Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 894
Filter
1.
Adv Healthc Mater ; 13(12): e2303928, 2024 May.
Article in English | MEDLINE | ID: mdl-38291861

ABSTRACT

Chirality is an intrinsic cellular property that describes cell polarization biases along the left-right axis, apicobasal axis, or front-rear axes. Cell chirality plays a significant role in the arrangement of organs in the body as well as in the orientation of organelles, cytoskeletons, and cells. Vascular networks within the endometrium, the mucosal inner lining of the uterus, commonly display spiral architectures that rapidly form across the menstrual cycle. Herein, the role of endometrial-relevant extracellular matrix stiffness, composition, and soluble signals on endometrial endothelial cell chirality is systematically examined using a high-throughput microarray. Endometrial endothelial cells display marked patterns of chirality as individual cells and as cohorts in response to substrate stiffness and environmental cues. Vascular networks formed from endometrial endothelial cells also display shifts in chirality as a function of exogenous hormones. Changes in cellular-scale chirality correlate with changes in vascular network parameters, suggesting a critical role for cellular chirality in directing endometrial vessel network organization.


Subject(s)
Endometrium , Endothelial Cells , Endometrium/cytology , Endometrium/blood supply , Endometrium/metabolism , Humans , Female , Endothelial Cells/cytology , Endothelial Cells/metabolism , Cell Polarity/physiology , Microvessels/cytology , Microvessels/physiology , Extracellular Matrix/metabolism , Cells, Cultured
2.
Arch Biochem Biophys ; 720: 109173, 2022 05 15.
Article in English | MEDLINE | ID: mdl-35300940

ABSTRACT

The effect of Esculetin on pyroptosis and its possible mechanism in endothelium were explored. 10 µg/mL LPS and 0.5 mM ATP were used to stimulate the rat intestinal microvascular endothelial cells. Then add different concentrations of Esculetin (20µM, 40 µM) to the culture medium containing LPS and ATP culturing for 24 h. The expression of p-NF-κB p65, NF-κB p65, I-κB, p-I-κB, NLRP3, ASC, caspase-1, and gasdermin-D were detected by Western blot, and the release level of IL-18 and IL-1ß were measured by ELISA. The NLRP3 inhibitor MCC950 was used at the concentration of 10 µM for 4 h to disentangle the potential mechanism of the influence of Esculetin on pyroptosis. In our experiments, the expression of gasdermin-d and important proteins of NF-κB and NLRP3 signaling pathways were inhibited by Esculetin. Besides, Esculetin also attenuated the morphological changes like swelling rupture and pores on the membrane caused by pyroptosis thereby protecting cells from being damaged by pyroptosis. Combining with the effect of Esculetin on proteins above and its protective effect on cell morphology, we believe that Esculetin has an anti-pyroptosis effect. The inhibiting pyroptosis effects mentioned above are similar to MCC950, which means the anti-pyroptosis effects of Esculetin are associated with the NLRP3 signaling pathway. In conclusion, Esculetin inhibits the pyroptosis of microvascular endothelial cells through the NF-κB/NLFP3 signaling pathway and is expected to be conducive in treating pyroptosis-related diseases.


Subject(s)
Endothelial Cells , Microvessels , NF-kappa B , Pyroptosis , Umbelliferones , Adenosine Triphosphate , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipopolysaccharides/pharmacology , Microvessels/cytology , Microvessels/drug effects , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Rats , Signal Transduction , Umbelliferones/pharmacology
3.
Bioengineered ; 13(2): 4385-4396, 2022 02.
Article in English | MEDLINE | ID: mdl-35139776

ABSTRACT

As one of the most frequently prescribed antidiabetic drugs, metformin can lower glucose levels, improve insulin resistance manage body weight. However, the effect of metformin on islet microcirculation remains unclear. In the present study, to explore the effect of metformin on islet endothelial cells and investigated the underlying mechanism, we assessed the effects of metformin on islet endothelial cell survival, proliferation, oxidative stress and apoptosis. Our results suggest that metformin stimulates the proliferation of pancreatic islet endothelial cells and inhibits the apoptosis and oxidative stress caused by high glucose levels. By activating farnesoid X receptor (FXR), metformin increases the expression of vascular endothelial growth factor-A (VEGF-A) and endothelial nitric oxide synthase (eNOS), improves the production of nitric oxide (NO) and decreases the production of ROS. After the inhibition of FXR or VEGF-A, all of the effects disappeared. Thus, metformin appears to regulate islet microvascular endothelial cell (IMEC) proliferation, apoptosis and oxidative stress by activating the FXR/VEGF-A/eNOS pathway. These findings provide a new mechanism underlying the islet-protective effect of metformin.


Subject(s)
Glucose/adverse effects , Islets of Langerhans , Metformin/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelium, Vascular/cytology , Islets of Langerhans/blood supply , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Mice , Microvessels/cytology , Oxidative Stress/drug effects
4.
Biochem Pharmacol ; 195: 114745, 2022 01.
Article in English | MEDLINE | ID: mdl-34454930

ABSTRACT

The repair of vascular endothelial cell dysfunction is an encouraging approach for the treatment of vascular complications associated with diabetes. It has been demonstrated that members of C1q/tumor necrosis factor-related protein (CTRP) family may improve endothelial function. Nevertheless, the protective properties of CTRPs in diabetic microvascular complications continue to be mostly unknown. Here, we demonstrate that the C1q-like globular domain of CTRP3, CTRP5, and CTRP9 (gCTRP3, 5, 9) exerted a vasorelaxant effect on the microvasculature, of which gCTRP3 was the most powerful one. In a murine model of type 2 diabetes mellitus, serum gCTRP3 level and endothelial function decreased markedly compared with controls. Two weeks of gCTRP3 treatment (0.5 µg/g/d) enhanced endothelium-dependent relaxation in microvessels, increased nitric oxide (NO·) production, and reduced retinal vascular leakage. In addition, Western blotting in human retinal microvascular endothelial cells indicated that gCTRP3 triggered AMP-activated protein kinase-α (AMPKα), hence increasing the endothelial NO synthase (eNOS) level and NO· production. In addition, incubation with gCTRP3 in vitro ameliorated the endothelial dysfunction induced by high glucose in the branch of the mesenteric artery. Blockade of either eNOS or AMPKα completely abolished the effects of gCTRP3 described above. Taken together, we demonstrate for the first time that gCTRP3 improves impaired vasodilatation of microvasculature in diabetes by ameliorating endothelial cell function through the AMPK/eNOS/NO· signaling pathway. This finding may suggest an effective intervention against diabetes-associated microvascular complications.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipokines/pharmacology , Diabetes Mellitus, Type 2/physiopathology , Endothelial Cells/drug effects , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Signal Transduction/drug effects , Adipokines/blood , Adipokines/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Endothelial Cells/physiology , Humans , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiology , Mice, Inbred C57BL , Microvessels/cytology , Tumor Necrosis Factors/metabolism , Vasodilation/drug effects
5.
Am J Physiol Heart Circ Physiol ; 322(1): H71-H86, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34767485

ABSTRACT

Microvessels-on-a-chip have enabled in vitro studies to closely simulate in vivo microvessel environment. However, assessing microvessel permeability, a functional measure of microvascular exchange, has not been attainable in nonpermeable microfluidic platforms. This study developed a new approach that enables permeability coefficients (Ps) to be quantified in microvessels developed in nonpermeable chip platforms by integrating avidin-biotin technology. Microvessels were developed on biotinylated fibronectin-coated microfluidic channels. Solute transport was assessed by perfusing microvessels with fluorescence-labeled avidin. Avidin molecules that crossed endothelium were captured by substrate biotin and recorded with real-time confocal images. The Ps was derived from the rate of avidin-biotin accumulation at the substrate relative to solute concentration difference across microvessel wall. Avidin tracers with different physiochemical properties were used to characterize the barrier properties of the microvessel wall. The measured baseline Ps and inflammatory mediator-induced increases in Ps and endothelial cell (EC) [Ca2+]i resembled those observed in intact microvessels. Importantly, the spatial accumulation of avidin-biotin at substrate defines the transport pathways. Glycocalyx layer is well formed on endothelium and its degradation increased transcellular transport without affecting EC junctions. This study demonstrated that in vitro microvessels developed in this simply designed microfluidics structurally possess in vivo-like glycocalyx layer and EC junctions and functionally recapitulate basal barrier properties and stimuli-induced responses observed in intact microvessels. This new approach overcomes the limitations of nonpermeable microfluidics and provides an easily executed highly reproducible in vitro microvessel model with in vivo microvessel functionality, suitable for a wide range of applications in blood and vascular research and drug development.NEW & NOTEWORTHY Our study developed a novel method that allows permeability coefficient to be measured in microvessels developed in nonpermeable microfluidic platforms using avidin-biotin technology. It overcomes the major limitation of nonpermeable microfluidic system and provides a simply designed easily executed and highly reproducible in vitro microvessel model with permeability accessibility. This model with in vivo-like endothelial junctions, glycocalyx, and permeability properties advances microfluidics in microvascular research, suitable for a wide range of biomedical and clinical applications.


Subject(s)
Avidin , Biotin , Capillary Permeability , Lab-On-A-Chip Devices , Microfluidics/methods , Microvessels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Glycocalyx/metabolism , Microfluidics/instrumentation , Microvessels/cytology , Rats
6.
J Mater Chem B ; 10(2): 271-281, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34897348

ABSTRACT

Anti-seizure medicines constitute a common yet important modality to treat epilepsy. However, some of them are associated with serious side effects including hepatotoxicity and hypersensitivity. Furthermore, the blood-brain barrier (BBB) is an insurmountable obstacle for brain drug delivery. Fortunately, the introduction of the nanoparticles for drug delivery is a feasible approach to overcome these obstacles. Encapsulating drugs into nanoparticles and delivering them to specific sites shows great potential for improving the efficiency of drug delivery and reducing systemic toxicity. Several in vivo studies have investigated the effect of nanoparticle size on biodistribution in mice, but very few have investigated its effects on efficient drug delivery while crossing the BBB. Therefore, we designed a methoxy poly(lactide-co-glycolide)-b-poly(ethylene glycol) methyl ether (mPEG-PLGA) nanoparticle delivery system and explored the cell uptake efficiency of nanoparticles with different sizes and their ability to penetrate the BBB while carrying carbamazepine (CBZ). CBZ-loaded nanoparticles could significantly reduce the cytotoxicity of CBZ to L929 cells at high concentrations. Results from the endocytosis experiment involving human cerebral microvessel endothelial cell/D3 showed that the DiR-loaded mPEG5K-PLGA10K nanoparticles possessed the highest cell uptake efficiency. The endocytosis efficiency was 90% at 30 min, which far exceeded that of the other groups. Moreover, similar results were obtained from subsequent experiments where fluorescence images of the isolated organs of the mice were acquired. To summarize, our study demonstrated that drug delivery to the brain using nanocarriers is size dependent. Nanoparticles with the smallest particle size can be internalized more effectively, and easily penetrate the BBB, and accumulate in the brain.


Subject(s)
Anticonvulsants/pharmacokinetics , Blood-Brain Barrier/physiology , Carbamazepine/pharmacokinetics , Drug Carriers/chemistry , Nanoparticles/chemistry , Animals , Anticonvulsants/chemistry , Brain/cytology , Carbamazepine/chemistry , Cell Line , Drug Carriers/metabolism , Drug Carriers/toxicity , Endocytosis/physiology , Female , Humans , Mice , Microvessels/cytology , Nanoparticles/metabolism , Nanoparticles/toxicity , Particle Size , Polyesters/chemistry , Polyesters/metabolism , Polyesters/toxicity , Polyethylene Glycols/chemistry , Polyethylene Glycols/metabolism , Polyethylene Glycols/toxicity
7.
Am J Physiol Cell Physiol ; 322(2): C185-C196, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34878922

ABSTRACT

The Notch pathway regulates complex patterning events in many species and is critical for the proper formation and function of the vasculature. Despite this importance, how the various components of the Notch pathway work in concert is still not well understood. For example, NOTCH1 stabilizes homotypic endothelial junctions, but the role of NOTCH1 in heterotypic interactions is not entirely clear. NOTCH3, on the other hand, is essential for heterotypic interactions of pericytes with the endothelium, but how NOTCH3 signaling in pericytes impacts the endothelium remains elusive. Here, we use in vitro vascular models to investigate whether pericyte-induced stabilization of the vasculature requires the cooperation of NOTCH1 and NOTCH3. We observe that both pericyte NOTCH3 and endothelial NOTCH1 are required for the stabilization of the endothelium. Loss of either NOTCH3 or NOTCH1 decreases the accumulation of VE-cadherin at endothelial adherens junctions and increases the frequency of wider, more motile junctions. We found that DLL4 was the key ligand for simulating NOTCH1 activation in endothelial cells and observed that DLL4 expression in pericytes is dependent on NOTCH3. Altogether, these data suggest that an interplay between pericyte NOTCH3 and endothelial NOTCH1 is critical for pericyte-induced vascular stabilization.


Subject(s)
Endothelial Cells/metabolism , Microvessels/metabolism , Pericytes/metabolism , Receptor, Notch1/metabolism , Receptor, Notch3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , Cells, Cultured , Coculture Techniques , Endothelial Cells/drug effects , HEK293 Cells , Humans , Microvessels/cytology , Microvessels/drug effects , Pericytes/drug effects , Receptor, Notch1/agonists , Receptor, Notch3/agonists
8.
Neurosci Lett ; 770: 136381, 2022 01 23.
Article in English | MEDLINE | ID: mdl-34906568

ABSTRACT

BACKGROUND: The maintenance of human brain microvascular endothelial cell (HBMEC) function is crucial to improve the outcomes of ischemic stroke (IS). Emerging evidence shows that circular RNAs (circRNAs) are involved in IS progression. This study aimed to investigate the role of circRNA FUN14 domain containing 1 (circFUNDC1) in oxygen-glucose deprivation (OGD)-treated HBMECs. METHODS: The expression of circFUNDC1, miR-375 and phosphatase and tensin homolog (PTEN) mRNA was detected by quantitative real-time PCR (qPCR). Cell viability, apoptosis, migration and angiogenesis were determined by CCK-8 assay, flow cytometry assay, transwell assay and tube formation assay. The protein level of PTEN was detected by western blot. The relationship between miR-375 and circFUNDC1 or PTEN was confirmed by pull-down assay, dual-luciferase reporter assay and RIP assay. Exosomes were identified by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). RESULTS: CircFUNDC1 expression was increased in peripheral blood of IS patients and OGD-treated HBMECs. CircFUNDC1 knockdown alleviated OGD-induced cell apoptosis and promoted OGD-blocked cell viability, migration and angiogenesis of HBMECs. MiR-375 was a target of circFUNDC1, and miR-375 restoration played similar effects with circFUNDC1 knockdown. The inhibition of miR-375 reversed the effects of circFUNDC1 knockdown. In addition, PTEN was a downstream target of miR-375, and PTEN overexpression abolished the effects of miR-375 restoration. The expression of circFUNDC1 was elevated in serum-derived exosomes of IS patients, and circFUNDC1 harbored diagnostic values. CONCLUSION: CircFUNDC1 knockdown alleviates OGD-induced HBMECs injuries by inhibiting PTEN via enriching miR-375.


Subject(s)
Endothelial Cells/metabolism , Ischemic Stroke/metabolism , Membrane Proteins/genetics , MicroRNAs/metabolism , Mitochondrial Proteins/genetics , PTEN Phosphohydrolase/genetics , RNA, Circular/metabolism , Aged , Brain/blood supply , Cell Hypoxia , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Exosomes/metabolism , Female , Glucose/deficiency , Humans , Ischemic Stroke/genetics , Male , MicroRNAs/genetics , Microvessels/cytology , Microvessels/metabolism , Middle Aged , Oxygen/metabolism , PTEN Phosphohydrolase/metabolism , RNA, Circular/genetics
9.
Front Immunol ; 12: 759176, 2021.
Article in English | MEDLINE | ID: mdl-34956190

ABSTRACT

Pulmonary microvascular endothelial cells (PMECs) and the extracellular vesicles (EVs) derived from PMECs participate in maintaining pulmonary homeostasis and mediating the inflammatory response. However, obtaining a high-purity population of PMECs and their EVs from mouse is still notoriously difficult. Herein we provide a method to isolate primary mouse PMECs (pMPMECs) and to transduce SV40 lentivirus into pMPMECs to establish an immortalized cell line (iMPMECs), which provides sufficient quantities of EVs for further studies. pMPMECs and iMPMECs can be identified using morphologic criteria, a phenotypic expression profile (e.g., CD31, CD144, G. simplicifolia lectin binding), and functional properties (e.g., Dil-acetylated low-density protein uptake, Matrigel angiogenesis). Furthermore, pMPMEC-EVs and iMPMEC-EVs can be identified and compared. The characteristics of pMPMEC-EVs and iMPMEC-EVs are ascertained by transmission electron microscopy, nanoparticle tracking analysis, and specific protein markers. iMPMECs produce far more EVs than pMPMECs, while their particle size distribution is similar. Our detailed protocol to isolate and immortalize MPMECs will provide researchers with an in vitro model to investigate the specific roles of EVs in pulmonary physiology and diseases.


Subject(s)
Endothelial Cells/chemistry , Extracellular Vesicles/chemistry , Microvessels/chemistry , Animals , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/immunology , Extracellular Vesicles/immunology , Mice , Microvessels/cytology , Microvessels/immunology , Particle Size , Single-Cell Analysis
10.
Dev Cell ; 56(22): 3042-3051, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34813766

ABSTRACT

Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.


Subject(s)
Endothelial Cells/cytology , Kidney/blood supply , Microvessels/cytology , Neovascularization, Physiologic/physiology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Humans , Kidney/cytology
11.
Int J Mol Sci ; 22(20)2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34681708

ABSTRACT

Ischemia-reperfusion injury (IRI) is an inevitable consequence of organ transplant procedure and associated with acute and chronic organ rejection in transplantation. IRI leads to various forms of programmed cell death, which worsens tissue damage and accelerates transplant rejection. We recently demonstrated that necroptosis participates in murine cardiac microvascular endothelial cell (MVEC) death and murine cardiac transplant rejection. However, MVEC death under a more complex IRI model has not been studied. In this study, we found that simulating IRI conditions in vitro by hypoxia, reoxygenation and treatment with inflammatory cytokines induced necroptosis in MVECs. Interestingly, the apoptosis-inducing factor (AIF) translocated to the nucleus during MVEC necroptosis, which is regulated by the mitochondrial permeability molecule cyclophilin D (CypD). Furthermore, CypD deficiency in donor cardiac grafts inhibited AIF translocation and mitigated graft IRI and rejection (n = 7; p = 0.002). Our studies indicate that CypD and AIF play significant roles in MVEC necroptosis and cardiac transplant rejection following IRI. Targeting CypD and its downstream AIF may be a plausible approach to inhibit IRI-caused cardiac damage and improve transplant survival.


Subject(s)
Apoptosis Inducing Factor/metabolism , Necroptosis , Peptidyl-Prolyl Isomerase F/metabolism , Animals , Apoptosis Inducing Factor/antagonists & inhibitors , Apoptosis Inducing Factor/genetics , Cell Hypoxia , Cell Nucleus/metabolism , Peptidyl-Prolyl Isomerase F/deficiency , Peptidyl-Prolyl Isomerase F/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Interferon-gamma/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microvessels/cytology , Models, Biological , Necroptosis/drug effects , Oxygen/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/pharmacology
12.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638868

ABSTRACT

Mechanical unloading contributes to significant cardiovascular deconditioning. Endothelial dysfunction in the sites of microcirculation may be one of the causes of the cardiovascular degeneration induced by unloading, but the detailed mechanism is still unclear. Here, we first demonstrated that mechanical unloading inhibited brain microvascular endothelial cell proliferation and downregulated histone deacetylase 6 (HDAC6) expression. Furthermore, HDAC6 promoted microvascular endothelial cell proliferation and attenuated the inhibition of proliferation caused by clinorotation unloading. To comprehensively identify microRNAs (miRNAs) that are regulated by HDAC6, we analyzed differential miRNA expression in microvascular endothelial cells after transfection with HDAC6 siRNA and selected miR-155-5p, which was the miRNA with the most significantly increased expression. The ectopic expression of miR-155-5p inhibited microvascular endothelial cell proliferation and directly downregulated Ras homolog enriched in brain (RHEB) expression. Moreover, RHEB expression was downregulated under mechanical unloading and was essential for the miR-155-5p-mediated promotion of microvascular endothelial cell proliferation. Taken together, these results are the first to elucidate the role of HDAC6 in unloading-induced cell growth inhibition through the miR-155-5p/RHEB axis, suggesting that the HDAC6/miR-155-5p/RHEB pathway is a specific target for the preventative treatment of cardiovascular deconditioning.


Subject(s)
Cell Proliferation , Endothelial Cells/metabolism , Gene Expression Regulation , Histone Deacetylase 6/metabolism , MicroRNAs/biosynthesis , Microvessels/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Animals , Cell Line , Endothelial Cells/cytology , Mice , Microvessels/cytology
13.
PLoS One ; 16(9): e0256734, 2021.
Article in English | MEDLINE | ID: mdl-34469488

ABSTRACT

OBJECTIVES: This study is to investigate whether the cardiac microvascular endothelial cells (CMECs) can regulate the autophagy of cardiomyocytes (CMs) by secreting lncRNA-ANRIL/miR-181b exosomes, thus participating in the occurrence of uremic cardiovascular disease (CVD). METHODS: A 5/6 nephrectomy uremia model was established, with the mice injected with ANRIL-shRNA lentivirus vector, miR-181b agomir, and related control reagents, containing the serum creatinine and urea nitrogen measured. The renal tissue sections of mice were stained with Periodic Acid-Schiff (PAS), TUNEL, and Hematoxylin-Eosin (HE) performed on myocardial tissue sections of mice. ANRIL-shRNA, miR-181b mimics, and related control reagents were transfected into CMECs, in which the exosomes were extracted and co-cultured with CMs. The expressions of ANRIL, miR-181b and ATG5 were detected by qRT-PCR, and the expressions of autophagy related proteins by Western blot, as well as the binding of ANRIL and miR-181b by the double luciferase reporter gene experiment. RESULTS: ANRIL down-regulation or miR-181b up-regulation can increase the weight of mice with uremia, as well as the expressions of p62 and miR-181b, and reduce the content of serum creatinine and urea nitrogen, the damage of kidney and myocardial tissues, the number of apoptotic cells in myocardial tissues, as well as the expressions of ANRIL, ATG5, Beclin1, and LC3. CMs can absorb the exosomes of CMECs. Compared with IS+ CMEC-Exo group, the expressions of ANRIL and ATG5 in CMs of IS+ CMEC-Exo + sh lncRNA ANRIL and IS+CMEC-Exo+miR-181b mimics groups was down-regulated, as well as the expressions of ATG5, Beclin1, and LC3, while miR-181b expression was up-regulated as well as P62 expression. CONCLUSIONS: CMECs can regulate autophagy of CMs by releasing exosomes containing ANRIL and miR-181b.


Subject(s)
Autophagy-Related Protein 5/genetics , Autophagy/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Uremia/immunology , Animals , Autophagy-Related Protein 5/metabolism , Coronary Vessels/cytology , Coronary Vessels/metabolism , Disease Models, Animal , Down-Regulation/immunology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Exosomes/metabolism , Humans , Male , Mice , MicroRNAs/genetics , Microvessels/cytology , Myocardium/cytology , Myocardium/immunology , Myocardium/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , RNA, Long Noncoding/genetics , Up-Regulation/immunology , Uremia/genetics , Uremia/pathology
14.
Nat Biomed Eng ; 5(8): 847-863, 2021 08.
Article in English | MEDLINE | ID: mdl-34385693

ABSTRACT

The therapeutic efficacy of stem cells transplanted into an ischaemic brain depends primarily on the responses of the neurovascular unit. Here, we report the development and applicability of a functional neurovascular unit on a microfluidic chip as a microphysiological model of ischaemic stroke that recapitulates the function of the blood-brain barrier as well as interactions between therapeutic stem cells and host cells (human brain microvascular endothelial cells, pericytes, astrocytes, microglia and neurons). We used the model to track the infiltration of a number of candidate stem cells and to characterize the expression levels of genes associated with post-stroke pathologies. We observed that each type of stem cell showed unique neurorestorative effects, primarily by supporting endogenous recovery rather than through direct cell replacement, and that the recovery of synaptic activities is correlated with the recovery of the structural and functional integrity of the neurovascular unit rather than with the regeneration of neurons.


Subject(s)
Ischemic Stroke/therapy , Lab-On-A-Chip Devices , Stem Cell Transplantation , Astrocytes/cytology , Astrocytes/metabolism , Blood-Brain Barrier/chemistry , Blood-Brain Barrier/metabolism , Coculture Techniques , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Microglia/cytology , Microglia/metabolism , Microvessels/cytology , Models, Biological , Neurons/cytology , Neurons/metabolism , Pericytes/cytology , Pericytes/metabolism , Stem Cells/cytology , Stem Cells/metabolism
15.
Molecules ; 26(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34443317

ABSTRACT

Endothelial cell injury is an early event in systemic sclerosis (SSc) pathogenesis and several studies indicate oxidative stress as the trigger of SSc-associated vasculopathy. Here, we show that circulating factors present in sera of SSc patients increased reactive oxygen species (ROS) production and collagen synthesis in human pulmonary microvascular endothelial cells (HPMECs). In addition, the possibility that iloprost, a drug commonly used in SSc therapy, might modulate the above-mentioned biological phenomena has been also investigated. In this regard, as compared to sera of SSc patients, sera of iloprost-treated SSc patients failed to increased ROS levels and collagen synthesis in HPMEC, suggesting a potential antioxidant mechanism of this drug.


Subject(s)
Collagen/biosynthesis , Endothelial Cells/drug effects , Iloprost/pharmacology , Microvessels/cytology , Oxidative Stress/drug effects , Scleroderma, Systemic/blood , Serum/metabolism , Adult , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Humans , Male , Reactive Oxygen Species/metabolism
16.
Int Rev Cell Mol Biol ; 362: 55-110, 2021.
Article in English | MEDLINE | ID: mdl-34253298

ABSTRACT

Blood brain barrier (BBB) is formed by the brain microvascular endothelial cells (BMVECs) lining the wall of brain capillaries. Its integrity is regulated by multiple mechanisms, including up/downregulation of tight junction proteins or adhesion molecules, altered Ca2+ homeostasis, remodeling of cytoskeleton, that are confined at the level of BMVECs. Beside the contribution of BMVECs to BBB permeability changes, other cells, such as pericytes, astrocytes, microglia, leukocytes or neurons, etc. are also exerting direct or indirect modulatory effects on BBB. Alterations in BBB integrity play a key role in multiple brain pathologies, including neurological (e.g. epilepsy) and neurodegenerative disorders (e.g. Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis etc.). In this review, the principal Ca2+ signaling pathways in brain microvascular endothelial cells are discussed and their contribution to BBB integrity is emphasized. Improving the knowledge of Ca2+ homeostasis alterations in BMVECa is fundamental to identify new possible drug targets that diminish/prevent BBB permeabilization in neurological and neurodegenerative disorders.


Subject(s)
Brain/blood supply , Calcium/metabolism , Endothelial Cells/metabolism , Homeostasis , Microvessels/cytology , Animals , Humans , Ion Channels/metabolism
17.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article in English | MEDLINE | ID: mdl-34260400

ABSTRACT

Understanding the potential of nanomaterials (NMs) to cross the blood-brain barrier (BBB), as a function of their physicochemical properties and subsequent behavior, fate, and adverse effect beyond that point, is vital for evaluating the neurological effects arising from their unintentional entry into the brain, which is yet to be fully explored. This is not only due to the complex nature of the brain but also the existing analytical limitations for characterization and quantification of NMs in the complex brain environment. By using a fit-for-purpose analytical workflow and an in vitro BBB model, we show that the physiochemical properties of metallic NMs influence their biotransformation in biological matrices, which in turn modulates the transport form, efficiency, amounts, and pathways of NMs through the BBB and, consequently, their neurotoxicity. The data presented here will support in silico modeling and prediction of the neurotoxicity of NMs and facilitate the tailored design of safe NMs.


Subject(s)
Blood-Brain Barrier/metabolism , Metals/chemistry , Nanostructures/chemistry , Astrocytes/metabolism , Biotransformation , Brain/blood supply , Endothelial Cells/metabolism , Exocytosis , Humans , Microvessels/cytology , Models, Biological , Permeability , Transcytosis
18.
Neurotox Res ; 39(5): 1449-1458, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34216363

ABSTRACT

It has been widely reported that severe neurotoxicity can be induced by the application of propofol, which is closely related to the disruption of the blood-brain barrier (BBB) induced by inflammation and injury in the human brain microvascular endothelial cells (HBMVECs). Benzbromarone is a classic anti-gout agent that has been recently reported to exert anti-inflammatory and anti-oxidative stress effects. In the present study, we aim to investigate the protective property of Benzbromarone against propofol-induced injury on HBMVECs and the underlying mechanism. CCK8 assay was used to detect the cell viability of treated HBMVECs. Oxidative stress in HBMVECs was evaluated by measuring the levels of MDA and mitochondrial ROS. ELISA and qRT-PCR assay were used to determine the production of IL-1ß, IL-8, MCP-1, ICAM-1, and VCAM-1 by treated HBMVECs. Calcein-AM staining was utilized to evaluate the attachment of U937 monocytes to HBMVECs. The expression level of Egr-1 was determined by qRT-PCR and Western blot assay. Firstly, the decreased cell viability of HBMVECs induced by propofol was significantly elevated by treatment with Benzbromarone. The increased levels of MDA and mitochondrial ROS induced by propofol were dramatically suppressed by Benzbromarone. Secondly, the excessive production of inflammatory factors (IL-1ß, IL-8, and MCP-1) and adhesion molecules (ICAM-1 and VCAM-1) triggered by propofol was pronouncedly inhibited by Benzbromarone. Benzbromarone ameliorated propofol-induced attachment of U937 monocytes to HBMVECs. Lastly, Benzbromarone downregulated propofol-induced expression of the transcriptional factor Egr-1 in HBMVECs. Benzbromarone protected against propofol-induced inflammation and injury through suppressing Egr-1 in human brain vascular endothelial cells.


Subject(s)
Benzbromarone/pharmacology , Brain/drug effects , Endothelial Cells/drug effects , Microvessels/drug effects , Neuroprotective Agents/pharmacology , Propofol/toxicity , Anesthetics, Intravenous/toxicity , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/cytology , Brain/pathology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Endothelial Cells/pathology , Humans , Microvessels/cytology , Microvessels/pathology
19.
Mol Med Rep ; 24(4)2021 10.
Article in English | MEDLINE | ID: mdl-34328190

ABSTRACT

Pulmonary microvascular endothelial cell (PMVEC) apoptosis is the initial stage of adult pulmonary hypertension (PH), which involves high pulmonary arterial pressure and pulmonary vascular remodeling. However, the mechanism regulating PMVEC apoptosis and its involvement in the early stages of neonatal hypoxic PH (HPH) pathogenesis are currently unclear. The present study aimed to investigate the effects of heat shock protein 70 (HSP70) on hypoxia­induced apoptosis in PMVECs. PMVECs isolated from neonatal Sprague­Dawley rats were transfected with lentivirus with or without HSP70, or treated with the synthetic HSP70 inhibitor N­formyl­3,4­methylenedioxy­benzylidene-g-butyrolactam under hypoxic conditions (5% O2) for 24, 48 or 72 h. PMVEC apoptosis was evaluated by performing flow cytometry and mitochondrial membrane potential (MMP) assays. The expression levels of HSP70, hypoxia­inducible factor­1α (HIF­1α) and apoptosis­associated proteins were determined by conducting reverse transcription­quantitative PCR and western blotting. Following 24, 48 or 72 h of hypoxia, the apoptotic rates of PMVECs were significantly elevated compared with cells under normoxic conditions. The MMP was significantly reduced, whereas the mRNA and protein expression levels of HIF­1α, cytochrome c (cyt C), caspase­3 and HSP70 were enhanced by hypoxia compared with those under normoxic conditions. Additionally, the mRNA and protein expression levels of B­cell lymphoma 2 (Bcl­2) were significantly downregulated in the hypoxia group compared with those in the normoxia group. In hypoxic PMVECs, HSP70 overexpression decreased the apoptotic rate and the expression levels of cyt C, downregulated the expression levels of caspase­3 and HIF­1α, and increased the MMP and the expression levels of Bcl­2. HSP70 inhibition resulted in the opposite outcomes compared with those of HSP70 overexpression. Therefore, the results of the present study suggested that HSP70 may inhibit mitochondrial pathway­mediated apoptosis in isolated neonatal rat PMVECs in early­stage hypoxia, which may be associated with HSP70­mediated HIF­1α downregulation. Overall, HSP70 may be protective against neonatal HPH through the HSP70/HIF­1α pathway.


Subject(s)
Apoptosis , Endothelial Cells , HSP70 Heat-Shock Proteins , Hypertension, Pulmonary , Microvessels , Animals , Animals, Newborn , Apoptosis/genetics , Caspase 3/genetics , Caspase 3/metabolism , Cell Hypoxia , Down-Regulation , Endothelial Cells/cytology , Endothelial Cells/metabolism , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Hypertension, Pulmonary/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Microvessels/cytology , Microvessels/metabolism , Mitochondria/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats, Sprague-Dawley , Signal Transduction/genetics , Up-Regulation
20.
STAR Protoc ; 2(2): 100563, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34151293

ABSTRACT

We describe the extended endothelial cell culture method (EECM) for the differentiation of human pluripotent stem cells (hPSCs) into brain microvascular endothelial cell (BMEC)-like cells. EECM-BMEC-like cells resemble primary human BMECs in morphology, molecular junctional architecture, and diffusion barrier characteristics. A mature immune phenotype with proper endothelial adhesion molecule expression makes this model distinct from any other hPSC-derived in vitro blood-brain barrier (BBB) model and suitable to study immune cell migration across the BBB in a disease relevant and personalized fashion. For complete details on the use and execution of this protocol, please refer to Lian et al. (2014), Nishihara et al. (2020a).


Subject(s)
Brain/blood supply , Cell Differentiation , Endothelium, Vascular/cytology , Microvessels/cytology , Pluripotent Stem Cells/cytology , Cells, Cultured , Endothelium, Vascular/immunology , Humans , Microvessels/immunology , Pluripotent Stem Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...