Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Pharmaceuticals (Basel) ; 15(12)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36558997

ABSTRACT

Cancers and neurological disorders are two major types of diseases. We previously developed a new concept termed "Aberrant Cell Cycle Diseases" (ACCD), revealing that these two diseases share a common mechanism of aberrant cell cycle re-entry. The aberrant cell cycle re-entry is manifested as kinase/oncogene activation and tumor suppressor inactivation, which are hallmarks of both tumor growth in cancers and neuronal death in neurological disorders. Therefore, some cancer therapies (e.g., kinase inhibition, tumor suppressor elevation) can be leveraged for neurological treatments. The United States Food and Drug Administration (US FDA) has so far approved 74 kinase inhibitors, with numerous other kinase inhibitors in clinical trials, mostly for the treatment of cancers. In contrast, there are dire unmet needs of FDA-approved drugs for neurological treatments, such as Alzheimer's disease (AD), intracerebral hemorrhage (ICH), ischemic stroke (IS), traumatic brain injury (TBI), and others. In this review, we list these 74 FDA-approved kinase-targeted drugs and identify those that have been reported in preclinical and/or clinical trials for neurological disorders, with a purpose of discussing the feasibility and applicability of leveraging these cancer drugs (FDA-approved kinase inhibitors) for neurological treatments.

2.
Nat Prod Res ; 36(5): 1380-1384, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33459059

ABSTRACT

1,2,3,4,6-Penta-O-galloyl-ß-D-glucopyranose (ß-PGG) is a compound commonly available in vegetables and fruits. It exhibited potential inhibition of α-glucosidase and hypoglycemic effect in vivo. This study explored its dynamics properties inhibiting α-glucosidase by Lineweaver - Burk plots, spectral analysis, docking analysis, and molecular dynamics simulations. ß-PGG showed a mix-type inhibition when it was interacting with α-glucosidase. The fluorescence quenching indicated that the PGG-glucosidase complex formed in a spontaneous exothermic process and was driven by enthalpy. The synchronous fluorescence and ECD spectra indicate that ß-PGG induced and changed the enzyme conformation in the complex formation. Docking results revealed multiple hydrogen bonds between the phenols and the amino acid residues. Further dynamic simulations indicated that the residues Asp345, Phe153, Arg435, Glu300, Pro305, and Phe296 played a more critical role in the interactions between ß-PGG and α-glucosidase.


Subject(s)
Hydrolyzable Tannins , Polyphenols , alpha-Glucosidases , Diet , Hydrolyzable Tannins/pharmacology , Molecular Docking Simulation , alpha-Glucosidases/metabolism
3.
Pharmaceuticals (Basel) ; 14(7)2021 Jul 11.
Article in English | MEDLINE | ID: mdl-34358088

ABSTRACT

Insulin mimic can promote transporting glucose to muscle tissue and accelerate glucose consumption. It is commonly occurring in many functional foods or traditional medicines. Anti-diabetes molecules from food sources are highly safe and suitable for long-term use to prevent early diabetes. The leaves of Acer truncatum was found glucose uptake promotion in our phenotypic screening. However, its bioactive components and mechanism are still unclear. We collected leaves from trees of different ages (2, 3, 4, 7 and 11 years old) and profiled the ingredients by LC-MS/MS. The essential active component (myricitrin) was acquired following bio-guide on a whole organism Zebrafish (Danio rerio). Its content in the leaves was not affected by tree ages. Therefore, myricitrin can serve as a quality mark for functional foods derived from A. truncatum leaves. The transcriptomic and metabolomic analysis in Zebrafish explored the differentially expressed genes and metabolites. Based on joint-pathway enrichment and qRT-PCR verification, the critical bioactive component myricitrin was found to affect toll-like receptors signaling pathways to regulate glucose uptake. Our findings disclosed a bioactive marker (myricitrin) in A. truncatum leaves and explored its regulation mechanism, which rationalized the anti-diabetes function of the herbal food.

4.
J Neuroinflammation ; 16(1): 56, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30836997

ABSTRACT

BACKGROUND: Intracerebral hemorrhage (ICH) has a high morbidity and mortality. The peripheral immune system and cross-talk between peripheral blood and brain have been implicated in the ICH immune response. Thus, we delineated the gene networks associated with human ICH in the peripheral blood transcriptome. We also compared the differentially expressed genes in blood following ICH to a prior human study of perihematomal brain tissue. METHODS: We performed peripheral blood whole-transcriptome analysis of ICH and matched vascular risk factor control subjects (n = 66). Gene co-expression network analysis identified groups of co-expressed genes (modules) associated with ICH and their most interconnected genes (hubs). Mixed-effects regression identified differentially expressed genes in ICH compared to controls. RESULTS: Of seven ICH-associated modules, six were enriched with cell-specific genes: one neutrophil module, one neutrophil plus monocyte module, one T cell module, one Natural Killer cell module, and two erythroblast modules. The neutrophil/monocyte modules were enriched in inflammatory/immune pathways; the T cell module in T cell receptor signaling genes; and the Natural Killer cell module in genes regulating alternative splicing, epigenetic, and post-translational modifications. One erythroblast module was enriched in autophagy pathways implicated in experimental ICH, and NRF2 signaling implicated in hematoma clearance. Many hub genes or module members, such as IARS, mTOR, S1PR1, LCK, FYN, SKAP1, ITK, AMBRA1, NLRC4, IL6R, IL17RA, GAB2, MXD1, PIK3CD, NUMB, MAPK14, DDX24, EVL, TDP1, ATG3, WDFY3, GSK3B, STAT3, STX3, CSF3R, PIP4K2A, ANXA3, DGAT2, LRP10, FLOT2, ANK1, CR1, SLC4A1, and DYSF, have been implicated in neuroinflammation, cell death, transcriptional regulation, and some as experimental ICH therapeutic targets. Gene-level analysis revealed 1225 genes (FDR p < 0.05, fold-change > |1.2|) have altered expression in ICH in peripheral blood. There was significant overlap of the 1225 genes with dysregulated genes in human perihematomal brain tissue (p = 7 × 10-3). Overlapping genes were enriched for neutrophil-specific genes (p = 6.4 × 10-08) involved in interleukin, neuroinflammation, apoptosis, and PPAR signaling. CONCLUSIONS: This study delineates key processes underlying ICH pathophysiology, complements experimental ICH findings, and the hub genes significantly expand the list of novel ICH therapeutic targets. The overlap between blood and brain gene responses underscores the importance of examining blood-brain interactions in human ICH.


Subject(s)
Autophagy/physiology , Cerebral Hemorrhage , Cytokines/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks , Signal Transduction/physiology , Case-Control Studies , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Cytokines/genetics , Female , Gene Expression Profiling , Humans , Immune System , Male , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Transcriptome/physiology
5.
J Cereb Blood Flow Metab ; 39(9): 1818-1835, 2019 09.
Article in English | MEDLINE | ID: mdl-29651892

ABSTRACT

Understanding how the blood transcriptome of human intracerebral hemorrhage (ICH) differs from ischemic stroke (IS) and matched controls (CTRL) will improve understanding of immune and coagulation pathways in both disorders. This study examined RNA from 99 human whole-blood samples using GeneChip® HTA 2.0 arrays to assess differentially expressed transcripts of alternatively spliced genes between ICH, IS and CTRL. We used a mixed regression model with FDR-corrected p(Dx) < 0.2 and p < 0.005 and |FC| > 1.2 for individual comparisons. For time-dependent analyses, subjects were divided into four time-points: 0(CTRL), <24 h, 24-48 h, >48 h; 489 transcripts were differentially expressed between ICH and CTRL, and 63 between IS and CTRL. ICH had differentially expressed T-cell receptor and CD36 genes, and iNOS, TLR, macrophage, and T-helper pathways. IS had more non-coding RNA. ICH and IS both had angiogenesis, CTLA4 in T lymphocytes, CD28 in T helper cells, NFAT regulation of immune response, and glucocorticoid receptor signaling pathways. Self-organizing maps revealed 4357 transcripts changing expression over time in ICH, and 1136 in IS. Understanding ICH and IS transcriptomes will be useful for biomarker development, treatment and prevention strategies, and for evaluating how well animal models recapitulate human ICH and IS.


Subject(s)
Brain Ischemia/genetics , Cerebral Hemorrhage/genetics , Stroke/genetics , Transcriptome , Aged , Alternative Splicing , Brain Ischemia/blood , Cerebral Hemorrhage/blood , Female , Humans , Male , Middle Aged , Stroke/blood
6.
Front Neurosci ; 12: 767, 2018.
Article in English | MEDLINE | ID: mdl-30405345

ABSTRACT

Aim: Our previous study demonstrated miR-122 mimic decreased NOS2 expression in blood leucocytes and improved stroke outcomes when given immediately after middle cerebral artery occlusion (MCAO) in rats. Since NOS2 is associated with neuro-inflammation in stroke and decreasing NOS2 expression alone in leucocytes is insufficient to improve stroke outcomes, we hypothesized that miR-122 mimic may also decrease NOS2 expression in brain microvascular endothelial cells (BMVECs) even at extended time windows. Methods: We administered PEG-liposome wrapped miR-122 mimic (2.4 mg/kg, i.v.) 0 or 6 h after MCAO, and assessed stroke volume and NOS2 expression in BMVECs 24 h following MCAO in rats. Luciferase reporter assays were used to determine if miR-122 binds to 3' untranslated regions (3'UTR) of NOS2. Results: The data showed that miR-122 mimic decreased infarct volumes and decreased MCAO-induced NOS2 over-expression in BMVECs. However, miR-122 did not bind to 3'UTR of NOS2 in the luciferase assays. Conclusion: The data show the 6-h period of therapeutic efficacy of miR-122 mimic which could relate to indirect knockdown of NOS2 in both BMVECs and leucocytes.

7.
J Cereb Blood Flow Metab ; 38(7): 1125-1148, 2018 07.
Article in English | MEDLINE | ID: mdl-29708005

ABSTRACT

Central nervous system (CNS) injuries, such as stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), are important causes of death and long-term disability worldwide. MicroRNA (miRNA), small non-coding RNA molecules that negatively regulate gene expression, can serve as diagnostic biomarkers and are emerging as novel therapeutic targets for CNS injuries. MiRNA-based therapeutics include miRNA mimics and inhibitors (antagomiRs) to respectively decrease and increase the expression of target genes. In this review, we summarize current miRNA-based therapeutic applications in stroke, TBI and SCI. Administration methods, time windows and dosage for effective delivery of miRNA-based drugs into CNS are discussed. The underlying mechanisms of miRNA-based therapeutics are reviewed including oxidative stress, inflammation, apoptosis, blood-brain barrier protection, angiogenesis and neurogenesis. Pharmacological agents that protect against CNS injuries by targeting specific miRNAs are presented along with the challenges and therapeutic potential of miRNA-based therapies.


Subject(s)
Brain Injuries, Traumatic/therapy , Drug Delivery Systems/methods , MicroRNAs/therapeutic use , Spinal Cord Injuries/therapy , Stroke/therapy , Animals , Apoptosis/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/therapy , Oxidative Stress/drug effects , Spinal Cord Injuries/metabolism , Stroke/metabolism , Stroke/pathology
8.
9.
J Cereb Blood Flow Metab ; 37(7): 2359-2367, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27624844

ABSTRACT

Intraventricular hemorrhage causes spatial memory loss, but the mechanism remains unknown. Our recent studies demonstrated that traumatic brain injury activates Src family kinases, which cause spatial memory loss. To test whether the spatial memory loss was due to blood in the ventricles, which activated Src family kinases, we infused autologous whole blood or thrombin into the lateral ventricles of adult rats to model non-traumatic intraventricular hemorrhage. Hippocampal neuron loss was examined 1 day to 5 weeks later. Spatial memory function was assessed 29 to 33 days later using the Morris water maze. Five weeks after the ventricular injections of blood or thrombin, there was death of most hippocampal neurons and significant memory deficits compared with sham operated controls. These data show that intraventricular thrombin is sufficient to kill hippocampal neurons and produce spatial memory loss. In addition, systemic administration of the non-specific Src family kinase inhibitor PP2 or intraventricular injection of siRNA-Fyn, a Src family kinase family member, prevented hippocampal neuronal loss and spatial memory deficits following intraventricular hemorrhage. The data support the conclusions that thrombin mediates the hippocampal neuronal cell death and spatial memory deficits produced by intraventricular blood and that these can be blocked by non-specific inhibition of Src family kinases or by inhibiting Fyn.


Subject(s)
Cerebral Ventricles/blood supply , Cognitive Dysfunction/enzymology , Intracranial Hemorrhages/drug therapy , Thrombin/metabolism , src-Family Kinases/antagonists & inhibitors , Animals , Cerebral Ventricles/enzymology , Cognitive Dysfunction/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Hippocampus/enzymology , Hippocampus/pathology , Injections, Intraventricular , Intracranial Hemorrhages/enzymology , Intracranial Hemorrhages/pathology , Intracranial Hemorrhages/psychology , Male , Maze Learning/drug effects , Neurons/enzymology , Neurons/pathology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Spatial Memory/drug effects , Thrombin/administration & dosage , src-Family Kinases/genetics
10.
J Cereb Blood Flow Metab ; 36(8): 1374-83, 2016 08.
Article in English | MEDLINE | ID: mdl-26661204

ABSTRACT

Because our recent studies have demonstrated that miR-122 decreased in whole blood of patients and in whole blood of rats following ischemic stroke, we tested whether elevating blood miR-122 would improve stroke outcomes in rats. Young adult rats were subjected to a temporary middle cerebral artery occlusion (MCAO) or sham operation. A polyethylene glycol-liposome-based transfection system was used to administer a miR-122 mimic after MCAO. Neurological deficits, brain infarction, brain vessel integrity, adhesion molecule expression and expression of miR-122 target and indirect-target genes were examined in blood at 24 h after MCAO with or without miR-122 treatment. miR-122 decreased in blood after MCAO, whereas miR-122 mimic elevated miR-122 in blood 24 h after MCAO. Intravenous but not intracerebroventricular injection of miR-122 mimic decreased neurological deficits and brain infarction, attenuated ICAM-1 expression, and maintained vessel integrity after MCAO. The miR-122 mimic also down-regulated direct target genes (e.g. Vcam1, Nos2, Pla2g2a) and indirect target genes (e.g. Alox5, Itga2b, Timp3, Il1b, Il2, Mmp8) in blood after MCAO which are predicted to affect cell adhesion, diapedesis, leukocyte extravasation, eicosanoid and atherosclerosis signaling. The data show that elevating miR-122 improves stroke outcomes and we postulate this occurs via downregulating miR-122 target genes in blood leukocytes.


Subject(s)
Infarction, Middle Cerebral Artery/blood , MicroRNAs/blood , MicroRNAs/genetics , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Drug Delivery Systems , Infarction, Middle Cerebral Artery/genetics , Injections, Intravenous , Leukocytes/metabolism , Liposomes , Male , MicroRNAs/administration & dosage , Polyethylene Glycols/chemistry , Rats, Sprague-Dawley , Treatment Outcome
11.
J Neuromuscul Dis ; 2(4): 387-396, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-27858746

ABSTRACT

BACKGROUND: Duchenne Muscular Dystrophy (DMD) is a recessive X-linked form of muscular dystrophy. Steroid therapy has clinical benefits for DMD patients, but the mechanism remains unclear. OBJECTIVE: This study was designed to identify mRNAs and microRNAs regulated in Duchenne Muscular Dystrophy patients prior to and after steroid therapy. METHODS: Genome wide transcriptome profiling of whole blood was performed to identify mRNAs and microRNAs regulated in DMD patients. RESULTS: The data show many regulated mRNAs and some microRNAs, including some muscle-specific microRNAs (e.g., miR-206), that were significantly altered in blood of young (age 3-10) DMD patients compared to young controls. A total of 95 microRNAs, but no mRNAs, were differentially expressed in older DMD patients compared to matched controls (age 11-20). Steroid treatment reversed expression patterns of several microRNAs (miR-206, miR-181a, miR-4538, miR-4539, miR-606, and miR-454) that were altered in the young DMD patients. As an example, the over-expression of miR-206 in young DMD patients is predicted to down-regulate a set of target genes (e.g., RHGAP31, KHSRP, CORO1B, PTBP1, C7orf58, DLG4, and KLF4) that would worsen motor function. Since steroids decreased miR-206 expression to control levels, this could provide one mechanism by which steroids improve motor function. CONCLUSIONS: These identified microRNA-mRNA alterations will help better understand the pathophysiology of DMD and the response to steroid treatment.

12.
J Neurotrauma ; 31(14): 1268-76, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24428562

ABSTRACT

Traumatic brain injury (TBI) is often associated with intracerebral and intraventricular hemorrhage. Thrombin is a neurotoxin generated at bleeding sites fater TBI and can lead to cell death and subsequent cognitive dysfunction via activation of Src family kinases (SFKs). We hypothesize that inhibiting SFKs can protect hippocampal neurons and improve cognitive memory function after TBI. To test these hypotheses, we show that moderate lateral fluid percussion (LFP) TBI in adult rats produces bleeding into the cerebrospinal fluid (CSF) in both lateral ventricles, which elevates oxyhemoglobin and thrombin levels in the CSF, activates the SFK family member Fyn, and increases Rho-kinase 1(ROCK1) expression. Systemic administration of the SFK inhibitor, PP2, immediately after moderate TBI blocks ROCK1 expression, protects hippocampal CA2/3 neurons, and improves spatial memory function. These data suggest the possibility that inhibiting SFKs after TBI might improve clinical outcomes.


Subject(s)
Brain Injuries/enzymology , Hippocampus/enzymology , Neurons/enzymology , src-Family Kinases/metabolism , Animals , Blotting, Western , Brain Injuries/pathology , Cognition , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hippocampus/pathology , Immunohistochemistry , Male , Maze Learning/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley
13.
Transl Stroke Res ; 4(6): 685-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24323422

ABSTRACT

Heat shock proteins (Hsps) are induced by heat shock via heat shock factor proteins binding to heat shock elements in their promoters. Hsp70 is massively induced in response to misfolded proteins following cerebral ischemia in all cell types but is induced mainly in neurons in the ischemic penumbra. Overexpression of Hsp70 via transgenes and viruses or systemic administration of Hsp70 fusion proteins that allow it to cross the blood brain barrier protects the brain against ischemia in most reported studies. Hsp27 can exist as unphosphorylated large oligomers that prevent misfolded protein aggregates and improve cell survival. P-Hsp27 small oligomers bind specific protein targets to improve survival. In the brain, protein kinase D phosphorylates Hsp27 following ischemia which then binds apoptosis signal-regulating kinase 1 to prevent MKK4/7, c-Jun NH(2)-terminal kinase, and Jun-induced apoptosis, and decrease infarct volumes following focal cerebral ischemia. Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. CO activates cGMP to promote vasodilation, and biliverdin is converted to bilirubin which can serve as an anti-oxidant, both of which may contribute to the reported protective role of HO-1 in cerebral ischemia and subarachnoid hemorrhage. However, ferrous ion can react with hydrogen peroxide to produce pro-oxidant hydroxyl radicals which may explain the harmful role of HO-1 in intracerebral hemorrhage. Heat shock proteins as a class have great potential as treatments for cerebrovascular disease and have yet to be tested in the clinic.


Subject(s)
Brain Ischemia/metabolism , Cerebrum/metabolism , Heat-Shock Proteins/metabolism , Neurons/metabolism , Apoptosis , Biliverdine/metabolism , Carbon Monoxide/metabolism , HSP27 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heme Oxygenase-1/metabolism , Humans , Protein Folding , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Subarachnoid Hemorrhage/metabolism
14.
ScientificWorldJournal ; 2012: 491737, 2012.
Article in English | MEDLINE | ID: mdl-22547985

ABSTRACT

Since publishing our earlier report describing a strategy for the treatment of central nervous system (CNS) diseases by inhibiting the cell cycle and without disrupting neurogenesis (Liu et al. 2010), we now update and extend this strategy to applications in the treatment of cancers as well. Here, we put forth the concept of "aberrant cell cycle diseases" to include both cancer and CNS diseases, the two unrelated disease types on the surface, by focusing on a common mechanism in each aberrant cell cycle reentry. In this paper, we also summarize the pharmacological approaches that interfere with classical cell cycle molecules and mitogenic pathways to block the cell cycle of tumor cells (in treatment of cancer) as well as to block the cell cycle of neurons (in treatment of CNS diseases). Since cell cycle inhibition can also block proliferation of neural progenitor cells (NPCs) and thus impair brain neurogenesis leading to cognitive deficits, we propose that future strategies aimed at cell cycle inhibition in treatment of aberrant cell cycle diseases (i.e., cancers or CNS diseases) should be designed with consideration of the important side effects on normal neurogenesis and cognition.


Subject(s)
Cell Cycle , Central Nervous System Diseases/therapy , Neurogenesis , Cognition/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Neoplasms/therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use
15.
Gene ; 495(2): 120-7, 2012 Mar 10.
Article in English | MEDLINE | ID: mdl-22244746

ABSTRACT

Mature neurons (MNs), neural progenitor cells (NPCs) and neuroblastoma cells (NBCs) are all neural-derived cells. However, MNs are unable to divide once differentiated; NPCs are able to divide a limited number of times and differentiate to normal brain cell types; whereas NBCs can divide an unlimited number of times but rarely differentiate. Here, we perform whole transcriptome (mRNA, miRNA) profiling of these cell types and compare expression levels of each cell type to the others. Integrated mRNA-miRNA functional analyses reveal that: 1) several very highly expressed genes (e.g., Robo1, Nrp1, Epha3, Unc5c, Dcc, Pak3, Limk4) and a few under-expressed miRNAs (e.g., miR-152, miR-146b, miR-339-5p) in MNs are associated with one important cellular process-axon guidance; 2) some very highly expressed mitogenic pathway genes (e.g., Map2k1, Igf1r, Rara, Runx1) and under-expressed miRNAs (e.g., miR-370, miR-9, miR-672) in NBCs are associated with cancer pathways. These results provide a library of negative mRNAmiRNA networks that are likely involved in the cellular processes of differentiation and division.


Subject(s)
Gene Expression Regulation , MicroRNAs/biosynthesis , Neural Stem Cells/metabolism , Neuroblastoma/genetics , Neurons/metabolism , RNA, Messenger/biosynthesis , Animals , Gene Expression Profiling , Nerve Tissue Proteins/genetics , Neuroblastoma/pathology , Rats
16.
Transl Stroke Res ; 3(Suppl 1): 62-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-24323862

ABSTRACT

Intracerebral hemorrhage (ICH) results in the release of a large number of endogenous molecules, including glutamate, Ca(2+), ROS, thrombin, heme, iron, TNF-α, and others. These molecules participate in excitatory and mitogenic signaling transduction in which N-methyl-D-aspartate (NMDA) receptors and Src family kinases (SFKs) are implicated. Mitogenic signaling initiates the cell cycle for normal cell division of microglia and neural progenitor cells, whereas aberrant mitogenic signaling causes toxicity, killing neurons, astrocytes, and brain microvascular endothelial cells in neurological diseases including ICH. In this review, we summarize (1) how SFKs modulate NMDA receptors to kill neurons following ICH and (2) how SFKs modulate mitogenic signaling transduction to kill neurons and play a role in disrupting the blood-brain barrier (BBB) immediately following ICH and in repairing the BBB during the recovery phases weeks following ICH.

17.
Acta Neurochir Suppl ; 111: 77-81, 2011.
Article in English | MEDLINE | ID: mdl-21725735

ABSTRACT

Src kinase signaling has been implicated in multiple mechanisms of intracerebral hemorrhage (ICH). These include (1) thrombin-mediated mitogenic stress, (2) excitatory amino acid (AA)-mediated excitatory toxicity, (3) vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs)-mediated changes of vascular permeability, (4) cytokines-mediated inflammatory responses, and (5) others. These work together after ICH, causing brain injuries in the acute stage and self-repair in the recovery stage. We found that acute administration of the Src inhibitor, PP2, blocks the blood-brain barrier (BBB) breakdown and brain edema that occurs after ICH. However, delayed and chronic administration of PP2 prevents the BBB repair and edema resolution after ICH. These results led us to suggest that the two contradictory findings share the same principles at least in part via activation of Src kinases in acute or recovery stages after ICH. Acute Src kinase activation after ICH leads to BBB damage, and chronic Src kinase activation after ICH leads to BBB repair.


Subject(s)
Cerebral Hemorrhage/enzymology , src-Family Kinases/metabolism , Animals , Cell Cycle/physiology , Cell Proliferation , Cerebral Hemorrhage/physiopathology , Enzyme Activation , Humans , Models, Biological , Neural Stem Cells/physiology
18.
Ann Neurol ; 67(4): 526-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20437588

ABSTRACT

OBJECTIVE: Thrombin mediates the life-threatening cerebral edema that occurs after intracerebral hemorrhage. Therefore, we examined the mechanisms of thrombin-induced injury to the blood-brain barrier (BBB) and subsequent mechanisms of BBB repair. METHODS: Intracerebroventricular injection of thrombin (20U) was used to model intraventricular hemorrhage in adult rats. RESULTS: Thrombin reduced brain microvascular endothelial cell (BMVEC) and perivascular astrocyte immunoreactivity-indicating either cell injury or death-and functionally disrupted the BBB as measured by increased water content and extravasation of sodium fluorescein and Evans blue dyes 24 hours later. Administration of nonspecific Src family kinase inhibitor (PP2) immediately after thrombin injections blocked brain edema and BBB disruption. At 7 to 14 days after thrombin injections, newborn endothelial cells and astrocytes were observed around cerebral vessels at the time when BBB permeability and cerebral water content resolved. Delayed administration of PP2 on days 2 through 6 after thrombin injections prevented resolution of the edema and abnormal BBB permeability. INTERPRETATION: Thrombin, via its protease-activated receptors, is postulated to activate Src kinase phosphorylation of molecules that acutely injure the BBB and produce edema. Thus, acute administration of Src antagonists blocks edema. In contrast, Src blockade for 2 to 6 days after thrombin injections is postulated to prevent resolution of edema and abnormal BBB permeability in part because Src kinase proto-oncogene members stimulate proliferation of newborn BMVECs and perivascular astrocytes in the neurovascular niche that repair the damaged BBB. Thus, Src kinases not only mediate acute BBB injury but also mediate chronic BBB repair after thrombin-induced injury.


Subject(s)
Blood-Brain Barrier , Brain Edema/prevention & control , Hemostatics/adverse effects , Pyrimidines/therapeutic use , Thrombin/adverse effects , src-Family Kinases/antagonists & inhibitors , Animals , Antigens, Surface/metabolism , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/injuries , Blood-Brain Barrier/physiopathology , Brain Edema/chemically induced , Brain Edema/pathology , Bromodeoxyuridine/metabolism , Capillary Permeability/drug effects , Cell Proliferation/drug effects , Evans Blue , Fluorescein , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Injections, Intraventricular/methods , Male , Rats , Rats, Sprague-Dawley , Time Factors , Water/metabolism
19.
Neurobiol Dis ; 37(3): 549-57, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19944161

ABSTRACT

Classically, the cell cycle is regarded as the process leading to cellular proliferation. However, increasing evidence over the last decade supports the notion that neuronal cell cycle re-entry results in post-mitotic death. A mature neuron that re-enters the cell cycle can neither advance to a new G0 quiescent state nor revert to its earlier G0 state. This presents a critical dilemma to the neuron from which death may be an unavoidable but necessary outcome for adult neurons attempting to complete the cell cycle. In contrast, tumor cells that undergo aberrant cell cycle re-entry divide and can survive. Thus, cell cycle inhibition strategies are of interest in cancer treatment but may also represent an important means of protecting neurons. In this review, we put forth the concept of the "expanded cell cycle" and summarize the cell cycle proteins, signal transduction events and mitogenic molecules that can drive a neuron into the cell cycle in various CNS diseases. We also discuss the pharmacological approaches that interfere with the mitogenic pathways and prevent mature neurons from attempting cell cycle re-entry, protecting them from cell death. Lastly, future attempts at blocking the cell cycle to rescue mature neurons from injury should be designed so as to not block normal neurogenesis.


Subject(s)
Brain Diseases/physiopathology , Cell Cycle/physiology , Nerve Degeneration/physiopathology , Brain Diseases/drug therapy , Brain Diseases/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Death/physiology , Drug Evaluation, Preclinical/methods , Genes, cdc/drug effects , Genes, cdc/physiology , Humans , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Signal Transduction/drug effects , Signal Transduction/physiology
20.
J Cereb Blood Flow Metab ; 30(1): 92-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19724284

ABSTRACT

MicroRNAs (miRNAs) regulate gene expression and have a critical role in many biologic and pathologic processes. We hypothesized that miRNA expression profiles in injured brain (hippocampus) would show common as well as unique profiles when compared with those of blood. Adult, untouched, control rats were compared with rats with sham surgeries, ischemic strokes, brain hemorrhage (lysed blood, fresh blood, or thrombin), and kainate-induced seizures. Brain and whole-blood miRNA expression profiles were assessed 24 h later using TaqMan rodent miRNA arrays. MicroRNA response profiles were different for each condition. Many miRNAs changed more than 1.5-fold in brain and blood after each experimental manipulation, and several miRNAs were upregulated or downregulated in both brain and blood after a given injury. A few miRNAs (e.g., miR-298, miR-155, and miR-362-3p) were upregulated or downregulated more than twofold in both brain and blood after several different injuries. The results show the possible use of blood miRNAs as biomarkers for brain injury; that selected blood miRNAs may correlate with miRNA changes in the brain; and that many of the mRNAs, previously shown to be regulated in brain and blood after brain injury, are likely accounted for by changes in miRNA expression.


Subject(s)
Brain Chemistry/physiology , Brain Ischemia/metabolism , Cerebral Hemorrhage/metabolism , Excitatory Amino Acid Antagonists , Kainic Acid , MicroRNAs/biosynthesis , Seizures/metabolism , Stroke/metabolism , Animals , Male , MicroRNAs/blood , Rats , Rats, Sprague-Dawley , Seizures/chemically induced , Seizures/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...