Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters








Database
Language
Publication year range
1.
Cell Mol Neurobiol ; 39(2): 223-240, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30539420

ABSTRACT

The irrefutable change in the expression of brain-enriched microRNAs (miRNAs) following ischemic stroke has promoted the development of radical miRNA-based therapeutics encompassing neuroprotection and neuronal restoration. Our previous report on the systems-level prediction of miR-9 in post-stroke-induced neurogenesis served as a premise to experimentally uncover the functional role of miR-9 in post-ischemic neuronal survival and regeneration. The oxygen-glucose deprivation (OGD) in SH-SY5Y cells significantly reduced miR-9 expression, while miR-9 mimic transfection enhanced post-ischemic neuronal cell viability. The next major objective involved the execution of a drug repositioning strategy to augment miR-9 expression via structure-based screening of Food and Drug Administration (FDA)-approved drugs that bind to Histone Deacetylase 4 (HDAC4), a known miR-9 target. Glucosamine emerged as the top hit and its binding potential to HDAC4 was verified by Molecular Dynamics (MD) Simulation, Drug Affinity Responsive Target Stability (DARTS) assay, and MALDI-TOF MS. It was intriguing that the glucosamine treatment 1-h post-OGD was associated with the increased miR-9 level as well as enhanced neuronal viability. miR-9 mimic or post-OGD glucosamine treatment significantly increased the cellular proliferation (BrdU assay), while the neurite outgrowth assay displayed elongated neurites. The enhanced BCL2 and VEGF parallel with the reduced NFκB1, TNF-α, IL-1ß, and iNOS mRNA levels in miR-9 mimic or glucosamine-treated cells further substantiated their post-ischemic neuroprotective and regenerative efficacy. Hence, this study unleashes a potential therapeutic approach that integrates neuronal survival and regeneration via small-molecule-based regulation of miR-9 favoring long-term recovery against ischemic stroke.


Subject(s)
Brain Ischemia/genetics , Brain Ischemia/pathology , MicroRNAs/genetics , Nerve Regeneration , Up-Regulation/genetics , Apoptosis , Brain Ischemia/physiopathology , Catalytic Domain , Cell Line, Tumor , Cell Survival/genetics , Glucosamine/metabolism , Glucose/deficiency , Histone Deacetylases/metabolism , Humans , Inflammation Mediators/metabolism , Ligands , MicroRNAs/metabolism , Molecular Docking Simulation , Necrosis , Neurites/metabolism , Neuroprotection , Oxygen , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism
2.
Mol Neurobiol ; 55(11): 8251-8262, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29524052

ABSTRACT

The complex and interlinked cascade of events regulated by microRNAs (miRNAs), transcription factors (TF), and target genes highlight the multifactorial nature of ischemic stroke pathology. The complexity of ischemic stroke requires a wider assessment than the existing experimental research that deals with only a few regulatory components. Here, we assessed a massive set of genes, miRNAs, and transcription factors to build a miRNA-gene-transcription factor regulatory network to elucidate the underlying post-transcriptional mechanisms in ischemic stroke. Feed-forward loops (three-node, four-node, and novel five-node) were converged to establish regulatory relationships between miRNAs, TFs, and genes. The synergistic function of miRNAs in ischemic stroke was predicted and incorporated into a novel five-node feed-forward loop. Significant miRNA-TF pairs were identified using cumulative hypergeometric distribution. Two subnetworks were derived from the extensive miRNA-TF regulatory network and analyzed to predict the molecular mechanism relating the regulatory components. NFKB and STAT were identified to be the chief regulators of innate inflammatory and neuronal survival mechanisms, respectively. Exclusive novel interactions between miR-9 and miR-124 with TLX, BCL2, and HDAC4 were identified to explain the post-stroke induced neurogenesis mechanism. Therefore, this network-based approach to delineate miRNA, TF, and gene interactions might promote the development of effective therapeutics against ischemic stroke.


Subject(s)
Brain Ischemia/genetics , Gene Regulatory Networks , MicroRNAs/genetics , Stroke/genetics , Transcription Factors/metabolism , Humans , MicroRNAs/metabolism , Neurogenesis/genetics , Signal Transduction/genetics
3.
Mol Neurobiol ; 55(11): 8328-8345, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29542054

ABSTRACT

The constant failure of single-target drug therapies for ischemic stroke necessitates the development of novel pleiotropic pharmacological treatment approaches, to effectively combat the aftermath of this devastating disorder. The major objective of our study involves a multi-target drug repurposing strategy to stabilize hypoxia-inducible factor-1 α (HIF-1α) via a structure-based screening approach to simultaneously inhibit its regulatory proteins, PHD2, FIH, and pVHL. Out of 1424 Food and Drug Administration (FDA)-approved drugs that were screened, folic acid (FA) emerged as the top hit and its binding potential to PHD2, FIH, and pVHL was further verified by re-docking, molecular dynamics (MD) simulation and by Drug Affinity Responsive Target Stability (DARTS) assay. HIF-1α stabilization by FA was demonstrated by the nuclear translocation and increased green fluorescence emission of HIF-1α using HIF1α-GFPSpark tag vector. Further, FA treatment enhanced the cell survival following oxygen glucose deprivation and its neuroprotective mechanism was elucidated by measuring the expression of BAX, NFE2L2, VEGF, and EPO genes in a time-dependent manner (5 and 11 h following FA treatment). VEGF and EPO expressions were significantly increased by 5.41- and 1.35-folds, respectively, whereas BAX expression reduced by 4-fold at 11 h post-FA treatment. NFE2L2 expression was elevated (1.65-fold) at 5 h with no major difference at 11 h post-FA treatment. The chicken chorioallantoic membrane (CAM) assay demonstrated the pro-angiogenic potential of FA as evidenced by an increased blood vessel density and branching. The present study elucidates for the first time that the post-ischemic neuroprotection exerted by FA may be attributed to its HIF-1α stabilization and pro-angiogenic properties.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Folic Acid/pharmacology , Neuroprotection/drug effects , Animals , Biological Assay , Cell Line, Tumor , Chickens , Folic Acid/chemistry , Gene Expression Regulation/drug effects , Glucose/deficiency , Humans , Hydrogen Bonding , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Ligands , Mixed Function Oxygenases/metabolism , Molecular Docking Simulation , Neovascularization, Physiologic/drug effects , Oxygen , Protein Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Thermodynamics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
4.
Mol Neurobiol ; 54(3): 2003-2011, 2017 04.
Article in English | MEDLINE | ID: mdl-26910816

ABSTRACT

Neurogenesis generates fledgling neurons that mature to form an intricate neuronal circuitry. The delusion on adult neurogenesis was far resolved in the past decade and became one of the largely explored domains to identify multifaceted mechanisms bridging neurodevelopment and neuropathology. Neurogenesis encompasses multiple processes including neural stem cell proliferation, neuronal differentiation, and cell fate determination. Each neurogenic process is specifically governed by manifold signaling pathways, several growth factors, coding, and non-coding RNAs. A class of small non-coding RNAs, microRNAs (miRNAs), is ubiquitously expressed in the brain and has emerged to be potent regulators of neurogenesis. It functions by fine-tuning the expression of specific neurogenic gene targets at the post-transcriptional level and modulates the development of mature neurons from neural progenitor cells. Besides the commonly discussed intrinsic factors, the neuronal morphogenesis is also under the control of several extrinsic temporal cues, which in turn are regulated by miRNAs. This review enlightens on dicer controlled switch from neurogenesis to gliogenesis, miRNA regulation of neuronal maturation and the differential expression of miRNAs in response to various extrinsic cues affecting neurogenesis.


Subject(s)
DEAD-box RNA Helicases/metabolism , MicroRNAs/physiology , Neurogenesis/physiology , Neurons/physiology , Ribonuclease III/metabolism , Animals , Brain/physiology , Cell Differentiation/physiology , DEAD-box RNA Helicases/genetics , Humans , Neural Stem Cells/physiology , Ribonuclease III/genetics , Synapses/genetics , Synapses/metabolism
5.
Mol Neurobiol ; 54(9): 6750-6761, 2017 11.
Article in English | MEDLINE | ID: mdl-27752994

ABSTRACT

Despite years of research, most preclinical trials on ischemic stroke have remained unsuccessful owing to poor methodological and statistical standards leading to "translational roadblocks." Various behavioral tests have been established to evaluate traits such as sensorimotor function, cognitive and social interactions, and anxiety-like and depression-like behavior. A test's validity is of cardinal importance as it influences the chance of a successful translation of preclinical results to clinical settings. The mission of choosing a behavioral test for a particular project is, therefore, imperative and the present review aims to provide a structured way to evaluate rodent behavioral tests with implications in ischemic stroke.


Subject(s)
Brain Ischemia/diagnosis , Brain Ischemia/psychology , Motor Activity/physiology , Stroke/diagnosis , Stroke/psychology , Animals , Exploratory Behavior/physiology , Hand Strength/physiology , Mice , Rats , Rotarod Performance Test/methods , Rotarod Performance Test/psychology
6.
CNS Neurol Disord Drug Targets ; 15(10): 1299-1305, 2016.
Article in English | MEDLINE | ID: mdl-27549144

ABSTRACT

KCa3.1 protein is part of a heterotetrameric voltage-independent potassium channel, the activity of which depends on the intracellular calcium binding to calmodulin. KCa3.1 is immensely significant in regulating immune responses and primarily expressed in cells of hematopoietic lineage. It is one of the attractive pharmacological targets that are known to inhibit neuroinflammation. KCa3.1 blockers mediate neuroprotection through multiple mechanisms, such as by targeting microglia-mediated neuronal killing. KCa3.1 modulators may provide alternative treatment options for neurological disorders like ischemic stroke, Alzheimer disease, glioblastoma multiforme, multiple sclerosis and spinal cord injury. This review is an attempt to draw attention towards KCa3.1 channel, which was never exploited to its full potential as a viable therapeutic candidate against various neurological disorders.


Subject(s)
Central Nervous System Diseases/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Central Nervous System Diseases/pathology , Humans
8.
Curr Drug Targets ; 17(7): 800-10, 2016.
Article in English | MEDLINE | ID: mdl-25981603

ABSTRACT

MicroRNAs (miRNAs) are small (19-25 nucleotides) non-coding single-stranded RNAs that control post-transcriptional gene expression. miRNAs are abundantly expressed in the brain, where they play key roles during neuronal differentiation, synaptogenesis, and plasticity. It is also becoming increasingly evident that miRNAs are involved in the etiology of several neurological disorders. Mounting evidence indicates that miRNAs have the ability to regulate the expression profiles of genes in signaling pathways associated with cerebrovascular diseases such as ischemic stroke, subarachnoid hemorrhage, and vascular dementia. For instance, miR-21 is involved in ischemic stroke pathology through atherosclerosis and provides neuroprotection by its anti-apoptotic features. miR-497 induces neuronal death and miR-210 is upregulated in hypoxic cells. Deregulated expression of miRNAs in response to ischemic stroke has enabled the use of miRNA as an efficient non-invasive biomarker. Antagomirs are often effective against neuronal apoptosis and can induce neuroregeneration following ischemia. Moreover, the advent of systems biology has introduced novel computational tools to identify the link between miRNAs, target genes and transcription factors involved in the stroke pathology and its treatment. This review describes the emerging role of miRNAs in neuroprotection and focuses on a subset of miRNAs that act as central players in ischemic stroke.


Subject(s)
Antagomirs/therapeutic use , Cerebrovascular Disorders/genetics , MicroRNAs/genetics , Animals , Antagomirs/pharmacology , Apoptosis , Cerebrovascular Disorders/drug therapy , Computational Biology/methods , Female , Gene Expression Regulation , Gene Regulatory Networks , Genetic Markers/genetics , Humans , Male , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL