Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 319
Filtrar
1.
Mediators Inflamm ; 2022: 5171525, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091666

RESUMEN

Inflammation is the body's biological reaction to endogenous and exogenous stimuli. Recent studies have demonstrated several anti-inflammatory properties of Ferula species. In this paper, we decided to study the anti-inflammatory effect of ethanolic extract of Ferula assafoetida oleo-gum-resin (asafoetida) against TNF-α-stimulated human umbilical vein endothelial cells (HUVECs). HUVECs were cultured in a flat-bottom plate and then treated with ethanolic extract of asafoetida (EEA, 0-500 µg/ml) and TNF-α (0-100 ng/ml) for 24 h. We used the MTT test to assess cell survival. In addition, the LC-MS analysis was performed to determine the active substances. HUVECs were pretreated with EEA and then induced by TNF-α. Intracellular reactive oxygen species (ROS) and adhesion of peripheral blood mononuclear cells (PBMCs) to HUVECs were evaluated with DCFH-DA and CFSE fluorescent probes, respectively. Gene expression of intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin and surface expression of ICAM-1 protein were measured using real-time PCR and flow cytometry methods, respectively. While TNF-α significantly increased intracellular ROS formation and PBMC adhesion to TNF-α-induced HUVECs, the pretreatment of HUVECs with EEA (125 and 250 µg/ml) significantly reduced the parameters. In addition, EEA pretreatment decreased TNF-α-induced mRNA expression of VCAM-1 and surface protein expression of ICAM-1 in the target cells. Taken together, the results indicated that EEA prevented ROS generation, triggered by TNF-α, and inhibited the expression of VCAM-1 and ICAM-1, leading to reduced PBMC adhesion. These findings suggest that EEA can probably have anti-inflammatory properties.


Asunto(s)
Antiinflamatorios , Moléculas de Adhesión Celular , Ferula , Células Endoteliales de la Vena Umbilical Humana , Extractos Vegetales , Antiinflamatorios/farmacología , Adhesión Celular , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Selectina E/biosíntesis , Selectina E/genética , Selectina E/inmunología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Leucocitos Mononucleares/inmunología , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
2.
Clin Exp Pharmacol Physiol ; 49(8): 805-812, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35577580

RESUMEN

Atherosclerosis is associated with a haemostatic imbalance characterized by excessive activation of pro-inflammatory and pro-coagulant pathways. Non-vitamin K antagonists oral anticoagulant (NOACs) may reduce the incidence of cardiovascular events, cerebral ischemia, thromboembolic events and atherosclerosis. Chronic inflammation, vascular proliferation and the development of atherosclerosis is also influenced by 25-hydroxycholesterol (25-OHC). The aim of the study was to assess the effect of rivaroxaban and dabigatran on the messenger RNA (mRNA) expression of anti-inflammatory cytokines transforming growth factor ß (TGF-ß), interleukin (IL)-37, IL-35 as well as of pro-inflammatory cytokines IL-18 and IL-23, in endothelial cells damaged by 25-OHC. Human umbilical vascular endothelial cells (HUVECs) were treated with 25-OHC (10 µg/mL), rivaroxaban (100, 500 ng/mL), dabigatran (100, 500 ng/mL), 25-OHC + rivaroxaban, and 25-OHC + dabigatran. The mRNA expression of TGF-ß, IL-37, IL-35 subunits EBI3 and p35, IL-18, and IL-23 was analysed using real-time polymerase chain reaction (PCR). The results showed that 25-OHC decreased TGF-ß and IL-37 mRNA expression and increased EBI3, p35, IL-18, IL-23 mRNA expression in endothelial cell as compared to an untreated control (P < .05). Messenger RNA expression of TGF-ß and IL-37 significantly increased following stimulation with rivaroxaban and dabigatran as compared to an untreated control (P < .01). In HUVECs pre-treated with oxysterol, rivaroxaban and dabigatran increased mRNA expression of TGF-ß, IL-37 and decreased mRNA expression of EBI3, p35, IL-23 and IL-18 as compared to 25-OHC (P < .01). Our finding suggests that both rivaroxaban and dabigatran inhibit the inflammatory activation caused by oxysterol in vitro.


Asunto(s)
Aterosclerosis , Citocinas , Dabigatrán , Células Endoteliales de la Vena Umbilical Humana , Hidroxicolesteroles , Rivaroxabán , Administración Oral , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/inmunología , Fibrilación Atrial/tratamiento farmacológico , Citocinas/genética , Citocinas/inmunología , Dabigatrán/farmacología , Dabigatrán/uso terapéutico , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Hidroxicolesteroles/administración & dosificación , Hidroxicolesteroles/efectos adversos , Hidroxicolesteroles/farmacología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Oxiesteroles/administración & dosificación , Oxiesteroles/efectos adversos , Oxiesteroles/farmacología , ARN Mensajero/genética , ARN Mensajero/inmunología , Rivaroxabán/farmacología , Rivaroxabán/uso terapéutico , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología
3.
Biochem Pharmacol ; 201: 115078, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35551917

RESUMEN

Endothelial cell senescence contributes to chronic inflammation and endothelial dysfunction, while favoring cardiovascular disorders and frailty. Senescent cells acquire a pro-inflammatory secretory phenotype that further propagates inflammation and senescence to neighboring cells. Cell senescence can be provoked by plethora of stressors, including inflammatory molecules and chemotherapeutic drugs. Doxorubicin (Doxo) is a powerful anthracycline anticancer drug whose clinical application is constrained by a dose-limiting cardiovascular toxicity. We here investigated whether cell senescence can contribute to the vascular damage elicited by Doxo. In human umbilical vein endothelial cells (HUVEC) cultures, Doxo (10-100 nM) increased the number of SA-ß-gal positive cells and the levels of γH2AX, p21 and p53, used as markers of senescence. Moreover, we identified Doxo-induced senescence to be mediated by the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome, a key player of the immune innate system capable of releasing interleukin (IL)-1ß. In fact, IL-1ß itself mimicked the stimulatory action of Doxo on both NLRP3 activation and cellular senescence, while the pharmacological blockade of IL-1 receptors markedly attenuated the pro-senescence effects of Doxo. In search of additional pharmacological strategies to attenuate Doxo-induced endothelial senescence, we identified resolvin E1 (RvE1), an endogenous pro-resolving mediator, as capable of reducing cell senescence induced by both Doxo and IL-1ß by interfering with the increased expression of pP65, NLRP3, and pro-IL-1ß proteins and with the formation of active NLRP3 inflammasome complexes. Overall, RvE1 and the blockade of the NLRP3 inflammasome-IL-1ß axis may offer a novel therapeutic approach against Doxo-induced cardiovascular toxicity and subsequent sequelae.


Asunto(s)
Doxorrubicina , Ácido Eicosapentaenoico , Células Endoteliales de la Vena Umbilical Humana , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Senescencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Interacciones Farmacológicas , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo
4.
Int J Mol Sci ; 23(4)2022 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-35216428

RESUMEN

As a cytokine, gamma-interferon (IFN-γ) is considered a key player in the fine-tuned orchestration of immune responses. The extreme cellular sensitivity to cytokines is attested by the fact that very few of these bioactive molecules per cell are enough to trigger cellular functions. These findings can, at least partially, explain how/why homeopathically-prepared cytokines, and especially micro-immunotherapy (MI) medicines, are able to drive cellular responses. We focused our fundamental research on a unitary MI preparation of IFN-γ, specifically employed at 4 CH, manufactured and impregnated onto sucrose-lactose pillules as all other MI medicines. We assessed the IFN-γ concentration in the medium after dilution of the IFN-γ (4 CH)-bearing pillules and we evaluated in vitro drug responses in a wide range of immune cells, and in endothelial cells. Our results showed that IFN-γ (4 CH) stimulated the proliferation, the activation and the phagocytic capabilities of primary immune cells, as well as modulated their cytokine-secretion and immunity-related markers' expression in a trend that is quite comparable with the well-recognized biological effects induced by IFN-γ. Altogether, these data provide novel and additional evidences on MI medicines, and specifically when active substances are prepared at 4 CH, thus suggesting the need for more investigations.


Asunto(s)
Inmunomodulación/inmunología , Interferón gamma/inmunología , Línea Celular Tumoral , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Inmunidad/inmunología , Factores Inmunológicos/inmunología , Inmunoterapia/métodos , Leucocitos Mononucleares/inmunología , Células THP-1
5.
Eur J Pharmacol ; 918: 174715, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35026193

RESUMEN

Evidence has demonstrated that a new class of anti-diabetic drugs, sodium-glucose co-transporter 2 (SGLT2) inhibitors, could exert beneficial effects on atherosclerotic complications of diabetes. Atherosclerosis is widely accepted as an inflammatory disease. Therefore, we aimed to assess the direct anti-inflammatory effects of SGLT2 inhibitors dapagliflozin (DAPA) on two cell types involved in the process of atherogenesis. Human umbilical vein endothelial cells (HUVECs) and macrophages were exposed to DAPA and lipopolysaccharide (LPS 20 ng/mL) for 24 h under normal (5.5 mmol/L, NG) or high glucose (25 mmol/L, HG) conditions. Then, levels of TLR-4/p-NF-κB, inflammatory cytokines, inflammation-related miR-146a and miR-155 as well as alteration in the ratio of M1/M2 macrophage polarization was assessed. DAPA (0.5 µM) could significantly attenuate LPS-induced TLR-4 overexpression (23.9% and 33.1% under NG and HG conditions in HUVECs and 53.3% and 52.4% under NG and HG states in macrophages, respectively). NF-κB p65 phosphorylation was also significantly decreased to 30.1% under NG condition in HUVECs and 51.9% and 34.5% under NG and HG states in macrophages by 0.5 µM DAPA. Moreover, DAPA elevated expression levels of anti-inflammatory miR-146a, while values of miR-155 decreased in those cells. DAPA also caused a shift from inflammatory M1 macrophages toward M2-dominant macrophages. These data suggest that regardless of glucose concentrations, DAPA could exert direct anti-inflammatory effects, at least partly, by inhibiting the expression of TLR-4 and activation of NF-κB along with the secretion of pro-inflammatory mediators.


Asunto(s)
Aterosclerosis , Compuestos de Bencidrilo/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucósidos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Macrófagos , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Antiinflamatorios/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/inmunología , Aterosclerosis/patología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Hipoglucemiantes/farmacología , Mediadores de Inflamación/antagonistas & inhibidores , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Transducción de Señal , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
6.
FASEB J ; 36(1): e22085, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34888952

RESUMEN

Allergic rhinitis (AR) is one of the most common allergic inflammatory diseases worldwide. In AR, increased blood flow and vascular permeability in nasal mucosa cause rhinorrhea and nasal congestion. We investigated the role of an 11Z,14Z-eicosadienoic acid-derived metabolite, 15-hydroxy-11Z,13Z-eicosadienoic acid (15-HEDE), in functional changes in vasculature and nasal congestion in AR. Repeated intranasal administration of Ovalbumin (OVA) caused AR symptoms, such as sneezing and nasal congestion, in mice. OVA administration increased the level of 15-HEDE in nasal lavage fluid, which reached approximately 0.6 ng/ml after ten OVA treatments. Upon measuring vascular contraction, treatment with 0.1-3 µM 15-HEDE did not cause contraction in mouse aortae, while it dilated aortae that were pre-contracted by thromboxane receptor stimulation. Pretreatment with the voltage-gated K+ (KV ) channel inhibitor 4-aminopyridine significantly inhibited the 15-HEDE-induced vascular relaxation. Intravital imaging showed that administration of 1 µg 15-HEDE dilated blood vessels, and Mile's assay demonstrated that this administration also caused dye leakage, indicating vascular hyperpermeability in mouse ears. Computed tomography scanning and morphological study revealed that administration of 3 µg 15-HEDE narrowed nasal passages and thickened nasal mucosa in mice. Finally, we confirmed that treating mice with 3 µg 15-HEDE caused rhinitis symptoms, such as abdominal breathing, and reduced respiratory frequency, suggesting nasal congestion. 15-HEDE caused vasodilation by activating KV channels and increased vascular permeability, which may lead to nasal congestion. Furthermore, 15-HEDE might be a new lipid mediator that exacerbates nasal congestion in AR.


Asunto(s)
Ácidos Eicosanoicos/toxicidad , Mucosa Nasal/inmunología , Ovalbúmina/toxicidad , Rinitis Alérgica , Administración Intranasal , Animales , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Rinitis Alérgica/inducido químicamente , Rinitis Alérgica/inmunología
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(12): 1057-1065, 2021 Dec.
Artículo en Chino | MEDLINE | ID: mdl-34906292

RESUMEN

Objective To establish the immortalized human umbilical vein vascular endothelial cells (HUVECs-hTERT) by introducing hTERT gene into primary HUVECs. In order to evaluate the potential of HUVECs-hTERT as a research model of HTNV infection, we explored the infection efficiency of Hantaan virus (HTNV) in HUVECs-hTERT and the influence of celluar innate immune regulation. Methods hTERT gene was cloned into lentivirus vector pCDH-CMV-MCS-EF1-puro, resulting in pCDH-CMV-hTERT-EF1-puro plasmid which was packaged into lentivirus. Then it was infected with HUVECs, and the HUVECs which stably express hTERT gene was selected by using puromycin and named HUVECs-hTERT. The morphology of HUVECs-hTERT and endothelial cell marker molecules, such as human von Willebrand factor (vWF), CD31 and vascular endothelial cell cadherin (VE-cadherin) were identified by microscopic observation and immunofluorescence assay. The percentage of nucleocapsid protein (NP)-positive cells after HTNV infection was detected by immunofluorescence assay to identify the difference of infection efficiency in HTNV between HUVECs and HUVECs-hTERT. Subsequently, real-time quantitative PCR (RT-qPCR) and Western blot analysis were used to detect the expression of HTNV S mRNA and NP after HTNV infection to verify amplification efficiency of HTNV in HUVECs and HUVECs-hTERT. RT-qPCR were used to detect the mRNA expression level of interferon ß (IFN-ß), interferon stimulating gene (ISG), including myxovirus resistance protein A (MxA), myxovirus resistance protein B (MxB), interferon inducing protein 2 (IFIT2), interferon-induced transmembrane protein 3 (IFITM3) and inflammatory factors, such as cyclooxygenase -2 (COX2), intercellular adhesion molecule (ICAM), C-C motif chemokine ligand 5 (CCL5) and the protein expression level of IFIT2, IFITM3 and MxA in the two types of cells after HTNV infection to determine whether the cellular innate immune response between HUVECs and HUVECs-hTERT are consistent. Results The immortalized cell line HUVECs-hTERT was screened successfully and the identification results showed that HUVECs-hTERT and HUVECs are with the same phenotype and express endothelial cell marker molecules, such as vWF, CD31 and VE-cadherin. HTNV can infect HUVECs-hTERT and HUVECs with approximately the same efficiency. In HTNV infection, the expression of innate immune molecules, such as IFN-ß, MxA, MxB, IFIT2, IFITM3, COX2, ICAM, CCL5 are similar between HUVECs and HUVECs-hTERT, indicating that the innate immune regulation of HUVECs-hTERT has not changed. Conclusion HUVECs-hTERT can replace primary HUVECs for the study of innate immune response regulation during HTNV infection under certain conditions.


Asunto(s)
Virus Hantaan , Células Endoteliales de la Vena Umbilical Humana , Inmunidad Innata , Línea Celular , Virus Hantaan/patogenicidad , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos
8.
mBio ; 12(6): e0211321, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34933450

RESUMEN

The interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral proteins that inhibit the entry of enveloped viruses. We analyzed the effect of IFITMs on the gamma-2 herpesviruses Kaposi's sarcoma-associated herpesvirus (KSHV) and the closely related rhesus monkey rhadinovirus (RRV). We used CRISPR/Cas9-mediated gene knockout to generate A549 cells, human foreskin fibroblasts (HFF), and human umbilical vein endothelial cells (HUVEC) with combined IFITM1/2/3 knockout and identified IFITMs as cell-dependent inhibitors of KSHV and RRV infection in A549 cells and HFF but not HUVEC. IFITM overexpression revealed IFITM1 as the relevant IFITM that inhibits KSHV and RRV infection. Fluorescent KSHV particles did not pronouncedly colocalize with IFITM-positive compartments. However, we found that KSHV and RRV glycoprotein-mediated cell-cell fusion is enhanced upon IFITM1/2/3 knockout. Taken together, we identified IFITM1 as a cell-dependent restriction factor of KSHV and RRV that acts at the level of membrane fusion. Of note, our results indicate that recombinant IFITM overexpression may lead to results that are not representative for the situation at endogenous levels. Strikingly, we observed that the endotheliotropic KSHV circumvents IFITM-mediated restriction in HUVEC despite high IFITM expression, while influenza A virus (IAV) glycoprotein-driven entry into HUVEC is potently restricted by IFITMs even in the absence of interferon. Mechanistically, we found that KSHV colocalizes less with IFITM1 and IFITM2 in HUVEC than in A549 cells immediately after attachment, potentially contributing to the observed difference in restriction. IMPORTANCE IFITM proteins are the first line of defense against infection by many pathogens and may also have therapeutic importance, as they, among other effectors, mediate the antiviral effect of interferons. Neither their function against herpesviruses nor their mechanism of action is well understood. We report here that in some cells but not in, for example, primary umbilical vein endothelial cells, IFITM1 restricts KSHV and RRV and that, mechanistically, this is likely effected by reducing the fusogenicity of the cell membrane. Further, we demonstrate potent inhibition of IAV glycoprotein-driven infection of cells of extrapulmonary origin by high constitutive IFITM expression.


Asunto(s)
Antígenos de Diferenciación/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Humano 8/fisiología , Proteínas de la Membrana/inmunología , Proteínas de Unión al ARN/inmunología , Rhadinovirus/fisiología , Animales , Antígenos de Diferenciación/genética , Coinfección/genética , Coinfección/inmunología , Coinfección/virología , Fibroblastos/inmunología , Fibroblastos/virología , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/virología , Humanos , Proteínas de la Membrana/genética , Proteínas de Unión al ARN/genética , Rhadinovirus/genética , Especificidad de la Especie , Internalización del Virus , Replicación Viral
9.
Int Immunopharmacol ; 101(Pt A): 108295, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34735917

RESUMEN

Toll-like receptors (TLRs) are among the players of inflammation during atherosclerosis. We assessed the effects of Eritoran, a TLR-4 antagonist, on lipopolysaccharide (LPS)-induced cytokines production by Peripheral Blood Mononuclear Cells (PBMCs) of patients with high-stenosis (HS) (n = 6) and healthy controls (HCs) (n = 6) co-cultured with Human Umbilical Vein Endothelial Cells (HUVECs). LPS stimulation significantly increased the levels of IL-6 (P = 0.007 and P = 0.005), TNF-α (P = 0.006 and P = 0.005), IL-2 (P = 0.007 and P = 0.002), IFN-γ (P = 0.006 and P = 0.003), IL-17A (P = 0.004 and P = 0.003), IL-17F (P = 0.005 and P = 0.003), IL-5 (P = 0.007 and P = 0.005), IL-13 (P = 0.006 and P = 0.005), IL-9 (P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.005) in HUVECs co-cultured with HC and HS PBMCs as compared with un-stimulated co-culture condition, respectively. Eritoran treatment (50 µg/mL and 100 µg/mL) significantly reduced the levels of LPS-induced IL-6 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), TNF-α (P = 0.005 and P = 0.003; P = 0.007 and P = 0.005), IL-2 (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IFN-γ (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IL-17A (P = 0.005 and P = 0.002; P = 0.005 and P = 0.002), IL-17F (P = 0.006 and P = 0.006; P = 0.005 and P = 0.005), IL-5 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), IL-9 (P = 0.005 and P = 0.005; P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.007; P = 0.005 and P = 0.005) in stimulated HUVECs co-cultured with HC and HS PBMCs, compared to un-treated condition, respectively. Our results demonstrate that attenuating effect of Eritoran on the inflammatory responses to LPS is higher in PBMCs of patients with high stenosis, suggesting its potential role in ameliorating inflammatory conditions in atherosclerosis.


Asunto(s)
Aterosclerosis/inmunología , Citocinas/metabolismo , Disacáridos/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Fosfatos de Azúcar/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Adulto , Aterosclerosis/tratamiento farmacológico , Estudios de Casos y Controles , Técnicas de Cocultivo , Disacáridos/uso terapéutico , Relación Dosis-Respuesta a Droga , Femenino , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-6/metabolismo , Interleucina-9/metabolismo , Interleucinas/metabolismo , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Masculino , Persona de Mediana Edad , Fosfatos de Azúcar/uso terapéutico , Factor de Necrosis Tumoral alfa/metabolismo
10.
Exp Cell Res ; 409(2): 112941, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34822812

RESUMEN

OBJECTIVE: The objective was to evaluate the expression levels of CD31+CD54+ and CD31+CD105+ endothelial microparticles (EMPs) before and after intravenous immunoglobulin (IVIG) treatment of Kawasaki disease (KD). To explore the role of human umbilical cord mesenchymal stem cells (hucMSCs) in inhibiting endothelial inflammation in KD, the effects of hucMSCs on the expression of CD54 and CD105 in endothelial cells in KD were analyzed in vivo and in vitro. METHODS: The concentrations of IL-1ß and VEGF in the peripheral blood of KD or healthy children were detected, and the distributions of CD31+CD54+ and CD31+CD105+ EMPs in platelet-poor plasma (PPP) were analyzed by flow cytometry. Human umbilical vein endothelial cells (HUVECs) were first cocultured with the patients' peripheral blood mononuclear cells (PBMCs). Next, HUVECs were cocultured with hucMSCs after stimulation with inactivated serum from patients. Cell proliferation and migration activities were assessed, and the expression of CD54, CD105 and IL-1ß was analyzed. In an in vivo study, hucMSCs were transplanted into KD mice. The locations and expression levels of CD54, CD105 and IL-1ß in the heart tissues of mice were analyzed. RESULTS: The levels of IL-1ß and CD31+CD54+ EMPs were significantly higher before IVIG treatment and 2 weeks after treatment in KD patients (P < 0.01). However, the levels of VEGF and CD31+CD105+ EMPs increased significantly in KD only after IVIG treatment (P < 0.01). KD-inactivated serum stimulation combined with cocultivation of PBMCs can activate inflammation in HUVECs, leading to reduced cell proliferation and migration activities. Cocultivation also increased the expression of CD54 and decreased the expression of CD105 (P < 0.001). Cocultivation with hucMSCs can reverse these changes. Additionally, hucMSC transplantation downregulated the expression of IL-1ß and CD54 and significantly upregulated the expression of CD105 in KD mice. CONCLUSION: The expression levels of CD31+CD54+ and CD31+CD105+ EMPs showed inconsistent changes at different KD statuses, providing potential markers for clinical application. HucMSCs suppress inflammation and regulate the expression levels of CD54 and CD105 in vascular endothelial cells in KD, possibly providing a new basis for stem cell therapy for KD.


Asunto(s)
Endoglina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Síndrome Mucocutáneo Linfonodular/terapia , Cordón Umbilical/citología , Vasculitis/prevención & control , Animales , Biomarcadores/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Preescolar , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Síndrome Mucocutáneo Linfonodular/complicaciones , Síndrome Mucocutáneo Linfonodular/metabolismo , Síndrome Mucocutáneo Linfonodular/patología , Pronóstico , Vasculitis/etiología , Vasculitis/patología
11.
Front Immunol ; 12: 718136, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34646263

RESUMEN

Angiotensin-converting enzyme 2 (ACE2) is a receptor for the spike protein of SARS-COV-2 that allows viral binding and entry and is expressed on the surface of several pulmonary and non-pulmonary cell types, with induction of a "cytokine storm" upon binding. Other cell types present the receptor and can be infected, including cardiac, renal, intestinal, and endothelial cells. High ACE2 levels protect from inflammation. Despite the relevance of ACE2 levels in COVID-19 pathogenesis, experimental studies to comprehensively address the question of ACE2 regulations are still limited. A relevant observation from the clinic is that, besides the pro-inflammatory cytokines, such as IL-6 and IL-1ß, the anti-inflammatory cytokine IL-10 is also elevated in worse prognosis patients. This could represent somehow a "danger signal", an alarmin from the host organism, given the immuno-regulatory properties of the cytokine. Here, we investigated whether IL-10 could increase ACE2 expression in the lung-derived Calu-3 cell line. We provided preliminary evidence of ACE2 mRNA increase in cells of lung origin in vitro, following IL-10 treatment. Endothelial cell infection by SARS-COV-2 is associated with vasculitis, thromboembolism, and disseminated intravascular coagulation. We confirmed ACE2 expression enhancement by IL-10 treatment also on endothelial cells. The sartans (olmesartan and losartan) showed non-statistically significant ACE2 modulation in Calu-3 and endothelial cells, as compared to untreated control cells. We observed that the antidiabetic biguanide metformin, a putative anti-inflammatory agent, also upregulates ACE2 expression in Calu-3 and endothelial cells. We hypothesized that IL-10 could be a danger signal, and its elevation could possibly represent a feedback mechanism fighting inflammation. Although further confirmatory studies are required, inducing IL-10 upregulation could be clinically relevant in COVID-19-associated acute respiratory distress syndrome (ARDS) and vasculitis, by reinforcing ACE2 levels.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antiinflamatorios/farmacología , COVID-19/enzimología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Interleucina-10/farmacología , Pulmón/efectos de los fármacos , ARN Mensajero/metabolismo , SARS-CoV-2/patogenicidad , Enzima Convertidora de Angiotensina 2/genética , COVID-19/genética , COVID-19/inmunología , Línea Celular , Interacciones Huésped-Patógeno , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Pulmón/enzimología , Pulmón/inmunología , Metformina/farmacología , ARN Mensajero/genética , SARS-CoV-2/inmunología , Regulación hacia Arriba
12.
Biomed Pharmacother ; 144: 112255, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34607110

RESUMEN

Chronic inflammation is characterized by persisting leukocyte infiltration of the affected tissue, which is enabled by activated endothelial cells (ECs). Chronic inflammatory diseases remain a major pharmacotherapeutic challenge, and thus the search for novel drugs and drug targets is an ongoing demand. We have identified the natural product vioprolide A (vioA) to exert anti-inflammatory actions in vivo and in ECs in vitro through inhibition of its cellular target nucleolar protein 14 (NOP14). VioA attenuated the infiltration of microglia and macrophages during laser-induced murine choroidal neovascularization and the leukocyte trafficking through the vascular endothelium in the murine cremaster muscle. Mechanistic studies revealed that vioA downregulates EC adhesion molecules and the tumor necrosis factor receptor (TNFR) 1 by decreasing the de novo protein synthesis in ECs. Most importantly, we found that inhibition of importin-dependent NF-ĸB p65 nuclear translocation is a crucial part of the action of vioA leading to reduced NF-ĸB promotor activity and inflammatory gene expression. Knockdown experiments revealed a causal link between the cellular target NOP14 and the anti-inflammatory action of vioA, classifying the natural product as unique drug lead for anti-inflammatory therapeutics.


Asunto(s)
Antiinflamatorios/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Inflamación/tratamiento farmacológico , Carioferinas/metabolismo , Leucocitos/efectos de los fármacos , Compuestos Macrocíclicos/farmacología , Proteínas Nucleares/metabolismo , Factor de Transcripción ReIA/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Transporte Activo de Núcleo Celular , Animales , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/inmunología , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Carioferinas/genética , Leucocitos/inmunología , Leucocitos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Proteínas Nucleares/genética , Factor de Transcripción ReIA/genética
13.
Immunity ; 54(9): 1989-2004.e9, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34363750

RESUMEN

The migration of neutrophils from the blood circulation to sites of infection or injury is a key immune response and requires the breaching of endothelial cells (ECs) that line the inner aspect of blood vessels. Unregulated neutrophil transendothelial cell migration (TEM) is pathogenic, but the molecular basis of its physiological termination remains unknown. Here, we demonstrated that ECs of venules in inflamed tissues exhibited a robust autophagic response that was aligned temporally with the peak of neutrophil trafficking and was strictly localized to EC contacts. Genetic ablation of EC autophagy led to excessive neutrophil TEM and uncontrolled leukocyte migration in murine inflammatory models, while pharmacological induction of autophagy suppressed neutrophil infiltration into tissues. Mechanistically, autophagy regulated the remodeling of EC junctions and expression of key EC adhesion molecules, facilitating their intracellular trafficking and degradation. Collectively, we have identified autophagy as a modulator of EC leukocyte trafficking machinery aimed at terminating physiological inflammation.


Asunto(s)
Autofagia/fisiología , Células Endoteliales/fisiología , Infiltración Neutrófila/fisiología , Migración Transendotelial y Transepitelial/fisiología , Animales , Quimiotaxis de Leucocito/fisiología , Células Endoteliales/patología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Uniones Intercelulares/fisiología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/fisiología
14.
Mol Pharm ; 18(9): 3235-3246, 2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34387081

RESUMEN

Nanoparticles (NPs) have wide potential applications in the biomedical field. To promote targeted and controlled delivery of encapsulated drugs, it is fundamentally important to understand the factors regulating NP uptake by different cells. Thus, the goal of the present study is to assess the internalization rates of different NPs under normal and proinflammatory states in primary human articular chondrocytes (hACs), human umbilical vein endothelial cells (EA), and human monocytes (THP-1). Here, we compared chitosan-hyaluronic acid (Ch-HA) polymeric NPs, methoxypolyethylene glycol amine-glutathione-palmitic acid (mPEG-GSHn-PA) micelles, and cholesterol/l-α-phosphatidylcholine/DSPE-PEG-Mal (Chol/EPC/DSPE-PEG-Mal) unilamellar liposomes (LUVs). Our results reveal the importance of surface charge and chemistry in determining the levels of NP internalization. Under normal conditions, the cellular uptake was ≈30% for Ch-HA NPs and ≈100% for mPEG-GSHn-PA micelles and Chol/EPC/DSPE-PEG-Mal LUVs. A proinflammatory cell state promoted a higher uptake of the Ch-HA NPs by EA cells (93% after 24 h). Since the therapeutic efficacy of the NP-loaded cargo is dependent on trafficking routes after cellular internalization, we tested their internalization pathways. Accordingly, caveolae-mediated endocytosis or energy-independent non-endocytic pathways, which circumvent lysosomal degradation, were accomplished in hACs and EA by LUVs and in M1 polarized macrophages by micelles. The present outcomes highlight the importance of considering cellular uptake and internalization pathways by the target cell when designing functional NPs for therapeutic applications.


Asunto(s)
Antiinflamatorios/administración & dosificación , Artritis/tratamiento farmacológico , Sistema de Administración de Fármacos con Nanopartículas/farmacocinética , Artritis/inmunología , Condrocitos/inmunología , Condrocitos/metabolismo , Endocitosis , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Micelas , Polímeros/química , Cultivo Primario de Células , Células THP-1
15.
Front Immunol ; 12: 623610, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093519

RESUMEN

Severe cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) strongly hampered the broad clinical applicability of chimeric antigen receptor T cell (CAR-T) therapy. Vascular endothelial activation has been suggested to contribute to the development of CRS and ICANS after CAR-T therapy. However, therapeutic strategies targeting endothelial dysfunction during CAR-T therapy have not been well studied yet. Here, we found that tumor necrosis factor α (TNFα) produced by CAR-T cells upon tumor recognition and interleukin 1ß (IL1ß) secreted by activated myeloid cells were the main cytokines in inducing endothelial activation. Therefore, we investigated the potential effectiveness of TNFα and IL1ß signaling blockade on endothelial activation in CAR-T therapy. The blockade of TNFα and IL1ß with adalimumab and anti-IL1ß antibody respectively, as well as the application of focal adhesion kinase (FAK) inhibitor, effectively ameliorated endothelial activation induced by CAR-T, tumor cells, and myeloid cells. Moreover, adalimumab and anti-IL1ß antibody exerted synergistic effect on the prevention of endothelial activation induced by CAR-T, tumor cells, and myeloid cells. Our results indicate that TNFα and IL1ß blockade might have therapeutic potential for the treatment of CAR-T therapy-associated CRS and neurotoxicity.


Asunto(s)
Adalimumab/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inmunoterapia Adoptiva , Interleucina-1beta/antagonistas & inhibidores , Células Mieloides/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/efectos de los fármacos , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Antígenos CD19/genética , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/metabolismo , Síndrome de Liberación de Citoquinas/prevención & control , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/inmunología , Humanos , Inmunoterapia Adoptiva/efectos adversos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Síndromes de Neurotoxicidad/inmunología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/prevención & control , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
16.
Int Immunol ; 33(8): 447-458, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34089617

RESUMEN

The extracellular matrix (ECM) is the basis for virtually all cellular processes and is also related to tumor metastasis. Fibronectin (FN), a major ECM macromolecule expressed by different cell types and also present in plasma, consists of multiple functional modules that bind to ECM-associated, plasma, and cell-surface proteins such as integrins and FN itself, thus ensuring its cell-adhesive and modulatory role. Here we show that FN constitutes an immune checkpoint. Thus, FN was identified as a physiological ligand for a tumor/leukemia/lymphoma- as well as autoimmune-associated checkpoint, ILT3/LILRB4 (B4, CD85k). Human B4 and the murine ortholog, gp49B, bound FN with sub-micromolar affinities as assessed by bio-layer interferometry. The major B4-binding site in FN was located at the N-terminal 30-kDa module (FN30), which is apart from the major integrin-binding site present at the middle of the molecule. Blockade of B4-FN binding such as with B4 antibodies or a recombinant FN30-Fc fusion protein paradoxically ameliorated autoimmune disease in lupus-prone BXSB/Yaa mice. The unexpected nature of the B4-FN checkpoint in autoimmunity is discussed, referring to its potential role in tumor immunity.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Fibronectinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Comunicación Celular/inmunología , Línea Celular Tumoral , Células Cultivadas , Fibronectinas/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Glicoproteínas de Membrana/inmunología , Ratones , Fagocitosis/inmunología , Células RAW 264.7 , Receptores Inmunológicos/inmunología , Células THP-1/inmunología , Células THP-1/metabolismo
17.
Methods Mol Biol ; 2308: 235-251, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34057727

RESUMEN

Over the last 20 years, significant progress has been made in the development of immunodeficient mouse models that now represents the gold standard tool in stem cell biology research. The latest major improvement has been the use of biomaterials in these xenogeneic mouse models to generate human "bone marrow like" tissues, which not only provides a more relevant xenograft model but can also potentially enable us to delineate the interactions that are specific between human bone marrow cells. There are a number of biomaterials and strategies to create humanized niches in immunodeficient mouse models, and the methods can also differ significantly among various research institutes. Here, we describe a protocol to create a humanized 3D collagen-based scaffold human niche in immunodeficient mouse model(s). This humanized in vivo model provides a powerful technique for understanding the human BM microenvironment and the role it plays in the regulation of normal as well as malignant hematopoiesis.


Asunto(s)
Hematopoyesis , Trasplante de Células Madre Hematopoyéticas/instrumentación , Células Madre Hematopoyéticas/fisiología , Huésped Inmunocomprometido , Nicho de Células Madre , Andamios del Tejido , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Células Cultivadas , Técnicas de Cocultivo , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/fisiología , Ratones , Ratones Mutantes , Fenotipo , Trasplante Heterólogo
18.
Front Immunol ; 12: 650522, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33897698

RESUMEN

Proinflammatory stimuli lead to endothelial injury, which results in pathologies such as cardiovascular diseases, autoimmune diseases, and contributes to alloimmune responses after organ transplantation. Both mesenchymal stromal cells (MSC) and the extracellular vesicles (EV) released by them are widely studied as regenerative therapy for the endothelium. However, for therapeutic application, the manipulation of living MSC and large-scale production of EV are major challenges. Membrane particles (MP) generated from MSC may be an alternative to the use of whole MSC or EV. MP are nanovesicles artificially generated from the membranes of MSC and possess some of the therapeutic properties of MSC. In the present study we investigated whether MP conserve the beneficial MSC effects on endothelial cell repair processes under inflammatory conditions. MP were generated by hypotonic shock and extrusion of MSC membranes. The average size of MP was 120 nm, and they showed a spherical shape. The effects of two ratios of MP (50,000; 100,000 MP per target cell) on human umbilical vein endothelial cells (HUVEC) were tested in a model of inflammation induced by TNFα. Confocal microscopy and flow cytometry showed that within 24 hours >90% of HUVEC had taken up MP. Moreover, MP ended up in the lysosomes of the HUVEC. In a co-culture system of monocytes and TNFα activated HUVEC, MP did not affect monocyte adherence to HUVEC, but reduced the transmigration of monocytes across the endothelial layer from 138 ± 61 monocytes per microscopic field in TNFα activated HUVEC to 61 ± 45 monocytes. TNFα stimulation induced a 2-fold increase in the permeability of the HUVEC monolayer measured by the translocation of FITC-dextran to the lower compartment of a transwell system. At a dose of 1:100,000 MP significantly decreased endothelial permeability (1.5-fold) respect to TNFα Stimulated HUVEC. Finally, MP enhanced the angiogenic potential of HUVEC in an in vitro Matrigel assay by stimulating the formation of angiogenic structures, such as percentage of covered area, total tube length, total branching points, total loops. In conclusion, MP show regenerative effects on endothelial cells, opening a new avenue for treatment of vascular diseases where inflammatory processes damage the endothelium.


Asunto(s)
Tejido Adiposo/citología , Vesículas Extracelulares/inmunología , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Madre Mesenquimatosas/inmunología , Monocitos/inmunología , Adhesión Celular/inmunología , Permeabilidad de la Membrana Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Microscopía por Crioelectrón , ADN/genética , ADN/aislamiento & purificación , Vesículas Extracelulares/genética , Vesículas Extracelulares/ultraestructura , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Microscopía Electrónica de Transmisión , Monocitos/citología , Tamaño de la Partícula , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Int J Mol Sci ; 22(4)2021 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-33670600

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the synovial joints. Inflammation, new blood vessel formation (angiogenesis) and bone resorption (osteoclastogenesis) are three key processes involved in the joint damage and deformities of arthritis. Various gut microbiota-derived metabolites are implicated in RA pathogenesis. However, there is barely any information about the impact of two such metabolites, indole-3-aldehyde (IAld) and indole-3-acetic acid (I3AA), on arthritis-related processes. We conducted a comparative analysis of IAld and I3AA using established cell-based models to understand how they might influence RA pathogenesis. Although structurally similar, the bioactivities of these two metabolites were profoundly different. IAld but not I3AA, inhibited the expression of pro-inflammatory cytokines (IL-1ß and IL-6) in RAW 264.7 (RAW) cells stimulated with heat-killed M. tuberculosis sonicate (Mtb) and lipopolysaccharide (LPS). IAld also exhibited pro-angiogenic activity and pro-osteoclastogenic activity. In contrast, I3AA exhibited anti-angiogenic activity on endothelial cell tube formation but had no effect on osteoclastogenesis. Both IAld and I3AA have been proposed as aryl hydrocarbon receptor (AhR) agonists. Use of CH-223191, an inhibitor of the AhR, suppressed the anti-angiogenic activity of I3AA but failed to mitigate the effects of IAld. Further investigation of the anti-inflammatory activities of IAld and I3AA in LPS-treated RAW cells indicated that inhibition of MyD88-dependent activation of NF-κB and MAPK pathways was not likely involved. Our results suggest that the relative bioavailability of these indole derivatives may differentially impact RA progression and possibly other diseases that share similar cellular processes.


Asunto(s)
Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Citocinas/inmunología , Ácidos Indolacéticos/inmunología , Indoles/inmunología , Microbiota/inmunología , Animales , Artritis Reumatoide/metabolismo , Enfermedades Autoinmunes/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Células Cultivadas , Citocinas/metabolismo , Calor , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ácidos Indolacéticos/metabolismo , Ácidos Indolacéticos/farmacología , Indoles/metabolismo , Indoles/farmacología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/inmunología , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/inmunología , Células RAW 264.7
20.
Int Immunopharmacol ; 95: 107476, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33676147

RESUMEN

BACKGROUND: Vitamin C (ascorbic acid, AscH2) has been shown to enhance immunity. Here, we studied its immunomodulatory effect on human endothelial cells (ECs) during S. aureus infection. MATERIALS AND METHODS: The ex vivo effects of AscH2 were performed on primary human umbilical vein endothelial cells (HUVECs) infected or not with S. aureus. RESULTS: AscH2 treatment induced a marked downregulation of nitric oxide (NO) production and a moderate upregulation of arginase activity in S. aureus-infected HUVECs (respectively, p < 0.05 and p > 0.05). Although the upregulated release levels of soluble intercellular adhesion molecular 1 (sICAM-1/sCD54) and sE-selectin (sCD62E) molecules were not significantly different between treated and untreated S. aureus-infected HUVECs, AscH2 treatment induced reversing effect on sICAM-1 release when comparing to uninfected control HUVECs. Moreover, AscH2 treatment appears to have a significant effect on preventing HUVEC necrosis induced by S. aureus infection (p < 0.05). Furthermore, AscH2 treatment induced a significant upregulation of cell protective redox biomarker in S. aureus-infected, as shown by superoxide dismutase (SOD) activity (p < 0.05), but not by catalase activity (p > 0.05). Additionally, S. aureus infection markedly downregulated total bound calcium ions (bCa2+) levels as compared to control HUVECs, whereas, AscH2 treatment induced a slight upregulation of bCa2+ levels in infected HUVECs as compared to infected and untreated HUVECs (p > 0.05). On the other hand, AscH2 treatment downregulated increased total cellular cholesterol content (tccCHOL) levels in HUVECs induced by S. aureus infection (p < 0.05). In addition, AscH2 treatment markedly reversed S. aureus effect on upregulation of intracellular glucose (iGLU) levels within infected HUVECs (p < 0.05). Moreover, AscH2 treatment significantly downregulated S. aureus growth (p < 0.05), and significantly upregulated bacterial internalization and intracellular killing by HUVECs (p < 0.05), as well as their cell cycle activation (p < 0.01). Finally, AscH2 treatment has a slight effect on the production of interleukin 6 (IL-6), but induced a marked downregulation of that of IL-1ß in S. aureus-infected HUVECs (respectively, p > 0.05, and p < 0.05). CONCLUSIONS: Our outcomes demonstrated that, during S. aureus infection, AscH2 treatment promotes human ECs survival and function, as well as prevents inflammatory response exacerbation, while inducing bactericidal activity.


Asunto(s)
Antibacterianos/farmacología , Ácido Ascórbico/análogos & derivados , Ácido Ascórbico/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Factores Inmunológicos/farmacología , Staphylococcus aureus , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/microbiología , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Óxido Nítrico/inmunología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...