Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cell Tissue Res ; 377(2): 245-257, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30919048

ABSTRACT

Despite extensive investigation focused on both the molecular characteristics and the expression level of Toll-like receptors (TLRs) during the inflammatory response in vertebrates, few data are available in the literature on the role of these proteins in invertebrate's immune response. Here, we propose the medicinal leech as a valuable model to better elucidate the role of TLR4 and its related products, such as tumor necrosis factor (TNF-α), after activation of the leech peripheral immune system with the endogenous medicinal leech recombinant allograft inflammatory factor-1 (rHmAIF-1) or with an exogenous stimulus, such as lipopolysaccharide (LPS). Our results indicate that activated macrophages (HmAIF-1+) and granulocytes (CD11b+) express both TLR4 and its coreceptor CD14. Moreover, functional studies performed by injecting a cyanobacterium selective TLR4 antagonist CyP demonstrated that only the TLR4 pathway was blocked, while the immune response caused by lipoteichoic acid (LTA) treatment is not affected. These results are consistent with literature on vertebrates, indicating that TLR4 functions as a LPS receptor while the recognition of LTA may involve other pathways.


Subject(s)
Disease Models, Animal , Granulocytes/immunology , Inflammation/immunology , Leeches , Macrophages/immunology , Toll-Like Receptor 4 , Animals , Calcium-Binding Proteins/immunology , Granulocytes/cytology , Leeching , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/cytology , Microfilament Proteins/immunology , Teichoic Acids/pharmacology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/physiology , Tumor Necrosis Factor-alpha/immunology
2.
Toxins (Basel) ; 10(7)2018 07 16.
Article in English | MEDLINE | ID: mdl-30012943

ABSTRACT

Lipopolysaccharide (LPS) from Porphyromonas gingivalis (Pg-LPS) is a key bacterial structure involved in the maintenance of a chronic pro-inflammatory environment during periodontitis. Similar to other gram-negative LPS, Pg-LPS induces the release of pro-inflammatory cytokines through interaction with Toll-Like Receptor 4 (TLR4) and is able to stimulate negative TLR4 regulatory pathways, such as those involving microRNA (miRNA). In this work, we employed CyP, an LPS with TLR4-MD2 antagonist activity obtained from the cyanobacterium Oscillatoria planktothrix FP1, to study the effects on pro-inflammatory cytokine production and miRNA expression in human monocytic THP-1 cells stimulated with Pg-LPS or E. coli LPS (Ec-LPS). Results showed that CyP inhibited TNF-α, IL-1ß and IL-8 expression more efficiently when co-incubated with Pg-LPS rather than with Ec-LPS. The inhibition of pro-inflammatory cytokine production was maintained even when CyP was added 2 h after LPS. The analysis of the effects of CyP on miRNA expression showed that, although being an antagonist, CyP did not inhibit miR-146a induced by Pg-LPS or Ec-LPS, whereas it significantly inhibited miR-155 only in the cultures stimulated with Ec-LPS. These results suggest that CyP may modulate the pro-inflammatory response induced by Pg-LPS, not only by blocking TLR4-MD2 complex, but also by preserving miR-146a expression.


Subject(s)
Cyanobacteria , Cytokines/metabolism , Lipopolysaccharides/antagonists & inhibitors , MicroRNAs/metabolism , Porphyromonas gingivalis , Cytokines/genetics , Escherichia coli , Humans , Lipopolysaccharides/pharmacology , THP-1 Cells
3.
Sci Rep ; 8(1): 11367, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054544

ABSTRACT

Endotoxin tolerance is a phenomenon characterized by a reduced capacity of monocytes and macrophages to respond to repeated stimulation with lipopolysaccharide (LPS) which has been suggested to represent a way of controlling the intensity and duration of innate immune response. During endotoxin tolerance, monocytes undergo functional re-programming primarily by epigenetic regulation. Recently, micro-RNA (miR)-146a has been demonstrated to be the major player of the negative regulation of the pro-inflammatory response, affecting TNF-α production. In this study, we have employed CyP, a cyanobacterial LPS antagonist acting on TLR4-MD2 complex, for priming human monocytes and evaluating their response to a subsequent challenge with E. coli LPS. Results show that CyP is able to induce cross-tolerance to E. coli LPS by inhibiting TNF-α production. The mechanism of action is mediated by a specific induction of miR-146a and reduction of IRAK1 and TRAF6 expressions in human monocytes by CyP priming. Up-regulation of miR-146a by CyP alone, affects subsequent cell response in term of TNF-α production even when monocytes are incubated with other TLR ligands, as lipoteichoic acid (LTA), thus confirming miR-146a as a critical player mediating TNF-α regulation during cross-tolerance with CyP.


Subject(s)
Cyanobacteria/metabolism , Endotoxins/toxicity , Immune Tolerance , Lipopolysaccharides/toxicity , MicroRNAs/metabolism , Down-Regulation/drug effects , Humans , Immune Tolerance/drug effects , Interleukin-1 Receptor-Associated Kinases/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , Monocytes/drug effects , Monocytes/metabolism , THP-1 Cells , TNF Receptor-Associated Factor 6/metabolism , Teichoic Acids/pharmacology , Tumor Necrosis Factor-alpha/biosynthesis
4.
Int J Inflam ; 2018: 2859135, 2018.
Article in English | MEDLINE | ID: mdl-29686833

ABSTRACT

Toll-Like Receptors (TLRs) are the innate immunity receptors that play an activating role when interacting with molecules released by bacteria and viruses (PAMPs, pathogen-associated molecular patterns) or with molecules released by injured cells and tissues (DAMPs, danger-associated molecular patterns). TLR triggering leads to the induction of proinflammatory cytokines and chemokines, driving the activation of both innate and adaptive immunity. In particular, Toll-Like Receptor 4 (TLR4) has been described to be involved in the inflammatory processes observed in several pathologies (such as ischemia/reperfusion injury, neuropathic pain, neurodegenerative diseases, and cancer). Molecules obtained by natural sources have been discovered to exert an anti-inflammatory action by targeting TLR4 activation pathways. This review focuses on TLR4 antagonists obtained from bacteria, cyanobacteria, and plants.

5.
J Neuroimmunol ; 313: 109-115, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153601

ABSTRACT

Increasing evidence supports the notion that the neurodegenerative process occurring in Alzheimer's disease (AD), Parkinson's disease (PD) and Amyotrophic Lateral Sclerosis (ALS) does not only imply the neuronal compartment but also involves a strong interaction with the immunological cells of the Central Nervous System (CNS), primarily microglia. Starting from the observation that the neurodegenerative disorders are frequent in elderly individuals, who have an immunological background that possibly favors this process, it is evident that a dysregulation of innate immune response triggered by misfolded and aggregated proteins, or by endogenous molecules released by injured neurons, directly contributes to disease pathogenesis and progression. There are important differences in the immunological processes occurring in AD, PD, ALS involving microglial function. Furthermore, although the contribution of adaptive immune cells in AD seems to be modest, in PD and especially in ALS models, T cells can influence microglial phenotype, inducing neuroprotection. A better understanding of the immunological mechanisms involved in the different phases of the neurodegenerative processes observed in AD, PD, ALS could effectively contribute to the development of new preventive and therapeutic strategies for such diseases.


Subject(s)
Central Nervous System/immunology , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/pathology , Animals , Humans
6.
Neurobiol Dis ; 99: 12-23, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27939857

ABSTRACT

We recently discovered that forebrain activation of the IL-1 receptor/Toll-like receptor (IL-1R1/TLR4) innate immunity signal plays a pivotal role in neuronal hyperexcitability underlying seizures in rodents. Since this pathway is activated in neurons and glia in human epileptogenic foci, it represents a potential target for developing drugs interfering with the mechanisms of epileptogenesis that lead to spontaneous seizures. The lack of such drugs represents a major unmet clinical need. We tested therefore novel therapies inhibiting the IL-1R1/TLR4 signaling in an established murine model of acquired epilepsy. We used an epigenetic approach by injecting a synthetic mimic of micro(mi)RNA-146a that impairs IL1R1/TLR4 signal transduction, or we blocked receptor activation with antiinflammatory drugs. Both interventions when transiently applied to mice after epilepsy onset, prevented disease progression and dramatically reduced chronic seizure recurrence, while the anticonvulsant drug carbamazepine was ineffective. We conclude that IL-1R1/TLR4 is a novel potential therapeutic target for attaining disease-modifications in patients with diagnosed epilepsy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anticonvulsants/administration & dosage , Epilepsy/therapy , MicroRNAs/administration & dosage , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Carbamazepine/pharmacology , Cyanobacteria , Dipeptides/administration & dosage , Disease Models, Animal , Epilepsy/drug therapy , Epilepsy/physiopathology , Hippocampus/physiopathology , Kainic Acid , Lipopolysaccharides/administration & dosage , Male , Mice, Inbred C57BL , Oligonucleotides/administration & dosage , Random Allocation , Receptors, Interleukin-1 Type I/metabolism , Time Factors , Toll-Like Receptor 4/metabolism , para-Aminobenzoates/administration & dosage
7.
Brain Behav Immun ; 60: 188-197, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27751869

ABSTRACT

BACKGROUND: Amyloid-ß oligomers (AßO) are species mainly involved in the synaptic and cognitive dysfunction in Alzheimer's disease. Although their action has been described mainly at neuronal level, it is now clear that glial cells govern synaptic activity in their resting state, contributing to new learning and memory establishment. In contrast, when activated, they may lead to synaptic and cognitive dysfunction. Using a reliable acute AßO-mediated mouse model of AD, we explored whether the memory alteration AßOs induce relies on the activation of glial cells, and if Toll-like receptor 4 (TLR4), pivotal in the initiation of an immune response, is involved. METHODS: C57 naïve mice were given a single intracerebroventricular injection of synthetic AßO-containing solution (1µM), which induces substantial impairment in the establishment of recognition memory. Then, first we assessed glial cell activation at different times post-injection by western blot, immunohistochemistry and ELISA in the hippocampus. After that we explored the efficacy of pre-treatment with anti-inflammatory drugs (indomethacin and an IL-1ß receptor antagonist) to prevent impairment in the novel object recognition task, and compared AßO's effects in TLR4 knockout mice. RESULTS: A single AßO injection rapidly activated glial cells and increased pro-inflammatory cytokine expression. Both anti-inflammatory drugs prevented the AßO-mediated impairment in memory establishment. A selective TLR4 receptor antagonist abolished AßO's action on memory, and in TLR4 knockout mice it had no effect on either memory or glial activation. CONCLUSIONS: These data provide new information on AßO's mechanism of action, indicating that besides direct action at the synapses, they also act through the immune system, with TLR4 playing a major role. This suggests that in a potential therapeutic setting inflammation must be considered as well.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Memory/drug effects , Microglia/metabolism , Toll-Like Receptor 4/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Neurons/metabolism , Synapses/metabolism
8.
Mediators Inflamm ; 2016: 6978936, 2016.
Article in English | MEDLINE | ID: mdl-27293318

ABSTRACT

Toll-like receptor 4 (TLR4) belongs to the family of pattern recognition receptors (PRRs). They are highly conserved receptors that recognize conserved pathogen-associated molecular patterns (PAMPs), thus representing the first line of defense against infections. TLR4 has been long recognized as the sensing receptor for gram-negative lipopolysaccharide (LPS). In addition, it also binds endogenous molecules produced as a result of tissue injury. Hence, TLR4 represents a key receptor on which both infectious and noninfectious stimuli converge to induce a proinflammatory response. TLR4-mediated inflammation, triggered by exogenous or endogenous ligands, is also involved in several acute and chronic diseases, having a pivotal role as amplifier of the inflammatory response. This review focuses on the research progress about the role of TLR4 activation in infectious and noninfectious (e.g., sterile) inflammation and the effects of TLR4 signaling in some pathological conditions.


Subject(s)
Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Animals , Humans , Receptors, Pattern Recognition/physiology , Signal Transduction/physiology
9.
Mol Med ; 18: 971-81, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22562723

ABSTRACT

Sustained inflammatory reactions are common pathological events associated with neuron loss in neurodegenerative diseases. Reported evidence suggests that Toll-like receptor 4 (TLR4) is a key player of neuroinflammation in several neurodegenerative diseases. However, the mechanisms by which TLR4 mediates neurotoxic signals remain poorly understood. We investigated the role of TLR4 in in vitro and in vivo settings of motor neuron degeneration. Using primary cultures from mouse spinal cords, we characterized both the proinflammatory and neurotoxic effects of TLR4 activation with lipopolysaccharide (activation of microglial cells, release of proinflammatory cytokines and motor neuron death) and the protective effects of a cyanobacteria-derived TLR4 antagonist (VB3323). With the use of TLR4-deficient cells, a critical role of the microglial component with functionally active TLR4 emerged in this setting. The in vivo experiments were carried out in a mouse model of spontaneous motor neuron degeneration, the wobbler mouse, where we preliminarily confirmed a protective effect of TLR4 antagonism. Compared with vehicle- and riluzole-treated mice, those chronically treated with VB3323 showed a decrease in microglial activation and morphological alterations of spinal cord neurons and a better performance in the paw abnormality and grip-strength tests. Taken together, our data add new understanding of the role of TLR4 in mediating neurotoxicity in the spinal cord and suggest that TLR4 antagonists could be considered in future studies as candidate protective agents for motor neurons in degenerative diseases.


Subject(s)
Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroprotective Agents/metabolism , Spinal Cord/pathology , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Cell Culture Techniques , Cell Shape/drug effects , Cell Survival/drug effects , Disease Models, Animal , Ligands , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Motor Neurons/drug effects , Muscles/drug effects , Muscles/pathology , Neurotoxins/toxicity , Spinal Cord/drug effects , Spinal Cord/metabolism , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Antimicrob Chemother ; 67(5): 1145-54, 2012 May.
Article in English | MEDLINE | ID: mdl-22334603

ABSTRACT

OBJECTIVES: In this study, the objective was to determine the anti-inflammatory properties of CyP, a cyanobacterial lipopolysaccharide (LPS) antagonist, used in combination with antibiotic chemotherapy during infection of an in vitro meningitis model infected with Neisseria meningitidis (meningococcus). METHODS: Monocultures of human meningioma cells and meningioma-primary human macrophage co-cultures were infected with meningococci (10(2)-10(8) cfu/monolayer) or treated with isolated outer membranes or purified LPS (0.1-100 ng/monolayer) from N. meningitidis. CyP (1-20 µg/monolayer) was added at intervals from t = 0 to 4 h, with and without benzylpenicillin (1-20 µg/monolayer). The antagonistic effect of CyP and its adjunctive properties to benzylpenicillin administration was determined by measuring cytokine levels in culture supernatants after 24 h. RESULTS: CyP significantly inhibited (P < 0.05) the secretion of interleukin (IL)-6, IL-8, monocyte chemoattractant protein (MCP)-1 and RANTES ('regulated upon activation, normal T cell expressed and secreted') (overall reduction levels from 50% to >95%) by meningioma cell lines and meningioma-macrophage co-cultures challenged with either live meningococci or bacterial components. Inhibition was effective when CyP was added within 2 h of challenge (P < 0.05) and was still pronounced by 4 h. In the co-culture model, CyP alone partially inhibited IL-1ß secretion, but did not prevent tumour necrosis factor (TNF)-α secretion, whereas penicillin alone inhibited IL-1ß and TNF-α but conversely did not reduce MCP-1 and RANTES secretion. However, coadministration of CyP and penicillin in both models had an additive effect and restored the overall inhibitory profile. CONCLUSIONS: CyP inhibits cytokine production in an in vitro meningitis model and augments the anti-inflammatory response when combined with benzylpenicillin. Administration of an LPS antagonist with antibiotic merits consideration in the emergency treatment of patients presenting with meningococcal infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cytokines/metabolism , Immunologic Factors/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Meningitis, Meningococcal/immunology , Neisseria meningitidis/immunology , Neisseria meningitidis/pathogenicity , Cells, Cultured , Coculture Techniques , Epithelial Cells/immunology , Humans , Macrophages/immunology , Meningitis, Meningococcal/physiopathology , Penicillin G/pharmacology
11.
Chemistry ; 17(25): 7053-60, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21557359

ABSTRACT

Pseudoalteromonas haloplanktis TAB 23 is a Gram-negative psychrophilic bacterium isolated from the Antarctic coastal sea. To survive in these conditions psychrophilic bacteria have evolved typical membrane lipids and "antifreeze" proteins to protect the inner side of the microorganism. As for Gram-negative bacteria, the outer membrane is mainly constituted by lipopoly- or lipooligosaccharides (LPS or LOS, respectively), which lean towards the external environment. Despite this, very little is known about the peculiarity of LPS from Gram-negative psychrophilic bacteria and what their role is in adaptation to cold temperature. Here we report the complete structure of the LOS from P. haloplanktis TAB 23. The lipid A was characterized by MALDI-TOF MS analysis and was tested in vitro showing a significant inhibitory effect on the LPS-induced pro-inflammatory cytokine production when added in culture with LPS from Escherichia coli. The product obtained after de-O-acylation of the LPS was analyzed by MALDI-TOF MS revealing the presence of several molecular species, differing in phosphorylation degree and oligosaccharide length. The oligosaccharide portion released after strong alkaline hydrolysis was purified by anion-exchange chromatography-pulsed amperometric detection (HPAEC-PAD) to give three main fractions, characterized by means of 2D NMR spectroscopy, which showed a very short highly phosphorylated saccharidic chain with the following general structure. α-Hepp3R,6R,4R'-(1→5)-α-Kdop4P-(2→6)-ß-GlcpN4R-(1→6)-α-GlcpN1P (R=-H(2)PO(3) or -H; R'=α-Galp-(1→4)-ß-Galp-(1→ or H-).


Subject(s)
Escherichia coli/chemistry , Lipid A/chemistry , Lipopolysaccharides/chemistry , Oligosaccharides/chemistry , Pseudoalteromonas/chemistry , Antarctic Regions , Carbohydrate Conformation , Cold Temperature , Lipid A/isolation & purification , Lipopolysaccharides/isolation & purification , Magnetic Resonance Spectroscopy , Models, Chemical , Molecular Sequence Data , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Int Immunopharmacol ; 11(2): 194-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21115122

ABSTRACT

CyP is a lipopolysaccharide (LPS)-like molecule extracted from the freshwater cyanobacterium Oscillatoria planktothrix FP1, which has been reported to be a potent competitive inhibitor of bacterial LPS. In the present study the ability of CyP to affect human polymorphonuclear leukocyte (PMN) function was investigated. PMNs were isolated from venous blood by standard density-gradient centrifugation. Cell migration was measured by use of the Boyden chamber assay. Interleukin (IL)-8 and tumor necrosis factor (TNF)-α production was measured using a sandwich-type enzyme-linked immunosorbent assay. PMN intracellular reactive oxygen species (ROS) levels were assessed by the use of a fluorescent probe coupled to spectrophotometry. CyP 10-100 µg/ml was chemotactic for PMNs without affecting the chemotactic response to either E. coli LPS or N-formyl-Met-Leu-Phe (fMLP). CyP per se did not affect PMN production of either IL-8 or TNF-α, but concentration-dependently reduced LPS-induced production of both cytokines. On the contrary, CyP had no effect either on fMLP-induced production of IL-8 or on PMN oxidative burst (at rest and after stimulation with fMLP), a response which is known to be independent from LPS-operated pathways. In human PMNs CyP behaves as a selective and effective LPS antagonist. These findings support the therapeutic potential of CyP in endotoxin-dependent disease.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Cyanobacteria/chemistry , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Neutrophils/drug effects , Calcium/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Humans , Interleukin-8/immunology , Lipopolysaccharides/isolation & purification , Neutrophils/immunology , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/immunology
13.
Nat Med ; 16(4): 413-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20348922

ABSTRACT

Brain inflammation is a major factor in epilepsy, but the impact of specific inflammatory mediators on neuronal excitability is incompletely understood. Using models of acute and chronic seizures in C57BL/6 mice, we discovered a proconvulsant pathway involving high-mobility group box-1 (HMGB1) release from neurons and glia and its interaction with Toll-like receptor 4 (TLR4), a key receptor of innate immunity. Antagonists of HMGB1 and TLR4 retard seizure precipitation and decrease acute and chronic seizure recurrence. TLR4-defective C3H/HeJ mice are resistant to kainate-induced seizures. The proconvulsant effects of HMGB1, like those of interleukin-1beta (IL-1beta), are partly mediated by ifenprodil-sensitive N-methyl-d-aspartate (NMDA) receptors. Increased expression of HMGB1 and TLR4 in human epileptogenic tissue, like that observed in the mouse model of chronic seizures, suggests a role for the HMGB1-TLR4 axis in human epilepsy. Thus, HMGB1-TLR4 signaling may contribute to generating and perpetuating seizures in humans and might be targeted to attain anticonvulsant effects in epilepsies that are currently resistant to drugs.


Subject(s)
HMGB1 Protein/physiology , Seizures/physiopathology , Toll-Like Receptor 4/physiology , Animals , Anticonvulsants/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Epilepsy/physiopathology , HMGB1 Protein/antagonists & inhibitors , Hippocampus/physiology , Humans , Interleukin-1beta/physiology , Kainic Acid/pharmacology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Seizures/chemically induced , Seizures/prevention & control , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 4/antagonists & inhibitors
14.
Infect Immun ; 76(7): 3156-63, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18443097

ABSTRACT

Septicemia caused by Neisseria meningitidis is characterized by increasing levels of meningococcal lipopolysaccharide (Nm-LPS) and cytokine production in the blood. We have used an in vitro human whole-blood model of meningococcal septicemia to investigate the potential of CyP, a selective Toll-like receptor 4 (TLR4)-MD-2 antagonist derived from the cyanobacterium Oscillatoria planktothrix FP1, for reducing LPS-mediated cytokine production. CyP (> or = 1 microg/ml) inhibited the secretion of the proinflammatory cytokines tumor necrosis factor alpha, interleukin-1beta (IL-1beta), and IL-6 (by >90%) and chemokines IL-8 and monocyte chemoattractant protein 1 (by approximately 50%) induced by the treatment of blood with pure Nm-LPS, by isolated outer membranes, and after infection with live meningococci of different serogroups. In vitro studies with human dendritic cells and TLR4-transfected Jurkat cells demonstrated that CyP competitively inhibited Nm-LPS interactions with TLR4 and subsequent NF-kappaB activation. These data demonstrate that CyP is a potent antagonist of meningococcal LPS and could be considered a new adjunctive therapy for treating septicemia.


Subject(s)
Bacteremia/immunology , Cyanobacteria/immunology , Cytokines/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Neisseria meningitidis/pathogenicity , Toll-Like Receptor 4/antagonists & inhibitors , Bacteremia/microbiology , Cytokines/biosynthesis , Dendritic Cells , Humans , Jurkat Cells , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/genetics , Transfection
15.
Biochem Biophys Res Commun ; 349(4): 1294-300, 2006 Nov 03.
Article in English | MEDLINE | ID: mdl-16978583

ABSTRACT

Dopaminergic human neuroblastoma SH-SY5Y cells were stably transformed to increase expression of alpha-synuclein, a Parkinson's disease-related protein. Transformed cells were more resistant to oxidative insults, showing a cytoprotective role of alpha-synuclein. The expression of redox chaperonins (DJ-1, HSP70, and 14-3-3) was evaluated by Western blotting. Expression of alpha-synuclein reduced HSP70 levels even in the presence of dopamine, with a twofold increase of DJ-1 in the absence of oxidants. DJ-1 is significantly reduced by dopamine, and even more by dopamine and Cu(II). Increased alpha-synuclein expression did not affect 14-3-3, although dopamine increased its level by 60% in wild-type cells. alpha-Synuclein not only upregulated DJ-1, but also shifted all DJ-1 forms to a single spot at pI=5.7 not observed in wild-type cells. Dopamine gradually restored the distribution of DJ-1 forms to a situation similar to wild-type cells, with the form at pI=6.1 progressively enriched under oxidative conditions.


Subject(s)
Apoptosis/drug effects , Cell Survival/drug effects , Dopamine/administration & dosage , Neuroblastoma/pathology , alpha-Synuclein/administration & dosage , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Combinations , Humans
16.
J Exp Med ; 203(6): 1481-92, 2006 Jun 12.
Article in English | MEDLINE | ID: mdl-16717116

ABSTRACT

Toll-like receptors (TLRs) function as primary sensors that elicit coordinated innate immune defenses through recognition of microbial products and induction of immune and proinflammatory genes. Here we report the identification and biological characterization of a lipopolysaccharide (LPS)-like molecule extracted from the cyanobacterium Oscillatoria Planktothrix FP1 (cyanobacterial product [CyP]) that is not stimulatory per se but acts as a potent and selective antagonist of bacterial LPS. CyP binds to MD-2 and efficiently competes with LPS for binding to the TLR4-MD-2 receptor complex. The addition of CyP together with LPS completely inhibited both MyD88- and TRIF-dependent pathways and suppressed the whole LPS-induced gene transcription program in human dendritic cells (DCs). CyP protected mice from endotoxin shock in spite of a lower capacity to inhibit LPS stimulation of mouse DCs. Interestingly, the delayed addition of CyP to DCs responding to LPS strongly inhibited signaling and cytokine production by immediate down-regulation of inflammatory cytokine mRNAs while not affecting other aspects of DC maturation, such as expression of major histocompatibility complex molecules, costimulatory molecules, and CCR7. Collectively, these results indicate that CyP is a potent competitive inhibitor of LPS in vitro and in vivo and reveal the requirement of sustained TLR4 stimulation for induction of cytokine genes in human DCs.


Subject(s)
Cyanobacteria/immunology , Cytokines/genetics , Dendritic Cells/immunology , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Shock, Septic/prevention & control , Toll-Like Receptor 4/immunology , Animals , Dendritic Cells/drug effects , Humans , Mice , Transcription, Genetic/drug effects
17.
Cancer Lett ; 235(1): 75-83, 2006 Apr 08.
Article in English | MEDLINE | ID: mdl-15922507

ABSTRACT

Endotoxin/lipopolysacccharide (LPS) is a potent inflammatory stimulus, which acts on tumour infiltrating leukocytes by eliciting a wide range of factors promoting invasion and metastasis. Less known is the effect of LPS directly on tumour cells. In this study, we analysed whether tumour cell lines from different origin (melanoma, ovarian carcinoma, neuroblastoma) are responsive to LPS in vitro. Results showed that only melanoma cells significantly up-regulated the production of IL-8 and cell adhesion, when triggered with LPS. These effects were associated with the constitutive expression of TLR-4 mRNA in these cells and the expression on the cell membrane of the complete LPS-binding receptor.


Subject(s)
Interleukin-8/metabolism , Lipopolysaccharides/pharmacology , Melanoma/drug therapy , Toll-Like Receptor 4/genetics , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Escherichia coli , Female , Humans , Melanoma/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , RNA, Messenger/metabolism , Toll-Like Receptor 4/metabolism , Tumor Cells, Cultured
18.
Cancer Detect Prev ; 27(3): 167-74, 2003.
Article in English | MEDLINE | ID: mdl-12787722

ABSTRACT

One of the mechanisms ensuring immunological unresponsiveness or tolerance depends on the action of CD8(+) lymphocytes. In this paper, we report that, in healthy subjects, a subset of CD8(+)CD28(-) T cells suppresses the specific response to TSH receptor (TSHR) of CD4(+) clones. Suppression was highly specific, required cell-cell interaction, and was not mediated by cytotoxicity. Co-incubation of CD8(+) and CD4(+) clones, followed by the removal of the CD8(+) cells from the cultures before testing CD4(+) responsiveness to TSHR, demonstrated that CD4(+) cells were anergic since they showed low response to the antigen and a significant impairment of IL-2 production. In CD8-mediated anergy induction, the T-cell receptor (TCR) on both CD4(+) and CD8(+) cells seems to play a role. Our results indicate that one of the mechanisms ensuring peripheral tolerance involve CD8(+)CD28(-) cells. A disregulation in the control of autoreactive clones by this subset might be important for the onset of autoimmune thyroid diseases.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Receptors, Thyrotropin/immunology , Self Tolerance , Cell Line , Clonal Anergy , Humans , Interleukin-2/biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/immunology
19.
Immunol Lett ; 83(2): 111-7, 2002 Sep 02.
Article in English | MEDLINE | ID: mdl-12067759

ABSTRACT

In normal human subjects a small proportion of peripheral blood T-cells simultaneously express both CD4 and CD8 differentiation antigens. In this study we characterized a subset of CD4+ clones, from a healthy donor, that is specific for the thyrotropin receptor (TSHR) and that showed cells co-expressing the CD8 receptor. To address whether the expression of the CD8 receptor on the cell membrane was associated to differences in the physiology of the T-cells, we isolated, from the same clone, CD4 single positive (SP) cells from those co-expressing CD4/CD8 receptors (DP cells) and stimulated them in vitro with antigen presenting cells (APC) carrying TSHR. The results demonstrated that CD8 co-expression has a profound effect on the physiology of T helper (Th) cells. In comparison to cells expressing the CD4 receptor alone, DP T-cells showed: (1) increased proliferation; (2) higher and more sustained release of free Ca2+ in the cytosol, under stimulus; (3) lower levels of IL-2 and IL-4 released in the supernatants; (4) increased amounts of IFN-gamma released.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cytokines/biosynthesis , Receptors, Antigen, T-Cell/physiology , CD4-Positive T-Lymphocytes/immunology , Cell Line , Humans , Receptors, Antigen, T-Cell/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...