Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.201
Filter
1.
Int J Biol Sci ; 19(5): 1490-1508, 2023.
Article in English | MEDLINE | ID: mdl-37056922

ABSTRACT

IFNγ has long been recognised as a key mediator of tumour immunity and angiostasis. However, IFNγ modulation for cancer therapy is still unsuccessful due to its complex effects on various host cells. In this study, we found that treatment of Lewis lung carcinoma transplants with cisplatin often caused IFNγ-dependent tumour vascular damage. IFNγ induced endothelial glycolysis and lactate production, leading to enhanced endocytosis of vascular endothelial (VE)-cadherin and vessel leakage. We have also developed anti-IFNγ nanoparticles coated with a clot-binding peptide CREKA (CREKA-lipo-anti-IFNγ), which targets the fibrin-fibronectin complex that appears in the leaky site of damaged tumour blood vessels. Blocking IFNγ activity in the leakage site of capillaries using nanoparticles rescued VE-cadherin distribution on the endothelial cellular surface, promoted blood vessel integrity, and improved drug delivery. In conclusion, IFNγ blockade in capillary leak site protected tumour blood vessels from lactate-dependent VE-cadherin loss and enhanced drug delivery during chemotherapy, which provides a basis for tissue-specific IFNγ blockade for tumour therapy.


Subject(s)
Lactic Acid , Neoplasms , Humans , Cadherins/metabolism , Capillary Permeability , Endocytosis , Lactic Acid/pharmacology , Interferon-gamma/antagonists & inhibitors
2.
Viruses ; 13(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34960717

ABSTRACT

The evasion of the Interferon response has important implications in Zika virus (ZIKV) disease. Mutations in ZIKV viral protein NS4B, associated with modulation of the interferon (IFN) system, have been linked to increased pathogenicity in animal models. In this study, we unravel ZIKV NS4B as antagonist of the IFN signaling cascade. Firstly, we reported the genomic characterization of NS4B isolated from a strain of the 2016 outbreak, ZIKV Brazil/2016/INMI1, and we predicted its membrane topology. Secondly, we analyzed its phylogenetic correlation with other flaviviruses, finding a high similarity with dengue virus 2 (DEN2) strains; in particular, the highest conservation was found when NS4B was aligned with the IFN inhibitory domain of DEN2 NS4B. Hence, we asked whether ZIKV NS4B was also able to inhibit the IFN signaling cascade, as reported for DEN2 NS4B. Our results showed that ZIKV NS4B was able to strongly inhibit the IFN stimulated response element and the IFN-γ-activated site transcription, blocking IFN-I/-II responses. mRNA expression levels of the IFN stimulated genes ISG15 and OAS1 were also strongly reduced in presence of NS4B. We found that the viral protein was acting by suppressing the STAT1 phosphorylation and consequently blocking the nuclear transport of both STAT1 and STAT2.


Subject(s)
Interferon Type I/metabolism , Interferon-gamma/metabolism , STAT1 Transcription Factor/metabolism , Viral Nonstructural Proteins/metabolism , Zika Virus Infection/virology , Zika Virus/metabolism , 2',5'-Oligoadenylate Synthetase/genetics , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytokines/genetics , HEK293 Cells , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Interferon-beta/biosynthesis , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Phosphorylation , Phylogeny , Protein Conformation , Response Elements , Signal Transduction , Ubiquitins/genetics , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Zika Virus/chemistry , Zika Virus/isolation & purification , Zika Virus/pathogenicity
3.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768860

ABSTRACT

Vitiligo is a chronic autoimmune dermatosis of which the pathogenesis remains scarcely known. A wide variety of clinical studies have been proposed to investigate the immune mediators which have shown the most recurrency. However, such trials have produced controversial results. The aim of this review is to summarize the main factors involved in the pathogenesis of vitiligo, the latest findings regarding the cytokines involved and to evaluate the treatments based on the use of biological drugs in order to stop disease progression and achieve repigmentation. According to the results, the most recurrent studies dealt with inhibitors of IFN-gamma and TNF-alpha. It is possible that, given the great deal of cytokines involved in the lesion formation process of vitiligo, other biologics could be developed in the future to be used as adjuvants and/or to entirely replace the treatments that have proven to be unsatisfactory so far.


Subject(s)
Autoimmune Diseases/pathology , Biological Products/therapeutic use , Cytokines/metabolism , Vitiligo/drug therapy , Vitiligo/pathology , Autoimmune Diseases/drug therapy , Exonucleases/genetics , HLA-DQ beta-Chains/genetics , HLA-DRB1 Chains/genetics , Humans , Interferon-gamma/antagonists & inhibitors , Keratinocytes/metabolism , Melanocytes/pathology , Pigmentation/physiology , Skin/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
4.
Biomed Res Int ; 2021: 1124055, 2021.
Article in English | MEDLINE | ID: mdl-34812407

ABSTRACT

Autoimmune disorder is a chronic immune imbalance which is developed through a series of pathways. The defect in B cells, T cells, and lack of self-tolerance has been greatly associated with the onset of many types of autoimmune complications including rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis, and chronic inflammatory demyelinating polyneuropathy. The SLE is an autoimmune disease with a common type of lupus that causes tissue and organ damage due to the wide spread of inflammation. In the current study, twenty anti-inflammatory peptides derived from plant and animal sources were docked as ligands or peptides counter to proinflammatory cytokines. Interferon gamma (IFN-γ), interleukin 3 (IL-3), and tumor necrosis factor alpha (TNF-α) were targeted in this study as these are involved in the pathogenesis of SLE in many clinical studies. Two docking approaches (i.e., protein-ligand docking and peptide-protein docking) were employed in this study using Molecular Operating Environment (MOE) software and HADDOCK web server, respectively. Amongst docked twenty peptides, the peptide DEDTQAMMPFR with S-score of -11.3018 and HADDOCK score of -10.3 ± 2.5 kcal/mol showed the best binding interactions and energy validation with active amino acids of IFN-γ protein in both docking approaches. Depending upon these results, this peptide could be used as a potential drug candidate to target IFN-γ, IL-3, and TNF-α proteins to control inflammatory events. Other peptides (i.e., QEPQESQQ and FRDEHKK) also revealed good binding affinity with IFN-γ with S-scores of -10.98 and -10.55, respectively. Similarly, the peptides KHDRGDEF, FRDEHKK, and QEPQESQQ showed best binding interactions with IL-3 with S-scores of -8.81, -8.64, and -8.17, respectively.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Interferon-gamma/antagonists & inhibitors , Interleukin-3/antagonists & inhibitors , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Oligopeptides/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Drug Discovery/methods , Humans , Ligands , Molecular Docking Simulation , Molecular Dynamics Simulation , Oligopeptides/chemistry , Oligopeptides/genetics , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/pharmacology
5.
Nat Rev Rheumatol ; 17(11): 678-691, 2021 11.
Article in English | MEDLINE | ID: mdl-34611329

ABSTRACT

Interferon-γ (IFNγ) is a pleiotropic cytokine with multiple effects on the inflammatory response and on innate and adaptive immunity. Overproduction of IFNγ underlies several, potentially fatal, hyperinflammatory or immune-mediated diseases. Several data from animal models and/or from translational research in patients point to a role of IFNγ in hyperinflammatory diseases, such as primary haemophagocytic lymphohistiocytosis, various forms of secondary haemophagocytic lymphohistiocytosis, including macrophage activation syndrome, and cytokine release syndrome, all of which are often managed by rheumatologists or in consultation with rheumatologists. Given the effects of IFNγ on B cells and T follicular helper cells, a role for IFNγ in systemic lupus erythematosus pathogenesis is emerging. To improve our understanding of the role of IFNγ in human disease, IFNγ-related biomarkers that are relevant for the management of hyperinflammatory diseases are progressively being identified and studied, especially because circulating levels of IFNγ do not always reflect its overproduction in tissue. These biomarkers include STAT1 (specifically the phosphorylated form), neopterin and the chemokine CXCL9. IFNγ-neutralizing agents have shown efficacy in the treatment of primary haemophagocytic lymphohistiocytosis in clinical trials and initial promising results have been obtained in various forms of secondary haemophagocytic lymphohistiocytosis, including macrophage activation syndrome. In clinical practice, there is a growing body of evidence supporting the usefulness of circulating CXCL9 levels as a biomarker reflecting IFNγ production.


Subject(s)
Immune System Diseases/immunology , Inflammation/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/blood , Chemokine CXCL9/blood , Chemokine CXCL9/immunology , Crohn Disease/blood , Crohn Disease/drug therapy , Crohn Disease/immunology , Disease Models, Animal , Humans , Immune System Diseases/blood , Immune System Diseases/drug therapy , Immunity/immunology , Inflammation/blood , Inflammation/drug therapy , Interferon-gamma/biosynthesis , Interferon-gamma/blood , Lymphohistiocytosis, Hemophagocytic/blood , Lymphohistiocytosis, Hemophagocytic/drug therapy , Macrophage Activation Syndrome/blood , Macrophage Activation Syndrome/drug therapy , Macrophage Activation Syndrome/immunology , Mice , Neopterin/blood , Neopterin/immunology , STAT1 Transcription Factor/blood , STAT1 Transcription Factor/immunology
6.
Front Immunol ; 12: 754106, 2021.
Article in English | MEDLINE | ID: mdl-34691073

ABSTRACT

Macrophage polarization is critical for liver tissue repair following acute liver injury. However, the underlying mechanisms of macrophage phenotype switching are not well defined. Invariant natural killer T (iNKT) cells orchestrate tissue inflammation and tissue repair by regulating cytokine production. Herein, we examined whether iNKT cells played an important role in liver repair after hepatic ischemia-reperfusion (I/R) injury by affecting macrophage polarization. To this end, we subjected male C57BL/6 mice to hepatic I/R injury, and mice received an intraperitoneal (ip) injection of α-galactosylceramide (α-GalCer) or vehicle. Compared with that of the vehicle, α-GalCer administration resulted in the promotion of liver repair accompanied by acceleration of macrophage differentiation and by increases in the numbers of Ly6Chigh pro-inflammatory macrophages and Ly6Clow reparative macrophages. iNKT cells activated with α-GalCer produced interleukin (IL)-4 and interferon (IFN)-γ. Treatment with anti-IL-4 antibodies delayed liver repair, which was associated with an increased number of Ly6Chigh macrophages and a decreased number of Ly6Clow macrophages. Treatment with anti-IFN-γ antibodies promoted liver repair, associated with reduced the number of Ly6Chigh macrophages, but did not change the number of Ly6Clow macrophages. Bone marrow-derived macrophages up-regulated the expression of genes related to both a pro-inflammatory and a reparative phenotype when co-cultured with activated iNKT cells. Anti-IL-4 antibodies increased the levels of pro-inflammatory macrophage-related genes and decreased those of reparative macrophage-related genes in cultured macrophages, while anti-IFN-γ antibodies reversed the polarization of macrophages. Cd1d-deficient mice showed delayed liver repair and suppressed macrophage switching, compared with that in wild-type mice. These results suggest that the activation of iNKT cells by α-GalCer facilitated liver repair after hepatic I/R injury by both IL-4-and IFN-γ-mediated acceleration of macrophage polarization. Therefore, the activation of iNKT cells may represent a therapeutic tool for liver repair after hepatic I/R injury.


Subject(s)
Galactosylceramides/pharmacology , Liver Regeneration/physiology , Liver/immunology , Macrophage Activation , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Cells, Cultured , Coculture Techniques , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interleukin-4/antagonists & inhibitors , Interleukin-4/biosynthesis , Liver/blood supply , Liver Regeneration/immunology , Lymphocyte Activation/drug effects , Macrophages/classification , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/metabolism , Reperfusion Injury
7.
Sci Rep ; 11(1): 20659, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667205

ABSTRACT

Gut lactobacilli and bifidobacteria on the immune homeostasis. Therefore, to understand the mechanism in vivo, we selected human fecal Lactobacillus rhamnosus NK210 and Bifidobacterium longum NK219, which strongly suppressed the IFN-γ to IL-10 expression (IIE) ratio in lipopolysaccharide-stimulated macrophages. Thereafter, we examined their effects on the endotoxin, antibiotics, or antitumor drug-stimulated immune imbalance in mice. Intraperitoneal injection of lipopolysaccharide and oral gavage of ampicillin increased IFN-γ and TNF-α expression in the spleen, colon, and hippocampus, while IL-10 expression decreased. However, intraperitoneal injection of cyclophosphamide suppressed IFN-γ, TNF-α, and IL-10 expression. LPS exposure induced splenic natural killer cell cytotoxicity against YAC-1 cells (sNK-C) and peritoneal macrophage phagocytosis against Candida albicans (pMA-P) activities, while cyclophosphamide and ampicillin treatments suppressed sNK-C and pMA-P activities. However, LPS, ampicillin, cyclophosphamide all increased IIE and TNF-α to IL-10 expression (TIE) ratios. Oral administration of NK210 and/or NK219 significantly reduced LPS-induced sNK-C, pMA-P, and IFN-γ expression, while cyclophosphamide- or ampicillin-suppressed sNK-C and pMA-P activities, cyclophosphamide-suppressed IFN-γ, TNF-α, and IL-10 expression, and ampicillin-suppressed IL-10 expression increased. Nevertheless, they suppressed LPS-, ampicillin-, or cyclophosphamide-induced IIE and TIE ratios, cognitive impairment, and gut dysbiosis. In particular, NK219, but not NK210, increased the IIE expression ratio in vitro and in vivo, and enhanced sNK-C and pMA-P activities in normal control mice, while cognitive function and gut microbiota composition were not significantly affected. These findings suggest that NK210, Lactobacillus sp, and NK219, Bifidobacterium additively or synergistically alleviate gut dysbiosis, inflammation, and cognitive impairment with immune imbalance by controlling IIE and TIE ratios.


Subject(s)
Bifidobacterium longum/metabolism , Dysbiosis/therapy , Lacticaseibacillus rhamnosus/metabolism , Animals , Bifidobacterium/metabolism , Bifidobacterium longum/pathogenicity , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/therapy , Colitis/microbiology , Colitis/therapy , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Inflammation/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lactobacillus/metabolism , Lacticaseibacillus rhamnosus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Probiotics/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
8.
Angew Chem Int Ed Engl ; 60(48): 25468-25476, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34580976

ABSTRACT

A versatile terpene synthase (LcTPS2) producing unconventional macrocyclic terpenoids was characterized from Leucosceptrum canum. Engineered Escherichia coli and Nicotiana benthamiana expressing LcTPS2 produced six 18-/14-membered sesterterpenoids including five new ones and two 14-membered diterpenoids. These products represent the first macrocyclic sesterterpenoids from plants and the largest sesterterpenoid ring system identified to date. Two variants F516A and F516G producing approximately 3.3- and 2.5-fold, respectively, more sesterterpenoids than the wild-type enzyme were engineered. Both 18- and 14-membered ring sesterterpenoids displayed significant inhibitory activity on the IL-2 and IFN-γ production of T cells probably via inhibition of the MAPK pathway. The findings will contribute to the development of efficient biocatalysts to create bioactive macrocyclic sesterterpenoids, and also herald a new potential in the well-trodden territory of plant terpenoid biosynthesis.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Immunosuppressive Agents/pharmacology , Interferon-gamma/antagonists & inhibitors , Interleukin-2/antagonists & inhibitors , Macrocyclic Compounds/pharmacology , Terpenes/pharmacology , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lamiaceae/chemistry , Lamiaceae/metabolism , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/metabolism , Molecular Structure , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Terpenes/chemistry , Terpenes/metabolism
9.
Front Immunol ; 12: 623087, 2021.
Article in English | MEDLINE | ID: mdl-34262557

ABSTRACT

Background: Seasonal variations have been reported for immune markers. However, the relative contributions of sunlight and vitamin D variability on such seasonal changes are unknown. Objective: This double-blind, randomized, placebo-controlled trial tested whether daily 400 IU vitamin D3 supplementation affected short-term (12 weeks) and long-term (43 weeks) natural regulatory T cell (nTreg) populations in healthy participants. Design: 62 subjects were randomized equally to vitamin D versus placebo in March and assessed at baseline, April (4w), June (12w), September (25w) and January (43w). Circulating nTregs, ex vivo proliferation, IL-10 and IFN-γ productions were measured. Vitamin D metabolites and sunlight exposure were also assessed. Results: Mean serum 25-hydroxyvitamin D (25(OH)D) increased from 35.8(SD 3.0) to 65.3(2.6) nmol/L in April and remained above 75 nmol/L with vitamin D supplementation, whereas it increased from 36.4(3.2) to 49.8(3.5) nmol/L in June to fall back to 39.6(3.5) nmol/L in January with placebo. Immune markers varied similarly between groups according to the season, but independently of 25(OH)D. For nTregs, the mean (%CD3+CD4+CD127lo cells (SEM)) nadir observed in March (2.9(0.1)%) peaked in September at 4.0(0.2)%. Mean T cell proliferation peaked in June (33156(1813) CPM) returning to the nadir in January (17965(978) CPM), while IL-10 peaked in June and reached its nadir in September (median (IQR) of 262(283) to (121(194) pg/ml, respectively). Vitamin D attenuated the seasonal increase in IFN-γ by ~28% with mean ng/ml (SEM) for placebo vs vitamin D, respectively, for April 12.5(1.4) vs 10.0(1.2) (p=0.02); June 13.9(1.3) vs 10.2(1.7) (p=0.02) and January 7.4(1.1) vs 6.0(1.1) (p=0.04). Conclusions: Daily low dose Vitamin D intake did not affect the nTregs population. There were seasonal variation in nTregs, proliferative response and cytokines, suggesting that environmental changes influence immune response, but the mechanism seems independent of vitamin D status. Vitamin D attenuated the seasonal change in T cell-produced IFN-γ, suggesting a decrease in effector response which could be associated with inflammation. Clinical Trial Registration: https://www.isrctn.com, identifier (ISRCTN 73114576).


Subject(s)
Cell Proliferation/drug effects , Cholecalciferol/administration & dosage , Cholecalciferol/immunology , Interferon-gamma/analysis , Seasons , T-Lymphocytes, Regulatory/immunology , Adult , Cholecalciferol/blood , Cholecalciferol/pharmacology , Dietary Supplements , Double-Blind Method , Female , Humans , Inflammation/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-10/analysis , Interleukin-10/immunology , Male , Middle Aged , Sunlight , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/physiology
10.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R112-R124, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34075808

ABSTRACT

Preeclampsia (PE) is characterized by maternal hypertension, intrauterine growth restriction, and increased cytolytic natural killer cells (cNKs), which secrete interferon γ (IFNγ). However, the precise role of IFNγ in contributing to PE pathophysiology remains unclear. Using the reduced uterine perfusion pressure (RUPP) rat model of placental ischemia, we tested the hypothesis that neutralization of IFNγ in RUPPs will decrease placental reactive oxygen species (ROS) and improve vascular function resulting in decreased MAP and improved fetal growth. On gestation day (GD) 14, the RUPP procedure was performed and on GDs 15 and 18, a subset of normal pregnant rats (NP) and RUPP rats were injected with 10 µg/kg of an anti-rat IFNγ monoclonal antibody. On GD 18, uterine artery resistance index (UARI) was measured via Doppler ultrasound and on GD 19, mean arterial pressure (MAP) was measured, animals were euthanized, and blood and tissues were collected for analysis. Increased MAP was observed in RUPP rats compared with NP and was reduced in RUPP + anti-IFNγ. Placental ROS was also increased in RUPP rats compared with NP rats and was normalized in RUPP + anti-IFNγ. Fetal and placental weights were reduced in RUPP rats, but were not improved following anti-IFNγ treatment. However, UARI was elevated in RUPP compared with NP rats and was reduced in RUPP + anti-IFNγ. In conclusion, we observed that IFNγ neutralization reduced MAP, UARI, and placental ROS in RUPP recipients. These data suggest that IFNγ is a potential mechanism by which cNKs contribute to PE pathophysiology and may represent a therapeutic target to improve maternal outcomes in PE.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arterial Pressure/drug effects , Interferon-gamma/antagonists & inhibitors , Killer Cells, Natural/drug effects , Oxidative Stress/drug effects , Placenta/blood supply , Placenta/drug effects , Pre-Eclampsia/prevention & control , Uterine Artery/drug effects , Vascular Resistance/drug effects , Angiogenic Proteins/metabolism , Animals , Disease Models, Animal , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/physiopathology , Fetal Growth Retardation/prevention & control , Interferon-gamma/metabolism , Ischemia/metabolism , Ischemia/physiopathology , Killer Cells, Natural/metabolism , Placenta/metabolism , Placental Circulation , Pre-Eclampsia/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Uterine Artery/metabolism , Uterine Artery/physiopathology
11.
Indian J Pharmacol ; 53(3): 226-228, 2021.
Article in English | MEDLINE | ID: mdl-34169908

ABSTRACT

Although many potent drugs have been used for cytokine storm, mortality is high for patients with coronavirus disease-2019 (COVID-19), which is followed up in the intensive care unit. Interferons (IFNs) are the major cytokines of the antiviral defense system released from many cell types. However, IFN-γ plays a key role in both primary and secondary cytokine storms. If the cytokine storm is not treated urgently, it will be fatal; therefore, it should be treated immediately. Anakinra, an interleukin-1 (IL-1) antagonist, tocilizumab, an IL-6 antagonist, and Janus kinase (JAK) inhibitors are successfully used in cytokine storm caused by COVID-19. However, sometimes, despite these treatments, the patient's clinical course does not improve. Emapalumab (Eb) is the human immunoglobulin G1 monoclonal antibody and is a potent and noncompetitive antagonist of IFN-γ. Eb can be life saving for cytokine storm caused by COVID-19, which is resistant to anakinra, tocilizumab, and JAK inhibitors.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , COVID-19 Drug Treatment , Cytokine Release Syndrome/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Neutralizing/pharmacology , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , COVID-19/epidemiology , COVID-19/immunology , Cytokine Release Syndrome/epidemiology , Cytokine Release Syndrome/immunology , Disease Progression , Drug Resistance, Viral , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukins/antagonists & inhibitors , Interleukins/immunology , Janus Kinase Inhibitors/pharmacology , Recurrence
12.
Int Immunopharmacol ; 96: 107687, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33965879

ABSTRACT

Paeoniflorin (PF) has been demonstrated to have an anti-allergic and anti-inflammatory effect in the treatment of allergic contact dermatitis (ACD). However, its clinical application is hampered by the lacking of comprehensive mechanical explanation. This research aimed to study the effect of PF on the proliferation, apoptosis and cytokines secretion as well as the expression of nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways of T lymphocytes activation in vitro and in vivo. We found that PF depressed human T lymphocytes activation via inhibition ofinterferon-gamma (IFN-γ) production and NF-κB/IκBα and p38 MAPK signaling pathway in vitro, also PF could attenuate such ACD responses by inhibiting the production of IFN-γ and NF-κB/IκBα pathway in T lymphocytes of ACD mouse model, suggesting that PF might be useful for the treatment of T cell-mediated allergic inflammatory disorders such as ACD. This would make PF a promising T cell-targeted drug candidate for further study because of its immunosuppressive and anti-inflammatory effects.


Subject(s)
Dermatitis, Allergic Contact/drug therapy , Glucosides/pharmacology , Inflammation/diet therapy , Interferon-gamma/antagonists & inhibitors , Monoterpenes/pharmacology , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , T-Lymphocytes/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cells, Cultured , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/pathology , Disease Models, Animal , Humans , Inflammation/immunology , Inflammation/pathology , NF-KappaB Inhibitor alpha/genetics , NF-kappa B/genetics , Signal Transduction , T-Lymphocytes/drug effects
13.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525403

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disease that is characterized by an impaired skin barrier and intense itchiness, which decreases the individual's quality of life. No fully effective therapeutic agents have prevailed for AD due to an insufficient grasp of the complex etiology. Ellagic acid (EA), a natural compound, has anti-inflammatory properties in chronic diseases. The effects of EA on AD have not yet been explored. The present study investigated the effects of EA on TNF-α/IFN-γ-stimulated HaCaT keratinocytes and house dust mite-induced AD-like skin lesions in NC/Nga mice. Treatment with EA suppressed inflammatory responses in keratinocytes by regulating critical inflammatory signaling pathways, such as mitogen-activated protein kinases and signal transducers and activators of transcription. In vivo studies using a DfE-induced AD mouse model showed the effects of EA administration through ameliorated skin lesions via decremented histological inflammatory reactions. These results suggest that EA could be a potential therapeutic alternative for the treatment of AD by inhibiting inflammatory signaling pathways.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dermatitis, Atopic/drug therapy , Dermatophagoides farinae/chemistry , Ellagic Acid/pharmacology , Mitogen-Activated Protein Kinases/genetics , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Animals , Antigens, Dermatophagoides/administration & dosage , Chemokine CCL17/genetics , Chemokine CCL17/immunology , Chemokine CCL22/genetics , Chemokine CCL22/immunology , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Complex Mixtures/administration & dosage , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/genetics , Dermatitis, Atopic/immunology , Dermatophagoides farinae/immunology , Disease Models, Animal , Female , Gene Expression Regulation , HaCaT Cells , Humans , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , Mice , Mitogen-Activated Protein Kinases/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , Thymic Stromal Lymphopoietin
14.
Pediatr Blood Cancer ; 68(4): e28900, 2021 04.
Article in English | MEDLINE | ID: mdl-33484058

ABSTRACT

Interferon-gamma (IFN-γ) plays a key role in the pathophysiology of hemophagocytic lymphohistiocytosis (HLH), and available evidence also points to a role in other conditions, including aplastic anemia (AA) and graft failure following allogeneic hematopoietic stem cell transplantation. Recently, the therapeutic potential of IFN-γ inhibition has been documented; emapalumab, an anti-IFN-γ monoclonal antibody, has been approved in the United States for treatment of primary HLH that is refractory, recurrent or progressive, or in patients with intolerance to conventional therapy. Moreover, ruxolitinib, an inhibitor of JAK/STAT intracellular signaling, is currently being investigated for treating HLH. In AA, IFN-γ inhibits hematopoiesis by disrupting the interaction between thrombopoietin and its receptor, c-MPL. Eltrombopag, a small-molecule agonist of c-MPL, acts at a different binding site to IFN-γ and is thus able to circumvent its inhibitory effects. Ongoing trials will elucidate the role of IFN-γ neutralization in secondary HLH and future studies could explore this strategy in controlling hyperinflammation due to CAR T cells.


Subject(s)
Hematologic Neoplasms/metabolism , Hematologic Neoplasms/therapy , Interferon-gamma/metabolism , Animals , Antineoplastic Agents/therapeutic use , Benzoates/therapeutic use , Child , Drug Discovery , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Humans , Hydrazines/therapeutic use , Immunotherapy , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/immunology , Janus Kinases/antagonists & inhibitors , Nitriles , Pyrazoles/therapeutic use , Pyrimidines , Signal Transduction/drug effects
15.
J Microbiol Immunol Infect ; 54(3): 396-403, 2021 Jun.
Article in English | MEDLINE | ID: mdl-31780358

ABSTRACT

BACKGROUND/PURPOSE: Severe infection with influenza A (H1N1)pdm09 virus is characterized by acute lung injury. The limited efficacy of anti-viral drugs indicates an urgent need for additional therapies. We have previously reported that neutralization of gamma interferon (IFN-γ) could significantly rescue the thymic atrophy induced by severe influenza A (H1N1)pdm09 infection in BALB/c mice. A deeper investigation was conducted into the influence of neutralizing IFN-γ to the BALB/c mice weight, survival rate, and lung injury. METHODS: The BALB/c mice was infected with severe influenza A (H1N1)pdm09. Monoclonal antibodies against IFN-γ were injected into the abdominal cavities of the mice. After neutralization of IFN-γ occurred in mice infected by severe ∖ influenza A (H1N1)pdm09, observing the influence of neutralizing IFN-γ to the BALB/c mice weight, survival rate, lung injury. RESULT: Our results here showed that anti-IFN-γ therapy alleviated the acute lung injury in this mouse model. Neutralization of IFN-γ led to a significant reduction in the lung microvascular leak and the cellular infiltrate in the lung tissue, and also improved the outcome in mice mortality. Several pro-inflammatory cytokines, including interleukin (IL)-1α, tumor necrosis factor (TNF)-α and granulocyte-colony stimulating factor (G-CSF) in the bronchoalveolar lavage fluid (BALF), and the chemokines including G-CSF, monocyte chemoattractant protein-1 (MCP-1) in serum samples were found to be significantly reduced after anti-IFN-γ treatment. CONCLUSION: These results suggested that IFN-γ plays an important role in acute lung injury induced by severe influenza A (H1N1)pdm09 infection, and monoclonal antibodies against IFN-γ could be useful as a potential therapeutic remedy for future influenza pandemics.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/prevention & control , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Interferon-gamma/antagonists & inhibitors , Lung/virology , Orthomyxoviridae Infections/drug therapy , Animals , Cytokines/analysis , Cytokines/immunology , Disease Models, Animal , Female , Influenza A Virus, H1N1 Subtype/pathogenicity , Lung/pathology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/virology
16.
Clin Transl Oncol ; 23(4): 750-756, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32737836

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) represents an important neoplasm with high mortality. Although PD-L1/PD-1 system-based immunotherapy has benefits for a certain type of CRC, many efforts should be made to enhance the responses to anti-PD-1/PD-L1 drugs. DNA methylation has been critically implicated in the regulation of tumor immunity. Here, we examined the effects of the natural alkaloid oxymatrine on PD-L1 expression in CRC cells and to elucidate the underlying mechanism. METHODS: Human CRC SW620 and HCT116 cells were treated with interferon γ (IFNγ) and/or oxymatrine. Cell viability was determined using MTT assays. PD-L1 expression was detected by real-time PCR and Western blot analyses. DNA demethylase activity was measured using kits. RESULTS: Oxymatrine did not apparently affect the viability of normal human intestinal epithelial cells. IFNγ at 20 ng/ml increased the viability of CRC cells, but oxymatrine concentration-dependently reduced the viability in the absence or presence of IFNγ. IFNγ increased the mRNA and protein expression of PD-L1 in the two cell lines, but oxymatrine significantly abolished IFNγ-elevated PD-L1 levels at both mRNA and protein levels. Furthermore, DNA demethylase activity was remarkably increased in IFNγ-treated CRC cells, which was abolished by oxymatrine concentration-dependently. In addition, DNA methyltransferase inhibitor 5-azacytidine considerably abrogated oxymatrine-induced downregulation of PD-L1 mRNA and protein levels in IFNγ-stimulated CRC cells. CONCLUSION: Oxymatrine suppressed viability and reduced PD-L1 expression in IFNγ-stimulated CRC cells, which was attributed to enhanced DNA demethylation. Our current discoveries suggested oxymatrine as an epigenetic modulatory agent for immunotherapy against CRC via PD-1/PD-L1 blockade.


Subject(s)
Alkaloids/pharmacology , B7-H1 Antigen/metabolism , Colorectal Neoplasms/metabolism , DNA Demethylation/drug effects , Quinolizines/pharmacology , Azacitidine/pharmacology , B7-H1 Antigen/genetics , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , HCT116 Cells , Humans , Immune Checkpoint Inhibitors , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/pharmacology , RNA, Messenger/metabolism
17.
Biochem Pharmacol ; 183: 114356, 2021 01.
Article in English | MEDLINE | ID: mdl-33285108

ABSTRACT

Abnormal activation of transforming growth factor (TGF)-ß is a common cause of fibroblast activation and fibrosis. In bleomycin (BLM)-induced lung fibrosis, the marked expression of phospho-Src homology-2 domain-containing phosphatase (SHP) 2, phospho-signal transducer and activator of transcription (STAT) 3, and suppressor of cytokine signaling (SOCS) 3 was highly associated with pulmonary parenchymal lesions and collagen deposition. Human pulmonary fibroblasts differentiated into myofibroblasts exhibited activation of SHP2, SOCS3, protein inhibitor of activated STAT1, STAT3, interleukin (IL)-6, and IL-10. The significant retardation of interferon (IFN)-γ signaling in myofibroblasts was revealed by the decreased expression of phospho-STAT1, IFN-γ-associated genes, and IFN-γ-inducible protein (IP) 10. Microarray analysis showed an induction of fibrotic genes in TGF-ß1-differentiated myofibroblasts, whereas IFN-γ-regulated anti-fibrotic genes were suppressed. Interestingly, BIBF 1120 treatment effectively inhibited both STAT3 and SHP2 phosphorylation in TGF-ß1-differentiated myofibroblasts and BLM fibrotic lung tissues, which was accompanied by suppression of fibroblast-myofibroblast transition. Moreover, the combined treatment of BIBF 1120 plus IFN-γ or SHP2 inhibitor PHPS1 plus IFN-γ markedly reduced TGF-ß1-induced α-smooth muscle actin and further ameliorated BLM lung fibrosis. Accordingly, myofibroblasts were hyporesponsiveness to IFN-γ, while blockade of SHP2 contributed to the anti-fibrotic efficacy of IFN-γ.


Subject(s)
Bleomycin/toxicity , Fibroblasts/metabolism , Interferon-gamma/metabolism , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Interferon-gamma/antagonists & inhibitors , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta/toxicity
18.
Biochem Biophys Res Commun ; 533(4): 1519-1526, 2020 12 17.
Article in English | MEDLINE | ID: mdl-33158480

ABSTRACT

Cognitive impairment, acute or long-term, is a common complication in patients with severe bacterial infection. However, the underlying mechanisms are not fully verified and effective medicine is not available in clinics. Interferon gamma (IFNγ) is a pivotal cytokine against infection and is believed to be a tune in homeostasis of cognitive function. Here, we collected blood and cerebrospinal fluid (CF) from human subjects and mice, and found that plasma and CF levels of IFNγ were significantly increased in septic patients and endotoxin-challenged mice when compared with healthy controls. IFNγ signaling was boosted in the hippocampus of mice after a challenge of lipopolysaccharide (LPS), which was accompanied with cognitive impairment and decline of neurogenesis. Deficiency of IFNγ or its receptor (IFNγR) dramatically attenuated microglia-induced A1 astrocytes and consequently restored neurogenesis and cognitive function in endotoxemia mice model. Using primary microglia, astrocytes and neurons, we found that IFNγ remarkably increased LPS-mediated release of TNFα and IL-1α in microglia and consequently induced the transformation of astrocyte to A1 subtype, which ultimately resulted in neuron damage. Thus, IFNγ promotes cognitive impairment in endotoxemia by enhancing microglia-induced A1 astrocytes. Targeting IFNγ would be a novel strategy for preventing or treating cognitive dysfunction in patients with Gram-negative infection.


Subject(s)
Astrocytes/physiology , Cognitive Dysfunction/physiopathology , Endotoxemia/physiopathology , Interferon-gamma/antagonists & inhibitors , Neurogenesis/physiology , Animals , Astrocytes/pathology , Case-Control Studies , Cells, Cultured , Cognitive Dysfunction/genetics , Cognitive Dysfunction/therapy , Disease Models, Animal , Endotoxemia/genetics , Endotoxemia/psychology , Gene Silencing , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Microglia/physiology , Neurogenesis/genetics , RNAi Therapeutics , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Receptors, Interferon/physiology , Interferon gamma Receptor
19.
J Immunol ; 205(10): 2821-2833, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33028619

ABSTRACT

A hallmark of secondary hemophagocytic lymphohistiocytosis (sHLH), a severe form of cytokine storm syndrome, is the emergence of overactivated macrophages that engulf healthy host blood cells (i.e., hemophagocytosis) and contribute to the dysregulated inflammation-driven pathology. In this study, we show that depleting SIRPα (SIRPα-/-) in mice during TLR9-driven inflammation exacerbates and accelerates the onset of fulminant sHLH, in which systemic hemophagocytosis, hypercytokinemia, consumptive cytopenias, hyperferritinemia, and other hemophagocytic lymphohistiocytosis hallmarks were apparent. In contrast, mice expressing SIRPα, including those deficient of the SIRPα ligand CD47 (CD47-/-), do not phenocopy SIRPα deficiency and fail to fully develop sHLH, albeit TLR9-inflamed wild-type and CD47-/- mice exhibited hemophagocytosis, anemia, and splenomegaly. Although IFN-γ is largely considered a driver of hemophagocytic lymphohistiocytosis pathology, IFN-γ neutralization did not preclude the precipitation of sHLH in TLR9-inflamed SIRPα-/- mice, whereas macrophage depletion attenuated sHLH in SIRPα-/- mice. Mechanistic studies confirmed that SIRPα not only restrains macrophages from acquiring a hemophagocytic phenotype but also tempers their proinflammatory cytokine and ferritin secretion by negatively regulating Erk1/2 and p38 activation downstream of TLR9 signaling. In addition to TLR9 agonists, TLR2, TLR3, or TLR4 agonists, as well as TNF-α, IL-6, or IL-17A, but not IFN-γ, similarly induced sHLH in SIRPα-/- mice but not SIRPα+ mice. Collectively, our study suggests that SIRPα plays a previously unappreciated role in sHLH/cytokine storm syndrome pathogenesis by preventing macrophages from becoming both hemophagocytic and hyperactivated under proinflammation.


Subject(s)
Cytokine Release Syndrome/immunology , Interferon-gamma/metabolism , Lymphohistiocytosis, Hemophagocytic/immunology , Macrophages/immunology , Receptors, Immunologic/metabolism , Animals , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cytokine Release Syndrome/genetics , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Interferon-gamma/antagonists & inhibitors , Lymphohistiocytosis, Hemophagocytic/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Knockout , Receptors, Immunologic/genetics , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/metabolism
20.
Biomed Pharmacother ; 130: 110587, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32763819

ABSTRACT

Doxorubicin (DOX) is commonly used as an anti-cancer agent. However, its severe cardiotoxicity often makes it life threatening even long after DOX therapy during childhood. We recently reported interferon-γ (IFN-γ) necessary for DOX-induced acute cardiotoxicity in a p38 dependent way and, asked here for the potential of IFN-γ blockade to prevent DOX-induced chronic cardiotoxicity during tumor therapy. In our model system, mice without or with growing tumors repeatedly received DOX treatment. Simultaneous injection of anti-IFN-γ antibody R46-A2 with DOX to block IFN-γ signal efficiently protected the cardiac function of DOX treated recipients. Importantly, a single late injection of R46-A2 after DOX exposure also ameliorated DOX induced cardiac dysfunction in tumor-bearing mice. The anti-IFN-γ treatment did not affect the DOX-mediated tumor suppression effect and it left the main cellular immune response intact. Therefore, temporary blockade of IFN-γ may represent a novel strategy to ameliorate established DOX induced cardiotoxicity (DIC) or prevent its development in tumor therapy.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Antibodies/therapeutic use , Cardiotoxicity/drug therapy , Doxorubicin/therapeutic use , Interferon-gamma/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/adverse effects , Cell Line, Tumor , Doxorubicin/adverse effects , Female , Mice, Inbred BALB C , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...