Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.974
Filter
1.
BMC Cardiovasc Disord ; 24(1): 231, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679696

ABSTRACT

BACKGROUND: Oxidized low-density lipoprotein (ox-LDL) can initiate and affect almost all atherosclerotic events including endothelial dysfunction. In this text, the role and underlying molecular basis of procyanidin B2 (PCB2) with potential anti-oxidant and anti-inflammatory activities in ox-LDL-induced HUVEC injury were examined. METHODS: HUVECs were treated with ox-LDL in the presence or absence of PCB2. Cell viability and apoptotic rate were examined by CCK-8 assay and flow cytometry, respectively. The mRNA and protein levels of genes were tested by RT-qPCR and western blot assays, respectively. Potential downstream targets and pathways of apple procyanidin oligomers were examined by bioinformatics analysis for the GSE9647 dataset. The effect of PCB2 on THP-1 cell migration was examined by recruitment assay. The effect of PCB2 on oxidative stress was assessed by reactive oxygen species (ROS) level, malondialdehyde (MDA) content, and mitochondrial membrane potential (MMP). RESULTS: ox-LDL reduced cell viability, induced cell apoptosis, and facilitated the expression of oxidized low-density lipoprotein receptor 1 (LOX-1), C-C motif chemokine ligand 2 (MCP-1), vascular cell adhesion protein 1 (VCAM-1) in HUVECs. PCB2 alleviated ox-LDL-induced cell injury in HUVECs. Apple procyanidin oligomers triggered the differential expression of 592 genes in HUVECs (|log2fold-change| > 0.58 and adjusted p-value < 0.05). These dysregulated genes might be implicated in apoptosis, endothelial cell proliferation, inflammation, and monocyte chemotaxis. PCB2 inhibited C-X-C motif chemokine ligand 1/8 (CXCL1/8) expression and THP-1 cell recruitment in ox-LDL-stimulated HUVECs. PCB2 inhibited ox-LDL-induced oxidative stress and nuclear factor kappa-B (NF-κB) activation in HUVECs. CONCLUSION: PCB2 weakened ox-LDL-induced cell injury, inflammation, monocyte recruitment, and oxidative stress by inhibiting the NF-κB pathway in HUVECs.


Subject(s)
Anti-Inflammatory Agents , Apoptosis , Biflavonoids , Catechin , Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , NF-kappa B , Oxidative Stress , Proanthocyanidins , Signal Transduction , Humans , Lipoproteins, LDL/toxicity , Catechin/pharmacology , Proanthocyanidins/pharmacology , Oxidative Stress/drug effects , Biflavonoids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Signal Transduction/drug effects , NF-kappa B/metabolism , Apoptosis/drug effects , Anti-Inflammatory Agents/pharmacology , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Antioxidants/pharmacology , THP-1 Cells , Chemotaxis, Leukocyte/drug effects , Reactive Oxygen Species/metabolism , Scavenger Receptors, Class E/metabolism , Scavenger Receptors, Class E/genetics
2.
Cell Death Dis ; 13(1): 33, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013119

ABSTRACT

Aged microglia display augmented inflammatory activity after neural injury. Although aging is a risk factor for poor outcome after brain insults, the precise impact of aging-related alterations in microglia on neural injury remains poorly understood. Microglia can be eliminated via pharmacological inhibition of the colony-stimulating factor 1 receptor (CSF1R). Upon withdrawal of CSF1R inhibitors, microglia rapidly repopulate the entire brain, leading to replacement of the microglial compartment. In this study, we investigated the impact of microglial replacement in the aged brain on neural injury using a mouse model of intracerebral hemorrhage (ICH) induced by collagenase injection. We found that replacement of microglia in the aged brain reduced neurological deficits and brain edema after ICH. Microglial replacement-induced attenuation of ICH injury was accompanied with alleviated blood-brain barrier disruption and leukocyte infiltration. Notably, newly repopulated microglia had reduced expression of IL-1ß, TNF-α and CD86, and upregulation of CD206 in response to ICH. Our findings suggest that replacement of microglia in the aged brain restricts neuroinflammation and brain injury following ICH.


Subject(s)
Aging/drug effects , Brain/drug effects , Cerebral Hemorrhage/drug therapy , Microglia/drug effects , Neuroinflammatory Diseases/drug therapy , Aging/pathology , Aminopyridines/administration & dosage , Aminopyridines/pharmacology , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/immunology , Brain/pathology , Brain Injuries/drug therapy , Brain Injuries/etiology , Brain Injuries/immunology , Brain Injuries/pathology , Cell Death/drug effects , Cerebral Hemorrhage/complications , Cerebral Hemorrhage/immunology , Cerebral Hemorrhage/pathology , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Mice , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
3.
Front Immunol ; 12: 777932, 2021.
Article in English | MEDLINE | ID: mdl-34899737

ABSTRACT

Complement activation is key to anti-microbial defenses by directly acting on microbes and indirectly by triggering cellular immune responses. Complement activation may also contribute to the pathogenesis of numerous inflammatory and immunological diseases. Consequently, intense research focuses on developing therapeutics that block pathology-causing complement activation while preserving anti-microbial complement activities. However, the pace of research is slowed down significantly by the limitations of current tools for evaluating complement-targeting therapeutics. Moreover, the effects of potential therapeutic agents on innate immune cells, like neutrophils, are not fully understood. Here, we employ microfluidic assays and measure chemotaxis, phagocytosis, and swarming changes in human neutrophils ex vivo in response to various complement-targeting agents. We show that whereas complement factor 5 (C5) cleavage inhibitor eculizumab blocks all neutrophil anti-microbial functions, newer compounds like the C5 cleavage inhibitor RA101295 and C5a receptor antagonist avacopan inhibit chemotaxis and swarming while preserving neutrophil phagocytosis. These results highlight the utility of microfluidic neutrophil assays in evaluating potential complement-targeting therapeutics.


Subject(s)
Aniline Compounds/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Complement Activation/drug effects , Complement Inactivating Agents/pharmacology , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Neutrophils/drug effects , Nipecotic Acids/pharmacology , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Complement C3/pharmacology , Complement C3-C5 Convertases/antagonists & inhibitors , Complement C3-C5 Convertases/metabolism , Complement C5a/pharmacology , Humans , Neutrophil Activation/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/drug effects , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/metabolism
4.
Front Immunol ; 12: 779076, 2021.
Article in English | MEDLINE | ID: mdl-34899738

ABSTRACT

Graft versus host disease (GVHD) is the major non-relapse complication associated with allogeneic hematopoietic stem cell transplantation (HSCT). Unfortunately, GVHD occurs in roughly half of patients following this therapy and can induce severe life-threatening side effects and premature mortality. The pathophysiology of GVHD is driven by alloreactive donor T cells that induce a proinflammatory environment to cause pathological damage in the skin, gastrointestinal (GI) tract, lung, and liver during the acute phase of this disease. Recent work has demonstrated that the GI tract is a pivotal target organ and a primary driver of morbidity and mortality in patients. Prevention of this complication has therefore emerged as an important goal of prophylaxis strategies given the primacy of this tissue site in GVHD pathophysiology. In this review, we summarize foundational pre-clinical studies that have been conducted in animal models to prevent GI tract GVHD and examine the efficacy of these approaches upon subsequent translation into the clinic. Specifically, we focus on therapies designed to block inflammatory cytokine pathways, inhibit cellular trafficking of alloreactive donor T cells to the GI tract, and reconstitute impaired regulatory networks for the prevention of GVHD in the GI tract.


Subject(s)
Gastrointestinal Diseases/prevention & control , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunotherapy , T-Lymphocytes/immunology , Adoptive Transfer , Animals , Anti-Inflammatory Agents/pharmacology , Chemotaxis, Leukocyte/drug effects , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/metabolism , Humans , Immunosuppressive Agents/pharmacology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Translational Research, Biomedical , Transplantation, Homologous/adverse effects
5.
Bull Exp Biol Med ; 172(2): 137-142, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855095

ABSTRACT

3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) along with their blood lipid-lowering effect exhibit anti-inflammatory and immunomodulatory activity. We studied the effects of long-term (72-h or longer) exposure of human T lymphocytes in culture to atorvastatin and rosuvastatin (5-80 nM) on their functional activity. Treatment with statins inhibited PHA/IL-2-induced proliferation of CD4+ T lymphocytes isolated from the peripheral blood of healthy donors. This was accompanied by a decrease in the relative content of cells expressing active caspase-3. Addition of mevalonate or fetal bovine serum simultaneously with statins restored proliferative activity of cells. Culturing of CD4+ T lymphocytes with statins in the presence of IL-2 did not significantly affect the expression of chemokine receptors CCR4, CCR5, CXCR3, and CXCR4. Pretreatment with statins suppressed spontaneous and SDF-1-stimulated migration of CD4+ T lymphocytes, but little changed the content of intracellular phosphorylated protein kinases Akt, p38 and p42/44 (ERK1/2). The cellular effects of "lipophilic" atorvastatin were observed at lower concentrations compared to "hydrophilic" rosuvastatin.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Atorvastatin/pharmacology , CD4-Positive T-Lymphocytes/physiology , Cell Proliferation/drug effects , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Humans , Receptors, CCR4/metabolism , Receptors, CCR5/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR4/metabolism , Rosuvastatin Calcium/pharmacology , Signal Transduction/drug effects
6.
Inflammopharmacology ; 29(6): 1829-1833, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34792671

ABSTRACT

Ouabain is a cardiac steroid hormone with immunomodulatory effects. It inhibits neutrophils migration induced by different stimuli, but little is known about the mechanisms involved in this effect. Thus, the aim of this study was to evaluate the ouabain effect on chemotactic signaling pathways in neutrophils. For that, mice neutrophils were isolated from bone marrow, treated with ouabain (1, 10, and 100 nM) for 2 h, submitted to transwell chemotaxis assay and flow cytometry analysis of Akt, ERK, JNK, and p38 phosphorylation induced by zymosan. Ouabain treatment (1, 10 and, 100 nM) reduces neutrophil chemotaxis induced by chemotactic peptide fMLP, but this substance did not inhibit Akt, ERK, and JNK activation induced by zymosan. However, ouabain (1 and 10 nM) reduced p38 phosphorylation in zymosan-stimulated neutrophils. These results suggest that ouabain may interfere in neutrophil migration through p38 MAPK inhibition.


Subject(s)
Neutrophils/drug effects , Ouabain/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Movement/drug effects , Chemotaxis, Leukocyte/drug effects , Dose-Response Relationship, Drug , Female , Flow Cytometry , Mice , Neutrophils/metabolism , Ouabain/administration & dosage , Phosphorylation/drug effects , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Pharmacol Res Perspect ; 9(5): e00873, 2021 10.
Article in English | MEDLINE | ID: mdl-34632734

ABSTRACT

We hypothesized whether propofol or active propofol component (2,6-diisopropylphenol [DIPPH] and lipid excipient [LIP-EXC]) separately may alter inflammatory mediators expressed by macrophages and neutrophils in lean and obese rats. Male Wistar rats (n = 10) were randomly assigned to receive a standard (lean) or obesity-inducing diet (obese) for 12 weeks. Animals were euthanized, and alveolar macrophages and neutrophils from lean and obese animals were exposed to propofol (50 µM), active propofol component (50 µM, 2,6-DIPPH), and lipid excipient (soybean oil, purified egg phospholipid, and glycerol) for 1 h. The primary outcome was IL-6 expression after propofol and its components exposure by alveolar macrophages extracted from bronchoalveolar lavage fluid. The secondary outcomes were the production of mediators released by macrophages from adipose tissue, and neutrophils from lung and adipose tissues, and neutrophil migration. IL-6 increased after the exposure to both propofol (median [interquartile range] 4.14[1.95-5.20]; p = .04) and its active component (2,6-DIPPH) (4.09[1.67-5.91]; p = .04) in alveolar macrophages from obese animals. However, only 2,6-DIPPH increased IL-10 expression (7.59[6.28-12.95]; p = .001) in adipose tissue-derived macrophages. Additionally, 2,6-DIPPH increased C-X-C chemokine receptor 2 and 4 (CXCR2 and CXCR4, respectively) in lung (10.08[8.23-29.01]; p = .02; 1.55[1.49-3.43]; p = .02) and adipose tissues (8.78[4.15-11.57]; p = .03; 2.86[2.17-3.71]; p = .01), as well as improved lung-derived neutrophil migration (28.00[-3.42 to 45.07]; p = .001). In obesity, the active component of propofol affected both the M1 and M2 markers as well as neutrophils in both alveolar and adipose tissue cells, suggesting that lipid excipient may hinder the effects of active propofol.


Subject(s)
Adipose Tissue/drug effects , Anesthetics, Intravenous/pharmacology , Excipients/pharmacology , Interleukin-6/metabolism , Lung/drug effects , Macrophages, Alveolar/drug effects , Neutrophils/drug effects , Obesity/metabolism , Propofol/pharmacology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Chemotaxis, Leukocyte/drug effects , Glycerol/pharmacology , Interleukin-10/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Neutrophils/metabolism , Phospholipids/pharmacology , Rats , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/drug effects , Receptors, Interleukin-8B/metabolism , Soybean Oil/pharmacology
9.
Front Immunol ; 12: 737245, 2021.
Article in English | MEDLINE | ID: mdl-34580585

ABSTRACT

Allergic asthma is a common inflammatory airway disease in which Th2 immune response and inflammation are thought to be triggered by inhalation of environmental allergens. Many studies using mouse models and human tissues and genome-wide association have indicated that Sonic Hedgehog (Shh) and the Hedgehog (Hh) signaling pathway are involved in allergic asthma and that Shh is upregulated in the lung on disease induction. We used a papain-induced mouse model of allergic airway inflammation to investigate the impact of systemic pharmacological inhibition of the Hh signal transduction molecule smoothened on allergic airway disease induction and severity. Smoothened-inhibitor treatment reduced the induction of Shh, IL-4, and IL-13 in the lung and decreased serum IgE, as well as the expression of Smo, Il4, Il13, and the mucin gene Muc5ac in lung tissue. Smoothened inhibitor treatment reduced cellular infiltration of eosinophils, mast cells, basophils, and CD4+ T-cells to the lung, and eosinophils and CD4+ T-cells in the bronchoalveolar lavage. In the mediastinal lymph nodes, smoothened inhibitor treatment reduced the number of CD4+ T-cells, and the cell surface expression of Th2 markers ST2 and IL-4rα and expression of Th2 cytokines. Thus, overall pharmacological smoothened inhibition attenuated T-cell infiltration to the lung and Th2 function and reduced disease severity and inflammation in the airway.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Asthma/drug therapy , Benzimidazoles/administration & dosage , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Lung/drug effects , Phenylurea Compounds/administration & dosage , Pneumonia/drug therapy , Smoothened Receptor/antagonists & inhibitors , Th2 Cells/drug effects , Animals , Asthma/immunology , Asthma/metabolism , Disease Models, Animal , Female , Injections, Intraperitoneal , Lung/immunology , Lung/metabolism , Male , Mice, Inbred C57BL , Pneumonia/immunology , Pneumonia/metabolism , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
10.
Int Immunopharmacol ; 100: 108090, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34507108

ABSTRACT

BACKGROUND: Immunomodulatory therapies are claimed to enhance antimicrobial immunity and counterbalance antimicrobial resistance mechanisms of pathogenic bacteria. PURPOSE: To investigate whether caffeine can be useful for control of inflammation derived from experimental systemic infection with Listeria monocytogenes. METHODS: Peritoneal macrophages (pMØ) from Swiss mice were cultured with caffeine in 96-well plates, and then infected with virulent L. monocytogenes 619. In another experiment, the pMØ were first infected with the bacterium and then treated with caffeine. Swiss mice were inoculated intraperitoneally with L. monocytogenes and then treated intravenously with caffeine (0.05; 0.5 or 5 mg/Kg). RESULTS: Caffeine did not exert direct antibacterial activity in vitro against L. monocytogenes. Macrophages exposed to caffeine before or after infection with L. monocytogenes had increased cell viability, although the intracellular bacterial loads were similar to the control groups. Caffeine treatments of Swiss mice reduced leukocyte infiltration into the peritoneal cavity after L. monocytogenes infection. However, the bacterial burden was reduced in the spleen and liver. The mRNA expressions of IL-1ß, IL-6 and the enzyme inducible nitric oxide synthase (iNOS) were reduced whereas IL-10 was increased. CONCLUSION: Caffeine has an anti-infectious potential and ameliorated infection-derived inflammation following experimental infection with L. monocytogenes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Caffeine/pharmacology , Inflammation/drug therapy , Listeria monocytogenes/pathogenicity , Listeriosis/drug therapy , Macrophages, Peritoneal/drug effects , Animals , Caffeine/analogs & derivatives , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Host-Pathogen Interactions , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation Mediators/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/metabolism , Listeriosis/microbiology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Virulence
11.
J Immunol Res ; 2021: 9957451, 2021.
Article in English | MEDLINE | ID: mdl-34337087

ABSTRACT

BACKGROUND: Solanum diploconos (Mart.) Bohs is a native Brazilian plant belonging to the Solanaceae family, popularly known as "tomatinho do mato" and poorly investigated. Herein, we presented for the first time evidence for the anti-inflammatory and wound healing activities of S. diploconos fruit hydroalcoholic extract. Material and Methods. In vitro fMLP-induced chemotaxis, LPS-induced inflammatory mediator levels (cytokines by ELISA and NO release by Griess reaction), and adhesion molecule expression (CD62L, CD49d, and CD18, by flow-cytometry) were assessed in neutrophils treated with different concentrations of the extract. Inflammation resolution was measured by the efferocytosis assay and the healing activity by in vivo and in vitro assays. The air pouch model of carrageenan-induced inflammation in Swiss mice was used to investigate the in vivo anti-inflammatory effects of the extract. Leukocyte influx (by optical microscopy) and cytokine release were quantified in the pouch exudates. Additionally, the acute and subacute toxic and genotoxic effects of the extract were evaluated. RESULTS: In vitro, the extract impaired neutrophil chemotaxis and its ability to produce and/or release cytokines (TNFα, IL-1ß, and IL-6) and NO upon LPS stimuli (p < 0.01). LPS-treated neutrophils incubated with the extract presented increased CD62L expression (p < 0.01), indicating a reduced activation. An enhanced efferocytosis of apoptotic neutrophils by macrophages was observed and accompanied by higher IL-10 and decreased TNFα secretion (p < 0.01). In vivo, similar results were noted, including reduction of neutrophil migration, protein exudation, and cytokine release (p < 0.01). Also, the extract increased fibroblast proliferation and promoted skin wound healing (p < 0.01). No signs of toxicity or genotoxicity were observed for the extract. CONCLUSION: S. diploconos fruit extract is anti-inflammatory by modulating neutrophil migration/activation as well macrophage-dependent efferocytosis and inflammatory mediator release. It also indicates its potential use as a healing agent. Finally, the absence of acute toxic and genotoxic effects reinforces its possible use as medicinal product.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Plant Extracts/pharmacology , Solanum/chemistry , Wound Healing/drug effects , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/therapeutic use , Carrageenan/administration & dosage , Carrageenan/immunology , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Female , Fruit/chemistry , Humans , Inflammation/immunology , Male , Mice , Neutrophils/drug effects , Neutrophils/immunology , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Rats , Toxicity Tests, Acute , Toxicity Tests, Subacute , Wound Healing/immunology
12.
Front Immunol ; 12: 633540, 2021.
Article in English | MEDLINE | ID: mdl-34295325

ABSTRACT

Sepsis is one of the most common comorbidities observed in diabetic patients, associated with a deficient innate immune response. Recently, we have shown that glucagon possesses anti-inflammatory properties. In this study, we investigated if hyperglucagonemia triggered by diabetes might reduce the migration of neutrophils, increasing sepsis susceptibility. 21 days after diabetes induction by intravenous injection of alloxan, we induced moderate sepsis in Swiss-Webster mice through cecum ligation and puncture (CLP). The glucagon receptor (GcgR) antagonist des-his1-[Glu9]-glucagon amide was injected intraperitoneally 24h and 1h before CLP. We also tested the effect of glucagon on CXCL1/KC-induced neutrophil migration to the peritoneal cavity in mice. Neutrophil chemotaxis in vitro was tested using transwell plates, and the expression of total PKA and phospho-PKA was evaluated by western blot. GcgR antagonist restored neutrophil migration, reduced CFU numbers in the peritoneal cavity and improved survival rate of diabetic mice after CLP procedure, however, the treatment did no alter hyperglycemia, CXCL1/KC plasma levels and blood neutrophilia. In addition, glucagon inhibited CXCL1/KC-induced neutrophil migration to the peritoneal cavity of non-diabetic mice. Glucagon also decreased the chemotaxis of neutrophils triggered by CXCL1/KC, PAF, or fMLP in vitro. The inhibitory action of glucagon occurred in parallel with the reduction of CXCL1/KC-induced actin polymerization in neutrophils in vitro, but not CD11a and CD11b translocation to cell surface. The suppressor effect of glucagon on CXCL1/KC-induced neutrophil chemotaxis in vitro was reversed by pre-treatment with GcgR antagonist and adenylyl cyclase or PKA inhibitors. Glucagon also increased PKA phosphorylation directly in neutrophils in vitro. Furthermore, glucagon impaired zymosan-A-induced ROS production by neutrophils in vitro. Human neutrophil chemotaxis and adherence to endothelial cells in vitro were inhibited by glucagon treatment. According to our results, this inhibition was independent of CD11a and CD11b translocation to neutrophil surface or neutrophil release of CXCL8/IL-8. Altogether, our results suggest that glucagon may be involved in the reduction of neutrophil migration and increased susceptibility to sepsis in diabetic mice. This work collaborates with better understanding of the increased susceptibility and worsening of sepsis in diabetics, which can contribute to the development of new effective therapeutic strategies for diabetic septic patients.


Subject(s)
Cell Movement/drug effects , Diabetes Mellitus, Experimental/complications , Disease Susceptibility/etiology , Glucagon/administration & dosage , Neutrophils/drug effects , Sepsis/etiology , Sepsis/immunology , Adult , Animals , Cell Movement/immunology , Chemotaxis, Leukocyte/drug effects , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/microbiology , Female , Glucagon/metabolism , Humans , Male , Mice , Mice, Inbred Strains , Neutrophils/immunology
13.
Nat Immunol ; 22(7): 851-864, 2021 07.
Article in English | MEDLINE | ID: mdl-34099918

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Subject(s)
Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytotoxicity, Immunologic/drug effects , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
14.
Front Immunol ; 12: 679856, 2021.
Article in English | MEDLINE | ID: mdl-34135907

ABSTRACT

Neutrophil trafficking, homeostatic and pathogen elicited, depends upon chemoattractant receptors triggering heterotrimeric G-protein Gαißγ signaling, whose magnitude and kinetics are governed by RGS protein/Gαi interactions. RGS proteins typically limit Gαi signaling by reducing the duration that Gαi subunits remain GTP bound and able to activate downstream effectors. Yet how in totality RGS proteins shape neutrophil chemoattractant receptor activated responses remains unclear. Here, we show that C57Bl/6 mouse neutrophils containing a genomic knock-in of a mutation that disables all RGS protein-Gαi2 interactions (G184S) cannot properly balance chemoattractant receptor signaling, nor appropriately respond to inflammatory insults. Mutant neutrophils accumulate in mouse bone marrow, spleen, lung, and liver; despite neutropenia and an intrinsic inability to properly mobilize from the bone marrow. In vitro they rapidly adhere to ICAM-1 coated plates, but in vivo they poorly adhere to blood vessel endothelium. Those few neutrophils that cross blood vessels and enter tissues migrate haphazardly. Following Concanavalin-A administration fragmented G184S neutrophils accumulate in liver sinusoids leading to thrombo-inflammation and perivasculitis. Thus, neutrophil Gαi2/RGS protein interactions both limit and facilitate Gαi2 signaling thereby promoting normal neutrophil trafficking, aging, and clearance.


Subject(s)
Cellular Senescence , Chemotaxis, Leukocyte , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction , Animals , Bone Marrow Transplantation , Cellular Senescence/genetics , Cellular Senescence/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Humans , Immunophenotyping , Male , Mice , Neutropenia/etiology , Neutrophils/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/metabolism
15.
Front Immunol ; 12: 624419, 2021.
Article in English | MEDLINE | ID: mdl-34140948

ABSTRACT

Integrin regulation by Rap1 is indispensable for lymphocyte recirculation. In mice having B-cell-specific Rap1a/b double knockouts (DKO), the number of B cells in lymph nodes decreased to approximately 4% of that of control mice, and B cells were present in the spleen and blood. Upon the immunization with NP-CGG, DKO mice demonstrated the defective GC formation in the spleen, and the reduced NP-specific antibody production. In vitro, Rap1 deficiency impaired the movement of activated B cells along the gradients of chemoattractants known to be critical for their localization in the follicles. Furthermore, B-1a cells were almost completely absent in the peritoneal cavity, spleen and blood of adult DKO mice, and the number of B-cell progenitor/precursor (B-p) were reduced in neonatal and fetal livers. However, DKO B-ps normally proliferated, and differentiated into IgM+ cells in the presence of IL-7. CXCL12-dependent migration of B-ps on the VCAM-1 was severely impaired by Rap1 deficiency. Immunostaining study of fetal livers revealed defects in the co-localization of DKO B-ps and IL-7-producing stromal cells. This study proposes that the profound effects of Rap1-deficiency on humoral responses and B-1a cell generation may be due to or in part caused by impairments of the chemoattractant-dependent positioning and the contact with stromal cells.


Subject(s)
B-Lymphocytes/metabolism , Chemotaxis, Leukocyte , Germinal Center/metabolism , rap GTP-Binding Proteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Chemokine CXCL12/pharmacology , Chemotaxis, Leukocyte/drug effects , Germinal Center/cytology , Germinal Center/drug effects , Germinal Center/immunology , Immunity, Humoral , Immunization , Intercellular Adhesion Molecule-1/metabolism , Liver/immunology , Liver/metabolism , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Spleen/immunology , Spleen/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , gamma-Globulins/pharmacology , rap GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/genetics
16.
MAbs ; 13(1): 1917484, 2021.
Article in English | MEDLINE | ID: mdl-33944659

ABSTRACT

Lymph node (LN) is a key tissue in the pathophysiology of mature blood cancers, especially for chronic lymphocytic leukemia (CLL). Within the multiple de-regulated pathways affecting CLL homeostasis, the CC-chemokine receptor 7 (CCR7) grants homing of CLL cells into the LN where protective environments foster tumor progression. To cover the lack of specific therapies targeting the CCR7-dependence of CLL to enter into the LN, and aiming to displace the disease from LN, we generated CAP-100, an antibody that specifically binds to hCCR7 and neutralizes its ligand-binding site and signaling. In various in vitro and in vivo preclinical models CAP-100 strongly inhibited CCR7-induced migration, extravasation, homing, and survival in CLL samples. Moreover, it triggered potent tumor cell killing, mediated by host immune mechanisms, and was effective in xenograft models of high-risk disease. Additionally, CAP-100 showed a favorable toxicity profile on relevant hematopoietic subsets. Our results validated CAP-100 as a novel therapeutic tool to prevent the access of CLL cells, and other neoplasia with nodal-dependence, into the LN niches, thus hitting a central hub in the pathogenesis of cancer. The first-in-human clinical trial (NCT04704323), which will evaluate this novel therapeutic approach in CLL patients, is pending.


Subject(s)
Antibodies, Blocking/pharmacology , Chemotaxis, Leukocyte/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphatic Metastasis/pathology , Receptors, CCR7/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Humans , Immunoglobulin G/pharmacology , Macaca fascicularis , Mice
17.
Vet Immunol Immunopathol ; 237: 110269, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34023617

ABSTRACT

Pectin is a dietary fibre composed of galacturonic acid, primarily found in the citrus fruits' cell walls. Citrus pectin (CP) has demonstrated antioxidative, anticancer, and anti-inflammatory properties in humans and animals. In broilers, CP supplementation improves energy utilization and nutrient digestibility, but limited information on its effects on chicken immunity is available so far. This study aimed to assess the in vitro impact of CP on chicken monocytes' immune response. Cells were purified from whole blood of healthy chickens and incubated with increasing concentrations (0, 0.25, 0.5, 0.75, 1 mg/mL) of CP to determine CP working concentration. The effects of different CP concentrations on cells' apoptosis and viability were assessed by measuring caspase-3 and -7 and the cells' metabolic activity (MTT assay), respectively. CP had no dose-dependent effect on monocyte apoptosis and viability.Then, the effects of CP (0.5 mg/mL) on chicken monocytes' chemotaxis and phagocytosis were assessed by measuring transwell migration and fluorescein-labelled E. coli incorporation, respectively. CP inhibited both monocytes' chemotaxis and phagocytosis.These data demonstrate that CP exerts an immunomodulatory role in chicken monocytes, supporting its integration in nutrition strategies that might be beneficial for the animal's immunity and health.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Citrus/chemistry , Monocytes/drug effects , Pectins/pharmacology , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Chickens , Immunologic Factors/pharmacology , Monocytes/immunology , Phagocytosis/drug effects
18.
Front Immunol ; 12: 628090, 2021.
Article in English | MEDLINE | ID: mdl-33841411

ABSTRACT

The ability to study migratory behavior of immune cells is crucial to understanding the dynamic control of the immune system. Migration induced by chemokines is often assumed to be directional (chemotaxis), yet commonly used end-point migration assays are confounded by detecting increased cell migration that lacks directionality (chemokinesis). To distinguish between chemotaxis and chemokinesis we used the classic "under-agarose assay" in combination with video-microscopy to monitor migration of CCR7+ human monocyte-derived dendritic cells and T cells in response to a concentration gradient of CCL19. Formation of the gradients was visualized with a fluorescent marker and lasted several hours. Monocyte-derived dendritic cells migrated chemotactically towards the CCL19 gradient. In contrast, T cells exhibited a biased random walk that was largely driven by increased exploratory chemokinesis towards CCL19. This dominance of chemokinesis over chemotaxis in T cells is consistent with CCR7 ligation optimizing T cell scanning of antigen-presenting cells in lymphoid tissues.


Subject(s)
Chemokine CCL19/pharmacology , Chemotaxis, Leukocyte/drug effects , Dendritic Cells/drug effects , Microscopy, Fluorescence , T-Lymphocytes/drug effects , Time-Lapse Imaging , Cell Communication , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Humans , T-Lymphocytes/immunology , Time Factors
19.
Sci Rep ; 11(1): 8207, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859291

ABSTRACT

Methamphetamine (METH) is a strong addictive central nervous system stimulant. METH abuse can alter biological processes and immune functions necessary for host defense. The acquisition and transmission of HIV, hepatitis, and other communicable diseases are possible serious infectious consequences of METH use. METH also accumulates extensively in major organs. Despite METH being a major public health and safety problem globally, there are limited studies addressing the impact of this popular recreational psychostimulant on tissue adaptive immune responses after exposure to T cell dependent [ovalbumin (OVA)] and independent [lipopolysaccharide (LPS)] antigens. We hypothesized that METH administration causes pulmonary and splenic tissue alterations and reduces T cell responses to OVA and LPS in vivo, suggesting the increased susceptibility of users to infection. Using a murine model of METH administration, we showed that METH causes tissue injury, apoptosis, and alters helper and cytotoxic T cell recruitment in antigen challenged mice. METH also reduces the expression and distribution of CD3 and CD28 molecules on the surface of human Jurkat T cells. In addition, METH decreases the production of IL-2 in these T-like cells, suggesting a negative impact on T lymphocyte activation and proliferation. Our findings demonstrate the pleotropic effects of METH on cell-mediated immunity. These alterations have notable implications on tissue homeostasis and the capacity of the host to respond to infection.


Subject(s)
Lung Injury/chemically induced , Methamphetamine/pharmacology , Splenic Diseases/chemically induced , T-Lymphocytes/drug effects , Amphetamine-Related Disorders/immunology , Amphetamine-Related Disorders/pathology , Animals , Antigens, Bacterial , Apoptosis/drug effects , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/physiology , Disease Models, Animal , Female , Humans , Inflammation/chemically induced , Inflammation/immunology , Inflammation/pathology , Jurkat Cells , Lipopolysaccharides , Lung/drug effects , Lung/immunology , Lung/pathology , Lung Injury/immunology , Lung Injury/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Spleen/drug effects , Spleen/immunology , Spleen/injuries , Spleen/pathology , Splenic Diseases/immunology , Splenic Diseases/pathology , T-Lymphocytes/physiology
20.
Front Immunol ; 12: 634368, 2021.
Article in English | MEDLINE | ID: mdl-33717174

ABSTRACT

Immunological rejection is an important factor resulting in allograft dysfunction, and more valid therapeutic methods need to be explored to improve allograft outcomes. Many researches have indicated that artemisinin and its derivative exhibits immunosuppressive functions, apart from serving as a traditional anti-malarial drug. In this assay, we further explored the therapeutic effects of artemisinin for transplant rejection in a rat cardiac transplantation model. We found that it markedly attenuated allograft rejection and histological injury and significantly prolonged the survival of allograft. Upon further exploring the mechanism, we demonstrated that artemisinin not only attenuated T cell-mediated rejection (TCMR) by reducing effector T cell infiltration and inflammatory cytokine secretion and increasing regulatory T cell infiltration and immunoregulatory cytokine levels, but also attenuated antibody-mediated rejection (ABMR) through inhibition of B cells activation and antibody production. Furthermore, artemisinin also reduced macrophage infiltration in allografts, which was determined to be important for TCMR and ABMR. Moreover, we demonstrated that artemisinin significantly inhibited the function of pure T cells, B cells, and macrophages in vitro. All in all, this study provide evidence that artemisinin significantly attenuates TCMR and ABMR by targeting multiple effectors. Therefore, this agent might have potential for use in clinical settings to protect against transplant rejection.


Subject(s)
Artemisinins/pharmacology , B-Lymphocytes/drug effects , Graft Rejection/prevention & control , Graft Survival/drug effects , Heart Transplantation/adverse effects , Immunosuppressive Agents/pharmacology , Skin Transplantation/adverse effects , T-Lymphocytes/drug effects , Adolescent , Adult , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Cytokines/metabolism , Female , Graft Rejection/immunology , Graft Rejection/metabolism , Humans , Isoantibodies/blood , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Phenotype , Rats, Inbred BN , Rats, Inbred Lew , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...