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.
Brain ; 147(3): 1057-1074, 2024 03 01.
Article in English | MEDLINE | ID: mdl-38153327

ABSTRACT

Incomplete reperfusion of the microvasculature ('no-reflow') after ischaemic stroke damages salvageable brain tissue. Previous ex vivo studies suggest pericytes are vulnerable to ischaemia and may exacerbate no-reflow, but the viability of pericytes and their association with no-reflow remains under-explored in vivo. Using longitudinal in vivo two-photon single-cell imaging over 7 days, we showed that 87% of pericytes constrict during cerebral ischaemia and remain constricted post reperfusion, and 50% of the pericyte population are acutely damaged. Moreover, we revealed ischaemic pericytes to be fundamentally implicated in capillary no-reflow by limiting and arresting blood flow within the first 24 h post stroke. Despite sustaining acute membrane damage, we observed that over half of all cortical pericytes survived ischaemia and responded to vasoactive stimuli, upregulated unique transcriptomic profiles and replicated. Finally, we demonstrated the delayed recovery of capillary diameter by ischaemic pericytes after reperfusion predicted vessel reconstriction in the subacute phase of stroke. Cumulatively, these findings demonstrate that surviving cortical pericytes remain both viable and promising therapeutic targets to counteract no-reflow after ischaemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Humans , Pericytes/physiology , Cerebral Infarction
2.
J Neuroinflammation ; 20(1): 301, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102677

ABSTRACT

Ischemic stroke is a major global health issue and characterized by acute vascular dysfunction and subsequent neuroinflammation. However, the relationship between these processes remains elusive. In the current study, we investigated whether alleviating vascular dysfunction by restoring vascular nitric oxide (NO) reduces post-stroke inflammation. Mice were subjected to experimental stroke and received inhaled NO (iNO; 50 ppm) after reperfusion. iNO normalized vascular cyclic guanosine monophosphate (cGMP) levels, reduced the elevated expression of intercellular adhesion molecule-1 (ICAM-1), and returned leukocyte adhesion to baseline levels. Reduction of vascular pathology significantly reduced the inflammatory cytokines interleukin-1ß (Il-1ß), interleukin-6 (Il-6), and tumor necrosis factor-α (TNF-α), within the brain parenchyma. These findings suggest that vascular dysfunction is responsible for leukocyte adhesion and that these processes drive parenchymal inflammation. Reversing vascular dysfunction may therefore emerge as a novel approach to diminish neuroinflammation after ischemic stroke and possibly other ischemic disorders.


Subject(s)
Ischemic Stroke , Stroke , Mice , Animals , Nitric Oxide , Neuroinflammatory Diseases , Stroke/complications , Stroke/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Inflammation/drug therapy , Inflammation/pathology , Intercellular Adhesion Molecule-1/metabolism
3.
Adv Sci (Weinh) ; 9(4): e2103265, 2022 02.
Article in English | MEDLINE | ID: mdl-34904402

ABSTRACT

Stroke is the second leading cause of death and disability worldwide. Current treatments, such as pharmacological thrombolysis or mechanical thrombectomy, reopen occluded arteries but do not protect against ischemia-induced damage that occurs before reperfusion or neuronal damage induced by ischemia/reperfusion. It has been shown that disrupting the conversion of glyoxal to glycolic acid (GA) results in a decreased tolerance to anhydrobiosis in Caenorhabditis elegans dauer larva and that GA itself can rescue this phenotype. During the process of desiccation/rehydration, a metabolic stop/start similar to the one observed during ischemia/reperfusion occurs. In this study, the protective effect of GA is tested in different ischemia models, i.e., in commonly used stroke models in mice and swine. The results show that GA, given during reperfusion, strongly protects against ischemic damage and improves functional outcome. Evidence that GA exerts its effect by counteracting the glutamate-dependent increase in intracellular calcium during excitotoxicity is provided. These results suggest that GA treatment has the potential to reduce mortality and disability in stroke patients.


Subject(s)
Brain Ischemia/drug therapy , Calcium/metabolism , Glycolates/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Animals , Brain Ischemia/metabolism , Desiccation , Disease Models, Animal , Glycolates/administration & dosage , Glycolates/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotection/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Reperfusion Injury/metabolism , Swine
4.
J Neurotrauma ; 38(11): 1572-1584, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33779289

ABSTRACT

Traumatic brain injury (TBI) causes long-lasting neurodegeneration and cognitive impairments; however, the underlying mechanisms of these processes are not fully understood. Acid-sensing ion channels 1a (ASIC1a) are voltage-gated Na+- and Ca2+-channels shown to be involved in neuronal cell death; however, their role for chronic post-traumatic brain damage is largely unknown. To address this issue, we used ASIC1a-deficient mice and investigated their outcome up to 6 months after TBI. ASIC1a-deficient mice and their wild-type (WT) littermates were subjected to controlled cortical impact (CCI) or sham surgery. Brain water content was analyzed 24 h and behavioral outcome up to 6 months after CCI. Lesion volume was assessed longitudinally by magnetic resonance imaging and 6 months after injury by histology. Brain water content was significantly reduced in ASIC1a-/- animals compared to WT controls. Over time, ASIC1a-/- mice showed significantly reduced lesion volume and reduced hippocampal damage. This translated into improved cognitive function and reduced depression-like behavior. Microglial activation was significantly reduced in ASIC1a-/- mice. In conclusion, ASIC1a deficiency resulted in reduced edema formation acutely after TBI and less brain damage, functional impairments, and neuroinflammation up to 6 months after injury. Hence, ASIC1a seems to be involved in chronic neurodegeneration after TBI.


Subject(s)
Acid Sensing Ion Channels/deficiency , Brain Damage, Chronic/etiology , Brain Damage, Chronic/pathology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Animals , Brain Damage, Chronic/psychology , Brain Injuries, Traumatic/psychology , Disease Models, Animal , Male , Mice , Mice, Transgenic , Motor Activity
5.
J Neurochem ; 159(4): 710-728, 2021 11.
Article in English | MEDLINE | ID: mdl-33694332

ABSTRACT

Progressive neuronal injury following ischaemic stroke is associated with glutamate-induced depolarization, energetic stress and activation of AMP-activated protein kinase (AMPK). We here identify a molecular signature associated with neuronal AMPK activation, as a critical regulator of cellular response to energetic stress following ischaemia. We report a robust induction of microRNA miR-210-3p both in vitro in primary cortical neurons in response to acute AMPK activation and following ischaemic stroke in vivo. Bioinformatics and reverse phase protein array analysis of neuronal protein expression changes in vivo following administration of a miR-210-3p mimic revealed altered expression of phosphatase and tensin homolog (PTEN), 3-phosphoinositide-dependent protein kinase 1 (PDK1), ribosomal protein S6 kinase (p70S6K) and ribosomal protein S6 (RPS6) signalling in response to increasing miR-210-3p. In vivo, we observed a corresponding reduction in p70S6K activity following ischaemic stroke. Utilizing models of glutamate receptor over-activation in primary neurons, we demonstrated that induction of miR-210-3p was accompanied by sustained suppression of p70S6K activity and that this effect was reversed by miR-210-3p inhibition. Collectively, these results provide new molecular insight into the regulation of cell signalling during ischaemic injury, and suggest a novel mechanism whereby AMPK regulates miR-210-3p to control p70S6K activity in ischaemic stroke and excitotoxic injury.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ischemic Stroke/pathology , MicroRNAs/genetics , Neurons/pathology , Phosphatidylinositol 3-Kinases/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Animals , Cerebral Cortex/pathology , Computational Biology , Enzyme Activation , Female , Male , Mice, Inbred C57BL , PTEN Phosphohydrolase/metabolism , Polymerase Chain Reaction , Primary Cell Culture , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Ribosomal Protein S6/metabolism , Signal Transduction
6.
J Cereb Blood Flow Metab ; 37(6): 2084-2097, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27449604

ABSTRACT

Experimental stroke models producing clinically relevant functional deficits are often associated with high mortality. Because the mechanisms that underlie post-stroke mortality are largely unknown, results obtained using these models are often difficult to interpret, thereby limiting their translational potential. Given that specific forms of post-stroke care reduce mortality in patients, we hypothesized that inadequate food and water intake may underlie mortality following experimental stroke. C57BL/6 mice were subjected to 1 h of intraluminal filament middle cerebral artery occlusion. Nutritional support beginning on the second day after filament middle cerebral artery occlusion reduced the 14-day mortality rate from 59% to 15%. The surviving mice in the post-stroke support group had the same infarct size as non-surviving control mice, suggesting that post-stroke care was not neuroprotective and that inadequate food and/or water intake are the main reasons for filament middle cerebral artery occlusion-induced mortality. This notion was supported by the presence of significant hypoglycemia, ketonemia, and dehydration in control mice. Taken together, these data suggest that post-filament middle cerebral artery occlusion mortality in mice is not primarily caused by ischemic brain damage, but secondarily by inadequate food and/or water intake. Thus, providing nutritional support following filament middle cerebral artery occlusion greatly minimizes mortality bias and allows the study of long-term morphological and functional sequelae of stroke in mice.


Subject(s)
Drinking/physiology , Eating/physiology , Nutritional Support , Stroke/physiopathology , Stroke/therapy , Animals , Disease Models, Animal , Male , Mice, Inbred C57BL , Survival Analysis
7.
Sci Transl Med ; 7(299): 299ra121, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26246166

ABSTRACT

Numerous treatments have been reported to provide a beneficial outcome in experimental animal stroke models; however, these treatments (with the exception of tissue plasminogen activator) have failed in clinical trials. To improve the translation of treatment efficacy from bench to bedside, we have performed a preclinical randomized controlled multicenter trial (pRCT) to test a potential stroke therapy under circumstances closer to the design and rigor of a clinical randomized control trial. Anti-CD49d antibodies, which inhibit the migration of leukocytes into the brain, were previously investigated in experimental stroke models by individual laboratories. Despite the conflicting results from four positive and one inconclusive preclinical studies, a clinical trial was initiated. To confirm the preclinical results and to test the feasibility of conducting a pRCT, six independent European research centers investigated the efficacy of anti-CD49d antibodies in two distinct mouse models of stroke in a centrally coordinated, randomized, and blinded approach. The results pooled from all research centers revealed that treatment with CD49d-specific antibodies significantly reduced both leukocyte invasion and infarct volume after the permanent distal occlusion of the middle cerebral artery, which causes a small cortical infarction. In contrast, anti-CD49d treatment did not reduce lesion size or affect leukocyte invasion after transient proximal occlusion of the middle cerebral artery, which induces large lesions. These results suggest that the benefits of immune-targeted approaches may depend on infarct severity and localization. This study supports the feasibility of performing pRCTs.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Ischemia/drug therapy , Disease Models, Animal , Drug Evaluation, Preclinical , Integrin alpha4/immunology , Acute Disease , Animals , Brain Ischemia/immunology , Humans , Mice , Random Allocation , Treatment Outcome
8.
Circ Res ; 110(5): 727-38, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22207711

ABSTRACT

RATIONALE: Stroke is the third most common cause of death in industrialized countries. The main therapeutic target is the ischemic penumbra, potentially salvageable brain tissue that dies within the first few hours after blood flow cessation. Hence, strategies to keep the penumbra alive until reperfusion occurs are needed. OBJECTIVE: To study the effect of inhaled nitric oxide on cerebral vessels and cerebral perfusion under physiological conditions and in different models of cerebral ischemia. METHODS AND RESULTS: This experimental study demonstrates that inhaled nitric oxide (applied in 30% oxygen/70% air mixture) leads to the formation of nitric oxide carriers in blood that distribute throughout the body. This was ascertained by in vivo microscopy in adult mice. Although under normal conditions inhaled nitric oxide does not affect cerebral blood flow, after experimental cerebral ischemia induced by transient middle cerebral artery occlusion it selectively dilates arterioles in the ischemic penumbra, thereby increasing collateral blood flow and significantly reducing ischemic brain damage. This translates into significantly improved neurological outcome. These findings were validated in independent laboratories using two different mouse models of cerebral ischemia and in a clinically relevant large animal model of stroke. CONCLUSIONS: Inhaled nitric oxide thus may provide a completely novel strategy to improve penumbral blood flow and neuronal survival in stroke or other ischemic conditions.


Subject(s)
Arterioles/physiology , Brain Ischemia/prevention & control , Collateral Circulation/physiology , Nitric Oxide/therapeutic use , Stroke/prevention & control , Vasodilation/physiology , Administration, Inhalation , Animals , Arterioles/drug effects , Brain/blood supply , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cell Survival/drug effects , Cell Survival/physiology , Collateral Circulation/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neurons/pathology , Nitric Oxide/administration & dosage , Nitric Oxide/pharmacology , Regional Blood Flow/drug effects , Regional Blood Flow/physiology , Sheep , Stroke/pathology , Stroke/physiopathology , Vasodilation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...