Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Cell ; 165(4): 882-95, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27133169

RESUMO

High-fat diet (HFD) feeding induces rapid reprogramming of systemic metabolism. Here, we demonstrate that HFD feeding of mice downregulates glucose transporter (GLUT)-1 expression in blood-brain barrier (BBB) vascular endothelial cells (BECs) and reduces brain glucose uptake. Upon prolonged HFD feeding, GLUT1 expression is restored, which is paralleled by increased expression of vascular endothelial growth factor (VEGF) in macrophages at the BBB. In turn, inducible reduction of GLUT1 expression specifically in BECs reduces brain glucose uptake and increases VEGF serum concentrations in lean mice. Conversely, myeloid-cell-specific deletion of VEGF in VEGF(Δmyel) mice impairs BBB-GLUT1 expression, brain glucose uptake, and memory formation in obese, but not in lean mice. Moreover, obese VEGF(Δmyel) mice exhibit exaggerated progression of cognitive decline and neuroinflammation on an Alzheimer's disease background. These experiments reveal that transient, HFD-elicited reduction of brain glucose uptake initiates a compensatory increase of VEGF production and assign obesity-associated macrophage activation a homeostatic role to restore cerebral glucose metabolism, preserve cognitive function, and limit neurodegeneration in obesity.


Assuntos
Encéfalo/metabolismo , Dieta Hiperlipídica , Glucose/metabolismo , Obesidade/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Cognição , Células Endoteliais/metabolismo , Ácidos Graxos/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Camundongos , Células Mieloides/metabolismo
3.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38731908

RESUMO

In atrial fibrillation (AF), multifactorial pathologic atrial alterations are manifested by structural and electrophysiological changes known as atrial remodeling. AF frequently develops in the context of underlying cardiac abnormalities. A critical mechanistic role played by atrial stretch is played by abnormal substrates in a number of conditions that predispose to AF, including obesity, heart failure, hypertension, and sleep apnea. The significant role of overweight and obesity in the development of AF is known; however, the differential effect of overweight, obesity, cardiovascular comorbidities, lifestyle, and other modifiable risk factors on the occurrence and recurrence of AF remains to be determined. Reverse remodeling of the atrial substrate and subsequent reduction in the AF burden by conversion into a typical sinus rhythm has been associated with weight loss through lifestyle changes or surgery. This makes it an essential pillar in the management of AF in obese patients. According to recently published research, microRNAs (miRs) may function as post-transcriptional regulators of genes involved in atrial remodeling, potentially contributing to the pathophysiology of AF. The focus of this review is on their modulation by both weight loss and catheter ablation interventions to counteract atrial remodeling in AF. Our analysis outlines the experimental and clinical evidence supporting the synergistic effects of weight loss and catheter ablation (CA) in reversing atrial electrical and structural remodeling in AF onset and in recurrent post-ablation AF by attenuating pro-thrombotic, pro-inflammatory, pro-fibrotic, arrhythmogenic, and male-sex-associated hypertrophic remodeling pathways. Furthermore, we discuss the promising role of miRs with prognostic potential as predictive biomarkers in guiding approaches to AF recurrence prevention.


Assuntos
Fibrilação Atrial , Biomarcadores , Ablação por Cateter , MicroRNAs , Redução de Peso , Fibrilação Atrial/metabolismo , Fibrilação Atrial/genética , Fibrilação Atrial/etiologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Ablação por Cateter/métodos , Recidiva , Remodelamento Atrial , Animais , Obesidade/metabolismo , Obesidade/complicações
4.
Int J Mol Sci ; 25(6)2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38542355

RESUMO

Breast cancer brain metastasis (BCBM) is a challenging condition with limited treatment options and poor prognosis. Understanding the interactions between tumor cells and the blood-brain barrier (BBB) is critical for developing novel therapeutic strategies. One promising target is estrogen receptor ß (ERß), which promotes the expression of key tight junction proteins, sealing the BBB and reducing its permeability. In this study, we investigated the effects of 17ß-estradiol (E2) and the selective ERß agonist diarylpropionitrile (DPN) on endothelial and cancer cells. Western blot analysis revealed the expression patterns of ERs in these cell lines, and estrogen treatment upregulated claudin-5 expression in brain endothelial cells. Using in vitro models of the BBB, we found that DPN treatment significantly increased BBB tightness about suppressed BBB transmigration activity of representative Her2-positive (BT-474) and triple-negative (MDA-MB-231) breast cancer cell lines. However, the efficacy of DPN treatment decreased when cancer cells were pre-differentiated in the presence of E2. Our results support ERß as a potential target for the prevention and treatment of BCBM and suggest that targeted vector-based approaches may be effective for future preventive and therapeutic implications.


Assuntos
Neoplasias Encefálicas , Neoplasias da Mama , Humanos , Feminino , Barreira Hematoencefálica/metabolismo , Estrogênios/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Receptor beta de Estrogênio/metabolismo , Células Endoteliais/metabolismo , Encéfalo/metabolismo , Estradiol/farmacologia , Estradiol/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/prevenção & controle , Neoplasias Encefálicas/metabolismo , Células MCF-7 , Receptor alfa de Estrogênio/metabolismo
5.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36232860

RESUMO

Intracranial hemorrhage results in devastating forms of cerebral damage. Frequently, these results also present with cardiac dysfunction ranging from ECG changes to Takotsubo syndrome (TTS). This suggests that intracranial bleeding due to subarachnoid hemorrhage (SAH) disrupts the neuro-cardiac axis leading to neurogenic stress cardiomyopathy (NSC) of different degrees. Following this notion, SAH and secondary TTS could be directly linked, thus contributing to poor outcomes. We set out to test if blood circulation is the driver of the brain-heart axis by investigating serum samples of TTS patients. We present a novel in vitro model combining SAH and secondary TTS to mimic the effects of blood or serum, respectively, on blood-brain barrier (BBB) integrity using in vitro monolayers of an established murine model. We consistently demonstrated decreased monolayer integrity and confirmed reduced Claudin-5 and Occludin levels by RT-qPCR and Western blot and morphological reorganization of actin filaments in endothelial cells. Both tight junction proteins show a time-dependent reduction. Our findings highlight a faster and more prominent disintegration of BBB in the presence of TTS and support the importance of the bloodstream as a causal link between intracerebral bleeding and cardiac dysfunction. This may represent potential targets for future therapeutic inventions in SAH and TTS.


Assuntos
Hemorragia Subaracnóidea , Cardiomiopatia de Takotsubo , Animais , Barreira Hematoencefálica/metabolismo , Claudina-5/metabolismo , Células Endoteliais/metabolismo , Humanos , Camundongos , Ocludina/metabolismo , Ratos , Ratos Sprague-Dawley , Hemorragia Subaracnóidea/metabolismo , Cardiomiopatia de Takotsubo/etiologia , Cardiomiopatia de Takotsubo/metabolismo , Proteínas de Junções Íntimas/metabolismo
6.
Histochem Cell Biol ; 156(3): 283-292, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34043058

RESUMO

Progressive deterioration of the central nervous system (CNS) is commonly associated with aging. An important component of the neurovasculature is the blood-brain barrier (BBB), majorly made up of endothelial cells joined together by intercellular junctions. The relationship between senescence and changes in the BBB has not yet been thoroughly explored. Moreover, the lack of in vitro models for the study of the mechanisms involved in those changes impede further and more in-depth investigations in the field. For this reason, we herein present an in vitro model of the senescent BBB and an initial attempt to identify senescence-associated alterations within.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Animais , Barreira Hematoencefálica/citologia , Células Cultivadas , Senescência Celular , Células Endoteliais/citologia , Camundongos , Modelos Biológicos
7.
Clin Auton Res ; 31(2): 179-185, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33259005

RESUMO

The forebrain cerebral network including the insular cortex plays a crucial role in the regulation of the central autonomic nervous system in relation to emotional stress. Numerous studies have recently shown that the insular cortex also has roles as a vestibular area in addition to auditory function. In this review, we summarize the recent literature regarding the relationship between the insular cortex and vestibular function, and we describe our hypothesis that the insular cortex has a pivotal role in vestibular-cardiovascular integration.


Assuntos
Sistema Nervoso Autônomo , Córtex Cerebral , Humanos , Prosencéfalo
8.
Ann Neurol ; 85(5): 667-680, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30843275

RESUMO

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.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Fibrinólise/fisiologia , Serpina E2/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Fibrinólise/efeitos dos fármacos , Ácidos Indolacéticos/farmacologia , Ácidos Indolacéticos/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Serpina E2/antagonistas & inibidores
9.
J Neurochem ; 147(2): 190-203, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30022488

RESUMO

Acute cerebral lesions are associated with dysregulation of brain glucose homeostasis. Previous studies showed that knockdown of Na+ -D-glucose cotransporter SGLT1 impaired outcome after middle cerebral artery occlusion and that widely expressed intracellular RS1 (RSC1A1) is involved in transcriptional and post-translational down-regulation of SGLT1. In the present study, we investigated whether SGLT1 is up-regulated during traumatic brain injury (TBI) and whether removal of RS1 in mice (RS1-KO) influences SGLT1 expression and outcome. Unexpectedly, brain SGLT1 mRNA in RS1-KO was similar to wild-type whereas it was increased in small intestine and decreased in kidney. One day after TBI, SGLT1 mRNA in the ipsilateral cortex was increased 160% in wild-type and 40% in RS1-KO. After RS1 removal lesion volume 1 day after TBI was reduced by 12%, brain edema was reduced by 28%, and motoric disability determined by a beam walking test was improved. In contrast, RS1 removal did neither influence glucose and glycogen accumulation 1 day after TBI nor up-regulation of inflammatory cytokines TNF-α, IL-1ß and IL-6 or microglia activation 1 or 5 days after TBI. The data provide proof of principle that inhibition or down-regulation of SGLT1 by targeting RS1 in brain could be beneficial for early treatment of TBI.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Encéfalo/patologia , Moléculas de Adesão Celular/deficiência , Transportador 1 de Glucose-Sódio/biossíntese , Animais , Química Encefálica/genética , Edema Encefálico/patologia , Edema Encefálico/prevenção & controle , Moléculas de Adesão Celular/genética , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Citocinas/metabolismo , Proteínas do Olho/genética , Expressão Gênica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/prevenção & controle , Regulação para Cima
10.
Histochem Cell Biol ; 149(5): 451-459, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29721642

RESUMO

In silico methods and models in the pathology of the blood-brain barrier (BBB) or also called BBB "computational pathology", are based on using mathematical approaches together with complex, high-dimensional experimental data to evaluate and predict disease-related impacts on the CNS. These computational methods and tools have been designed to deal with BBB-linked neuropathology at the molecular, cellular, tissue, and organ levels. The molecular and cellular levels mainly include molecular docking and molecular dynamics simulations (atomistic and coarse-grain) of mutated or misfolded tight junction proteins, receptors, and various BBB transporters. The tissue and organ levels encompass the mechanistic and pharmacokinetic models as well as finite-element method and pathway analyses enriched with multiple sources of raw data (e.g., in vitro and in vivo, histopathological records, "-omics", and imaging data). Overall, this review discusses comprehensive computational techniques and strategies at different levels of complexity, providing new insights and future directions for diagnosis, treatment improvement, and a deeper understanding of BBB-related neuropathological events.


Assuntos
Barreira Hematoencefálica/patologia , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Animais , Barreira Hematoencefálica/metabolismo , Humanos
11.
Biol Cell ; 106(7): 219-35, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24749543

RESUMO

BACKGROUND INFORMATION: Glucocorticoids (GCs), including the synthetic GC derivate dexamethasone, are widely used as immunomodulators. One of the numerous side effects of dexamethasone therapy is hypertension arising from reduced release of the endothelium-derived vasodilator nitric oxide (NO). RESULTS: Herein, we described the role of dexamethasone and its glucocorticoid receptor (GR) in the regulation of NO synthesis in vitro using the mouse myocardial microvascular endothelial cell line, MyEND. GC treatment caused a firm decrease of extracellular NO levels, whereas the expression of endothelial NO synthase (eNOS) was not affected. However, GC application induced an impairment of tetrahydrobiopterin (BH4 ) concentrations as well as GTP cyclohydrolase-1 (GTPCH-1) expression, both essential factors for NO production upstream of eNOS. Moreover, dexamethasone stimulation resulted in a substantially decreased GR gene and protein expression in MyEND cells. Importantly, inhibition of proteasome-mediated proteolysis of the GR or overexpression of an ubiquitination-defective GR construct improved the bioavailability of BH4 and strengthened GTPCH-1 expression and eNOS activity. CONCLUSIONS: Summarising our results, we propose a new mechanism involved in the regulation of NO signalling by GCs in myocardial endothelial cells. We suggest that a sufficient GR protein expression plays a crucial role for the management of GC-induced harmful adverse effects, including deregulations of vasorelaxation arising from disturbed NO biosynthesis.


Assuntos
Dexametasona/farmacologia , Células Endoteliais/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Receptores de Glucocorticoides/metabolismo , Animais , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Linhagem Celular , Vasos Coronários/metabolismo , Dexametasona/metabolismo , GTP Cicloidrolase/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Receptores de Glucocorticoides/genética
13.
Biomolecules ; 14(2)2024 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-38397404

RESUMO

Takotsubo syndrome (TTS) is a cardiomyopathy that clinically presents as a transient and reversible left ventricular wall motion abnormality (LVWMA). Recovery can occur spontaneously within hours or weeks. Studies have shown that it mainly affects older people. In particular, there is a higher prevalence in postmenopausal women. Physical and emotional stress factors are widely discussed and generally recognized triggers. In addition, the hypothalamic-pituitary-adrenal (HPA) axis and the associated glucocorticoid-dependent negative feedback play an important role in the resulting immune response. This review aims to highlight the unstudied aspects of the trigger factors of TTS. The focus is on emotional stress/chronic unpredictable mild stress (CUMS), which is influenced by estrogen concentration and noradrenaline, for example, and can lead to changes in the behavioral, hormonal, and autonomic systems. Age- and gender-specific aspects, as well as psychological effects, must also be considered. We hypothesize that this leads to a stronger corticosteroid response and altered feedback of the HPA axis. This may trigger proinflammatory markers and thus immunosuppression, inflammaging, and sympathetic overactivation, which contributes significantly to the development of TTS. The aim is to highlight the importance of CUMS and psychological triggers as risk factors and to make an exploratory proposal based on the new knowledge. Based on the imbalance between the sympathetic and parasympathetic nervous systems, transcutaneous vagus nerve stimulation (tVNS) is presented as a possible new therapeutic approach.


Assuntos
Cardiomiopatia de Takotsubo , Humanos , Feminino , Idoso , Sistema Hipotálamo-Hipofisário , Sistema Hipófise-Suprarrenal , Estresse Psicológico , Glucocorticoides
14.
Crit Care Med ; 41(5): 1305-15, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23474678

RESUMO

OBJECTIVES: To establish the molecular background for glucocorticoid insensitivity, that is, failure to reduce edema formation and to protect blood-brain barrier integrity after acute traumatic brain injury. DESIGN: Controlled animal study. SETTING: University research laboratory. SUBJECTS: Male C57Bl/6N mice. INTERVENTIONS: Mechanical brain lesion by controlled cortical impact. MEASUREMENTS AND MAIN RESULTS: Our study demonstrates that 1) proteasomal glucocorticoid receptor degradation is established in brain endothelial cells after traumatic brain injury as a form of posttranslational glucocorticoid receptor modification; 2) inhibition of the proteasomal degradation pathway with bortezomib (0.2 mg/kg) in combination with the glucocorticoid dexamethasone (10 mg/kg) by subcutaneous injection 30 minutes postinjury restores levels of barrier sealing glucocorticoid receptor target occludin in brain endothelial cells, improves blood-brain barrier integrity, reduces edema formation, and limits neuronal damage after brain trauma. CONCLUSIONS: The results indicate that the stabilizing effect of glucocorticoids on the blood-brain barrier is hampered after cerebral lesions by proteasomal glucocorticoid receptor degradation in brain endothelial cells and restored by inhibition of proteasomal degradation pathways. The results provide underlying mechanisms for the clinically observed inefficacy of glucocorticoids. The novel combined treatment strategy might help to attenuate trauma-induced brain edema formation and neuronal damage as secondary effects of brain trauma.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas/tratamento farmacológico , Dexametasona/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/efeitos dos fármacos , Animais , Gasometria , Western Blotting , Ácidos Borônicos/farmacologia , Bortezomib , Edema Encefálico/tratamento farmacológico , Edema Encefálico/prevenção & controle , Lesões Encefálicas/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise Multivariada , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Pirazinas/farmacologia , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Glucocorticoides/metabolismo , Valores de Referência , Sensibilidade e Especificidade , Estatísticas não Paramétricas
15.
Differentiation ; 84(4): 294-304, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23023065

RESUMO

The blood-air barrier in the lung consists of the alveolar epithelium, the underlying capillary endothelium, their basement membranes and the interstitial space between the cell layers. Little is known about the interactions between the alveolar and the blood compartment. The aim of the present study was to gain first insights into the possible interplay between these two neighbored cell layers. We established an in vitro Transwell model of the alveolar epithelium based on human cell line H441 and investigated the influence of conditioned medium obtained from human lung endothelial cell line HPMEC-ST1.6R on the barrier properties of the H441 layers. As control for tissue specificity H441 layers were exposed to conditioned medium from human brain endothelial cell line hCMEC/D3. Addition of dexamethasone was necessary to obtain stable H441 cell layers. Moreover, dexamethasone increased expression of cell type I markers (caveolin-1, RAGE) and cell type II marker SP-B, whereas decreased the transepithelial electrical resistance (TEER) in a concentration dependent manner. Soluble factors obtained from the lung endothelial cell line increased the barrier significantly proven by TEER values and fluorescein permeability on the functional level and by the differential expression of tight junctional proteins on the molecular level. In contrast to this, soluble factors derived from brain endothelial cells weakened the barrier significantly. In conclusion, soluble factors from lung endothelial cells can strengthen the alveolar epithelium barrier in vitro, which suggests communication between endothelial and epithelial cells regulating the integrity of the blood-air barrier.


Assuntos
Barreira Alveolocapilar , Encéfalo/fisiologia , Células Endoteliais/fisiologia , Endotélio Vascular/fisiologia , Pulmão/fisiologia , Alvéolos Pulmonares/fisiologia , Caveolina 1/biossíntese , Linhagem Celular , Técnicas de Cocultura , Dexametasona/farmacologia , Impedância Elétrica , Humanos , Permeabilidade , Proteína B Associada a Surfactante Pulmonar/biossíntese , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Junções Íntimas/fisiologia
16.
Front Hum Neurosci ; 17: 1149449, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033910

RESUMO

Introduction: While central blood pressure (BP) has been recognized as a major indicator of left ventricular (LV) afterload, the reduction of central pressure decreases LV afterload and may prevent heart failure (HF) decompensation. Non-invasive transcutaneous vagus nerve stimulation (tVNS) was shown to improve cardiac function in HF patients. In this study, the relationship between active tVNS and reduction of central BP was investigated in patients with acute HF (AHF). Methods: The 22 patients hospitalized for AHF after initial stabilization (median 80 yrs, males 60%) were randomly assigned to active or sham group. For 1 h daily over 5 days, low-level transcutaneous electrical stimulation (LLTS) (20 Hz, 1 mA) was performed after attaching an ear clip to the tragus (active group) or the earlobe (sham control group). Before and after stimulation, central aortic systolic pressure (CASP), brachial systolic BP (SBP), diastolic BP (DBP) as well as heart rate (HR) were noninvasively measured. Results: No significant differences in baseline characteristics were observed between the active and sham groups. In the active group, CASP, SBP, DBP, and HR each decreased significantly after stimulation (all p < 0.05), whereas in the sham group, CASP, SBP, DBP, and HR each increased significantly after stimulation (all p < 0.05). All the changes in CASP, SBP, DBP and HR before and after stimulation were also significantly different between active and sham groups (all p < 0.01). There were no device-related side effects. Conclusion: In this study, the left tragus tVNS resulted in an acute afterload reduction in the elderly AHF patients. Non-invasive LLTS may be useful and safe for reducing afterload in AHF. Clinical trial registration: ClinicalTrials.gov, identifier UMIN000044121.

17.
Anesth Analg ; 114(2): 371-4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22025492

RESUMO

Renal failure is a common complication of critically ill patients. Colloids such as hydroxyethyl starch (HES), gelatin, or albumin are regularly used for intravascular volume resuscitation, but there are increasing reports about the nephrotoxic side effects of synthetic colloids in septic patients. Therefore, we investigated the influence of colloids (HES130/0.4 (Voluven®), gelatin (Gelafundin®), human albumin, and the crystalloid Sterofundin® ISO on cell viability of human proximal tubular (HK-2) cells. HK-2 cells were incubated with colloids (0.1%-4%) and with equivalent volumes of the crystalloid solution Sterofundin ISO. After 21 hours, cell viability of HK-2 cells was measured by EZ4U assay (dye XTT). Application of HES130/0.4 decreased cell viability significantly in a concentration-dependent manner (86.80% ± 10.79% by 0.5% HES down to 24.02% ± 4.27% by 4% HES). Human albumin (>1.25%) as well as gelatin (>1%) also showed deleterious effects on HK-2 cells. Interestingly, in lower concentrations, human albumin and the crystalloid solution Sterofundin ISO were cytoprotective in comparison with the NaCl control. In conclusion, synthetic and natural colloids showed a harmful impact on HK-2 cells in higher concentrations without any prior proinflammatory stimulus. HES130/0.4 exhibited the most distinctive harmful impact, whereas the application of crystalloid Sterofundin ISO revealed cytoprotective effects.


Assuntos
Albuminas/toxicidade , Gelatina/toxicidade , Derivados de Hidroxietil Amido/toxicidade , Soluções Isotônicas/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Substitutos do Plasma/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Coloides , Soluções Cristaloides , Citoproteção , Relação Dose-Resposta a Droga , Humanos , Túbulos Renais Proximais/patologia , Compostos Orgânicos/toxicidade , Fatores de Tempo
18.
Biomolecules ; 12(11)2022 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-36421731

RESUMO

Alzheimer's disease (AD), the most common cause of dementia in the elderly, is a neurodegenerative disorder associated with neurovascular dysfunction, cognitive decline, and the accumulation of amyloid ß peptide (Aß) in the brain and tau-related lesions in neurons termed neurofibrillary tangles (NFTs). Aß deposits and NFT formation are the central pathological hallmarks in AD brains, and the majority of AD cases have been shown to exhibit a complex combination of systemic comorbidities. While AD is the foremost common cause of dementia in the elderly, age-related hearing loss (ARHL) is the most predominant sensory deficit in the elderly. During aging, chronic inflammation and resulting endothelial dysfunction have been described and might be key contributors to AD; we discuss an intriguing possible link between inner ear strial microvascular pathology and blood-brain barrier pathology and present ARHL as a potentially modifiable and treatable risk factor for AD development. We present compelling evidence that ARHL might well be seen as an important risk factor in AD development: progressive hearing impairment, leading to social isolation, and its comorbidities, such as frailty, falls, and late-onset depression, link ARHL with cognitive decline and increased risk of dementia, rendering it tempting to speculate that ARHL might be a potential common molecular and pathological trigger for AD. Additionally, one could speculate that amyloid-beta might damage the blood-labyrinth barrier as it does to the blood-brain barrier, leading to ARHL pathology. Finally, there are options for the treatment of ARHL by targeted neurotrophic factor supplementation to the cochlea to improve cognitive outcomes; they can also prevent AD development and AD-related comorbidity in the future.


Assuntos
Doença de Alzheimer , Humanos , Idoso , Doença de Alzheimer/diagnóstico , Doença de Alzheimer/terapia , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Barreira Hematoencefálica/metabolismo , Neurônios/metabolismo
19.
J Pain ; 23(6): 967-980, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34974173

RESUMO

Blood nerve barrier disruption and edema are common in neuropathic pain as well as in complex regional pain syndrome (CRPS). MicroRNAs (miRNA) are epigenetic multitarget switches controlling neuronal and non-neuronal cells in pain. The miR-183 complex attenuates hyperexcitability in nociceptors, but additional non-neuronal effects via transcription factors could contribute as well. This study explored exosomal miR-183 in CRPS and murine neuropathy, its effect on the microvascular barrier via transcription factor FoxO1 and tight junction protein claudin-5, and its antihyperalgesic potential. Sciatic miR-183 decreased after CCI. Substitution with perineural miR-183 mimic attenuated mechanical hypersensitivity and restored blood nerve barrier function. In vitro, serum from CCI mice und CRPS patients weakened the microvascular barrier of murine cerebellar endothelial cells, increased active FoxO1 and reduced claudin-5, concomitant with a lack of exosomal miR-183 in CRPS patients. Cellular stress also compromised the microvascular barrier which was rescued either by miR-183 mimic via FoxO1 repression or by prior silencing of Foxo1. PERSPECTIVE: Low miR-183 leading to barrier impairment via FoxO1 and subsequent claudin-5 suppression is a new aspect in the pathophysiology of CRPS and neuropathic pain. This pathway might help untangle the wide symptomatic range of CRPS and nurture further research into miRNA mimics or FoxO1 inhibitors.


Assuntos
Síndromes da Dor Regional Complexa , Proteína Forkhead Box O1 , MicroRNAs , Neuralgia , Animais , Claudina-5/genética , Claudina-5/metabolismo , Síndromes da Dor Regional Complexa/genética , Síndromes da Dor Regional Complexa/metabolismo , Células Endoteliais/metabolismo , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Humanos , Camundongos , MicroRNAs/genética , Neuralgia/metabolismo
20.
Biomolecules ; 12(8)2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-36009007

RESUMO

Diabetes mellitus is a common disease affecting more than 537 million adults worldwide. The microvascular complications that occur during the course of the disease are widespread and affect a variety of organ systems in the body. Diabetic retinopathy is one of the most common long-term complications, which include, amongst others, endothelial dysfunction, and thus, alterations in the blood-retinal barrier (BRB). This particularly restrictive physiological barrier is important for maintaining the neuroretina as a privileged site in the body by controlling the inflow and outflow of fluid, nutrients, metabolic end products, ions, and proteins. In addition, people with diabetic retinopathy (DR) have been shown to be at increased risk for systemic vascular complications, including subclinical and clinical stroke, coronary heart disease, heart failure, and nephropathy. DR is, therefore, considered an independent predictor of heart failure. In the present review, the effects of diabetes on the retina, heart, and kidneys are described. In addition, a putative common microRNA signature in diabetic retinopathy, nephropathy, and heart failure is discussed, which may be used in the future as a biomarker to better monitor disease progression. Finally, the use of miRNA, targeted neurotrophin delivery, and nanoparticles as novel therapeutic strategies is highlighted.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Insuficiência Cardíaca , Nefropatias , MicroRNAs , Adulto , Barreira Hematorretiniana/metabolismo , Diabetes Mellitus/metabolismo , Retinopatia Diabética/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Nefropatias/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Crescimento Neural
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA