Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 904
1.
J Alzheimers Dis ; 80(3): 973-977, 2021.
Article En | MEDLINE | ID: mdl-33612548

There is a dire need for due innovative therapeutic modalities to improve outcomes of AD patients. In this study, we tested whether cannabidiol (CBD) improves outcomes in a translational model of familial AD and to investigate if CBD regulates interleukin (IL)-33 and triggering receptor expressed on myeloid cells 2 (TREM2), which are associated with improved cognitive function. CBD was administered to 5xFAD mice, which recapitulate early onset, familial AD. Behavioral tests and immunoassays were used to evaluate cognitive and motor outcomes. Our findings suggest that CBD treatment enhanced IL-33 and TREM2 expression, ameliorated the symptoms of AD, and retarded cognitive decline.


Alzheimer Disease/metabolism , Cannabidiol/pharmacology , Cognition/drug effects , Interleukin-33/drug effects , Membrane Glycoproteins/drug effects , Receptors, Immunologic/drug effects , Alzheimer Disease/pathology , Animals , Disease Models, Animal , Humans , Interleukin-33/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Receptors, Immunologic/metabolism , Up-Regulation
2.
Expert Opin Ther Pat ; 31(6): 549-561, 2021 Jun.
Article En | MEDLINE | ID: mdl-33507843

Introduction: Triggering receptors expressed on myeloid cells (TREMs) are inflammatory amplifiers with defined pathophysiological role in various infectious diseases, acute and chronic aseptic inflammations, and a variety of cancers, depicting TREMs as prominent therapeutic targets.Areas covered: Herein, updates from 2015 to 2020 are discussed to divulge the TREM ligands, as well as their peptide blockers, claimed to modulate their expression. The article also presents different strategies employed during the last five years to block interactions between TREMs and their ligands to treat various disease conditions by modulating their expression and activity.Expert opinion: There has been significant progress in the discovery of novel ligands and modulators of TREMs in the last five years that mainly revolved around the function of TREM molecules. A few peptides showed encouraging results to modulate the expression and activity of TREMs in preclinical studies, and these peptides are currently under clinical investigation. Based on the findings so far in several careful studies, we expect novel therapeutics in the near future which could have the ability to treat various disease conditions associated with TREM expression.


Membrane Glycoproteins/drug effects , Molecular Targeted Therapy , Receptors, Immunologic/drug effects , Triggering Receptor Expressed on Myeloid Cells-1/drug effects , Animals , Drug Development , Drug Discovery , Humans , Ligands , Membrane Glycoproteins/metabolism , Patents as Topic , Receptors, Immunologic/metabolism , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
5.
Osteoarthritis Cartilage ; 28(5): 603-612, 2020 05.
Article En | MEDLINE | ID: mdl-31730805

OBJECTIVE: A number of studies have demonstrated that molecules called 'alarmins' or danger-associated molecular patterns (DAMPs), contribute to inflammatory processes in the OA joint. Metabolic reprogramming of immune cells, including macrophages, is emerging as a prominent player in determining immune cell phenotype and function. The aim of this study was to investigate if basic calcium phosphate (BCP) crystals which are OA-associated DAMPs, impact on macrophage phenotype and metabolism. METHODS: Human monocyte derived macrophages were treated with BCP crystals and expression of M1 (CXCL9, CXCL10) and M2 (MRC1, CCL13)-associated markers was assessed by real-time PCR while surface maturation marker (CD40, CD80 & CD86) expression was assessed by flow cytometry. BCP induced metabolic changes were assessed by Seahorse analysis and glycolytic marker expression (hexokinase 2(HK2), Glut1 and HIF1α) was examined using real-time PCR and immunoblotting. RESULTS: Treatment with BCP crystals upregulated mRNA levels of CXCL9 and CXCL10 while concomitantly downregulating expression of CCL13 and MRC1. Furthermore, BCP-treated macrophages enhanced surface expression of the maturation makers, CD40, CD80 and CD86. BCP-treated cells also exhibited a shift towards glycolysis as evidenced by an increased ECAR/OCR ratio and enhanced expression of the glycolytic markers, HK2, Glut1 and HIF1α. Finally, BCP-induced macrophage activation and alarmin expression was reduced in the presence of the glycolytic inhibitor, 2-DG. CONCLUSIONS: This study not only provides further insight into how OA-associated DAMPs impact on immune cell function, but also highlights metabolic reprogramming as a potential therapeutic target for calcium crystal-related arthropathies.


Calcium Phosphates/pharmacology , Cytokines/drug effects , Glycolysis/drug effects , Macrophages/drug effects , Osteoarthritis/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD40 Antigens/metabolism , Chemokine CXCL10/drug effects , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL9/drug effects , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Cytokines/genetics , Down-Regulation , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/genetics , Hexokinase/drug effects , Hexokinase/genetics , Hexokinase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monocyte Chemoattractant Proteins/drug effects , Monocyte Chemoattractant Proteins/genetics , Monocyte Chemoattractant Proteins/immunology , Osteoarthritis/genetics , Phenotype , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Immunologic/drug effects , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Up-Regulation
6.
Front Immunol ; 10: 2677, 2019.
Article En | MEDLINE | ID: mdl-31824485

Dendritic cells (DC), which are involved in orchestrating early immune responses against pathogens, are dysregulated in their function by HIV infection. This dysregulation likely contributes to tip the balance toward viral persistence. Different DC subpopulations, including classical (cDCs) and plasmacytoid (pDCs) dendritic cells, are subjected to concomitant inflammatory and immunoregulatory events during HIV infection, which hampers the precise characterization of their regulation through classical approaches. Here, we carried out mass cytometry analysis of blood samples from early HIV-infected patients that were longitudinally collected before and after 1 year of effective combination antiretroviral therapy (cART). Blood samples from HIV controller patients who naturally control the infection were also included. Our data revealed that plasma HIV RNA level was positively associated with a loss of cDC and pDC subpopulations that display high expression of LILR immunomodulatory receptors. Conversely, specific monocyte populations co-expressing high levels of HLA-I, 3 immunomodulatory receptors, CD64, LILRA2, and LILRB4, and the restriction factor CD317 (also known as BST2/Tetherin), were more abundant in early HIV-infection. Finally, our analysis revealed that the blood of HIV controller patients contained in a higher abundance a particular subtype of CD1c+ cDCs, characterized by elevated co-expression of CD32b inhibitory receptor and HLA-DR antigen-presentation molecules. Overall, this study unravels the modifications induced in DC and monocyte subpopulations in different HIV+ conditions, and provides a better comprehension of the immune regulation/dysregulation mechanisms induced during this viral infection.


Dendritic Cells/immunology , HIV Infections/immunology , Monocytes/immunology , Adult , Anti-HIV Agents/therapeutic use , Dendritic Cells/drug effects , Female , Flow Cytometry , HIV Infections/drug therapy , HIV-1/immunology , Humans , Male , Middle Aged , Monocytes/drug effects , Receptors, Immunologic/drug effects , Receptors, Immunologic/immunology
7.
Hypertension ; 74(6): 1499-1506, 2019 12.
Article En | MEDLINE | ID: mdl-31587572

This study tested whether brain L-PGDS (lipocalin-type prostaglandin [PG] D synthase), through prostanoid signaling, might increase neurogenic pressor activity and thereby cause hypertension. Sprague Dawley rats on high-salt diet received either vehicle or Ang II (angiotensin II) infusion. On day 4, the developmental stage of hypertension, brains from different sets of control and Ang II-treated rats were collected for measuring L-PGDS expression, PGD2 levels, and DP1R (type 1 PGD2 receptor) expression. In a different set of 14-day Ang II-salt-treated rats, mini-osmotic pumps were used to infuse either a nonselective COX (cyclooxygenase) inhibitor ketorolac, L-PGDS inhibitor AT56, or DP1R inhibitor BWA868C to test the role of brain COX-PGD2-DP1R signaling in Ang II-salt hypertension. The acute depressor response to ganglion blockade with hexamethonium was used to quantify neurogenic pressor activity. During the developmental stage of Ang II-salt hypertension, L-PGDS expression was higher in cerebrospinal fluid, and PGD2 levels were increased in the choroid plexus, cerebrospinal fluid, and the cardioregulatory brain region rostral ventrolateral medulla. DP1R expression was decreased in rostral ventrolateral medulla. Both brain COX inhibition with ketorolac and L-PGDS inhibition with AT56 lowered mean arterial pressure by altering neurogenic pressor activity compared with vehicle controls. Blockade of DP1R with BWA868C, however, increased the magnitude of Ang II-salt hypertension and significantly increased neurogenic pressor activity. In summary, we establish that the development of Ang II-salt hypertension requires increased COX- and L-PGDS-derived PGD2 production in the brain, making L-PGDS a possible target for treating neurogenic hypertension.


Angiotensin II/pharmacology , Arterial Pressure/physiology , Brain/diagnostic imaging , Hypertension/physiopathology , Ketorolac/pharmacology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Animals , Arterial Pressure/drug effects , Biomarkers/blood , Brain/drug effects , Brain/metabolism , Chromatography, Liquid/methods , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Hypertension/drug therapy , Hypertension/metabolism , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Immunologic/drug effects , Receptors, Prostaglandin/drug effects , Reference Values , Risk Assessment , Sodium Chloride, Dietary/administration & dosage , Sodium Chloride, Dietary/metabolism , Tandem Mass Spectrometry/methods
8.
Neuropharmacology ; 158: 107727, 2019 11 01.
Article En | MEDLINE | ID: mdl-31356825

Apoptosis following hypoxic-ischemic injury to the brain plays a major role in neuronal cell death. The neonatal brain is more susceptible to injury as the cortical neurons are immature and there are lower levels of antioxidants. Slit2, an extracellular matrix protein, has been shown to be neuroprotective in various models of neurological diseases. However, there is no information about the role of Slit2 in neonatal hypoxia-ischemia. In this study, we evaluated the effect of Slit2 and its receptor Robo1 in a rat model with neonatal HIE. 10-day old rat pups were used to create the neonatal HIE model. The right common carotid artery was ligated followed by 2.5 h of hypoxia. Recombinant Slit2 was administered intranasally 1 h post HI, recombinant Robo1 was used as a decoy receptor and administered intranasally 1h before HI and srGAP1-siRNA was administered intracerebroventricularly 24 h before HI. Brain infarct area measurement, short-term and long-term neurological function tests, Western blot, immunofluorescence staining, Fluoro-Jade C staining, Nissl staining and TUNEL staining were the assessments done following drug administration. Recombinant Slit2 administration reduced neuronal apoptosis and neurological deficits after neonatal HIE which were reversed by co-administration of recombinant Robo1 and srGAP1-siRNA administration. Recombinant Slit2 showed improved outcomes possibly via the robo1-srGAP1 pathway which mediated the inhibition of RhoA. In this study, the results suggest that Slit2 may help in attenuation of apoptosis and could be a therapeutic agent for treatment of neonatal hypoxic ischemic encephalopathy.


Apoptosis/drug effects , GTPase-Activating Proteins/drug effects , Hypoxia-Ischemia, Brain/physiopathology , Intercellular Signaling Peptides and Proteins/pharmacology , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/pharmacology , Neurons/drug effects , Receptors, Immunologic/drug effects , Administration, Intranasal , Animals , Animals, Newborn , GTPase-Activating Proteins/metabolism , Hypoxia-Ischemia, Brain/metabolism , In Situ Nick-End Labeling , Injections, Intraventricular , Nerve Tissue Proteins/metabolism , RNA, Small Interfering , Rats , Receptors, Immunologic/metabolism , Recombinant Proteins , Signal Transduction , rho GTP-Binding Proteins/drug effects , rho GTP-Binding Proteins/metabolism , Roundabout Proteins
9.
Gastroenterology ; 156(2): 311-324, 2019 01.
Article En | MEDLINE | ID: mdl-30243618

Hepatitis B virus (HBV) infection affects approximately 300 million people worldwide. Although antiviral therapies have improved the long-term outcomes, patients often require life-long treatment and there is no cure for HBV infection. New technologies can help us learn more about the pathogenesis of HBV infection and develop therapeutic agents to reduce its burden. We review recent advances in development of direct-acting antiviral and host-targeting agents, some of which have entered clinical trials. We also discuss strategies for unbiased high-throughput screens to identify compounds that inhibit HBV and for repurposing existing drugs.


Antiviral Agents/pharmacology , Drug Development , Hepatitis B/therapy , Capsid/drug effects , Humans , Immunologic Factors/pharmacology , Receptors, Immunologic/drug effects , Receptors, Virus/drug effects , Virus Replication/drug effects
10.
J Cell Physiol ; 234(6): 8541-8549, 2019 06.
Article En | MEDLINE | ID: mdl-30511409

Among the main promising systems to triggering therapeutic antitumor immunity is the blockade of immune checkpoints. Immune checkpoint pathways regulate the control and eradication of infections, malignancies, and resistance against a host of autoantigens. Initiation point of the immune response is T cells, which have a critical role in this pathway. As several immune checkpoints are initiated by ligand-receptor interactions, they can be freely blocked by antibodies or modulated by recombinant forms of ligands or receptors. Antibodies against cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) were the first immunotherapeutics that achieved the US Food and Drug Administration approval. Preliminary clinical results with the blockers of additional immune checkpoint proteins, such as programmed cell death protein 1 (PD-1) indicate extensive and different chances to boost antitumor immunity with the objective of conferring permanent clinical effects. This study provides an overview of the immune checkpoint pathways, including CTLA-4, PD-1, lymphocyte activation gene 3, T-cell immunoglobulin and mucin domain 3, B7-H3, and diacylglycerol kinase α and implications of their inhibition in the cancer therapy.


Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy , Neoplasms/drug therapy , Receptors, Immunologic/drug effects , Animals , Antineoplastic Agents, Immunological/adverse effects , Drug Resistance, Neoplasm , Humans , Immunotherapy/adverse effects , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction , Tumor Microenvironment
11.
Sci Rep ; 8(1): 13543, 2018 09 10.
Article En | MEDLINE | ID: mdl-30201974

T lymphocytes have a crucial role in initiating and promoting type I allergies. Their responses are tightly regulated by numerous activating and inhibitory signals provided by APCs. Here we have addressed the role of the major coinhibitory receptors PD-1, CTLA-4, BTLA and LAG-3 in allergen-specific CD4+ T cell responses. PBMCs of healthy individuals and 41 patients allergic to house dust mites, birch, grass or mugwort pollen were stimulated with allergenic extracts and expression of coinhibitory receptors on responding CD4+ T cells was assessed. Blocking antibodies to PD-1, CTLA-4, BTLA and LAG-3 were used to evaluate the role of coinhibitory pathways. Allergen-specific CD4+ T cells showed strong upregulation of PD-1, LAG-3 and CTLA-4 upon stimulation, whereas BTLA was downregulated. Blockade of PD-1 strongly enhanced proliferation and cytokine production (IL-10; TH1 cytokines IFN-γ, TNF-α; TH2 cytokines IL-5, IL-13) of allergen-specific CD4+ T cells derived from allergic as well as non-allergic individuals. BTLA blockade enhanced proliferation but not cytokine production in response to house dust mite extract. Blocking LAG-3 was ineffective and surprisingly, we observed reduced proliferation and cytokine production in presence of a CTLA-4 antibody. Our results point to a unique potency of PD-1 pathways to dampen allergen-specific human T cells.


Allergens/immunology , CD4-Positive T-Lymphocytes/immunology , Hypersensitivity/immunology , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/immunology , Adult , Antigens, CD/drug effects , Antigens, CD/immunology , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/drug effects , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Proliferation/drug effects , Cohort Studies , Cytokines/immunology , Cytokines/metabolism , Down-Regulation/immunology , Female , Healthy Volunteers , Humans , Hypersensitivity/blood , Hypersensitivity/drug therapy , Lymphocyte Activation , Male , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/drug effects , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Up-Regulation/immunology , Lymphocyte Activation Gene 3 Protein
12.
Nutrients ; 10(7)2018 Jul 14.
Article En | MEDLINE | ID: mdl-30011891

Immunomodulatory protein hydrolysate consumption may delay or prevent western immune-related diseases. In order to purposively develop protein hydrolysates with an optimal and reproducible immunomodulatory effect, knowledge is needed on which components in protein hydrolysates are responsible for the immune effects. Important advances have been made on this aspect. Also, knowledge on mechanisms underlying the immune modulating effects is indispensable. In this review, we discuss the most promising application possibilities for immunomodulatory protein hydrolysates. In order to do so, an overview is provided on reported in vivo immune effects of protein hydrolysates in both local intestinal and systemic organs, and the current insights in the underlying mechanisms of these effects. Furthermore, we discuss current knowledge and physicochemical approaches to identify the immune active protein sequence(s). We conclude that multiple hydrolysate compositions show specific immune effects. This knowledge can improve the efficacy of existing hydrolysate-containing products such as sports nutrition, clinical nutrition, and infant formula. We also provide arguments for why immunomodulatory protein hydrolysates could be applied to manage the immune response in the increasing number of individuals with a higher risk of immune dysfunction due to, for example, increasing age or stress.


Dietary Supplements , Immune System/drug effects , Immunologic Factors/administration & dosage , Protein Hydrolysates/administration & dosage , Animals , Dietary Supplements/adverse effects , Endocytosis , Humans , Immune System/immunology , Immune System/metabolism , Immunity, Mucosal/drug effects , Immunologic Factors/adverse effects , Immunologic Factors/immunology , Immunologic Factors/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Intestines/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/metabolism , Peptide Transporter 1/metabolism , Protein Hydrolysates/adverse effects , Protein Hydrolysates/immunology , Protein Hydrolysates/metabolism , Receptors, Immunologic/drug effects , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism
13.
CNS Neurol Disord Drug Targets ; 16(7): 749-762, 2017.
Article En | MEDLINE | ID: mdl-28745241

BACKGROUND & OBJECTIVE: Epilepsy is one of the most complex neurological disorders and its study requires a broad knowledge of neurology and neuroscience. It comprises a diverse group of neurological disorders that share the central feature of spontaneous recurrent seizures, and are often accompanied by cognitive deficits and mood disorder. This condition is one of the most common neurological disorders. Until recently, alterations of neuronal activities had been the focus of epilepsy research. This neurocentric emphasis did not address issues that arise in more complex models of epileptogenesis. An important factor in epilepsy that is not regulated directly by neurons is inflammation and the immune response of the brain. Recent evidence obtained in rodent epilepsy models supports the role of immune responses in the initiation and maintenance of epilepsy. Recognition of exogenous pathogens by the innate immune system is mediated by some pattern recognition receptors such as Toll-like receptors leading to cell activation and cytokine production. Currently, these receptors have been the focus of epilepsy studies looking to determine whether the innate immune activation is neuroprotective or neurotoxic for the brain. CONCLUSION: Here, we present the evidence in the literature of the involvement of key innate immune receptors in the development of epilepsy. We address some of the contradictory findings in these studies and also mention possible avenues for research into epilepsy treatments that target these receptors.


Epilepsy/physiopathology , Immunity, Innate/physiology , Receptors, Immunologic/physiology , Animals , Humans , Molecular Targeted Therapy/methods , Receptors, Immunologic/drug effects
14.
Postepy Hig Med Dosw (Online) ; 71(0): 352-358, 2017 May 09.
Article En | MEDLINE | ID: mdl-28513459

The antimicrobial function of neutrophils, which is dependent on opsonin receptors, deteriorates in severe acute pancreatitis (SAP). Granulocyte colony-stimulating factor (G-CSF) putatively enhanced levels of the opsonin receptors CD11b and CD32/16 in healthy human subjects, and provided protection against infection in animal models of SAP. A statistically convincing study of the effect of G-CSF on CD32/16 expression in an SAP model is lacking. We used a mouse model of SAP to investigate the association between G-CSF administration and CD32/16 levels on neutrophils and bacterial translocation. G-CSF or saline was subcutaneously injected into SAP-induced mice. The pancreases were histologically examined, and leukocytes were stained to count neutrophils. The expression of CD11b and CD32/16 on neutrophils was measured by flow cytometry, and bacterial translocation was observed by bacterial culture. The numbers of CD11b and CD32/16-positive neutrophils were significantly elevated in the SAP mice treated with G-CSF, and the mean fluorescence intensities of these receptors on neutrophils were significantly elevated. Bacterial translocations to cavity organs were suppressed from 17% to 6% by G-CSF treatment. Our results indicated that the number of neutrophils significantly increased with increasing expression of CD11b and CD32/16 and their mean fluorescence intensities (MFIs). This inhibited bacterial translocation to other organs. These results are in accord with other studies in SAP dogs and SAP mice. Our findings suggest that G-CSF was effective in protecting against bacterial infection in SAP mice.


Disease Models, Animal , Granulocyte Colony-Stimulating Factor/therapeutic use , Neutrophil Activation/drug effects , Pancreatitis/physiopathology , Receptors, Immunologic/drug effects , Acute Disease , Animals , Bacterial Infections/drug therapy , Bacterial Infections/immunology , Female , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/pharmacology , Mice , Pancreatitis/drug therapy , Pancreatitis/immunology , Receptors, Immunologic/genetics
15.
J Clin Pharmacol ; 57(2): 161-172, 2017 02.
Article En | MEDLINE | ID: mdl-27402064

We report pharmacokinetics, pharmacodynamics, and safety of a novel anti-CD28 domain antibody antagonist (lulizumab pegol) in healthy subjects following single- or multiple-dose administration. A minimal anticipated biological effect level approach was used to select a 0.01 mg starting dose for a single-ascending-dose (SAD), double-blind, first-in-human study. Part 1 included 9 intravenous (IV; 0.01-100 mg) and 3 subcutaneous (SC; 9-50 mg) doses or placebo. In part 2, a keyhole limpet hemocyanin (KLH) immunization was performed in 16 subjects/panel, who received 1 of 3 IV doses (9-100 mg) or placebo. In a double-blind, multiple-ascending-dose (MAD) study, subjects received SC lulizumab 6.25 mg every 2 weeks, 12.5 mg weekly, 37.5 mg weekly, or placebo. Among 180 treated subjects, 169 completed the studies. Peak concentrations and areas under the curve from time 0 to infinity increased dose proportionally. Estimated SC bioavailability was 68.2%. Receptor occupancy of approximately ≥80% was maintained for ≥2 weeks at ≥9-mg doses (SAD) and throughout the dosing interval (MAD). IV doses ≥9 mg inhibited antibody production against KLH for 2 weeks. No significant cytokine or immune cell changes were observed. No immunogenicity responses persisted, and there was no correlation to adverse events. Headache occurred in 21 SAD and 4 MAD subjects receiving lulizumab; in the MAD study 5 lulizumab subjects experienced infections. Lulizumab IV or SC was safe at all doses studied, without evidence of cytokine release.


Antibodies, Blocking/metabolism , CD28 Antigens/immunology , Polyethylene Glycols/pharmacokinetics , Administration, Intravenous , Adolescent , Adult , Antibodies/adverse effects , Antibodies, Blocking/adverse effects , Biological Availability , Cytokines/blood , Dose-Response Relationship, Drug , Double-Blind Method , Female , Headache/chemically induced , Healthy Volunteers , Hemocyanins/immunology , Humans , Injections, Subcutaneous , Male , Middle Aged , Polyethylene Glycols/adverse effects , Receptors, Immunologic/drug effects , Young Adult
16.
Drug Discov Ther ; 11(6): 329-335, 2017.
Article En | MEDLINE | ID: mdl-29332891

Phytohemagglutinin (PHA) isolated from the family of Phaseolus vulgaris beans is a promising agent against viral infection; however, it has not yet been demonstrated in vivo. We herein investigated this issue using Drosophila as a host. Adult flies were fed lectin approximately 12 h before they were subjected to a systemic viral infection. After a fatal infection with Drosophila C virus, death was delayed and survival was longer in flies fed PHA-P, a mixture of L4, L3E1, and L2E2, than in control unfed flies. We then examined PHA-L4, anticipating subunit L as the active form, and confirmed the protective effects of this lectin at markedly lower concentrations than PHA-P. In both experiments, lectin feeding reduced the viral load prior to the onset of fly death. Furthermore, we found a dramatic increase in the levels of the mRNAs of phagocytosis receptors in flies after feeding with PHA-L4 while a change in the levels of the mRNAs of antimicrobial peptides was marginal. We concluded that P. vulgaris PHA protects Drosophila against viral infection by augmenting the level of host immunity.


Dicistroviridae , Drosophila/drug effects , Mitogens/pharmacology , Phytohemagglutinins/pharmacology , Survival Rate , Virus Diseases/virology , Animals , Drosophila/genetics , Drosophila/virology , Phaseolus , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Immunologic/drug effects , Receptors, Immunologic/genetics , Viral Load/drug effects
17.
Am J Physiol Renal Physiol ; 312(4): F716-F731, 2017 04 01.
Article En | MEDLINE | ID: mdl-27558558

Increasing incidences of obesity and diabetes have made diabetic kidney disease (DKD) the leading cause of chronic kidney disease and end-stage renal disease worldwide. Despite current pharmacological treatments, including strategies for optimizing glycemic control and inhibitors of the renin-angiotensin system, DKD still makes up almost one-half of all cases of end-stage renal disease in the United States. Compelling and mounting evidence has clearly demonstrated that immunity and inflammation play a paramount role in the pathogenesis of DKD. This article reviews the involvement of the immune system in DKD and identifies important roles of key immune and inflammatory mediators. One of the most recently identified biomarkers is serum amyloid A, which appears to be relatively specific for DKD. Novel and evolving treatment approaches target protein kinases, transcription factors, chemokines, adhesion molecules, growth factors, advanced glycation end-products, and other inflammatory molecules. This is the beginning of a new era in the understanding and treatment of DKD, and we may have finally reached a tipping point in our fight against the growing burden of DKD.


Anti-Inflammatory Agents/therapeutic use , Diabetic Nephropathies/drug therapy , Drug Discovery/methods , Inflammation Mediators/antagonists & inhibitors , Kidney/drug effects , Nephritis/drug therapy , Receptors, Immunologic/drug effects , Animals , Biomarkers/metabolism , Diabetic Nephropathies/immunology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Molecular Targeted Therapy , Nephritis/immunology , Nephritis/metabolism , Nephritis/pathology , Predictive Value of Tests , Receptors, Immunologic/metabolism , Signal Transduction/drug effects
18.
Inflammation ; 40(1): 13-20, 2017 Feb.
Article En | MEDLINE | ID: mdl-27696333

Triggering receptors expressed on myeloid cell-1 (TREM-1) is a superimmunoglobulin receptor expressed on myeloid cells. TREM-1 amplifies the inflammatory response. Epoxyeicosatrienoic acids (EETs), the metabolites of arachidonic acid derived from the cytochrome P450 enzyme, have anti-inflammatory properties. However, the effects of EETs on TREM-1 expression under inflammatory stimulation remain unclear. Therefore, inhibition of soluble epoxide hydrolase (sEH) with a highly selective inhibitor [1-trifluoromethoxyphenyl-3-(1-propionylpiperidin-4-yl) urea, TPPU] was used to stabilize EETs. LPS was intratracheally injected into mice to induce pulmonary inflammation, after TPPU treatment for 3 h. Histological examination showed TPPU treatment-alleviated LPS-induced pulmonary inflammation. TPPU decreased TREM-1 expression, but not DAP12 or MyD88 expression. Murine peritoneal macrophages were challenged with LPS in vitro. We found that TPPU reduced LPS-induced TREM-1 expression in a dose-dependent manner, but not DAP12 or MyD88 expression. TPPU also decreased downstream signal from TREM-1, reducing pro-inflammatory cytokine TNF-α and IL-1ß mRNA expression. Furthermore, TPPU treatment inhibited IkB degradation in vivo and in vitro. Our results indicate that the inhibition of sEH suppresses LPS-induced TREM-1 expression and inflammation via inhibiting NF-kB activation in murine macrophage.


Epoxide Hydrolases/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Myeloid Cells/metabolism , NF-kappa B/metabolism , Receptors, Immunologic/metabolism , Animals , Cells, Cultured , Cytokines/drug effects , Cytokines/metabolism , Gene Expression/drug effects , I-kappa B Proteins/drug effects , I-kappa B Proteins/metabolism , Macrophages/metabolism , Membrane Glycoproteins/drug effects , Mice , Mice, Inbred C57BL , Phenylurea Compounds/pharmacology , Piperidines/pharmacology , Pneumonia/drug therapy , Receptors, Immunologic/drug effects , Solubility , Triggering Receptor Expressed on Myeloid Cells-1
19.
Int J Mol Sci ; 17(3): 375, 2016 Mar 15.
Article En | MEDLINE | ID: mdl-26999109

(1) BACKGROUND: A growing body of literature suggest that polymorphisms (SNPs) from inflammation-related genes could possibly play a role in cytokine production and then interact with dietary n-3 fatty acids (FAs) to modulate inflammation. The aim of the present study was to test whether gene expression of selected inflammatory genes was altered following an n-3 PUFA supplementation and to test for gene-diet interactions modulating plasma inflammatory biomarker levels. (2) METHODS: 191 subjects completed a 6-week n-3 FA supplementation with 5 g/day of fish oil. Gene expression of TNF-α and IL6 was assessed in peripheral blood mononuclear cells (PBMCs) using the TaqMan technology. Genotyping of 20 SNPs from the TNF-LTA gene cluster, IL1ß, IL6 and CRP genes was performed. (3) RESULTS: There was no significant reduction of plasma IL-6, TNF-α and C-reactive protein (CRP) levels after the 6-week fish oil supplementation. TNF-α and IL6 were slightly overexpressed in PBMCs after the supplementation (fold changes of 1.05 ± 0.38 and 1.18 ± 0.49, respectively (n = 191)), but relative quantification (RQ) within the -0.5 to 2.0 fold are considered as nonbiologically significant. In a MIXED model for repeated measures adjusted for the effects of age, sex and BMI, gene by supplementation interaction effects were observed for rs1143627, rs16944, rs1800797, and rs2069840 on IL6 levels, for rs2229094 on TNF-α levels and for rs1800629 on CRP levels (p < 0.05 for all). (4) CONCLUSIONS: This study shows that a 6-week n-3 FA supplementation with 5 g/day of fish oil did not alter gene expression levels of TNF-α and IL6 in PBMCs and did not have an impact on inflammatory biomarker levels. However, gene-diet interactions were observed between SNPs within inflammation-related genes modulating plasma inflammatory biomarker levels.


Dietary Supplements , Fatty Acids, Omega-3/pharmacology , Fish Oils/pharmacology , Inflammation/metabolism , Interleukin-6/genetics , Receptors, Immunologic/genetics , Tumor Necrosis Factor-alpha/genetics , Adolescent , Adult , Biomarkers/blood , Female , Fish Oils/chemistry , Gene Expression , Gene-Environment Interaction , Humans , Inflammation/blood , Inflammation/diet therapy , Interleukin-6/blood , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Immunologic/blood , Receptors, Immunologic/drug effects , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/drug effects , Young Adult
20.
J Pharmacol Exp Ther ; 357(2): 273-80, 2016 May.
Article En | MEDLINE | ID: mdl-26945085

Prostaglandin D2(PGD2) is involved in the pathogenesis of allergic rhinitis. However, the sensory nervous system-mediated contributions of PGD2to the symptoms of allergic rhinitis remain unclear. We investigated the involvement of PGD2in these symptoms and in neuronal excitation by in vivo and ex vivo experiments. In an ovalbumin-induced model of allergic rhinitis in guinea pigs, the number of sneezing, nasal rubbing, and nasal secretion events were assessed after the nasal cavity instillation of PGD2, histamine, or a combination of PGD2and histamine. In situ hybridization for PGD2receptor 1 (DP1) mRNA transcripts and immunohistochemical analysis of histamine H1receptor protein expression in guinea pig trigeminal ganglion (TRG) were performed. The effects of DP1receptor activation on the excitability of TRG neurons to electrical and histamine stimuli were assessed using whole-cell patch-clamp recordings. Histamine induced more sneezing, nasal rubbing, and nasal secretion events than PGD2 PGD2augmented histamine-induced responses, whereas pretreatment with a DP1receptor-selective antagonist completely suppressed PGD2-induced augmentation. DP1receptor mRNA transcripts and H1receptor protein expression could be detected in TRG neurons. Moreover, a DP1receptor agonist caused significant increases in the number of histamine-induced action potentials and depolarization, and reduced the current threshold in small-diameter neurons. Our findings show that PGD2-DP1receptor signaling augments the symptoms of allergic rhinitis via the sensory nervous system by modulating nasal neuronal activation to various stimuli, such as histamine. These findings suggest that DP1receptor antagonist has therapeutic potential for the treatment of allergic rhinitis.


Neurons/drug effects , Prostaglandin D2/pharmacology , Rhinitis, Allergic/chemically induced , Trigeminal Ganglion/drug effects , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Electric Stimulation , Guinea Pigs , Histamine/pharmacology , Male , Ovalbumin/immunology , Patch-Clamp Techniques , Receptors, Immunologic/drug effects , Receptors, Prostaglandin/drug effects , Rhinitis, Allergic/physiopathology , Rhinitis, Allergic/psychology , Sneezing/drug effects
...