1
|
Sampath D, Zardeneta ME, Akbari Z, Singer J, Gopalakrishnan B, Hurst DA, Villarreal M, McDaniel EA, Noarbe BP, Obenaus A, Sohrabji F. Loss of white matter tracts and persistent microglial activation in the chronic phase of ischemic stroke in female rats and the effect of miR-20a-3p treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.01.636074. [PMID: 39975179 PMCID: PMC11838816 DOI: 10.1101/2025.02.01.636074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Our previous studies showed that intravenous injections of the small non-coding RNA mir-20a-3p is neuroprotective for stroke in the acute phase and attenuates long-term cognitive impairment in middle-aged female rats. In this study, we evaluated postmortem brain pathology at 100+d after stroke in a set of behaviorally characterized animals. This included Sham (no stroke) controls or stroke animals that received either mir20a-3p at 4h, 24h and 70d iv post stroke (MCAo+mir20a-3p) or a scrambled oligo (MCAo+Scr). Brain volumetric features were analyzed with T2 weighted and Diffusion Tensor magnetic resonance imaging (MRI) followed by histological analysis. Principal component analysis of Fractional Anisotropy (FA)-diffusion tensor MRI measures showed that MCAo+Scr and MCAo+mir20a-3p groups differed significantly in the volume of white matter but not gray matter. Weil myelin-stained sections confirmed decreased volume of the corpus callosum, internal capsule and the anterior commissure in the ischemic hemisphere of MCAo+Scr animals compared to the non-ischemic hemisphere, while sham and MCAo+Mir-20a-3p showed no hemispheric asymmetries. The MCAo+Scr group also exhibited asymmetry in hemisphere and lateral ventricle volumes, with ventricular enlargement in the ischemic hemisphere as compared to the non-ischemic hemisphere. The numbers of microglia were significantly elevated in white matter tracts in the MCAo+Scr group, with a trend towards increased myelin phagocytic microglia in these tracts. Regression analysis indicated that performance on an episodic memory test (novel object recognition test; NORT) was associated with decreased white matter volume and increased microglial numbers. These data support the hypothesis that stroke-induced cognitive impairment is accompanied by white matter attrition and persistent microglial activation and is consistent with reports that cognitive deterioration resulting from vascular diseases, such as stroke, is associated with secondary neurodegeneration in regions distal from the initial infarction.
Collapse
|
2
|
El-Hakim Y, Mani KK, Pickle KA, Akbari Z, Samiya N, Pham C, Salas G, Pilla R, Sohrabji F. Peripheral, but not central, IGF-1 treatment attenuates stroke-induced cognitive impairment in middle-aged female Sprague Dawley rats: The gut as a therapeutic target. Brain Behav Immun 2024; 122:150-166. [PMID: 39142422 PMCID: PMC11972691 DOI: 10.1016/j.bbi.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 08/16/2024] Open
Abstract
Stroke results in immediate sensory or motor disability and increases the risk for long term cognitive-affective impairments. Thus, therapies are urgently needed to improve quality of life for stroke survivors, especially women who are at a greater risk for severe stroke after menopause. Most current research on stroke therapies target the central nervous system; however, stroke also impacts peripheral organ systems. Our studies using acyclic (estrogen-deficient) middle aged female Sprague Dawley rats show that this group not only displays worse outcomes after stroke as compared to adult females, but also has lower levels of the neuroprotective peptide Insulin-like Growth Factor (IGF1) in circulation. Intracerebroventricular (ICV) administration of IGF1 to this group decreases infarct volume and improves sensory motor performance in the acute phase. In this study, we show that, despite this neuroprotection, ICV-IGF1 did not reduce peripheral inflammation or improve post stroke cognitive impairment in the chronic phase. In view of the evidence that stroke induces rapid gut dysfunction, we tested whether systemic delivery of IGF1 (intraperitoneal, IP) would promote gut health and consequently improve long-term behavioral outcomes. Surprisingly, while IP-IGF1, delivered 4 h and 24 h after ischemic stroke, did not reduce infarct volume or acute sensory motor impairment, it significantly attenuated circulating levels of pro-inflammatory cytokines, and attenuated stroke-induced cognitive impairment. In addition, IP-IGF1 treatment reduced gut dysmorphology and gut dysbiosis. Our data support the conclusion that therapeutics targeting peripheral targets are critical for long-term stroke recovery, and that gut repair is a novel therapeutic target to improve brain health in aging females.
Collapse
Affiliation(s)
- Yumna El-Hakim
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Kathiresh Kumar Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Kaylin A Pickle
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Zara Akbari
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Nadia Samiya
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Chloe Pham
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Gianna Salas
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA
| | - Rachel Pilla
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine Texas A&M University, College Station, TX Brazos
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University-Health Science Center, Bryan TX-77807 USA.
| |
Collapse
|
3
|
De Assis GG, Murawska-Ciałowicz E. BDNF Modulation by microRNAs: An Update on the Experimental Evidence. Cells 2024; 13:880. [PMID: 38786102 PMCID: PMC11119608 DOI: 10.3390/cells13100880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/25/2024] Open
Abstract
MicroRNAs can interfere with protein function by suppressing their messenger RNA translation or the synthesis of its related factors. The function of brain-derived neurotrophic factor (BDNF) is essential to the proper formation and function of the nervous system and is seen to be regulated by many microRNAs. However, understanding how microRNAs influence BDNF actions within cells requires a wider comprehension of their integrative regulatory mechanisms. Aim: In this literature review, we have synthesized the evidence of microRNA regulation on BDNF in cells and tissues, and provided an analytical discussion about direct and indirect mechanisms that appeared to be involved in BDNF regulation by microRNAs. Methods: Searches were conducted on PubMed.gov using the terms "BDNF" AND "MicroRNA" and "brain-derived neurotrophic factor" AND "MicroRNA", updated on 1 September 2023. Papers without open access were requested from the authors. One hundred and seventy-one papers were included for review and discussion. Results and Discussion: The local regulation of BDNF by microRNAs involves a complex interaction between a series of microRNAs with target proteins that can either inhibit or enhance BDNF expression, at the core of cell metabolism. Therefore, understanding this homeostatic balance provides resources for the future development of vector-delivery-based therapies for the neuroprotective effects of BDNF.
Collapse
Affiliation(s)
- Gilmara Gomes De Assis
- Department of Restorative Dentistry, Araraquara School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-385, SP, Brazil
| | - Eugenia Murawska-Ciałowicz
- Department of Physiology and Biochemistry, Wroclaw University of Health and Sport Sciences, 51-612 Wrocław, Poland;
| |
Collapse
|
4
|
Zhu F, Ding S, Liu Y, Wang X, Wu Z. Ozone-mediated cerebral protection: Unraveling the mechanism through ferroptosis and the NRF2/SLC7A11/GPX4 signaling pathway. J Chem Neuroanat 2024; 136:102387. [PMID: 38182039 DOI: 10.1016/j.jchemneu.2023.102387] [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: 10/20/2023] [Revised: 12/16/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND The pathogenesis of brain ischemic/reperfusion (I/R) insult is characterized by neuronal loss due to excessive oxidative stress responses. Ferroptosis, a form of oxidative cell death, can be triggered when the balance between antioxidants and pro-oxidants in cells is disrupted. Ozone, a natural bioactive molecule with antioxidant/anti-apoptotic and pro-autophagic properties, has been shown to enhance the antioxidant system's capacity and ameliorate oxidative stress. However, its role in neuronal ferroptosis remains unclear. Therefore, we investigated the functions and possible mechanisms of ozone in cerebral I/R-induced ferroptotic neuronal death. METHODS A cerebral ischemia-reperfusion injury model was induced in Sprague-Dawley (SD) rats pre-treated with ozone. Intraperitoneal administration of the NRF2 inhibitor ML385, the SLC7A11 inhibitor Erastin, and the GPX4 inhibitor RSL3 was performed one hour prior to model establishment. RESULTS Our results showed that ozone preconditioning mitigated neuronal damage caused by cerebral I/R, reduced the severity of neurological deficits, lowered cerebral infarct volume in middle cerebral artery occlusion (MCAO) rats, and decreased the volume of cerebral infarcts. Transmission electron microscopy, immunofluorescence, and Western blotting indicated ferroptosis following MCAO-induced brain damage. MCAO resulted in morphological damage to neuronal mitochondria, increased lipid peroxidation accumulation, and elevated malondialdehyde (MDA) production. Furthermore, MCAO decreased levels of FTH1 and GPX4 (negative regulators of ferroptosis) and increased ACSL4 levels (a positive regulator of ferroptosis). Ozone preconditioning demonstrated a neuroprotective effect by increasing NRF2 nuclear translocation and the expression of SLC7A11 and GPX4. Treatment with ML385, Erastin, and RSL3 significantly reversed ozone preconditioning's protective effect on neuronal ferroptosis. CONCLUSION Our findings demonstrated that ozone treatment attenuates ferroptosis in a cerebral ischemia/reperfusion injury rat model via the NRF2/SLC7A11/GPX4 pathway, providing a theoretical basis for ozone's potential use as a therapy to prevent ischemic stroke.
Collapse
Affiliation(s)
- Farong Zhu
- Department of Anesthesiology, Nanjing Medical University, Jiangning, Nanjing 211166, People's Republic of China
| | - Shengyang Ding
- Department of Anesthesiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 29 Xinglong Lane, Changzhou, Jiangsu 213000, People's Republic of China
| | - Yu Liu
- Department of Graduate School of Dalian Medical University, Dalian 116044, People's Republic of China
| | - Xinlei Wang
- Department of Graduate School of Dalian Medical University, Dalian 116044, People's Republic of China
| | - Zhouquan Wu
- Department of Anesthesiology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, 29 Xinglong Lane, Changzhou, Jiangsu 213000, People's Republic of China.
| |
Collapse
|
5
|
Qian L, Huang S, Liu X, Jiang Y, Jiang Y, Hu Y, Yang Z. Morroniside improves the symptoms of post-stroke depression in mice through the BDNF signaling pathway mediated by MiR-409-3p. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155224. [PMID: 38006805 DOI: 10.1016/j.phymed.2023.155224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/22/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Post-stroke depression (PSD) is a common psychiatric symptom after a stroke. Morroniside, an iridoid glycoside found in Cornus officinalis, has garnered significant attention for its potential to alleviate symptoms associated with depression. PURPOSE This study aims to highlight the potential use of morroniside in the treatment of PSD and elucidate the underlying molecular mechanisms. METHODS To establish a reliable PSD model, male C57BL/6 mice were subjected to brief MCAO in conjunction with CUMS. Post-morroniside administration, neuronal viability, and hippocampal cell apoptosis were evaluated by Nissl staining and TUNEL detection, respectively. Depression-like behaviors were evaluated using SPT, TST, and FST. The Longa score and cylinder test were used to evaluate the effect of morroniside on motor function. Furthermore, to investigate the underlying molecular mechanisms, bioinformatic analysis and the dual luciferase assay were performed to investigate the MiR-409-3p-BDNF interaction. In addition, subsequent to MiR-409-3p overexpression via AAV virus, we assessed mRNA expression and protein levels of key components within the BDNF/TrkB signaling pathway using RT-qPCR, immunohistochemistry, and western blot analysis. RESULTS The observed decrease in apoptosis and amelioration of depression-like behaviors strongly indicate the potential of morroniside as a therapeutic agent for PSD. Furthermore, the upregulation of key proteins within the BDNF/TrkB signaling pathway in the cortex suggests that morroniside activates this pathway. Through bioinformatics analysis, MiR-409-3p was identified and found to bind to the BDNF gene, resulting in the inhibition of BDNF expression. Importantly, we demonstrate that morroniside mitigates this inhibitory effect of MiR-409-3p on BDNF, thereby facilitating the activation of the BDNF/TrkB signaling pathway. CONCLUSION The findings suggest that morroniside demonstrates the ability to improve PSD symptoms through the BDNF/TrkB signaling pathway mediated by MiR-409-3p. These results emphasize the importance of the BDNF signaling pathway in improving PSD symptoms and provide a possible mechanism for morroniside to treat PSD.
Collapse
Affiliation(s)
- Lihui Qian
- Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, 222004, Lianyungang, Jiangsu, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, Jiangsu, China
| | - Sirui Huang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, Jiangsu, China
| | - Xiaoli Liu
- Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, 222004, Lianyungang, Jiangsu, China
| | - Yongxia Jiang
- Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, 222004, Lianyungang, Jiangsu, China
| | - Yongqu Jiang
- Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, 222004, Lianyungang, Jiangsu, China
| | - Yue Hu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, Jiangsu, China; Shen Chun-ti Nation-Famous Experts Studio for Traditional Chinese Medicine Inheritance, Changzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 213003, Changzhou, Jiangsu, China.
| | - Zhou Yang
- Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, 222004, Lianyungang, Jiangsu, China.
| |
Collapse
|
6
|
Zhang Y, Yang Y, Li H, Feng Q, Ge W, Xu X. Investigating the Potential Mechanisms and Therapeutic Targets of Inflammatory Cytokines in Post-stroke Depression. Mol Neurobiol 2024; 61:132-147. [PMID: 37592185 DOI: 10.1007/s12035-023-03563-w] [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: 05/05/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Post-stroke depression (PSD) affects approximately one-third of stroke survivors, severely impacting general recovery and quality of life. Despite extensive studies, the exact mechanisms underlying PSD remain elusive. However, emerging evidence implicates proinflammatory cytokines, including interleukin-1β, interleukin-6, tumor necrosis factor-alpha, and interleukin-18, play critical roles in PSD development. These cytokines contribute to PSD through various mechanisms, including hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neurotransmitter alterations, neurotrophic factor changes, gut microbiota imbalances, and genetic predispositions. This review is aimed at exploring the role of cytokines in stroke and PSD while identifying their potential as specific therapeutic targets for managing PSD. A more profound understanding of the mechanisms regulating inflammatory cytokine expression and anti-inflammatory cytokines like interleukin-10 in PSD may facilitate the development of innovative interventions to improve outcomes for stroke survivors experiencing depression.
Collapse
Affiliation(s)
- Yutong Zhang
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuehua Yang
- Department of Neurology, Suzhou Yongding Hospital, Suzhou, 215028, China
| | - Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Qian Feng
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wei Ge
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221600, China.
| | - Xingshun Xu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
7
|
Pinson MR, Bake S, Hurst DA, Samiya NT, Sohrabji F, Miranda RC. Prenatal alcohol alters inflammatory signatures in enteric portal tissues following adult-onset cerebrovascular ischemic stroke. iScience 2023; 26:107920. [PMID: 37810225 PMCID: PMC10550726 DOI: 10.1016/j.isci.2023.107920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Prenatal alcohol exposure (PAE) impairs recovery from cerebrovascular ischemic stroke in adult rodents. Since the gut becomes dysbiotic following stroke, we assessed links between PAE and enteric portal inflammation. Adult control and PAE rat offspring received a unilateral endothelin-1-induced occlusion of the middle cerebral artery. Post-stroke behavioral disabilities and brain cytokines were assessed. Mesenteric adipose and liver transcriptomes were assessed from stroke-exposed and stroke-naive offspring. We identified, in the liver of stroke-naive animals, a moderate correlation between PAE and a gene network for inflammatory necroptosis. PAE inhibited the acute-phase brain inflammatory cytokine response to stroke. Post-stroke neurological function was correlated with an adipose gene network associated with B-lymphocyte differentiation and nuclear factor κB (NF-κB) signaling and with a liver pro-inflammatory gene network. Collectively, PAE inhibits brain inflammation but results in an inflammatory signature in enteric portal tissues after stroke, suggesting that PAE persistently and adversely impacts the gut-brain axis following adult-onset disease.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - David A Hurst
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Nadia T Samiya
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| |
Collapse
|
8
|
Sampath D, Branyan TE, Markowsky KG, Gunda R, Samiya N, Obenaus A, Sohrabji F. Sex differences in cognitive impairment after focal ischemia in middle-aged rats and the effect of iv miR-20a-3p treatment. Neurobiol Aging 2023; 129:168-177. [PMID: 37336171 DOI: 10.1016/j.neurobiolaging.2023.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/02/2023] [Accepted: 05/02/2023] [Indexed: 06/21/2023]
Abstract
Stroke is a major cause of death and disability worldwide and is also a leading cause of vascular dementia and Alzheimer's disease, with older women experiencing accelerated decline. Our previous studies show that intravenous (iv) injections of miR-20a-3p, a small noncoding RNA (miRNA) delivered after stroke improves acute stroke outcomes in middle-aged male and female rats. The present study tested whether mir-20a-3p treatment would also ameliorate stroke-induced cognitive decline in the chronic phase. Acyclic middle-aged females and age-matched male Sprague Dawley rats were subjected to middle cerebral artery occlusion using endothelin-1 or sham surgery, and treated iv with miR-20a-3p mimics or scrambled oligos at 4 hours, 24 hours, and 70 days post-stroke. Stroke resulted in a significant sensory motor deficit, while miR-20a-3p treatment reduced these deficits in both sexes. Cognitive impairment was assessed periodically for 3 months after stroke using contextual fear conditioning and the novel object recognition task. Overall, the tests of associative and episodic memory were affected by focal ischemia only in female rats, and miR-20a-3p ameliorated the rate of decline.
Collapse
Affiliation(s)
- Dayalan Sampath
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Taylor E Branyan
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Kylee G Markowsky
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Rithvik Gunda
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Nadia Samiya
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA
| | - Andre Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine and Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX, USA.
| |
Collapse
|
9
|
Assessing Depression and Cognitive Impairment Following Stroke and Neurotrauma: Behavioral Methods for Quantifying Impairment and Functional Recovery. Methods Mol Biol 2023; 2616:263-277. [PMID: 36715941 DOI: 10.1007/978-1-0716-2926-0_20] [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: 01/31/2023]
Abstract
Rodent models of stroke and neural injury are reliable and useful tools for testing new interventions and therapeutics. In addition to physical (motor) impairment, cognitive deficits and depressive behaviors are often observed due to neurotrauma. Proper experimental design of pre- and post-assessments of these behaviors that reduce or minimize the confounding effects of motor impairment are essential for determining markers of progression of impairment or recovery. This chapter provides step-by-step laboratory protocols for assessing cognition using the Barnes maze and the novel object recognition test (NORT) and depressive-like behaviors using the sucrose preference test, the three-chambered sociability approach test, and the burrowing test.
Collapse
|
10
|
Mani KK, El-Hakim Y, Branyan TE, Samiya N, Pandey S, Grimaldo MT, Habbal A, Wertz A, Sohrabji F. Intestinal epithelial stem cell transplants as a novel therapy for cerebrovascular stroke. Brain Behav Immun 2023; 107:345-360. [PMID: 36328163 PMCID: PMC11906171 DOI: 10.1016/j.bbi.2022.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/24/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Almost 2/3rds of stroke survivors exhibit vascular cognitive impairment and a third of stroke patients will develop dementia 1-3 years after stroke. These dire consequences underscore the need for effective stroke therapies. In addition to its damaging effects on the brain, stroke rapidly dysregulates the intestinal epithelium, resulting in elevated blood levels of inflammatory cytokines and toxic gut metabolites due to a 'leaky' gut. We tested whether repairing the gut via intestinal epithelial stem cell (IESC) transplants would also improve stroke recovery. Organoids containing IESCs derived from young rats transplanted into older rats after stroke were incorporated into the gut, restored stroke-induced gut dysmorphology and decreased gut permeability, and reduced circulating levels of endotoxin LPS and the inflammatory cytokine IL-17A. Remarkably, IESC transplants also improved stroke-induced acute (4d) sensory-motor disability and chronic (30d) cognitive-affective function. Moreover, IESCs from older animals displayed senescent features and were not therapeutic for stroke. These data underscore the gut as a critical therapeutic target for stroke and demonstrate the effectiveness of gut stem cell therapy.
Collapse
Affiliation(s)
- Kathiresh Kumar Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States; Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX 77807, United States
| | - Yumna El-Hakim
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Taylor E Branyan
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States; Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX 77807, United States
| | - Nadia Samiya
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Sivani Pandey
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Maria T Grimaldo
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Ali Habbal
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Anna Wertz
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, College of Medicine, Mail Stop 1359 | 8447 Riverside Pkwy, Bryan, TX 77807-3260, United States; Texas A&M Institute for Neuroscience, Texas A&M University, Bryan, TX 77807, United States.
| |
Collapse
|
11
|
Branyan TE, Selvamani A, Park MJ, Korula KE, Kosel KF, Srinivasan R, Sohrabji F. Functional Assessment of Stroke-Induced Regulation of miR-20a-3p and Its Role as a Neuroprotectant. Transl Stroke Res 2022; 13:432-448. [PMID: 34570349 PMCID: PMC9046320 DOI: 10.1007/s12975-021-00945-x] [Citation(s) in RCA: 9] [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: 05/02/2021] [Revised: 08/12/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022]
Abstract
MicroRNAs have gained popularity as a potential treatment for many diseases, including stroke. This study identifies and characterizes a specific member of the miR-17-92 cluster, miR-20a-3p, as a possible stroke therapeutic. A comprehensive microRNA screening showed that miR-20a-3p was significantly upregulated in astrocytes of adult female rats, which typically have better stroke outcomes, while it was profoundly downregulated in astrocytes of middle-aged females and adult and middle-aged males, groups that typically have more severe stroke outcomes. Assays using primary human astrocytes and neurons show that miR-20a-3p treatment alters mitochondrial dynamics in both cell types. To assess whether stroke outcomes could be improved by elevating astrocytic miR-20a-3p, we created a tetracycline (Tet)-induced recombinant adeno-associated virus (rAAV) construct where miR-20a-3p was located downstream a glial fibrillary acidic protein promoter. Treatment with doxycycline induced miR-20-3p expression in astrocytes, reducing mortality and modestly improving sensory motor behavior. A second Tet-induced rAAV construct was created in which miR-20a-3p was located downstream of a neuron-specific enolase (NSE) promoter. These experiments demonstrate that neuronal expression of miR-20a-3p is vastly more neuroprotective than astrocytic expression, with animals receiving the miR-20a-3p vector showing reduced infarction and sensory motor improvement. Intravenous injections, which are a therapeutically tractable treatment route, with miR-20a-3p mimic 4 h after middle cerebral artery occlusion (MCAo) significantly improved stroke outcomes including infarct volume and sensory motor performance. Improvement was not observed when miR-20a-3p was given immediately or 24 h after MCAo, identifying a unique delayed therapeutic window. Overall, this study identifies a novel neuroprotective microRNA and characterizes several key pathways by which it can improve stroke outcomes.
Collapse
Affiliation(s)
- Taylor E Branyan
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
- Texas A&M Institute for Neuroscience, College Station, TX, 77840, USA
| | - Amutha Selvamani
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Min Jung Park
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Kriti E Korula
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Kelby F Kosel
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
| | - Rahul Srinivasan
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA
- Texas A&M Institute for Neuroscience, College Station, TX, 77840, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, 77807, USA.
- Texas A&M Institute for Neuroscience, College Station, TX, 77840, USA.
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, 8447 Riverside Pkwy, Bryan, TX, 77807, USA.
| |
Collapse
|
12
|
Lopez MS, Morris-Blanco KC, Ly N, Maves C, Dempsey RJ, Vemuganti R. MicroRNA miR-21 Decreases Post-stroke Brain Damage in Rodents. Transl Stroke Res 2022; 13:483-493. [PMID: 34796453 PMCID: PMC11846127 DOI: 10.1007/s12975-021-00952-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022]
Abstract
Due to their role in controlling translation, microRNAs emerged as novel therapeutic targets to modulate post-stroke outcomes. We previously reported that miR-21 is the most abundantly induced microRNA in the brain of rodents subjected to preconditioning-induced cerebral ischemic tolerance. We currently show that intracerebral administration of miR-21 mimic decreased the infarct volume and promoted better motor function recovery in adult male and female C57BL/6 mice subjected to transient middle cerebral artery occlusion. The miR-21 mimic treatment is also efficacious in aged mice of both sexes subjected to focal ischemia. Mechanistically, miR-21 mimic treatment decreased the post-ischemic levels of several pro-apoptotic and pro-inflammatory RNAs, which might be responsible for the observed neuroprotection. We further observed post-ischemic neuroprotection in adult mice administered with miR-21 mimic intravenously. Overall, the results of this study implicate miR-21 as a promising candidate for therapeutic translation after stroke.
Collapse
Affiliation(s)
- Mary S Lopez
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
- Cell & Molecular Pathology Training Program, University of Wisconsin, Madison, WI, 53792, USA
| | | | - Nancy Ly
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Carly Maves
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, 53792, USA.
- Cell & Molecular Pathology Training Program, University of Wisconsin, Madison, WI, 53792, USA.
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
13
|
Stewart CE, Branyan TE, Sampath D, Sohrabji F. Sex Differences in the Long-Term Consequences of Stroke. Curr Top Behav Neurosci 2022; 62:287-308. [PMID: 35332459 DOI: 10.1007/7854_2022_311] [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: 11/24/2022]
Abstract
Stroke is the fifth leading cause of death and as healthcare intervention improves, the number of stroke survivors has also increased. Furthermore, there exists a subgroup of younger adults, who suffer stroke and survive. Given the overall improved survival rate, bettering our understanding of long-term stroke outcomes is critical. In this review we will explore the causes and challenges of known long-term consequences of stroke and if present, their corresponding sex differences in both old and young survivors. We have separated these long-term post-stroke consequences into three categories: mobility and muscle weakness, memory and cognitive deficits, and mental health and mood. Lastly, we discuss the potential of common preclinical stroke models to contribute to our understanding of long-term outcomes following stroke.
Collapse
Affiliation(s)
- Courtney E Stewart
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Taylor E Branyan
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA.,Texas A&M Institute for Neuroscience, College Station, TX, USA
| | - Dayalan Sampath
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX, USA. .,Texas A&M Institute for Neuroscience, College Station, TX, USA.
| |
Collapse
|
14
|
Bake S, Pinson MR, Pandey S, Chambers JP, Mota R, Fairchild AE, Miranda RC, Sohrabji F. Prenatal alcohol-induced sex differences in immune, metabolic and neurobehavioral outcomes in adult rats. Brain Behav Immun 2021; 98:86-100. [PMID: 34390803 PMCID: PMC8591773 DOI: 10.1016/j.bbi.2021.08.207] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 07/10/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can result in neurobehavioral anomalies, that may be exacerbated by co-occurring metabolic and immune system deficits. To test the hypothesis that the peripheral inflammation in adult PAE offspring is linked to poor glucose metabolism and neurocognitive deficits, pregnant Sprague-Dawley rats were exposed to ethanol vapor or ambient air during the latter half of gestation. We assessed, in adult offspring of both sexes, performance on a battery of neurocognitive behaviors, glucose tolerance, circulating and splenic immune cells by flow-cytometry, and circulating and tissue (liver, mesenteric adipose, and spleen) cytokines by multiplexed assays. PAE reduced both the ratio of spleen to body weight and splenic regulatory T-cell (Treg) numbers. PAE males, but not females exhibited an increase in circulating monocytes. Overall, PAE males exhibited a suppression of cytokine levels, while PAE females exhibited elevated cytokines in mesenteric adipose tissue (IL-6 and IL1α) and liver (IFN-γ, IL-1β, IL-13, IL-18, IL-12p70, and MCP-1), along with increased glucose intolerance. Behavioral analysis also showed sex-dependent PAE effects. PAE-males exhibited increased anxiety-like behavior while PAE-females showed decreased social interaction. PAE offspring of both sexes exhibited impaired recognition of novel objects. Multilinear regression modeling to predict the association between peripheral immune status, glucose intolerance and behavioral outcomes, showed that in PAE offspring, higher levels of adipose leptin and liver TNF- α predicted higher circulating glucose levels. Lower liver IL-1 α and higher plasma fractalkine predicted more time spent in the center of an open-field with sex being an additional predictor. Higher circulating and splenic Tregs predicted better social interaction in the PAE-offspring. Collectively, our data show that peripheral immune status is a persistent, sex-dependent predictor of glucose intolerance and neurobehavioral function in adult PAE offspring.
Collapse
Affiliation(s)
- Shameena Bake
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Marisa R Pinson
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Sivani Pandey
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joanna P Chambers
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Roxanna Mota
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Ashlyn E Fairchild
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Rajesh C Miranda
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, and Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| |
Collapse
|
15
|
Le Marois M, Tzavara E, Ibrahim EC, Blin O, Belzeaux R. RNA therapeutics for mood disorders: current evidence toward clinical trials. Expert Opin Investig Drugs 2021; 30:721-736. [PMID: 33966550 DOI: 10.1080/13543784.2021.1928073] [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: 10/21/2022]
Abstract
INTRODUCTION Mood disorders are severe yet frequent psychiatric disorders worldwide, comprising major depressive disorder (MDD) and bipolar disorders (BD). Their treatment remains poorly effective. Recently, growing evidence for epigenetic mechanisms has emerged. Consequently, a great interest in a novel pharmacological class arose: RNA therapeutics. AREAS COVERED We conducted a systematic review of RNA therapeutics -antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), short hairpin RNAs (shRNAs), and micro-RNA (miRNA) therapeutics- for the treatment of mood disorders studied in pre-clinical animal models listed in PubMed, in clinical trials registered in ClinicalTrials.gov and available on the market by combining literature search and Food and Drug Administration and European Medicine Agency online databases. Eighteen pre-clinical studies investigated the antidepressant effects of RNA therapeutics. However, even though there is an increasing number of marketing authorizations and clinical trials for the past twenty years, no RNA therapeutic has reached the clinical development pipeline for the treatment of psychiatric disorders yet. EXPERT OPINION Several promising RNA therapeutics have been tested in pre-clinical studies for MDD, whereas no molecule has been developed for BD. There are several issues to address before reaching clinical development and new challenges include stratifying patient population and predicting therapeutic response.
Collapse
Affiliation(s)
- Marguerite Le Marois
- Aix Marseille Univ, APHM, INSERM, Inst Neurosci Syst, University Hospital Federation DHUNE, Service de Pharmacologie Clinique et Pharmacovigilance, Marseille, France.,Aix Marseille Univ, CNRS, Inst Neurosci Timone, Marseille, France
| | - Eleni Tzavara
- Fondation FondaMental, Créteil, France.,Pôle De Psychiatrie, Hôpital Sainte-Marguerite, AP-HM, Marseille, France
| | - El Chérif Ibrahim
- Aix Marseille Univ, CNRS, Inst Neurosci Timone, Marseille, France.,Fondation FondaMental, Créteil, France
| | - Olivier Blin
- Aix Marseille Univ, APHM, INSERM, Inst Neurosci Syst, University Hospital Federation DHUNE, Service de Pharmacologie Clinique et Pharmacovigilance, Marseille, France
| | - Raoul Belzeaux
- Aix Marseille Univ, CNRS, Inst Neurosci Timone, Marseille, France.,Fondation FondaMental, Créteil, France.,Pôle De Psychiatrie, Hôpital Sainte-Marguerite, AP-HM, Marseille, France
| |
Collapse
|
16
|
Martins HC, Schratt G. MicroRNA-dependent control of neuroplasticity in affective disorders. Transl Psychiatry 2021; 11:263. [PMID: 33941769 PMCID: PMC8093191 DOI: 10.1038/s41398-021-01379-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/17/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
Affective disorders are a group of neuropsychiatric disorders characterized by severe mood dysregulations accompanied by sleep, eating, cognitive, and attention disturbances, as well as recurring thoughts of suicide. Clinical studies consistently show that affective disorders are associated with reduced size of brain regions critical for mood and cognition, neuronal atrophy, and synaptic loss in these regions. However, the molecular mechanisms that mediate these changes and thereby increase the susceptibility to develop affective disorders remain poorly understood. MicroRNAs (miRNAs or miRs) are small regulatory RNAs that repress gene expression by binding to the 3'UTR of mRNAs. They have the ability to bind to hundreds of target mRNAs and to regulate entire gene networks and cellular pathways implicated in brain function and plasticity, many of them conserved in humans and other animals. In rodents, miRNAs regulate synaptic plasticity by controlling the morphology of dendrites and spines and the expression of neurotransmitter receptors. Furthermore, dysregulated miRNA expression is frequently observed in patients suffering from affective disorders. Together, multiple lines of evidence suggest a link between miRNA dysfunction and affective disorder pathology, providing a rationale to consider miRNAs as therapeutic tools or molecular biomarkers. This review aims to highlight the most recent and functionally relevant studies that contributed to a better understanding of miRNA function in the development and pathogenesis of affective disorders. We focused on in vivo functional studies, which demonstrate that miRNAs control higher brain functions, including mood and cognition, in rodents, and that their dysregulation causes disease-related behaviors.
Collapse
Affiliation(s)
- Helena Caria Martins
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, 8057, Zurich, Switzerland
| | - Gerhard Schratt
- Lab of Systems Neuroscience, Institute for Neuroscience, Department of Health Science and Technology, Swiss Federal Institute of Technology ETH, 8057, Zurich, Switzerland.
| |
Collapse
|
17
|
Miao C, Chang J. The important roles of microRNAs in depression: new research progress and future prospects. J Mol Med (Berl) 2021; 99:619-636. [PMID: 33641067 DOI: 10.1007/s00109-021-02052-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/04/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) are non-encoding, single-stranded RNA molecules of about 22 nucleotides in length encoded by endogenous genes involved in posttranscriptional gene expression regulation. Studies have shown that miRNAs participate in a series of important pathophysiological processes, including the pathogenesis of depression. This article systematically summarized the research results published in the field of miRNAs and depression, which mainly involved three topics: circulating miRNAs as markers for diagnosis and prognosis of depression, the regulatory roles of miRNAs in the pathogenesis of depression, and the roles of miRNAs in the mechanisms of depression treatment. By summarizing and analyzing the research literature in recent years, we found that some circulating miRNAs can be potential biomarkers for the diagnosis and prognostic evaluation of depression. miRNAs that disorderly expressed during the disease play important roles in the depression pathogenesis, and miRNAs also play roles in the mechanisms of psychotherapy and drug therapy for depression. Elucidating the important roles of miRNAs in depression will bring people's understanding of the pathogenesis of depression to a new level. In addition, these miRNAs may be developed as new biomarkers for diagnosing depression, or as drug targets, or these molecules may be used as new drugs, which may provide new means for the treatment of depression. KEY MESSAGES: • The research results of miRNAs and depression are reviewed. • Circulating miRNAs can be potential biomarkers for depression. • MiRNAs play important roles in the depression pathogenesis. • MiRNAs play important roles in drug therapy for depression.
Collapse
Affiliation(s)
- Chenggui Miao
- Department of Pharmacology, College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 1 Qianjiang Road, Xinzhan District, Hefei, 230012, Anhui Province, China. .,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China. .,Anhui Provincial Key Laboratory of Chinese Medicine Compound, Anhui University of Chinese Medicine, Hefei, 230012, China. .,Institute of Life and Health Sciences, Anhui University of Science and Technology, Fengyang, 233100, China.
| | - Jun Chang
- Fourth Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
18
|
Boltze J, Aronowski JA, Badaut J, Buckwalter MS, Caleo M, Chopp M, Dave KR, Didwischus N, Dijkhuizen RM, Doeppner TR, Dreier JP, Fouad K, Gelderblom M, Gertz K, Golubczyk D, Gregson BA, Hamel E, Hanley DF, Härtig W, Hummel FC, Ikhsan M, Janowski M, Jolkkonen J, Karuppagounder SS, Keep RF, Koerte IK, Kokaia Z, Li P, Liu F, Lizasoain I, Ludewig P, Metz GAS, Montagne A, Obenaus A, Palumbo A, Pearl M, Perez-Pinzon M, Planas AM, Plesnila N, Raval AP, Rueger MA, Sansing LH, Sohrabji F, Stagg CJ, Stetler RA, Stowe AM, Sun D, Taguchi A, Tanter M, Vay SU, Vemuganti R, Vivien D, Walczak P, Wang J, Xiong Y, Zille M. New Mechanistic Insights, Novel Treatment Paradigms, and Clinical Progress in Cerebrovascular Diseases. Front Aging Neurosci 2021; 13:623751. [PMID: 33584250 PMCID: PMC7876251 DOI: 10.3389/fnagi.2021.623751] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
The past decade has brought tremendous progress in diagnostic and therapeutic options for cerebrovascular diseases as exemplified by the advent of thrombectomy in ischemic stroke, benefitting a steeply increasing number of stroke patients and potentially paving the way for a renaissance of neuroprotectants. Progress in basic science has been equally impressive. Based on a deeper understanding of pathomechanisms underlying cerebrovascular diseases, new therapeutic targets have been identified and novel treatment strategies such as pre- and post-conditioning methods were developed. Moreover, translationally relevant aspects are increasingly recognized in basic science studies, which is believed to increase their predictive value and the relevance of obtained findings for clinical application.This review reports key results from some of the most remarkable and encouraging achievements in neurovascular research that have been reported at the 10th International Symposium on Neuroprotection and Neurorepair. Basic science topics discussed herein focus on aspects such as neuroinflammation, extracellular vesicles, and the role of sex and age on stroke recovery. Translational reports highlighted endovascular techniques and targeted delivery methods, neurorehabilitation, advanced functional testing approaches for experimental studies, pre-and post-conditioning approaches as well as novel imaging and treatment strategies. Beyond ischemic stroke, particular emphasis was given on activities in the fields of traumatic brain injury and cerebral hemorrhage in which promising preclinical and clinical results have been reported. Although the number of neutral outcomes in clinical trials is still remarkably high when targeting cerebrovascular diseases, we begin to evidence stepwise but continuous progress towards novel treatment options. Advances in preclinical and translational research as reported herein are believed to have formed a solid foundation for this progress.
Collapse
Affiliation(s)
- Johannes Boltze
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Jaroslaw A. Aronowski
- Institute for Stroke and Cerebrovascular Diseases, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jerome Badaut
- NRS UMR 5287, INCIA, Brain Molecular Imaging Team, University of Bordeaux, Bordeaux cedex, France
| | - Marion S. Buckwalter
- Departments of Neurology and Neurological Sciences, and Neurosurgery, Wu Tsai Neurosciences Institute, Stanford School of Medicine, Stanford, CA, United States
| | - Mateo Caleo
- Neuroscience Institute, National Research Council, Pisa, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
| | - Kunjan R. Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nadine Didwischus
- School of Life Sciences, University of Warwick, Warwick, United Kingdom
| | - Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, Netherlands
| | - Thorsten R. Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens P. Dreier
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Department of Experimental Neurology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Karim Fouad
- Faculty of Rehabilitation Medicine and Institute for Neuroscience and Mental Health, University of Alberta, Edmonton, AB, Canada
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Karen Gertz
- Department of Neurology, Center for Stroke Research Berlin, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, University of Warmia and Mazury, Olsztyn, Poland
| | - Barbara A. Gregson
- Neurosurgical Trials Group, Institute of Neuroscience, The University of Newcastle upon Tyne, Newcastle upon Tyne, United Kingdom
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Daniel F. Hanley
- Division of Brain Injury Outcomes, Johns Hopkins University, Baltimore, MD, United States
| | - Wolfgang Härtig
- Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Friedhelm C. Hummel
- Clinical Neuroengineering, Center for Neuroprosthetics and Brain Mind Institute, Swiss Federal Institute of Technology Valais, Clinique Romande de Réadaptation, Sion, Switzerland
- Clinical Neuroscience, University of Geneva Medical School, Geneva, Switzerland
| | - Maulana Ikhsan
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Miroslaw Janowski
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jukka Jolkkonen
- Department of Neurology, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Saravanan S. Karuppagounder
- Burke Neurological Institute, White Plains, NY, United States
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Inga K. Koerte
- Psychiatric Neuroimaging Laboratory, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Child and Adolescent Psychiatry, Psychosomatic, and Psychotherapy, Ludwig Maximilians University, Munich, Germany
| | - Zaal Kokaia
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fudong Liu
- Department of Neurology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, United States
| | - Ignacio Lizasoain
- Unidad de Investigación Neurovascular, Departamento Farmacología y Toxicología, Facultad de Medicina, Instituto Universitario de Investigación en Neuroquímica, Universidad Complutense de Madrid, Madrid, Spain
| | - Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerlinde A. S. Metz
- Department of Neuroscience, Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Andre Obenaus
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Alex Palumbo
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| | - Monica Pearl
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Miguel Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna M. Planas
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Àrea de Neurociències, Barcelona, Spain
- Department d’Isquèmia Cerebral I Neurodegeneració, Institut d’Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), Munich University Hospital, Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Ami P. Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maria A. Rueger
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Farida Sohrabji
- Women’s Health in Neuroscience Program, Neuroscience and Experimental Therapeutics, Texas A&M College of Medicine, Bryan, TX, United States
| | - Charlotte J. Stagg
- Nuffield Department of Clinical Neurosciences, Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom
- MRC Brain Network Dynamics Unit, University of Oxford, Oxford, United Kingdom
| | - R. Anne Stetler
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ann M. Stowe
- Department of Neurology and Neurotherapeutics, Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, United States
| | - Dandan Sun
- Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, PA, United States
| | - Akihiko Taguchi
- Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Mickael Tanter
- Institute of Physics for Medicine Paris, INSERM U1273, ESPCI Paris, CNRS FRE 2031, PSL University, Paris, France
| | - Sabine U. Vay
- Faculty of Medicine and University Hospital, Department of Neurology, University of Cologne, Cologne, Germany
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, United States
| | - Denis Vivien
- UNICAEN, INSERM, INSERM UMR-S U1237, Physiopathology and Imaging for Neurological Disorders (PhIND), Normandy University, Caen, France
- CHU Caen, Clinical Research Department, CHU de Caen Côte de Nacre, Caen, France
| | - Piotr Walczak
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland, Baltimore, MD, United States
| | - Jian Wang
- Department of Human Anatomy, College of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, United States
| | - Marietta Zille
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
- Fraunhofer Research Institution for Marine Biotechnology and Cell Technology, Lübeck, Germany
- Institute for Medical and Marine Biotechnology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
19
|
Sarkar A, Sarmah D, Datta A, Kaur H, Jagtap P, Raut S, Shah B, Singh U, Baidya F, Bohra M, Kalia K, Borah A, Wang X, Dave KR, Yavagal DR, Bhattacharya P. Post-stroke depression: Chaos to exposition. Brain Res Bull 2020; 168:74-88. [PMID: 33359639 DOI: 10.1016/j.brainresbull.2020.12.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 12/09/2020] [Accepted: 12/18/2020] [Indexed: 12/12/2022]
Abstract
Cerebral ischemia contributes to significant disabilities worldwide, impairing cognitive function and motor coordination in affected individuals. Stroke has severe neuropsychological outcomes, the major one being a stroke. Stroke survivors begin to show symptoms of depression within a few months of the incidence that overtime progresses to become a long-term ailment. As the pathophysiology for the progression of the disease is multifactorial and complex, it limits the understanding of the disease mechanism completely. Meta-analyses and randomized clinical trials have shown that intervening early with tricyclic antidepressants and selective serotonin receptor inhibitors can be effective. However, these pharmacotherapies possess several limitations that have given rise to newer approaches such as brain stimulation, psychotherapy and rehabilitation therapy, which in today's time are gaining attention for their beneficial results in post-stroke depression (PSD). The present review highlights numerous factors like lesion location, inflammatory mediators and genetic abnormalities that play a crucial role in the development of depression in stroke patients. Further, we have also discussed various mechanisms involved in post-stroke depression (PSD) and strategies for early detection and diagnosis using biomarkers that may revolutionize treatment for the affected population. Towards the end, along with the preclinical scenario, we have also discussed the various treatment approaches like pharmacotherapy, traditional medicines, psychotherapy, electrical stimulation and microRNAs being utilized for effectively managing PSD.
Collapse
Affiliation(s)
- Ankan Sarkar
- 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
| | - Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Priya Jagtap
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Swapnil Raut
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Birva Shah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Upasna Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Falguni Baidya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Mariya Bohra
- 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, USA
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Dileep R Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India.
| |
Collapse
|
20
|
Gonçalves NP, Yan Y, Ulrichsen M, Venø MT, Poulsen ET, Enghild JJ, Kjems J, Vægter CB. Modulation of Small RNA Signatures in Schwann-Cell-Derived Extracellular Vesicles by the p75 Neurotrophin Receptor and Sortilin. Biomedicines 2020; 8:E450. [PMID: 33114403 PMCID: PMC7694014 DOI: 10.3390/biomedicines8110450] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
Schwann cells (SCs) are the main glial cells of the peripheral nervous system (PNS) and are known to be involved in various pathophysiological processes, such as diabetic neuropathy and nerve regeneration, through neurotrophin signaling. Such glial trophic support to axons, as well as neuronal survival/death signaling, has previously been linked to the p75 neurotrophin receptor (p75NTR) and its co-receptor Sortilin. Recently, SC-derived extracellular vesicles (EVs) were shown to be important for axon growth and nerve regeneration, but cargo of these glial cell-derived EVs has not yet been well-characterized. In this study, we aimed to characterize signatures of small RNAs in EVs derived from wild-type (WT) SCs and define differentially expressed small RNAs in EVs derived from SCs with genetic deletions of p75NTR (Ngfr-/-) or Sortilin (Sort1-/-). Using RNA sequencing, we identified a total of 366 miRNAs in EVs derived from WT SCs of which the most highly expressed are linked to the regulation of axonogenesis, axon guidance and axon extension, suggesting an involvement of SC EVs in axonal homeostasis. Signaling of SC EVs to non-neuronal cells was also suggested by the presence of several miRNAs important for regulation of the endothelial cell apoptotic process. Ablated p75NTR or sortilin expression in SCs translated into a set of differentially regulated tRNAs and miRNAs, with impact in autophagy and several cellular signaling pathways such as the phosphatidylinositol signaling system. With this work, we identified the global expression profile of small RNAs present in SC-derived EVs and provided evidence for a regulatory function of these vesicles on the homeostasis of other cell types of the PNS. Differentially identified miRNAs can pave the way to a better understanding of p75NTR and sortilin roles regarding PNS homeostasis and disease.
Collapse
Affiliation(s)
- Nádia P. Gonçalves
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| | - Yan Yan
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Omiics ApS, 8000 Aarhus, Denmark
| | - Maj Ulrichsen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| | - Morten T. Venø
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Omiics ApS, 8000 Aarhus, Denmark
| | - Ebbe T. Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Jan J. Enghild
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000 Aarhus, Denmark; (Y.Y.); (M.T.V.); (J.K.)
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark; (E.T.P.); (J.J.E.)
| | - Christian B. Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic-EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark; (M.U.); (C.B.V.)
| |
Collapse
|
21
|
Panta A, Montgomery K, Nicolas M, Mani KK, Sampath D, Sohrabji F. Mir363-3p Treatment Attenuates Long-Term Cognitive Deficits Precipitated by an Ischemic Stroke in Middle-Aged Female Rats. Front Aging Neurosci 2020; 12:586362. [PMID: 33132904 PMCID: PMC7550720 DOI: 10.3389/fnagi.2020.586362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 01/29/2023] Open
Abstract
Cognitive impairment and memory loss are commonly seen after stroke and a third of patients will develop signs of dementia a year after stroke. Despite a large number of studies on the beneficial effects of neuroprotectants, few studies have examined the effects of these compounds/interventions on long-term cognitive impairment. Our previous work showed that the microRNA mir363-3p reduced infarct volume and sensory-motor impairment in the acute stage of stroke in middle-aged females but not males. Thus, the present study determined the impact of mir363-3p treatment on stroke-induced cognitive impairment in middle-aged females. Sprague–Dawley female rats (12 months of age) were subjected to middle cerebral artery occlusion (MCAo; or sham surgery) and injected (iv) with mir363-3p mimic (MCAo + mir363-3p) or scrambled oligos (MCAo + scrambled) 4 h later. Sensory-motor performance was assessed in the acute phase (2–5 days after stroke), while all other behaviors were tested 6 months after MCAo (18 months of age). Cognitive function was assessed by the novel object recognition test (declarative memory) and the Barnes maze (spatial memory). The MCAo + scrambled group showed reduced preference for a novel object after the stroke and poor learning in the spatial memory task. In contrast, mir363-3p treated animals were similar to either their baseline performance or to the sham group. Histological analysis showed significant deterioration of specific white matter tracts due to stroke, which was attenuated in mir363-3p treated animals. The present data builds on our previous finding to show that a neuroprotectant can abrogate the long-term effects of stroke.
Collapse
Affiliation(s)
- Aditya Panta
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Karienn Montgomery
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Marissa Nicolas
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Kathiresh K Mani
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Dayalan Sampath
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Farida Sohrabji
- Women's Health in Neuroscience Program, Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
22
|
Mukhopadhyay D, Mussa BM. Identification of Novel Hypothalamic MicroRNAs as Promising Therapeutics for SARS-CoV-2 by Regulating ACE2 and TMPRSS2 Expression: An In Silico Analysis. Brain Sci 2020; 10:E666. [PMID: 32992681 PMCID: PMC7601472 DOI: 10.3390/brainsci10100666] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neuroinvasion of severe acute respiratory syndrome coronavirus (SARS-CoV) is well documented and, given the similarities between this virus and SARS-CoV-2, it seems that the neurological impairment that is associated with coronavirus disease 2019 (COVID-19) is due to SARS-CoV-2 neuroinvasion. Hypothalamic circuits are exposed to the entry of the virus via the olfactory bulb and interact centrally with crucial respiratory nuclei. Hypothalamic microRNAs are considered as potential biomarkers and modulators for various diseases and future therapeutic targets. The present study aims to investigate the microRNAs that regulate the expression of hypothalamic angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), essential elements for SARS-CoV-2 cell entry. METHODS To determine potential hypothalamic miRNAs that can directly bind to ACE2 and TMPRSS2, multiple target bioinformatics prediction algorithms were used, including miRBase, Target scan, and miRWalk2.029. RESULTS Our in silico analysis has revealed that, although there are over 5000 hypothalamic miRNAs, around 31 miRNAs and 29 miRNAs have shown binding sites and strong binding capacity against ACE2 and TMPRSS2, respectively. CONCLUSION These novel potential hypothalamic miRNAs can be used to identify new therapeutic targets to treat neurological symptoms in COVID-19 patients via regulation of ACE2 and TMPRSS2 expression.
Collapse
Affiliation(s)
- Debasmita Mukhopadhyay
- Biomedical & Chemical Engineering Department, American University of Sharjah, Sharjah 26666, UAE;
| | - Bashair M. Mussa
- Basic Medical Science Department, College of Medicine, University of Sharjah, Sharjah 27272, UAE
| |
Collapse
|
23
|
Kaushik P, Ali M, Tabassum H, Parvez S. Post-ischemic administration of dopamine D2 receptor agonist reduces cell death by activating mitochondrial pathway following ischemic stroke. Life Sci 2020; 261:118349. [PMID: 32853654 DOI: 10.1016/j.lfs.2020.118349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 08/17/2020] [Accepted: 08/22/2020] [Indexed: 12/21/2022]
Abstract
AIMS Cerebral ischemic stroke leads to mitochondrial alterations which are key factors for initiation of various cascades resulting in neuronal damage. Dopamine D2 receptor (D2R) agonist, Sumanirole (SUM) has been reported to possess anti-inflammatory, anti-oxidant, and anti-apoptotic properties. However, the role of SUM in ischemic stroke (IS) has not been studied yet. The aim of the present study was to investigate the neuroprotective efficiency of SUM against ischemic injury and its possible effect on mitochondrial restorative mechanisms. MATERIALS AND METHODS Transient middle cerebral artery occlusion (tMCAO) was performed in Wistar rats for 90 min occlusion and 22.5 h reperfusion to mimic ischemic stroke. Post- treatment with Sumanirole (0.1 mg/kg and 1 mg/kg; s.c.) was done at 1 h, 6 h, 12 hand 18 h after surgery. In addition, neurobehavioral analysis, mitochondrial reactive oxygen species and mitochondrial membrane potential by flow cytometric analysis, mitochondrial complexes analysis, infarct size evaluation and histological analysis were performed. KEY FINDINGS Sumanirole restored behavioural alterations as measured by rotarod performance, grip strength, adhesive tape removal analysis and neurological deficits. In addition, it also refurbished mitochondrial dysfunction by decreasing mitochondrial reactive oxygen species production, elevating mitochondrial membrane potential and by protecting the activity of mitochondrial complexes along with histological alterations. As a result, infarct sizes were markedly reduced in tMCAO surgery animals. SIGNIFICANCE Findings from the study provide evidence that SUM promotes neuronal survival in in vivo model of IS through mitochondria mediated neuroprotective features.
Collapse
Affiliation(s)
- Pooja Kaushik
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Mubashshir Ali
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Heena Tabassum
- Division of Basic Medical Sciences, Indian Council of Medical Research, Ministry of Health and Family Welfare, Government of India, V. Ramalingaswamy Bhawan, New Delhi 110029, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|