Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Am J Physiol Heart Circ Physiol ; 327(2): H390-H398, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38874615

ABSTRACT

The endothelial glycocalyx (EG), covering the luminal side of endothelial cells, regulates vascular permeability and senses wall shear stress. In sepsis, EG undergoes degradation leading to increased permeability and edema formation. We hypothesized that restoring EG integrity using liposomal nanocarriers of preassembled glycocalyx (LNPG) will restore normal venular permeability in lipopolysaccharide (LPS)-induced sepsis model of mice. To test this hypothesis, we designed a unique perfusion microchamber in which the permeability of isolated venules could be assessed by measuring the concentration of Evans blue dye (EBD) in microliter samples of extravascular solution (ES). Histamine-induced time- and dose-dependent increases in EBD in the ES could be measured, confirming the sensitivity of the microchamber system. Notably, the histamine-induced increase in permeability was significantly attenuated by histamine receptor (H1) antagonist, triprolidine hydrochloride. Subsequently, mice were treated with LPS or LPS + LNPG. When compared with control mice, venules from LPS-treated mice showed a significant increased permeability, which was significantly reduced by LNPG administration. Moreover, in the presence of wall shear stress, intraluminal administration of LNPG significantly reduced the permeability in isolated venules from LPS-treated mice. We have found no sex differences. In conclusion, our newly developed microchamber system allows us to quantitatively measure the permeability of isolated venules. LPS-induced sepsis increases permeability of mesenteric venules that is attenuated by in vivo LNPG administration, which also reestablished endothelial responses to shear stress. Thus, LNPG presents a promising therapeutic potential for restoring EG function and thereby mitigating vasogenic edema due to increased permeability in sepsis.NEW & NOTEWORTHY In sepsis, the degradation of the endothelial glycocalyx leads to increased venular permeability. In this study, we developed a potentially new therapeutic approach by in vivo administration of liposomal nanocarriers of preassembled glycocalyx to mice, which restored venular sensitivity to wall shear stress and permeability in lipopolysaccharide-induced sepsis, likely by restoring the integrity of the endothelial glycocalyx. Using a new microchamber system, the permeability of Evans blue dye could be quantitatively determined.


Subject(s)
Capillary Permeability , Glycocalyx , Lipopolysaccharides , Liposomes , Sepsis , Stress, Mechanical , Animals , Glycocalyx/metabolism , Glycocalyx/drug effects , Capillary Permeability/drug effects , Lipopolysaccharides/toxicity , Venules/metabolism , Venules/physiopathology , Venules/drug effects , Male , Sepsis/physiopathology , Sepsis/metabolism , Sepsis/drug therapy , Mice , Mice, Inbred C57BL , Disease Models, Animal , Histamine/metabolism
2.
J Appl Physiol (1985) ; 136(6): 1488-1495, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38722754

ABSTRACT

Advancing age increases cardiovascular disease risk, in part, because of impaired glycocalyx thickness and endothelial dysfunction. Glycocalyx-targeted therapies, such as Endocalyx Pro, could improve both glycocalyx thickness and endothelial function in older adults; however, this has yet to be tested. We hypothesized that Endocalyx Pro supplementation would increase glycocalyx thickness and endothelial function in older adults. Twenty-three older adults aged 66 ± 7 yr (52% female) were enrolled in a randomized, double-blind, placebo-controlled, parallel-arms study to investigate the effect of 12-wk Endocalyx Pro supplementation (3,712 mg/day) on glycocalyx thickness and endothelial function. Glycocalyx thickness was assessed using the GlycoCheck, and endothelial function was determined via brachial artery flow-mediated dilation (FMD). Between-group comparisons revealed Endocalyx Pro did not increase glycocalyx thickness in microvessels 4-25 µm (P = 0.33), 4-7 µm (P = 0.07), or 10-25 µm (P = 0.47) in diameter when compared with placebo. In addition, Endocalyx Pro did not significantly improve FMD [mean ratio (95%) confidence interval [CI]) for between-group comparisons, 1.16 (0.77-1.74); P = 0.48]. However, Endocalyx Pro improved FMD normalized to shear rate (SR) area under the curve [mean ratio (95% CI) for between-group comparisons, 2.41 (1.14,4.13); P = 0.001]. Moreover, Endocalyx Pro increased capillary glycocalyx thickness more than placebo in individuals not taking antihypertensive medication [mean difference (95% CI) for between-group comparison, -0.08 (-0.15, -0.01); P = 0.02]. Our pilot study suggests that Endocalyx Pro supplementation is feasible in older adults but has no measurable effect on overall glycocalyx thickness and FMD. However, Endocalyx Pro may have select effects on capillary glycocalyx thickness and FMD normalized to shear rate among older adults, but further investigation is warranted.NEW & NOTEWORTHY Endothelial glycocalyx thickness and vascular endothelial function decline with advancing age. Endocalyx Pro is a glycocalyx-targeted therapy that may improve endothelial glycocalyx thickness and vascular endothelial function in older adults. This study demonstrated that 12-wk Endocalyx Pro supplementation did not improve overall endothelial glycocalyx thickness or flow-mediated dilation in older adults; however, Endocalyx Pro did increase capillary glycocalyx thickness in individuals not taking antihypertensive medication and improve flow-mediated dilation normalized to the shear stimulus.


Subject(s)
Brachial Artery , Endothelium, Vascular , Glycocalyx , Humans , Glycocalyx/drug effects , Glycocalyx/metabolism , Female , Male , Aged , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Double-Blind Method , Brachial Artery/drug effects , Brachial Artery/physiology , Brachial Artery/diagnostic imaging , Vasodilation/drug effects , Vasodilation/physiology , Middle Aged , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology
3.
Sci Rep ; 14(1): 10477, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714743

ABSTRACT

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.


Subject(s)
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.
Microvasc Res ; 154: 104684, 2024 07.
Article in English | MEDLINE | ID: mdl-38663724

ABSTRACT

The endothelial glycocalyx (EG) undergoes early degradation in sepsis. Our recent work introduced a novel therapeutic approach involving liposomal nanocarriers of preassembled glycocalyx (LNPG) to restore EG in lipopolysaccharide (LPS)-induced sepsis model of mice. While short-term effects were promising, this study focuses on the long-term impact of LNPG on mouse cerebral microcirculation. Utilizing cranial window, we assessed the stability of vascular density (VD) and perfused boundary region (PBR), an index of EG thickness, over a five-day period in normal control mice. In septic groups (LPS, LPS + 1-dose LNPG, and LPS + 2-dose LNPG), the exposure of mice to LPS significantly reduced VD and increased PBR within 3 h. Without LNPG treatment, PBR returned to the normal control level by endogenous processes at 48 h, associated with the recovery of VD to the baseline level at 72 h. However, mice receiving LNPG treatment significantly reduced the increment of PBR at 3 h. The therapeutic effect of 1-dose LNPG persisted for 6 h while the 2-dose LNPG treatment further reduced PBR and significantly increased VD at 12 h compared to LPS group. This study provides valuable insights into the potential therapeutic benefits of LNPG in mitigating EG degradation in sepsis.


Subject(s)
Cerebrovascular Circulation , Disease Models, Animal , Glycocalyx , Lipopolysaccharides , Liposomes , Mice, Inbred C57BL , Microcirculation , Sepsis , Animals , Glycocalyx/metabolism , Glycocalyx/drug effects , Glycocalyx/pathology , Sepsis/drug therapy , Sepsis/metabolism , Male , Time Factors , Cerebrovascular Circulation/drug effects , Microcirculation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Mice , Nanoparticles , Drug Carriers
5.
J Trauma Acute Care Surg ; 97(1): 32-38, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38444065

ABSTRACT

INTRODUCTION: The endothelial glycocalyx on the luminal surface of endothelial cells contributes to the permeability barrier of the pulmonary vasculature. Dimethyl sulfoxide (DMSO) has a disordering effect on plasma membranes, which prevents the formation of ordered membrane domains important in the shedding of the endothelial glycocalyx. We hypothesized that DMSO would protect against protein leak by preserving the endothelial glycocalyx in a murine model of acute respiratory distress syndrome (ARDS). METHODS: C57BL/6 mice were given ARDS via intratracheally administered lipopolysaccharide (LPS). Dimethyl sulfoxide (220 mg/kg) was administered intravenously for 4 days. Animals were sacrificed postinjury day 4 after bronchoalveolar lavage (BAL). Bronchoalveolar lavage cell counts and protein content were quantified. Lung sections were stained with fluorescein isothiocyanate-labeled wheat germ agglutinin to quantify the endothelial glycocalyx. Human umbilical vein endothelial cells (HUVECs) were exposed to LPS. Endothelial glycocalyx was measured using fluorescein isothiocyanate-labeled wheat germ agglutinin, and co-immunoprecipitation was performed to measure interaction between sheddases and syndecan-1. RESULTS: Dimethyl sulfoxide treatment resulted in greater endothelial glycocalyx staining intensity in the lung when compared with sham (9,641 vs. 36,659 arbitrary units, p < 0.001). Total BAL cell counts were less for animals receiving DMSO (6.93 × 10 6 vs. 2.49 × 10 6 cells, p = 0.04). The treated group had less BAL macrophages (189.2 vs. 76.9 cells, p = 0.02) and lymphocytes (527.7 vs. 200.0 cells, p = 0.02). Interleukin-6 levels were lower in DMSO treated. Animals that received DMSO had less protein leak in BAL (1.48 vs. 1.08 µg/µL, p = 0.02). Dimethyl sulfoxide prevented LPS-induced endothelial glycocalyx loss in HUVECs and reduced the interaction between matrix metalloproteinase 16 and syndecan-1. CONCLUSION: Systemically administered DMSO protects the endothelial glycocalyx in the pulmonary vasculature, mitigating pulmonary capillary leak after acute lung injury. Dimethyl sulfoxide also results in decreased inflammatory response. Dimethyl sulfoxide reduced the interaction between matrix metalloproteinase 16 and syndecan-1 and prevented LPS-induced glycocalyx damage in HUVECs. Dimethyl sulfoxide may be a novel therapeutic for ARDS.


Subject(s)
Acute Lung Injury , Dimethyl Sulfoxide , Disease Models, Animal , Glycocalyx , Mice, Inbred C57BL , Animals , Mice , Glycocalyx/metabolism , Glycocalyx/drug effects , Dimethyl Sulfoxide/pharmacology , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Lipopolysaccharides , Male , Humans , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/pathology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Human Umbilical Vein Endothelial Cells/drug effects
6.
Diabetes ; 73(6): 964-976, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38530908

ABSTRACT

Adiponectin has vascular anti-inflammatory and protective effects. Although adiponectin protects against the development of albuminuria, historically, the focus has been on podocyte protection within the glomerular filtration barrier (GFB). The first barrier to albumin in the GFB is the endothelial glycocalyx (eGlx), a surface gel-like barrier covering glomerular endothelial cells (GEnCs). In diabetes, eGlx dysfunction occurs before podocyte damage; hence, we hypothesized that adiponectin could protect from eGlx damage to prevent early vascular damage in diabetic kidney disease (DKD). Globular adiponectin (gAd) activated AMPK signaling in human GEnCs through AdipoR1. It significantly reduced eGlx shedding and the tumor necrosis factor-α (TNF-α)-mediated increase in syndecan-4 (SDC4) and MMP2 mRNA expression in GEnCs in vitro. It protected against increased TNF-α mRNA expression in glomeruli isolated from db/db mice and against expression of genes associated with glycocalyx shedding (namely, SDC4, MMP2, and MMP9). In addition, gAd protected against increased glomerular albumin permeability (Ps'alb) in glomeruli isolated from db/db mice when administered intraperitoneally and when applied directly to glomeruli (ex vivo). Ps'alb was inversely correlated with eGlx depth in vivo. In summary, adiponectin restored eGlx depth, which was correlated with improved glomerular barrier function, in diabetes.


Subject(s)
Adiponectin , Diabetes Mellitus, Type 2 , Glycocalyx , Kidney Glomerulus , Animals , Glycocalyx/metabolism , Glycocalyx/drug effects , Adiponectin/metabolism , Adiponectin/genetics , Mice , Diabetes Mellitus, Type 2/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Kidney Glomerulus/drug effects , Humans , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Glomerular Filtration Barrier/metabolism , Glomerular Filtration Barrier/drug effects , Tumor Necrosis Factor-alpha/metabolism , Syndecan-4/metabolism , Syndecan-4/genetics , Disease Models, Animal , Mice, Inbred C57BL
7.
Shock ; 61(5): 776-782, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38517274

ABSTRACT

ABSTRACT: Background : This study aims to determine the impact and mechanism of miR-21-3p on intestinal injury and intestinal glycocalyx during fluid resuscitation in traumatic hemorrhagic shock (THS), and the different impacts of sodium lactate Ringer's solution (LRS) and sodium bicarbonate Ringer's solution (BRS) for resuscitation on intestinal damage. Methods : A rat model of THS was induced by hemorrhage from the left femur fracture. The pathological changes of intestinal tissues and glycocalyx structure were observed by hematoxylin-eosin staining and transmission electron microscope. MiR-21-3p expression in intestinal tissues was detected by real-time quantitative polymerase chain reaction. The expression of glycocalyx-, cell junction-, and PI3K/Akt/NF-κB signaling pathway-related proteins was analyzed by western blot. Results : MiR-21-3p expression was increased in THS rats, which was suppressed by resuscitation with BRS. BRS or LRS aggravated the intestinal injury and damaged intestinal glycocalyx in THS rats. The expression of SDC-1, HPA, ß-catenin, MMP2, and MMP9 was upregulated, the expression of E-cad was downregulated, and the PI3K/Akt/NF-κB signaling pathway was activated in THS rats, which were further aggravated by BRS or LRS. The adverse effect of LRS was more serious than BRS. MiR-21-3p overexpression deteriorated the injury of intestinal tissues and intestinal glycocalyx; increased the expression of SDC-1, HPA, ß-catenin, MMP2, and MMP9 while decreasing E-cad expression; and activated the PI3K/Akt/NF-κB signaling pathway in BRS-resuscitated THS rats. Conclusion : MiR-21-3p aggravated intestinal tissue injury and intestinal glycocalyx damage through activating PI3K/Akt/NF-κB signaling pathway in rats with THS resuscitated with BRS.


Subject(s)
Intestines , MicroRNAs , Ringer's Solution , Shock, Hemorrhagic , Animals , Male , Rats , Glycocalyx/drug effects , Glycocalyx/metabolism , Glycocalyx/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestines/pathology , Intestines/drug effects , Intestines/injuries , Isotonic Solutions/pharmacology , Isotonic Solutions/therapeutic use , MicroRNAs/metabolism , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Resuscitation , Shock, Hemorrhagic/drug therapy , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/complications , Signal Transduction/drug effects , Sodium Bicarbonate/therapeutic use , Sodium Bicarbonate/pharmacology , Ringer's Solution/pharmacology , Ringer's Solution/therapeutic use
8.
J Appl Toxicol ; 44(6): 908-918, 2024 06.
Article in English | MEDLINE | ID: mdl-38396353

ABSTRACT

Cadmium (Cd) is one of the most polluting heavy metal in the environment. Cd exposure has been elucidated to cause dysfunction of the glomerular filtration barrier (GFB). However, the underlying mechanism remains unclear. C57BL/6J male mice were administered with 2.28 mg/kg cadmium chloride (CdCl2) dissolved in distilled water by oral gavage for 14 days. The expression of SDC4 in the kidney tissues was detected. Human renal glomerular endothelial cells (HRGECs) were exposed to varying concentrations of CdCl2 for 24 h. The mRNA levels of SDC4, along with matrix metalloproteinase (MMP)-2 and 9, were analyzed by quantitative PCR. Additionally, the protein expression levels of SDC4, MMP-2/9, and both total and phosphorylated forms of Smad2/3 (P-Smad2/3) were detected by western blot. The extravasation rate of fluorescein isothiocyanate-dextran through the Transwell was used to evaluate the permeability of HRGECs. SB431542 was used as an inhibitor of transforming growth factor (TGF)-ß signaling pathway to further investigate the role of TGF-ß. Cd reduced SDC4 expression in both mouse kidney tissues and HRGECs. In addition, Cd exposure increased permeability and upregulated P-Smad2/3 levels in HRGECs. SB431542 treatment inhibited the phosphorylation of Smad2/3, Cd-induced SDC4 downregulation, and hyperpermeability. MMP-2/9 levels increased by Cd exposure was also blocked by SB431542, demonstrating the involvement of TGF-ß/Smad pathway in low-dose Cd-induced SDC4 reduction in HRGECs. Given that SDC4 is an essential component of glycocalyx, protection or repair of endothelial glycocalyx is a potential strategy for preventing or treating kidney diseases associated with environmental Cd exposure.


Subject(s)
Cadmium , Endothelial Cells , Glycocalyx , Kidney Glomerulus , Syndecan-4 , Animals , Humans , Male , Mice , Cadmium/toxicity , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Glycocalyx/drug effects , Glycocalyx/metabolism , Kidney Glomerulus/drug effects , Kidney Glomerulus/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , Signal Transduction/drug effects , Syndecan-4/metabolism , Syndecan-4/genetics , Transforming Growth Factor beta/metabolism
9.
J Pain ; 25(6): 104462, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38211844

ABSTRACT

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.


Subject(s)
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.
Viruses ; 15(7)2023 07 05.
Article in English | MEDLINE | ID: mdl-37515191

ABSTRACT

Endothelial glycocalyx (EG) derangement has been associated with cardiovascular disease (CVD). Studies on EG integrity among people living with HIV (PLWH), are lacking. We conducted a prospective cohort study among treatment-naïve PLWH who received emtricitabine/tenofovir alafenamide, combined with either an integrase strand transfer inhibitor (INSTI, dolutegravir, raltegravir or elvitegravir/cobicistat), or a protease inhibitor (PI, darunavir/cobicistat). We assessed EG at baseline, 24 (±4) and 48 (±4) weeks, by measuring the perfused boundary region (PBR, inversely proportional to EG thickness), in sublingual microvessels. In total, 66 consecutive PLWH (60 (90.9%) males) with a median age (interquartile range, IQR) of 37 (12) years, were enrolled. In total, 40(60.6%) received INSTI-based regimens. The mean (standard deviation) PBR decreased significantly from 2.17 (0.29) µm at baseline to 2.04 (0.26) µm (p = 0.019), and then to 1.93 (0.3) µm (p < 0.0001) at 24 (±4) and 48 (±4) weeks, respectively. PBR did not differ among treatment groups. PLWH on INSTIs had a significant PBR reduction at 48 (±4) weeks. Smokers and PLWH with low levels of viremia experienced the greatest PBR reduction. This study is the first to report the benefit of antiretroviral treatment on EG improvement in treatment-naïve PLWH and depicts a potential bedside biomarker and therapeutic target for CVD in PLWH.


Subject(s)
Anti-HIV Agents , Endothelium , Glycocalyx , HIV Infections , HIV Infections/drug therapy , HIV Infections/pathology , Glycocalyx/drug effects , Glycocalyx/pathology , Endothelium/drug effects , Endothelium/pathology , Humans , Anti-HIV Agents/therapeutic use , Male , Female , Adult , Middle Aged , Cohort Studies , CD4 Lymphocyte Count , Viral Load , Smoking
11.
Pflugers Arch ; 474(10): 1069-1076, 2022 10.
Article in English | MEDLINE | ID: mdl-35867189

ABSTRACT

Proinflammatory cytokines target vascular endothelial cells during COVID-19 infections. In particular, the endothelial glycocalyx (eGC), a proteoglycan-rich layer on top of endothelial cells, was identified as a vulnerable, vasoprotective structure during infections. Thus, eGC damage can be seen as a hallmark in the development of endothelial dysfunction and inflammatory processes. Using sera derived from patients suffering from COVID-19, we could demonstrate that the eGC became progressively worse in relation to disease severity (mild vs severe course) and in correlation to IL-6 levels. This could be prevented by administering low doses of spironolactone, a well-known and highly specific aldosterone receptor antagonist. Our results confirm that SARS-CoV-2 infections cause eGC damage and endothelial dysfunction and we outline the underlying mechanisms and suggest potential therapeutic options.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Glycocalyx , Mineralocorticoid Receptor Antagonists , SARS-CoV-2 , Spironolactone , COVID-19/blood , COVID-19/pathology , Cytokines/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/pathology , Glycocalyx/drug effects , Glycocalyx/pathology , Humans , Interleukin-6/blood , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Proteoglycans/analysis , Proteoglycans/blood , Spironolactone/pharmacology , Spironolactone/therapeutic use
12.
Mar Drugs ; 20(2)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35200650

ABSTRACT

We previously reported that rhamnan sulfate (RS) purified from Monostroma nitidum significantly suppressed lipopolysaccharide (LPS)-induced inflammation in cultured human vascular endothelial cells. Here, we analyzed the effect of orally administered RS on LPS-induced damage to mouse organs and vascular endothelium. RS (1 mg) was orally administered daily to BALB/c mice, 50 µg of LPS was intraperitoneally administered on day 8, and Evans blue was injected into the tail vein 6 h later. After 30 min, LPS-treated mice showed pulmonary Evans blue leakage and elevated plasma levels of liver damage markers, whereas this reaction was suppressed in LPS + RS-treated mice. Immunohistochemical and Western blot analysis of mouse organs 24 h after LPS treatment showed significant neutrophil infiltration into the lung, liver, and jejunum tissues of LPS-treated mice and high expression levels of inflammation-related factors in these tissues. Expression levels of these factors were significantly suppressed in LPS + RS-treated mice. Analysis of lung glycocalyx showed a significant reduction in glycocalyx in LPS-treated mice but not in LPS + RS-treated mice. Levels of syndecan-4, one of the glycocalyx components, decreased in LPS-treated mice and increased in LPS + RS-treated mice. The current results suggest that orally administered RS protects organs and vascular endothelium from LPS-induced inflammation and maintains blood circulation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chlorophyta/chemistry , Deoxy Sugars/pharmacology , Inflammation/drug therapy , Mannans/pharmacology , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/isolation & purification , Deoxy Sugars/administration & dosage , Deoxy Sugars/isolation & purification , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Glycocalyx/drug effects , Glycocalyx/metabolism , Inflammation/pathology , Lipopolysaccharides , Male , Mannans/administration & dosage , Mannans/isolation & purification , Mice , Mice, Inbred BALB C , Neutrophils/metabolism
13.
Surgery ; 171(3): 825-832, 2022 03.
Article in English | MEDLINE | ID: mdl-34865862

ABSTRACT

BACKGROUND: Hemorrhagic shock leads to endothelial glycocalyx shedding, endothelial cellular inflammation, and increased vascular permeability. Early plasma administration improves survival in severely injured patients; this may be due in part to its ability to ameliorate this trauma-induced endotheliopathy. The protective effect of early plasma administration may be due to its sphingosine 1-phosphate content. Principle carriers of plasma sphingosine 1-phosphate include apolipoprotein M and albumin. The relative roles of these carriers on sphingosine 1-phosphate protective effects are unknown and were studied in an in vitro model of microcirculation. METHODS: Endothelial cell monolayers were established in microfluidic perfusion devices and exposed to control or biomimetic shock conditions. Sphingosine 1-phosphate, albumin + sphingosine 1-phosphate, or apolipoprotein M + sphingosine 1-phosphate were added later to the perfusate. Biomarkers of endothelial and glycocalyx activation and damage were then determined. RESULTS: Sphingosine 1-phosphate preserved endothelial and glycocalyx barrier function after exposure to conditions of shock in the microcirculation. The protective effect was related to sphingosine 1-phosphate chaperones; the apolipoprotein M loaded with sphingosine 1-phosphate had the most profound effect. CONCLUSION: Carrier-based sphingosine 1-phosphate may be a useful adjunct in early hemorrhagic shock resuscitation.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Lysophospholipids/pharmacology , Shock/pathology , Sphingosine/analogs & derivatives , Albumins/pharmacology , Apolipoproteins M/pharmacology , Cell Culture Techniques , Glycocalyx/drug effects , Humans , Microcirculation , Sphingosine/pharmacology
14.
Front Immunol ; 12: 789142, 2021.
Article in English | MEDLINE | ID: mdl-34917096

ABSTRACT

Fabry disease (FD) is an X-linked multisystemic lysosomal storage disease due to a deficiency of α-galactosidase A (GLA/AGAL). Progressive cellular accumulation of the AGAL substrate globotriaosylceramide (Gb3) leads to endothelial dysfunction. Here, we analyzed endothelial function in vivo and in vitro in an AGAL-deficient genetic background to identify the processes underlying this small vessel disease. Arterial stiffness and endothelial function was prospectively measured in five males carrying GLA variants (control) and 22 FD patients under therapy. AGAL-deficient endothelial cells (EA.hy926) and monocytes (THP1) were used to analyze endothelial glycocalyx structure, function, and underlying inflammatory signals. Glycocalyx thickness and small vessel function improved significantly over time (p<0.05) in patients treated with enzyme replacement therapy (ERT, n=16) and chaperones (n=6). AGAL-deficient endothelial cells showed reduced glycocalyx and increased monocyte adhesion (p<0.05). In addition, increased expression of angiopoietin-2, heparanase and NF-κB was detected (all p<0.05). Incubation of wild-type endothelial cells with pathological globotriaosylsphingosine concentrations resulted in comparable findings. Treatment of AGAL-deficient cells with recombinant AGAL (p<0.01), heparin (p<0.01), anti-inflammatory (p<0.001) and antioxidant drugs (p<0.05), and a specific inhibitor (razuprotafib) of angiopoietin-1 receptor (Tie2) (p<0.05) improved glycocalyx structure and endothelial function in vitro. We conclude that chronic inflammation, including the release of heparanases, appears to be responsible for the degradation of the endothelial glycocalyx and may explain the endothelial dysfunction in FD. This process is partially reversible by FD-specific and anti-inflammatory treatment, such as targeted protective Tie2 treatment.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Fabry Disease/metabolism , Glycocalyx/metabolism , Vascular Stiffness , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/therapeutic use , Adult , Aged , Anti-Inflammatory Agents/pharmacology , Case-Control Studies , Coculture Techniques , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Enzyme Replacement Therapy , Fabry Disease/drug therapy , Fabry Disease/pathology , Fabry Disease/physiopathology , Genetic Predisposition to Disease , Glycocalyx/drug effects , Glycocalyx/pathology , Humans , Male , Middle Aged , Mutation , Phenotype , Prospective Studies , THP-1 Cells , Vascular Stiffness/drug effects , alpha-Galactosidase/genetics , alpha-Galactosidase/therapeutic use
15.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830227

ABSTRACT

Ricin toxin isolated from the castor bean (Ricinus communis) is one of the most potent and lethal molecules known. While the pathophysiology and clinical consequences of ricin poisoning by the parenteral route, i.e., intramuscular penetration, have been described recently in various animal models, the preceding mechanism underlying the clinical manifestations of systemic ricin poisoning has not been completely defined. Here, we show that following intramuscular administration, ricin bound preferentially to the vasculature in both mice and swine, leading to coagulopathy and widespread hemorrhages. Increased levels of circulating VEGF and decreased expression of vascular VE-cadherin caused blood vessel impairment, thereby promoting hyperpermeability in various organs. Elevated levels of soluble heparan sulfate, hyaluronic acid and syndecan-1 were measured in blood samples following ricin intoxication, indicating that the vascular glycocalyx of both mice and swine underwent extensive damage. Finally, by using side-stream dark field intravital microscopy imaging, we determined that ricin poisoning leads to microvasculature malfunctioning, as manifested by aberrant blood flow and a significant decrease in the number of diffused microvessels. These findings, which suggest that glycocalyx shedding and microcirculation dysfunction play a major role in the pathology of systemic ricin poisoning, may serve for the formulation of specifically tailored therapies for treating parenteral ricin intoxication.


Subject(s)
Endothelial Cells/drug effects , Glycocalyx/drug effects , Ricin/toxicity , Ricinus/chemistry , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Gene Expression/drug effects , Glycocalyx/chemistry , Glycocalyx/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Hydrolysis , Injections, Intramuscular , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Microcirculation/drug effects , Ricin/isolation & purification , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Swine , Syndecan-1/chemistry , Syndecan-1/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Shock ; 56(4): 593-600, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34524269

ABSTRACT

ABSTRACT: Heat stroke is characterized by excessive oxidative stress and inflammatory responses, both of which are implicated in vascular endothelial glycocalyx shedding and heat-stroke mortality. Although molecular hydrogen has antioxidation and anti-inflammatory potency, its effect on the vascular endothelial glycocalyx in heat stroke has not been examined. Therefore, the aim of this study was to investigate the influence of hydrogen inhalation on the survival and thickness of the vascular endothelial glycocalyx of rats subjected to heat stroke. Altogether, 98 Wistar rats were assigned to the experiments. A heat-controlled chamber set at 40°C temperature and 60% humidity was used to induce heat stroke. After preparation, the anesthetized rats that underwent the heating process were subjected to an hour of stabilization in which 0%, 2%, or 4% hydrogen gas was inhaled and maintained until the experiment ended. In addition to survival rate assessments, blood samples and left ventricles were collected to evaluate the thickness of the vascular endothelial glycocalyx and relevant biomarkers. The results showed that 2% hydrogen gas significantly improved survival in the heat-stroked rats and partially preserved the thickness of the endothelial glycocalyx. In addition, serum levels of endotoxin, syndecan-1, malondialdehyde, and tumor necrosis factor-α decreased, whereas superoxide dismutase levels increased, indicating that inhalation of 2% hydrogen attenuated the damage to the vascular endothelial glycocalyx through its antioxidative and anti-inflammatory effects.


Subject(s)
Deuterium/administration & dosage , Endothelial Cells/drug effects , Glycocalyx/drug effects , Heat Stroke/metabolism , Heat Stroke/therapy , Administration, Inhalation , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Glycocalyx/metabolism , Heat Stroke/pathology , Male , Rats , Rats, Wistar
17.
Cells ; 10(9)2021 09 04.
Article in English | MEDLINE | ID: mdl-34571967

ABSTRACT

Glycomic profiling methods were used to determine the effect of metabolic inhibitors on glycan production. These inhibitors are commonly used to alter the cell surface glycosylation. However, structural analysis of the released glycans has been limited. In this research, the cell membranes were enriched and the glycans were released to obtain the N-glycans of the glycocalyx. Glycomic analysis using liquid chromatography-mass spectrometry (LC-MS) with a PGC chip column was used to profile the structures in the cell membrane. Glycans of untreated cells were compared to glycans of cells treated with inhibitors, including kifunensine, which inhibits the formation of complex- and hybrid-type structures, 2,4,7,8,9-Penta-O-acetyl-N-acetyl-3-fluoro-b-d-neuraminic acid methyl ester for sialylated glycans, 2-deoxy-2-fluorofucose, and 6-alkynyl fucose for fucosylated glycans. Kifunensine was the most effective, converting nearly 95% of glycans to high mannose types. The compound 6-alkynyl fucose inhibited some fucosylation but also incorporated into the glycan structure. Proteomic analysis of the enriched membrane for the four inhibitors showed only small changes in the proteome accompanied by large changes in the N-glycome for Caco-2. Future works may use these inhibitors to study the cellular behavior associated with the alteration of glycosylation in various biological systems, e.g., viral and bacterial infection, drug binding, and cell-cell interactions.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycocalyx/drug effects , Glycomics , Glycoproteins/metabolism , Glycosyltransferases/antagonists & inhibitors , Polysaccharides/metabolism , A549 Cells , Alkaloids/chemistry , Alkaloids/pharmacology , Caco-2 Cells , Chromatography, Liquid , Enzyme Inhibitors/chemistry , Fucose/analogs & derivatives , Fucose/chemistry , Fucose/pharmacology , Glycocalyx/enzymology , Glycomics/instrumentation , Glycosylation , Glycosyltransferases/metabolism , Humans , Lab-On-A-Chip Devices , Mass Spectrometry , Microfluidic Analytical Techniques/instrumentation , Molecular Structure , Neuraminic Acids/chemistry , Neuraminic Acids/pharmacology , Proteomics , Structure-Activity Relationship
18.
Physiol Rep ; 9(17): e15019, 2021 09.
Article in English | MEDLINE | ID: mdl-34472715

ABSTRACT

Vascular endothelial cells are covered with glycocalyx comprising heparan sulfate, hyaluronan, chondroitin sulfate, and associated proteins. Glomerular endothelial glycocalyx is involved in protecting against induction of proteinuria and structural damage, but the specific components in glycocalyx that represent therapeutic targets remain unclear. Anti-vascular endothelial growth factor (VEGF) therapy is associated with an increased risk of glomerular endothelial injury. This study investigated whether hyaluronan could provide a therapeutic target to protect against proteinuria. We conducted ex vivo and in vivo experiments to explore the effects of degrading glomerular hyaluronan by administering hyaluronidase and of supplementation with hyaluronan. We investigated hyaluronan expression using biotin-labeled hyaluronan-binding protein (HABP) in human kidney specimens or serum hyaluronan in endothelial injuries under inhibition of VEGF signaling. We directly demonstrated hyaluronan in glomerular endothelial layers using HABP staining. Ex vivo and in vivo experiments showed the development of proteinuria after digestion of hyaluronan in glomerular capillaries. Supplementation with hyaluronan after hyaluronidase treatment suppressed proteinuria. Mice in the in vivo study developed albuminuria after intraperitoneal injection of hyaluronidase with decreased glomerular hyaluronan and increased serum hyaluronan. In human kidneys with endothelial cell dysfunction and proteinuria due to inhibition of VEGF, glomerular expression of hyaluronan was reduced even in normal-appearing glomeruli. Serum hyaluronan levels were elevated in patients with pre-eclampsia with VEGF signaling inhibition. Our data suggest that hyaluronan itself plays crucial roles in preventing proteinuria and preserving the integrity of endothelial cells. Hyaluronan could provide a therapeutic target for preventing glomerular endothelial glycocalyx damage, including VEGF signaling inhibition.


Subject(s)
Endothelial Cells/metabolism , Glycocalyx/metabolism , Hyaluronic Acid/biosynthesis , Kidney Glomerulus/metabolism , Proteinuria/metabolism , Animals , Cattle , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Glycocalyx/drug effects , Glycocalyx/pathology , Humans , Hyaluronoglucosaminidase/administration & dosage , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Pregnancy , Proteinuria/pathology , Rats , Rats, Inbred Lew
19.
Anesth Analg ; 133(4): 1036-1047, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34269720

ABSTRACT

BACKGROUND: The endothelial glycocalyx, a carbohydrate-rich layer coating all endothelial surfaces, plays a fundamental role in the function of microcirculation. The primary aim of this study was to evaluate the feasibility of using dexamethasone and albumin to protect the endothelial glycocalyx in patients undergoing abdominal surgery. Secondary and exploratory outcomes included efficacy and safety. METHODS: We conducted a multicenter, open-label, blinded end point, phase 2, randomized trial. Patients undergoing colorectal, pancreas, or liver surgery were recruited and randomized to receive either intravenous dexamethasone (16 mg) and 20% albumin (100 mL) at induction of anesthesia, then 200 mL of 20% albumin with each subsequent 1000 mL of crystalloid administered (dexamethasone and albumin [Dex-Alb] group), or crystalloid fluid only with no dexamethasone (control group). Feasibility end points included patient recruitment and retention, consent rate, and successful study drug administration. The primary efficacy end point was the measurement of plasma syndecan-1 level on postoperative day (POD) 1, and secondary end points were heparan sulfate levels and inflammatory markers measured at 4 perioperative timepoints. Safety end points included errors in administration of the intervention, hyperglycemia, occurrence of postoperative complications, and patient retention. RESULTS: Seventy-two patients were randomized. All feasibility end points were achievable. There were no statistically significant differences observed in median (interquartile range) syndecan-1 levels on POD 1 (39 ng·mL-1 [20-97] in the Dex-Alb group versus 41 ng·mL-1 [19-84] in the control group; difference in medians -2.1, 95% confidence interval [CI], -13 to 8.6; P = .69). The Dex-Alb group had lower POD 1 heparan sulfate levels (319 ng·mL-1 [161-717] in the Dex-Alb group versus 1422 [670-2430] ng·mL-1 in the control group; difference in medians -1085, 95% CI, -1779 to -391) and C-reactive protein (CRP) levels on POD 1 (48 [29-77] mg·L-1 in the Dex-Alb group versus 85 mg·L-1 [49-133] in the control group; difference in medians -48, 95% CI, -75 to -21). Fewer patients had one or more postoperative complication in the Dex-Alb group than in the control group (6 [17%] vs 18 patients [50%]; odds ratio = 0.2, 95% CI, 0.06-0.6). CONCLUSIONS: Intravenous dexamethasone and albumin administration was feasible but did not reduce syndecan-1 on POD 1 in patients undergoing abdominal surgery. Given the clinically important CIs observed between the groups for heparan sulfate, CRP, and postoperative complications, a larger trial assessing the associations between dexamethasone and albumin administration and these outcomes is warranted.


Subject(s)
Abdomen/surgery , Albumins/administration & dosage , Crystalloid Solutions/administration & dosage , Dexamethasone/administration & dosage , Digestive System Surgical Procedures , Endothelium, Vascular/drug effects , Glucocorticoids/administration & dosage , Microvessels/drug effects , Postoperative Complications/prevention & control , Aged , Albumins/adverse effects , Biomarkers/blood , C-Reactive Protein/metabolism , Crystalloid Solutions/adverse effects , Dexamethasone/adverse effects , Digestive System Surgical Procedures/adverse effects , Endothelium, Vascular/metabolism , Feasibility Studies , Female , Glucocorticoids/adverse effects , Glycocalyx/drug effects , Glycocalyx/metabolism , Heparitin Sulfate/blood , Humans , Infusions, Intravenous , Male , Microvessels/metabolism , Middle Aged , New Zealand , Postoperative Complications/blood , Postoperative Complications/etiology , Preoperative Care , Syndecan-1/blood , Time Factors , Treatment Outcome , Victoria
20.
Respir Res ; 22(1): 193, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34217286

ABSTRACT

BACKGROUND: Endothelial glycocalyx loss is integral to increased pulmonary vascular permeability in sepsis-related acute lung injury. Protectin conjugates in tissue regeneration 1 (PCTR1) is a novel macrophage-derived lipid mediator exhibiting potential anti-inflammatory and pro-resolving benefits. METHODS: PCTR1 was administrated intraperitoneally with 100 ng/mouse after lipopolysaccharide (LPS) challenged. Survival rate and lung function were used to evaluate the protective effects of PCTR1. Lung inflammation response was observed by morphology and inflammatory cytokines level. Endothelial glycocalyx and its related key enzymes were measured by immunofluorescence, ELISA, and Western blot. Afterward, related-pathways inhibitors were used to identify the mechanism of endothelial glycocalyx response to PCTR1 in mice and human umbilical vein endothelial cells (HUVECs) after LPS administration. RESULTS: In vivo, we show that PCTR1 protects mice against lipopolysaccharide (LPS)-induced sepsis, as shown by enhanced the survival and pulmonary function, decreased the inflammatory response in lungs and peripheral levels of inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and interleukin-1ß. Moreover, PCTR1 restored lung vascular glycocalyx and reduced serum heparin sulphate (HS), syndecan-1 (SDC-1), and hyaluronic acid (HA) levels. Furthermore, we found that PCTR1 downregulated heparanase (HPA) expression to inhibit glycocalyx degradation and upregulated exostosin-1 (EXT-1) protein expression to promote glycocalyx reconstitution. Besides, we observed that BAY11-7082 blocked glycocalyx loss induced by LPS in vivo and in vitro, and BOC-2 (ALX antagonist) or EX527 (SIRT1 inhibitor) abolished the restoration of HS in response to PCTR1. CONCLUSION: PCTR1 protects endothelial glycocalyx via ALX receptor by regulating SIRT1/NF-κB pathway, suggesting PCTR1 may be a significant therapeutic target for sepsis-related acute lung injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents/pharmacology , Glycocalyx/metabolism , NF-kappa B/metabolism , Respiratory Mucosa/metabolism , Sirtuin 1/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Animals , Docosahexaenoic Acids/pharmacology , Glycocalyx/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipopolysaccharides/toxicity , Male , Mice , NF-kappa B/antagonists & inhibitors , Respiratory Mucosa/drug effects , Sirtuin 1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL