Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 184
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-39043180

RESUMEN

The medical burden of stroke extends beyond the brain injury itself and is largely determined by chronic comorbidities that develop secondarily. We hypothesized that these comorbidities might share a common immunological cause, yet chronic effects post-stroke on systemic immunity are underexplored. Here, we identify myeloid innate immune memory as a cause of remote organ dysfunction after stroke. Single-cell sequencing revealed persistent pro-inflammatory changes in monocytes/macrophages in multiple organs up to 3 months after brain injury, notably in the heart, leading to cardiac fibrosis and dysfunction in both mice and stroke patients. IL-1ß was identified as a key driver of epigenetic changes in innate immune memory. These changes could be transplanted to naive mice, inducing cardiac dysfunction. By neutralizing post-stroke IL-1ß or blocking pro-inflammatory monocyte trafficking with a CCR2/5 inhibitor, we prevented post-stroke cardiac dysfunction. Such immune-targeted therapies could potentially prevent various IL-1ß-mediated comorbidities, offering a framework for secondary prevention immunotherapy.

2.
Lancet ; 403(10442): 2395-2404, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38761811

RESUMEN

BACKGROUND: It is unknown whether decompressive craniectomy improves clinical outcome for people with spontaneous severe deep intracerebral haemorrhage. The SWITCH trial aimed to assess whether decompressive craniectomy plus best medical treatment in these patients improves outcome at 6 months compared to best medical treatment alone. METHODS: In this multicentre, randomised, open-label, assessor-blinded trial conducted in 42 stroke centres in Austria, Belgium, Finland, France, Germany, the Netherlands, Spain, Sweden, and Switzerland, adults (18-75 years) with a severe intracerebral haemorrhage involving the basal ganglia or thalamus were randomly assigned to receive either decompressive craniectomy plus best medical treatment or best medical treatment alone. The primary outcome was a score of 5-6 on the modified Rankin Scale (mRS) at 180 days, analysed in the intention-to-treat population. This trial is registered with ClincalTrials.gov, NCT02258919, and is completed. FINDINGS: SWITCH had to be stopped early due to lack of funding. Between Oct 6, 2014, and April 4, 2023, 201 individuals were randomly assigned and 197 gave delayed informed consent (96 decompressive craniectomy plus best medical treatment, 101 best medical treatment). 63 (32%) were women and 134 (68%) men, the median age was 61 years (IQR 51-68), and the median haematoma volume 57 mL (IQR 44-74). 42 (44%) of 95 participants assigned to decompressive craniectomy plus best medical treatment and 55 (58%) assigned to best medical treatment alone had an mRS of 5-6 at 180 days (adjusted risk ratio [aRR] 0·77, 95% CI 0·59 to 1·01, adjusted risk difference [aRD] -13%, 95% CI -26 to 0, p=0·057). In the per-protocol analysis, 36 (47%) of 77 participants in the decompressive craniectomy plus best medical treatment group and 44 (60%) of 73 in the best medical treatment alone group had an mRS of 5-6 (aRR 0·76, 95% CI 0·58 to 1·00, aRD -15%, 95% CI -28 to 0). Severe adverse events occurred in 42 (41%) of 103 participants receiving decompressive craniectomy plus best medical treatment and 41 (44%) of 94 receiving best medical treatment. INTERPRETATION: SWITCH provides weak evidence that decompressive craniectomy plus best medical treatment might be superior to best medical treatment alone in people with severe deep intracerebral haemorrhage. The results do not apply to intracerebral haemorrhage in other locations, and survival is associated with severe disability in both groups. FUNDING: Swiss National Science Foundation, Swiss Heart Foundation, Inselspital Stiftung, and Boehringer Ingelheim.


Asunto(s)
Hemorragia Cerebral , Craniectomía Descompresiva , Humanos , Persona de Mediana Edad , Masculino , Craniectomía Descompresiva/métodos , Femenino , Hemorragia Cerebral/cirugía , Anciano , Adulto , Resultado del Tratamiento , Terapia Combinada
3.
Brain ; 147(3): 1057-1074, 2024 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-38153327

RESUMEN

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.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Pericitos/fisiología , Infarto Cerebral
4.
Cereb Cortex ; 33(4): 1074-1089, 2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-35353195

RESUMEN

At present, many studies support the notion that after stroke, remote regions connected to the infarcted area are also affected and may contribute to functional outcome. In the present study, we have analyzed possible microanatomical alterations in pyramidal neurons from the contralesional hemisphere after induced stroke. We performed intracellular injections of Lucifer yellow in pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere in an ischemic stroke mouse model. A detailed 3-dimensional analysis of the neuronal complexity and morphological alterations of dendritic spines was then performed. Our results demonstrate that pyramidal neurons from layer III in the somatosensory cortex of the contralesional hemisphere show selective changes in their dendritic arbors, namely, less dendritic complexity of the apical dendritic arbor-but no changes in the basal dendritic arbor. In addition, we found differences in spine morphology in both apical and basal dendrites comparing the contralesional hemisphere with the lesional hemisphere. Our results show that pyramidal neurons of remote areas connected to the infarct zone exhibit a series of selective changes in neuronal complexity and morphological distribution of dendritic spines, supporting the hypothesis that remote regions connected to the peri-infarcted area are also affected after stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratones , Animales , Corteza Somatosensorial , Células Piramidales/fisiología , Neuronas , Dendritas/fisiología
5.
Cell Mol Life Sci ; 80(12): 369, 2023 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-37989805

RESUMEN

Mutations of large conductance Ca2+- and voltage-activated K+ channels (BK) are associated with cognitive impairment. Here we report that CA1 pyramidal neuron-specific conditional BK knock-out (cKO) mice display normal locomotor and anxiety behavior. They do, however, exhibit impaired memory acquisition and retrieval in the Morris Water Maze (MWM) when compared to littermate controls (CTRL). In line with cognitive impairment in vivo, electrical and chemical long-term potentiation (LTP) in cKO brain slices were impaired in vitro. We further used a genetically encoded fluorescent K+ biosensor and a Ca2+-sensitive probe to observe cultured hippocampal neurons during chemical LTP (cLTP) induction. cLTP massively reduced intracellular K+ concentration ([K+]i) while elevating L-Type Ca2+ channel- and NMDA receptor-dependent Ca2+ oscillation frequencies. Both, [K+]i decrease and Ca2+ oscillation frequency increase were absent after pharmacological BK inhibition or in cells lacking BK. Our data suggest that L-Type- and NMDAR-dependent BK-mediated K+ outflow significantly contributes to hippocampal LTP, as well as learning and memory.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio , Potenciación a Largo Plazo , Ratones , Animales , Potenciación a Largo Plazo/fisiología , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Plasticidad Neuronal/fisiología , Hipocampo/fisiología , Neuronas , Ratones Noqueados
6.
Stroke ; 54(10): 2666-2670, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37675614

RESUMEN

BACKGROUND: The only established pharmacological treatment option improving outcomes for patients suffering from subarachnoid hemorrhage (SAH) is the L-type-calcium channel inhibitor nimodipine. However, the exact mechanisms of action of nimodipine conferring neuroprotection after SAH have yet to be determined. More recently, spasms of the cerebral microcirculation were suggested to play an important role in reduced cerebral perfusion after SAH and, ultimately, outcome. It is unclear whether nimodipine may influence microvasospasms and, thus, microcirculatory dysfunction. The aim of the current study was, therefore, to assess the effect of nimodipine on microvasospasms after experimental SAH. METHODS: Male C57Bl/6 N mice (n=3-5/group) were subjected to SAH using the middle cerebral artery perforation model. Six hours after SAH induction, a cranial window was prepared, and the diameter of cortical microvessels was assessed in vivo by 2-photon-microscopy before, during, and after nimodipine application. RESULTS: Nimodipine significantly reduced the number of posthemorrhagic microvasospasms. The diameters of nonspastic vessels were not affected. CONCLUSIONS: Our results show that nimodipine reduces the formation of microvasospasms, thus, shedding new light on the mode of action of a drug routinely used for the treatment of SAH for >3 decades. Furthermore, L-type Ca2+ channels may be involved in the pathophysiology of microvasospasm formation.


Asunto(s)
Nimodipina , Hemorragia Subaracnoidea , Humanos , Animales , Ratones , Masculino , Nimodipina/farmacología , Nimodipina/uso terapéutico , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/tratamiento farmacológico , Microcirculación , Ratones Endogámicos C57BL , Microvasos
7.
Stroke ; 54(8): 2126-2134, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37325921

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) is characterized by acute and delayed reductions of cerebral blood flow (CBF) caused, among others, by spasms of cerebral arteries and arterioles. Recently, the inactivation of perivascular macrophages (PVM) has been demonstrated to improve neurological outcomes after experimental SAH, but the underlying mechanisms of protection remain unclear. The aim of our exploratory study was, therefore, to investigate the role of PVM in the formation of acute microvasospasms after experimental SAH. METHODS: PVMs were depleted in 8- to 10-week-old male C57BL/6 mice (n=8/group) by intracerebroventricular application of clodronate-loaded liposomes and compared with mice with vehicle liposome injections. Seven days later, SAH was induced by filament perforation under continuous monitoring of CBF and intracranial pressure. Results were compared with sham-operated animals and animals who underwent SAH induction but no liposome injection (n=4/group each). Six hours after SAH induction or sham surgery, numbers of microvasospasms per volume of interest and % of affected pial and penetrating arterioles were examined in 9 standardized regions of interest per animal by in vivo 2-photon microscopy. Depletion of PVMs was proven by quantification of PVMs/mm3 identified by immunohistochemical staining for CD206 and Collagen IV. Statistical significance was tested with t tests for parametric data and Mann-Whitney U test for nonparametric data. RESULTS: PVMs were located around pial and intraparenchymal arterioles and were effectively depleted by clodronate from 671±28 to 46±14 PVMs/mm3 (P<0.001). After SAH, microvasospasms was observed in pial arteries and penetrating and precapillary arterioles and were accompanied by an increase to 1405±142 PVMs/mm3. PVM depletion significantly reduced the number of microvasospasms from 9 IQR 5 to 3 IQR 3 (P<0.001). CONCLUSIONS: Our results suggest that PVMs contribute to the formation of microvasospasms after experimental SAH.


Asunto(s)
Hemorragia Subaracnoidea , Ratones , Masculino , Animales , Hemorragia Subaracnoidea/complicaciones , Ácido Clodrónico , Ratones Endogámicos C57BL , Arteriolas , Circulación Cerebrovascular/fisiología , Modelos Animales de Enfermedad
8.
Stroke ; 54(8): 2172-2177, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37381898

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) is characterized by an acute reduction of cerebral blood flow and subsequent cortical infarcts, but the underlying mechanisms are not well understood. Since pericytes regulate cerebral perfusion on the capillary level, we hypothesize that pericytes may reduce cerebral perfusion after SAH. METHODS: Pericytes and vessel diameters of cerebral microvessels were imaged in vivo using NG2 (neuron-glial antigen 2) reporter mice and 2-photon microscopy before and 3 hours after sham surgery or induction of SAH by perforating the middle cerebral artery with an intraluminal filament. Twenty-four hours after, SAH pericyte density was assessed by immunohistochemistry. RESULTS: SAH caused pearl-string-like constrictions of pial arterioles, slowed down blood flow velocity in pial arterioles by 50%, and reduced the volume of intraparenchymal arterioles and capillaries by up to 70% but did not affect pericyte density or induce capillary constriction by pericytes. CONCLUSIONS: Our results suggest that perfusion deficits after SAH are not induced by pericyte-mediated capillary constrictions.


Asunto(s)
Pericitos , Hemorragia Subaracnoidea , Ratones , Animales , Pericitos/fisiología , Capilares , Hemorragia Subaracnoidea/complicaciones , Vasoconstricción/fisiología , Perfusión
9.
J Neuroinflammation ; 20(1): 301, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38102677

RESUMEN

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.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratones , Animales , Óxido Nítrico , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Molécula 1 de Adhesión Intercelular/metabolismo
10.
Medicina (Kaunas) ; 59(12)2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38138271

RESUMEN

Background and Objectives: Dissecting the complex pathological cascade of an ischemic stroke in preclinical models is highly warranted to understand the course of this disease in humans. Neurogenesis and angiogenesis are integral for post-stroke recovery, yet it is not clear how these processes are altered months after an ischemic stroke. In this study, we investigated the changes that take place subacutely after focal cerebral ischemia in experimental adult male mice. Materials and Methods: Male 12-week-old C57BL/6 mice underwent a 60 min long fMCAo or sham surgery. Two months after the procedure, we examined the immunohistochemistry to assess the changes in neuroblast (DCX) and differentiated neuron (NeuN) numbers, as well as the density of the pro-angiogenic factor VEGF. Results: We found decreased neuroblast numbers in both brain hemispheres of the fMCAo mice: by more than 85% in the dentate gyrus and by more than 70% in the subventricular zone. No neuroblasts were found in the contralateral hemisphere of the fMCAO mice or the sham controls, but a small population was detected in the ipsilateral ischemic core of the fMCAo mice. Intriguingly, the number of differentiated neurons in the ipsilateral ischemic core was lower by 20% compared to the contralateral hemisphere. VEGF expression was diminished in both brain hemispheres of the fMCAo mice. Conclusions: Our current report shows that focal cerebral ischemia induces changes in neuroblast numbers and the pro-angiogenic factor VEGF in both cerebral hemispheres 2 months after an fMCAo in mice. Our data show that focal cerebral ischemia induces a long-term regenerative response in both brain hemispheres.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Humanos , Ratones , Masculino , Animales , Inductores de la Angiogénesis , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones Endogámicos C57BL , Isquemia Encefálica/complicaciones , Neuronas/metabolismo , Infarto Cerebral/patología , Isquemia/patología
11.
Small ; 18(18): e2200302, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35384294

RESUMEN

The current lack of understanding about how nanocarriers cross the blood-brain barrier (BBB) in the healthy and injured brain is hindering the clinical translation of nanoscale brain-targeted drug-delivery systems. Here, the bio-distribution of lipid nano-emulsion droplets (LNDs) of two sizes (30 and 80 nm) in the mouse brain after traumatic brain injury (TBI) is investigated. The highly fluorescent LNDs are prepared by loading them with octadecyl rhodamine B and a bulky hydrophobic counter-ion, tetraphenylborate. Using in vivo two-photon and confocal imaging, the circulation kinetics and bio-distribution of LNDs in the healthy and injured mouse brain are studied. It is found that after TBI, LNDs of both sizes accumulate at vascular occlusions, where specifically 30 nm LNDs extravasate into the brain parenchyma and reach neurons. The vascular occlusions are not associated with bleedings, but instead are surrounded by processes of activated microglia, suggesting a specific opening of the BBB. Finally, correlative light-electron microscopy reveals 30 nm LNDs in endothelial vesicles, while 80 nm particles remain in the vessel lumen, indicating size-selective vesicular transport across the BBB via vascular occlusions. The data suggest that microvascular occlusions serve as "gates" for the transport of nanocarriers across the BBB.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nanopartículas , Animales , Barrera Hematoencefálica , Encéfalo , Portadores de Fármacos/química , Liposomas , Ratones , Nanopartículas/química
12.
J Neurosci Res ; 100(5): 1201-1217, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33789358

RESUMEN

Traumatic brain injuries (TBIs) are common with an estimated 27.1 million cases per year. Approximately 80% of TBIs are categorized as mild TBI (mTBI) based on initial symptom presentation. While in most individuals, symptoms resolve within days to weeks, in some, symptoms become chronic. Advanced neuroimaging has the potential to characterize brain morphometric, microstructural, biochemical, and metabolic abnormalities following mTBI. However, translational studies are needed for the interpretation of neuroimaging findings in humans with respect to the underlying pathophysiological processes, and, ultimately, for developing novel and more targeted treatment options. In this review, we introduce the most commonly used animal models for the study of mTBI. We then summarize the neuroimaging findings in humans and animals after mTBI and, wherever applicable, the translational aspects of studies available today. Finally, we highlight the importance of translational approaches and outline future perspectives in the field of translational neuroimaging in mTBI.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Encéfalo/diagnóstico por imagen , Conmoción Encefálica/diagnóstico por imagen , Humanos , Imagen por Resonancia Magnética/métodos , Neuroimagen/métodos
13.
FASEB J ; 35(11): e21962, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34613632

RESUMEN

Proteolytic ectodomain shedding of membrane proteins is a fundamental mechanism to control the communication between cells and their environment. A key protease for membrane protein shedding is ADAM17, which requires a non-proteolytic subunit, either inactive Rhomboid 1 (iRhom1) or iRhom2 for its activity. While iRhom1 and iRhom2 are co-expressed in most tissues and appear to have largely redundant functions, the brain is an organ with predominant expression of iRhom1. Yet, little is known about the spatio-temporal expression of iRhom1 in mammalian brain and about its function in controlling membrane protein shedding in the nervous system. Here, we demonstrate that iRhom1 is expressed in mouse brain from the prenatal stage to adulthood with a peak in early postnatal development. In the adult mouse brain iRhom1 was widely expressed, including in cortex, hippocampus, olfactory bulb, and cerebellum. Proteomic analysis of the secretome of primary neurons using the hiSPECS method and of cerebrospinal fluid, obtained from iRhom1-deficient and control mice, identified several membrane proteins that require iRhom1 for their shedding in vitro or in vivo. One of these proteins was 'multiple-EGF-like-domains protein 10' (MEGF10), a phagocytic receptor in the brain that is linked to the removal of amyloid ß and apoptotic neurons. MEGF10 was further validated as an ADAM17 substrate using ADAM17-deficient mouse embryonic fibroblasts. Taken together, this study discovers a role for iRhom1 in controlling membrane protein shedding in the mouse brain, establishes MEGF10 as an iRhom1-dependent ADAM17 substrate and demonstrates that iRhom1 is widely expressed in murine brain.


Asunto(s)
Proteína ADAM17/metabolismo , Encéfalo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Embrionarias de Ratones
14.
Nanomedicine ; 40: 102511, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34915181

RESUMEN

The potential of poly(lactic-co-glycolic acid) (PLGA) to design nanoparticles (NPs) and target the central nervous system remains to be exploited. In the current study we designed fluorescent 70-nm PLGA NPs, loaded with bulky fluorophores, thereby making them significantly brighter than quantum dots in single-particle fluorescence measurements. The high brightness of NPs enabled their visualization by intravital real-time 2-photon microscopy. Subsequently, we found that PLGA NPs coated with pluronic F-68 circulated in the blood substantially longer than uncoated NPs and were taken up by cerebro-vascular endothelial cells. Additionally, confocal microscopy revealed that coated PLGA NPs were present in late endothelial endosomes of cerebral vessels within 1 h after systemic injection and were more readily taken up by endothelial cells in peripheral organs. The combination of ultra-bright NPs and in vivo imaging may thus represent a promising approach to reduce the gap between development and clinical application of nanoparticle-based drug carriers.


Asunto(s)
Nanopartículas , Poloxámero , Portadores de Fármacos , Células Endoteliales , Glicoles , Microscopía , Tamaño de la Partícula , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
15.
Stroke ; 52(12): 4033-4042, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34749506

RESUMEN

BACKGROUND AND PURPOSE: Subarachnoid hemorrhage (SAH) is associated with acute and delayed cerebral ischemia resulting in high acute mortality and severe chronic neurological deficits. Spasms of the pial and intraparenchymal microcirculation (microvasospasms) contribute to acute cerebral ischemia after SAH; however, the underlying mechanisms remain unknown. We hypothesize that free iron (Fe3+) released from hemolytic red blood cells into the subarachnoid space may be involved in microvasospasms formation. METHODS: Male C57BL/6 mice (n=8/group) received 200 mg/kg of the iron scavenger deferoxamine or vehicle intravenously and were then subjected to SAH by filament perforation. Microvasospasms of pial and intraparenchymal vessels were imaged three hours after SAH by in vivo 2-photon microscopy. RESULTS: Microvasospasms occurred in all investigated vessel categories down to the capillary level. Deferoxamine significantly reduced the number of microvasospasms after experimental SAH. The effect was almost exclusively observed in larger pial arterioles (>30 µm) covered with blood. CONCLUSIONS: These results provide proof-of-principle evidence that Fe3+ is involved in the formation of arteriolar microvasospasms after SAH and that arteriolar and capillary microvasospasms are triggered by different mechanisms. Deciphering the mechanisms of Fe3+-induced microvasospasms may result in novel therapeutic strategies for SAH patients.


Asunto(s)
Hierro/metabolismo , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/metabolismo , Vasoespasmo Intracraneal/etiología , Vasoespasmo Intracraneal/metabolismo , Animales , Arteriolas , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Capilares , Deferoxamina/farmacología , Masculino , Ratones Endogámicos C57BL , Sideróforos/farmacología
16.
J Neurochem ; 159(4): 710-728, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33694332

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Accidente Cerebrovascular Isquémico/patología , MicroARNs/genética , Neuronas/patología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Animales , Corteza Cerebral/patología , Biología Computacional , Activación Enzimática , Femenino , Masculino , Ratones Endogámicos C57BL , Fosfohidrolasa PTEN/metabolismo , Reacción en Cadena de la Polimerasa , Cultivo Primario de Células , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Proteína S6 Ribosómica/metabolismo , Transducción de Señal
18.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445151

RESUMEN

Subarachnoid hemorrhage (SAH) is associated with acute and delayed cerebral ischemia. We suggested spasms of pial arterioles as a possible mechanism; however, it remained unclear whether and how pial microvasospasms (MVSs) induce cerebral ischemia. Therefore, we used in vivo deep tissue imaging by two-photon microscopy to investigate MVSs together with the intraparenchymal microcirculation in a clinically relevant murine SAH model. Male C57BL/6 mice received a cranial window. Cerebral vessels and leukocytes were labelled with fluorescent dyes and imaged by in vivo two-photon microscopy before and three hours after SAH induced by filament perforation. After SAH, a large clot formed around the perforation site at the skull base, and blood distributed along the perivascular space of the middle cerebral artery up to the cerebral cortex. Comparing the cerebral microvasculature before and after SAH, we identified three different patterns of constrictions: pearl string, global, and bottleneck. At the same time, the volume of perfused intraparenchymal vessels and blood flow velocity in individual arterioles were significantly reduced by more than 60%. Plugging of capillaries by leukocytes was observed but infrequent. The current study demonstrates that perivascular blood is associated with spasms of pial arterioles and that these spasms result in a significant reduction in cortical perfusion after SAH. Thus, the pial microvasospasm seems to be an important mechanism by which blood in the subarachnoid space triggers cerebral ischemia after SAH. Identifying the mechanisms of pial vasospasm may therefore result in novel therapeutic options for SAH patients.


Asunto(s)
Encéfalo/irrigación sanguínea , Leucocitos/patología , Microvasos/patología , Hemorragia Subaracnoidea/patología , Vasoespasmo Intracraneal/patología , Animales , Encéfalo/patología , Circulación Cerebrovascular , Microscopía Intravital , Masculino , Ratones Endogámicos C57BL , Microscopía de Fluorescencia por Excitación Multifotónica
19.
Crit Care Med ; 48(8): 1157-1164, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32697486

RESUMEN

OBJECTIVES: Traumatic brain injury is the number one cause of death in children and young adults and has become increasingly prevalent in the elderly. Decompressive craniectomy prevents intracranial hypertension but does not clearly improve physical outcome 6 months after traumatic brain injury. However, it has not been analyzed if decompressive craniectomy affects traumatic brain injury patients' quality of life in the long term. DESIGN: Therefore, we conducted a cross-sectional study assessing health-related quality of life in traumatic brain injury patients with or without decompressive craniectomy up to 10 years after injury. SETTING: Former critical care patients. PATIENTS: Chronic traumatic brain injury patients having not (n = 37) or having received (n = 98) decompressive craniectomy during the acute treatment. MEASUREMENTS AND MAIN RESULTS: Decompressive craniectomy was necessary in all initial traumatic brain injury severity groups. Eight percent more decompressive craniectomy patients reported good health-related quality of life with a Quality of Life after Brain Injury total score greater than or equal to 60 compared with the no decompressive craniectomy patients up to 10 years after traumatic brain injury (p = 0.004). Initially, mild classified traumatic brain injury patients had a median Quality of Life after Brain Injury total score of 83 (decompressive craniectomy) versus 62 (no decompressive craniectomy) (p = 0.028). Health-related quality of life regarding physical status was better in decompressive craniectomy patients (p = 0.025). Decompressive craniectomy showed a trend toward better health-related quality of life in the 61-85-year-old reflected by median Quality of Life after Brain Injury total scores of 62 (no decompressive craniectomy) versus 79 (decompressive craniectomy) (p = 0.06). CONCLUSIONS: Our results suggest that decompressive craniectomy is associated with good health-related quality of life up to 10 years after traumatic brain injury. Thus, decompressive craniectomy may have an underestimated therapeutic potential after traumatic brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/cirugía , Craniectomía Descompresiva , Calidad de Vida , Factores de Edad , Anciano , Anciano de 80 o más Años , Lesiones Traumáticas del Encéfalo/rehabilitación , Estudios Transversales , Craniectomía Descompresiva/métodos , Femenino , Escala de Coma de Glasgow , Humanos , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento
20.
Ann Neurol ; 85(5): 667-680, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30843275

RESUMEN

OBJECTIVE: Plasminogen activator inhibitor-1 (PAI-1) is the key endogenous inhibitor of fibrinolysis, and enhances clot formation after injury. In traumatic brain injury, dysregulation of fibrinolysis may lead to sustained microthrombosis and accelerated lesion expansion. In the present study, we hypothesized that PAI-1 mediates post-traumatic malfunction of coagulation, with inhibition or genetic depletion of PAI-1 attenuating clot formation and lesion expansion after brain trauma. METHODS: We evaluated PAI-1 as a possible new target in a mouse controlled cortical impact (CCI) model of traumatic brain injury. We performed the pharmacological inhibition of PAI-1 with PAI-039 and stimulation by tranexamic acid, and we confirmed our results in PAI-1-deficient animals. RESULTS: PAI-1 mRNA was time-dependently upregulated, with a 305-fold peak 12 hours after CCI, which effectively counteracted the 2- to 3-fold increase in cerebral tissue-type/urokinase plasminogen activator expression. PAI-039 reduced brain lesion volume by 26% at 24 hours and 43% at 5 days after insult. This treatment also attenuated neuronal apoptosis and improved neurofunctional outcome. Moreover, intravital microscopy demonstrated reduced post-traumatic thrombus formation in the pericontusional cortical microvasculature. In PAI-1-deficient mice, the therapeutic effect of PAI-039 was absent. These mice also displayed 13% reduced brain damage compared with wild type. In contrast, inhibition of fibrinolysis with tranexamic acid increased lesion volume by 25% compared with vehicle. INTERPRETATION: This study identifies impaired fibrinolysis as a critical process in post-traumatic secondary brain damage and suggests that PAI-1 may be a central endogenous inhibitor of the fibrinolytic pathway, promoting a procoagulatory state and clot formation in the cerebral microvasculature. Ann Neurol 2019;85:667-680.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Encéfalo/metabolismo , Encéfalo/patología , Fibrinólisis/fisiología , Serpina E2/metabolismo , Animales , Encéfalo/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Fibrinólisis/efectos de los fármacos , Ácidos Indolacéticos/farmacología , Ácidos Indolacéticos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Serpina E2/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA