioengineering and Biosciences, The Associate Laboratory Institute for Health and Bioeconomy (i4HB), Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
2
|
Jones TB, Mackey T, Juba AN, Amin K, Atyam A, McDole M, Yancy J, Thomas TC, Buhlman LM. Mild traumatic brain injury in Drosophila melanogaster alters reactive oxygen and nitrogen species in a sex-dependent manner. Exp Neurol 2024; 372:114621. [PMID: 38029809 PMCID: PMC10872660 DOI: 10.1016/j.expneurol.2023.114621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
Traumatic brain injury (TBI) is an outside force causing a modification in brain function and/or structural brain pathology that upregulates brain inducible nitric oxide synthase (iNOS), instigating increased levels of nitric oxide activity which is implicated in secondary pathology leading to behavioral deficits (Hall et al., 2012; Garry et al., 2015; Kozlov et al., 2017). In mammals, TBI-induced NO production activates an immune response and potentiates metabolic crisis through mitochondrial dysfunction coupled with vascular dysregulation; however, the direct influence on pathology is complicated by the activation of numerous secondary cascades and activation of other reactive oxygen species. Drosophila TBI models have demonstrated key features of mammalian TBI, including temporary incapacitation, disorientation, motor deficits, activation of innate immunity (inflammation), and autophagy responses observed immediately after injury (Katzenberger et al., 2013; Barekat et al., 2016; Simon et al., 2017; Anderson et al., 2018; Buhlman et al., 2021b). We hypothesized that acute behavioral phenotypes would be associated with deficits in climbing behavior and increased oxidative stress. Because flies lack mammalian-like cardiovascular and adaptive immune systems, we were able to make our observations in the absence of vascular disruption and adaptive immune system interference in a system where highly targeted interventions can be rapidly evaluated. To demonstrate the induction of injury, ten-day-old transgenic flies received an injury of increasing angles from a modified high impact trauma (HIT) device where angle-dependent increases occurred for acute neurological behavior assessments and twenty-four-hour mortality, and survival was significantly decreased. Injury caused sex-dependent effects on climbing activity and measures of oxidative stress. Specifically, after a single 60-degree HIT, female flies exhibited significant impairments in climbing activity beyond that observed in male flies. We also found that several measures of oxidative stress, including Drosophila NOS (dNOS) expression, protein nitration, and hydrogen peroxide production were significantly decreased in female flies. Interestingly, protein nitration was also decreased in males, but surpassed sham levels with a more severe injury. We also observed decreased autophagy demand in vulnerable dopaminergic neurons in female, but not male flies. In addition, mitophagy initiation was decreased in females. Collectively, our data suggest that TBI in flies induces acute behavioral phenotypes and climbing deficits that are analogous to mammalian TBI. We also observed that various indices of oxidative stress, including dNOS expression, protein tyrosine nitration, and hydrogen peroxide levels, as well as basal levels of autophagy, are altered in response to injury, an effect that is more pronounced in female flies.
Collapse
Affiliation(s)
- T Bucky Jones
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA; Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Tracy Mackey
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Amber N Juba
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA
| | - Kush Amin
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Amruth Atyam
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Madison McDole
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Jarod Yancy
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Phoenix VA Health Care System, Phoenix, AZ, USA.
| | - Lori M Buhlman
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
3
|
Doenyas-Barak K, Kutz I, Lang E, Merzbach R, Lev Wiesel R, Boussi-Gross R, Efrati S. The use of hyperbaric oxygen for veterans with PTSD: basic physiology and current available clinical data. Front Neurosci 2023; 17:1259473. [PMID: 38027524 PMCID: PMC10630921 DOI: 10.3389/fnins.2023.1259473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) affects up to 30% of veterans returning from the combat zone. Unfortunately, a substantial proportion of them do not remit with the current available treatments and thus continue to experience long-term social, behavioral, and occupational dysfunction. Accumulating data implies that the long-standing unremitting symptoms are related to changes in brain activity and structure, mainly disruption in the frontolimbic circuit. Hence, repair of brain structure and restoration of function could be a potential aim of effective treatment. Hyperbaric oxygen therapy (HBOT) has been effective in treating disruptions of brain structure and functions such as stroke, traumatic brain injury, and fibromyalgia even years after the acute insult. These favorable HBOT brain effects may be related to recent protocols that emphasize frequent fluctuations in oxygen concentrations, which in turn contribute to gene expression alterations and metabolic changes that induce neuronal stem cell proliferation, mitochondrial multiplication, angiogenesis, and regulation of the inflammatory cascade. Recently, clinical findings have also demonstrated the beneficial effect of HBOT on veterans with treatment-resistant PTSD. Moderation of intrusive symptoms, avoidance, mood and cognitive symptoms, and hyperarousal were correlated with improved brain function and with diffusion tensor imaging-defined structural changes. This article reviews the current data on the regenerative biological effects of HBOT, and the ongoing research of its use for veterans with PTSD.
Collapse
Affiliation(s)
- Keren Doenyas-Barak
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Kutz
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
| | - Erez Lang
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rachel Merzbach
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
- The Louis and Gabi Weisfeld School of Social Work, Bar-Ilan University, Ramat Gan, Israel
| | - Rachel Lev Wiesel
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
- The Emili Sagol Creative Arts Therapies Research Center, University of Haifa, Haifa, Israel
| | - Rahav Boussi-Gross
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
| | - Shai Efrati
- Sagol Center for Hyperbaric Medicine and Research, Shamir Medical Center, Zerifin, Israel
- School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Sanchez-Bezanilla S, Hood RJ, Collins-Praino LE, Turner RJ, Walker FR, Nilsson M, Ong LK. More than motor impairment: A spatiotemporal analysis of cognitive impairment and associated neuropathological changes following cortical photothrombotic stroke. J Cereb Blood Flow Metab 2021; 41:2439-2455. [PMID: 33779358 PMCID: PMC8393292 DOI: 10.1177/0271678x211005877] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
There is emerging evidence suggesting that a cortical stroke can cause delayed and remote hippocampal dysregulation, leading to cognitive impairment. In this study, we aimed to investigate motor and cognitive outcomes after experimental stroke, and their association with secondary neurodegenerative processes. Specifically, we used a photothrombotic stroke model targeting the motor and somatosensory cortices of mice. Motor function was assessed using the cylinder and grid walk tasks. Changes in cognition were assessed using a mouse touchscreen platform. Neuronal loss, gliosis and amyloid-β accumulation were investigated in the peri-infarct and ipsilateral hippocampal regions at 7, 28 and 84 days post-stroke. Our findings showed persistent impairment in cognitive function post-stroke, whilst there was a modest spontaneous motor recovery over the investigated period of 84 days. In the peri-infarct region, we detected a reduction in neuronal loss and decreased neuroinflammation over time post-stroke, which potentially explains the spontaneous motor recovery. Conversely, we observed persistent neuronal loss together with concomitant increased neuroinflammation and amyloid-β accumulation in the hippocampus, which likely accounts for the persistent cognitive dysfunction. Our findings indicate that cortical stroke induces secondary neurodegenerative processes in the hippocampus, a region remote from the primary infarct, potentially contributing to the progression of post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Rebecca J Hood
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Lyndsey E Collins-Praino
- Department of Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Renée J Turner
- Department of Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia.,Centre for Rehab Innovations, The University of Newcastle, Callaghan, NSW, Australia
| | - Michael Nilsson
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia.,Centre for Rehab Innovations, The University of Newcastle, Callaghan, NSW, Australia.,LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia.,Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| |
Collapse
|
5
|
Datta A, Sarmah D, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Advances in Studies on Stroke-Induced Secondary Neurodegeneration (SND) and Its Treatment. Curr Top Med Chem 2020; 20:1154-1168. [DOI: 10.2174/1568026620666200416090820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/23/2022]
Abstract
Background:
The occurrence of secondary neurodegeneration has exclusively been observed
after the first incidence of stroke. In humans and rodents, post-stroke secondary neurodegeneration
(SND) is an inevitable event that can lead to progressive neuronal loss at a region distant to initial infarct.
SND can lead to cognitive and motor function impairment, finally causing dementia. The exact
pathophysiology of the event is yet to be explored. It is seen that the thalami, in particular, are susceptible
to cause SND. The reason behind this is because the thalamus functioning as the relay center and is
positioned as an interlocked structure with direct synaptic signaling connection with the cortex. As SND
proceeds, accumulation of misfolded proteins and microglial activation are seen in the thalamus. This
leads to increased neuronal loss and worsening of functional and cognitive impairment.
Objective:
There is a necessity of specific interventions to prevent post-stroke SND, which are not properly
investigated to date owing to sparsely reproducible pre-clinical and clinical data. The basis of this
review is to investigate about post-stroke SND and its updated treatment approaches carefully.
Methods:
Our article presents a detailed survey of advances in studies on stroke-induced secondary neurodegeneration
(SND) and its treatment.
Results:
This article aims to put forward the pathophysiology of SND. We have also tabulated the latest
treatment approaches along with different neuroimaging systems that will be helpful for future reference
to explore.
Conclusion:
In this article, we have reviewed the available reports on SND pathophysiology, detection
techniques, and possible treatment modalities that have not been attempted to date.
Collapse
Affiliation(s)
- Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, India
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Kunjan R. Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Dileep R. Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| |
Collapse
|
6
|
Kitchen P, Salman MM, Halsey AM, Clarke-Bland C, MacDonald JA, Ishida H, Vogel HJ, Almutiri S, Logan A, Kreida S, Al-Jubair T, Winkel Missel J, Gourdon P, Törnroth-Horsefield S, Conner MT, Ahmed Z, Conner AC, Bill RM. Targeting Aquaporin-4 Subcellular Localization to Treat Central Nervous System Edema. Cell 2020; 181:784-799.e19. [PMID: 32413299 PMCID: PMC7242911 DOI: 10.1016/j.cell.2020.03.037] [Citation(s) in RCA: 303] [Impact Index Per Article: 60.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/09/2020] [Accepted: 03/17/2020] [Indexed: 01/07/2023]
Abstract
Swelling of the brain or spinal cord (CNS edema) affects millions of people every year. All potential pharmacological interventions have failed in clinical trials, meaning that symptom management is the only treatment option. The water channel protein aquaporin-4 (AQP4) is expressed in astrocytes and mediates water flux across the blood-brain and blood-spinal cord barriers. Here we show that AQP4 cell-surface abundance increases in response to hypoxia-induced cell swelling in a calmodulin-dependent manner. Calmodulin directly binds the AQP4 carboxyl terminus, causing a specific conformational change and driving AQP4 cell-surface localization. Inhibition of calmodulin in a rat spinal cord injury model with the licensed drug trifluoperazine inhibited AQP4 localization to the blood-spinal cord barrier, ablated CNS edema, and led to accelerated functional recovery compared with untreated animals. We propose that targeting the mechanism of calmodulin-mediated cell-surface localization of AQP4 is a viable strategy for development of CNS edema therapies.
Collapse
Affiliation(s)
- Philip Kitchen
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Mootaz M Salman
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pharmacology, College of Pharmacy, University of Mosul, Mosul 41002, Iraq
| | - Andrea M Halsey
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Charlotte Clarke-Bland
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Justin A MacDonald
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Hiroaki Ishida
- Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hans J Vogel
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Department of Biological Sciences, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Sharif Almutiri
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Clinical Laboratory Science, College of Applied Medical Science, Shaqra University, Shaqra, Saudi Arabia
| | - Ann Logan
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Stefan Kreida
- Department of Biochemistry and Structural Biology, Lund University, PO Box 124, 221 00 Lund, Sweden
| | - Tamim Al-Jubair
- Department of Biochemistry and Structural Biology, Lund University, PO Box 124, 221 00 Lund, Sweden
| | - Julie Winkel Missel
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Pontus Gourdon
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Department of Experimental Medical Science, Lund University, PO Box 118, 221 00 Lund, Sweden
| | | | - Matthew T Conner
- School of Sciences, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Alex C Conner
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Roslyn M Bill
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B4 7ET, UK.
| |
Collapse
|
7
|
Guennoun R, Zhu X, Fréchou M, Gaignard P, Slama A, Liere P, Schumacher M. Steroids in Stroke with Special Reference to Progesterone. Cell Mol Neurobiol 2019; 39:551-568. [PMID: 30302630 PMCID: PMC11469871 DOI: 10.1007/s10571-018-0627-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/05/2018] [Indexed: 12/21/2022]
Abstract
Both sex and steroid hormones are important to consider in human ischemic stroke and its experimental models. Stroke initiates a cascade of changes that lead to neural cell death, but also activates endogenous protective processes that counter the deleterious consequences of ischemia. Steroids may be part of these cerebroprotective processes. One option to provide cerebroprotection is to reinforce these intrinsic protective mechanisms. In the current review, we first summarize studies describing sex differences and the influence of steroid hormones in stroke. We then present and discuss our recent results concerning differential changes in endogenous steroid levels in the brains of male and female mice and the importance of progesterone receptors (PR) during the early phase after stroke. In the third part, we give an overview of experimental studies, including ours, that provide evidence for the pleiotropic beneficial effects of progesterone and its promising cerebroprotective potential in stroke. We also highlight the key role of PR signaling as well as potential additional mechanisms by which progesterone may provide cerebroprotection.
Collapse
Affiliation(s)
- Rachida Guennoun
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France.
| | - Xiaoyan Zhu
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Magalie Fréchou
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France
| | - Pauline Gaignard
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Abdelhamid Slama
- Biochemistry Laboratory, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Philippe Liere
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France
| | - Michael Schumacher
- U1195 Inserm and University Paris-Sud and University Paris-Saclay, 80 rue du Général Leclerc, 94276, Le Kremlin-Bicêtre, France
| |
Collapse
|
8
|
Shih CH, Chen JK, Kuo LW, Cho KH, Hsiao TC, Lin ZW, Lin YS, Kang JH, Lo YC, Chuang KJ, Cheng TJ, Chuang HC. Chronic pulmonary exposure to traffic-related fine particulate matter causes brain impairment in adult rats. Part Fibre Toxicol 2018; 15:44. [PMID: 30413208 PMCID: PMC6234801 DOI: 10.1186/s12989-018-0281-1 10.1186/s12989-018-0281-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Effects of air pollution on neurotoxicity and behavioral alterations have been reported. The objective of this study was to investigate the pathophysiology caused by particulate matter (PM) in the brain. We examined the effects of traffic-related particulate matter with an aerodynamic diameter of < 1 μm (PM1), high-efficiency particulate air (HEPA)-filtered air, and clean air on the brain structure, behavioral changes, brainwaves, and bioreactivity of the brain (cortex, cerebellum, and hippocampus), olfactory bulb, and serum after 3 and 6 months of whole-body exposure in 6-month-old Sprague Dawley rats. RESULTS The rats were exposed to 16.3 ± 8.2 (4.7~ 68.8) μg/m3 of PM1 during the study period. An MRI analysis showed that whole-brain and hippocampal volumes increased with 3 and 6 months of PM1 exposure. A short-term memory deficiency occurred with 3 months of exposure to PM1 as determined by a novel object recognition (NOR) task, but there were no significant changes in motor functions. There were no changes in frequency bands or multiscale entropy of brainwaves. Exposure to 3 months of PM1 increased 8-isoporstance in the cortex, cerebellum, and hippocampus as well as hippocampal inflammation (interleukin (IL)-6), but not in the olfactory bulb. Systemic CCL11 (at 3 and 6 months) and IL-4 (at 6 months) increased after PM1 exposure. Light chain 3 (LC3) expression increased in the hippocampus after 6 months of exposure. Spongiosis and neuronal shrinkage were observed in the cortex, cerebellum, and hippocampus (neuronal shrinkage) after exposure to air pollution. Additionally, microabscesses were observed in the cortex after 6 months of PM1 exposure. CONCLUSIONS Our study first observed cerebral edema and brain impairment in adult rats after chronic exposure to traffic-related air pollution.
Collapse
Affiliation(s)
- Chi-Hsiang Shih
- 0000 0000 9337 0481grid.412896.0School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jen-Kun Chen
- 0000000406229172grid.59784.37Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- 0000000406229172grid.59784.37Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Kuan-Hung Cho
- 0000000406229172grid.59784.37Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ta-Chih Hsiao
- 0000 0004 0546 0241grid.19188.39Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Zhe-Wei Lin
- 0000 0000 9337 0481grid.412896.0School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Syuan Lin
- 0000 0000 9337 0481grid.412896.0School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiunn-Horng Kang
- 0000 0004 0639 0994grid.412897.1Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei, Taiwan ,0000 0000 9337 0481grid.412896.0Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chun Lo
- 0000 0000 9337 0481grid.412896.0The Ph.D Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- 0000 0000 9337 0481grid.412896.0School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan ,0000 0000 9337 0481grid.412896.0Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsun-Jen Cheng
- 0000 0004 0546 0241grid.19188.39Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- 0000 0000 9337 0481grid.412896.0School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan ,0000 0000 9337 0481grid.412896.0School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan ,0000 0000 9337 0481grid.412896.0Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
9
|
Shih CH, Chen JK, Kuo LW, Cho KH, Hsiao TC, Lin ZW, Lin YS, Kang JH, Lo YC, Chuang KJ, Cheng TJ, Chuang HC. Chronic pulmonary exposure to traffic-related fine particulate matter causes brain impairment in adult rats. Part Fibre Toxicol 2018; 15:44. [PMID: 30413208 PMCID: PMC6234801 DOI: 10.1186/s12989-018-0281-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/24/2018] [Indexed: 11/30/2022] Open
Abstract
Background Effects of air pollution on neurotoxicity and behavioral alterations have been reported. The objective of this study was to investigate the pathophysiology caused by particulate matter (PM) in the brain. We examined the effects of traffic-related particulate matter with an aerodynamic diameter of < 1 μm (PM1), high-efficiency particulate air (HEPA)-filtered air, and clean air on the brain structure, behavioral changes, brainwaves, and bioreactivity of the brain (cortex, cerebellum, and hippocampus), olfactory bulb, and serum after 3 and 6 months of whole-body exposure in 6-month-old Sprague Dawley rats. Results The rats were exposed to 16.3 ± 8.2 (4.7~ 68.8) μg/m3 of PM1 during the study period. An MRI analysis showed that whole-brain and hippocampal volumes increased with 3 and 6 months of PM1 exposure. A short-term memory deficiency occurred with 3 months of exposure to PM1 as determined by a novel object recognition (NOR) task, but there were no significant changes in motor functions. There were no changes in frequency bands or multiscale entropy of brainwaves. Exposure to 3 months of PM1 increased 8-isoporstance in the cortex, cerebellum, and hippocampus as well as hippocampal inflammation (interleukin (IL)-6), but not in the olfactory bulb. Systemic CCL11 (at 3 and 6 months) and IL-4 (at 6 months) increased after PM1 exposure. Light chain 3 (LC3) expression increased in the hippocampus after 6 months of exposure. Spongiosis and neuronal shrinkage were observed in the cortex, cerebellum, and hippocampus (neuronal shrinkage) after exposure to air pollution. Additionally, microabscesses were observed in the cortex after 6 months of PM1 exposure. Conclusions Our study first observed cerebral edema and brain impairment in adult rats after chronic exposure to traffic-related air pollution. Electronic supplementary material The online version of this article (10.1186/s12989-018-0281-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chi-Hsiang Shih
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Kuan-Hung Cho
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ta-Chih Hsiao
- Graduate Institute of Environmental Engineering, National Taiwan University, Taipei, Taiwan
| | - Zhe-Wei Lin
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Syuan Lin
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jiunn-Horng Kang
- Department of Physical Medicine and Rehabilitation, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Physical Medicine and Rehabilitation, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Chun Lo
- The Ph.D Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Kai-Jen Chuang
- School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan.,Department of Public Health, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsun-Jen Cheng
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,School of Public Health, College of Public Health, Taipei Medical University, Taipei, Taiwan. .,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
10
|
Gong W, Qie S, Huang P, Xi J. Deletion of long noncoding RNA EFNA3 aggravates hypoxia-induced injury in PC-12 cells by upregulation of miR-101a. J Cell Biochem 2018; 120:836-847. [PMID: 30125989 DOI: 10.1002/jcb.27444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/16/2018] [Indexed: 11/07/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been reported to be involved in several neurological pathogenesis conditions including cerebral ischemia. In the current study, the functions of lncRNA EFNA3 on hypoxia-injured rat adrenal pheochromocytoma (PC-12) cells and the underlying molecular mechanism were studied. The expression of lncRNA EFNA3 was silenced by short hairpin RNA transfection, after which the cells were subjected with hypoxia. Cell viability, migration, invasion, and apoptosis were, respectively, determined by trypan blue staining, Transwell assay, annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double-staining, and Western blot analysis. The cross regulation between lncRNA EFNA3 and miR-101a, as well as between miR-101a and Rho associated coiled-coil containing protein kinase 2 (ROCK2) were detected by performing quantitative real-time polymerase chain reaction, RNA pull-down, RNA immunoprecipitation, luciferase activity assay, and Western blot analysis. Studies showed that lncRNA EFNA3 was highly expressed in response to hypoxia. Deletion of lncRNA EFNA3 significantly aggravated hypoxia-induced injury in PC-12 cells, as the impairment of cell viability, migration, and invasion, and the inducement of apoptosis. LncRNA EFNA3 worked as a sponging molecule for miR-101a and miR-101a suppression-protected PC-12 cells against hypoxia-induced injury even when lncRNA EFNA3 was silenced. ROCK2 was a target gene of miR-101a. ROCK2 overexpression exhibited neuroprotective activities. Besides, ROCK2 overexpression activated Wnt/β-catenin pathway whereas it deactivated JAK/STAT pathway upon hypoxia. Our study suggests that deletion of lncRNA EFNA3 aggravates hypoxia-induced injury in PC-12 cells by upregulating miR-101a, which further targets ROCK2.
Collapse
Affiliation(s)
- Weijun Gong
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Shuyan Qie
- Department of Rehabilitation Therapy, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Peiling Huang
- Beijing Rehabilitation Medicine Academy, Capital Medical University, Beijing, China
| | - Jianing Xi
- Department of Neurological Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Human Neural Stem Cell Extracellular Vesicles Improve Tissue and Functional Recovery in the Murine Thromboembolic Stroke Model. Transl Stroke Res 2017; 9:530-539. [PMID: 29285679 PMCID: PMC6132936 DOI: 10.1007/s12975-017-0599-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 02/08/2023]
Abstract
Over 700 drugs have failed in stroke clinical trials, an unprecedented rate thought to be attributed in part to limited and isolated testing often solely in “young” rodent models and focusing on a single secondary injury mechanism. Here, extracellular vesicles (EVs), nanometer-sized cell signaling particles, were tested in a mouse thromboembolic (TE) stroke model. Neural stem cell (NSC) and mesenchymal stem cell (MSC) EVs derived from the same pluripotent stem cell (PSC) line were evaluated for changes in infarct volume as well as sensorimotor function. NSC EVs improved cellular, tissue, and functional outcomes in middle-aged rodents, whereas MSC EVs were less effective. Acute differences in lesion volume following NSC EV treatment were corroborated by MRI in 18-month-old aged rodents. NSC EV treatment has a positive effect on motor function in the aged rodent as indicated by beam walk, instances of foot faults, and strength evaluated by hanging wire test. Increased time with a novel object also indicated that NSC EVs improved episodic memory formation in the rodent. The therapeutic effect of NSC EVs appears to be mediated by altering the systemic immune response. These data strongly support further preclinical development of a NSC EV-based stroke therapy and warrant their testing in combination with FDA-approved stroke therapies.
Collapse
|
12
|
Hobson BA, Rowland DJ, Supasai S, Harvey DJ, Lein PJ, Garbow JR. A magnetic resonance imaging study of early brain injury in a rat model of acute DFP intoxication. Neurotoxicology 2017; 66:170-178. [PMID: 29183789 DOI: 10.1016/j.neuro.2017.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 11/18/2022]
Abstract
Current treatments for seizures induced by organophosphates do not protect sufficiently against progressive neurodegeneration or delayed cognitive impairment. Developing more effective therapeutic approaches has been challenging because the pathogenesis of these delayed consequences is poorly defined. Using magnetic resonance imaging (MRI), we previously reported brain lesions that persist for months in a rat model of acute intoxication with the OP, diisopropylfluorophosphate (DFP). However, the early spatiotemporal progression of these lesions remains unknown. To address this data gap, we used in vivo MRI to longitudinally monitor brain lesions during the first 3 d following acute DFP intoxication. Adult male Sprague Dawley rats acutely intoxicated with DFP (4mg/kg, sc) were MR imaged at 6, 12, 18, 24, 48, 72h post-DFP, and their brains then taken for correlative histology to assess neurodegeneration using FluoroJade C (FJC) staining. Acute DFP intoxication elicited moderate-to-severe seizure activity. T2-weighted (T2w) anatomic imaging revealed prominent lesions within the thalamus, piriform cortex, cerebral cortex, hippocampus, corpus striatum, and substantia nigra that corresponded to neurodegeneration, evident as bands of FJC positive cells. Semi-quantitative assessment of lesion severity demonstrated significant regional variation in the onset and progression of injury, and suggested that lesion severity may be modulated by isoflurane anesthesia. These results imply that the timing of therapeutic intervention for attenuating brain injury following OP intoxication may be regionally dependent, and that longitudinal assessment of OP-induced damage by MRI may be a powerful tool for assessing therapeutic response.
Collapse
Affiliation(s)
- Brad A Hobson
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Douglas J Rowland
- Center for Molecular and Genomic Imaging, University of California, Davis, College of Engineering, Davis, CA, 95616, United States.
| | - Suangsuda Supasai
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Danielle J Harvey
- Department of Public Health Sciences, University of California, Davis, School of Medicine, Davis, CA, 95616, United States.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, School of Veterinary Medicine, Davis, CA, 95616, United States.
| | - Joel R Garbow
- Biomedical Magnetic Resonance Laboratory, Mallinckrodt Institute of Radiology, School of Medicine,Washington University in St. Louis, St. Louis, MO, 63110, United States.
| |
Collapse
|
13
|
Kluge MG, Kracht L, Abdolhoseini M, Ong LK, Johnson SJ, Nilsson M, Walker FR. Impaired microglia process dynamics post-stroke are specific to sites of secondary neurodegeneration. Glia 2017; 65:1885-1899. [PMID: 28836304 DOI: 10.1002/glia.23201] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/06/2017] [Accepted: 07/23/2017] [Indexed: 01/28/2023]
Abstract
Stroke induces tissue death both at the site of infarction and at secondary sites connected to the primary infarction. This latter process has been referred to as secondary neurodegeneration (SND). Using predominantly fixed tissue analyses, microglia have been implicated in regulating the initial response at both damage sites post-stroke. In this study, we used acute slice based multiphoton imaging, to investigate microglia dynamic process movement in mice 14 days after a photothrombotic stroke. We evaluated the baseline motility and process responses to locally induced laser damage in both the peri-infarct (PI) territory and the ipsilateral thalamus, a major site of post-stroke SND. Our findings show that microglia process extension toward laser damage within the thalamus is lost, yet remains robustly intact within the PI territory. However, microglia at both sites displayed an activated morphology and elevated levels of commonly used activation markers (CD68, CD11b), indicating that the standardly used fixed tissue metrics of microglial "activity" are not necessarily predictive of microglia function. Analysis of the purinergic P2 Y12 receptor, a key regulator of microglia process extension, revealed an increased somal localization on nonresponsive microglia in the thalamus. To our knowledge, this is the first study to identify a non-responsive microglia phenotype specific to areas of SND post-stroke, which cannot be identified by the classical assessment of microglia activation but rather the localization of P2 Y12 to the soma.
Collapse
Affiliation(s)
- Murielle G Kluge
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Laura Kracht
- Department of Neuroscience, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - Mahmoud Abdolhoseini
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, New South Wales, Australia
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, Victoria, Australia
| | - Sarah J Johnson
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, New South Wales, Australia
| | - Michael Nilsson
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, Victoria, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, New South Wales, Australia.,Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, Victoria, Australia
| |
Collapse
|
14
|
Andrabi SS, Parvez S, Tabassum H. Progesterone induces neuroprotection following reperfusion-promoted mitochondrial dysfunction after focal cerebral ischemia in rats. Dis Model Mech 2017; 10:787-796. [PMID: 28363987 PMCID: PMC5482998 DOI: 10.1242/dmm.025692] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 03/28/2017] [Indexed: 12/31/2022] Open
Abstract
Organelle damage and increases in mitochondrial permeabilization are key events in the development of cerebral ischemic tissue injury because they cause both modifications in ATP turnover and cellular apoptosis/necrosis. Early restoration of blood flow and improvement of mitochondrial function might reverse the situation and help in recovery following an onset of stroke. Mitochondria and related bioenergetic processes can be effectively used as pharmacological targets. Progesterone (P4), one of the promising neurosteroids, has been found to be neuroprotective in various models of neurological diseases, through a number of mechanisms. This influenced us to investigate the possible role of P4 in the mitochondria-mediated neuroprotective mechanism in an ischemic stroke model of rat. In this study, we have shown the positive effect of P4 administration on behavioral deficits and mitochondrial health in an ischemic stroke injury model of transient middle cerebral artery occlusion (tMCAO). After induction of tMCAO, the rats received an initial intraperitoneal injection of P4 (8 mg/kg body weight) or vehicle at 1 h post-occlusion followed by subcutaneous injections at 6, 12 and 18 h. Behavioral assessment for functional deficits included grip strength, motor coordination and gait analysis. Findings revealed a significant improvement with P4 treatment in tMCAO animals. Staining of isolated brain slices from P4-treated rats with 2,3,5-triphenyltetrazolium chloride (TTC) showed a reduction in the infarct area in comparison to the vehicle group, indicating the presence of an increased number of viable mitochondria. P4 treatment was also able to attenuate mitochondrial reactive oxygen species (ROS) production, as well as block the mitochondrial permeability transition pore (mPTP), in the tMCAO injury model. In addition, it was also able to ameliorate the altered mitochondrial membrane potential and respiration ratio in the ischemic animals, thereby suggesting that P4 has a positive effect on mitochondrial bioenergetics. In conclusion, these results demonstrate that P4 treatment is beneficial in preserving the mitochondrial functions that are altered in cerebral ischemic injury and thus can help in defining better therapies. Summary: Progesterone treatment is beneficial in preserving the altered mitochondrial functions in cerebral ischemic injury and thus can help in defining better therapies.
Collapse
Affiliation(s)
- Syed Suhail Andrabi
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| | - Heena Tabassum
- Department of Biochemistry, Jamia Hamdard (Hamdard University), New Delhi 110062, India
| |
Collapse
|
15
|
Tamargo IA, Bader M, Li Y, Yu SJ, Wang Y, Talbot K, DiMarchi RD, Pick CG, Greig NH. Novel GLP-1R/GIPR co-agonist "twincretin" is neuroprotective in cell and rodent models of mild traumatic brain injury. Exp Neurol 2017; 288:176-186. [PMID: 27845037 PMCID: PMC5878017 DOI: 10.1016/j.expneurol.2016.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 12/12/2022]
Abstract
Several single incretin receptor agonists that are approved for the treatment of type 2 diabetes mellitus (T2DM) have been shown to be neuroprotective in cell and animal models of neurodegeneration. Recently, a synthetic dual incretin receptor agonist, nicknamed "twincretin," was shown to improve upon the metabolic benefits of single receptor agonists in mouse and monkey models of T2DM. In the current study, the neuroprotective effects of twincretin are probed in cell and mouse models of mild traumatic brain injury (mTBI), a prevalent cause of neurodegeneration in toddlers, teenagers and the elderly. Twincretin is herein shown to have activity at two different receptors, dose-dependently increase levels of intermediates in the neurotrophic CREB pathway and enhance viability of human neuroblastoma cells exposed to toxic concentrations of glutamate and hydrogen peroxide, insults mimicking the inflammatory conditions in the brain post-mTBI. Additionally, twincretin is shown to improve upon the neurotrophic effects of single incretin receptor agonists in these same cells. Finally, a clinically translatable dose of twincretin, when administered post-mTBI, is shown to fully restore the visual and spatial memory deficits induced by mTBI, as evaluated in a mouse model of weight drop close head injury. These results establish twincretin as a novel neuroprotective agent and suggest that it may improve upon the effects of the single incretin receptor agonists via dual agonism.
Collapse
MESH Headings
- Animals
- Body Temperature/drug effects
- Brain Injuries, Traumatic/complications
- Brain Injuries, Traumatic/drug therapy
- CREB-Binding Protein/metabolism
- Cell Line, Tumor
- Cells, Cultured
- Disease Models, Animal
- Embryo, Mammalian
- Glucagon-Like Peptide 1/metabolism
- Glucagon-Like Peptide-1 Receptor/agonists
- Glucagon-Like Peptide-1 Receptor/metabolism
- Humans
- Incretins/therapeutic use
- Male
- Maze Learning/drug effects
- Memory Disorders/etiology
- Memory Disorders/prevention & control
- Mice
- Mice, Inbred ICR
- Neuroblastoma/pathology
- Neuroprotective Agents/therapeutic use
- Rats
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Receptors, Gastrointestinal Hormone/agonists
- Receptors, Gastrointestinal Hormone/metabolism
- Recognition, Psychology/drug effects
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Ian A Tamargo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Miaad Bader
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | | | | | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
16
|
Chiu CC, Liao YE, Yang LY, Wang JY, Tweedie D, Karnati HK, Greig NH, Wang JY. Neuroinflammation in animal models of traumatic brain injury. J Neurosci Methods 2016; 272:38-49. [PMID: 27382003 PMCID: PMC5201203 DOI: 10.1016/j.jneumeth.2016.06.018] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and morbidity worldwide. Neuroinflammation is prominent in the short and long-term consequences of neuronal injuries that occur after TBI. Neuroinflammation involves the activation of glia, including microglia and astrocytes, to release inflammatory mediators within the brain, and the subsequent recruitment of peripheral immune cells. Various animal models of TBI have been developed that have proved valuable to elucidate the pathophysiology of the disorder and to assess the safety and efficacy of novel therapies prior to clinical trials. These models provide an excellent platform to delineate key injury mechanisms that associate with types of injury (concussion, contusion, and penetration injuries) that occur clinically for the investigation of mild, moderate, and severe forms of TBI. Additionally, TBI modeling in genetically engineered mice, in particular, has aided the identification of key molecules and pathways for putative injury mechanisms, as targets for development of novel therapies for human TBI. This Review details the evidence showing that neuroinflammation, characterized by the activation of microglia and astrocytes and elevated production of inflammatory mediators, is a critical process occurring in various TBI animal models, provides a broad overview of commonly used animal models of TBI, and overviews representative techniques to quantify markers of the brain inflammatory process. A better understanding of neuroinflammation could open therapeutic avenues for abrogation of secondary cell death and behavioral symptoms that may mediate the progression of TBI.
Collapse
Affiliation(s)
- Chong-Chi Chiu
- Department of General Surgery, Chi Mei Medical Center, Tainan and Liouying, Taiwan
| | - Yi-En Liao
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Ling-Yu Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jing-Ya Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hanuma K Karnati
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
17
|
Pessah IN, Rogawski MA, Tancredi DJ, Wulff H, Zolkowska D, Bruun DA, Hammock BD, Lein PJ. Models to identify treatments for the acute and persistent effects of seizure-inducing chemical threat agents. Ann N Y Acad Sci 2016; 1378:124-136. [PMID: 27467073 DOI: 10.1111/nyas.13137] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/15/2016] [Accepted: 05/19/2016] [Indexed: 12/27/2022]
Abstract
Exposures to seizure-inducing chemical threat agents are a major public health concern. Of particular need is improved treatment to terminate convulsions and to prevent the long-term neurological sequelae in survivors. We are studying the organophosphorus cholinesterase inhibitor diisopropyl fluorophosphate (DFP) and the GABA receptor inhibitor tetramethylenedisulfotetramine (TETS), which arguably encompass the mechanistic spectrum of seizure-inducing chemical threats, with the goal of identifying therapeutic approaches with broad-spectrum efficacy. Research efforts have focused on developing translational models and translational diagnostic approaches, including (1) in vivo models of DFP- and TETS-induced seizures for studying neuropathological mechanisms and identifying treatment approaches; (2) in vivo imaging modalities for noninvasive longitudinal monitoring of neurological damage and response to therapeutic candidates; and (3) higher-throughput in vitro platforms for rapid screening of compounds to identify potential antiseizure and neuroprotective agents, as well as mechanistically relevant novel drug targets. This review summarizes our progress toward realizing these goals and discusses best practices and mechanistic insights derived from our modeling efforts.
Collapse
Affiliation(s)
- Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine
| | | | | | - Heike Wulff
- Department of Pharmacology, School of Medicine
| | | | - Donald A Bruun
- Department of Molecular Biosciences, School of Veterinary Medicine
| | - Bruce D Hammock
- Department of Entomology, College of Agricultural and Environmental Sciences, University of California, Davis, Davis, California
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine.
| |
Collapse
|
18
|
Li Y, Bader M, Tamargo I, Rubovitch V, Tweedie D, Pick CG, Greig NH. Liraglutide is neurotrophic and neuroprotective in neuronal cultures and mitigates mild traumatic brain injury in mice. J Neurochem 2015; 135:1203-1217. [PMID: 25982185 DOI: 10.1111/jnc.13169] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 04/29/2015] [Accepted: 05/07/2015] [Indexed: 01/21/2023]
Abstract
Traumatic brain injury (TBI), a brain dysfunction for which there is no present effective treatment, is often caused by a concussive impact to the head and affects an estimated 1.7 million Americans annually. Our laboratory previously demonstrated that exendin-4, a long-lasting glucagon-like peptide 1 receptor (GLP-1R) agonist, has neuroprotective effects in cellular and animal models of TBI. Here, we demonstrate neurotrophic and neuroprotective effects of a different GLP-1R agonist, liraglutide, in neuronal cultures and a mouse model of mild TBI (mTBI). Liraglutide promoted dose-dependent proliferation in SH-SY5Y cells and in a GLP-1R over-expressing cell line at reduced concentrations. Pre-treatment with liraglutide rescued neuronal cells from oxidative stress- and glutamate excitotoxicity-induced cell death. Liraglutide produced neurotrophic and neuroprotective effects similar to those of exendin-4 in vitro. The cAMP/PKA/pCREB pathway appears to play an important role in this neuroprotective activity of liraglutide. Furthermore, our findings in cell culture were well-translated in a weight drop mTBI mouse model. Post-treatment with a clinically relevant dose of liraglutide for 7 days in mice ameliorated memory impairments caused by mTBI when evaluated 7 and 30 days post trauma. These data cross-validate former studies of exendin-4 and suggest that liraglutide holds therapeutic potential for the treatment of mTBI. Exendin-4, a long-lasting glucagon-like peptide 1 receptor (GLP-1R) agonist, has neuroprotective effects in cellular and animal models of traumatic brain injury (TBI). Here, we demonstrate neurotrophic and neuroprotective effects of a different GLP-1R agonist, liraglutide, in neuronal cultures and a mouse model of mild TBI (mTBI). Liraglutide promoted dose-dependent proliferation in SH-SY5Y cells and in a GLP-1R over-expressing cell line at reduced concentrations. Pretreatment with liraglutide rescued neuronal cells from oxidative stress- and glutamate excitotoxicity-induced cell death. Liraglutide produced neurotrophic and neuroprotective effects similar to those of exendin-4 in vitro, likely involving the cAMP/PKA/pCREB pathway. Our findings in cell culture were well-translated in a weight-drop mTBI mouse model. Post-treatment with a clinically relevant dose of liraglutide for 7 days in mice ameliorated memory impairments caused by mTBI.
Collapse
Affiliation(s)
- Yazhou Li
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Miaad Bader
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - Ian Tamargo
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - David Tweedie
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Chaim G Pick
- Department of Anatomy and Anthropology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978 Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, 69978 Israel
| | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
19
|
Panagiotou S, Saha S. Therapeutic benefits of nanoparticles in stroke. Front Neurosci 2015; 9:182. [PMID: 26041986 PMCID: PMC4436818 DOI: 10.3389/fnins.2015.00182] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/08/2015] [Indexed: 12/30/2022] Open
Abstract
Stroke represents one of the major causes of death and disability worldwide, for which no effective treatments are available. The thrombolytic drug alteplase (tissue plasminogen activator or tPA) is the only treatment for acute ischemic stroke but its use is limited by several factors including short therapeutic window, selective efficacy, and subsequent haemorrhagic complications. Numerous preclinical studies have reported very promising results using neuroprotective agents but they have failed at clinical trials because of either safety issues or lack of efficacy. The delivery of many potentially therapeutic neuroprotectants and diagnostic compounds to the brain is restricted by the blood-brain barrier (BBB). Nanoparticles (NPs), which can readily cross the BBB without compromising its integrity, have immense applications in the treatment of ischemic stroke. In this review, potential uses of NPs will be summarized for the treatment of ischemic stroke. Additionally, an overview of targeted NPs will be provided, which could be used in the diagnosis of stroke. Finally, the potential limitations of using NPs in medical applications will be mentioned. Since the use of NPs in stroke therapy is now emerging and is still in development, this review is far from comprehensive or conclusive. Instead, examples of NPs and their current use will be provided, as well as the potentials of NPs in an effort to meet the high demand of new therapies in stroke.
Collapse
Affiliation(s)
- Stavros Panagiotou
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds Leeds, UK
| | - Sikha Saha
- Division of Cardiovascular and Diabetes Research, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds Leeds, UK
| |
Collapse
|