Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Curr Issues Mol Biol ; 35: 127-144, 2020.
Article in English | MEDLINE | ID: mdl-31422937

ABSTRACT

Brain ischaemia is a severe form of metabolic stress that activates a cascade of pathological events involving many signalling pathways. Modulation of these pathways is largely mediated by post-translational modifications (PTMs). Indeed, PTMs can rapidly modify pre-existing proteins by attaching chemical or polypeptide moieties to selected amino acid residues, altering their functions, stability, subcellular localizations, or interactions with other proteins. Subsequently, related signalling pathways can be substantially affected. Thus, PTMs are widely deployed by cells as an adaptive strategy at the front line to efficiently cope with internal and external stresses. Many types of PTMs have been identified, including phosphorylation, O-GlcNAcylation, small ubiquitin-like modifier (SUMO) modification (SUMOylation), and ubiquitination. All these PTMs have been studied in brain ischaemia to some extent. In particular, a large body of evidence has demonstrated that both global SUMOylation and ubiquitination are massively activated after brain ischaemia, and this activation may play a critical role in defining the fate and function of cells in the post-ischaemic brain. The goal of this review will be to summarize the current findings on SUMOylation and ubiquitination in brain ischaemia and discuss their clinical implications.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/metabolism , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Ubiquitination , Animals , Brain Ischemia/pathology , Brain Ischemia/therapy , Cell Line , Humans , Proteome/genetics , Proteome/metabolism , Signal Transduction/genetics
2.
FASEB J ; 32(3): 1677-1691, 2018 03.
Article in English | MEDLINE | ID: mdl-29146736

ABSTRACT

The development of novel neuroprotective treatments for acute stroke has been fraught with failures, which supports the view of ischemic brain damage as a highly complex multifactorial process. Post-translational modifications such as small ubiquitin-like modifier (SUMO)ylation have emerged as critical molecular regulatory mechanisms in states of both homeostasis and ischemic stress, as evidenced by our previous work. Accordingly, the clinical significance of the selective control of the global SUMOylation process has become apparent in studies of ischemic pathobiology and pathophysiology. Herein, we describe a process capable of identifying and characterizing small molecules with the potential of targeting the SUMO system through inhibition of SUMO deconjugation in an effort to develop novel stroke therapies.-Bernstock, J. D., Ye, D., Smith, J. A., Lee, Y.-J., Gessler, F. A., Yasgar, A., Kouznetsova, J., Jadhav, A., Wang, Z., Pluchino, S., Zheng, W., Simeonov, A., Hallenbeck, J. M., Yang, W. Quantitative high-throughput screening identifies cytoprotective molecules that enhance SUMO-conjugation via the inhibition of SUMO-specific protease (SENP)2.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cysteine Endopeptidases/metabolism , Protease Inhibitors/pharmacology , SUMO-1 Protein/metabolism , Sumoylation , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Transformed , Cysteine Endopeptidases/genetics , Humans , Rats , SUMO-1 Protein/genetics , Stroke/drug therapy , Stroke/genetics , Stroke/metabolism , Stroke/pathology
3.
Circ Res ; 121(12): 1360-1369, 2017 Dec 08.
Article in English | MEDLINE | ID: mdl-29051340

ABSTRACT

RATIONALE: Cryptogenic strokes, those of unknown cause, have been estimated as high as 30% to 40% of strokes. Inflammation has been suggested as a critical etiologic factor. However, there is lack of experimental evidence. OBJECTIVE: In this study, we investigated inflammation-associated stroke using a mouse model that developed spontaneous stroke because of myeloid deficiency of TGF-ß (transforming growth factor-ß) signaling. METHODS AND RESULTS: We report that mice with deletion of Tgfbr2 in myeloid cells (Tgfbr2Myeko) developed cerebrovascular inflammation in the absence of significant pathology in other tissues, culminating in stroke and severe neurological deficits with 100% penetrance. The stroke phenotype can be transferred to syngeneic wild-type mice via Tgfbr2Myeko bone marrow transplant and can be rescued in Tgfbr2Myeko mice with wild-type bone marrow. The underlying mechanisms involved an increased type 1 inflammation and cerebral endotheliopathy, characterized by elevated NF-κB (nuclear factor-κB) activation and TNF (tumor necrosis factor) production by myeloid cells. A high-fat diet accelerated stroke incidence. Anti-TNF treatment, as well as metformin and methotrexate, which are associated with decreased stroke risk in population studies, delayed stroke occurrence. CONCLUSIONS: Our studies show that TGF-ß signaling in myeloid cells is required for maintenance of vascular health and provide insight into inflammation-mediated cerebrovascular disease and stroke.


Subject(s)
Myeloid Cells/metabolism , Signal Transduction , Stroke/metabolism , Transforming Growth Factor beta/genetics , Animals , Cell Line , Immunosuppressive Agents/therapeutic use , Inflammation/complications , Inflammation/metabolism , Metformin/therapeutic use , Methotrexate/therapeutic use , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Penetrance , Stroke/etiology , Stroke/genetics , Stroke/prevention & control , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
Chromosome Res ; 26(4): 307-315, 2018 12.
Article in English | MEDLINE | ID: mdl-30443803

ABSTRACT

Сonstitutive heterochromatin areas are revealed by differential staining as C-positive chromosomal regions. These C-positive bands may greatly vary by location, size, and nucleotide composition. CBG-banding is the most commonly used method to detect structural heterochromatin in animals. The difficulty in identification of individual chromosomes represents an unresolved problem of this method as the body of the chromosome is stained uniformly and does not have banding pattern beyond C-bands. Here, we present the method that we called CDAG for sequential heterochromatin staining after differential GTG-banding. The method uses G-banding followed by heat denaturation in the presence of formamide with consecutive fluorochrome staining. The new technique is valid for the concurrent revealing of heterochromatin position due to differential banding of chromosomes and heterochromatin composition (AT-/GC-rich) in animal karyotyping.


Subject(s)
Chromosome Banding/methods , Heterochromatin/chemistry , Animals , Base Composition , Fluorescent Dyes , Formamides/pharmacology , Karyotyping , Nucleic Acid Denaturation , Staining and Labeling
5.
Stroke ; 49(3): 718-726, 2018 03.
Article in English | MEDLINE | ID: mdl-29440474

ABSTRACT

BACKGROUND AND PURPOSE: MAGL (monoacylglycerol lipase) is an enzyme that hydrolyzes the endocannabinoid 2-arachidonoylglycerol and regulates the production of arachidonic acid and prostaglandins-substances that mediate tissue inflammatory response. Here, we have studied the effects of the selective MAGL inhibitors JZL184 and MJN110 and their underlying molecular mechanisms on 3 different experimental models of focal cerebral ischemia. METHODS: SHR (spontaneously hypertensive rats) and normotensive WKY (Wistar Kyoto) rats were subject to an intracortical injection of the potent vasoconstrictor endothelin-1, permanent occlusion of a distal segment of the middle cerebral artery via craniectomy, or transient occlusion of the middle cerebral artery by the intraluminal suture method. JZL184 or MJN110 was administered 60 minutes after focal cerebral ischemia. Infarct volumes, hemispheric swelling, and functional outcomes were assessed between days 1 to 28 by magnetic resonance imaging, histology, and behavioral tests. RESULTS: Pharmacological inhibition of MAGL significantly attenuated infarct volume and hemispheric swelling. MAGL inhibition also ameliorated sensorimotor deficits, suppressed inflammatory response, and decreased the number of degenerating neurons. These beneficial effects of MAGL inhibition were not fully abrogated by selective antagonists of cannabinoid receptors, indicating that the anti-inflammatory effects are caused by inhibition of eicosanoid production rather than by activation of cannabinoid receptors. CONCLUSIONS: Our results suggest that MAGL may contribute to the pathophysiology of focal cerebral ischemia and is thus a promising therapeutic target for the treatment of ischemic stroke.


Subject(s)
Benzodioxoles/pharmacology , Brain Ischemia/drug therapy , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Monoacylglycerol Lipases/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Stroke/drug therapy , Succinimides/pharmacology , Animals , Brain Ischemia/enzymology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Disease Models, Animal , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/enzymology , Stroke/pathology , Stroke/physiopathology
6.
Lab Invest ; 98(6): 799-813, 2018 06.
Article in English | MEDLINE | ID: mdl-29472640

ABSTRACT

The intestinal epithelium constitutes a crucial defense to the potentially life-threatening effects of gut microbiota. However, due to a complex underlying vasculature, hypoperfusion and resultant tissue ischemia pose a particular risk to function and integrity of the epithelium. The small ubiquitin-like modifier (SUMO) conjugation pathway critically regulates adaptive responses to metabolic stress and is of particular significance in the gut, as inducible knockout of the SUMO-conjugating enzyme Ubc9 results in rapid intestinal epithelial disintegration. Here we analyzed the pattern of individual SUMO isoforms in intestinal epithelium and investigated their roles in intestinal ischemia/reperfusion (I/R) damage. Immunostaining revealed that epithelial SUMO2/3 expression was almost exclusively limited to crypt epithelial nuclei in unchallenged mice. However, intestinal I/R or overexpression of Ubc9 caused a remarkable enhancement of epithelial SUMO2/3 staining along the crypt-villus axis. Unexpectedly, a similar pattern was found in SUMO1 knockout mice. Ubc9 transgenic mice, but also SUMO1 knockout mice were protected from I/R injury as evidenced by better preserved barrier function and blunted inflammatory responses. PCR array analysis of microdissected villus-tip epithelia revealed a specific epithelial contribution to reduced inflammatory responses in Ubc9 transgenic mice, as key chemotactic signaling molecules such as IL17A were significantly downregulated. Together, our data indicate a critical role particularly of the SUMO2/3 isoforms in modulating responses to I/R and provide the first evidence that SUMO1 deletion activates a compensatory process that protects from ischemic damage.


Subject(s)
Intestinal Mucosa/blood supply , Reperfusion Injury/prevention & control , SUMO-1 Protein/physiology , Ubiquitin-Conjugating Enzymes/physiology , Animals , Chemokines/analysis , Intestinal Mucosa/chemistry , Laser Capture Microdissection , Mice , Mice, Inbred C57BL , Mice, Knockout , SUMO-1 Protein/deficiency , Small Ubiquitin-Related Modifier Proteins/analysis , Small Ubiquitin-Related Modifier Proteins/physiology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitins/analysis , Ubiquitins/physiology
7.
J Neurosci Res ; 96(4): 487-500, 2018 04.
Article in English | MEDLINE | ID: mdl-28463430

ABSTRACT

Traumatic brain injuries (TBIs) pose a massive burden of disease and continue to be a leading cause of morbidity and mortality throughout the world. A major obstacle in developing effective treatments is the lack of comprehensive understanding of the underlying mechanisms that mediate tissue damage and recovery after TBI. As such, our work aims to highlight the development of a novel experimental platform capable of fully characterizing the underlying pathobiology that unfolds after TBI. This platform encompasses an empirically optimized multiplex immunohistochemistry staining and imaging system customized to screen for a myriad of biomarkers required to comprehensively evaluate the extent of neuroinflammation, neural tissue damage, and repair in response to TBI. Herein, we demonstrate that our multiplex biomarker screening platform is capable of evaluating changes in both the topographical location and functional states of resident and infiltrating cell types that play a role in neuropathology after controlled cortical impact injury to the brain in male Sprague-Dawley rats. Our results demonstrate that our multiplex biomarker screening platform lays the groundwork for the comprehensive characterization of changes that occur within the brain after TBI. Such work may ultimately lead to the understanding of the governing pathobiology of TBI, thereby fostering the development of novel therapeutic interventions tailored to produce optimal tissue protection, repair, and/or regeneration with minimal side effects, and may ultimately find utility in a wide variety of other neurological injuries, diseases, and disorders that share components of TBI pathobiology.


Subject(s)
Brain Injuries, Traumatic/physiopathology , Immunohistochemistry/methods , Neuroimaging/methods , Animals , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers/metabolism , Brain/physiopathology , Brain Injuries, Traumatic/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Male , Oligodendroglia/metabolism , Oligodendroglia/pathology , Rats, Sprague-Dawley , Tongue Diseases/metabolism , Tongue Diseases/pathology
8.
J Neurochem ; 138(1): 101-16, 2016 07.
Article in English | MEDLINE | ID: mdl-27087120

ABSTRACT

The putative neuroprotective properties of various flavonoids have long been reported. Among this class of chemicals, quercetin, a major flavone/flavonol naturally occurring in plants, deserves focused attention because of the myriad of beneficial effects observed in various in vitro and in vivo models of central nervous system damage/degeneration. However, the mechanisms governing the beneficial outcomes mediated by quercetin remain to be elucidated. In an effort to define the underlying molecular mechanisms, our study employed human/rat neuroblastoma cell lines (SHSY5Y and B35, respectively) and E18-derived rat primary cortical neurons upon which the effects of various flavonoids were examined. Of note, increases in the levels of global SUMOylation, a post-translational modification with the Small Ubiquitin-like MOdifier (SUMO) were pronounced. Quercetin treatment increased SUMOylation levels in both SHSY5Y cells and rat cortical neurons in a dose and time-dependent manner, possibly via the direct inactivation of certain SENPs (SUMO-specific isopeptidases). Of particular interest, cells treated with quercetin displayed increased tolerance to oxygen/glucose deprivation exposure, an in vitro model of ischemia. SHSY5Y cells treated with quercetin also increased the expression of Nrf2 (via a decrease in the levels of Keap1), heme oxygenase-1 (HO-1), and nitric oxide synthase 1 (NOS1), which provide further protection from oxidative stress. In addition, the increased SUMOylation of HIF-1α was noted and deemed to be significant. We hypothesize that SUMOylated HIF-1α plays a fundamental role in the protection afforded and may underlie some of quercetin's ability to protect cells from oxygen/glucose deprivation-induced cell death, via an up-regulation of HO-1 and NOS1, which ultimately leads to the induction of pro-life NOS1/protein kinase G signaling. Quercetin acts to increase survival in the face of ischemia via an increase of SENP3 expression, the possible inactivation of SENPs 1/2, and via a decrease in KEAP1 levels (thereby increasing Nrf2 stability). These changes may then lead to increase in HIF-1α SUMOylation and HO-1 activation, followed by an up-regulation of NOS1/PKG signaling. Pathways altered via quercetin treatment within our experimental system are represented by blue arrowheads. Solid black arrows represent relationships that have been explored while a dotted arrow represents a relationship that has yet to be confirmed.


Subject(s)
Glucose/deficiency , Hypoxia/drug therapy , Neuroprotective Agents/pharmacology , Quercetin/pharmacology , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation/drug effects , Animals , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Female , Humans , L-Lactate Dehydrogenase/metabolism , Neuroblastoma/pathology , Neurons/drug effects , Pregnancy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
9.
Biochim Biophys Acta ; 1843(3): 640-51, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333653

ABSTRACT

Poly(ADP-ribose) polymerase-1 (PARP-1) and its cleavage products regulate cell viability and NF-kappaB activity when expressed in neurons. PARP-1 cleavage generates a 24 kDa (PARP-1(24)) and an 89 kDa fragment (PARP-1(89)). Compared to WT (PARP-1WT), the expression of an uncleavable PARP-1 (PARP-1(UNCL)) or of PARP-1(24) conferred protection from oxygen/glucose deprivation (OGD) or OGD/restoration of oxygen and glucose (ROG) damage in vitro, whereas expression of PARP-1(89) was cytotoxic. Viability experiments were performed in SH-SY5Y, a human neuroblastoma cell line, as well as in rat primary cortical neurons. Following OGD, the higher viability in the presence of PARP-1UNCL or PARP-1(24) was not accompanied with decreased formation of poly(ADP-riboses) or higher NAD levels. PARP-1 is a known cofactor for NF-kappaB, hence we investigated whether PARP-1 cleavage influences the inflammatory response. All PARP-1 constructs mimicked PARP-1WT in regard to induction of NF-kappaB translocation into the nucleus and its increased activation during ischemic challenge. However, expression of PARP-1(89) construct induced significantly higher NF-kB activity than PARP-1WT; and the same was true for NF-kappaB-dependent iNOS promoter binding activity. At a protein level, PARP-1UNCL and PARP-1(24) decreased iNOS (and lower levels of iNOS transcript) and COX-2, and increased Bcl-xL The increased levels of NF-kB and iNOS transcriptional activities, seen with cytotoxic PARP-189, were accompanied by higher protein expression of COX-2 and iNOS (and higher levels of INOS transcript) and lower protein expression of Bcl-xL Taken together, these findings suggest that PARP-1 cleavage products may regulate cellular viability and inflammatory responses in opposing ways during in vitro models of "ischemia".


Subject(s)
NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Animals , Caspases/metabolism , Cell Death/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Cyclooxygenase 2/metabolism , Humans , Neurons/metabolism , Nitric Oxide Synthase Type II/metabolism , Poly (ADP-Ribose) Polymerase-1 , Rats , Rats, Sprague-Dawley , Signal Transduction
10.
J Neurochem ; 135(5): 943-57, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26375300

ABSTRACT

Thirteen-lined ground squirrels (Ictidomys tridecemlineatus) have an extraordinary capacity to withstand prolonged and profound reductions in blood flow and oxygen delivery to the brain without incurring any cellular damage. As such, the hibernation torpor of I. tridecemlineatus provides a valuable model of tolerance to ischemic stress. Herein, we report that during hibernation torpor, a marked reduction in the phosphorylation of the ribosomal protein S6 (rpS6) occurs within the brains of I. tridecemlineatus. Of note, rpS6 phosphorylation was shown to increase in the brains of rats that underwent an occlusion of the middle cerebral artery. However, such an increase was attenuated after the implementation of an ischemic preconditioning paradigm. In addition, cultured cortical neurons treated with the rpS6 kinase (S6K) inhibitors, D-glucosamine or PF4708671, displayed a decrease in rpS6 phosphorylation and a subsequent increase in tolerance to oxygen/glucose deprivation, an in vitro model of ischemic stroke. Collectively, such evidence suggests that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning. Further identification and characterization of the mechanisms used by hibernating species to increase ischemic tolerance may eventually clarify how the loss of homeostatic control that occurs during and after cerebral ischemia in the clinic can ultimately be minimized and/or prevented. Mammalian hibernation provides a valuable model of tolerance to ischemic stress. Herein, we demonstrate that marked reductions in the phosphorylation of ribosomal protein S6 (rpS6), extracellular signal-regulated kinase family of mitogen-activated protein (MAP) kinase p44/42 (p44/42MAPK) and ribosomal protein S6 kinase (S6K) occur within the brains of both hibernating squirrels and rats, which have undergone an ischemic preconditioning paradigm. We therefore propose that the down-regulation of rpS6 signal transduction may account for a substantial part of the observed increase in cellular tolerance to brain ischemia that occurs during hibernation torpor and after ischemic preconditioning, via a suppression of protein synthesis and/or energy consumption.


Subject(s)
Gene Expression Regulation/physiology , Hibernation , Infarction, Middle Cerebral Artery/prevention & control , Ischemic Preconditioning , Ribosomal Protein S6/metabolism , Animals , Cells, Cultured , Cerebral Cortex , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/genetics , Glucose/deficiency , Male , Molecular Sequence Data , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6/genetics , Sciuridae , Signal Transduction/genetics , Signal Transduction/physiology , Time Factors
11.
bioRxiv ; 2023 Mar 30.
Article in English | MEDLINE | ID: mdl-37205496

ABSTRACT

Ischemic stroke results in a loss of tissue homeostasis and integrity, the underlying pathobiology of which stems primarily from the depletion of cellular energy stores and perturbation of available metabolites 1 . Hibernation in thirteen-lined ground squirrels (TLGS), Ictidomys tridecemlineatus , provides a natural model of ischemic tolerance as these mammals undergo prolonged periods of critically low cerebral blood flow without evidence of central nervous system (CNS) damage 2 . Studying the complex interplay of genes and metabolites that unfolds during hibernation may provide novel insights into key regulators of cellular homeostasis during brain ischemia. Herein, we interrogated the molecular profiles of TLGS brains at different time points within the hibernation cycle via RNA sequencing coupled with untargeted metabolomics. We demonstrate that hibernation in TLGS leads to major changes in the expression of genes involved in oxidative phosphorylation and this is correlated with an accumulation of the tricarboxylic acid (TCA) cycle intermediates citrate, cis-aconitate, and α-ketoglutarate-αKG. Integration of the gene expression and metabolomics datasets led to the identification of succinate dehydrogenase (SDH) as the critical enzyme during hibernation, uncovering a break in the TCA cycle at that level. Accordingly, the SDH inhibitor dimethyl malonate (DMM) was able to rescue the effects of hypoxia on human neuronal cells in vitro and in mice subjected to permanent ischemic stroke in vivo . Our findings indicate that studying the regulation of the controlled metabolic depression that occurs in hibernating mammals may lead to novel therapeutic approaches capable of increasing ischemic tolerance in the CNS.

12.
Stroke ; 43(2): 585-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22246691

ABSTRACT

After an unconventional beginning in stroke research, I veered off the main path repeatedly to view problems from a different perspective. In this lecture summary, I would like to return to several points along the byways that led to research with some continuity.


Subject(s)
Neurology/education , Animals , Brain Ischemia/blood , Brain Ischemia/pathology , Capillaries/pathology , Disease Models, Animal , Endothelium, Vascular/pathology , History, 20th Century , History, 21st Century , Humans , Immunity/physiology , Inflammation Mediators/physiology , Internship and Residency , Military Personnel , Neurology/history , Rats , Stroke
13.
J Neuroinflammation ; 9: 174, 2012 Jul 16.
Article in English | MEDLINE | ID: mdl-22799573

ABSTRACT

BACKGROUND: Deletion of some Toll-like receptors (TLRs) affords protection against cerebral ischemia, but disruption of their known major downstream adaptors does not. To determine whether compensation in the production of downstream effectors by one pathway when the other is disrupted can explain these findings, we examined cytokine/chemokine expression and inflammatory infiltrates in wild-type (WT), MyD88(-/-) and TRIF-mutant mice following permanent middle cerebral artery occlusion (pMCAO). METHODS: Cytokine/chemokine expression was measured with a 25-plex bead array in the serum and brains of all three groups of mice at baseline (no surgery/naïve) and at 3 hours and 24 hours following pMCAO. Brain inflammatory and neutrophil infiltrates were examined 24 hours following pMCAO. RESULTS: IL-6, keratinocyte chemoattractant (KC), granulocyte colony-stimulating factor (G-CSF) and IL-10 were significantly decreased in MyD88(-/-) mice compared to WT mice following pMCAO. Significantly, decreased levels of the neutrophil chemoattractants KC and G-CSF corresponded with a trend toward fewer neutrophils in the brains of MyD88(-/-) mice. IP-10 was significantly decreased when either pathway was disrupted. MIP-1 α was significantly decreased in TRIF-mutant mice, consistent with TRIF-dependent production. MyD88(-/-) mice showed elevations of a number of Th2 cytokines, such as IL-13, at baseline, which became significantly decreased following pMCAO. CONCLUSIONS: Both MyD88 and TRIF mediate pathway-specific cytokine production following focal cerebral ischemia. Our results also suggest a compensatory Th2-type skew at baseline in MyD88(-/-) mice and a paradoxical switch to a Th1 phenotype following focal cerebral ischemia. The MyD88 pathway directs the expression of neutrophil chemoattractants following cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Cytokines/biosynthesis , Gene Expression Regulation , Signal Transduction/genetics , Toll-Like Receptors/physiology , Animals , Brain Ischemia/pathology , Cytokines/genetics , Male , Mice , Myeloid Differentiation Factor 88/physiology , Random Allocation
14.
Stroke ; 41(3): e123-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20035078

ABSTRACT

BACKGROUND AND PURPOSE: Matrix metalloproteinases (MMP) may play a role in blood-brain barrier (BBB) disruption after ischemic stroke. We hypothesized that plasma concentrations of MMP-9 are associated with a marker of BBB disruption in patients evaluated for acute stroke. METHODS: Patients underwent MRI on presentation and approximately 24 hours later. The MRI marker, termed hyperintense acute reperfusion injury marker (HARM), is gadolinium enhancement of cerebrospinal fluid on fluid-attenuated inversion recovery MRI. Plasma MMP-9 and tissue inhibitor of matrix metalloproteinase-1 were measured by enzyme-linked immunosorbent assay. Logistic regression models tested for predictors of HARM on 24-hour follow-up scans separately for MMP-9 and the ratio of MMP-9 to TIMP-1. RESULTS: For the 41 patients enrolled, diagnoses were: acute ischemic cerebrovascular syndrome, 33 (80.6%); intracerebral hemorrhage, 6 (14.6%); stroke mimic, 1 (2.4%); and no stroke, 1 (2.4%). HARM was present in 17 (41.5%) patients. In model 1, HARM was associated with baseline plasma MMP-9 concentration (odds ratio [OR], 1.01; 95% confidence interval [CI], 1.001-1.019; P=0.033). In model 2, HARM was associated with the ratio of MMP-9 to tissue inhibitor of matrix metalloproteinase-1 (OR, 4.94; 95% CI, 1.27-19.14; P=0.021). CONCLUSIONS: Baseline MMP-9 was a significant predictor of HARM at 24-hour follow-up, supporting the hypothesis that MMP-9 is associated with BBB disruption. If the association between MMP-9 and BBB disruption is confirmed in future studies, HARM may be a useful imaging marker to evaluate MMP-9 inhibition in ischemic stroke and other populations with BBB disruption.


Subject(s)
Blood-Brain Barrier/enzymology , Blood-Brain Barrier/pathology , Cerebrovascular Disorders/blood , Cerebrovascular Disorders/enzymology , Matrix Metalloproteinase 9/blood , Adult , Aged , Aged, 80 and over , Biomarkers/blood , Cerebrovascular Disorders/physiopathology , Enzyme Activation/physiology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prospective Studies , Reperfusion Injury/blood , Reperfusion Injury/enzymology , Reperfusion Injury/physiopathology
15.
J Neurochem ; 109(1): 257-67, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19200349

ABSTRACT

Ground squirrels in hibernation torpor have been shown to have striking increases in global SUMOylation on tissue immunoblots. Here, we find evidence that global SUMOylation is also involved in ischemic tolerance in primary cortical neuronal cultures (from rats and mice) and SHSY5Y human neuroblastoma cells. Cultured cortical neurons preconditioned by sublethal oxygen/glucose deprivation (OGD) were less vulnerable to severe OGD than non-preconditioned neurons. Preconditioned neurons maintained elevated SUMO-1 conjugation levels (and, to a lesser extent those of SUMO-2/3) on western blots in contrast to non-preconditioned cells. Further, cortical neurons and SHSY5Y cells in which transfected SUMO-1 or SUMO-2 were over-expressed showed increased survival after severe OGD. In contrast, cell cultures subjected to depletion of endogenous SUMO-1 protein by RNAi had reduced survival after exposure to this form of in vitro ischemia and an attenuated protective response to preconditioning. These findings suggest that maintenance of a globally elevated SUMO-1 (and maybe SUMO-2/3) conjugation level as revealed by immunoblot assays is a component of ischemic tolerance.


Subject(s)
Brain Ischemia/etiology , Brain Ischemia/metabolism , SUMO-1 Protein/metabolism , Animals , Brain Ischemia/pathology , Cell Death/physiology , Cell Hypoxia/physiology , Cell Line, Tumor , Cells, Cultured , Humans , Ischemic Preconditioning , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley
16.
Front Mol Neurosci ; 12: 190, 2019.
Article in English | MEDLINE | ID: mdl-31507371

ABSTRACT

E-selectin plays an important role in mediating the rolling of leukocytes along and thus, the subsequent extravasation across activated endothelial cells comprising the microvasculature of the blood brain barrier (BBB). In multiple sclerosis (MS) and other inflammatory disorders of the central nervous system (CNS), the microvasculature is altered and immune cells infiltrate the brain and spinal cord contributing to damage, demyelination and ultimately disability. While mucosal administration is typically used to affect lymphocyte hyporesponsiveness or tolerance to suspect autoantigens, intranasal administration to E-selectin has previously been shown to protect against CNS inflammatory insults. We characterized the potential for mucosal administration of E-selectin to modulate CNS autoimmunity in the experimental autoimmune encephalomyelitis (EAE) model of MS. Intranasally administered E-selectin reduced swelling by as much as 50% in delayed-type hypersensitivity reactions compared to ovalbumin-tolerized controls. Intranasal E-selectin delivery prior to disease induction with myelin oligodendrocyte glycoprotein (MOG)35-55 reduced disease severity and total disease burden by more than 50% compared to PBS-tolerized animals; this protection was not associated with differences in the magnitude of the autoimmune response. Examination after the onset of disease showed that protection was associated with significant reductions in inflammatory infiltrates throughout the spinal cord. Tolerization to E-selectin did not influence encephalitogenic characteristics of autoreactive T cells such as IFN-gamma or IL-17 production. Clinical disease was also significantly reduced when E-selectin was first delivered after the onset of clinical symptoms. Splenic and lymph node (LN) populations from E-selectin-tolerized animals showed E-selectin-specific T cell responses and production of the immunomodulatory cytokine IL-10. Transfer of enriched CD4+ T cells from E-selectin tolerized mice limited disability in the passive SJL model of relapsing remitting MS. These results suggest a role for influencing E-selectin specific responses to limit neuroinflammation that warrants further exploration and characterization to better understand its potential to mitigate neurodegeneration in disorders such as MS.

17.
Ann Clin Transl Neurol ; 6(4): 817-820, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31020007

ABSTRACT

We aimed to characterize peripheral blood gene expression profile of penumbra defined as MRI perfusion-diffusion mismatch (PD MM) in peripheral blood of patients with acute ischemic stroke. We studied 23 patients. Perfusion-diffusion mismatch volume was observed to be associated and significantly correlated with the expression of 34 genes including those related to inflammation, SUMOylation, and coagulation; while lipopolysaccharide inhibition was identified to be a candidate upstream regulator of these processes (z-score -2.38, P = 0.04). Penumbral volume is correlated with a specific gene expression profile in the peripheral blood characterized by overlap of inflammatory and neuroprotective pathways that are regulated by lipopolysaccharide inhibition.


Subject(s)
Brain Ischemia/genetics , Brain/pathology , Cerebrovascular Circulation/physiology , Stroke/genetics , Aged , Aged, 80 and over , Brain/metabolism , Brain Ischemia/diagnosis , Diffusion Magnetic Resonance Imaging , Female , Humans , Male , Middle Aged , Research Design , Stroke/diagnosis
18.
EBioMedicine ; 42: 214-224, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30905846

ABSTRACT

BACKGROUND: Neural stem cell (NSC)-based therapies hold great promise for treating diseases of the central nervous system (CNS). However, several fundamental problems still need to be overcome to fully exploit the clinical potential of NSC therapeutics. Chief among them is the limited survival of NSC grafts within hostile microenvironments. METHODS: Herein, we sought to engineer NSCs in an effort to increase graft survival within ischemic brain lesions via upregulation of global SUMOylation, a post-translational modification critically involved in mediating tolerance to ischemia/reperfusion. FINDINGS: NSCs overexpressing the SUMO E2-conjugase Ubc9 displayed resistance to oxygen-glucose-deprivation/restoration of oxygen/glucose (OGD/ROG) and enhanced neuronal differentiation in vitro, as well as increased survival and neuronal differentiation when transplanted in mice with transient middle cerebral artery occlusion in vivo. INTERPRETATION: Our work highlights a critical role for SUMOylation in NSC biology and identifies a biological pathway that can be targeted to increase the effectiveness of exogenous stem cell medicines in ischemic stroke. FUND: Intramural Research Program of the NINDS/NIH, the Italian Multiple Sclerosis Foundation (FISM), the Bascule Charitable Trust, NIH-IRTA-OxCam and Wellcome Trust Research Training Fellowships.


Subject(s)
Cell Survival , Neural Stem Cells/metabolism , Stroke/metabolism , Animals , Biomarkers , Cell Cycle/genetics , Cell Survival/genetics , Computational Biology/methods , Energy Metabolism , Gene Expression , Gene Expression Profiling , Glucose/metabolism , Male , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis/genetics , Neurons/cytology , Neurons/metabolism , Oxygen/metabolism , Signal Transduction , Stem Cell Transplantation , Stroke/etiology , Sumoylation , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
19.
Stroke ; 39(12): 3405-10, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18757286

ABSTRACT

BACKGROUND AND PURPOSE: Symptomatic hemorrhagic transformation (HT) is the most important complicating factor after treatment with intravenous tissue plasminogen activator. In this study, we used multimodal magnetic resonance imaging to investigate the incidence and severity of reperfusion-based HT in spontaneously hypertensive rats after ischemia/reperfusion. METHODS: Twenty male spontaneously hypertensive rats were subjected to 30 minutes of middle cerebral artery occlusion via the suture model. Diffusion-weighted, T(2)-weighted, and gradient-echo imaging were performed on days 1, 2, 3, 4, and 7 for longitudinal evaluation of lesion evolution, vasogenic edema, and HT, respectively. Findings on gradient-echo images were classified according to the severity of hemorrhage: no HT; punctate or small petechial hemorrhage (HI-1); confluent petechial hemorrhage (HI-2); hematoma with absent/mild space-occupying effect (PH-1, 30% lesion volume). Histopathologic evaluation of HT was performed after final imaging for comparison with magnetic resonance imaging results. RESULTS: Final hemorrhage scores based on severity were as follows: HI-1 23.1%, HI-2 30.8%, PH-1 30.8%, and PH-2 15.4%. Similar to clinical observations, only PH-2 was associated with neurologic deterioration and associated weight loss. CONCLUSIONS: This model has a high incidence of parenchymal hematomas (46.2%) and therefore is appropriate for the evaluation of novel therapeutics targeting blood-brain barrier integrity and the reduction of symptomatic HT events (PH-2), as well as those potentially "at risk" for neurologic deterioration (PH-1).


Subject(s)
Cerebral Hemorrhage/pathology , Fibrinolytic Agents/toxicity , Hematoma/pathology , Infarction, Middle Cerebral Artery/drug therapy , Reperfusion Injury/pathology , Thrombolytic Therapy/adverse effects , Tissue Plasminogen Activator/toxicity , Animals , Blood-Brain Barrier/drug effects , Brain Edema/etiology , Cerebral Hemorrhage/chemically induced , Disease Progression , Fibrinolytic Agents/therapeutic use , Hematoma/etiology , Hypertension/complications , Hypertension/genetics , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Magnetic Resonance Imaging , Male , Movement Disorders/etiology , Rats , Rats, Inbred SHR , Recombinant Proteins/therapeutic use , Recombinant Proteins/toxicity , Time Factors , Tissue Plasminogen Activator/therapeutic use , Weight Loss
20.
J Cereb Blood Flow Metab ; 28(2): 341-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17637705

ABSTRACT

Vascular cognitive impairment (VCI) is the second most prevalent type of dementia in the world. The white matter damage that characterizes the common subcortical ischemic form of VCI can be modeled by ligating both common carotid arteries in the Wistar rat to induce protracted cerebral hypoperfusion. In this model, we find that repetitive intranasal administration of recombinant E-selectin to induce mucosal tolerance and to target immunomodulation to activating blood vessels potently suppresses both white matter (and possibly gray matter) damage and markers of vessel activation (tumor necrosis factor and E-selectin); it also preserves behavioral function in T-maze spontaneous alternation, T-maze spatial discrimination memory retention, and object recognition tests. Immunomodulation may be an effective novel strategy to prevent progression of VCI.


Subject(s)
Brain/pathology , Dementia, Vascular/prevention & control , E-Selectin/pharmacology , Immune Tolerance/physiology , Immunity, Mucosal/physiology , Memory Disorders/prevention & control , Animals , Dementia, Vascular/pathology , Dementia, Vascular/physiopathology , Discrimination, Psychological/drug effects , Female , Hypersensitivity, Delayed/physiopathology , Immune Tolerance/drug effects , Immunity, Mucosal/drug effects , Immunoassay , Immunologic Factors/pharmacology , Male , Maze Learning/drug effects , Memory Disorders/pathology , Memory Disorders/physiopathology , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL