Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.687
1.
J Control Release ; 350: 146-157, 2022 10.
Article En | MEDLINE | ID: mdl-35973471

Chronic ulcerative oral mucosal inflammatory diseases, including oral lichen planus and recurrent aphthous stomatitis, are painful and highly prevalent, yet lack effective clinical management. In recent years, systemic biologic therapies, including monoclonal antibodies that block the activity of cytokines, have been increasingly used to treat a range of immune-mediated inflammatory conditions such as rheumatoid arthritis and psoriasis. The ability to deliver similar therapeutic agents locally to the oral epithelium could radically alter treatment options for oral mucosal inflammatory diseases, where pro-inflammatory cytokines, in particular tumour-necrosis factor-α (TNFα), are major drivers of pathogenesis. To address this, an electrospun dual-layer mucoadhesive patch comprising medical-grade polymers was investigated for the delivery of F(ab) biologics to the oral mucosa. A fluorescent-labelled F(ab) was incorporated into mucoadhesive membranes using electrospinning with 97% v/v ethanol as a solvent. The F(ab) was detected within the fibres in aggregates when visualised by confocal microscopy. Biotinylated F(ab) was rapidly eluted from the patch (97 ± 5% released within 3 h) without loss of antigen-binding activity. Patches applied to oral epithelium models successfully delivered the F(ab), with fluorescent F(ab) observed within the tissue and 5.1 ± 1.5% cumulative transepithelial permeation reached after 9 h. Neutralising anti-TNFα F(ab) fragments were generated from whole IgG by papain cleavage, as confirmed by SDS-PAGE, then incorporated into patches. F(ab)-containing patches had TNFα neutralising activity, as shown by the suppression of TNFα-mediated CXCL8 release from oral keratinocytes cultured as monolayers. Patches were applied to lipopolysaccharide-stimulated immune-competent oral mucosal ulcer equivalents that contained primary macrophages. Anti-TNFα patch treatment led to reduced levels of active TNFα along with a reduction in the levels of disease-implicated T-cell chemokines (CCL3, CCL5, and CXCL10) to baseline concentrations. This is the first report of an effective device for the delivery of antibody-based biologics to the oral mucosa, enabling the future development of new therapeutic strategies to treat painful conditions.


Mucositis , Humans , Immunoglobulin Fab Fragments/administration & dosage , Immunoglobulin Fab Fragments/immunology , Mucositis/drug therapy , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/immunology
2.
Int Immunopharmacol ; 101(Pt B): 108365, 2021 Dec.
Article En | MEDLINE | ID: mdl-34815190

Preeclampsia (PE) is characterized by hypertension, autonomic imbalance and inflammation. The subfornical organ (SFO) reportedly relays peripheral inflammatory mediator's signals to the paraventricular nucleus (PVN), a brain autonomic center shown to mediate hypertension in hypertensive rat but not yet in PE rat models. Additionally, we previously showed that Pyridostigmine (PYR), an acetylcholinesterase inhibitor, attenuated placental inflammation and hypertension in PE models. In this study, we investigated the effect of PYR on the activities of these brain regions in PE model. PYR (20 mg/kg/day) was administered to reduced uterine perfusion pressure (RUPP) Sprague-Dawley rat from gestational day (GD) 14 to GD19. On GD19, the mean arterial pressure (MAP) was recorded and samples were collected for analysis. RUPP rats exhibited increased MAP (P = 0.0025), elevated circulating tumor necrosis factor-α (TNF-α, P = 0.0075), reduced baroreflex sensitivity (BRS), increased neuroinflammatory markers including TNF-α, interleukin-1ß (IL-1ß), microglial activation (P = 0.0039), oxidative stress and neuronal excitation within the PVN and the SFO. Changes in MAP, in molecular and cellular expression induced by RUPP intervention were improved by PYR. The ability of PYR to attenuate TNF-α mediated central effect was evaluated in TNF-α-infused pregnant rats. TNF-α infusion-promoted neuroinflammation in the PVN and SFO in dams was abolished by PYR. Collectively, our data suggest that PYR improves PE-like symptoms in rat by dampening placental ischemia and TNF-α-promoted inflammation and pro-hypertensive activity in the PVN. This broadens the therapeutical potential of PYR in PE.


Cholinesterase Inhibitors/pharmacology , Hypertension/drug therapy , Neuroinflammatory Diseases/drug therapy , Paraventricular Hypothalamic Nucleus/drug effects , Pre-Eclampsia/drug therapy , Pyridostigmine Bromide/pharmacology , ATP-Binding Cassette Transporters , Animals , Bacterial Proteins , Baroreflex/drug effects , Biomarkers/metabolism , Blood Pressure/drug effects , Female , Heart Rate/drug effects , Oxidative Stress/drug effects , Pregnancy , Random Allocation , Rats , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/toxicity
3.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article En | MEDLINE | ID: mdl-34201546

Tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) are two cytokines involved in the perpetuation of the chronic inflammation state characterizing rheumatoid arthritis (RA). Significant advances in the treatment of this pathology have been made over the past ten years, partially through the development of anti-TNF and anti-IL-1 therapies. However, major side effects still persist and new alternative therapies should be considered. The formulation of the micro-immunotherapy medicine (MIM) 2LARTH® uses ultra-low doses (ULD) of TNF-α, IL-1ß, and IL-2, in association with other immune factors, to gently restore the body's homeostasis. The first part of this review aims at delineating the pivotal roles played by IL-1ß and TNF-α in RA physiopathology, leading to the development of anti-TNF and anti-IL-1 therapeutic agents. In a second part, an emphasis will be made on explaining the rationale of using multiple therapeutic targets, including both IL-1ß and TNF-α in 2LARTH® medicine. Particular attention will be paid to the ULD of those two main pro-inflammatory factors in order to counteract their overexpression through the lens of their molecular implication in RA pathogenesis.


Arthritis, Rheumatoid/drug therapy , Cytokines/administration & dosage , Immunotherapy/methods , Interleukin-1beta/administration & dosage , Tumor Necrosis Factor-alpha/administration & dosage , Administration, Oral , Animals , Arthritis, Rheumatoid/physiopathology , Dose-Response Relationship, Drug , Humans , Interleukin-1beta/adverse effects , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/physiology , Interleukin-2/administration & dosage , Interleukin-2/adverse effects , Molecular Targeted Therapy/methods , Precision Medicine , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/physiology
4.
Pediatr Infect Dis J ; 40(10): 912-916, 2021 10 01.
Article En | MEDLINE | ID: mdl-34260494

In this retrospective cohort study, the response to routinely administered Haemophilus influenzae type B vaccine, pneumococcal and pertussis vaccinations in 27 children exposed to antitumor necrosis factor alpha (anti-TNFα) during pregnancy was measured. The overall vaccination response seems comparable for children exposed to anti-TNFα and healthy infants. After primary vaccination series, inadequate response was present in some patients and might be related to exposure to anti-TNFα.


Antibodies, Bacterial/blood , Diphtheria-Tetanus-Pertussis Vaccine/immunology , Haemophilus Vaccines/immunology , Immune System/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Diphtheria-Tetanus-Pertussis Vaccine/administration & dosage , Female , Haemophilus Vaccines/administration & dosage , Humans , Immune System/immunology , Immunoglobulin G/blood , Infant , Infant, Newborn , Lymphocyte Subsets/drug effects , Pregnancy , Retrospective Studies , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/blood , Vaccination
5.
Int J Mol Sci ; 22(14)2021 Jul 19.
Article En | MEDLINE | ID: mdl-34299302

Short-chain fatty acid (SCFA) acetate, a byproduct of dietary fiber metabolism by gut bacteria, has multiple immunomodulatory functions. The anti-inflammatory role of acetate is well documented; however, its effect on monocyte chemoattractant protein-1 (MCP-1) production is unknown. Similarly, the comparative effect of SCFA on MCP-1 expression in monocytes and macrophages remains unclear. We investigated whether acetate modulates TNFα-mediated MCP-1/CCL2 production in monocytes/macrophages and, if so, by which mechanism(s). Monocytic cells were exposed to acetate with/without TNFα for 24 h, and MCP-1 expression was measured. Monocytes treated with acetate in combination with TNFα resulted in significantly greater MCP-1 production compared to TNFα treatment alone, indicating a synergistic effect. On the contrary, treatment with acetate in combination with TNFα suppressed MCP-1 production in macrophages. The synergistic upregulation of MCP-1 was mediated through the activation of long-chain fatty acyl-CoA synthetase 1 (ACSL1). However, the inhibition of other bioactive lipid enzymes [carnitine palmitoyltransferase I (CPT I) or serine palmitoyltransferase (SPT)] did not affect this synergy. Moreover, MCP-1 expression was significantly reduced by the inhibition of p38 MAPK, ERK1/2, and NF-κB signaling. The inhibition of ACSL1 attenuated the acetate/TNFα-mediated phosphorylation of p38 MAPK, ERK1/2, and NF-κB. Increased NF-κB/AP-1 activity, resulting from acetate/TNFα co-stimulation, was decreased by ACSL1 inhibition. In conclusion, this study demonstrates the proinflammatory effects of acetate on TNF-α-mediated MCP-1 production via the ACSL1/MAPK/NF-κB axis in monocytic cells, while a paradoxical effect was observed in THP-1-derived macrophages.


Acetates/pharmacology , Chemokine CCL2/biosynthesis , Fatty Acids, Volatile/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Acetates/administration & dosage , Chemokine CCL2/genetics , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/metabolism , Drug Synergism , Enzyme Inhibitors/pharmacology , Fatty Acids, Volatile/administration & dosage , Humans , MAP Kinase Signaling System , Models, Biological , Monocytes/immunology , NF-kappa B/metabolism , Obesity/etiology , Obesity/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , THP-1 Cells , Triazenes/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology
6.
Immunopharmacol Immunotoxicol ; 43(5): 554-561, 2021 Oct.
Article En | MEDLINE | ID: mdl-34314307

BACKGROUND: Indoleamine 2,3-dioxygenase (IDO) inhibition has received much attention in cancer immunotherapy due to its role in immune escape in cancer cells. Additionally, changes in the pro-inflammatory cytokine levels can affect tumor growth and metastasis as well as the effectiveness of immunotherapy. The purpose of this study was for the first time to determine the effects of indoximod as an IDO inhibitor on triple-negative breast cancer (TNBC) and to assess the link between the efficacy of indoximod and IFN-γ or TNF-α stimulation. METHODS: The cytotoxic and apoptotic effects of indoximod alone or IFN-γ or TNF-α induction to mimic an inflammatory environment were evaluated by WST-1, Annexin V, cell cycle analysis, and acridine orange (AO)/ethidium bromide (EtBr) staining. Furthermore, the expression levels of IDO1 and PD-L1 expression were analyzed by RT-PCR. RESULTS: Our results demonstrated that indoximod significantly decreased the TNBC cell viability through apoptotic cell death (p < .05). The combination of indoximod and TNF-α was more effective than indoximod and IFN-γ stimulation or indoximod alone in TNBC cells. Additionally, PD-L1 expression level was significantly up-regulated after treatment with indoximod and TNF-α or IFN-γ combinations (p < .05). CONCLUSIONS: Indoximod exhibited a therapeutic potential in TNBC cells and pro-inflammatory cytokines could affect the effectiveness of indoximod. However, further studies are required to identify the role of the IDO-associated signaling pathways, the molecular mechanisms of indoximod induced apoptotic cell death, and the relationship between IDO inhibition by IDO inhibitors and pro-inflammatory cytokine levels.


Cytokines/administration & dosage , Inflammation Mediators/administration & dosage , Triple Negative Breast Neoplasms/metabolism , Tryptophan/analogs & derivatives , B7-H1 Antigen/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Interferon-gamma/administration & dosage , Treatment Outcome , Triple Negative Breast Neoplasms/drug therapy , Tryptophan/administration & dosage , Tumor Necrosis Factor-alpha/administration & dosage
7.
Allergol. immunopatol ; 49(4): 149-154, jul. 2021. tab
Article En | IBECS | ID: ibc-214295

Introduction: Juvenile idiopathic arthritis (JIA) is the most common rheumatological disease of childhood. The therapy with tumor necrosis factor (TNF) inhibitors (TNFi) in JIA patients has demonstrated efficacy and safety. The most reported adverse event is the high susceptibility to infections. Preventive vaccination helps to decrease these risks. The information on response to vaccines in JIA patients having treatment with anti-TNF is limited. Objectives: To evaluate the response to pneumococcal vaccine in JIA patients undergoing treatment with mAb. Materials and methods Analytical observational mixed cohort study. Data obtained from the clinical records of an immunorheumatology polyclinic of a metropolitan hospital in Santiago (Chile). Treatments, pneumococcal vaccine schedules, immunological laboratory, and measurement of specific antibodies against 10 pneumococcal serotypes were recorded. Results: Nineteen patients were included; average age was 13.8 years; and average evolution time of the disease was 46.2 months. Adalimumab (Humira®) was indicated in case of 13 patients (68.4%) and etanercept (Enbrel®) to 6 (31.5%). The most indicated scheme was a dose of 13-valent pneumococcal conjugate vaccine (PCV13) followed at 8 weeks by a dose of pneumococcal polysaccharide vaccine (PPSV23) in nine (47.3%) patients. Seventeen (89.4%) patients were on immunosuppressive treatment at the time of vaccination. Only one patient did not meet the criteria for response to vaccine. Conclusions: The pneumococcal vaccine induces protective levels of serum antibodies in JIA patients undergoing TNFi treatment. The vaccination schedule and the lymphocyte count could influence the response capacity (AU)


Humans , Male , Female , Adolescent , Arthritis, Juvenile/drug therapy , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Cohort Studies
8.
Brain Res Bull ; 174: 53-62, 2021 09.
Article En | MEDLINE | ID: mdl-34090933

Surgical intervention is necessary following nerve trauma. Tubular prostheses can guide growing axons and inserting substances within these prostheses can be positive for the regeneration, making it an alternative for the current standard tools for nerve repair. Our aim was to investigate the effects of fibrin glue BthTL when combined with a synthetic TNF mimetic-action peptide on nerve regeneration. Male Wistar rats suffered left sciatic nerve transection. For repairing, we used empty silicon tubes (n = 10), tubes filled with fibrin glue BthTL (Tube + Glue group, n = 10) or tubes filled with fibrin glue BThTL mixed with TNF mimetic peptide (Tube + Glue + Pep group, n = 10). Animals were euthanized after 45 days. We collected nerves to perform immunostaining (neurofilament, GAP43, S100-ß, NGFRp75 and Iba-1), light and transmission electron microscopy (for counting myelinated, unmyelinated and degenerated fibers; and for the evaluation of morphometric aspects of regenerated fibers) and collagen staining. All procedures were approved by local ethics committee (protocol 063/17). Tube + Glue + Pep group showed intense inflammatory infiltrate, higher Iba-1 expression, increased immunostaining for NGFRp75 receptor (which characterizes Schwann cell regenerative phenotype), higher myelin thickness and fiber diameter and more type III collagen deposition. Tube + Glue group showed intermediate results between empty tube and Tube + Glue + Pep groups for anti-NGFRp75 immunostaining, inflammation and collagen; on fiber counts, this group showed more degenerate fibers and fewer unmyelinated axons than others. Empty tube group showed superiority only in GAP43 immunostaining. A combination of BthTL glue and TNF mimetic peptide induced greater axonal regrowth and remyelination.


Fibrin Tissue Adhesive , Nerve Regeneration/drug effects , Peptidomimetics/administration & dosage , Peptidomimetics/pharmacology , Peripheral Nerves/drug effects , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Animals , Axons/drug effects , Collagen/metabolism , Immunohistochemistry , Male , Myelin Sheath/drug effects , Nerve Fibers, Myelinated/drug effects , Nerve Tissue Proteins/metabolism , Peptidomimetics/chemistry , Rats , Rats, Wistar , Schwann Cells/drug effects , Schwann Cells/ultrastructure , Sciatic Nerve/injuries , Tumor Necrosis Factor-alpha/chemistry
9.
J Clin Invest ; 131(16)2021 08 16.
Article En | MEDLINE | ID: mdl-34166230

Without cystic fibrosis transmembrane conductance regulator-mediated (CFTR-mediated) HCO3- secretion, airway epithelia of newborns with cystic fibrosis (CF) produce an abnormally acidic airway surface liquid (ASL), and the decreased pH impairs respiratory host defenses. However, within a few months of birth, ASL pH increases to match that in non-CF airways. Although the physiological basis for the increase is unknown, this time course matches the development of inflammation in CF airways. To learn whether inflammation alters CF ASL pH, we treated CF epithelia with TNF-α and IL-17 (TNF-α+IL-17), 2 inflammatory cytokines that are elevated in CF airways. TNF-α+IL-17 markedly increased ASL pH by upregulating pendrin, an apical Cl-/HCO3- exchanger. Moreover, when CF epithelia were exposed to TNF-α+IL-17, clinically approved CFTR modulators further alkalinized ASL pH. As predicted by these results, in vivo data revealed a positive correlation between airway inflammation and CFTR modulator-induced improvement in lung function. These findings suggest that inflammation is a key regulator of HCO3- secretion in CF airways. Thus, they explain earlier observations that ASL pH increases after birth and indicate that, for similar levels of inflammation, the pH of CF ASL is abnormally acidic. These results also suggest that a non-cell-autonomous mechanism, airway inflammation, is an important determinant of the response to CFTR modulators.


Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Interleukin-17/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Tumor Necrosis Factor-alpha/metabolism , Aminophenols/administration & dosage , Benzodioxoles/administration & dosage , Bicarbonates/metabolism , Cells, Cultured , Cystic Fibrosis/drug therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Drug Combinations , Humans , Hydrogen-Ion Concentration , Indoles/administration & dosage , Infant , Infant, Newborn , Interleukin-17/administration & dosage , Ion Transport , Mutation , Pyrazoles/administration & dosage , Pyridines/administration & dosage , Quinolines/administration & dosage , Respiratory Mucosa/drug effects , Sulfate Transporters/genetics , Sulfate Transporters/metabolism , Tumor Necrosis Factor-alpha/administration & dosage
10.
Eur J Pharmacol ; 904: 174152, 2021 Aug 05.
Article En | MEDLINE | ID: mdl-33964292

Antigen-binding fragments (Fabs) are preferred alternatives to antibodies for medical application, whereas their short half-lives limit therapeutic effectiveness. Albumin binding domain (ABD) with high affinity for albumin possesses a great potential in enhancing in vivo performance of biotherapeutics. In this study, to mitigate the poor pharmacokinetics of adalimumab Fab targeting tumor necrosis factor-α (TNFα), an ABD fusion strategy was applied innovatively using GA3, ABD035, ABD094 and ABDCon with high affinities for albumin. The prokaryotic expression, bioactivities and half-lives of those novel Fab-ABD fusions were investigated in vitro and in vivo. All Fab-ABD fusions were successfully purified, and they retained similar TNFα-binding activities with the unmodified Fab control, also presented high affinities for human/mouse serum albumin (HSA/MSA). Additionally, the simultaneous binding of the difunctional Fab-ABD fusions to TNFα and albumin was verified, and ABD fused to Fab neither interfered with Fab-TNFα binding nor impaired the association between Fc fragment of IgG receptor and transporter (FcRn) and albumin. Based on the highest binding affinity for HSA and maximal yield, Fab-ABDCon was selected for further evaluation. Fab-ABDCon showed similar thermostability with the Fab control and robust stability in human and mouse plasma. Most notably, the pharmacokinetics of Fab-ABDCon in mice was significantly improved with a 22-fold longer plasma half-life (28.2 h) compared with that of Fab control (1.31 h), which have contributed to its satisfactory therapeutic efficacy in murine TNFα-induced hepatonecrosis model. Thus, Fab-ABDCon could be a promising long-acting candidate suitable for drug development targeting TNFα-mediated inflammatory disease.


Adalimumab/biosynthesis , Adalimumab/pharmacology , Albumins/metabolism , Anti-Inflammatory Agents/pharmacology , Immunoglobulin Fab Fragments/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Albumins/immunology , Animals , Anti-Inflammatory Agents/immunology , Cell Death/drug effects , Cell Line, Tumor , Chemical and Drug Induced Liver Injury/prevention & control , Drug Design , Female , Galactosamine/administration & dosage , Galactosamine/toxicity , Half-Life , Humans , Hydrogen-Ion Concentration , Immunoglobulin Fab Fragments/pharmacology , Injections, Intraperitoneal , Mice, Inbred BALB C , Necrosis/chemically induced , Necrosis/prevention & control , Protein Binding/genetics , Protein Domains/genetics , Receptors, Fc/immunology , Receptors, Fc/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Serum Albumin, Human/immunology , Serum Albumin, Human/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/toxicity
11.
Toxicology ; 457: 152806, 2021 06 15.
Article En | MEDLINE | ID: mdl-33961948

Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer death. Benzo[a]pyrene (BaP) and 2-amino-1-methyl-6-phenylimidazol [4,5-b] pyridine (PhIP) present in cooked meat are pro-carcinogens and considered to be potential risk factors for CRC. Their carcinogenic and mutagenic effects require metabolic activation primarily by cytochrome P450 1 family enzymes (CYPs); the expression of these enzymes can be modulated by aryl hydrocarbon receptor (AhR) activation and the tumour microenvironment, involving mediators of inflammation. In this study, we hypothesized that tumour necrosis factor-α (TNF-α), a key mediator of inflammation, modulates BaP- and PhIP-induced DNA damage in colon cancer epithelial cells. Importantly, we observed that TNF-α alone (0.1-100 pg/ml) induced DNA damage (micronuclei formation) in HCT-116 cells and co-treatment of TNF-α with BaP or PhIP showed higher levels of DNA damage compared to the individual single treatments. TNF-α alone or in combination with BaP or PhIP did not affect the expression levels of CYP1A1 and CYP1B1 (target genes of AhR signaling pathways). The DNA damage induced by TNF-α was elevated in p53 null HTC-116 cells compared to wild type cells, suggesting that TNF-α-induced DNA damage is suppressed by functional p53. In contrast, p53 status failed to affect BaP and PhIP induced micronucleus frequency. Furthermore, JNK and NF-κB signaling pathway were activated by TNF-α treatment but only inhibition of JNK significantly reduced TNF-α-induced DNA damage. Collectively, these findings suggest that TNF-α induced DNA damage involves JNK signaling pathway rather than AhR and NF-κB pathways in colon cancer epithelial cells.


Carcinogens/toxicity , Colorectal Neoplasms/metabolism , DNA Damage/drug effects , Epithelial Cells/drug effects , MAP Kinase Signaling System/drug effects , Tumor Necrosis Factor-alpha/toxicity , Carcinogens/administration & dosage , Cell Survival/drug effects , Cell Survival/physiology , Colorectal Neoplasms/pathology , DNA Damage/physiology , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , HCT116 Cells , Humans , MAP Kinase Signaling System/physiology , Tumor Necrosis Factor-alpha/administration & dosage
12.
Drug Des Devel Ther ; 15: 1981-1992, 2021.
Article En | MEDLINE | ID: mdl-34007158

BACKGROUND: Fingolimod (FTY720), a novel immunomodulator, was found to suppress the severity of collagen-induced arthritis (CIA) in mice. However, the potential molecular mechanisms are still unknown, and the effect of FTY720 on the recruitment of immune cells in the affected joints in the CIA model is not clear. MATERIALS AND METHODS: Following the oral administration of FTY720 (2 mg/kg) was treated into CIA mice per day for 35 days, intravital microscopy and immunofluorescence assays were performed to examine immune cell recruitment in the affected joints. Human MH7A synoviocytes were stimulated with tumour necrosis factor (TNF)-α and incubated with FTY720. Interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and interleukin-8 (IL-8) mRNA and protein expression were evaluated using RT-PCR and enzyme-linked immunosorbent assay, respectively. Signal transduction pathway protein expression was measured by Western blotting. Nuclear translocation of nuclear factor (NF)-κB was also analyzed by fluorescence microscopy. RESULTS: In vivo experiments showed that FTY720 inhibited the recruitment of CD4+ lymphocytes in the affected joints of CIA mice. FTY720 reduced the secretion of IL-1ß, IL-6, and IL-8 from TNF-α-stimulated MH7A cells in a dose-dependent manner. FTY720 also inhibited TNF-α-induced phosphorylation of NF-κBp65 and IκBα, as well as NF-κBp65 nuclear translocation, in a dose- and time-dependent manner. Interestingly, FTY720 blocked PI3K/Akt, the upstream targets of the NF-κB pathway. CONCLUSION: Our findings demonstrated that oral administration of FTY720 exerted beneficial effects in CIA mice by inhibiting CD4+ T lymphocyte recruitment to the affected joints. Our data also indicated that FTY720 inhibited TNF-α-induced inflammation by suppressing the AKT/PI3K/NF-κB pathway in MH7A cells.


Arthritis, Experimental/prevention & control , CD4-Positive T-Lymphocytes/immunology , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/pharmacology , Animals , Arthritis, Experimental/pathology , Dose-Response Relationship, Drug , Fingolimod Hydrochloride/administration & dosage , Humans , Immunosuppressive Agents/administration & dosage , Inflammation/drug therapy , Inflammation/pathology , Male , Mice , Mice, Inbred DBA , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Synoviocytes/drug effects , Synoviocytes/metabolism , Time Factors , Tumor Necrosis Factor-alpha/administration & dosage
13.
Pharm Biol ; 59(1): 401-409, 2021 Dec.
Article En | MEDLINE | ID: mdl-33794116

CONTEXT: Pomelo peel oil (PPO) [Citrus maxima (Burm.) Merr. (Rutaceae)] is reported to possess antioxidant and antimelanogenic activities. OBJECTIVE: To investigate the effect of PPO [Citrus maxima (Burm.) Merr. cv. Shatian Yu] on tumour necrosis factor-α (TNF-α)-induced necroptosis in cerebral ischaemia-reperfusion injury (CIRI) after cardiac arrest (CA). MATERIALS AND METHODS: Male Sprague Dawley rats were randomly assigned to six groups: sham group, PP0-L (10 mg/kg), PPO-M (20 mg/kg), PPO-H (40 mg/kg) and two control groups (CA, 0.9% saline; Gly, 10% glycerol). All drugs were administered intravenously to the CA/CPR rats within 10 min after return of spontaneous circulation (ROSC). After 24 h, rats were assessed for neuronal injury via the neurological deficit score (NDS), cerebral cortex staining and transmission electron microscopy (TEM) and expression levels of TNF-α and necroptosis-related proteins by immunoreactivity staining and western blotting. RESULTS: Compared to those in the sham group (survival rate, 100% and NDS, 80), the survival rate and NDS were significantly reduced in the model groups (CA, 56.25%, 70; Gly, 62.5%, 71; PPO-L, 75%, 72; PPO-M, 87.5%, 75; PPO-H, 81.25%, 74). In the PPO-M group, Nissl bodies were significantly increased (43.67 ± 1.906 vs. 17 ± 1.732), the incidence of pathomorphological injury was lower and the necroptosis markers (TNF-α, RIPK1, RIPK3, p-MLKL/MLKL) expression was downregulated compared to those in the CA group (p < 0.05). DISCUSSION AND CONCLUSIONS: The neuroprotective effects of PPO in the CA rats suggested that PPO possibility as a health product enhances the resistance ability against brain injury for humans.


Citrus/chemistry , Heart Arrest/drug therapy , Plant Oils/pharmacology , Reperfusion Injury/drug therapy , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Heart Arrest/physiopathology , Male , Necroptosis/drug effects , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/pharmacology , Oils, Volatile/administration & dosage , Oils, Volatile/isolation & purification , Oils, Volatile/pharmacology , Plant Oils/administration & dosage , Plant Oils/isolation & purification , Random Allocation , Rats , Rats, Sprague-Dawley , Reperfusion Injury/physiopathology , Survival Rate , Tumor Necrosis Factor-alpha/administration & dosage
14.
J Vis Exp ; (168)2021 02 14.
Article En | MEDLINE | ID: mdl-33645590

Symptomatic intervertebral disc (IVD) degeneration (IDD) is a major socioeconomic burden and is characterized by inflammation and tissue degradation. Due to the lack of causative therapies, there is an urgent need for innovative experimental organ culture models to study the mechanisms involved in the progression of the disease, find therapeutic targets, and reduce the need for animal models. We here present a novel, three-dimensional organ culture model protocol mimicking the proinflammatory and catabolic microenvironment, which is present during IDD. Initially, bovine caudal IVDs were dissected, cleaned, and cultured in the tissue culture medium. Dynamic physiologic or pathologic loading was applied in a custom-made bioreactor for 2 hours per day. IVDs were assigned to a control group (high glucose medium, physiological loading, phosphate-buffered saline injection) and a pathological group (low glucose medium, pathological loading, tumor necrosis factor-alpha injection) for four days. Gene expression analysis from collected nucleus pulposus cells of the IVDs and enzyme-linked immunosorbent assay of the conditioned organ culture media was performed. Our data revealed a higher expression of inflammatory markers and reduced disc heights after loading in the pathological group compared to the control group. This protocol is reliable to simulate IVD inflammation and degeneration and can be further expanded to broaden its application scope.


Inflammation/pathology , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Displacement/pathology , Models, Biological , Organ Culture Techniques/methods , Animals , Annulus Fibrosus/pathology , Cattle , Culture Media, Conditioned/metabolism , Gene Expression Regulation , Injections , Interleukin-8/metabolism , Nucleus Pulposus/pathology , Tumor Necrosis Factor-alpha/administration & dosage
15.
Eur J Med Chem ; 217: 113327, 2021 May 05.
Article En | MEDLINE | ID: mdl-33730678

Necroptosis is a form of regulated necrotic cell death that is independent of caspases. Receptor-interacting protein kinase 3 (RIPK3) has been identified as a key regulator for necroptosis, and has been proposed as a potential therapeutic target for the treatment of diseases associated with necroptosis. In this report, we describe the design, synthesis, and evaluation of a series of novel RIPK3 inhibitors. The lead compound 38 exhibited potent activity (EC50 = 0.42 µM) in blocking TNFα, Smac mimetic and z-VAD (TSZ) induced cell death in HT-29 cells. Mechanistic studies showed that compound 38 bound to RIPK3 with high affinity (Kd = 7.1 nM), and inhibited RIPK3 kinase activity in a ADP-Glo functional assay. In addition, compound 38 displayed good selectivity over another necroptosis regulator RIPK1 (Kd = 6000 nM). Furthermore, compound 38 demonstrated excellent in vitro safety profiles with minimal inhibition of CYP isozymes and hERG potassium channel. Lastly, compound 38 efficiently blocked hypothermia and death in mice in the TNFα-induced systemic inflammatory response syndrome model.


Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hypothermia/drug therapy , Hypothermia/metabolism , Injections, Intravenous , Mice , Mice, Inbred C57BL , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology
16.
J Neurotrauma ; 38(15): 2186-2191, 2021 08 01.
Article En | MEDLINE | ID: mdl-33397170

After a severe, high-level spinal cord injury (SCI), plasticity to intraspinal circuits below injury results in heightened spinal sympathetic reflex activity and detrimentally impacts peripheral organ systems. Such sympathetic hyperreflexia is immediately apparent as an episode of autonomic dysreflexia (AD), a life-threatening condition characterized by sudden hypertension and reflexive bradycardia following below-level sensory inputs; for example, pressure sores or impacted fecal matter. Over time, plasticity within the spinal sympathetic reflex (SSR) circuit contributes to the progressive intensification of AD events, as the frequency and severity of AD events increase greatly beginning ∼2 weeks post-injury (wpi). The neuroimmune system has been implicated in driving sympathetic hyperreflexia, as inhibition of the cytokine soluble tumor necrosis factor-alpha (sTNFα) using the biological mimetic XPro1595 beginning within days post-SCI has been shown to attenuate the development of AD. Here, we sought to further understand the effective therapeutic time window of XPro1595 to diminish sympathetic hyperreflexia, as indicated by AD. We delayed the commencement of continuous intrathecal administration of XPro1595 until 2 weeks after a complete, thoracic level 3 injury in adult rats. We examined the severity of colorectal distension-induced AD biweekly. We found that initiation of sTNFα inhibition at 2 wpi does not attenuate the severity or intensification of sympathetic hyperreflexia compared with saline-treated controls. Coupled with previous data from our group, these findings suggest that central sTNFα signaling must be targeted prior to 2 weeks post-SCI in order to decrease sympathetic hyperreflexia.


Autonomic Dysreflexia/prevention & control , Spinal Cord Injuries/complications , Spinal Cord Injuries/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Autonomic Dysreflexia/etiology , Disease Models, Animal , Drug Administration Schedule , Female , Injections, Spinal , Rats , Rats, Wistar , Thoracic Vertebrae , Time Factors , Tumor Necrosis Factor-alpha/administration & dosage
17.
Dig Liver Dis ; 53(9): 1112-1117, 2021 Sep.
Article En | MEDLINE | ID: mdl-33376073

BACKGROUND: Anti-tumour necrosis factor (TNF)-α drugs are used by increasing numbers of reproductive-age women. Although the neonatal outcomes have been described, there are concerns regarding the risk of infection in offspring following exposure to anti-TNF-α. METHODS: A literature search was conducted using Pubmed, EMBASE, and the Cochrane Database, from inception through August 2020. We evaluated the risk of infection in autoimmune disease (AID) offspring unexposed to anti-TNF-α compared to AID offspring exposed to anti-TNF-α, as well as to unexposed non-AID offspring. RESULTS: Our primary analysis showed that both AID offspring unexposed to anti-TNF-α [risk ratio (RR) 1.09; 95% confidence interval (CI), 1.03-1.16; I2=0%] and AID offspring exposed to anti-TNF-α (RR 1.39; 95% CI, 1.2-1.61; I2=0%] was associated with an increased risk of infection during the first year of life compared with the unexposed non-AID offspring. However, our secondary analysis demonstrated that AID offspring exposed to anti-TNF-α was not associated with an increased risk of infection when compared with AID offspring unexposed to anti-TNF-α (RR=1.1; 95% CI, 0.86-1.4). CONCLUSION: Our results suggest that in utero exposure to anti-TNF-α does not appear to increase the risk of infection during the first year of life in the offspring; however, AID itself was associated with a marked excess risk of infection in the children.


Autoimmune Diseases/drug therapy , Pregnancy Complications/drug therapy , Tumor Necrosis Factor-alpha/adverse effects , Female , Humans , Infant , Infant, Newborn , Pregnancy , Prenatal Exposure Delayed Effects , Risk Assessment , Tumor Necrosis Factor-alpha/administration & dosage
18.
Surg Today ; 51(1): 32-43, 2021 Jan.
Article En | MEDLINE | ID: mdl-32277281

Tumor necrosis factor-alpha inhibitor (TNFi) treatment is effective for ulcerative colitis (UC) and Crohn's disease (CD). Although several meta-analyses have been performed to evaluate the association between TNFi treatment and surgical morbidity, the results are controversial. We conducted a systematic review and meta-analysis of the prevention of surgical site infection (SSI) after surgery for UC and CD in patients on TNFis, based on literature published between January 2000 and May 2019 (registered on PROSPERO, No. CRD42019134156). Overall, 2175 UC patients in 13 observational studies (OBSs) and 7084 CD patients in 16 OBSs were included. The incidences of incisional (INC) SSI and organ/space (O/S) SSI after surgery for UC were 179/1985 (9.0%) and 176/2175 (8.1%), respectively. TNFi use was not associated with the incidences of INC SSI (odds ratio (OR) 1.04, 95% confidence interval (CI) (0.47-2.32) or O/S SSI (OR 1.85, 95% CI (0.82-4.20)) after surgery for UC. The INC SSI and O/S SSI incidences after surgery for CD were 289/3089 (9.4%) and 526/7,084 (7.4%), respectively. Preoperative TNFi use was not associated with INC SSI (OR 0.98, 95% CI (0.52-1.83)) or O/S SSI incidence (OR 1.09, 95% CI (0.78-1.52)) after surgery for CD. We did not find a significant association between preoperative TNFi use and SSI in surgery for UC or CD.


Colitis, Ulcerative/surgery , Crohn Disease/surgery , Digestive System Surgical Procedures/adverse effects , Surgical Wound Infection/epidemiology , Surgical Wound Infection/etiology , Tumor Necrosis Factor-alpha/therapeutic use , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Digestive System Surgical Procedures/methods , Humans , Incidence , Preoperative Care , Tumor Necrosis Factor-alpha/administration & dosage
19.
Front Immunol ; 12: 788638, 2021.
Article En | MEDLINE | ID: mdl-34975882

Intestinal inflammation is a major threat to the health and growth of young animals such as piglets. As a next-generation probiotics, limited studies have shown that Akkermansia muciniphila could alleviate inflammation of intestinal epithelial cells (IECs). In this study, a TNF-α-induced inflammatory model of IPEC-J2 cells, the intestinal porcine enterocytes, was built to evaluate the effects of active or inactive A. muciniphila on the inflammation of IECs. The viability of IPEC-J2 cells was the highest when treated with active (108 copies/mL) or inactive (109 copies/mL) A. muciniphila for 7.5 h (P < 0.01). Treated with 20 ng/mL of TNF-α and followed by a treatment of A. muciniphila, the mRNA level of proinflammatory cytokines (IL-8, IL-1ß, IL-6 and TNF-α) was remarkably reduced (P < 0.05) along with the increased mRNA level of tight junction proteins (ZO-1 and Occludin, P < 0.05). Flow cytometry analysis showed that active or inactive A. muciniphila significantly suppressed the rate of the early and total apoptotic of the inflammatory IPEC-J2 cells (P < 0.05). According to results of transcriptome sequencing, active and inactive A. muciniphila may decline cell apoptosis by down-regulating the expression of key genes in calcium signaling pathway, or up-regulating the expression of key genes in cell cycle signaling pathway. And the bacterium may alleviate the inflammation of IECs by down-regulating the expression of PI3K upstream receptor genes. Our results indicate that A. muciniphila may be a promising NGP targeting intestinal inflammation.


Inflammation/diet therapy , Intestinal Mucosa/immunology , Probiotics/administration & dosage , Akkermansia/immunology , Animals , Calcium Signaling/immunology , Cell Line , Cell Survival/immunology , Disease Models, Animal , Down-Regulation/immunology , Epithelial Cells , Humans , Inflammation/immunology , Intestinal Mucosa/cytology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/immunology , Swine , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/immunology
20.
Hypertension ; 76(6): 1744-1752, 2020 12.
Article En | MEDLINE | ID: mdl-33131307

We showed that intrarenal suppression of TNF (tumor necrosis factor) production under low salt (LS) conditions increases renal cortical AGT (angiotensinogen) mRNA and protein expression. Intrarenal injection of murine recombinant TNF attenuated increases of AGT in mice ingesting LS. Moreover, AGT mRNA and protein expression increased ≈6-fold and 2-fold, respectively, in mice ingesting LS that also received an intrarenal injection of a lentivirus construct that specifically silenced TNF in the kidney (U6-TNF-ex4). Silencing of TNF under normal salt and high salt (HS) conditions also resulted in increased AGT expression. Since renal TNF production decreases in response to LS and increases in response to HS, the data suggest that alterations in TNF production under these conditions modulate the degree of AGT expression. We also tested the hypothesis that TNF inhibits intrarenal AGT expression by a mechanism involving miR-133a. Expression of miR-133a decreased in mice given LS and increased in response to HS for 7 days. Intrarenal silencing of TNF reversed the effects of HS on miR-133a-dependent AGT expression. In contrast, intrarenal TNF administration increased miR-133a expression in the kidney. Collectively, the data suggest that miR-133a is a salt-sensitive microRNA that inhibits AGT in the kidney and is increased by TNF. The HS-induced increase in blood pressure observed following silencing of TNF was markedly reduced upon intrarenal administration of miR-133a suggesting that intrinsic effects of TNF in the kidney to limit the blood pressure response to HS include an increase in miR-133a, which suppresses AGT expression.


Angiotensinogen/genetics , Gene Expression Regulation/genetics , Kidney Tubules, Proximal/metabolism , MicroRNAs/genetics , Tumor Necrosis Factor-alpha/genetics , Angiotensinogen/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Gene Expression Regulation/drug effects , Kidney/drug effects , Kidney/metabolism , Kidney Tubules, Proximal/drug effects , Male , Mice, Inbred C57BL , RNA Interference , Sodium Chloride, Dietary/administration & dosage , Tumor Necrosis Factor-alpha/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
...