Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Life Sci ; 335: 122242, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37952834

ABSTRACT

AIM: 2-Pentadecyl-2-oxazoline (PEA-OXA), the oxazoline derivative of N-palmitoylethanolamine, exerts anti-inflammatory activity; however, very little is known about the molecular mechanisms underlying this effect. Here, we tested the anti-neuroinflammatory effect of PEA-OXA in primary microglia and we also investigated the possible interaction of the molecule with the Toll-like receptor 4 (TLR4)-myeloid differentiation protein-2 (MD-2) complex. MAIN METHODS: The anti-inflammatory effect of PEA-OXA was analyzed by measuring the expression and release of pro-inflammatory mediators in primary microglia by real-time PCR and ELISA, respectively. The effect of PEA-OXA on the activation of TLR4 signaling was assessed using two stably TLR4-transfected cell lines (i.e., HEK-293 and Ba/F3 cells). Finally, the putative binding mode of PEA-OXA to TLR4-MD-2 was investigated by molecular docking simulations. KEY FINDINGS: Treatment with PEA-OXA resulted in the following effects: (i) it down-regulated gene expression of several pro-inflammatory molecules and the secretion of pro-inflammatory cytokines in LPS stimulated microglia cells; (ii) it did not prevent microglia activation after stimulation with TLR2 ligands; (iii) it prevented TLR4/NF-κB activation triggered by LPS in HEK-Blue™ hTLR4 cells; and (iv) it interfered with the binding of LPS to TLR4-MD-2 complex. Furthermore, molecular docking studies suggested that PEA-OXA could bind MD-2 with a 1:3 (MD-2/PEA-OXA) stoichiometry. CONCLUSION: We show for the first time that the anti-neuroinflammatory effect of PEA-OXA involves its activity against TLR4 signaling, making this molecule a valuable tool for the development of new compounds directed to control neuroinflammation via inhibiting TLR4 signaling.


Subject(s)
Inflammation , Lipopolysaccharides , Humans , Lipopolysaccharides/adverse effects , Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Molecular Docking Simulation , Microglia/metabolism , HEK293 Cells , Anti-Inflammatory Agents/pharmacology , NF-kappa B/metabolism
2.
Front Pharmacol ; 12: 698133, 2021.
Article in English | MEDLINE | ID: mdl-34276381

ABSTRACT

Remyelination in patients with multiple sclerosis frequently fails, especially in the chronic phase of the disease promoting axonal and neuronal degeneration and progressive disease disability. Drug-based therapies able to promote endogenous remyelination capability of oligodendrocytes are thus emerging as primary approaches to multiple sclerosis. We have recently reported that the co-ultramicronized composite of palmitoylethanolamide and the flavonoid luteolin (PEALut) promotes oligodendrocyte precursor cell (OPC) maturation without affecting proliferation. Since TAM receptor signaling has been reported to be important modulator of oligodendrocyte survival, we here evaluated the eventual involvement of TAM receptors in PEALut-induced OPC maturation. The mRNAs related to TAM receptors -Tyro3, Axl, and Mertk- were all present at day 2 in vitro. However, while Tyro3 gene expression significantly increased upon cell differentiation, Axl and Mertk did not change during the first week in vitro. Tyro3 gene expression developmental pattern resembled that of MBP myelin protein. In OPCs treated with PEALut the developmental increase of Tyro3 mRNA was significantly higher as compared to vehicle while was reduced gene expression related to Axl and Mertk. Rapamycin, an inhibitor of mTOR, prevented oligodendrocyte growth differentiation and myelination. PEALut, administered to the cultures 30 min after rapamycin, prevented the alteration of mRNA basal expression of the TAM receptors as well as the expression of myelin proteins MBP and CNPase. Altogether, data obtained confirm that PEALut promotes oligodendrocyte differentiation as shown by the increase of MBP and CNPase and Tyro3 mRNAs as well as CNPase and Tyro3 immunostainings. The finding that these effects are reduced when OPCs are exposed to rapamycin suggests an involvement of mTOR signaling in PEALut effects.

3.
J Neuroinflammation ; 16(1): 126, 2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31221190

ABSTRACT

BACKGROUND: Persistent and/or recurrent inflammatory processes are the main factor leading to multiple sclerosis (MS) lesions. The composite ultramicronized palmitoylethanolamide, an endogenous N-acylethanolamine, combined with the flavonoid luteolin, PEALut, have been found to exert neuroprotective activities in experimental models of spinal and brain injury and Alzheimer disease, as well as a clinical improvement in human stroke patients. Furthermore, PEALut enhances the expression of different myelin proteins in oligodendrocyte progenitor cells suggesting that this composite might have protective effects in MS experimental models. METHODS: The mouse model of experimental autoimmune encephalomyelitis (EAE) based on active immunization with a fragment of myelin oligodendrocyte glycoprotein (MOG35-55) was used. The daily assessment of clinical score and the expression of serum amyloid A (SAA1), proinflammatory cytokines TNF-α, IL-1ß, IFN-γ, and NLRP3 inflammasome, as well as TLR2, Fpr2, CD137, CD3-γ, and TCR-ζ chain, heterodimers that form T cell surface glycoprotein (TCR), and cannabinoid receptors CB1, CB2, and MBP, were evaluated in the brainstem and cerebellum at different postimmunization days (PIDs). RESULTS: Vehicle-MOG35-55-immunized (MOG35-55) mice developed ascending paralysis which peaked several days later and persisted until the end of the experiment. PEALut, given intraperitoneally daily starting on day 11 post-immunization, dose-dependently improved clinical score over the range 0.1-5 mg/kg. The mRNA expression of SAA1, TNF-α, IL-1ß, IFN-γ, and NLRP3 were significantly increased in MOG35-55 mice at 14 PID. In MOG35-55 mice treated with 5 mg /kg PEALut, the increase of SAA1, TNF- α, IL-1ß, and IFN-γ transcripts at 14 PID was statistically downregulated as compared to vehicle-MOG35-55 mice (p < 0.05). The expression of TLR2, Fpr2, CD137, CD3-γ, TCR-ζ chain, and CB2 receptors showed a significant upregulation in vehicle-MOG35-55 mice at 14 PID. Instead, CB1 and MBP transcripts have not changed in expression at any time. In MOG/PEALut-treated mice, TLR2, Fpr2, CD137, CD3-γ, TCR-ζ chain, and CB2 mRNAs were significantly downregulated as compared to vehicle MOG35-55 mice. CONCLUSIONS: The present results demonstrate that the intraperitoneal administration of the composite PEALut significantly reduces the development of clinical signs in the MOG35-55 model of EAE. The dose-dependent improvement of clinical score induced by PEALut was associated with a reduction in transcript expression of the acute-phase protein SAA1, TNF-α, IL-1ß, IFN-γ, and NLRP3 proinflammatory proteins and TLR2, Fpr2, CD137, CD3-γ, TCR-ζ chain, and CB2 receptors.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/pathology , Ethanolamines/pharmacology , Luteolin/pharmacology , Neuroprotective Agents/pharmacology , Palmitic Acids/pharmacology , Amides , Animals , Biomarkers/analysis , Cytokines/drug effects , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL
4.
J Alzheimers Dis ; 64(3): 671-688, 2018.
Article in English | MEDLINE | ID: mdl-29991138

ABSTRACT

Epigenetics is the study of changes in gene expression which may be triggered by both genetic and environmental factors, and independent from changes to the underlying DNA sequence-a change in phenotype without a change in genotype-which in turn affects how cells read genes. Epigenetic changes represent a regular and natural occurrence but can be influenced also by factors such as age, environment, and disease state. Epigenetic modifications can manifest themselves not only as the manner in which cells terminally differentiate, but can have also deleterious effects, resulting in diseases such as cancer. At least three systems including DNA methylation, histone modification, and non-coding RNA (ncRNA)-associated gene silencing are thought to initiate and sustain epigenetic change. For example, in Alzheimer's disease (AD), both genetic and non-genetic factors contribute to disease etiopathology. While over 250 gene mutations have been related to familial AD, less than 5% of AD cases are explained by known disease genes. More than likely, non-genetic factors, probably triggered by environmental factors, are causative factors of late-onset AD. AD is associated with dysregulation of DNA methylation, histone modifications, and ncRNAs. Among the classes of ncRNA, microRNAs (miRNAs) have a well-established regulatory relevance. MicroRNAs are highly expressed in CNS neurons, where they play a major role in neuron differentiation, synaptogenesis, and plasticity. MicroRNAs impact higher cognitive functions, as their functional impairment is involved in the etiology of neurological diseases, including AD. Alterations in the miRNA network contribute to AD disease processes, e.g., in the regulation of amyloid peptides, tau, lipid metabolism, and neuroinflammation. MicroRNAs, both as biomarkers for AD and therapeutic targets, are in the early stages of exploration. In addition, emerging data suggest that altered transcription of long ncRNAs, endogenous, ncRNAs longer than 200 nucleotides, may be involved in an elevated risk for AD.


Subject(s)
Alzheimer Disease/genetics , Epigenesis, Genetic/physiology , Alzheimer Disease/metabolism , DNA Methylation , Humans
5.
J Neuroinflammation ; 15(1): 164, 2018 May 26.
Article in English | MEDLINE | ID: mdl-29803222

ABSTRACT

BACKGROUND: Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves production of acute-phase proteins, including serum amyloid A (SAA). Interleukin-1ß (IL-1ß), a master regulator of neuroinflammation produced by activated inflammatory cells of the myeloid lineage, in particular microglia, plays a key role in the pathogenesis of acute and chronic diseases of the peripheral nervous system and CNS. IL-1ß release is promoted by ATP acting at the purinergic P2X7 receptor (P2X7R) in cells primed with toll-like receptor (TLR) ligands. METHODS: Purified (> 99%) microglia cultured from neonatal rat cortex and cerebellum were first primed with the putative TLR4/TLR2 agonist SAA (recombinant human Apo-SAA) or the established TLR4 agonist lipopolysaccharide (LPS) followed by addition of ATP. Expression of genes for the NLRP3 inflammasome, IL-1ß, tumor necrosis factor-α (TNF-α), and SAA1 was measured by quantitative real-time polymerase chain reaction (q-PCR). Intracellular and extracellular amounts of IL-1ß were determined by ELISA. RESULTS: Apo-SAA stimulated, in a time-dependent manner, the expression of NLRP3, IL-1ß, and TNF-α in cortical microglia, and produced a concentration-dependent increase in the intracellular content of IL-1ß in these cells. A 2-h 'priming' of the microglia with Apo-SAA followed by addition of ATP for 1 h, resulting in a robust release of IL-1ß into the culture medium, with a concomitant reduction in its intracellular content. The selective P2X7R antagonist A740003 blocked ATP-dependent release of IL-1ß. Microglia prepared from rat cerebellum displayed similar behaviors. As with LPS, Apo-SAA upregulated SAA1 and TLR2 mRNA, and downregulated that of TLR4. LPS was less efficacious than Apo-SAA, perhaps reflecting an action of the latter at TLR4 and TLR2. The TLR4 antagonist CLI-095 fully blocked the action of LPS, but only partially that of Apo-SAA. Although the TLR2 antagonist CU-CPT22 was inactive against Apo-SAA, it also failed to block the TLR2 agonist Pam3CSK4. CONCLUSIONS: Microglia are central to the inflammatory process and a major source of IL-1ß when activated. P2X7R-triggered IL-1ß maturation and export is thus likely to represent an important contributor to this cytokine pool. Given that SAA is detected in Alzheimer disease and multiple sclerosis brain, together with IL-1ß-immunopositive microglia, these findings propose a link between P2X7R, SAA, and IL-1ß in CNS pathophysiology.


Subject(s)
Adenosine Triphosphate/pharmacology , Interleukin-1beta/metabolism , Microglia/drug effects , Serum Amyloid A Protein/pharmacology , Animals , Animals, Newborn , Brain/cytology , Cells, Cultured , Interleukin-1beta/genetics , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , RNA, Messenger/metabolism , Rats , Time Factors
6.
Mol Neurobiol ; 55(1): 103-114, 2018 01.
Article in English | MEDLINE | ID: mdl-28822061

ABSTRACT

Oligodendrocytes, the myelin-producing cells of the central nervous system (CNS), have limited capability to bring about repair in chronic CNS neuroinflammatory demyelinating disorders such as multiple sclerosis (MS). MS lesions are characterized by a compromised pool of undifferentiated oligodendrocyte progenitor cells (OPCs) unable to mature into myelin-producing oligodendrocytes. An attractive strategy may be to replace lost OLs and/or promote their maturation. N-palmitoylethanolamine (PEA) is an endogenous fatty acid amide signaling molecule with anti-inflammatory and neuroprotective actions. Recent studies show a co-ultramicronized composite of PEA and the flavonoid luteolin (co-ultraPEALut) to be more efficacious than PEA in improving outcome in CNS injury models. Here, we examined the effects of co-ultraPEALut on development of OPCs from newborn rat cortex cultured under conditions favoring either differentiation (Sato medium) or proliferation (fibroblast growth factor-2 and platelet-derived growth factor (PDGF)-AA-supplemented serum-free medium ("SFM")). OPCs in SFM displayed high expression of PDGF receptor alpha gene and the proliferation marker Ki-67. In Sato medium, in contrast, OPCs showed rapid decreases in PDGF receptor alpha and Ki-67 expression with a concomitant rise in myelin basic protein (MBP) expression. In these conditions, co-ultraPEALut (10 µM) enhanced OPC morphological complexity and expression of MBP and the transcription factor TCF7l2. Surprisingly, co-ultraPEALut also up-regulated MBP mRNA expression in OPCs in SFM. MBP expression in all cases was sensitive to inhibition of mammalian target of rapamycin. Within the context of strategies to promote endogenous remyelination in MS which focus on enhancing long-term survival of OPCs and stimulating their differentiation into remyelinating oligodendrocytes, co-ultraPEALut may represent a novel pharmacological approach.


Subject(s)
Cell Differentiation/drug effects , Ethanolamines/pharmacology , Luteolin/pharmacology , Oligodendrocyte Precursor Cells/drug effects , Oligodendroglia/drug effects , Animals , Animals, Newborn , Cattle , Cell Differentiation/physiology , Cells, Cultured , Drug Combinations , Humans , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/physiology , Rats
7.
Methods Mol Biol ; 1727: 107-118, 2018.
Article in English | MEDLINE | ID: mdl-29222776

ABSTRACT

Dopaminergic neuronal cell degeneration is the principal characteristic feature of the neuropathology of Parkinson disease. Cultures of mesencephalic neurons are widely used as a source of dopaminergic neurons for the study of mechanisms implicated in dopaminergic cell death and for the evaluation of potential dopaminergic neuroprotective agents, including neurotrophic factors. This chapter presents a detailed protocol for the preparation of rat mesencephalic cell cultures and their application to evaluating the effect of the dopaminergic neurotoxin 1-methyl-4-phenylpyridinium and the neuroprotective action of brain-derived neurotrophic factor.


Subject(s)
Cell Culture Techniques/methods , Dopaminergic Neurons/cytology , Mesencephalon/cytology , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , 1-Methyl-4-phenylpyridinium/pharmacology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Cell Count , Cells, Cultured , Dopaminergic Neurons/drug effects , Mesencephalon/drug effects , Rats
8.
Methods Mol Biol ; 1727: 127-137, 2018.
Article in English | MEDLINE | ID: mdl-29222778

ABSTRACT

Glial cell activation plays an important role in the pathogenesis of various neurodegenerative disorders as well as in chronic and neuropathic pain. This chapter describes a model which allows one to assess the individual and combined contributions of astrocytes and microglia in response to a pro-inflammatory stimulus, with emphasis on ionotropic purinergic receptors.


Subject(s)
Astrocytes/cytology , Coculture Techniques/methods , Inflammation/metabolism , Microglia/cytology , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Female , Male , Microglia/metabolism , Models, Biological , Rats , Receptors, Purinergic/metabolism
9.
Methods Mol Biol ; 1727: 155-166, 2018.
Article in English | MEDLINE | ID: mdl-29222780

ABSTRACT

The protocol presented in this chapter covers the application of rat cortical oligodendrocyte progenitor cells cultured under conditions of differentiation for the evaluation of agents with potential trophic activity, that is, which are capable of promoting maturation under such conditions. As an example we have chosen a co-ultramicronized N-palmitoylethanolamine/luteolin composite, a formulation described in the literature as possessing anti-inflammatory, neuroprotective, and neuroregenerative actions.


Subject(s)
Cell Culture Techniques/methods , Nerve Regeneration/drug effects , Neuroprotective Agents/pharmacology , Oligodendrocyte Precursor Cells/cytology , Amides , Animals , Cell Differentiation , Cells, Cultured , Ethanolamines/pharmacology , Luteolin/pharmacology , Myelin Sheath/drug effects , Myelin Sheath/physiology , Palmitic Acids/pharmacology , Rats
10.
Sci Rep ; 7(1): 12158, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28939905

ABSTRACT

Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves hepatic production of acute-phase proteins, including serum amyloid A (SAA). Extrahepatically, SAA immunoreactivity is found in axonal myelin sheaths of cortex in Alzheimer's disease and multiple sclerosis (MS), although its cellular origin is unclear. We examined the responses of cultured rat cortical astrocytes, microglia and oligodendrocyte precursor cells (OPCs) to master pro-inflammatory cytokine tumour necrosis factor (TNF)-α and lipopolysaccaride (LPS). TNF-α time-dependently increased Saa1 (but not Saa3) mRNA expression in purified microglia, enriched astrocytes, and OPCs (as did LPS for microglia and astrocytes). Astrocytes depleted of microglia were markedly less responsive to TNF-α and LPS, even after re-addition of microglia. Microglia and enriched astrocytes showed complementary Saa1 expression profiles following TNF-α or LPS challenge, being higher in microglia with TNF-α and higher in astrocytes with LPS. Recombinant human apo-SAA stimulated production of both inflammatory mediators and its own mRNA in microglia and enriched, but not microglia-depleted astrocytes. Co-ultramicronized palmitoylethanolamide/luteolin, an established anti-inflammatory/ neuroprotective agent, reduced Saa1 expression in OPCs subjected to TNF-α treatment. These last data, together with past findings suggest that co-ultramicronized palmitoylethanolamide/luteolin may be a novel approach in the treatment of inflammatory demyelinating disorders like MS.


Subject(s)
Astrocytes/immunology , Microglia/immunology , Serum Amyloid A Protein/metabolism , Tumor Necrosis Factor-alpha/immunology , Acute-Phase Reaction/immunology , Animals , Cells, Cultured , Humans , Lipopolysaccharides/immunology , Oligodendrocyte Precursor Cells/immunology , RNA, Messenger/genetics , Rats , Up-Regulation
11.
Sci Rep ; 5: 16676, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26578323

ABSTRACT

Oligodendrocytes have limited ability to repair the damage to themselves or to other nerve cells, as seen in demyelinating diseases like multiple sclerosis. An important strategy may be to replace the lost oligodendrocytes and/or promote the maturation of undifferentiated oligodendrocyte precursor cells (OPCs). Recent studies show that a composite of co-ultramicronized N-palmitoylethanolamine (PEA) and luteolin (co-ultramicronized PEA/luteolin, 10:1 by mass) is efficacious in improving outcome in experimental models of spinal cord and traumatic brain injuries. Here, we examined the ability of co-ultramicronized PEA/luteolin to promote progression of OPCs into a more differentiated phenotype. OPCs derived from newborn rat cortex were placed in culture and treated the following day with 10 µM co-ultramicronized PEA/luteolin. Cells were collected 1, 4 and 8 days later and analyzed for expression of myelin basic protein (MBP). qPCR and Western blot analyses revealed a time-dependent increase in expression of both mRNA for MBP and MBP content, along with an increased expression of genes involved in lipid biogenesis. Ultramicronized PEA or luteolin, either singly or in simple combination, were ineffective. Further, co-ultramicronized PEA/luteolin promoted morphological development of OPCs and total protein content without affecting proliferation. Co-ultramicronized PEA/luteolin may represent a novel pharmacological strategy to promote OPC maturation.


Subject(s)
Cell Differentiation/drug effects , Ethanolamines/pharmacology , Luteolin/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Palmitic Acids/pharmacology , Amides , Animals , Cells, Cultured , Ethanolamines/chemistry , Gene Expression Regulation/genetics , Luteolin/chemistry , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Palmitic Acids/chemistry , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
12.
Mol Neurobiol ; 52(2): 1034-42, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26055231

ABSTRACT

Inflammation is fundamentally a protective cellular response aimed at removing injurious stimuli and initiating the healing process. However, when prolonged, it can override the bounds of physiological control and becomes destructive. Inflammation is a key element in the pathobiology of chronic pain, neurodegenerative diseases, stroke, spinal cord injury, and neuropsychiatric disorders. Glia, key players in such nervous system disorders, are not only capable of expressing a pro-inflammatory phenotype but respond also to inflammatory signals released from cells of immune origin such as mast cells. Chronic inflammatory processes may be counteracted by a program of resolution that includes the production of lipid mediators endowed with the capacity to switch off inflammation. These naturally occurring lipid signaling molecules include the N-acylethanolamines, N-arachidonoylethanolamine (an endocannabinoid), and its congener N-palmitoylethanolamine (palmitoylethanolamide or PEA). PEA may play a role in maintaining cellular homeostasis when faced with external stressors provoking, for example, inflammation. PEA is efficacious in mast cell-mediated models of neurogenic inflammation and neuropathic pain and is neuroprotective in models of stroke, spinal cord injury, traumatic brain injury, and Parkinson disease. PEA in micronized/ultramicronized form shows superior oral efficacy in inflammatory pain models when compared to naïve PEA. Intriguingly, while PEA has no antioxidant effects per se, its co-ultramicronization with the flavonoid luteolin is more efficacious than either molecule alone. Inhibiting or modulating the enzymatic breakdown of PEA represents a complementary therapeutic approach to treat neuroinflammation. This review is intended to discuss the role of mast cells and glia in neuroinflammation and strategies to modulate their activation based on leveraging natural mechanisms with the capacity for self-defense against inflammation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Ethanolamines/therapeutic use , Inflammation/drug therapy , Palmitic Acids/therapeutic use , Administration, Oral , Amides , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/physiology , Animals , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Biological Availability , Biotransformation , Depression/drug therapy , Disease Models, Animal , Drug Combinations , Ethanolamines/administration & dosage , Ethanolamines/pharmacokinetics , Ethanolamines/pharmacology , Homeostasis , Humans , Inflammation/immunology , Inflammation/physiopathology , Luteolin/administration & dosage , Luteolin/therapeutic use , Mast Cells/immunology , Mast Cells/metabolism , Mast Cells/pathology , Neurocognitive Disorders/drug therapy , Neurocognitive Disorders/pathology , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/immunology , Palmitic Acids/administration & dosage , Palmitic Acids/pharmacokinetics , Palmitic Acids/pharmacology , Particle Size , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology
13.
Sci Rep ; 4: 6824, 2014 Oct 29.
Article in English | MEDLINE | ID: mdl-25351234

ABSTRACT

Interleukin-1ß (IL-1ß) is a crucial mediator in the pathogenesis of inflammatory diseases at the periphery and in the central nervous system (CNS). Produced as an unprocessed and inactive pro-form which accumulates intracellularly, release of the processed cytokine is strongly promoted by ATP acting at the purinergic P2X7 receptor (P2X7R) in cells primed with lipopolysaccharide (LPS), a Toll-like receptor (TLR) 4 ligand. Microglia are central to the inflammatory process and a major source of IL-1ß when activated. Here we show that purified (>99%) microglia cultured from rat cortex, spinal cord and cerebellum respond robustly to ATP-dependent IL-1ß release, upon priming with a number of TLR isoform ligands (zymosan and Pam3CSK4 for TLR2, poly(I:C) for TLR3). Cytokine release was prevented by a P2X7R antagonist and inhibitors of stress-activated protein kinases. Enriched astrocytes (≤ 5% microglia) from these CNS regions displayed responses qualitatively similar to microglia but became unresponsive upon eradication of residual microglia with the lysosomotropic agent Leu-Leu-OMe. Activation of multiple TLR isoforms in nervous system pathology, coupled with elevated extracellular ATP levels and subsequent P2X7R activation may represent an important route for microglia-derived IL-1ß. This phenomenon may have important consequences for neuroinflammation and its position to the common pathology of CNS diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Astrocytes/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Acetamides/pharmacology , Animals , Astrocytes/drug effects , Central Nervous System/drug effects , Central Nervous System/metabolism , Cerebellum/metabolism , Cerebral Cortex/metabolism , Ligands , Microglia/drug effects , Quinolines/pharmacology , Rats , Signal Transduction , Spinal Cord/metabolism , Stress, Physiological , Toll-Like Receptor 2/agonists , Toll-Like Receptor 3/agonists , Toll-Like Receptor 4/agonists
14.
CNS Neurol Disord Drug Targets ; 12(5): 608-18, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23574172

ABSTRACT

Glial cells not only serve supportive and nutritive roles for neurons, but also respond to protracted stress and insults by up-regulating inflammatory processes. The complexity of studying glial activation in vivo has led to the widespread adoption of in vitro approaches, for example the use of the bacterial toxin lipopolysaccharide (LPS, a ligand for toll-like receptor 4 (TLR4)) as an experimental model of glial activation. Astrocyte cultures frequently contain minor numbers of microglia, which can complicate interpretation of responses. In the present study, enriched (≤5% microglia) astrocytes cultured from neonatal rat cortex and spinal cord were treated with the lysosomotropic agent L-leucyl-L-leucine methyl ester to eliminate residual microglia, as confirmed by loss of microglia-specific marker genes. L-Leucyl-L-leucine methyl ester treatment led to a loss of LPS responsiveness, in terms of nitric oxide and cytokine gene up-regulation and mediator (pro-inflammatory cytokines, nitric oxide) output into the culture medium. Surprisingly, when astrocyte/microglia co-cultures were then reconstituted by adding defined numbers of purified microglia to microglia-depleted astrocytes, the LPS-induced up-regulation of pro-inflammatory gene and mediator output far exceeded that observed from cultures containing the same numbers of microglia only. Similar behaviors were found when examining interleukin-1ß release caused by activation of the purinergic P2X7 receptor. Given that astrocytes greatly outnumber microglia in the central nervous system, these data suggest that a similar interaction between microglia and astrocytes in vivo may be an important element in the evolution of an inflammatory pathology.


Subject(s)
Astrocytes/metabolism , Cytokines/metabolism , Gene Expression Regulation/physiology , Microglia/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Astrocytes/drug effects , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Cytokines/genetics , Dipeptides/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Lipopolysaccharides/pharmacology , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Rats , Spinal Cord/cytology
15.
PLoS One ; 8(4): e61983, 2013.
Article in English | MEDLINE | ID: mdl-23626760

ABSTRACT

Although the induction of cytochrome P450 (CYP) has long been investigated in patients with cirrhosis, the question whether liver dysfunction impairs the response to CYP inducers still remains unresolved. Moreover, the mechanism underlying the possible effect of cirrhosis on induction has not been investigated. Since ethical constraints do not permit methodologically rigorous studies in humans, this question was addressed by investigating the effect of the prototypical inducer benzo[a]pyrene (BP) on CYP1A1 and CYP1A2 in cirrhotic rats stratified according to the severity of liver dysfunction. We simultaneously assessed mRNA level, protein expression and enzymatic activity of the CYP1A enzymes, as well as mRNA and protein expressions of the aryl hydrocarbon receptor (AhR), which mediates the BP effect. Basal mRNA and protein expressions of CYP1A1 were virtually absent in both healthy and cirrhotic rats. On the contrary, CYP1A2 mRNA, protein and enzyme activity were constitutively present in healthy rats and decreased significantly as liver function worsened. BP treatment markedly increased the concentrations of mRNA and immunodetectable protein, and the enzymatic activities of both CYP1A enzymes to similar levels in healthy and non-ascitic cirrhotic rats. Induced mRNA levels, protein expressions and enzymatic activities of both CYPs were much lower in ascitic rats and were proportionally reduced. Both constitutive and induced protein expressions of AhR were significantly lower in ascitic than in healthy rats. These results indicate that the inducibility of CYP1A enzymes is well preserved in compensated cirrhosis, whereas it is markedly reduced when liver dysfunction becomes severe. Induction appears to be impaired at the transcriptional level, due to the reduced expression of AhR, which controls the transcription of CYP1A genes.


Subject(s)
Ascites/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochromes/metabolism , Enzyme Induction/genetics , Liver Cirrhosis, Experimental/genetics , Liver/enzymology , Receptors, Aryl Hydrocarbon/metabolism , Animals , Ascites/chemically induced , Ascites/enzymology , Ascites/pathology , Benzo(a)pyrene , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A2 , Cytochromes/genetics , Gene Expression Regulation , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/enzymology , Liver Cirrhosis, Experimental/pathology , Liver Function Tests , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Aryl Hydrocarbon/genetics , Severity of Illness Index , Transcription, Genetic
16.
Drug Metab Pharmacokinet ; 27(6): 640-52, 2012.
Article in English | MEDLINE | ID: mdl-22785257

ABSTRACT

The objective of this study was to compare RT-PCR, Western blot and determination of enzyme activity in the assessment of the induction of cytochromes P450 (CYPs) 1A1 and 1A2 by benzo[a]pyrene (BaP) in Sprague-Dawley and Wistar rats. Inhibition studies and kinetic analyses confirmed literature data indicating that methoxyresorufin is a specific CYP1A2 substrate in both uninduced and BaP-treated rats, whereas ethoxyresorufin is a specific CYP1A1 substrate only in BaP-treated rats. BaP treatment increased mRNA and protein expressions of both CYP1A enzymes to a greater extent in Wistar than Sprague-Dawley rats. It consistently caused a higher increase in mRNA and protein expression of the aryl hydrocarbon receptor in the former rats. By contrast, CYP1A2 enzyme activity was much more markedly increased in Sprague-Dawley than Wistar rats and CYP1A1 activity was induced to similar levels. A BaP-induced increase in the turnover number of CYP1A enzymes in Sprague-Dawley rats, relative to Wistar rats, may provide a plausible explanation for the differential effect of BaP on gene expression and enzyme activity. These results have methodological implications, since they show that RT-PCR and Western blot may not provide a quantitative measure of induction of CYP1A activity, which is the actual measure of the change in CYP1A-mediated metabolism.


Subject(s)
Benzo(a)pyrene/pharmacology , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Cytochromes/genetics , Cytochromes/metabolism , Animals , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A2 , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytochromes/biosynthesis , Gene Expression/drug effects , Kinetics , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxazines/pharmacology , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
17.
Methods Mol Biol ; 846: 67-77, 2012.
Article in English | MEDLINE | ID: mdl-22367802

ABSTRACT

The protocol described in this chapter covers the preparation and culture of enriched populations of microglia, astrocytes, and oligodendrocytes from the cortex and spinal cord of neonatal rat and mouse. The procedure is based on the enzymatic digestion of tissue, followed by the culture of a mixed glial cell population which is then utilized as the starting point for the isolation, via differential attachment, of the different cell types.


Subject(s)
Astrocytes/cytology , Cell Culture Techniques/methods , Cell Separation/methods , Cerebral Cortex/cytology , Microglia/cytology , Oligodendroglia/cytology , Spinal Cord/cytology , Animals , Animals, Newborn , Mice , Rats
18.
Methods Mol Biol ; 846: 235-46, 2012.
Article in English | MEDLINE | ID: mdl-22367816

ABSTRACT

Immunofluorescence is a technique allowing the visualization of a specific protein or antigen in cells or tissue sections by binding a specific antibody chemically conjugated with a fluorescent dye such as fluorescein isothiocyanate. There are two major types of immunofluorescence staining methods: (1) direct immunofluorescence staining in which the primary antibody is labeled with fluorescence dye and (2) indirect immunofluorescence staining in which a secondary antibody labeled with fluorochrome is used to recognize a primary antibody. This chapter describes procedures for the application of indirect immunofluorescence staining to neural cells in culture.


Subject(s)
Cells, Cultured/cytology , Fluorescent Antibody Technique, Indirect/methods , Neurons/cytology , Immunohistochemistry/methods , Microscopy, Fluorescence/methods
19.
Blood ; 113(19): 4525-33, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19196659

ABSTRACT

The peculiar site of development of primary effusion lymphoma (PEL) highlights a specific role of body cavities in the pathogenesis of this neoplasia. We used a xenograft murine model of PEL to characterize the contribution of the host microenvironment to PEL growth. The activity of a murine (ie, host-specific) interferon-alpha(1) (IFN-alpha(1))-expressing lentiviral vector (mIFN-alpha(1)-LV) was compared with that of a human (h) IFN-alpha(2)b-LV. LVs efficiently delivered the transgene to PEL cells and conferred long-term transgene expression in vitro and in vivo. Treatment of PEL-injected severe combined immunodeficiency mice with hIFN-alpha(2)b-LV significantly prolonged mice survival and reduced ascites development. Interestingly, mIFN-alpha(1)-LV showed an antineoplastic activity comparable with that observed with hIFN-alpha(2)b-LV. As mIFN-alpha(1) retained species-restricted activity in vitro, it probably acted in vivo on the intracavitary murine milieu. mIFN-alpha(1)-treated murine mesothelial cells were found to express tumor necrosis factor-related apoptosis-inducing ligand and to significantly trigger apoptosis of cocultured PEL cells in a tumor necrosis factor-related apoptosis-inducing ligand-dependent manner. These data suggest that the interaction between lymphomatous and mesothelial cells lining the body cavities may play a key role in PEL growth control and also indicate that the specific targeting of microenvironment may impair PEL development.


Subject(s)
Antineoplastic Agents/therapeutic use , Genetic Vectors , Interferon-alpha/therapeutic use , Lentivirus/genetics , Lymphoma, Primary Effusion/drug therapy , Animals , Cells, Cultured , Cytokines/metabolism , Drug Evaluation, Preclinical , Female , Humans , Interferon alpha-2 , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/pathology , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Mice, SCID , Peritoneum/cytology , Peritoneum/drug effects , Peritoneum/metabolism , Recombinant Proteins
20.
PLoS One ; 3(12): e4073, 2008.
Article in English | MEDLINE | ID: mdl-19115001

ABSTRACT

Herpesvirus infection of placenta may be harmful in pregnancy leading to disorders in fetal growth, premature delivery, miscarriage, or major congenital abnormalities. Although a correlation between human herpesvirus 8 (HHV-8) infection and abortion or low birth weight in children has been suggested, and rare cases of in utero or perinatal HHV-8 transmission have been documented, no direct evidence of HHV-8 infection of placenta has yet been reported. The aim of this study was to evaluate the in vitro and in vivo susceptibility of placental cells to HHV-8 infection. Short-term infection assays were performed on placental chorionic villi isolated from term placentae. Qualitative and quantitative HHV-8 detection were performed by PCR and real-time PCR, and HHV-8 proteins were analyzed by immunohistochemistry. Term placenta samples from HHV-8-seropositive women were analyzed for the presence of HHV-8 DNA and antigens. In vitro infected histocultures showed increasing amounts of HHV-8 DNA in tissues and supernatants; cyto- and syncitiotrophoblasts, as well as endothelial cells, expressed latent and lytic viral antigens. Increased apoptotic phenomena were visualized by the terminal deoxynucleotidyl transferase-mediated deoxyuridine nick end-labeling method in infected histocultures. Ex vivo, HHV-8 DNA and a latent viral antigen were detected in placenta samples from HHV-8-seropositive women. These findings demonstrate that HHV-8, like other human herpesviruses, may infect placental cells in vitro and in vivo, thus providing evidence that this phenomenon might influence vertical transmission and pregnancy outcome in HHV-8-infected women.


Subject(s)
Herpesviridae Infections/virology , Herpesvirus 8, Human/isolation & purification , Placenta Diseases/virology , Placenta/virology , Apoptosis , DNA, Viral/metabolism , Endothelial Cells/metabolism , Female , Herpesviridae Infections/immunology , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Humans , Immunohistochemistry , Placenta/immunology , Placenta Diseases/immunology , Pregnancy , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...