Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
Front Immunol ; 15: 1426526, 2024.
Article in English | MEDLINE | ID: mdl-39055717

ABSTRACT

Introduction: Complement-mediated damage to the myocardium during acute myocardial infarction (AMI), particularly the late components of the terminal pathway (C5-convertase and C5b-9), have previously been characterized. Unfortunately, only few studies have reported a direct association between dysregulated complement activation and endothelial function. Hence, little attention has been paid to the role of the anaphylatoxin C5a. The endothelial glycocalyx (eGC) together with the cellular actin cortex provide a vasoprotective barrier against chronic vascular inflammation. Changes in their nanomechanical properties (stiffness and height) are recognized as hallmarks of endothelial dysfunction as they correlate with the bioavailability of vasoactive substances, such as nitric oxide (NO). Here, we determined how the C5a:C5aR1 axis affects the eGC and endothelial function in AMI. Methods: Samples of fifty-five patients with ST-elevation myocardial infarction (STEMI) vs. healthy controls were analyzed in this study. eGC components and C5a levels were determined via ELISA; NO levels were quantified chemiluminescence-based. Endothelial cells were stimulated with C5a or patient sera (with/without C5a-receptor1 antagonist "PMX53") and the nanomechanical properties of eGC quantified using the atomic force microscopy (AFM)-based nanoindentation technique. To measure actin cytoskeletal tension regulator activation (RhoA and Rac1) G-LISA assays were applied. Vascular inflammation was examined by quantifying monocyte-endothelium interaction via AFM-based single-cell-force spectroscopy. Results: Serum concentrations of eGC components and C5a were significantly increased during STEMI. Serum and solely C5a stimulation decreased eGC height and stiffness, indicating shedding of the eGC. C5a enhanced RhoA activation, resulting in increased cortical stiffness with subsequent reduction in NO concentrations. Monocyte adhesion to the endothelium was enhanced after both C5a and stimulation with STEMI serum. eGC degradation- and RhoA-induced cortical stiffening with subsequent endothelial dysfunction were attenuated after administering PMX53. Conclusion: This study demonstrates that dysregulated C5a activation during AMI results in eGC damage with subsequent endothelial dysfunction and reduced NO bioavailability, indicating progressively developing vascular inflammation. This could be prevented by antagonizing C5aR1, highlighting the role of the C5a:C5a-Receptor1 axis in vascular inflammation development and endothelial dysfunction in AMI, offering new therapeutic approaches for future investigations.


Subject(s)
Complement Activation , Complement C5a , Glycocalyx , Myocardial Infarction , Receptor, Anaphylatoxin C5a , Humans , Glycocalyx/metabolism , Glycocalyx/pathology , Complement C5a/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Receptor, Anaphylatoxin C5a/metabolism , Male , Middle Aged , Female , Aged , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Signal Transduction , Nitric Oxide/metabolism
2.
FASEB J ; 38(13): e23785, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38949120

ABSTRACT

Cancer metastasis is the leading cause of death for those afflicted with cancer. In cancer metastasis, the cancer cells break off from the primary tumor, penetrate nearby blood vessels, and attach and extravasate out of the vessels to form secondary tumors at distant organs. This makes extravasation a critical step of the metastatic cascade. Herein, with a focus on triple-negative breast cancer, the role that the prospective secondary tumor microenvironment's mechanical properties play in circulating tumor cells' extravasation is reviewed. Specifically, the effects of the physically regulated vascular endothelial glycocalyx barrier element, vascular flow factors, and subendothelial extracellular matrix mechanical properties on cancer cell extravasation are examined. The ultimate goal of this review is to clarify the physical mechanisms that drive triple-negative breast cancer extravasation, as these mechanisms may be potential new targets for anti-metastasis therapy.


Subject(s)
Glycocalyx , Triple Negative Breast Neoplasms , Tumor Microenvironment , Glycocalyx/metabolism , Glycocalyx/pathology , Humans , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Female , Tumor Microenvironment/physiology , Animals , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Neoplasm Metastasis , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology
3.
Ageing Res Rev ; 99: 102362, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38830545

ABSTRACT

The integrity of the endothelial glycocalyx (eGCX), a mixture of carbohydrates attached to proteins expressed on the surface of blood vessel endothelial cells (EC), is critical for the maintenance of homeostasis of the cardiovascular system and all systems of the human body, the endothelium being the critical component of the stroma of all tissues. Consequently, dysfunction of eGCX results in a dysfunctional cardiovascular wall and severe downstream cardiovascular events, which contribute to the onset of cardio- and cerebrovascular diseases and neurodegenerative disorders, as well as other age-related diseases (ARDs). The key role of eGCX dysfunction in the onset of ARDs is examined here, with a focus on the most prevalent neurological diseases: ischemic stroke and Alzheimer's disease. Furthermore, the advantages and limitations of some treatment strategies for anti-eGCX dysfunction are described, ranging from experimental drug therapies, which need to be better tested and explored not only in animal models but also in humans, as well as reprogramming, the use of nutraceuticals, which are emerging as regenerative and new approaches. The promotion of these strategies is essential to keep eGCX and endothelium healthy, as is the development of intravital (e.g., intravascular) tools to estimate eGCX health status and treatment efficacy, which could lead to advanced solutions to address ARDs.


Subject(s)
Alzheimer Disease , Glycocalyx , Ischemic Stroke , Humans , Glycocalyx/metabolism , Glycocalyx/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Ischemic Stroke/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology
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.
Angiogenesis ; 27(3): 411-422, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38598083

ABSTRACT

Damage of the endothelial glycocalyx (eGC) plays a central role in the development of vascular hyperpermeability and organ damage during systemic inflammation. However, the specific signalling pathways for eGC damage remain poorly defined. Aim of this study was to combine sublingual video-microscopy, plasma proteomics and live cell imaging to uncover further pathways of eGC damage in patients with coronavirus disease 2019 (COVID-19) or bacterial sepsis. This secondary analysis of the prospective multicenter MICROCODE study included 22 patients with COVID-19 and 43 patients with bacterial sepsis admitted to intermediate or intensive care units and 10 healthy controls. Interleukin-6 (IL-6) was strongly associated with damaged eGC and correlated both with eGC dimensions (rs=0.36, p = 0.0015) and circulating eGC biomarkers. In vitro, IL-6 reduced eGC height and coverage, which was inhibited by blocking IL-6 signalling with the anti-IL-6 receptor antibody tocilizumab or the Janus kinase inhibitor tofacitinib. Exposure of endothelial cells to 5% serum from COVID-19 or sepsis patients resulted in a significant decrease in eGC height, which was attenuated by co-incubation with tocilizumab. In an external COVID-19 cohort of 219 patients from Massachusetts General Hospital, a previously identified proteomic eGC signature correlated with IL-6 (rs=-0.58, p < 0.0001) and predicted the combined endpoint of 28-day mortality and/or intubation (ROC-AUC: 0.86 [95% CI: 0.81-0.91], p < 0.001). The data suggest that IL-6 may significantly drive eGC damage in COVID-19 and bacterial sepsis. Our findings provide valuable insights into pathomechanisms of vascular dysfunction during systemic inflammation and highlight the need for further in vivo studies.


Subject(s)
COVID-19 , Glycocalyx , Interleukin-6 , Sepsis , Humans , COVID-19/pathology , COVID-19/metabolism , COVID-19/complications , Glycocalyx/metabolism , Glycocalyx/pathology , Interleukin-6/metabolism , Interleukin-6/blood , Male , Female , Middle Aged , Sepsis/pathology , Sepsis/metabolism , Sepsis/complications , Aged , Prospective Studies , SARS-CoV-2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/pathology , Antibodies, Monoclonal, Humanized
6.
Pediatr Dev Pathol ; 27(4): 318-326, 2024.
Article in English | MEDLINE | ID: mdl-38616561

ABSTRACT

The goal of this investigation was to identify the association between Syndecan-1 (S1) serum levels in preterm newborns exposed to chorioamnionitis (CA) in utero and the potential of S1 as a biomarker of early-onset neonatal sepsis. A cohort of preterm newborns born <33 weeks gestational age was recruited. Within 48 hours of birth, 0.5 mL of blood was drawn to obtain S1 levels, measured via ELISA. Placentas were examined and classified as having (1) no CA, (2) CA without umbilical cord involvement, or (3) CA with inflammation of the umbilical cord (funisitis). S1 levels were compared between preterm newborns without exposure to CA verus newborns with exposure to CA (including with and without funisitis). Preterm newborns exposed to CA were found to have significantly elevated S1 levels compared to those unexposed. Although S1 levels could not differentiate fetal exposure to CA from exposure to CA with funisitis, the combined CA groups had significantly higher S1 levels compared to those not exposed to CA. S1 level has the potential to become a clinically useful biomarker that could assist in the management of mothers and preterm newborns with CA and funisitis. Furthermore, S1 level could aid in the diagnosis and treatment of early-onset neonatal sepsis.


Subject(s)
Biomarkers , Chorioamnionitis , Infant, Premature , Neonatal Sepsis , Syndecan-1 , Humans , Chorioamnionitis/diagnosis , Chorioamnionitis/pathology , Chorioamnionitis/blood , Infant, Newborn , Female , Biomarkers/blood , Neonatal Sepsis/diagnosis , Neonatal Sepsis/blood , Pregnancy , Syndecan-1/blood , Male , Glycocalyx/metabolism , Glycocalyx/pathology , Placenta/metabolism , Placenta/pathology
7.
Cardiovasc Pathol ; 70: 107629, 2024.
Article in English | MEDLINE | ID: mdl-38461960

ABSTRACT

BACKGROUND: Abdominal aortic aneurysm is a weakening and expansion of the abdominal aorta. Currently, there is no drug treatment to limit abdominal aortic aneurysm growth. The glycocalyx is the outermost layer of the cell surface, mainly composed of glycosaminoglycans and proteoglycans. OBJECTIVE: The aim of this review was to identify a potential relationship between glycocalyx disruption and abdominal aortic aneurysm pathogenesis. METHODS: A narrative review of relevant published research was conducted. RESULTS: Glycocalyx disruption has been reported to enhance vascular permeability, impair immune responses, dysregulate endothelial function, promote extracellular matrix remodeling and modulate mechanotransduction. All these effects are implicated in abdominal aortic aneurysm pathogenesis. Glycocalyx disruption promotes inflammation through exposure of adhesion molecules and release of proinflammatory mediators. Glycocalyx disruption affects how the endothelium responds to shear stress by reducing nitric oxide availabilty and adversely affecting the storage and release of several antioxidants, growth factors, and antithromotic proteins. These changes exacerbate oxidative stress, stimulate vascular smooth muscle cell dysfunction, and promote thrombosis, all effects implicated in abdominal aortic aneurysm pathogenesis. Deficiency of key component of the glycocalyx, such as syndecan-4, were reported to promote aneurysm formation and rupture in the angiotensin-II and calcium chloride induced mouse models of abdominal aortic aneurysm. CONCLUSION: This review provides a summary of past research which suggests that glycocalyx disruption may play a role in abdominal aortic aneurysm pathogenesis. Further research is needed to establish a causal link between glycocalyx disruption and abdominal aortic aneurysm development.


Subject(s)
Aorta, Abdominal , Aortic Aneurysm, Abdominal , Glycocalyx , Glycocalyx/pathology , Glycocalyx/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/physiopathology , Humans , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/metabolism , Aorta, Abdominal/physiopathology , Oxidative Stress , Mechanotransduction, Cellular , Capillary Permeability , Signal Transduction , Vascular Remodeling
8.
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
9.
Am J Physiol Renal Physiol ; 326(5): F681-F693, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38205540

ABSTRACT

Intermittent fasting has become of interest for its possible metabolic benefits and reduction of inflammation and oxidative damage, all of which play a role in the pathophysiology of diabetic nephropathy. We tested in a streptozotocin (60 mg/kg)-induced diabetic apolipoprotein E knockout mouse model whether repeated fasting mimicking diet (FMD) prevents glomerular damage. Diabetic mice received 5 FMD cycles in 10 wk, and during cycles 1 and 5 caloric measurements were performed. After 10 wk, glomerular endothelial morphology was determined together with albuminuria, urinary heparanase-1 activity, and spatial mass spectrometry imaging to identify specific glomerular metabolic dysregulation. During FMD cycles, blood glucose levels dropped while a temporal metabolic switch was observed to increase fatty acid oxidation. Overall body weight at the end of the study was reduced together with albuminuria, although urine production was dramatically increased without affecting urinary heparanase-1 activity. Weight loss was found to be due to lean mass and water, not fat mass. Although capillary loop morphology and endothelial glycocalyx heparan sulfate contents were preserved, hyaluronan surface expression was reduced together with the presence of UDP-glucuronic acid. Mass spectrometry imaging further revealed reduced protein catabolic breakdown products and increased oxidative stress, not different from diabetic mice. In conclusion, although FMD preserves partially glomerular endothelial glycocalyx, loss of lean mass and increased glomerular oxidative stress argue whether such diet regimes are safe in patients with diabetes.NEW & NOTEWORTHY Repeated fasting mimicking diet (FMD) partially prevents glomerular damage in a diabetic mouse model; however, although endothelial glycocalyx heparan sulfate contents were preserved, hyaluronan surface expression was reduced in the presence of UDP-glucuronic acid. The weight loss observed was of lean mass, not fat mass, and increased glomerular oxidative stress argue whether such a diet is safe in patients with diabetes.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Fasting , Glycocalyx , Kidney Glomerulus , Oxidative Stress , Animals , Glycocalyx/metabolism , Glycocalyx/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Blood Glucose/metabolism , Albuminuria/metabolism , Mice , Glucuronidase/metabolism , Mice, Knockout, ApoE , Mice, Inbred C57BL , Diet
10.
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
11.
Front Immunol ; 14: 1251876, 2023.
Article in English | MEDLINE | ID: mdl-37854589

ABSTRACT

Lupus nephritis (LN) is a common and severe manifestation of systemic lupus erythematosus and an important cause of acute and chronic kidney injury. Early diagnosis of LN and preventing relapses are key to preserving renal reserve. However, due to the complexity and heterogeneity of the disease, clinical management remains challenging. Kidney biopsy remains the gold standard for confirming the diagnosis of LN and subsequent assessment of kidney histopathology, but it is invasive and cannot be repeated frequently. Current clinical indicators of kidney function such as proteinuria and serum creatinine level are non-specific and do not accurately reflect histopathological changes, while anti-dsDNA antibody and C3 levels reflect immunological status but not kidney injury. Identification of novel and specific biomarkers for LN is prerequisite to improve management. Renal function deterioration is associated with changes in the endothelial glycocalyx, a delicate gel-like layer located at the interface between the endothelium and bloodstream. Inflammation induces endothelial cell activation and shedding of glycocalyx constituents into the circulation. This review discusses the potential role of soluble glycocalyx components as biomarkers of active LN, especially in patients in whom conventional serological and biochemical markers do not appear helpful.


Subject(s)
Lupus Nephritis , Humans , Lupus Nephritis/pathology , Glycocalyx/pathology , Kidney/pathology , Biomarkers , Endothelial Cells/pathology
12.
Ren Fail ; 45(1): 2188966, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37563795

ABSTRACT

BACKGROUND: Renal dysfunction and disruption of renal endothelial glycocalyx are two important events during septic acute kidney injury (AKI). Here, the role and mechanism of hyaluronidase 1 (HYAL1) in regulating renal injury and renal endothelial glycocalyx breakdown in septic AKI were explored for the first time. METHODS: BALB/c mice were injected with lipopolysaccharide (LPS, 10 mg/kg) to induce AKI. HYAL1 was blocked in vivo using lentivirus-mediated short hairpin RNA targeting HYAL1 (LV-sh-HYAL1). Biochemical assays were performed to measure the levels and concentrations of biochemical parameters associated with AKI as well as levels of inflammatory cytokines. Renal pathological lesions were determined by hematoxylin-eosin (HE) staining. Cell apoptosis in the kidney was detected using terminal-deoxynucleoitidyl transferase-mediated nick end labeling (TUNEL) assay. Immunofluorescence and immunohistochemical (IHC) staining assays were used to examine the levels of hyaluronic acid in the kidney. The protein levels of adenosine monophosphate-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling, endothelial glycocalyx, and autophagy-associated indicators were assessed by western blotting. RESULTS: The knockdown of HYAL1 in LPS-subjected mice by LV-sh-HYAL1 significantly reduced renal inflammation, oxidative stress, apoptosis and kidney dysfunction in AKI, as well as alleviated renal endothelial glycocalyx disruption by preventing the release of hyaluronic acid to the bloodstream. Additionally, autophagy-related protein analysis indicated that knockdown of HYAL1 significantly enhanced autophagy in LPS mice. Furthermore, the beneficial actions of HYAL1 blockade were closely associated with the AMPK/mTOR signaling. CONCLUSION: HYAL1 deficiency attenuates LPS-triggered renal injury and endothelial glycocalyx breakdown in septic AKI in mice.


Subject(s)
Acute Kidney Injury , Hyaluronoglucosaminidase , Animals , Mice , Acute Kidney Injury/pathology , AMP-Activated Protein Kinases , Apoptosis , Glycocalyx/metabolism , Glycocalyx/pathology , Hyaluronic Acid , Hyaluronoglucosaminidase/genetics , Kidney/pathology , Lipopolysaccharides , TOR Serine-Threonine Kinases , Mice, Inbred BALB C
13.
Clin Rheumatol ; 42(10): 2541-2555, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37581758

ABSTRACT

This narrative review provides a comprehensive examination of the complex interplay between inflammatory arthritis (IA) and cardiovascular pathology. It particularly illuminates the roles of atherosclerosis initiation, endothelial dysfunction, and glycocalyx shedding. IA not only provokes tissue-specific inflammatory responses, but also engenders a considerable degree of non-specific systemic inflammation. This review underscores the accelerating influence of the chronic inflammatory milieu of IA on cardiovascular disease (CVD) progression. A focal point of our exploration is the critical function of the endothelial glycocalyx (EG) in this acceleration process, which possibly characterizes the earliest phases of atherosclerosis. We delve into the influence of inflammatory mediators on microtubule dynamics, EG modulation, immune cell migration and activation, and lipid dysregulation. We also illuminate the impact of microparticles and microRNA on endothelial function. Further, we elucidate the role of systemic inflammation and sheddases in EG degradation, the repercussions of complement activation, and the essential role of syndecans in preserving EG integrity. Our review provides insight into the complex and dynamic interface between systemic circulation and the endothelium.


Subject(s)
Arthritis , Atherosclerosis , Cardiovascular Diseases , Humans , Endothelium, Vascular , Glycocalyx/metabolism , Glycocalyx/pathology , Risk Factors , Arthritis/pathology , Inflammation/pathology , Heart Disease Risk Factors
14.
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
15.
Methods Mol Biol ; 2674: 285-293, 2023.
Article in English | MEDLINE | ID: mdl-37258975

ABSTRACT

Vascular dysfunction is a hallmark of systemic inflammatory responses such as bacterial sepsis. The luminal surface of the blood vessels is coated with a dense layer of glycans and proteoglycans, collectively known as the glycocalyx. Surface associated glycoproteins of endothelial origin, or derived from pericytes, intravascular leukocytes, and plasma, are other important components of the glycocalyx, constituting a vascular cell surface proteome that is dynamic, tissue-specific, and sensitive to changes in vascular homeostasis, blood infection, and inflammation. Here, we describe an experimental protocol to chemically tag and quantify the vascular cell surface proteome in murine models of bacteremia, in a time-resolved and organ-specific manner. This method facilitates the identification of markers of vascular activation and provides a molecular framework to understand the contribution of vascular dysfunction to the organ pathology of systemic inflammation.


Subject(s)
Bacteremia , Proteome , Humans , Animals , Mice , Proteome/metabolism , Disease Models, Animal , Glycocalyx/pathology , Inflammation/metabolism , Endothelium, Vascular
16.
J Clin Invest ; 133(9)2023 05 01.
Article in English | MEDLINE | ID: mdl-36917196

ABSTRACT

Dysfunction of vascular endothelial cells (ECs) facilitates imbalanced immune responses and tissue hyperinflammation. However, the heterogeneous functions of skin ECs and their underlying mechanism in dermatoses remain to be determined. Here, focusing on the pathogenic role of skin ECs in psoriasis, we characterized the molecular and functional heterogeneity of skin ECs from healthy individuals and psoriasis patients at the single-cell level. We found that endothelial glycocalyx destruction, a major feature of EC dysfunction in psoriasis, was a driving force during the process of T cell extravasation. Interestingly, we identified a skin EC subset, IGFBP7hi ECs, in psoriasis. This subset actively responded to psoriatic-related cytokine signaling, secreted IGFBP7, damaged the endothelial glycocalyx, exposed the adhesion molecules underneath, and prepared the endothelium for immune-cell adhesion and transmigration, thus aggravating skin inflammation. More importantly, we provided evidence in a psoriasis-like mouse model that anti-IGFBP7 treatment showed promising therapeutic effects for restoring the endothelial glycocalyx and alleviating skin inflammation. Taken together, our results depict the distinct functions of EC clusters in healthy and psoriatic skin, identify IGFBP7hi ECs as an active subset modulating vascular function and cutaneous inflammation, and indicate that targeting IGFBP7 is a potential therapeutic strategy in psoriasis.


Subject(s)
Glycocalyx , Psoriasis , Mice , Animals , Glycocalyx/metabolism , Glycocalyx/pathology , Endothelial Cells/metabolism , Psoriasis/pathology , T-Lymphocytes , Inflammation/metabolism
17.
Am J Pathol ; 193(4): 474-492, 2023 04.
Article in English | MEDLINE | ID: mdl-36669683

ABSTRACT

The outer layer of endothelial cells (ECs), consisting of the endothelial glycocalyx (eGC) and the cortex (CTX), provides a protective barrier against vascular diseases. Structural and functional impairments of their mechanical properties are recognized as hallmarks of endothelial dysfunction and can lead to cardiovascular events, such as acute myocardial infarction (AMI). This study investigated the effects of AMI on endothelial nanomechanics and function and the use of exogenous recombinant syndecan-1 (rSyn-1), a major component of the eGC, as recovering agent. ECs were exposed in vitro to serum samples collected from patients with AMI. In addition, in situ ECs of ex vivo aorta preparations derived from a mouse model for AMI were employed. Effects were quantified by using atomic force microscopy-based nanoindentation measurements, fluorescence staining, and histologic examination of the mouse hearts. AMI serum samples damaged eGC/CTX and augmented monocyte adhesion to the endothelial surface. In particular, the anaphylatoxins C3a and C5a played an important role in these processes. The impairment of endothelial function could be prevented by rSyn-1 treatment. In the mouse model of myocardial infarction, pretreatment with rSyn-1 alleviated eGC/CTX deterioration and reduced cardiomyocyte damage in histologic analyses. However, echocardiographic measurements did not indicate a functional benefit. These results provide new insights into the underlying mechanisms of AMI-induced endothelial dysfunction and perspectives for future studies on the benefit of rSyn-1 in post-AMI treatment.


Subject(s)
Endothelial Cells , Myocardial Infarction , Animals , Mice , Endothelial Cells/pathology , Glycocalyx/pathology , Syndecan-1 , Myocytes, Cardiac , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology
18.
Respir Res ; 23(1): 269, 2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36183124

ABSTRACT

BACKGROUND: The defects and imbalance in lung repair and structural maintenance contribute to the pathogenesis of chronic obstructive pulmonary diseases (COPD), yet the molecular mechanisms that regulate lung repair process are so far incompletely understood. We hypothesized that cigarette smoking causes glycocalyx impairment and endothelial apoptosis in COPD, which could be repaired by the stimulation of fibroblast growth factor 10 (FGF10)/FGF receptor 1 (FGFR1) signaling. METHODS: We used immunostaining (immunohistochemical [IHC] and immunofluorescence [IF]) and enzyme-linked immunosorbent assay (ELISA) to detect the levels of glycocalyx components and endothelial apoptosis in animal models and in patients with COPD. We used the murine emphysema model and in vitro studies to determine the protective and reparative role of FGF10/FGFR1. RESULTS: Exposure to cigarette smoke caused endothelial glycocalyx impairment and emphysematous changes in murine models and human specimens. Pretreatment of FGF10 attenuated the development of emphysema and the shedding of glycocalyx components induced by CSE in vivo. However, FGF10 did not attenuate the emphysema induced by endothelial-specific killing peptide CGSPGWVRC-GG-D(KLAKLAK)2. Mechanistically, FGF10 alleviated smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling in vitro. FGF10 was shown to repair pulmonary glycocalyx injury and endothelial apoptosis, and attenuate smoke-induced COPD through FGFR1 signaling. CONCLUSIONS: Our results suggest that FGF10 may serve as a potential therapeutic strategy against COPD via endothelial repair and glycocalyx reconstitution.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Animals , Apoptosis/physiology , Emphysema/complications , Fibroblast Growth Factor 10 , Glycocalyx/metabolism , Glycocalyx/pathology , Humans , Mice , Pulmonary Emphysema/metabolism , Receptors, Fibroblast Growth Factor/therapeutic use , Nicotiana
19.
Biomed Pharmacother ; 155: 113666, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36099790

ABSTRACT

Acute lung injury (ALI) and its more serious form; acute respiratory distress syndrome are major causes of COVID-19 related mortality. Finding new therapeutic targets for ALI is thus of great interest. This work aimed to prepare a biocompatible nanoformulation for effective pulmonary delivery of the herbal drug; tanshinone-IIA (TSIIA) for ALI management. A nanoemulsion (NE) formulation based on bioactive natural ingredients; rhamnolipid biosurfactant and tea-tree oil, was developed using a simple ultrasonication technique, optimized by varying oil concentration and surfactant:oil ratio. The selected TSIIA-NE formulation showed 105.7 nm diameter and a PDI âˆ¼ 0.3. EE exceeded 98 % with biphasic sustained drug release and good stability over 3-months. In-vivo efficacy was evaluated in lipopolysaccharide (LPS)-induced ALI model. TSIIA-NE (30 µg/kg) was administered once intratracheally 2 h after LPS instillation. Evaluation was performed 7days post-treatment. Pulmonary function assessment, inflammatory, oxidative stress and glycocalyx shedding markers analysis in addition to histopathological examination of lung tissue were performed. When compared to untreated rats, in-vivo efficacy study demonstrated 1.4 and 1.9-fold increases in tidal volume and minute respiratory volume, respectively, with 32 % drop in wet/dry lung weight ratio and improved levels of arterial blood gases. Lung histopathology and biochemical analysis of different biomarkers in tissue homogenate and bronchoalveolar lavage fluid indicated that treatment may ameliorate LPS-induced ALI symptoms thorough anti-oxidative, anti-inflammatory effects and inhibition of glycocalyx degradation. TSIIA-NE efficacy was superior to free medication and blank-NE. The enhanced efficacy of TSIIA bioactive nanoemulsion significantly suggests the pharmacotherapeutic potential of bioactive TSIIA-NE as a promising nanoplatform for ALI.


Subject(s)
Acute Lung Injury , COVID-19 Drug Treatment , Rats , Animals , Lipopolysaccharides/pharmacology , Glycocalyx/pathology , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Lung , Anti-Inflammatory Agents/pharmacology , Surface-Active Agents/pharmacology , Gases/adverse effects , Gases/metabolism , Tea/metabolism
20.
Compr Physiol ; 12(4): 3781-3811, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35997082

ABSTRACT

The glycocalyx is a polysaccharide structure that protrudes from the body of a cell. It is primarily conformed of glycoproteins and proteoglycans, which provide communication, electrostatic charge, ionic buffering, permeability, and mechanosensation-mechanotransduction capabilities to cells. In blood vessels, the endothelial glycocalyx that projects into the vascular lumen separates the vascular wall from the circulating blood. Such a physical location allows a number of its components, including sialic acid, glypican-1, heparan sulfate, and hyaluronan, to participate in the mechanosensation-mechanotransduction of blood flow-dependent shear stress, which results in the synthesis of nitric oxide and flow-mediated vasodilation. The endothelial glycocalyx also participates in the regulation of vascular permeability and the modulation of inflammatory responses, including the processes of leukocyte rolling and extravasation. Its structural architecture and negative charge work to prevent macromolecules greater than approximately 70 kDa and cationic molecules from binding and flowing out of the vasculature. This also prevents the extravasation of pathogens such as bacteria and virus, as well as that of tumor cells. Due to its constant exposure to shear and circulating enzymes such as neuraminidase, heparanase, hyaluronidase, and matrix metalloproteinases, the endothelial glycocalyx is in a continuous process of degradation and renovation. A balance favoring degradation is associated with a variety of pathologies including atherosclerosis, hypertension, vascular aging, metastatic cancer, and diabetic vasculopathies. Consequently, ongoing research efforts are focused on deciphering the mechanisms that promote glycocalyx degradation or limit its syntheses, as well as on therapeutic approaches to improve glycocalyx integrity with the goal of reducing vascular disease. © 2022 American Physiological Society. Compr Physiol 12: 1-31, 2022.


Subject(s)
Glycocalyx , Mechanotransduction, Cellular , Endothelium, Vascular/physiology , Glycocalyx/metabolism , Glycocalyx/pathology , Heparitin Sulfate/metabolism , Humans , Mechanotransduction, Cellular/physiology , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL