Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.140
Filter
1.
J Ethnopharmacol ; 336: 118711, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181286

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: According to the theory of Qi and blood in Traditional Chinese Medicine (TCM), the combination of Qi-reinforcing herbs and blood-activating herbs has a synergistic effect in improving blood stasis syndrome, especially in tumor treatment. The classic "Radix Astragali - Salvia miltiorrhiza" duo exemplifies this principle, renowned for invigorating Qi and activating blood flow, employed widely in tumor therapies. Our prior research underscores the potent inhibition of pancreatic tumor xenografts by the combination of Formononetin (from Radix Astragali) and Salvianolic acid B (from Salvia miltiorrhiza) in vitro. However, it remains unclear whether this combination can inhibit the abnormal vascularization of pancreatic tumors to achieve its anti-cancer effect. AIM OF THE STUDY: Abnormal vasculature, known to facilitate tumor growth and metastasis. Strategies to normalize tumor-associated blood vessels provide a promising avenue for anti-tumor therapy. This study aimed to unravel the therapeutic potential of Formononetin combined with Salvianolic acid B (FcS) in modulating pancreatic cancer's impact on endothelial cells, illuminate the underlying mechanisms that govern this therapeutic interaction, thereby advancing strategies to normalize tumor vasculature and combat cancer progression. MATERIALS AND METHODS: A co-culture system involving Human Umbilical Vein Endothelial Cells (HUVECs) and PANC-1 cells was established to investigate the potential of targeting abnormal vasculature as a novel anti-tumor therapeutic strategy. We systematically compared HUVEC proliferation, migration, invasion, and lumenogenesis in both mono- and co-culture conditions with PANC-1 (H-P). Subsequently, FcS treatment of the H-P system was evaluated for its anti-angiogenic properties. Molecular docking was utilized to predict the interactions between Formononetin and Salvianolic acid B with RhoA, and the post-treatment expression of RhoA in HUVECs was assessed. Furthermore, we utilized shRhoA lentivirus to elucidate the role of RhoA in FcS-mediated effects on HUVECs. In vivo, a zebrafish xenograft tumor model was employed to assess FcS's anti-tumor potential, focusing on cancer cell proliferation, migration, apoptosis, and vascular development. RESULTS: FcS treatment demonstrated a significant, dose-dependent inhibition of PANC-1-induced alterations in HUVECs, including proliferation, migration, invasion, and tube formation capabilities. Molecular docking analyses indicated potential interactions between FcS and RhoA. Further, FcS treatment was found to downregulate RhoA expression and modulated the PI3K/AKT signaling pathway in PANC-1-induced HUVECs. Notably, the phenotypic inhibitory effects of FcS on HUVECs were attenuated by RhoA knockdown. In vivo zebrafish studies validated FcS's anti-tumor activity, inhibiting cancer cell proliferation, metastasis, and vascular sprouting, while promoting tumor cell apoptosis. CONCLUSIONS: This study underscores the promising potential of FcS in countering pancreatic cancer-induced endothelial alterations. FcS exhibits pronounced anti-abnormal vasculature effects, potentially achieved through downregulation of RhoA and inhibition of the PI3K/Akt signaling pathway, thereby presenting a novel therapeutic avenue for pancreatic cancer management.


Subject(s)
Benzofurans , Cell Movement , Human Umbilical Vein Endothelial Cells , Isoflavones , Pancreatic Neoplasms , rhoA GTP-Binding Protein , Isoflavones/pharmacology , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Benzofurans/pharmacology , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/drug effects , Cell Movement/drug effects , Neovascularization, Pathologic/drug therapy , Zebrafish , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Depsides
2.
Elife ; 132024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365738

ABSTRACT

Artificially sweetened beverages containing noncaloric monosaccharides were suggested as healthier alternatives to sugar-sweetened beverages. Nevertheless, the potential detrimental effects of these noncaloric monosaccharides on blood vessel function remain inadequately understood. We have established a zebrafish model that exhibits significant excessive angiogenesis induced by high glucose, resembling the hyperangiogenic characteristics observed in proliferative diabetic retinopathy (PDR). Utilizing this model, we observed that glucose and noncaloric monosaccharides could induce excessive formation of blood vessels, especially intersegmental vessels (ISVs). The excessively branched vessels were observed to be formed by ectopic activation of quiescent endothelial cells (ECs) into tip cells. Single-cell transcriptomic sequencing analysis of the ECs in the embryos exposed to high glucose revealed an augmented ratio of capillary ECs, proliferating ECs, and a series of upregulated proangiogenic genes. Further analysis and experiments validated that reduced foxo1a mediated the excessive angiogenesis induced by monosaccharides via upregulating the expression of marcksl1a. This study has provided new evidence showing the negative effects of noncaloric monosaccharides on the vascular system and the underlying mechanisms.


Consuming too much sugar can damage blood vessels and contribute to diseases like diabetes and heart disease. Artificial sweeteners have been suggested as a healthier alternative, and are now included in many products like sodas and baked goods. However, some studies have suggested that people who consume large amounts of artificial sweeteners also have an increased risk of cardiovascular disease. Others suggest individuals may also experience spikes in blood sugar levels similar to those observed in people with diabetes. Yet few studies have examined how artificial sweeteners affect the network of vessels that transport blood and other substances around the body. To investigate this question, Wang, Zhao, Xu, et al. studied zebrafish embryos which had been exposed to sugar and a type of artificial sweetener known as non-caloric monosaccharides. Various imaging tools revealed that high levels of sugar caused the embryos to produce more new blood vessels via a process called angiogenesis. This excessive growth of blood vessels has previously been linked to diabetic complications, including cardiovascular disease. Wang, Zhao, Xu, et al. found that zebrafish embryos exposed to several different non-caloric monosaccharides developed similar blood vessel problems. All the sweeteners tested caused immature cells lining the blood vessels to develop into active tip cells that promote angiogenesis. This led to more new blood vessels forming that branch off already existing veins and arteries. These findings suggest that artificial sweeteners may cause the same kind of damage to blood vessels as sugar. This may explain why people who consume a lot of artificial sweeteners are at risk of developing heart disease and high blood sugar levels. Future studies could help scientists learn more about how genetics or other factors affect the health impact of sugars and artificial sweeteners. This may lead to a greater understanding of the long-term health effects of artificially sweetened foods.


Subject(s)
Forkhead Box Protein O1 , Monosaccharides , Neovascularization, Physiologic , Zebrafish , Animals , Neovascularization, Physiologic/drug effects , Monosaccharides/metabolism , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Glucose/metabolism , Glucose/pharmacology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Signal Transduction , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Angiogenesis
3.
J Nanobiotechnology ; 22(1): 620, 2024 Oct 12.
Article in English | MEDLINE | ID: mdl-39396002

ABSTRACT

Endothelial cells (ECs) are pivotal in maintaining vascular health, regulating hemodynamics, and modulating inflammatory responses. Nanocarriers hold transformative potential for precise drug delivery within the vascular system, particularly targeting ECs for therapeutic purposes. However, the complex interactions between vascular ECs and nanocarriers present significant challenges for the development and clinical translation of nanotherapeutics. This review assesses recent advancements and key strategies in employing nanocarriers for drug delivery to vascular ECs. It suggested that through precise physicochemical design and surface modifications, nanocarriers can enhance targeting specificity and improve drug internalization efficiency in ECs. Additionally, we elaborated on the applications of nanocarriers specifically designed for targeting ECs in the treatment of cardiovascular diseases, cancer metastasis, and inflammatory disorders. Despite these advancements, safety concerns, the complexity of in vivo processes, and the challenge of achieving subcellular drug delivery remain significant obstacles to the effective targeting of ECs with nanocarriers. A comprehensive understanding of endothelial cell biology and its interaction with nanocarriers is crucial for realizing the full potential of targeted drug delivery systems.


Subject(s)
Drug Carriers , Drug Delivery Systems , Endothelial Cells , Nanoparticles , Humans , Drug Carriers/chemistry , Animals , Drug Delivery Systems/methods , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Nanoparticles/chemistry , Endothelium, Vascular/drug effects , Cardiovascular Diseases/drug therapy , Neoplasms/drug therapy
4.
Biomed Res ; 45(5): 197-207, 2024.
Article in English | MEDLINE | ID: mdl-39370298

ABSTRACT

Our study explored the therapeutic effect and the mechanism of quercetin against hypoxia/reoxygenation (H/R)-induced injury in human coronary artery endothelial cells (CAECs). Quercetin was selected as a potential component for the BuShenKangShuaiPian formula (BSKSP) treatment via the Network pharmacology analysis. Cell viability and reactive oxygen species (ROS) production were measured by CCK8 assay and immunofluorescence, respectively. The expression of Bax, Bcl-2, Cle-caspase-3, cytochrome c (Cyt-C), NF-E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) protein was quantified by western blotting. The superoxide dismutase (SOD), catalase (CAT), malondialdehyde (MDA) activity, mtDNA copy number, and ATP production were measured via corresponding kits. Quercetin was selected from the BSKSP for its high degree value (Degree value: 22). Besides, quercetin protected CAECs against H/R-induced cytotoxicity and apoptosis. The H/R-induced increased ROS level, ATP production, Cyt-C release, and decreased mtDNA copy number were removed by the quercetin. Moreover, quercetin upregulated the Nrf2/ HO-1 axis, SOD, and CAT activity, and downregulated MDA levels in H/R treated CAECs, while knockdown Nrf2 reversed the protection of quercetin against H/R-induced oxidative stress, mitochondrial damage, and apoptosis. Quercetin protects CAECs against H/R-induced mitochondrial apoptosis via the Nrf2/HO-1 axis, which innovatively suggests the therapeutic potential of quercetin for coronary heart disease (CHD) treatment.


Subject(s)
Apoptosis , Coronary Vessels , Endothelial Cells , Heme Oxygenase-1 , Mitochondria , NF-E2-Related Factor 2 , Quercetin , Reactive Oxygen Species , Signal Transduction , Humans , Quercetin/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics , Reactive Oxygen Species/metabolism , Coronary Vessels/cytology , Coronary Vessels/metabolism , Coronary Vessels/drug effects , Signal Transduction/drug effects , Cell Survival/drug effects , Oxidative Stress/drug effects , Cell Hypoxia/drug effects
5.
J Biochem Mol Toxicol ; 38(9): e23843, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39253885

ABSTRACT

Hypertensive intracerebral hemorrhage (HICH) is a destructive disease with high mortality, incidence, and disability. Asiaticoside (AC) is a triterpenoid derivative that has demonstrated to exert a protective effect on neuron and blood vessel. To investigate the function and potential mechanism of AC on HICH. Human brain microvascular endothelial cells (hBMECs) were treated with 20 U/mL thrombin for 24 h to establish the HICH model in vitro, and AC with the concentration of 1, 2 and 4 µM were used to incubate hBMECs. The effect and potential mechanism of AC on HICH were investigated by using cell counting kit-8, flow cytometry, tube forming assays, vascular permeability experiments and western blot assays. In vivo, rats were injected with 20 µL hemoglobin with a concentration of 150 mg/mL, and then intragastrically administrated with 1.25, 2.5 and 5 mg/kg AC. Behavioral tests, brain water content measurement, hematoxylin-eosin (HE) staining, terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling assays, and western blot were used to assess the effect and potential mechanism of AC on HICH. AC (at 2 and 4 µM) improved the proliferation, apoptosis, angiogenesis and vascular permeability in thrombin-induced hBMECs (p < 0.05). Besides, AC (2.5 and 5 mg/kg) ameliorated behavioral scores, brain water content, pathological lesion, apoptosis and the expression of vascular permeability-related proteins in rats with HICH (p < 0.05). In addition, AC elevated the expression of PI3K/AKT pathway after HICH both in cell and animal models (p < 0.05). Application of LY294002, an inhibitor of PI3K/AKT pathway, reversed the ameliorative effect of AC on the proliferation, apoptosis, angiogenesis and vascular permeability in thrombin-induced hBMECs (p < 0.05). AC reduced brain damage by increasing the expression of the PI3K/AKT pathway after HICH.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Signal Transduction , Triterpenes , Animals , Proto-Oncogene Proteins c-akt/metabolism , Rats , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Male , Humans , Triterpenes/pharmacology , Intracranial Hemorrhage, Hypertensive/drug therapy , Intracranial Hemorrhage, Hypertensive/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Apoptosis/drug effects , Neuroprotective Agents/pharmacology , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/prevention & control , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebral Hemorrhage/complications
6.
Cardiovasc Diabetol ; 23(1): 331, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252021

ABSTRACT

BACKGROUND: Visceral adipose tissue in individuals with obesity is an independent cardiovascular risk indicator. However, it remains unclear whether adipose tissue influences common cardiovascular diseases, such as atherosclerosis, through its secreted exosomes. METHODS: The exosomes secreted by adipose tissue from diet-induced obesity mice were isolated to examine their impact on the progression of atherosclerosis and the associated mechanism. Endothelial apoptosis and the proliferation and migration of vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque were evaluated. Statistical significance was analyzed using GraphPad Prism 9.0 with appropriate statistical tests. RESULTS: We demonstrate that adipose tissue-derived exosomes (AT-EX) exacerbate atherosclerosis progression by promoting endothelial apoptosis, proliferation, and migration of VSMCs within the plaque in vivo. MicroRNA-132/212 (miR-132/212) was detected within AT-EX cargo. Mechanistically, miR-132/212-enriched AT-EX exacerbates palmitate acid-induced endothelial apoptosis via targeting G protein subunit alpha 12 and enhances platelet-derived growth factor type BB-induced VSMC proliferation and migration by targeting phosphatase and tensin homolog in vitro. Importantly, melatonin decreases exosomal miR-132/212 levels, thereby mitigating the pro-atherosclerotic impact of AT-EX. CONCLUSION: These data uncover the pathological mechanism by which adipose tissue-derived exosomes regulate the progression of atherosclerosis and identify miR-132/212 as potential diagnostic and therapeutic targets for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Exosomes , Mice, Inbred C57BL , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Male , Signal Transduction , Cells, Cultured , Obesity/metabolism , Obesity/pathology , Mice, Knockout, ApoE , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/drug effects , Aortic Diseases/pathology , Aortic Diseases/metabolism , Aortic Diseases/genetics , Becaplermin/pharmacology , Becaplermin/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Mice , Humans
7.
Anal Cell Pathol (Amst) ; 2024: 2348336, 2024.
Article in English | MEDLINE | ID: mdl-39290461

ABSTRACT

Background: The present study aimed to analyze the impact of astragaloside IV (AS-IV) on abdominal aortic aneurysm (AAA) and the glycocalyx, elucidating the potential mechanism of AS-IV. Methods: Rat models of AAA were established using porcine pancreatic elastase. The effects of intraperitoneal AS-IV injection on the morphology, diameter, and glycocalyx of the aorta and the expression of miR-17-3p and Syndecan-1 (SDC1) protein were examined. Differentially expressed miRNAs from peripheral blood samples of healthy individuals, untreated patients with AAA, and treated patients with AAA were identified through sequencing. The relationship between miR-17-3p and SDC1 was validated using a dual-luciferase reporter assay. In vitro, shear stress was induced in human aortic endothelial cells (HAECs) to simulate AAA. Overexpression of miR-17-3p was performed to assess the effects of AS-IV on miR-17-3p and SDC1 expressions, apoptosis, and glycocalyx in HAECs. Results: AS-IV mitigated aortic damage in AAA rats, reducing the aortic diameter and alleviating glycocalyx damage. In addition, it suppressed the increase in miR-17-3p expression and promoted SDC1 expression in AAA rats. Peripheral blood miR-17-3p levels were significantly higher in patients with AAA than in healthy individuals. miR-17-3p inhibited the SDC1 protein expression in HAECs. In the in vitro AAA environment, miR-17-3p was upregulated and SDC1 was downregulated in HAECs. AS-IV inhibited miR-17-3p expression, promoted SDC1 expression, and mitigated shear stress-induced apoptosis and glycocalyx damage in HAECs. Overexpression of miR-17-3p blocked AS-IV-induced SDC1 expression promotion, glycocalyx protection, and apoptosis suppression in HAECs. Conclusion: miR-17-3p may damage the glycocalyx of aortic endothelial cells by targeting SDC1. AS-IV may promote SDC1 expression by inhibiting miR-17-3p, thereby protecting the glycocalyx and alleviating AAA.


Subject(s)
Aortic Aneurysm, Abdominal , Glycocalyx , MicroRNAs , Rats, Sprague-Dawley , Saponins , Stress, Mechanical , Syndecan-1 , Triterpenes , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Saponins/pharmacology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Syndecan-1/metabolism , Humans , Triterpenes/pharmacology , Male , Rats , Glycocalyx/metabolism , Glycocalyx/drug effects , Apoptosis/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Disease Models, Animal , Protective Agents/pharmacology , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Abdominal/drug effects
8.
Cell Death Dis ; 15(9): 650, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231943

ABSTRACT

Acid sphingomyelinase (ASM) inhibitors are widely used for the treatment of post-stroke depression. They promote neurological recovery in animal stroke models via neurorestorative effects. In a previous study, we found that antidepressants including amitriptyline, fluoxetine, and desipramine increase cerebral angiogenesis post-ischemia/reperfusion (I/R) in an ASM-dependent way. To elucidate the underlying mechanisms, we investigated the effects of the functional ASM inhibitor amitriptyline in two models of I/R injury, that is, in human cerebral microvascular endothelial hCMEC/D3 cells exposed to oxygen-glucose deprivation and in mice exposed to middle cerebral artery occlusion (MCAO). In addition to our earlier studies, we now show that amitriptyline increased mitochondrial reactive oxygen species (ROS) formation in hCMEC/D3 cells and increased ROS formation in the vascular compartment of MCAO mice. ROS formation was instrumental for amitriptyline's angiogenic effects. ROS formation did not result in excessive endothelial injury. Instead, amitriptyline induced a profound metabolic reprogramming of endothelial cells that comprised reduced endothelial proliferation, reduced mitochondrial energy metabolism, reduced endoplasmic reticulum stress, increased autophagy/mitophagy, stimulation of antioxidant responses and inhibition of apoptotic cell death. Specifically, the antioxidant heme oxygenase-1, which was upregulated by amitriptyline, mediated amitriptyline's angiogenic effects. Thus, heme oxygenase-1 knockdown severely compromised angiogenesis and abolished amitriptyline's angiogenic responses. Our data demonstrate that ASM inhibition reregulates a complex network of metabolic and mitochondrial responses post-I/R that contribute to cerebral angiogenesis without compromising endothelial survival.


Subject(s)
Amitriptyline , Endothelial Cells , Mitochondria , Oxidative Stress , Reactive Oxygen Species , Reperfusion Injury , Sphingomyelin Phosphodiesterase , Animals , Sphingomyelin Phosphodiesterase/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Humans , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Reactive Oxygen Species/metabolism , Amitriptyline/pharmacology , Mice , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Male , Mice, Inbred C57BL , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/metabolism , Cell Survival/drug effects , Neovascularization, Physiologic/drug effects , Cell Line , Angiogenesis
9.
Cell Mol Life Sci ; 81(1): 401, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269632

ABSTRACT

Methylglyoxal (MGO), a reactive dicarbonyl metabolite of glucose, plays a prominent role in the pathogenesis of diabetes and vascular complications. Our previous studies have shown that MGO is associated with increased oxidative stress, inflammatory responses and apoptotic cell death in endothelial cells (ECs). Pyroptosis is a novel form of inflammatory caspase-1-dependent programmed cell death that is closely associated with the activation of the NOD-like receptor 3 (NLRP3) inflammasome. Recent studies have shown that sulforaphane (SFN) can inhibit pyroptosis, but the effects and underlying mechanisms by which SFN affects MGO-induced pyroptosis in endothelial cells have not been determined. Here, we found that SFN prevented MGO-induced pyroptosis by suppressing oxidative stress and inflammation in vitro and in vivo. Our results revealed that SFN dose-dependently prevented MGO-induced HUVEC pyroptosis, inhibited pyroptosis-associated biochemical changes, and attenuated MGO-induced morphological alterations in mitochondria. SFN pretreatment significantly suppressed MGO-induced ROS production and the inflammatory response by inhibiting the NLRP3 inflammasome (NLRP3, ASC, and caspase-1) signaling pathway by activating Nrf2/HO-1 signaling. Similar results were obtained in vivo, and we demonstrated that SFN prevented MGO-induced oxidative damage, inflammation and pyroptosis by reversing the MGO-induced downregulation of the NLRP3 signaling pathway through the upregulation of Nrf2. Additionally, an Nrf2 inhibitor (ML385) noticeably attenuated the protective effects of SFN on MGO-induced pyroptosis and ROS generation by inhibiting the Nrf2/HO-1 signaling pathway, and a ROS scavenger (NAC) and a permeability transition pore inhibitor (CsA) completely reversed these effects. Moreover, NLRP3 inhibitor (MCC950) and caspase-1 inhibitor (VX765) further reduced pyroptosis in endothelial cells that were pretreated with SFN. Collectively, these findings broaden our understanding of the mechanism by which SFN inhibits pyroptosis induced by MGO and suggests important implications for the potential use of SFN in the treatment of vascular diseases.


Subject(s)
Glucose , Human Umbilical Vein Endothelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Pyroptosis , Pyruvaldehyde , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Humans , Oxidative Stress/drug effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Glucose/metabolism , Isothiocyanates/pharmacology , Mice , Sulfoxides/pharmacology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
10.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273330

ABSTRACT

In this study, we assessed the impact of hepatocyte growth factor (HGF) on corneal endothelial cells (CECs), finding that HGF concentrations of 100-250 ng/mL significantly increased CEC proliferation by 30%, migration by 32% and improved survival under oxidative stress by 28% compared to untreated controls (p < 0.05). The primary objective was to identify non-fibrotic pharmacological strategies to enhance corneal endothelial regeneration, addressing a critical need in conditions like Fuchs' endothelial dystrophy (FED), where donor tissue is scarce. To confirm the endothelial nature of the cultured CECs, Na+/K+-ATPase immunohistochemistry was performed. Proliferation rates were determined through BrdU incorporation assays, while cell migration was assessed via scratch assays. Cell viability was evaluated under normal and oxidative stress conditions using WST-1 assays. To ensure that HGF treatment did not trigger epithelial-mesenchymal transition, which could lead to undesirable fibrotic changes, α-SMA staining was conducted. These comprehensive methodologies provided robust data on the effects of HGF, confirming its potential as a therapeutic agent for corneal endothelial repair without inducing harmful EMT, as indicated by the absence of α-SMA expression. These findings suggest that HGF holds therapeutic promise for enhancing corneal endothelial repair, warranting further investigation in in vivo models to confirm its clinical applicability.


Subject(s)
Cell Movement , Cell Proliferation , Endothelium, Corneal , Hepatocyte Growth Factor , Wound Healing , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Humans , Wound Healing/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cell Survival/drug effects , Cells, Cultured , Oxidative Stress/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fuchs' Endothelial Dystrophy/drug therapy , Fuchs' Endothelial Dystrophy/metabolism , Fuchs' Endothelial Dystrophy/pathology , Sodium-Potassium-Exchanging ATPase/metabolism
11.
Part Fibre Toxicol ; 21(1): 38, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39300536

ABSTRACT

BACKGROUND: The formation of secondary organic aerosols (SOA) by atmospheric oxidation reactions substantially contributes to the burden of fine particulate matter (PM2.5), which has been associated with adverse health effects (e.g., cardiovascular diseases). However, the molecular and cellular effects of atmospheric aging on aerosol toxicity have not been fully elucidated, especially in model systems that enable cell-to-cell signaling. METHODS: In this study, we aimed to elucidate the complexity of atmospheric aerosol toxicology by exposing a coculture model system consisting of an alveolar (A549) and an endothelial (EA.hy926) cell line seeded in a 3D orientation at the air‒liquid interface for 4 h to model aerosols. Simulation of atmospheric aging was performed on volatile biogenic (ß-pinene) or anthropogenic (naphthalene) precursors of SOA condensing on soot particles. The similar physical properties for both SOA, but distinct differences in chemical composition (e.g., aromatic compounds, oxidation state, unsaturated carbonyls) enabled to determine specifically induced toxic effects of SOA. RESULTS: In A549 cells, exposure to naphthalene-derived SOA induced stress-related airway remodeling and an early type I immune response to a greater extent. Transcriptomic analysis of EA.hy926 cells not directly exposed to aerosol and integration with metabolome data indicated generalized systemic effects resulting from the activation of early response genes and the involvement of cardiovascular disease (CVD) -related pathways, such as the intracellular signal transduction pathway (PI3K/AKT) and pathways associated with endothelial dysfunction (iNOS; PDGF). Greater induction following anthropogenic SOA exposure might be causative for the observed secondary genotoxicity. CONCLUSION: Our findings revealed that the specific effects of SOA on directly exposed epithelial cells are highly dependent on the chemical identity, whereas non directly exposed endothelial cells exhibit more generalized systemic effects with the activation of early stress response genes and the involvement of CVD-related pathways. However, a greater correlation was made between the exposure to the anthropogenic SOA compared to the biogenic SOA. In summary, our study highlights the importance of chemical aerosol composition and the use of cell systems with cell-to-cell interplay on toxicological outcomes.


Subject(s)
Aerosols , Coculture Techniques , Epithelial Cells , Particulate Matter , Signal Transduction , Transcriptome , Humans , Particulate Matter/toxicity , Signal Transduction/drug effects , Transcriptome/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , A549 Cells , Air Pollutants/toxicity , Metabolomics , Metabolome/drug effects
12.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(9): 967-973, 2024.
Article in Chinese | MEDLINE | ID: mdl-39267513

ABSTRACT

OBJECTIVES: To study the effects and mechanisms of tetramethylpyrazine (TMP) on tumor necrosis factor-α (TNF-α)-induced inflammatory injury in human coronary artery endothelial cells (HCAEC). METHODS: HCAEC were randomly divided into four groups: the control group (no treatment), the model group (treated with TNF-α, 50 ng/mL for 24 hours), the TMP group (pre-treated with TMP, 80 µg/mL for 12 hours followed by TNF-α treatment for 24 hours), and the SIRT1 inhibitor group (pre-treated with TMP and the specific SIRT1 inhibitor EX527 for 12 hours followed by TNF-α treatment for 24 hours). Cell viability was assessed using the CCK-8 method, lactate dehydrogenase (LDH) activity was measured using an LDH assay kit, reactive oxygen species (ROS) levels were observed using DCFH-DA staining, expression of pyroptosis-related proteins was detected by Western blot, and SIRT1 expression was analyzed using immunofluorescence staining. RESULTS: Compared to the control group, the model group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). Compared to the model group, the TMP group exhibited increased cell viability, decreased LDH activity, ROS level and expression of pyroptosis-related proteins, and increased SIRT1 expression (P<0.05). In comparison to the TMP group, the SIRT1 inhibitor group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). CONCLUSIONS: TMP may attenuate TNF-α-induced inflammatory injury in HCAEC, which is associated with the inhibition of pyroptosis and activation of the SIRT1 signaling pathway.


Subject(s)
Endothelial Cells , Pyrazines , Reactive Oxygen Species , Signal Transduction , Sirtuin 1 , Tumor Necrosis Factor-alpha , Sirtuin 1/metabolism , Sirtuin 1/physiology , Humans , Pyrazines/pharmacology , Signal Transduction/drug effects , Endothelial Cells/drug effects , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Cell Survival/drug effects , Pyroptosis/drug effects , Cells, Cultured , Inflammation/drug therapy
13.
Pak J Pharm Sci ; 37(4): 731-741, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39348637

ABSTRACT

The study proposed improving the arsenic encapsulation efficiency (EE) in liposomes and make it pH responsive. Liposomes were prepared using the ethanol injection method (EIM), thin film dispersion method (TFM) and CAGM with sodium arsenite (NaAsO2). The orthogonal experimental was used to optimize the preparation conditions of the CAGM. The arsenic-carrying liposomes were characterized by polydispersity index (PDI), transmission electron microscopy (TEM), in vitro release experiments and inductively coupled plasma emission spectrum (ICP). The toxicity was investigated by rat glioma cells (C6) and human brain micro vascular endothelial cells (HBMEC). The results indicated that the CAGM can effectively improve the EE of NaAsO2 and has a pH response compared with EIM and TFM. The size of nanoparticles prepared by CAGM was 118.8 ±56.67 nm, the arsenic EE was 54.3 ±9.82%, the drug loading rate was 7.13 ±0.72% (P <0.01), pH sensitivity was shown at pH 5.5. The optimal parameters of the CAGM were 3 mg NaAsO2, 5:1 egg phosphatidylcholine (EPC) to cholesterol (CHOL) and 240 mmol/L calcium acetate (CaAC2). The results showed that the CAGM has good biocompatibility and is one of the effective ways to improve the NaAsO2 encapsulation rate and pH response in liposome nanoparticles.


Subject(s)
Liposomes , Nanoparticles , Hydrogen-Ion Concentration , Animals , Rats , Humans , Arsenites/toxicity , Cell Line, Tumor , Arsenic/toxicity , Drug Compounding , Cell Survival/drug effects , Particle Size , Sodium Compounds/chemistry , Sodium Compounds/toxicity , Drug Liberation , Endothelial Cells/drug effects , Endothelial Cells/metabolism
14.
Nutrients ; 16(18)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39339760

ABSTRACT

Endothelial dysfunction occurs prior to atherosclerosis, which is an independent predictor of cardiovascular diseases (CVDs). Diabetes mellitus impairs endothelial function by triggering oxidative stress and inflammation in vascular tissues. Isoliquiritigenin (ISL), one of the major bioactive ingredients extracted from licorice, has been reported to inhibit inflammation and oxidative stress. However, the therapeutic effects of ISL on ameliorating type 2 diabetes (T2D)-associated endothelial dysfunction remain unknown. In our animal study, db/db male mice were utilized as a model for T2D-associated endothelial dysfunction, while their counterpart, heterozygote db/m+ male mice, served as the control. Mouse brain microvascular endothelial cells (mBMECs) were used for in vitro experiments. Interleukin-1ß (IL-1ß) was used to induce endothelial cell dysfunction. ISL significantly reversed the impairment of endothelium-dependent relaxations (EDRs) in db/db mouse aortas. ISL treatment decreased ROS (reactive oxygen species) levels in db/db mice aortic sections and IL-1ß-treated endothelial cells. Encouragingly, ISL attenuated the overexpression of pro-inflammatory factors MCP-1, TNF-α, and IL-6 in db/db mouse aortas and IL-1ß-impaired endothelial cells. The NOX2 (NADPH oxidase 2) overexpression was inhibited by ISL treatment. Notably, ISL treatment restored the expression levels of IL-10, SOD1, Nrf2, and HO-1 in db/db mouse aortas and IL-1ß-impaired endothelial cells. This study illustrates, for the first time, that ISL attenuates endothelial dysfunction in T2D mice, offering new insights into the pharmacological effects of ISL. Our findings demonstrate the potential of ISL as a promising therapeutic agent for the treatment of vascular diseases, paving the way for the further exploration of novel vascular therapies.


Subject(s)
Chalcones , Diabetes Mellitus, Type 2 , Endothelial Cells , Endothelium, Vascular , Glycyrrhiza , Oxidative Stress , Plant Extracts , Animals , Chalcones/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glycyrrhiza/chemistry , Male , Mice , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Plant Extracts/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Aorta/drug effects , Diabetes Mellitus, Experimental/drug therapy , Mice, Inbred C57BL , Interleukin-1beta/metabolism
15.
Int Heart J ; 65(5): 929-938, 2024.
Article in English | MEDLINE | ID: mdl-39343596

ABSTRACT

Pulmonary arterial hypertension is a progressive heart and lung disease that is caused by irreversible pulmonary vascular remodeling. Sinomenine hydrochloride is an alkaloid that is extracted from sinomenium acutum, which has strong anti-inflammatory effects. In this study, male rats were injected with monocrotaline, and endothelial cells were exposed to hypoxia for 24 hours to induce pulmonary arterial hypertension. Apoptosis, inflammation, and oxidative stress pathways were observed the in lungs and cells. Sinomenine hydrochloride repressed the increased right ventricular systolic pressure and attenuated the right ventricular hypertrophy and pulmonary artery remodeling in model rats. It reversed the expression of BCL2 and BAX and prevented the apoptosis of endothelial cells. Additionally, it increased the contents of IKBα and NRF2. P65, P-P65, TNFα, IL1ß, and IL6 levels in the lungs decreased by it. Malondialdehyde contents decreased, and the superoxide dismutase and glutathione peroxidase activity and HO-1 level increased in the treatment group. In vivo, it promoted apoptosis of pulmonary artery endothelial cells. Moreover, by activating PPAR-γ, sinomenine hydrochloride attains the above effects. These data suggested that sinomenine hydrochloride could protect endothelial cells, restrain inflammation and oxidative stress, and enhance pulmonary vascular remodeling.


Subject(s)
Apoptosis , Endothelial Cells , Hypertension, Pulmonary , Morphinans , Oxidative Stress , PPAR gamma , Morphinans/pharmacology , Morphinans/therapeutic use , Animals , Apoptosis/drug effects , Male , Rats , PPAR gamma/metabolism , Oxidative Stress/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Vascular Remodeling/drug effects , Cells, Cultured
16.
BMC Pulm Med ; 24(1): 477, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39334020

ABSTRACT

BACKGROUND: The mitochondria are essential organelles not only providing cellular energy in the form of ATP, but also regulating the inflammatory response and the cell death program. Mitochondrial dysfunction has been associated with various human diseases, including metabolic syndromes as well as inflammatory and neurodegenerative diseases. Acute respiratory distress syndrome (ARDS) is an acute pulmonary disorder characterized by uncontrolled alveolar inflammation, apoptotic lung epithelial/endothelial cells, and pulmonary edema. Despite the high mortality of ARDS, an effective pharmacotherapy to treat this disease has not been established yet. Therefore, identifying a novel targeted therapy for ARDS is important. Recently, exogenous mitochondrial transplantation was reported to be beneficial for treating mitochondrial dysfunction. The current study aimed to investigate the therapeutic effect of mitochondrial transplantation on ARDS in vitro and in vivo. METHODS: Mitochondria were isolated from human stem cells. For in vitro efficacy of mitochondrial transplantation on the inflammation and cell death, murine alveolar macrophages MH-S and human pulmonary microvascular endothelial cells HPMECs were exposed to LPS, respectively. The ARDS mice model established by a single intratracheal instillation of LPS was used for in vivo efficacy of intravenously treated mitochondria. RESULTS: Our results showed that the mitochondria isolated from human stem cells exhibited an anti-inflammatory effect against alveolar macrophages and an anti-apoptotic effect against the alveolar epithelial cells. Furthermore, intravenous mitochondrial treatment was associated with the attenuation of lung injury in the LPS-induced ARDS mice. CONCLUSION: Dual effects of mitochondria on anti-inflammation and anti-apoptosis support the potential of mitochondrial transplantation as a novel therapeutic strategy for ARDS.


Subject(s)
Apoptosis , Disease Models, Animal , Lipopolysaccharides , Mitochondria , Respiratory Distress Syndrome , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/chemically induced , Animals , Mitochondria/transplantation , Mitochondria/drug effects , Mice , Humans , Apoptosis/drug effects , Male , Macrophages, Alveolar/drug effects , Mice, Inbred C57BL , Endothelial Cells/drug effects
17.
Clin Sci (Lond) ; 138(18): 1131-1150, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39282930

ABSTRACT

Vascular endothelial growth factor receptor inhibitors (VEGFRis) improve cancer survival but are associated with treatment-limiting hypertension, often attributed to endothelial cell (EC) dysfunction. Using phosphoproteomic profiling of VEGFRi-treated ECs, drugs were screened for mitigators of VEGFRi-induced EC dysfunction and validated in primary aortic ECs, mice, and canine cancer patients. VEGFRi treatment significantly raised systolic blood pressure (SBP) and increased markers of endothelial and renal dysfunction in mice and canine cancer patients. α-Adrenergic-antagonists were identified as drugs that most oppose the VEGFRi proteomic signature. Doxazosin, one such α-antagonist, prevented EC dysfunction in murine, canine, and human aortic ECs. In mice with sorafenib-induced-hypertension, doxazosin mitigated EC dysfunction but not hypertension or glomerular endotheliosis, while lisinopril mitigated hypertension and glomerular endotheliosis without impacting EC function. Hence, reversing EC dysfunction was insufficient to mitigate VEGFRi-induced-hypertension in this mouse model. Canine cancer patients with VEGFRi-induced-hypertension were randomized to doxazosin or lisinopril and both agents significantly decreased SBP. The canine clinical trial supports safety and efficacy of doxazosin and lisinopril as antihypertensives for VEGFRi-induced-hypertension and the potential of trials in canines with spontaneous cancer to accelerate translation. The overall findings demonstrate the utility of phosphoproteomics to identify EC-protective agents to mitigate cardio-oncology side effects.


Subject(s)
Doxazosin , Endothelial Cells , Hypertension , Receptors, Vascular Endothelial Growth Factor , Animals , Dogs , Humans , Hypertension/drug therapy , Hypertension/metabolism , Hypertension/physiopathology , Doxazosin/pharmacology , Doxazosin/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Proteomics/methods , Blood Pressure/drug effects , Mice , Mice, Inbred C57BL , Lisinopril/pharmacology , Lisinopril/therapeutic use , Male , Neoplasms/drug therapy , Neoplasms/metabolism , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic alpha-1 Receptor Antagonists/therapeutic use , Sorafenib/pharmacology , Sorafenib/therapeutic use
18.
Biomolecules ; 14(9)2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39334925

ABSTRACT

Hyperlipidemia is a major risk factor for vascular lesions in diabetes mellitus and other metabolic disorders, although its basis remains poorly understood. One of the key pathogenetic events in this condition is mitochondrial dysfunction associated with the opening of the mitochondrial permeability transition (MPT) pore, a drop in the membrane potential, and ROS overproduction. Here, we investigated the effects of bongkrekic acid and carboxyatractyloside, a potent blocker and activator of the MPT pore opening, respectively, acting through direct interaction with the adenine nucleotide translocator, on the progression of mitochondrial dysfunction in mouse primary lung endothelial cells exposed to elevated levels of palmitic acid. Palmitate treatment (0.75 mM palmitate/BSA for 6 days) resulted in an 80% decrease in the viability index of endothelial cells, which was accompanied by mitochondrial depolarization, ROS hyperproduction, and increased colocalization of mitochondria with lysosomes. Bongkrekic acid (25 µM) attenuated palmitate-induced lipotoxicity and all the signs of mitochondrial damage, including increased spontaneous formation of the MPT pore. In contrast, carboxyatractyloside (10 µM) stimulated cell death and failed to prevent the progression of mitochondrial dysfunction under hyperlipidemic stress conditions. Silencing of gene expression of the predominate isoform ANT2, similar to the action of carboxyatractyloside, led to increased ROS generation and cell death under conditions of palmitate-induced lipotoxicity in a stably transfected HEK293T cell line. Altogether, these results suggest that targeted manipulation of the permeability transition pore through inhibition of ANT may represent an alternative approach to alleviate mitochondrial dysfunction and cell death in cell culture models of fatty acid overload.


Subject(s)
Bongkrekic Acid , Mitochondria , Mitochondrial Permeability Transition Pore , Palmitates , Reactive Oxygen Species , Animals , Mitochondrial Permeability Transition Pore/metabolism , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Bongkrekic Acid/pharmacology , Palmitates/pharmacology , Palmitic Acid/pharmacology , Atractyloside/pharmacology , Atractyloside/analogs & derivatives , Mitochondrial Membrane Transport Proteins/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondrial ADP, ATP Translocases/metabolism , Humans , Endothelial Cells/metabolism , Endothelial Cells/drug effects
19.
Redox Biol ; 76: 103342, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39265498

ABSTRACT

BACKGROUND: Disruption of the blood-brain barrier (BBB) is a major contributor to hemorrhagic transformation (HT) in patients with acute ischemic stroke (AIS) following intravenous thrombolysis (IVT). However, the clinical therapies aimed at BBB protection after IVT remain limited. METHODS: One hundred patients with AIS who underwent IVT were enrolled (42 with HT and 58 without HT 24 h after IVT). Based on the cytokine chip, the serum levels of several AIS-related proteins, including LCN2, ferritin, matrix metalloproteinase-3, vascular endothelial-derived growth factor, and X-linked inhibitor of apoptosis, were detected upon admission, and their associations with HT were analyzed. After finding that LCN2 was related to HT in patients with IVT, we clarified whether the modulation of LCN2 influenced BBB dysfunction and HT after thrombolysis and investigated the potential mechanism. RESULTS: In patients with AIS following IVT, logistic regression analysis showed that baseline serum LCN2 (p = 0.023) and ferritin (p = 0.046) levels were independently associated with HT. A positive correlation between serum LCN2 and ferritin levels was identified in patients with HT. In experimental studies, recombinant LCN2 (rLCN2) significantly aggravated BBB dysfunction and HT in the thromboembolic stroke rats after thrombolysis, whereas LCN2 inhibition by ZINC006440089 exerted opposite effects. Further mechanistic studies showed that, LCN2 promoted endothelial cell ferroptosis, accompanied by the induction of high mobility group box 1 (HMGB1) and the inhibition of nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) proteins. Ferroptosis inhibitor ferrostatin-1 (fer-1) significantly restricted the LCN2-mediated BBB disruption. Transfection of LCN2 and HMGB1 siRNA inhibited the endothelial cell ferroptosis, and this effects was reversed by Nrf2 siRNA. CONCLUSION: LCN2 aggravated BBB disruption after thrombolysis by promoting endothelial cell ferroptosis via regulating the HMGB1/Nrf2/HO-1 pathway, this may provide a promising therapeutic target for the prevention of HT after IVT.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Ferroptosis , HMGB1 Protein , Lipocalin-2 , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Humans , Animals , Male , Rats , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , HMGB1 Protein/metabolism , Ferroptosis/drug effects , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Female , Lipocalin-2/metabolism , Signal Transduction/drug effects , Aged , Middle Aged , Thrombolytic Therapy , Ischemic Stroke/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/pathology , Heme Oxygenase-1/metabolism , Heme Oxygenase-1/genetics
20.
Nat Commun ; 15(1): 8273, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333556

ABSTRACT

Disruption of ciliary homeostasis in vascular endothelial cells has been implicated in the development of atherosclerosis. However, the molecular basis for the regulation of endothelial cilia during atherosclerosis remains poorly understood. Herein, we provide evidence in male mice that the accumulation of lipid droplets in vascular endothelial cells induces ciliary loss and contributes to atherosclerosis. Triglyceride accumulation in vascular endothelial cells differentially affects the abundance of free fatty acid species in the cytosol, leading to stimulated lipid droplet formation and suppressed protein S-palmitoylation. Reduced S-palmitoylation of ciliary proteins, including ADP ribosylation factor like GTPase 13B, results in the loss of cilia. Restoring palmitic acid availability, either through pharmacological inhibition of stearoyl-CoA desaturase 1 or a palmitic acid-enriched diet, significantly restores endothelial cilia and mitigates the progression of atherosclerosis. These findings thus uncover a previously unrecognized role of lipid droplets in regulating ciliary homeostasis and provide a feasible intervention strategy for preventing and treating atherosclerosis.


Subject(s)
Atherosclerosis , Cilia , Endothelial Cells , Lipid Droplets , Palmitic Acid , Animals , Male , Palmitic Acid/pharmacology , Palmitic Acid/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Lipid Droplets/metabolism , Lipid Droplets/drug effects , Mice , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Cilia/metabolism , Cilia/drug effects , Mice, Inbred C57BL , Humans , Triglycerides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL