Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.675
1.
Int J Mol Med ; 54(1)2024 Jul.
Article En | MEDLINE | ID: mdl-38874017

In paraquat (PQ)­induced acute lung injury (ALI)/ acute respiratory distress syndrome, PQ disrupts endothelial cell function and vascular integrity, which leads to increased pulmonary leakage. Anthrahydroquinone­2,6­disulfonate (AH2QDS) is a reducing agent that attenuates the extent of renal injury and improves survival in PQ­intoxicated Sprague­Dawley (SD) rats. The present study aimed to explore the beneficial role of AH2QDS in PQ­induced ALI and its related mechanisms. A PQ­intoxicated ALI model was established using PQ gavage in SD rats. Human pulmonary microvascular endothelial cells (HPMECs) were challenged with PQ. Superoxide dismutase, malondialdehyde, reactive oxygen species and nitric oxide (NO) fluorescence were examined to detect the level of oxidative stress in HPMECs. The levels of TNF­α, IL­1ß and IL­6 were assessed using an ELISA. Transwell and Cell Counting Kit­8 assays were performed to detect the migration and proliferation of the cells. The pathological changes in lung tissues and blood vessels were examined by haematoxylin and eosin staining. Evans blue staining was used to detect pulmonary microvascular permeability. Western blotting was performed to detect target protein levels. Immunofluorescence and immunohistochemical staining were used to detect the expression levels of target proteins in HPMECs and lung tissues. AH2QDS inhibited inflammatory responses in lung tissues and HPMECs, and promoted the proliferation and migration of HPMECs. In addition, AH2QDS reduced pulmonary microvascular permeability by upregulating the levels of vascular endothelial­cadherin, zonula occludens­1 and CD31, thereby attenuating pathological changes in the lungs in rats. Finally, these effects may be related to the suppression of the phosphatidylinositol­3­kinase (PI3K)/protein kinase B (AKT)/endothelial­type NO synthase (eNOS) signalling pathway in endothelial cells. In conclusion, AH2QDS ameliorated PQ­induced ALI by improving alveolar endothelial barrier disruption via modulation of the PI3K/AKT/eNOS signalling pathway, which may be an effective candidate for the treatment of PQ­induced ALI.


Acute Lung Injury , Capillary Permeability , Lung , Nitric Oxide Synthase Type III , Paraquat , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Animals , Acute Lung Injury/metabolism , Acute Lung Injury/drug therapy , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Proto-Oncogene Proteins c-akt/metabolism , Nitric Oxide Synthase Type III/metabolism , Capillary Permeability/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Humans , Male , Signal Transduction/drug effects , Lung/pathology , Lung/metabolism , Lung/drug effects , Paraquat/adverse effects , Paraquat/toxicity , Rats , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Oxidative Stress/drug effects
2.
J Pathol ; 263(3): 347-359, 2024 Jul.
Article En | MEDLINE | ID: mdl-38734878

Vascular permeability is temporarily heightened during inflammation, but excessive inflammation-associated microvascular leakage can be detrimental, as evidenced in the inflamed lung. Formylated peptides regulate vascular leakage indirectly via formylated peptide receptor-1 (FPR1)-mediated recruitment and activation of neutrophils. Here we identify how the GTPase-activating protein ARAP3 protects against formylated peptide-induced microvascular permeability via endothelial cells and neutrophils. In vitro, Arap3-/- endothelial monolayers were characterised by enhanced formylated peptide-induced permeability due to upregulated endothelial FPR1 and enhanced vascular endothelial cadherin internalisation. In vivo, enhanced inflammation-associated microvascular leakage was observed in Arap3-/- mice. Leakage of plasma protein into the lungs of Arap3-/- mice increased within hours of formylated peptide administration. Adoptive transfer experiments indicated this was dependent upon ARAP3 deficiency in both immune and non-immune cells. Bronchoalveolar lavages of formylated peptide-challenged Arap3-/- mice contained neutrophil extracellular traps (NETs). Pharmacological inhibition of NET formation abrogated excessive microvascular leakage, indicating a critical function of NETs in this context. The observation that Arap3-/- mice developed more severe influenza suggests these findings are pertinent to pathological situations characterised by abundant formylated peptides. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Capillary Permeability , Endothelial Cells , Mice, Knockout , Neutrophils , Animals , Neutrophils/metabolism , Capillary Permeability/drug effects , Humans , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Mice , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Mice, Inbred C57BL , Extracellular Traps/metabolism , Lung/metabolism , Lung/pathology , Lung/blood supply
3.
Sci Rep ; 14(1): 10477, 2024 05 07.
Article En | MEDLINE | ID: mdl-38714743

Endothelial glycocalyx (eGC) covers the inner surface of the vessels and plays a role in vascular homeostasis. Syndecan is considered the "backbone" of this structure. Several studies have shown eGC shedding in sepsis and its involvement in organ dysfunction. Matrix metalloproteinases (MMP) contribute to eGC shedding through their ability for syndecan-1 cleavage. This study aimed to investigate if doxycycline, a potent MMP inhibitor, could protect against eGC shedding in lipopolysaccharide (LPS)-induced sepsis and if it could interrupt the vascular hyperpermeability, neutrophil transmigration, and microvascular impairment. Rats that received pretreatment with doxycycline before LPS displayed ultrastructural preservation of the eGC observed using transmission electronic microscopy of the lung and heart. In addition, these animals exhibited lower serum syndecan-1 levels, a biomarker of eGC injury, and lower perfused boundary region (PBR) in the mesenteric video capillaroscopy, which is inversely related to the eGC thickness compared with rats that only received LPS. Furthermore, this study revealed that doxycycline decreased sepsis-related vascular hyperpermeability in the lung and heart, reduced neutrophil transmigration in the peritoneal lavage and inside the lungs, and improved some microvascular parameters. These findings suggest that doxycycline protects against LPS-induced eGC shedding, and it could reduce vascular hyperpermeability, neutrophils transmigration, and microvascular impairment.


Doxycycline , Glycocalyx , Lipopolysaccharides , Sepsis , Glycocalyx/metabolism , Glycocalyx/drug effects , Animals , Sepsis/drug therapy , Sepsis/metabolism , Doxycycline/pharmacology , Rats , Male , Capillary Permeability/drug effects , Lung/pathology , Lung/metabolism , Lung/drug effects , Syndecan-1/metabolism , Rats, Wistar , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Neutrophils/metabolism , Neutrophils/drug effects , Matrix Metalloproteinase Inhibitors/pharmacology
4.
PLoS One ; 19(5): e0302628, 2024.
Article En | MEDLINE | ID: mdl-38723000

Blood vessels permit the selective passage of molecules and immune cells between tissues and circulation. Uncontrolled inflammatory responses from an infection can increase vascular permeability and edema, which can occasionally lead to fatal organ failure. We identified mexenone as a vascular permeability blocker by testing 2,910 compounds in the Clinically Applied Compound Library using the lipopolysaccharide (LPS)-induced vascular permeability assay. Mexenone suppressed the LPS-induced downregulation of junctional proteins and phosphorylation of VE-cadherin in Bovine Aortic Endothelial Cells (BAECs). The injection of mexenone 1 hr before LPS administration completely blocked LPS-induced lung vascular permeability and acute lung injury in mice after 18hr. Our results suggest that mexenone-induced endothelial cell (EC) barrier stabilization could be effective in treating sepsis patients.


Endothelial Cells , Lipopolysaccharides , Sepsis , Animals , Sepsis/drug therapy , Sepsis/chemically induced , Sepsis/metabolism , Mice , Cattle , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Capillary Permeability/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Acute Lung Injury/prevention & control , Male , Cadherins/metabolism , Mice, Inbred C57BL , Antigens, CD/metabolism
5.
Crit Care ; 28(1): 136, 2024 04 23.
Article En | MEDLINE | ID: mdl-38654391

BACKGROUND: In acute respiratory distress syndrome (ARDS), respiratory drive often differs among patients with similar clinical characteristics. Readily observable factors like acid-base state, oxygenation, mechanics, and sedation depth do not fully explain drive heterogeneity. This study evaluated the relationship of systemic inflammation and vascular permeability markers with respiratory drive and clinical outcomes in ARDS. METHODS: ARDS patients enrolled in the multicenter EPVent-2 trial with requisite data and plasma biomarkers were included. Neuromuscular blockade recipients were excluded. Respiratory drive was measured as PES0.1, the change in esophageal pressure during the first 0.1 s of inspiratory effort. Plasma angiopoietin-2, interleukin-6, and interleukin-8 were measured concomitantly, and 60-day clinical outcomes evaluated. RESULTS: 54.8% of 124 included patients had detectable respiratory drive (PES0.1 range of 0-5.1 cm H2O). Angiopoietin-2 and interleukin-8, but not interleukin-6, were associated with respiratory drive independently of acid-base, oxygenation, respiratory mechanics, and sedation depth. Sedation depth was not significantly associated with PES0.1 in an unadjusted model, or after adjusting for mechanics and chemoreceptor input. However, upon adding angiopoietin-2, interleukin-6, or interleukin-8 to models, lighter sedation was significantly associated with higher PES0.1. Risk of death was less with moderate drive (PES0.1 of 0.5-2.9 cm H2O) compared to either lower drive (hazard ratio 1.58, 95% CI 0.82-3.05) or higher drive (2.63, 95% CI 1.21-5.70) (p = 0.049). CONCLUSIONS: Among patients with ARDS, systemic inflammatory and vascular permeability markers were independently associated with higher respiratory drive. The heterogeneous response of respiratory drive to varying sedation depth may be explained in part by differences in inflammation and vascular permeability.


Biomarkers , Capillary Permeability , Inflammation , Respiratory Distress Syndrome , Humans , Respiratory Distress Syndrome/physiopathology , Respiratory Distress Syndrome/blood , Male , Female , Middle Aged , Capillary Permeability/physiology , Capillary Permeability/drug effects , Inflammation/physiopathology , Inflammation/blood , Aged , Biomarkers/blood , Biomarkers/analysis , Angiopoietin-2/blood , Angiopoietin-2/analysis , Interleukin-8/blood , Interleukin-8/analysis , Interleukin-6/blood , Interleukin-6/analysis , Respiratory Mechanics/physiology
6.
Arch Toxicol ; 98(7): 2247-2259, 2024 Jul.
Article En | MEDLINE | ID: mdl-38635053

3-Bromofluoranthene (3-BrFlu) is the secondary metabolite of fluoranthene, which is classified as a polycyclic aromatic hydrocarbon, through bromination and exists in the fine particulate matter of air pollutants. Endothelial dysfunction plays a critical role in the pathogenesis of cardiovascular and vascular diseases. Little is known about the molecular mechanism of 3-BrFlu on endothelial dysfunction in vivo and in vitro assay. In the present study, 3-BrFlu included concentration-dependent changes in ectopic angiogenesis of the sub-intestinal vein and dilation of the dorsal aorta in zebrafish. Disruption of vascular endothelial integrity and up-regulation of vascular endothelial permeability were also induced by 3-BrFlu in a concentration-dependent manner through pro-inflammatory responses in vascular endothelial cells, namely, SVEC4-10 cells. Generation of pro-inflammatory mediator PGE2 was induced by 3-BrFlu through COX2 expression. Expression of COX2 and generation of pro-inflammatory cytokines, including TNFα and IL-6, were induced by 3-BrFlu through phosphorylation of NF-κB p65, which was mediated by phosphorylation of MAPK, including p38 MAPK, ERK and JNK. Furthermore, generation of intracellular ROS was induced by 3-BrFlu, which is associated with the down-regulated activities of the antioxidant enzyme (AOE), including SOD and catalase. We also found that 3-BrFlu up-regulated expression of the AOE and HO-1 induced by 3-BrFlu through Nrf-2 expression. However, the 3-BrFlu-induced upregulation of AOE and HO-1 expression could not be revised the responses of vascular endothelial dysfunction. In conclusion, 3-BrFlu is a hazardous substance that results in vascular endothelial dysfunction through the MAPK-mediated-NFκB pro-inflammatory pathway and intracellular ROS generation.


Endothelium, Vascular , Fluorenes , NF-kappa B , Reactive Oxygen Species , Zebrafish , Animals , Reactive Oxygen Species/metabolism , Fluorenes/toxicity , NF-kappa B/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cell Line , Cyclooxygenase 2/metabolism , Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Inflammation/chemically induced , Inflammation/metabolism , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Capillary Permeability/drug effects
7.
Pflugers Arch ; 476(6): 993-1005, 2024 Jun.
Article En | MEDLINE | ID: mdl-38438679

Increase in transendothelial water permeability is an essential etiological factor in a variety of diseases like edema and shock. Despite the high clinical relevance, there has been no precise method to detect transendothelial water flow until now. The deuterium oxide (D2O) dilution method, already established for measuring transepithelial water transport, was used to precisely determine the transendothelial water permeability. It detected appropriate transendothelial water flow induced by different hydrostatic forces. This was shown in four different endothelial cell types. The general experimental setup was verified by gravimetry and absorbance spectroscopy. Determination of transendothelial electrical resistance (TEER) and immunocytochemical staining for proteins of the cell-cell contacts were performed to ensure that no damage to the endothelium occurred because of the measurements. Furthermore, endothelial barrier function was modulated. Measurement of transendothelial water flux was verified by measuring the TEER, the apparent permeability coefficient and the electrical capacity. The barrier-promoting substances cyclic adenosine monophosphate and iloprost reduced TEER and electrical capacity and increased permeability. This was accompanied by a reduced transendothelial water flux. In contrast, the barrier-damaging substances thrombin, histamine and bradykinin reduced TEER and electrical capacity, but increased permeability. Here, an increased water flow was shown. This newly established in vitro method for direct measurement of transendothelial water permeability was verified as a highly precise technique in various assays. The use of patient-specific endothelial cells enables individualized precision medicine in the context of basic edema research, for example regarding the development of barrier-protective pharmaceuticals.


Deuterium Oxide , Deuterium Oxide/metabolism , Humans , Electric Impedance , Water/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Permeability , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/drug effects , Capillary Permeability/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects
8.
Burns ; 50(4): 913-923, 2024 May.
Article En | MEDLINE | ID: mdl-38267288

BACKGROUND: Severe burns are devastating injuries with significant immune dysfunction and result in substantial mortality and morbidity due to sepsis induced organ failure. Acute lung injury is the most common type of organ injury in sepsis, however, the mechanisms of which are poorly understood and effective therapeutic measures are limited. This study is aimed to investigate the effect of a small Guanosine triphosphatase (GTPase), Adenosine diphosphate ribosylation factor 6 (ARF6), on burn sepsis induced lung injury, and discuss the possible mechanisms. METHODS: Burn sepsis was established in male C57BL/6 mice. Mice were anesthetised by intramuscular injection of ketamine and xylazine hydrochloride, then 30% TBSA full thickness burn followed by sub-eschar injection of lipopolysaccharide. Animals were treated with intraperitoneal injection of a small molecule inhibitor of ARF6: NAV-2729, or vehicle, right after the burn and sepsis stimuli were inflicted. Lung tissues were harvested for histopathological observation and the acute lung injury scores were calculated. Organ permeability, Vascular Endothelial Cadherin (VE-cadherin) expression, inflammatory cytokine levels and myeloperoxidase activity in lung tissues were detected. Rat pulmonary microvascular endothelial cells (PMVECs) were stimulated by burn sepsis serum with or without 10 µM NAV-2729. The ARF6 activation, VE-cadherin expression, inflammasome activity, adapter protein apoptosis speck-like protein containing a caspase recruiting domain (ASC) specks and cytokines secretion were determined. Student's t test was used for comparison between two groups. Multiple comparisons among groups were performed by using analysis of variance, with Tukey's test for the post hoc test. RESULTS: NAV-2729 treatment attenuated burn sepsis induced lung injury and promoted survival of burn septic mice by preserving VE-cadherin expression in endothelial cell adherent junction and limited vascular hyperpermeability in lung tissues. Moreover, inflammatory cytokine expression and inflammatory injury in lung tissues were alleviated. Mechanistically, NAV-2729 enhanced vascular integrity by inhibiting ARF6 activation and restoring VE-cadherin expression in PMVECs. In addition, NAV-2729 inhibited ARF6-dependent phagocytosis of ASC specks, thus preventing inflammation propagation mediated by cell-to-cell transmission of ASC specks. CONCLUSIONS: ARF6 inhibition preserved vascular integrity by restoring expression of VE-cadherin and suppressed the spread of inflammation by affecting phagocytosis of ASC specks, thus protected against sepsis induced lung injury and improve survival of burn septic animals. The findings of this study implied potential therapeutics by which ARF6 inhibition can protect lung function from septic induced lung injury and improve outcomes in burn sepsis.


ADP-Ribosylation Factor 6 , Acute Lung Injury , Burns , Cadherins , Inflammasomes , Mice, Inbred C57BL , Sepsis , Animals , Burns/complications , Burns/metabolism , Sepsis/complications , Sepsis/metabolism , Mice , Inflammasomes/metabolism , Inflammasomes/drug effects , Cadherins/metabolism , Male , Acute Lung Injury/prevention & control , Acute Lung Injury/metabolism , Acute Lung Injury/etiology , Antigens, CD/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Peroxidase/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Capillary Permeability/drug effects , Rats , Disease Models, Animal , Cytokines/metabolism
9.
J Pain ; 25(6): 104462, 2024 Jun.
Article En | MEDLINE | ID: mdl-38211844

Oxaliplatin, a platinum-based anticancer drug, is associated with peripheral neuropathy (oxaliplatin-induced peripheral neuropathy, OIPN), which can lead to worsening of quality of life and treatment interruption. The endothelial glycocalyx, a fragile carbohydrate-rich layer covering the luminal surface of endothelial cells, acts as an endothelial gatekeeper and has been suggested to protect nerves, astrocytes, and other cells from toxins and substances released from the capillary vessels. Mechanisms underlying OIPN and the role of the glycocalyx remain unclear. This study aimed to define changes in the three-dimensional ultrastructure of capillary endothelial glycocalyx near nerve fibers in the hind paws of mice with OIPN. The mouse model of OPIN revealed disruption of the endothelial glycocalyx in the peripheral nerve compartment, accompanied by vascular permeability, edema, and damage to the peripheral nerves. To investigate the potential treatment interventions, nafamostat mesilate, a glycocalyx protective agent was used in tumor-bearing male mice. Nafamostat mesilate suppressed mechanical allodynia associated with neuropathy. It also prevented intra-epidermal nerve fiber loss and improved vascular permeability in the peripheral paws. The disruption of endothelial glycocalyx in the capillaries that lie within peripheral nerve bundles is a novel finding in OPIN. Furthermore, these findings point toward the potential of a new treatment strategy targeting endothelial glycocalyx to prevent vascular injury as an effective treatment of neuropathy as well as of many other diseases. PERSPECTIVE: OIPN damages the endothelial glycocalyx in the peripheral capillaries, increasing vascular permeability. In order to prevent OIPN, this work offers a novel therapy approach that targets endothelial glycocalyx.


Antineoplastic Agents , Glycocalyx , Oxaliplatin , Animals , Glycocalyx/drug effects , Glycocalyx/metabolism , Glycocalyx/pathology , Oxaliplatin/toxicity , Mice , Male , Antineoplastic Agents/pharmacology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Capillaries/drug effects , Capillaries/pathology , Disease Models, Animal , Hyperalgesia/chemically induced , Hyperalgesia/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Mice, Inbred C57BL
10.
Environ Sci Technol ; 57(30): 10940-10950, 2023 08 01.
Article En | MEDLINE | ID: mdl-37467138

Urban stormwater runoff frequently contains the car tire transformation product 6PPD-quinone, which is highly toxic to juvenile and adult coho salmon (Onchorychus kisutch). However, it is currently unclear if embryonic stages are impacted. We addressed this by exposing developing coho salmon embryos starting at the eyed stage to three concentrations of 6PPD-quinone twice weekly until hatch. Impacts on survival and growth were assessed. Further, whole-transcriptome sequencing was performed on recently hatched alevin to address the potential mechanism of 6PPD-quinone-induced toxicity. Acute mortality was not elicited in developing coho salmon embryos at environmentally measured concentrations lethal to juveniles and adults, however, growth was inhibited. Immediately after hatching, coho salmon were sensitive to 6PPD-quinone mortality, implicating a large window of juvenile vulnerability prior to smoltification. Molecularly, 6PPD-quinone induced dose-dependent effects that implicated broad dysregulation of genomic pathways governing cell-cell contacts and endothelial permeability. These pathways are consistent with previous observations of macromolecule accumulation in the brains of coho salmon exposed to 6PPD-quinone, implicating blood-brain barrier disruption as a potential pathway for toxicity. Overall, our data suggests that developing coho salmon exposed to 6PPD-quinone are at risk for adverse health events upon hatching while indicating potential mechanism(s) of action for this highly toxic chemical.


Benzoquinones , Blood-Brain Barrier , Capillary Permeability , Oncorhynchus kisutch , Phenylenediamines , Water Pollutants, Chemical , Animals , Capillary Permeability/drug effects , Capillary Permeability/genetics , Oncorhynchus kisutch/metabolism , Water Pollutants, Chemical/toxicity , Water Pollutants, Chemical/metabolism , Phenylenediamines/analysis , Phenylenediamines/metabolism , Phenylenediamines/toxicity , Benzoquinones/analysis , Benzoquinones/metabolism , Benzoquinones/toxicity , Transcription, Genetic/drug effects , Blood-Brain Barrier/drug effects , Biotransformation
11.
Front Endocrinol (Lausanne) ; 13: 973058, 2022.
Article En | MEDLINE | ID: mdl-36060954

Protein kinase C (PKC) is a family of serine/threonine protein kinases, the activation of which plays an important role in the development of diabetic microvascular complications. The activation of PKC under high-glucose conditions stimulates redox reactions and leads to an accumulation of redox stress. As a result, various types of cells in the microvasculature are influenced, leading to changes in blood flow, microvascular permeability, extracellular matrix accumulation, basement thickening and angiogenesis. Structural and functional disorders further exacerbate diabetic microvascular complications. Here, we review the roles of PKC in the development of diabetic microvascular complications, presenting evidence from experiments and clinical trials.


Diabetic Angiopathies , Protein Kinase C , Capillary Permeability/drug effects , Capillary Permeability/physiology , Diabetes Mellitus , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , Humans , Microvessels/drug effects , Microvessels/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Protein Kinase C/adverse effects , Protein Kinase C/metabolism
12.
Cells ; 11(17)2022 08 30.
Article En | MEDLINE | ID: mdl-36078114

The aim of this study was to evaluate the effect of acute aldosterone (ALDO) administration on the vascular permeability of skin. ALDO was injected intradermally into rats, and vascular permeability was measured. Eplerenone (EPL), a selective mineralocorticoid receptor (MR) antagonist, was used. Skin biopsies were carried out for immunohistochemical (IHC) staining, and polymerase chain reactions were performed to analyze the expression of MR, 11ß-hydroxysteroid dehydrogenase type 2, von Willebrand factor (vWF), vascular endothelial growth factor (VEGF), and zonula occludens 1. Our study showed the presence of MR in the rat skin vasculature for the first time. It was found that ALDO injection resulted in a more than 30% increase in vascular permeability and enhanced the endothelial exocytosis of vWF. The effect of ALDO diminished after EPL administration. An accumulation of vWF and a reduction in VEGF IHC staining were observed following chronic EPL administration. No effect of ALDO or EPL on the mRNA expression of the studied genes or skin structure was observed. The results suggest that ALDO increases vascular permeability in the skin via an MR-dependent mechanism. This effect of ALDO on skin microcirculation may have important therapeutic implications for diseases characterized by increased levels of ALDO and coexisting skin microangiopathy.


Aldosterone , Capillary Permeability , Aldosterone/metabolism , Aldosterone/pharmacokinetics , Aldosterone/pharmacology , Animals , Capillary Permeability/drug effects , Eplerenone , Mineralocorticoid Receptor Antagonists , Rats , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/metabolism
13.
Front Immunol ; 13: 810376, 2022.
Article En | MEDLINE | ID: mdl-35185902

Exacerbated inflammatory response and altered vascular function are hallmarks of dengue disease. Reactive oxygen species (ROS) production has been associated to endothelial barrier disturbance and microvascular alteration in distinct pathological conditions. Increased ROS has been reported in in vitro models of dengue virus (DENV) infection, but its impact for endothelial cell physiology had not been fully investigated. Our group had previously demonstrated that infection of human brain microvascular endothelial cells (HBMEC) with DENV results in the activation of RNA sensors and production of proinflammatory cytokines, which culminate in cell death and endothelial permeability. Here, we evaluated the role of mitochondrial function and NADPH oxidase (NOX) activation for ROS generation in HBMEC infected by DENV and investigated whether altered cellular physiology could be a consequence of virus-induced oxidative stress. DENV-infected HBMECs showed a decrease in the maximal respiratory capacity and altered membrane potential, indicating functional mitochondrial alteration, what might be related to mtROS production. Indeed, mtROS was detected at later time points after infection. Specific inhibition of mtROS diminished virus replication, cell death, and endothelial permeability, but did not affect cytokine production. On the other hand, inhibition of NOX-associated ROS production decreased virus replication and cell death, as well as the secretion of inflammatory cytokines, including IL-6, IL-8, and CCL5. These results demonstrated that DENV replication in endothelial cells induces ROS production by different pathways, which impacts biological functions that might be relevant for dengue pathogenesis. Those data also indicate oxidative stress events as relevant therapeutical targets to avoid vascular permeability, inflammation, and neuroinvasion during DENV infection.


Antiviral Agents/pharmacology , Dengue Virus/drug effects , Endothelium, Vascular/virology , Reactive Oxygen Species/metabolism , Virus Replication/drug effects , Capillary Permeability/drug effects , Cell Line , Cells, Cultured , Cytokines/metabolism , Dengue/immunology , Dengue/virology , Dengue Virus/genetics , Endothelium, Vascular/drug effects , Humans , Oxidative Stress/drug effects
14.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article En | MEDLINE | ID: mdl-35054938

Midazolam is an anesthetic widely used for anxiolysis and sedation; however, to date, a possible role for midazolam in diabetic kidney disease remains unknown. Here, we investigated the effect of midazolam on hyperglycemia-induced glomerular endothelial dysfunction and elucidated its mechanism of action in kidneys of diabetic mice and human glomerular microvascular endothelial cells (HGECs). We found that, in diabetic mice, subcutaneous midazolam treatment for 6 weeks attenuated hyperglycemia-induced elevation in urine albumin/creatinine ratios. It also ameliorated hyperglycemia-induced adherens junction disruption and subsequent microvascular leakage in glomeruli of diabetic mice. In HGECs, midazolam suppressed high glucose-induced vascular endothelial-cadherin disruption and endothelial cell permeability via inhibition of intracellular Ca2+ elevation and subsequent generation of reactive oxygen species (ROS) and transglutaminase 2 (TGase2) activation. Notably, midazolam also suppressed hyperglycemia-induced ROS generation and TGase2 activation in glomeruli of diabetic mice and markedly improved pathological alterations in glomerular ultrastructure in these animals. Analysis of kidneys from diabetic Tgm2-/- mice further revealed that TGase2 played a critical role in microvascular leakage. Overall, our findings indicate that midazolam ameliorates hyperglycemia-induced glomerular endothelial dysfunction by inhibiting ROS-mediated activation of TGase2.


Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Endothelial Cells/metabolism , Hyperglycemia/complications , Kidney Glomerulus/metabolism , Midazolam/pharmacology , Protein Glutamine gamma Glutamyltransferase 2/antagonists & inhibitors , Animals , Biomarkers , Calcium/metabolism , Capillary Permeability/drug effects , Diabetes Mellitus, Experimental , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/pathology , Disease Management , Disease Models, Animal , Disease Susceptibility , Endothelial Cells/drug effects , Endothelial Cells/pathology , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Mice , Mice, Knockout , Models, Biological , Reactive Oxygen Species/metabolism
15.
Bioengineered ; 13(2): 3680-3693, 2022 02.
Article En | MEDLINE | ID: mdl-35081868

Sepsis is mainly characterized by severe inflammation triggered by infection, and sepsis-associated encephalopathy (SAE) is defined as brain damage caused by sepsis. Disruption of the blood-brain barrier (BBB) triggered by injured brain microvascular endothelial cells (BMECs) and damaged tight junction (TJ) structure is closely associated with the pathogenesis of SAE. The present research proposed to evaluate the potential therapeutic effects of Mirtazapine, a central presynaptic α2 receptor antagonist, on LPS-induced BBB disruption. The mice were administered with normal saline and 10 mg/kg Mirtazapine for 8 consecutive days, and from day 6, the experiment group of mice received LPS for 2 days to induce SAE. We found that the increased BBB permeability, elevated concentrations of inflammatory factors in brain tissues, and downregulated zonula occludens -1 (ZO-1) were observed in LPS-stimulated mice, all of which were reversed by 10 mg/kg Mirtazapine. In the in vitro assay, bEnd.3 brain endothelial cells were treated with 1 µM LPS in the absence or presence of Mirtazapine (25, 50 µM). We found that LPS-treated cells had significantly declined transendothelial electrical resistance (TEER), increased monolayer permeability, elevated production of inflammatory factors, and downregulated ZO-1. However, 25 and 50 µM Mirtazapine ameliorated all these LPS- induced aberrations. Mirtazapine also mitigated the decreased level of NF-E2-related factor 2 (Nrf2) in LPS-challenged endothelial cells. The protective effect of Mirtazapine on endothelial permeability against LPS was significantly abolished by the knockdown of Nrf2. Collectively, we concluded that Mirtazapine exerted protective effects on LPS-induced endothelial cells hyperpermeability by upregulating Nrf2.


Blood-Brain Barrier/metabolism , Capillary Permeability/drug effects , Lipopolysaccharides/toxicity , Mirtazapine/pharmacology , Sepsis-Associated Encephalopathy , Tight Junctions/metabolism , Animals , Male , Mice , Sepsis-Associated Encephalopathy/chemically induced , Sepsis-Associated Encephalopathy/drug therapy , Sepsis-Associated Encephalopathy/metabolism
16.
Microvasc Res ; 139: 104266, 2022 01.
Article En | MEDLINE | ID: mdl-34688627

The no-reflow phenomenon induced by ischemia-reperfusion (I/R) injury seriously limits the therapeutic value of coronary recanalization and leads to a poor prognosis. Previous studies have shown that luteolin (LUT) is a vasoprotective factor. However, whether LUT can be used to prevent the no-reflow phenomenon remains unknown. Positron emission tomography perfusion imaging, performed to detect the effects of LUT on the no-reflow phenomenon in vivo, revealed that LUT treatment was able to reduce the no-reflow area in rat I/R models. In vitro, LUT was shown to reduce the hypoxia-reoxygenation injury-induced endothelial permeability and apoptosis. The levels of malondialdehyde, reactive oxygen species and NADPH were also measured and the results indicated that LUT could inhibit the oxidative stress. Western blot analysis revealed that LUT protected endothelial cells from I/R injury by regulating the Wnt/ß-catenin pathway. Overall, we concluded that the use of LUT to minimize I/R induced microvascular damage is a feasible strategy to prevent the no-reflow phenomenon.


Coronary Circulation/drug effects , Coronary Vessels/drug effects , Endothelial Cells/drug effects , Luteolin/pharmacology , Myocardial Reperfusion Injury/prevention & control , No-Reflow Phenomenon/prevention & control , Wnt Signaling Pathway/drug effects , Animals , Apoptosis/drug effects , Capillary Permeability/drug effects , Cells, Cultured , Coronary Vessels/diagnostic imaging , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Myocardial Perfusion Imaging , Myocardial Reperfusion Injury/diagnostic imaging , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , No-Reflow Phenomenon/diagnostic imaging , No-Reflow Phenomenon/metabolism , No-Reflow Phenomenon/physiopathology , Oxidative Stress/drug effects , Positron-Emission Tomography , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
17.
J Allergy Clin Immunol ; 149(1): 275-291, 2022 01.
Article En | MEDLINE | ID: mdl-34111449

BACKGROUND: P17, a peptide isolated from Tetramorium bicarinatum ant venom, is known to induce an alternative phenotype of human monocyte-derived macrophages via activation of an unknown G protein-coupled receptor (GPCR). OBJECTIVE: We sought to investigate the mechanism of action and the immunomodulatory effects of P17 mediated through MRGPRX2 (Mas-related G protein-coupled receptor X2). METHODS: To identify the GPCR for P17, we screened 314 GPCRs. Upon identification of MRGPRX2, a battery of in silico, in vitro, ex vivo, and in vivo assays along with the receptor mutation studies were performed. In particular, to investigate the immunomodulatory actions, we used ß-hexosaminidase release assay, cytokine releases, quantification of mRNA expression, cell migration and differentiation assays, immunohistochemical labeling, hematoxylin and eosin, and immunofluorescence staining. RESULTS: P17 activated MRGPRX2 in a dose-dependent manner in ß-arrestin recruitment assay. In LAD2 cells, P17 induced calcium and ß-hexosaminidase release. Quercetin- and short hairpin RNA-mediated knockdown of MRGPRX2 reduced P17-evoked ß-hexosaminidase release. In silico and in vitro mutagenesis studies showed that residue Lys8 of P17 formed a cation-π interaction with the Phe172 of MRGPRX2 and [Ala8]P17 lost its activity partially. P17 activated LAD2 cells to recruit THP-1 and human monocytes in Transwell migration assay, whereas MRGPRX2-impaired LAD2 cells cannot. In addition, P17-treated LAD2 cells stimulated differentiation of THP-1 and human monocytes, as indicated by the enhanced expression of macrophage markers cluster of differentiation 11b and TNF-α by quantitative RT-PCR. Immunohistochemical and immunofluorescent staining suggested monocyte recruitment in mice ears injected with P17. CONCLUSIONS: Our data provide novel structural information regarding the interaction of P17 with MRGPRX2 and intracellular pathways for its immunomodulatory action.


Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites , Capillary Permeability/drug effects , Cell Differentiation/drug effects , Cell Line , Chemotaxis/drug effects , Cricetulus , Cytokines/metabolism , Edema/immunology , Edema/metabolism , Evans Blue/metabolism , Gene Silencing , Humans , Male , Mast Cells/drug effects , Mice, Inbred C57BL , Models, Molecular , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Receptors, G-Protein-Coupled/genetics
18.
Microvasc Res ; 139: 104265, 2022 01.
Article En | MEDLINE | ID: mdl-34662588

The investigation aimed to evaluate the effects of Mcc950, an inhibitor of the NLRP3 inflammasome, on diabetic retinopathy (DR) mice. The general physiological condition of each group of mice was recorded. Retinal blood vessels were stained for observation of the density of blood vessels, and retinas were used for further morphological examination and fluorescent staining after the intravitreal injection of Mcc950. Mcc950 partially reversed hyperglycemia-induced vascular damage and had reduced histological changes compared to DR mice. IL-1ß production in mice retinas in the diabetic model (DM) group increased, but pretreatment with Mcc950 significantly reversed these changes. Additionally, Mcc950 engineered reduced FITC dextran extravasation and vascular leakage. Therefore, it played an apparent protective role in DR and could be a new treatment strategy for DR.


Anti-Inflammatory Agents/pharmacology , Diabetic Retinopathy/prevention & control , Furans/pharmacology , Indenes/pharmacology , Inflammasomes/antagonists & inhibitors , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Retinal Neovascularization/prevention & control , Retinal Vessels/drug effects , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Blood Glucose/metabolism , Capillary Permeability/drug effects , Diabetic Retinopathy/immunology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Disease Models, Animal , Disease Progression , Furans/administration & dosage , Indenes/administration & dosage , Inflammasomes/metabolism , Intravitreal Injections , Male , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Retinal Neovascularization/immunology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Vessels/immunology , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Sulfonamides/administration & dosage
19.
Inflammation ; 45(1): 116-128, 2022 Feb.
Article En | MEDLINE | ID: mdl-34494203

Angioedema is characterized by swelling of the skin or mucous membranes. Overproduction of the vasodilator bradykinin (BK) is an important contributor to the disease pathology, which causes rapid increase in vascular permeability. BK formation on endothelial cells results from high molecular weight kininogen (HK) interacting with gC1qR, the receptor for the globular heads of C1q, the first component of the classical pathway of complement. Endothelial cells are sensitive to blood-flow-induced shear stress and it has been shown that shear stress can modulate gC1qR expression. This study aimed to determine the following: (1) how BK or angioedema patients' (HAE) plasma affected endothelial cell permeability and gC1qR expression under shear stress, and (2) if monoclonal antibody (mAb) 74.5.2, which recognizes the HK binding site on gC1qR, had an inhibitory effect in HK binding to endothelial cells. Human dermal microvascular endothelial cells (HDMECs) grown on Transwell inserts were exposed to shear stress in the presence of HAE patients' plasma. Endothelial cell permeability was measured using FITC-conjugated bovine serum albumin. gC1qR expression and HK binding to endothelial cell surface was measured using solid-phase ELISA. Cell morphology was quantified using immunofluorescence microscopy. The results demonstrated that BK at 1 µg/mL, but not HAE patients' plasma and/or shear stress, caused significant increases in HDMEC permeability. The mAb 74.5.2 could effectively inhibit HK binding to recombinant gC1qR, and reduce HAE patients' plasma-induced HDMEC permeability change. These results suggested that monoclonal antibody to gC1qR, i.e., 74.5.2, could be potentially used as an effective therapeutic reagent to prevent angioedema.


Angioedema/drug therapy , Antibodies, Monoclonal/pharmacology , Bradykinin/metabolism , Capillary Permeability/drug effects , Cardiovascular Agents/pharmacology , Carrier Proteins/immunology , Endothelial Cells/drug effects , Mitochondrial Proteins/immunology , Angioedema/immunology , Angioedema/metabolism , Angioedema/physiopathology , Antibodies, Monoclonal/therapeutic use , Biomarkers/metabolism , Capillary Permeability/immunology , Cardiovascular Agents/therapeutic use , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Permeability/drug effects , Shear Strength/drug effects
20.
J Neurosci ; 42(3): 362-376, 2022 01 19.
Article En | MEDLINE | ID: mdl-34819341

Multifaceted microglial functions in the developing brain, such as promoting the differentiation of neural progenitors and contributing to the positioning and survival of neurons, have been progressively revealed. Although previous studies have noted the relationship between vascular endothelial cells and microglia in the developing brain, little attention has been given to the importance of pericytes, the mural cells surrounding endothelial cells. In this study, we attempted to dissect the role of pericytes in microglial distribution and function in developing mouse brains. Our immunohistochemical analysis showed that approximately half of the microglia attached to capillaries in the cerebral walls. Notably, a magnified observation of the position of microglia, vascular endothelial cells and pericytes demonstrated that microglia were preferentially associated with pericytes that covered 79.8% of the total capillary surface area. Through in vivo pericyte depletion induced by the intraventricular administration of a neutralizing antibody against platelet-derived growth factor receptor (PDGFR)ß (clone APB5), we found that microglial density was markedly decreased compared with that in control antibody-treated brains because of their low proliferative capacity. Moreover, in vitro coculture of isolated CD11b+ microglia and NG2+PDGFRα- cells, which are mostly composed of pericytes, from parenchymal cells indicated that pericytes promote microglial proliferation via the production of soluble factors. Furthermore, pericyte depletion by APB5 treatment resulted in a failure of microglia to promote the differentiation of neural stem cells into intermediate progenitors. Taken together, our findings suggest that pericytes facilitate microglial homeostasis in the developing brains, thereby indirectly supporting microglial effects on neural progenitors.SIGNIFICANCE STATEMENT This study highlights the novel effect of pericytes on microglia in the developing mouse brain. Through multiple analyses using an in vivo pericyte depletion mouse model and an in vitro coculture study of isolated pericytes and microglia from parenchymal cells, we demonstrated that pericytes contribute to microglial proliferation and support microglia in efficiently promoting the differentiation of neural stem cells into intermediate progenitors. Our present data provide evidence that pericytes function not only in the maintenance of cerebral microcirculation and blood brain barrier (BBB) integrity but also in microglial homeostasis in the developing cerebral walls. These findings will expand our knowledge and help elucidate the mechanism of brain development both in healthy and disease conditions.


Cerebral Cortex/cytology , Homeostasis/physiology , Microglia/cytology , Neural Stem Cells/cytology , Pericytes/cytology , Animals , Antibodies, Neutralizing , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/embryology , Capillary Permeability/drug effects , Cell Line , Cell Proliferation/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Clodronic Acid/pharmacology , Homeostasis/drug effects , Liposomes , Mice , Microglia/drug effects , Neural Stem Cells/drug effects , Pericytes/drug effects , Receptor, Platelet-Derived Growth Factor beta
...