Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 319
Filter
1.
Mediators Inflamm ; 2022: 5171525, 2022.
Article in English | MEDLINE | ID: mdl-36091666

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents , Cell Adhesion Molecules , Ferula , Human Umbilical Vein Endothelial Cells , Plant Extracts , Anti-Inflammatory Agents/pharmacology , Cell Adhesion , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , E-Selectin/biosynthesis , E-Selectin/genetics , E-Selectin/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes, Mononuclear/immunology , Plant Extracts/pharmacology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
2.
Biochem Pharmacol ; 201: 115078, 2022 07.
Article in English | MEDLINE | ID: mdl-35551917

ABSTRACT

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.


Subject(s)
Doxorubicin , Eicosapentaenoic Acid , Human Umbilical Vein Endothelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Cellular Senescence/drug effects , Doxorubicin/pharmacology , Drug Interactions , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammasomes/drug effects , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/chemically induced , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
3.
Clin Exp Pharmacol Physiol ; 49(8): 805-812, 2022 08.
Article in English | MEDLINE | ID: mdl-35577580

ABSTRACT

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.


Subject(s)
Atherosclerosis , Cytokines , Dabigatran , Human Umbilical Vein Endothelial Cells , Hydroxycholesterols , Rivaroxaban , Administration, Oral , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/immunology , Atrial Fibrillation/drug therapy , Cytokines/genetics , Cytokines/immunology , Dabigatran/pharmacology , Dabigatran/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Hydroxycholesterols/administration & dosage , Hydroxycholesterols/adverse effects , Hydroxycholesterols/pharmacology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-23/genetics , Interleukin-23/immunology , Oxysterols/administration & dosage , Oxysterols/adverse effects , Oxysterols/pharmacology , RNA, Messenger/genetics , RNA, Messenger/immunology , Rivaroxaban/pharmacology , Rivaroxaban/therapeutic use , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
4.
Int J Mol Sci ; 23(4)2022 Feb 19.
Article in English | MEDLINE | ID: mdl-35216428

ABSTRACT

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.


Subject(s)
Immunomodulation/immunology , Interferon-gamma/immunology , Cell Line, Tumor , Cells, Cultured , Human Umbilical Vein Endothelial Cells/immunology , Humans , Immunity/immunology , Immunologic Factors/immunology , Immunotherapy/methods , Leukocytes, Mononuclear/immunology , THP-1 Cells
5.
Eur J Pharmacol ; 918: 174715, 2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35026193

ABSTRACT

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.


Subject(s)
Atherosclerosis , Benzhydryl Compounds/pharmacology , Gene Expression Regulation/drug effects , Glucosides/pharmacology , Human Umbilical Vein Endothelial Cells , Macrophages , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Anti-Inflammatory Agents/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/immunology , Atherosclerosis/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Hypoglycemic Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Signal Transduction , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
FASEB J ; 36(1): e22085, 2022 01.
Article in English | MEDLINE | ID: mdl-34888952

ABSTRACT

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.


Subject(s)
Eicosanoic Acids/toxicity , Nasal Mucosa/immunology , Ovalbumin/toxicity , Rhinitis, Allergic , Administration, Intranasal , Animals , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Rhinitis, Allergic/chemically induced , Rhinitis, Allergic/immunology
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(12): 1057-1065, 2021 Dec.
Article in Chinese | MEDLINE | ID: mdl-34906292

ABSTRACT

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.


Subject(s)
Hantaan virus , Human Umbilical Vein Endothelial Cells , Immunity, Innate , Cell Line , Hantaan virus/pathogenicity , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/virology , Humans
8.
mBio ; 12(6): e0211321, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34933450

ABSTRACT

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.


Subject(s)
Antigens, Differentiation/immunology , Herpesviridae Infections/immunology , Herpesvirus 8, Human/physiology , Membrane Proteins/immunology , RNA-Binding Proteins/immunology , Rhadinovirus/physiology , Animals , Antigens, Differentiation/genetics , Coinfection/genetics , Coinfection/immunology , Coinfection/virology , Fibroblasts/immunology , Fibroblasts/virology , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/virology , Humans , Membrane Proteins/genetics , RNA-Binding Proteins/genetics , Rhadinovirus/genetics , Species Specificity , Virus Internalization , Virus Replication
9.
Exp Cell Res ; 409(2): 112941, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34822812

ABSTRACT

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.


Subject(s)
Endoglin/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mucocutaneous Lymph Node Syndrome/therapy , Umbilical Cord/cytology , Vasculitis/prevention & control , Animals , Biomarkers/metabolism , Case-Control Studies , Cell Differentiation , Child, Preschool , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/immunology , Humans , Male , Mice , Mice, Inbred C57BL , Mucocutaneous Lymph Node Syndrome/complications , Mucocutaneous Lymph Node Syndrome/metabolism , Mucocutaneous Lymph Node Syndrome/pathology , Prognosis , Vasculitis/etiology , Vasculitis/pathology
10.
Int Immunopharmacol ; 101(Pt A): 108295, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34735917

ABSTRACT

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.


Subject(s)
Atherosclerosis/immunology , Cytokines/metabolism , Disaccharides/pharmacology , Leukocytes, Mononuclear/drug effects , Sugar Phosphates/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Adult , Atherosclerosis/drug therapy , Case-Control Studies , Coculture Techniques , Disaccharides/therapeutic use , Dose-Response Relationship, Drug , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-2/metabolism , Interleukin-6/metabolism , Interleukin-9/metabolism , Interleukins/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Male , Middle Aged , Sugar Phosphates/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
11.
Biomed Pharmacother ; 144: 112255, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607110

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Inflammation/drug therapy , Karyopherins/metabolism , Leukocytes/drug effects , Macrocyclic Compounds/pharmacology , Nuclear Proteins/metabolism , Transcription Factor RelA/metabolism , Transendothelial and Transepithelial Migration/drug effects , Active Transport, Cell Nucleus , Animals , Cell Adhesion/drug effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , Choroidal Neovascularization/drug therapy , Choroidal Neovascularization/immunology , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Coculture Techniques , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Karyopherins/genetics , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Nuclear Proteins/genetics , Transcription Factor RelA/genetics
12.
Front Immunol ; 12: 718136, 2021.
Article in English | MEDLINE | ID: mdl-34646263

ABSTRACT

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.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Anti-Inflammatory Agents/pharmacology , COVID-19/enzymology , Human Umbilical Vein Endothelial Cells/drug effects , Interleukin-10/pharmacology , Lung/drug effects , RNA, Messenger/metabolism , SARS-CoV-2/pathogenicity , Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , COVID-19/immunology , Cell Line , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Lung/enzymology , Lung/immunology , Metformin/pharmacology , RNA, Messenger/genetics , SARS-CoV-2/immunology , Up-Regulation
13.
Immunity ; 54(9): 1989-2004.e9, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34363750

ABSTRACT

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.


Subject(s)
Autophagy/physiology , Endothelial Cells/physiology , Neutrophil Infiltration/physiology , Transendothelial and Transepithelial Migration/physiology , Animals , Chemotaxis, Leukocyte/physiology , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Intercellular Junctions/physiology , Mice , Mice, Inbred C57BL , Neutrophils/physiology
14.
Mol Pharm ; 18(9): 3235-3246, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34387081

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Arthritis/drug therapy , Nanoparticle Drug Delivery System/pharmacokinetics , Arthritis/immunology , Chondrocytes/immunology , Chondrocytes/metabolism , Endocytosis , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Micelles , Polymers/chemistry , Primary Cell Culture , THP-1 Cells
15.
Front Immunol ; 12: 623610, 2021.
Article in English | MEDLINE | ID: mdl-34093519

ABSTRACT

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.


Subject(s)
Adalimumab/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Immunotherapy, Adoptive , Interleukin-1beta/antagonists & inhibitors , Myeloid Cells/drug effects , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/drug effects , Tumor Necrosis Factor Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, CD19/metabolism , Cell Line, Tumor , Coculture Techniques , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/prevention & control , HEK293 Cells , Human Umbilical Vein Endothelial Cells/immunology , Humans , Immunotherapy, Adoptive/adverse effects , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Neurotoxicity Syndromes/immunology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/prevention & control , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
Int Immunol ; 33(8): 447-458, 2021 07 23.
Article in English | MEDLINE | ID: mdl-34089617

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/metabolism , Fibronectins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Animals , Autoimmune Diseases/immunology , Autoimmunity/immunology , Cell Communication/immunology , Cell Line, Tumor , Cells, Cultured , Fibronectins/immunology , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Membrane Glycoproteins/immunology , Mice , Phagocytosis/immunology , RAW 264.7 Cells , Receptors, Immunologic/immunology , THP-1 Cells/immunology , THP-1 Cells/metabolism
17.
Methods Mol Biol ; 2308: 235-251, 2021.
Article in English | MEDLINE | ID: mdl-34057727

ABSTRACT

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.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cell Transplantation/instrumentation , Hematopoietic Stem Cells/physiology , Immunocompromised Host , Stem Cell Niche , Tissue Scaffolds , Animals , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Coculture Techniques , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/physiology , Mice , Mice, Mutant Strains , Phenotype , Transplantation, Heterologous
18.
Front Immunol ; 12: 650522, 2021.
Article in English | MEDLINE | ID: mdl-33897698

ABSTRACT

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.


Subject(s)
Adipose Tissue/cytology , Extracellular Vesicles/immunology , Human Umbilical Vein Endothelial Cells/immunology , Mesenchymal Stem Cells/immunology , Monocytes/immunology , Cell Adhesion/immunology , Cell Membrane Permeability/immunology , Cells, Cultured , Coculture Techniques , Cryoelectron Microscopy , DNA/genetics , DNA/isolation & purification , Extracellular Vesicles/genetics , Extracellular Vesicles/ultrastructure , Human Umbilical Vein Endothelial Cells/cytology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Transmission , Monocytes/cytology , Particle Size , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
19.
Immunopharmacol Immunotoxicol ; 43(2): 230-238, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33657977

ABSTRACT

Neuropilin-1 (NRP-1) regulates a range of physiological and pathological processes, including angiogenesis. Targeting of NRP1 is considered a significant approach in cancer therapy. In the present study, a novel antiNRP1 immunotoxin (αNRP1 IT) was developed by genetic fusion of a single domain (VHH) anti-NRP-1 antibody fragment to a truncated diphtheria toxin. The αNRP1 IT was expressed into bacterial cells as an inclusion body (IB). Expression of αNRP1 IT was confirmed by SDS-PAGE and western blotting. Recombinant αNRP1 IT was purified using nickel affinity chromatography. Toxicity and antiangiogenesis effect of αNRP1 IT was investigated both in vitro and in vivo. Results showed that αNRP1 IT significantly reduced the viability of human umbilical vein endothelial cell line (HUVEC) (p < .05). The αNRP1 IT significantly inhibited tube formation of HUVEC cells (p < .001). Furthermore, αNRP1 IT inhibited angiogenesis in Chick Chorioallantoic Membrane (CAM) Assay. These data suggest the potential of αNRP1 IT as a novel therapeutic in targeted cancer therapy.


Subject(s)
Diphtheria Toxin/administration & dosage , Human Umbilical Vein Endothelial Cells/drug effects , Immunotoxins/administration & dosage , Neovascularization, Pathologic/prevention & control , Neuropilin-1/antagonists & inhibitors , Single-Domain Antibodies/administration & dosage , Animals , Camelus , Cell Survival/drug effects , Cell Survival/immunology , Chickens , Dose-Response Relationship, Drug , HEK293 Cells , Human Umbilical Vein Endothelial Cells/immunology , Humans , MCF-7 Cells , Male , Neovascularization, Pathologic/immunology , Neuropilin-1/immunology
20.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670600

ABSTRACT

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.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Indoleacetic Acids/immunology , Indoles/immunology , Microbiota/immunology , Animals , Arthritis, Rheumatoid/metabolism , Autoimmune Diseases/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Hot Temperature , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Indoleacetic Acids/metabolism , Indoleacetic Acids/pharmacology , Indoles/metabolism , Indoles/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/immunology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/immunology , RAW 264.7 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...