Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Shock ; 2024 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-39194254

RESUMO

OBJECTIVE: Vascular endothelial cells (ECs) sense and respond to both trauma factors (histone proteins) and sepsis signals (bacterial lipopolysaccharide, LPS) with elevations in calcium (Ca2+), but it is not clear if the patterns of activation are similar or different. We hypothesized that within seconds of exposure, histones but not LPS would produce a large EC Ca2+ response. We also hypothesized that histones would produce different spatio-temporal patterns of Ca2+ events in veins than in arteries. METHODS: We studied cultured ECs (Ea.Hy926) and native endothelial cells from surgically-opened murine blood vessels. High-speed live cell imaging of Ca2+ events were acquired for 5 minutes before and after stimulation of cultured ECs with histones or LPS alone or in combination. Histone-induced EC Ca2+ events were also compared in native endothelial cells from resistance-sized arteries and veins. Ca2+ activity was quantified as "Ca2+ prevalence" using custom spatiotemporal analysis. Additionally, cultured ECs were collected after 6 hours of exposure to histones or LPS for RNA sequencing. RESULTS: ECs - both in culture and in blood vessels - rapidly increased Ca2+ activity within seconds of histone exposure. In contrast, LPS exposure produced only a slight increase in Ca2+ activity in cultured ECs and no effect on blood vessels over 5-minute recording periods. Histones evoked large aberrant Ca2+ events (>30 seconds in duration) in both veins and arteries, but with different spatio-temporal patterns. Ca2+ activity in arterial ECs appeared as "rosettes", with Ca2+ events that propagated from one cell to all adjacent surrounding cells. In veins, ECs responsed individually without spreading. Suprisingly, exposure of cultured ECs to LPS for 5-minutes before histones potentiated EC Ca2+ activity by an order of magnitude. Exposure of ECs to histones or LPS both increased gene expression, but different mRNAs were induced. CONCLUSIONS: LPS and histones activate ECs through mechanisms that are distinct and additive; only histones produce large aberrant Ca2+ events. ECs in arteries and veins display different patterns of Ca2+ responses to histones.

2.
J Cereb Blood Flow Metab ; 44(5): 680-688, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38420777

RESUMO

The accumulation of the microtubule-associated tau protein in and around blood vessels contributes to brain microvascular dysfunction through mechanisms that are incompletely understood. Delivery of nutrients to active neurons in the brain relies on capillary calcium (Ca2+) signals to direct blood flow. The initiation and amplification of endothelial cell Ca2+ signals require an intact microtubule cytoskeleton. Since tau accumulation in endothelial cells disrupts native microtubule stability, we reasoned that tau-induced microtubule destabilization would impair endothelial Ca2+ signaling. We tested the hypothesis that tau disrupts the regulation of local cerebral blood flow by reducing endothelial cell Ca2+ signals and endothelial-dependent vasodilation. We used a pathogenic soluble tau peptide (T-peptide) model of tau aggregation and mice with genetically encoded endothelial Ca2+ sensors to measure cerebrovascular endothelial responses to tau exposure. T-peptide significantly attenuated endothelial Ca2+ activity and cortical capillary blood flow in vivo. Further, T-peptide application constricted pressurized cerebral arteries and inhibited endothelium-dependent vasodilation. This study demonstrates that pathogenic tau alters cerebrovascular function through direct attenuation of endothelial Ca2+ signaling and endothelium-dependent vasodilation.


Assuntos
Sinalização do Cálcio , Circulação Cerebrovascular , Microvasos , Vasodilatação , Proteínas tau , Animais , Vasodilatação/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Camundongos , Proteínas tau/metabolismo , Microvasos/metabolismo , Microvasos/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Masculino , Endotélio Vascular/metabolismo , Endotélio Vascular/efeitos dos fármacos , Cálcio/metabolismo
3.
bioRxiv ; 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37609200

RESUMO

The accumulation of the microtubule-associated tau protein in and around blood vessels contributes to brain microvascular dysfunction through mechanisms that are incompletely understood. Delivery of nutrients to active neurons in the brain relies on capillary inositol 1,4,5-triphosphate receptor (IP3R)-mediated calcium (Ca2+) signals to direct blood flow. The initiation and amplification of endothelial cell IP3R-mediated Ca2+ signals requires an intact microtubule cytoskeleton. Since tau accumulation in endothelial cells disrupts native microtubule stability, we reasoned that tau-induced microtubule destabilization would impair endothelial IP3-evoked Ca2+ signaling. We tested the hypothesis that tau disrupts the regulation of local cerebral blood flow by reducing endothelial cell Ca2+ signals and endothelial-dependent vasodilation. We used a pathogenic soluble tau peptide (T-peptide) model of tau aggregation and mice with genetically encoded endothelial Ca2+ sensors to measure cerebrovascular endothelial responses to tau exposure. T-peptide significantly attenuated endothelial Ca2+ activity and cortical capillary blood flow in vivo within 120 seconds. Further, T-peptide application constricted pressurized cerebral arteries and inhibited endothelium-dependent vasodilation. This study demonstrates that pathogenic tau alters cerebrovascular function through direct attenuation of endothelial Ca2+ signaling and endothelium-dependent vasodilation.

4.
J Immunol ; 211(4): 648-657, 2023 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-37405700

RESUMO

Drugs are needed to protect against the neutrophil-derived histones responsible for endothelial injury in acute inflammatory conditions such as trauma and sepsis. Heparin and other polyanions can neutralize histones but challenges with dosing or side effects such as bleeding limit clinical application. In this study, we demonstrate that suramin, a widely available polyanionic drug, completely neutralizes the toxic effects of individual histones, but not citrullinated histones from neutrophil extracellular traps. The sulfate groups on suramin form stable electrostatic interactions with hydrogen bonds in the histone octamer with a dissociation constant of 250 nM. In cultured endothelial cells (Ea.Hy926), histone-induced thrombin generation was significantly decreased by suramin. In isolated murine blood vessels, suramin abolished aberrant endothelial cell calcium signals and rescued impaired endothelial-dependent vasodilation caused by histones. Suramin significantly decreased pulmonary endothelial cell ICAM-1 expression and neutrophil recruitment caused by infusion of sublethal doses of histones in vivo. Suramin also prevented histone-induced lung endothelial cell cytotoxicity in vitro and lung edema, intra-alveolar hemorrhage, and mortality in mice receiving a lethal dose of histones. Protection of vascular endothelial function from histone-induced damage is a novel mechanism of action for suramin with therapeutic implications for conditions characterized by elevated histone levels.


Assuntos
Histonas , Suramina , Camundongos , Animais , Histonas/metabolismo , Suramina/farmacologia , Células Endoteliais/metabolismo , Endotélio/metabolismo , Hemorragia
5.
Vascul Pharmacol ; 142: 106950, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34871770

RESUMO

Coagulopathy in severe COVID-19 is common but poorly understood. The purpose of this study was to determine how SARS-CoV-2 infection impacts histone levels, fibrin structure, and endogenous thrombin potential in the presence and absence of endothelial cells. We studied individuals with SARS-CoV-2 infection and acute respiratory distress syndrome at the time of initiation of mechanical ventilation compared to healthy controls. Circulating histone-DNA complexes were elevated in the plasma of COVID-19 patients relative to healthy controls (n=6, each group). Using calibrated automated thrombography, thrombin generation was altered in COVID-19 patient plasma samples. Despite having increased endogenous thrombin potential, patient plasma samples exhibited prolonged lag times and times to peak thrombin in the presence of added tissue factor and PCPS. Strikingly different results were observed when endothelial cells were used in place of tissue factor and PCPS. While healthy control plasma samples did not generate measurable thrombin after 60 min, plasma samples from COVID-19+ patients formed thrombin (mean lag time ~20 min). Consistent with the observed alterations in thrombin generation, clots from COVID-19 subjects exhibited a denser fibrin network, thinner fibers and lower fibrin resolvability. Elevated histones, aberrant fibrin formation, and increased endothelial-dependent thrombin generation may contribute to coagulopathy in COVID-19.


Assuntos
COVID-19 , Histonas , DNA , Células Endoteliais , Humanos , SARS-CoV-2 , Trombina
6.
Function (Oxf) ; 2(3)2021.
Artigo em Inglês | MEDLINE | ID: mdl-34568829

RESUMO

Trauma can lead to widespread vascular dysfunction, but the underlying mechanisms remain largely unknown. Inward-rectifier potassium channels (Kir2.1) play a critical role in the dynamic regulation of regional perfusion and blood flow. Kir2.1 channel activity requires phosphatidylinositol 4,5-bisphosphate (PIP2), a membrane phospholipid that is degraded by phospholipase A2 (PLA2) in conditions of oxidative stress or inflammation. We hypothesized that PLA2-induced depletion of PIP2 after trauma impairs Kir2.1 channel function. A fluid percussion injury model of traumatic brain injury (TBI) in rats was used to study mesenteric resistance arteries 24 hours after injury. The functional responses of intact arteries were assessed using pressure myography. We analyzed circulating PLA2, hydrogen peroxide (H2O2), and metabolites to identify alterations in signaling pathways associated with PIP2 in TBI. Electrophysiology analysis of freshly-isolated endothelial and smooth muscle cells revealed a significant reduction of Ba2+-sensitive Kir2.1 currents after TBI. Additionally, dilations to elevated extracellular potassium and BaCl2- or ML 133-induced constrictions in pressurized arteries were significantly decreased following TBI, consistent with an impairment of Kir2.1 channel function. The addition of a PIP2 analog to the patch pipette successfully rescued endothelial Kir2.1 currents after TBI. Both H2O2 and PLA2 activity were increased after injury. Metabolomics analysis demonstrated altered lipid metabolism signaling pathways, including increased arachidonic acid, and fatty acid mobilization after TBI. Our findings support a model in which increased H2O2-induced PLA2 activity after trauma hydrolyzes endothelial PIP2, resulting in impaired Kir2.1 channel function.


Assuntos
Lesões Encefálicas Traumáticas , Peróxido de Hidrogênio , Ratos , Animais , Hemodinâmica , Transdução de Sinais
7.
J Cereb Blood Flow Metab ; 41(6): 1313-1327, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33050826

RESUMO

Traumatic brain injury (TBI) acutely impairs dynamic regulation of local cerebral blood flow, but long-term (>72 h) effects on functional hyperemia are unknown. Functional hyperemia depends on capillary endothelial cell inward rectifier potassium channels (Kir2.1) responding to potassium (K+) released during neuronal activity to produce a regenerative, hyperpolarizing electrical signal that propagates from capillaries to dilate upstream penetrating arterioles. We hypothesized that TBI causes widespread disruption of electrical signaling from capillaries-to-arterioles through impairment of Kir2.1 channel function. We randomized mice to TBI or control groups and allowed them to recover for 4 to 7 days post-injury. We measured in vivo cerebral hemodynamics and arteriolar responses to local stimulation of capillaries with 10 mM K+ using multiphoton laser scanning microscopy through a cranial window under urethane and α-chloralose anesthesia. Capillary angio-architecture was not significantly affected following injury. However, K+-induced hyperemia was significantly impaired. Electrophysiology recordings in freshly isolated capillary endothelial cells revealed diminished Ba2+-sensitive Kir2.1 currents, consistent with a reduction in channel function. In pressurized cerebral arteries isolated from TBI mice, K+ failed to elicit the vasodilation seen in controls. We conclude that disruption of endothelial Kir2.1 channel function impairs capillary-to-arteriole electrical signaling, contributing to altered cerebral hemodynamics after TBI.


Assuntos
Arteríolas/metabolismo , Lesões Encefálicas Traumáticas/fisiopatologia , Capilares/metabolismo , Circulação Cerebrovascular/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Células Endoteliais/metabolismo , Hemodinâmica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia
8.
J Trauma Acute Care Surg ; 88(2): 258-265, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31999655

RESUMO

BACKGROUND: Both hyperfibrinolysis and fibrinolysis shutdown can occur after severe trauma. The subgroup of trauma patients with fibrinolysis shutdown resistant to tissue plasminogen activator (t-PA)-mediated fibrinolysis have increased mortality. Fibrin polymerization and structure may influence fibrinolysis subgroups in trauma, but fibrin architecture has not been characterized in acutely injured subjects. We hypothesized that fibrin polymerization measured in situ will correlate with fibrinolysis subgroups. METHODS: Blood samples were collected from trauma patients and noninjured controls. We selected samples across a range of fibrinolysis phenotypes (shutdown, physiologic, hyperfibrinolysis) and t-PA sensitivities (sensitive, physiologic, resistant) determined by thrombelastography. Plasma clots were created in situ with fluorescent fibrinogen and imaged using confocal microscopy for analysis of clot architecture in three dimensions. For each clot, we quantified the fiber resolvability, a metric of fiber distinctness or clarity, by mapping the variance of fluorescence intensity relative to background fluorescence. We also determined clot porosity by measuring the size and distribution of the gaps between fibrin fibers in three-dimensional space. We compared these measures across fibrinolysis subgroups. RESULTS: Fiber resolvability was significantly lower in all trauma subgroups compared with controls (n = 35 and 5, respectively; p < 0.05). We observed markedly different patterns of fibrin architecture among trauma patients stratified by fibrinolysis subgroup. Subjects with t-PA-resistant fibrinolysis shutdown exhibited abnormal, densely packed fibrin clots nearly devoid of pores. Individuals with t-PA-hypersensitive fibrinolysis shutdown had highly irregular clots with pores as large as 2500 µm to 20,000 µm, versus 78 µm to 1250 µm in noninjured controls. CONCLUSION: Fiber resolvability was significantly lower in trauma patients than controls, and subgroups of fibrinolysis differ in the porosity of the fibrin clot structure. The dense fibrin network in the t-PA-resistant group may prevent access to plasmin, suggesting a mechanism for thrombotic morbidity after injury.


Assuntos
Transtornos da Coagulação Sanguínea/sangue , Fibrina/química , Fibrinólise , Ativador de Plasminogênio Tecidual/sangue , Ferimentos e Lesões/complicações , Adulto , Biomarcadores/sangue , Transtornos da Coagulação Sanguínea/diagnóstico , Transtornos da Coagulação Sanguínea/mortalidade , Feminino , Humanos , Escala de Gravidade do Ferimento , Masculino , Pessoa de Meia-Idade , Polimerização , Estudos Retrospectivos , Tromboelastografia , Ferimentos e Lesões/sangue , Ferimentos e Lesões/diagnóstico , Adulto Jovem
9.
Microvasc Res ; 125: 103873, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30974113

RESUMO

The microvascular endothelium plays a key role in regulating solute permeability in the gut, but the contribution of vascular smooth muscle to barrier function is unknown. We sought to determine the role of vascular smooth muscle and its myogenic tone in the vascular barrier to solutes in mesenteric microvessels. We determined vascular permeability to 4.4 kDa and 70 kDa dextrans in isolated mouse mesenteric arteries at increasing pressure increments. The myogenic response was simultaneously monitored using video edge-detection of vessel diameter and wall thickness. We expressed permeability as the apparent permeability coefficient, or the solute flux per second normalized to surface area and concentration gradient. We compared the effects of myogenic tone, L-type calcium channel blockade, calcium elimination, and endothelial removal on the permeability of each dextran. We found arteries resisted changes in 4.4 kDa and 70 kDa dextran permeability coefficients at intravascular pressures associated with myogenic tone. Manipulations that reduced or eliminated myogenic tone (L-type calcium channel blockade or calcium elimination) caused vasodilation and increased permeability coefficients. Thus, the maintenance of a reactive mesenteric vascular smooth muscle layer and its myogenic tone prevents increases in vascular permeability that would otherwise occur with increasing pressure. Conditions that impact vascular tone, such as trauma, stroke, or major surgery could diminish the gut-vascular barrier against dissemination of the microbiome.


Assuntos
Permeabilidade Capilar , Artérias Mesentéricas/fisiologia , Microvasos/fisiologia , Músculo Liso Vascular/fisiologia , Vasoconstrição , Vasodilatação , Animais , Pressão Arterial , Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Dextranos/metabolismo , Endotélio Vascular/fisiologia , Técnicas In Vitro , Masculino , Artérias Mesentéricas/metabolismo , Camundongos Endogâmicos C57BL , Microvasos/metabolismo , Músculo Liso Vascular/metabolismo
10.
J Surg Res ; 213: 100-109, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28601302

RESUMO

INTRODUCTION: Understanding the extent to which murine models of traumatic brain injury (TBI) replicate clinically relevant neurologic outcomes is critical for mechanistic and therapeutic studies. We determined sensorimotor outcomes in a mouse model of TBI and validated the use of a standardized neurologic examination scoring system to quantify the extent of injury. MATERIALS AND METHODS: We used a lateral fluid percussion injury model of TBI and compared TBI animals to those that underwent sham surgery. We measured neurobehavioral deficits using a standardized 12-point neurologic examination, magnetic resonance imaging, a rotating rod test, and longitudinal acoustic startle testing. RESULTS: TBI animals had a significantly decreased ability to balance on a rotating rod and a marked reduction in the amplitude of acoustic startle response. The neurologic examination had a high inter-rater reliability (87% agreement) and correlated with latency to fall on a rotating rod (Rs = -0.809). CONCLUSIONS: TBI impairs sensorimotor function in mice, and the extent of impairment can be predicted by a standardized neurologic examination.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Animais , Lesões Encefálicas Traumáticas/diagnóstico , Lesões Encefálicas Traumáticas/psicologia , Escala de Gravidade do Ferimento , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Exame Neurológico , Testes Neuropsicológicos , Variações Dependentes do Observador , Equilíbrio Postural , Distribuição Aleatória , Reflexo de Sobressalto
11.
J Neurotrauma ; 34(1): 192-203, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26757855

RESUMO

Endothelial dysfunction is a hallmark of many chronic diseases, including diabetes and long-term hypertension. We show that acute traumatic brain injury (TBI) leads to endothelial dysfunction in rat mesenteric arteries. Endothelial-dependent dilation was greatly diminished 24 h after TBI because of impaired nitric oxide (NO) production. The activity of arginase, which competes with endothelial NO synthase (eNOS) for the common substrate l-arginine, were also significantly increased in arteries, suggesting that arginase-mediated depletion of l-arginine underlies diminished NO production. Consistent with this, substrate restoration by exogenous application of l-arginine or inhibition of arginase recovered endothelial function. Moreover, evidence for increased reactive oxygen species production, a consequence of l-arginine starvation-dependent eNOS uncoupling, was detected in endothelium and plasma. Collectively, our findings demonstrate endothelial dysfunction in a remote vascular bed after TBI, manifesting as impaired endothelial-dependent vasodilation, with increased arginase activity, decreased generation of NO, and increased O2- production. We conclude that blood vessels have a "molecular memory" of neurotrauma, 24 h after injury, because of functional changes in vascular endothelial cells; these effects are pertinent to understanding the systemic inflammatory response that occurs after TBI even in the absence of polytrauma.


Assuntos
Arginase/metabolismo , Lesões Encefálicas Traumáticas/enzimologia , Endotélio Vascular/enzimologia , Microcirculação/fisiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Acetilcolina/farmacologia , Animais , Lesões Encefálicas Traumáticas/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/patologia , Masculino , Microcirculação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
12.
J Am Heart Assoc ; 3(6): e001474, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25527626

RESUMO

BACKGROUND: Traumatic brain injury (TBI) has been reported to increase the concentration of nitric oxide (NO) in the brain and can lead to loss of cerebrovascular tone; however, the sources, amounts, and consequences of excess NO on the cerebral vasculature are unknown. Our objective was to elucidate the mechanism of decreased cerebral artery tone after TBI. METHODS AND RESULTS: Cerebral arteries were isolated from rats 24 hours after moderate fluid­percussion TBI. Pressure­induced increases in vasoconstriction (myogenic tone) and smooth muscle Ca2+ were severely blunted in cerebral arteries after TBI. However, myogenic tone and smooth muscle Ca2+ were restored by inhibition of NO synthesis or endothelium removal, suggesting that TBI increased endothelial NO levels. Live native cell NO, indexed by 4,5­diaminofluorescein (DAF­2 DA) fluorescence, was increased in endothelium and smooth muscle of cerebral arteries after TBI. Clamped concentrations of 20 to 30 nmol/L NO were required to simulate the loss of myogenic tone and increased (DAF­2T) fluorescence observed following TBI. In comparison, basal NO in control arteries was estimated as 0.4 nmol/L. Consistent with TBI causing enhanced NO­mediated vasodilation, inhibitors of guanylyl cyclase, protein kinase G, and large­conductance Ca2+­activated potassium (BK) channel restored function of arteries from animals with TBI. Expression of the inducible isoform of NO synthase was upregulated in cerebral arteries isolated from animals with TBI, and the inducible isoform of NO synthase inhibitor 1400W restored myogenic responses following TBI. CONCLUSIONS: The mechanism of profound cerebral artery vasodilation after TBI is a gain of function in vascular NO production by 60­fold over controls, resulting from upregulation of the inducible isoform of NO synthase in the endothelium.


Assuntos
Lesões Encefálicas/enzimologia , Artérias Cerebrais/enzimologia , Endotélio Vascular/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/metabolismo , Vasodilatação , Animais , Pressão Arterial , Lesões Encefálicas/fisiopatologia , Cálcio/metabolismo , Sinalização do Cálcio , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/fisiopatologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Masculino , Mecanotransdução Celular , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/fisiopatologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima , Vasoconstrição , Vasodilatação/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA