In Vitro Secretion Profile of Pro-Inflammatory Cytokines IL-1β, TNF-α, IL-6, and of Human Beta-Defensins (HBD)-1, HBD-2, and HBD-3 from Human Chorioamniotic Membranes After Selective Stimulation with Gardnerella vaginalis

2011 ◽  
Vol 67 (1) ◽  
pp. 34-43 ◽  
Author(s):  
Veronica Zaga-Clavellina ◽  
Ruiz Velasco-Muñoz Martha ◽  
Pilar Flores-Espinosa
2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Dan Li ◽  
Chenyu Li ◽  
Yan Xu

Abstract Background and Aims Acute kidney injury (AKI), commonly appeared in cardiac arrest, surgery and kidney transplantation which involved in ischemia-reperfusion (IR) injury of kidney. However, the mechanisms underlying inflammatory response in IR AKI is still unclear. Method Public dataset showed kruppel-like factor 6 (KLF6) was significantly highly expressed (P<0.05) in AKI, implies KLF6 might be associated with AKI. To evaluate the mechanism of KLF6 on IR AKI, 30 rats were randomly divided into sham and IR group, and were sacrificed at 0 h, 3 h, 6 h, 12 h or 24 h after IR. Results The results showed KLF6 expression was peaking at 6 h after IR, and the expression of pro-inflammatory cytokines MCP-1 and TNF-α were increased both in serum and kidney tissues after IR, while anti-inflammatory cytokine IL-10 was decreased after IR. Furthermore, in vitro results showed KLF6 knock-down reduced the pro-inflammatory cytokines expression and increased the anti-inflammatory cytokines expression. Conclusion These results suggest that (1) KLF6 might be a novel biomarker for early diagnosis of AKI and (2) targeting KLF6 expression may offer novel strategies to protect kidneys from IR AKI Figure KLF6, AKI, Control Inflammation


2016 ◽  
Vol 11 (6) ◽  
pp. 1934578X1601100
Author(s):  
Anna K Gazha ◽  
Lyudmila A. Ivanushko ◽  
Eleonora V. Levina ◽  
Sergey N. Fedorov ◽  
Tatyana S. Zaporozets ◽  
...  

The action of seven polyhydroxylated sterol mono- and disulfates (1-7), isolated from ophiuroids, on innate and adaptive immunity was examined in in vitro and in vivo experiments. At least, three of them (1, 2 and 4) increased the functional activities of neutrophils, including levels of oxygen-dependent metabolism, adhesive and phagocytic properties, and induced the expression of pro-inflammatory cytokines TNF-α and IL-8. Compound 4 was the most active for enhancing the production of antibody forming cells in the mouse spleen.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4094-4094
Author(s):  
Dorian Forte ◽  
Daria Sollazzo ◽  
Nicola Polverelli ◽  
Romano Marco ◽  
Lara Rossi ◽  
...  

Abstract Introduction. Myelofibrosis (MF), an acquired clonal disorder of the hematopoietic stem/progenitor cell (HSPC) with a dysregulation in JAK/STAT signalling (mutations in JAK2, MPL and Calreticulin (CALR) genes), is characterized by a state of chronic inflammation. It is argued that the up-regulated production of proinflammatory cytokines by both HSPCs and the surrounding stromal cells generates a microenvironment that selects for the malignant clone. Only recently, it has been hypothesized that the sustained inflammatory microenvironment of MF can alter crucial biological processes, leading to genomic instability and cancer progression. Here we tested the in vitro functional effects of pivotal players of the inflammatory microenvironment (the extracellular ATP nucleotide and selected cytokines, such as Interleukin (IL)-1β, Tumor Necrosis Factor (TNF)-α or the Tissue Inhibitor of Metalloproteinases-1 (TIMP-1)) on the HSPCs from MF patients. Methods: Circulating CD34+/CD34+ CD38- cells from MF patients (JAK2V617F (17 cases) and CALR (9 cases) mutations) or cord blood (CB; 8 samples) were phenotypically and functionally characterized after in vitro incubation with or without ATP (1000 μM), IL-1β (10 ng/mL), TNF-α (10 ng/mL) or TIMP-1 (100 ng/mL) (alone or in combination). Cells were then analyzed for survival/apoptosis (Annexin-V/Propidium Iodide staining), phenotype (evaluation of CD63 (TIMP-1 receptor), CXCR4 and CD38 expression), cell cycle and clonogenic capacity. Migration was assessed first towards a CXCL12 gradient in the presence or absence of the pro-inflammatory factors. In parallel experiments, CD34+ cells from MF patients were co-cultured with normal mesenchymal stromal cells (MSCs) in the presence or absence of the pro-inflammatory cytokines and then evaluated for their ability to migrate towards a CXCL12 gradient. Plasma TIMP-1, TNF-α, IL-1β and CXCL12 were measured by ELISA assay. Results: The plasma levels of TIMP-1, TNF-α, IL-1β, CXCL12 and the number of circulating CD34+, CD34+ CD38-, CD34+ CD63+, CD34+ CD184+ cells were increased in MF patients. According to mutational status, the CD34+ CD63+ cells were higher in the CALR+ patients. The survival of MF CD34+ cells was strongly stimulated by in vitro incubation with TNF-α or IL-1β as compared with the CB-derived CD34+ cells or untreated cells. By multiple cytokine combinations, IL-1β/TIMP-1, IL-1β /ATP or IL-1β /TNF-α treatments significantly promote the survival of MF CD34+ cells as compared with the normal counterparts or the untreated cells. Various combinations with IL-1β were also effective in stimulating survival of CD34+CD38- cells. IL-1β/TIMP-1 and IL-1β/TNF-α/TIMP-1, but not factors alone, significantly increased the CFU-C growth of MF patients as compared with the CB-derived counterparts and the untreated cells. Moreover, comparing CALR+ vs JAK2V617F+ patients, the colony formation of JAK2V617F+ patients was mainly promoted by the IL-1β/TNF-α treatment. Along with clonogenic capacity stimulation, exposure of CD34+ cells from MF patients to IL-1β/TNF-α/TIMP-1 significantly increases the S-phase cells, suggesting that these pro-inflammatory factors stimulated cell-cycle progression in dormant CD34+ MF cells. Migration of CD34+ cells from MF was significantly increased in CXCL12 treated cells. In addition, exposure of MF CD34+ cells to IL-1β/TNF-α, IL-1β/TIMP-1 or IL-1β/TNF-α/TIMP-1 significantly promotes cell migration in comparison with the CB-derived counterparts or SDF-1 alone. MF migrated cells in the presence of IL-1β/TNF-α significantly upregulate CD63 expression. Intriguingly, colony formation of MF migrated CD34+ cells in the presence of IL-1β/TNF-α or IL-1β/TNF-α/TIMP-1 was potently increased. Finally, co-culture systems with normal MSCs in the presence of pro-inflammatory factors revealed that MF CD34+ cells display increased migration ability toward CXCL12 gradient. Conclusions: Altogether our findings suggest that in MF the inflammatory niche plays a key role in the maintenance of the malignant clone. Thus, the interplay between the pro-inflammatory cytokines promote and select the HSPCs with higher proliferative activity, clonogenic potential and migration capability. Targeting these microenvironmental interactions may be a clinically relevant approach. D.F. and D.S. equally contributed Disclosures Martinelli: Pfizer: Consultancy; Ariad: Consultancy; Novartis: Consultancy, Speakers Bureau; MSD: Consultancy; AMGEN: Consultancy; BMS: Consultancy, Speakers Bureau; ROCHE: Consultancy.


2011 ◽  
Vol 49 (2) ◽  
pp. 168-173
Author(s):  
F. Sachse ◽  
K. Becker ◽  
T.J. Basel ◽  
D. Weiss ◽  
C. Rudack

BACKGROUND: Nasal polyposis (NP) is considered a subgroup within chronic rhinosinusitis. NP can be further subdivided into aspirin sensitive- and aspirin tolerant types (ASNP/ ATNP). Although the true etiology of NP has not been identified so far, it is agreed that NP represents an inflammatory disease of the nasal mucosa. Alterations of cellular kinase activities including that of IKK-2 might play a role in this inflammatory process. METHODS: Paraffin sections of ASNP, ATNP and controls were immunostained with Phospho-IkB-α antibody that detects the direct IKK-2 product (IkB-α. Intensity of epithelial staining was analysed semi-quantitatively by two independent observers. In cultured nasal polyp epithelial cells (NPECs) epithelial derived cytokines IL-8 and GRO α were induced by TNF-α or Staphylococcal supernatants and subsequently repressed by IKK-2 inhibitor TPCA-1. RESULTS: Significant Phospho-IkB-α staining was observed in the nasal epithelium of ASNP compared to ATNP and controls suggesting strong IKK-2 activation in patients with ASNP in vivo. In vitro, pro-inflammatory cytokines IL-8 and GRO-α in NPECs were significantly repressed by TPCA-1. CONCLUSION: IKK-2 activity is increased in the subgroup of ASNP. IL-8 and GRO-α responses were repressed by IKK-2 inhibitor TPCA-1 in vitro. IKK-2 inhibitors might represent a potential target for anti-inflammatory intervention in ASNP.


2021 ◽  
Vol 21 (02) ◽  
Author(s):  
Yaolei Ge

ABSTRACT The present study examined functions of miR-200a-3p accelerated progressions of HCM cells via IGF2R and Wnt/β-catenin signalling pathway after hypoxia/reoxygenation treatment in vitro. CCK-8 showed that cell viability of HCM was inhibited while apoptosis rates detected by flow cytometry were promoted in a time dependent manner after H/R (12 hours and 24 hours). Beyond that, Bcl-2 and c-IAP1 were decreased but Bax and caspase-3 were upregulated by H/R treatment. IL-1β, IL-6, TNF-α and NLRP3 were also increased after treatment. RT-qPCR showed increased expressions of miR-200a-3p by H/R treatment while its inhibitor elevated cell viability but depressed apoptosis rate and pro-inflammatory cytokines’ expressions. IGF2R was upregulated after H/R treatment and its downregulation magnified effects of suppressed miR-200a-3p. HIF-1α/Wnt/β -catenin signalling pathway was activated by miR-200a-3p and IGF2R while IWP-2 treatment abolished the activation of Wnt3a andβ -catenin, causing decreased apoptosis and pro-inflammatory cytokines’ expressions but accelerated the cell viability.


Author(s):  
N. Osakue ◽  
C. C. Onyenekwe ◽  
F. A. Ehiaghe ◽  
J. E. Ahaneku ◽  
J. I. Ikechebelu ◽  
...  

Background: In vitro fertilization (IVF) is an assisted reproductive technology (ART) that is widely used globally in the treatment of infertility. Infertility can occur due to male factors, female factors or both. Aim: This is the first Nigerian study that sets out to observe the levels and relationship between circulating pro-inflammatory cytokines (IFN-γ, TNF-α) and progesterone (PG) in Nigerian women undergoing in vitro fertilization pre and post treatment and their possible effect on pregnancy outcome. Materials and Methods: This observational study randomly selected sixty-two (62) infertile females below 45 year of age who enrolled in the IVF treatment at Lily Hospitals, Warri and Shepherd Specialist Hospital, Warri, Southern Nigeria. Only data of the thirteen (13) infertile females who became pregnant after the IVF treatment where followed up and presented in this study. Five (5) ml of whole blood were collected into plain tubes on day 3 of the menstrual cycle of all the participants from the ante-cubital vein before and after IVF procedure using standard laboratory collection technique. Ovarian stimulation was done using the long gonadotropin-releasing hormone agonist protocol. Oocyte retrieval transfer was done using ultrasound-guided fine-needle aspiration and embryo transfer was done using ultrasound-guided embryo transfer. IFN-γ, TNF-α and PG were estimated using enzyme-linked immunosorbent assay method. Results and Conclusion: Significant increase in the levels of TNF-α and PG at the second trimester and third trimester of pregnancy when compared with the first trimester of pregnancy (p = 0.000). While the level of IFN-γ was significantly increased in the second trimester of pregnancy when compared with the first trimester of pregnancy (p = 0.000). It is evident from the study that both pro-inflammatory cytokines (IFN-γ and TNF-α) act in synergy to maintain the level of progesterone which act as an anti-inflammatory agent to regulate the activities of the pro-inflammatory cytokines for successful oocytes implantation and maturation.


PeerJ ◽  
2020 ◽  
Vol 8 ◽  
pp. e9533 ◽  
Author(s):  
Zhiyu Wang ◽  
Yanfei Wang ◽  
Prachi Vilekar ◽  
Seung-Pil Yang ◽  
Mayuri Gupta ◽  
...  

The novel coronavirus SARS-CoV-2 has become a global health concern. The morbidity and mortality of the potentially lethal infection caused by this virus arise from the initial viral infection and the subsequent host inflammatory response. The latter may lead to excessive release of pro-inflammatory cytokines, IL-6 and IL-8, as well as TNF-α ultimately culminating in hypercytokinemia (“cytokine storm”). To address this immuno-inflammatory pathogenesis, multiple clinical trials have been proposed to evaluate anti-inflammatory biologic therapies targeting specific cytokines. However, despite the obvious clinical utility of such biologics, their specific applicability to COVID-19 has multiple drawbacks, including they target only one of the multiple cytokines involved in COVID-19’s immunopathy. Therefore, we set out to identify a small molecule with broad-spectrum anti-inflammatory mechanism of action targeting multiple cytokines of innate immunity. In this study, a library of small molecules endogenous to the human body was assembled, subjected to in silico molecular docking simulations and a focused in vitro screen to identify anti-pro-inflammatory activity via interleukin inhibition. This has enabled us to identify the loop diuretic furosemide as a candidate molecule. To pre-clinically evaluate furosemide as a putative COVID-19 therapeutic, we studied its anti-inflammatory activity on RAW264.7, THP-1 and SIM-A9 cell lines stimulated by lipopolysaccharide (LPS). Upon treatment with furosemide, LPS-induced production of pro-inflammatory cytokines was reduced, indicating that furosemide suppresses the M1 polarization, including IL-6 and TNF-α release. In addition, we found that furosemide promotes the production of anti-inflammatory cytokine products (IL-1RA, arginase), indicating M2 polarization. Accordingly, we conclude that furosemide is a reasonably potent inhibitor of IL-6 and TNF-α that is also safe, inexpensive and well-studied. Our pre-clinical data suggest that it may be a candidate for repurposing as an inhaled therapy against COVID-19.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1686-1686
Author(s):  
Martina Barone ◽  
Francesca Ricci ◽  
Marco Romano ◽  
Dorian Forte ◽  
Giuseppe Auteri ◽  
...  

Introduction: Myelofibrosis (MF) is a chronic myeloproliferative neoplasm characterized by hyper-activation of the JAK-STAT pathway. More than half of the patients carries the JAK2V617F mutation. Cytokine overproduction, which is the hallmark of MF, is driven by multiple signaling pathways (NF-KB and MAPK) beyond JAK-STAT and is reduced but not abrogated by Ruxolitinib (RUX). RUX, which is the only JAK1/2 inhibitor approved for the treatment of splenomegaly and symptoms associated with MF, suppresses myeloproliferation (JAK2-driven) and release of pro-inflammatory cytokines (JAK1-driven). Microvesicles (MVs), which have a role in the inflammatory network and are critical players in the regulation of immunity through cytokine signaling, are released from a broad variety of cells with effects on communication among cells. Infections are one of the main causes of morbidity and mortality in MF. The increased infectious risk is thought to arise from dysfunction of key immune cells, including T, natural killer and dendritic cells, which further aggravates after JAK1/2 inhibition therapy. In this scenario, even though MF monocytes (Mo) are over-activated and show inflammatory features, their contribution still needs to be clarified. Aims: To study the role of circulating Mo in the inflammatory network of MF and to investigate whether and to what extent in vivoJAK1/2 inhibition may affects their in vitro cytokine producing ability. Methods: EDTA-anticoagulated peripheral blood was collected from 12 JAK2V617F mutatedMF patients before (Baseline) and after 6 months of RUX therapy and from 10 age/sex-matched healthy donors (HD). After 4 hours in vitro stimulation of mononuclear cells (PBMCs) with lipopolysaccharides (LPS) and in the presence of brefeldin A, the Interleukin (IL)-1β, -6, -10 and Tumor Necrosis Factor (TNF)-α producing Mo (CD14+ cells) were measured by intracellular staining and flow cytometry analysis. In parallel experiments, upon LPS stimulation, free and MVs-bound cytokines (IL-1β, IL-6, IL-10 and TNF-α) have been measured in the supernatants of immunomagnetically isolated HD/MF-Mo by flow cytometry. In addition, after isolation with ultracentrifugation from platelet poor plasma, titrating doses of circulating HD/MF MVs were co-cultured for 24 hours with immunomagnetically isolated HD-Mo and, upon LPS stimulation, inflammatory cytokines secretion was analysed in the supernatants by flow cytometry. Results: To characterize the cytokine producing ability of Mo we analyzed the IL1-β, TNF-α, IL-6 and IL-10 positive MF/HD Mo in response to LPS stimulation. At baseline, the percentages of MF-Mo producing pro-inflammatory cytokines (IL-1β, IL-6, TNF-α) after 4 hours LPS stimulation was highly reduced as compared with the HD counterparts (Fig. 1). No IL-10-positive cells were detected with LPS stimulation (data not shown). To confirm the data, we analysed the free and MVs-bound cytokines in the culture supernatants upon LPS stimulation. At baseline, MF-Mo showed defective capacity to secrete free (Fig.2) and MVs-bound (Fig.3) IL-1β, IL-6, TNF-α and IL-10. Interestingly, the isolated circulating MF-MVs inhibited the LPS-driven inflammatory cytokines in vitro secretion of HD-Mo (Fig.4). Six months-RUX therapy reactivated the in vitro MF-Mo ability to produce intracellular inflammatory cytokines (Fig.1) and to secrete MVs-bound inflammatory cytokines in response to LPS stimulation (Fig.3). Conversely, the MF-Mo ability to secrete free cytokines in the supernatants in response to LPS stimulus remained lower than the HD counterparts (Fig.2). Conclusions: These data further refine the immune dysfunction of MF by demonstrating defective cytokine production/secretion of circulating Mo in response to infectious stimulus (LPS). This defect might be due, at least in part, to the inhibitory activity of circulating MF-MVs. Importantly, in vivoJAK1/2 inhibition ameliorates Mo cytokines production and promotes the MVs-based inflammatory cytokine signaling, suggesting that the increased infectious risk of MF patients undergoing RUX therapy is not due to defective inflammatory signals of circulating Mo. These findings contribute to better interpreting the off-target efficacy of JAK1/2 inhibition and to envisaging strategies aimed at facilitating the immune surveillance in MF. Disclosures Martinelli: BMS: Consultancy; Pfizer: Consultancy; ARIAD: Consultancy; Novartis: Consultancy; Roche: Consultancy. Cavo:janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel accommodations, Speakers Bureau; celgene: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: travel accommodations, Speakers Bureau; AbbVie: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; novartis: Honoraria; sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; bms: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Palandri:Novartis: Consultancy, Honoraria.


Dose-Response ◽  
2020 ◽  
Vol 18 (4) ◽  
pp. 155932582096172
Author(s):  
Ilaria Floris ◽  
Thorsten Rose ◽  
Juan Antonio Collado Rojas ◽  
Kurt Appel ◽  
Camille Roesch ◽  
...  

Tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) are pro-inflammatory cytokines involved in acute and chronic inflammatory diseases. Indeed, immunotherapy blocking these 2 cytokines has been developed. Micro-immunotherapy (MI) also uses ultra-low doses (ULD) of pro-inflammatory cytokines, impregnated on lactose-sucrose pillules, to counteract their overexpression. The study has been conducted with 2 objectives: examine the anti-inflammatory effect in vitro and the capacity of 2 unitary medicines, TNF-α (27 CH) and IL-1β (27 CH), to reduce the secretion of TNF-α in human primary monocytes and THP-1 cells differentiated with phorbol-12-myristate-13-acetate, after lipopolysaccharide (LPS) exposure; then, investigate the presence of particles possibly containing starting materials using tunable resistive pulse sensing technique. The results show that the unitary medicines, tested at 3 pillules concentrations (5.5, 11 and 22 mM), have reduced the secretion of TNF-α in both models by about 10−20% vs. vehicle control, depending on concentration. In this exploratory study, particles (150−1000 nm) have been detected in MI ULD-impregnated pillules and a hypothesis for MI medicines mode of action has been proposed. Conscious that more evaluations are necessary, authors are cautious in the conclusions because the findings described in the study are still limited, and future investigations may lead to different hypothesis.


2016 ◽  
Vol 33 (S1) ◽  
pp. S93-S93
Author(s):  
S. Gnanavel ◽  
A. Parmar ◽  
P. Sharan ◽  
V. Subbiah ◽  
U. Sharma ◽  
...  

IntroductionIn vitro studies have demonstrated possible neuroprotective effects of the following pro-inflammatory cytokines: IL-6, IL-1β and TNF-α against glutaminergic excitotoxicity in brain through different pathways.ObjectivesIn the current study, we aim to correlate level of the above pro-inflammatory cytokines in serum with glutamate levels in head of caudate nucleus measured using Proton Magnetic Resonance Spectroscopy (1H-MRS) in patients with obsessive-compulsive disorder (OCD), a neuropsychiatric illness with possible multifactorial aetiology including immunological and excitotoxic factors.MethodThirty psychotropic-naïve patients with OCD and an equal number of gender and age-matched normal controls were recruited in the study. A detailed psychiatric assessment was carried out including sociodemographic and clinical variables. A 3T MR imaging and spectroscopy session was carried out in head of caudate nucleus. Further, absolute quantification of glutamate level was obtained using LC model. Simultaneously, 5 mL of blood sample was collected and assayed for the above pro-inflammatory cytokines (Siemens, Immulite™). The level of glutamate was correlated with the cytokine levels in patients with OCD.ResultsThe level of Glx was significantly higher in patients with OCD as compared to controls (P < 0.05). The Glx level negatively correlated with two of the three pro-inflammatory cytokines: IL-6 and TNF-α (r = −0.807; r = −0.838; P < 0.05) while no significant correlation was demonstrated with IL-1β.ConclusionsThe findings provide preliminary evidence regarding possible neuroprotective effects of pro-inflammatory cytokines against glutaminergic excitotoxicity in patients with OCD. Further studies including patients with other psychiatric illnesses as controls are required for confirmation of the above findings.Disclosure of interestThe authors have not supplied their declaration of competing interest.


Sign in / Sign up

Export Citation Format

Share Document