Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters

Publication year range
1.
Purinergic Signal ; 20(1): 35-45, 2024 Feb.
Article in English | MEDLINE | ID: mdl-36918461

ABSTRACT

Interest has been focused in recent years on the analgesic effects exerted by adenosine and its receptors, A1, A2A, A2B, and A3 adenosine receptor (AR) subtypes, in different in vivo models of chronic pain. In particular, it was demonstrated that selective A3AR agonists reduced pro-nociceptive N-type Ca2+ channels in dorsal root ganglion (DRG) neurons isolated from rats and, by this mechanism, inhibit post inflammatory visceral hypersensitivity. In the present study, we investigate the effect of a previously reported irreversibly binding A3AR agonist, ICBM, on Ca2+ currents (ICa) in rat DRG neurons. Present data demonstrate that ICBM, an isothiocyanate derivative designed for covalent binding to the receptor, concentration-dependently inhibits ICa. This effect is irreversible, since it persists after drug removal, differently from the prototypical A3AR agonist, Cl-IB-MECA. ICBM pre-exposure inhibits the effect of a subsequent Cl-IB-MECA application. Thus, covalent A3AR agonists such as ICBM may represent an innovative, beneficial, and longer-lasting strategy to achieve efficacious chronic pain control versus commonly used, reversible, A3AR agonists. However, the possible limitations of this drug and other covalent drugs may be, for example, a characteristic adverse effect profile, suggesting that more pre-clinical studies are needed.


Subject(s)
Chronic Pain , Ganglia, Spinal , Rats , Animals , Ganglia, Spinal/metabolism , Chronic Pain/metabolism , Neurons/metabolism , Adenosine/metabolism , Receptors, Purinergic P1/metabolism , Receptor, Adenosine A3/metabolism , Adenosine A3 Receptor Agonists/pharmacology
2.
Molecules ; 27(6)2022 03 15.
Article in English | MEDLINE | ID: mdl-35335254

ABSTRACT

Ligands of the Gi protein-coupled adenosine A3 receptor (A3R) are receiving increasing interest as attractive therapeutic tools for the treatment of a number of pathological conditions of the central and peripheral nervous systems (CNS and PNS, respectively). Their safe pharmacological profiles emerging from clinical trials on different pathologies (e.g., rheumatoid arthritis, psoriasis and fatty liver diseases) confer a realistic translational potential to these compounds, thus encouraging the investigation of highly selective agonists and antagonists of A3R. The present review summarizes information on the effect of latest-generation A3R ligands, not yet available in commerce, obtained by using different in vitro and in vivo models of various PNS- or CNS-related disorders. This review places particular focus on brain ischemia insults and colitis, where the prototypical A3R agonist, Cl-IB-MECA, and antagonist, MRS1523, have been used in research studies as reference compounds to explore the effects of latest-generation ligands on this receptor. The advantages and weaknesses of these compounds in terms of therapeutic potential are discussed.


Subject(s)
Adenosine A3 Receptor Agonists , Arthritis, Rheumatoid , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/therapeutic use , Arthritis, Rheumatoid/drug therapy , Humans , Ligands , Peripheral Nervous System , Receptors, Purinergic P1
3.
J Neurochem ; 157(4): 1182-1195, 2021 05.
Article in English | MEDLINE | ID: mdl-33030215

ABSTRACT

The Nucleus Basalis of Meynert (NBM) is the main source of cholinergic neurons in the basal forebrain to be crucially involved in cognitive functions and whose degeneration correlates with cognitive decline in major degenerative pathologies as Alzheimer's and Parkinson's diseases. However, knowledge concerning NBM neurons derived from human brain is very limited to date. We recently characterized a primary culture of proliferating neuroblasts isolated from the human fetal NBM (hfNBM) as immature cholinergic neurons expressing the machinery to synthetize and release acetylcholine. Here we studied in detail electrophysiological features and cholinergic effects in this cell culture by patch-clamp recordings. Our data demonstrate that atropine-blocked muscarinic receptor activation by acetylcholine or carbachol enhanced IK and reduced INa currents by stimulating Gi -coupled M2 or phospholipase C-coupled M3 receptors, respectively. Inhibition of acetylcholine esterase activity by neostigmine unveiled a spontaneous acetylcholine release from hfNBM neuroblasts that might account for an autocrine/paracrine signaling during human brain development. Present data provide the first description of cholinergic effects in human NBM neurons and point to a role of acetylcholine as an autocrine/paracrine modulator of voltage-dependent channels. Our research could be of relevance in understanding the mechanisms of cholinergic system development and functions in the human brain, either in health or disease.


Subject(s)
Acetylcholine/metabolism , Action Potentials/physiology , Basal Forebrain/metabolism , Cholinergic Neurons/metabolism , Neural Stem Cells/metabolism , Basal Nucleus of Meynert/metabolism , Cells, Cultured , Fetus , Humans , Signal Transduction/physiology
4.
J Enzyme Inhib Med Chem ; 36(1): 964-976, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34056989

ABSTRACT

Ischaemic stroke is a leading cause of death and disability. One of the major pathogenic mechanisms after ischaemia includes the switch to the glycolytic pathway, leading to tissue acidification. Carbonic anhydrase (CA) contributes to pH regulation. A new generation of CA inhibitors, AN11-740 and AN6-277 and the reference compound acetazolamide (ACTZ) were investigated in two models of brain ischaemia: in rat hippocampal acute slices exposed to severe oxygen, glucose deprivation (OGD) and in an in vivo model of focal cerebral ischaemia induced by permanent occlusion of the middle cerebral artery (pMCAo) in the rat. In vitro, the application of selective CAIs significantly delayed the appearance of anoxic depolarisation induced by OGD. In vivo, sub-chronic systemic treatment with AN11-740 and ACTZ significantly reduced the neurological deficit and decreased the infarct volume after pMCAo. CAIs counteracted neuronal loss, reduced microglia activation and partially counteracted astrocytes degeneration inducing protection from functional and tissue damage.


Subject(s)
Brain Ischemia/drug therapy , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Disease Models, Animal , Neuroprotective Agents/pharmacology , Animals , Brain Ischemia/metabolism , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Dose-Response Relationship, Drug , Male , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Rats , Rats, Wistar , Structure-Activity Relationship
5.
BMC Biol ; 18(1): 197, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33317522

ABSTRACT

BACKGROUND: The mechanism underlying the pain symptoms associated with chemotherapeutic-induced peripheral neuropathy (CIPN) is poorly understood. Transient receptor potential ankyrin 1 (TRPA1), TRP vanilloid 4 (TRPV4), TRPV1, and oxidative stress have been implicated in several rodent models of CIPN-evoked allodynia. Thalidomide causes a painful CIPN in patients via an unknown mechanism. Surprisingly, the pathway responsible for such proalgesic response has not yet been investigated in animal models. RESULTS: Here, we reveal that a single systemic administration of thalidomide and its derivatives, lenalidomide and pomalidomide, elicits prolonged (~ 35 days) mechanical and cold hypersensitivity in C57BL/6J mouse hind paw. Pharmacological antagonism or genetic deletion studies indicated that both TRPA1 and TRPV4, but not TRPV1, contribute to mechanical allodynia, whereas cold hypersensitivity was entirely due to TRPA1. Thalidomide per se did not stimulate recombinant and constitutive TRPA1 and TRPV4 channels in vitro, which, however, were activated by the oxidative stress byproduct, hydrogen peroxide. Systemic treatment with an antioxidant attenuated mechanical and cold hypersensitivity, and the increase in oxidative stress in hind paw, sciatic nerve, and lumbar spinal cord produced by thalidomide. Notably, central (intrathecal) or peripheral (intraplantar) treatments with channel antagonists or an antioxidant revealed that oxidative stress-dependent activation of peripheral TRPA1 mediates cold allodynia and part of mechanical allodynia. However, oxidative stress-induced activation of central TRPV4 mediated the residual TRPA1-resistant component of mechanical allodynia. CONCLUSIONS: Targeting of peripheral TRPA1 and central TRPV4 may be required to attenuate pain associated with CIPN elicited by thalidomide and related drugs.


Subject(s)
Hyperalgesia/genetics , Oxidative Stress , Pain/genetics , TRPA1 Cation Channel/genetics , TRPV Cation Channels/genetics , Thalidomide/adverse effects , Animals , Hyperalgesia/chemically induced , Male , Mice , Mice, Inbred C57BL , Pain/chemically induced , Rats , Rats, Sprague-Dawley , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism
6.
Int J Mol Sci ; 22(19)2021 Sep 27.
Article in English | MEDLINE | ID: mdl-34638733

ABSTRACT

The importance of precise co- and post-transcriptional processing of RNA in the regulation of gene expression has become increasingly clear. RNA-binding proteins (RBPs) are a class of proteins that bind single- or double-chain RNA, with different affinities and selectivity, thus regulating the various functions of RNA and the fate of the cells themselves. ELAV (embryonic lethal/abnormal visual system)/Hu proteins represent an important family of RBPs and play a key role in the fate of newly transcribed mRNA. ELAV proteins bind AU-rich element (ARE)-containing transcripts, which are usually present on the mRNA of proteins such as cytokines, growth factors, and other proteins involved in neuronal differentiation and maintenance. In this review, we focused on a member of ELAV/Hu proteins, HuR, and its role in the development of neurodegenerative disorders, with a particular focus on demyelinating diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , ELAV-Like Protein 1 , Multiple Sclerosis , Muscular Atrophy, Spinal , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/therapy , Animals , Cell Differentiation/genetics , Cytokines/genetics , Cytokines/metabolism , ELAV-Like Protein 1/genetics , ELAV-Like Protein 1/metabolism , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/therapy , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/therapy , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Int J Mol Sci ; 22(14)2021 Jul 06.
Article in English | MEDLINE | ID: mdl-34298893

ABSTRACT

Multiple sclerosis (MS) is the most demyelinating disease of the central nervous system (CNS) characterized by neuroinflammation. Oligodendrocyte progenitor cells (OPCs) are cycling cells in the developing and adult CNS that, under demyelinating conditions, migrate to the site of lesions and differentiate into mature oligodendrocytes to remyelinate damaged axons. However, this process fails during disease chronicization due to impaired OPC differentiation. Moreover, OPCs are crucial players in neuro-glial communication as they receive synaptic inputs from neurons and express ion channels and neurotransmitter/neuromodulator receptors that control their maturation. Ion channels are recognized as attractive therapeutic targets, and indeed ligand-gated and voltage-gated channels can both be found among the top five pharmaceutical target groups of FDA-approved agents. Their modulation ameliorates some of the symptoms of MS and improves the outcome of related animal models. However, the exact mechanism of action of ion-channel targeting compounds is often still unclear due to the wide expression of these channels on neurons, glia, and infiltrating immune cells. The present review summarizes recent findings in the field to get further insights into physio-pathophysiological processes and possible therapeutic mechanisms of drug actions.


Subject(s)
Brain/metabolism , Ion Channels/metabolism , Myelin Sheath/metabolism , Remyelination/physiology , Animals , Cell Differentiation/physiology , Humans , Neurons/metabolism , Oligodendrocyte Precursor Cells/metabolism
8.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360719

ABSTRACT

Agonists of the Gi protein-coupled A3 adenosine receptor (A3AR) have shown important pain-relieving properties in preclinical settings of several pain models. Active as a monotherapy against chronic pain, A3AR agonists can also be used in combination with classic opioid analgesics. Their safe pharmacological profile, as shown by clinical trials for other pathologies, i.e., rheumatoid arthritis, psoriasis and fatty liver diseases, confers a realistic translational potential, thus encouraging research studies on the molecular mechanisms underpinning their antinociceptive actions. A number of pathways, involving central and peripheral mechanisms, have been proposed. Recent evidence showed that the prototypical A3AR agonist Cl-IB-MECA and the new, highly selective, A3AR agonist MRS5980 inhibit neuronal (N-type) voltage-dependent Ca2+ currents in dorsal root ganglia, a known pain-related mechanism. Other proposed pathways involve reduced cytokine production, immune cell-mediated responses, as well as reduced microglia and astrocyte activation in the spinal cord. The aim of this review is to summarize up-to-date information on A3AR in the context of pain, including cellular and molecular mechanisms underlying this effect. Based on their safety profile shown in clinical trials for other pathologies, A3AR agonists are proposed as novel, promising non-narcotic agents for pain control.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Calcium Signaling/drug effects , Ganglia, Spinal , Pain , Receptor, Adenosine A3/metabolism , Animals , Astrocytes/metabolism , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Humans , Microglia/metabolism , Pain/drug therapy , Pain/metabolism , Pain/physiopathology
9.
Int J Mol Sci ; 22(9)2021 May 10.
Article in English | MEDLINE | ID: mdl-34068564

ABSTRACT

Ischemic stroke is a leading cause of death and disability worldwide. The only pharmacological treatment available to date for cerebral ischemia is tissue plasminogen activator (t-PA) and the search for successful therapeutic strategies still remains a major challenge. The loss of cerebral blood flow leads to reduced oxygen and glucose supply and a subsequent switch to the glycolytic pathway, which leads to tissue acidification. Carbonic anhydrase (CA, EC 4.2.1.1) is the enzyme responsible for converting carbon dioxide into a protons and bicarbonate, thus contributing to pH regulation and metabolism, with many CA isoforms present in the brain. Recently, numerous studies have shed light on several classes of carbonic anhydrase inhibitor (CAI) as possible new pharmacological agents for the management of brain ischemia. In the present review we summarized pharmacological, preclinical and clinical findings regarding the role of CAIs in strokes and we discuss their potential protective mechanisms.


Subject(s)
Brain Ischemia/drug therapy , Carbonic Anhydrase Inhibitors/therapeutic use , Carbonic Anhydrases/genetics , Ischemic Stroke/drug therapy , Bicarbonates/metabolism , Brain/metabolism , Brain/pathology , Brain Ischemia/genetics , Brain Ischemia/pathology , Carbon Dioxide/metabolism , Humans , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Ischemic Stroke/genetics , Sulfonamides/therapeutic use
10.
Int J Mol Sci ; 21(24)2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33353217

ABSTRACT

Adenosine is a signaling molecule, which, by activating its receptors, acts as an important player after cerebral ischemia. Here, we review data in the literature describing A2BR-mediated effects in models of cerebral ischemia obtained in vivo by the occlusion of the middle cerebral artery (MCAo) or in vitro by oxygen-glucose deprivation (OGD) in hippocampal slices. Adenosine plays an apparently contradictory role in this receptor subtype depending on whether it is activated on neuro-glial cells or peripheral blood vessels and/or inflammatory cells after ischemia. Indeed, A2BRs participate in the early glutamate-mediated excitotoxicity responsible for neuronal and synaptic loss in the CA1 hippocampus. On the contrary, later after ischemia, the same receptors have a protective role in tissue damage and functional impairments, reducing inflammatory cell infiltration and neuroinflammation by central and/or peripheral mechanisms. Of note, demyelination following brain ischemia, or autoimmune neuroinflammatory reactions, are also profoundly affected by A2BRs since they are expressed by oligodendroglia where their activation inhibits cell maturation and expression of myelin-related proteins. In conclusion, data in the literature indicate the A2BRs as putative therapeutic targets for the still unmet treatment of stroke or demyelinating diseases.


Subject(s)
Brain Ischemia/drug therapy , Demyelinating Diseases/drug therapy , Neuroprotective Agents/pharmacology , Receptors, Adenosine A2/chemistry , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Signal Transduction
11.
J Cell Mol Med ; 23(3): 1976-1986, 2019 03.
Article in English | MEDLINE | ID: mdl-30636360

ABSTRACT

Safranal, contained in Crocus sativus L., exerts anti-inflammatory and analgesic effects. However, the underlying mechanisms for such effects are poorly understood. We explored whether safranal targets the transient receptor potential ankyrin 1 (TRPA1) channel, which in nociceptors mediates pain signals. Safranal by binding to specific cysteine/lysine residues, stimulates TRPA1, but not the TRP vanilloid 1 and 4 channels (TRPV1 and TRPV4), evoking calcium responses and currents in human cells and rat and mouse dorsal root ganglion (DRG) neurons. Genetic deletion or pharmacological blockade of TRPA1 attenuated safranal-evoked release of calcitonin gene-related peptide (CGRP) from rat and mouse dorsal spinal cord, and acute nociception in mice. Safranal contracted rat urinary bladder isolated strips in a TRPA1-dependent manner, behaving as a partial agonist. After exposure to safranal the ability of allyl isothiocyanate (TRPA1 agonist), but not that of capsaicin (TRPV1 agonist) or GSK1016790A (TRPV4 agonist), to evoke currents in DRG neurons, contraction of urinary bladder strips and CGRP release from spinal cord slices in rats, and acute nociception in mice underwent desensitization. As previously shown for other herbal extracts, including petasites or parthenolide, safranal might exert analgesic properties by partial agonism and selective desensitization of the TRPA1 channel.


Subject(s)
Analgesics/pharmacology , Crocus/chemistry , Cyclohexenes/pharmacology , Nociception/drug effects , TRPA1 Cation Channel/metabolism , Terpenes/pharmacology , Animals , Calcium Channels/metabolism , Cell Line , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Isothiocyanates/pharmacology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Sesquiterpenes/pharmacology , TRPV Cation Channels/metabolism
12.
Brain ; 139(Pt 5): 1361-77, 2016 05.
Article in English | MEDLINE | ID: mdl-26984186

ABSTRACT

Despite intense investigation, the mechanisms of the different forms of trigeminal neuropathic pain remain substantially unidentified. The transient receptor potential ankyrin 1 channel (encoded by TRPA1) has been reported to contribute to allodynia or hyperalgesia in some neuropathic pain models, including those produced by sciatic nerve constriction. However, the role of TRPA1 and the processes that cause trigeminal pain-like behaviours from nerve insult are poorly understood. The role of TRPA1, monocytes and macrophages, and oxidative stress in pain-like behaviour evoked by the constriction of the infraorbital nerve in mice were explored. C57BL/6 and wild-type (Trpa1(+/+)) mice that underwent constriction of the infraorbital nerve exhibited prolonged (20 days) non-evoked nociceptive behaviour and mechanical, cold and chemical hypersensitivity in comparison to sham-operated mice (P < 0.05-P < 0.001). Both genetic deletion of Trpa1 (Trpa1(-/-)) and pharmacological blockade (HC-030031 and A-967079) abrogated pain-like behaviours (both P < 0.001), which were abated by the antioxidant, α-lipoic acid, and the nicotinamide adenine dinucleotide phosphate oxidase inhibitor, apocynin (both P < 0.001). Nociception and hypersensitivity evoked by constriction of the infraorbital nerve was associated with intra- and perineural monocytic and macrophagic invasion and increased levels of oxidative stress by-products (hydrogen peroxide and 4-hydroxynonenal). Attenuation of monocyte/macrophage increase by systemic treatment with an antibody against the monocyte chemoattractant chemokine (C-C motif) ligand 2 (CCL2) or the macrophage-depleting agent, clodronate (both P < 0.05), was associated with reduced hydrogen peroxide and 4-hydroxynonenal perineural levels and pain-like behaviours (all P < 0.01), which were abated by perineural administration of HC-030031, α-lipoic acid or the anti-CCL2 antibody (all P < 0.001). The present findings propose that, in the constriction of the infraorbital nerve model of trigeminal neuropathic pain, pain-like behaviours are entirely mediated by the TRPA1 channel, targeted by increased oxidative stress by-products released from monocytes and macrophages clumping at the site of nerve injury.


Subject(s)
Hyperalgesia/physiopathology , Macrophages/drug effects , Monocytes/drug effects , Neuralgia/physiopathology , Oxidative Stress/drug effects , Transient Receptor Potential Channels/physiology , Acetanilides/antagonists & inhibitors , Acetanilides/pharmacology , Acetophenones/pharmacology , Animals , Chemokine CCL2/antagonists & inhibitors , Clodronic Acid/pharmacology , Hyperalgesia/metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout , Monocytes/metabolism , Neuralgia/metabolism , Oximes/antagonists & inhibitors , Oximes/pharmacology , Purines/antagonists & inhibitors , Purines/pharmacology , TRPA1 Cation Channel , Thioctic Acid/pharmacology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
13.
Eur J Nucl Med Mol Imaging ; 43(3): 499-508, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26341365

ABSTRACT

PURPOSE: The aim of this study was to evaluate the supportive role of molecular and structural biomarkers (CSF protein levels, FDG PET and MRI) in the early differential diagnosis of dementia in a large sample of patients with neurodegenerative dementia, and in determining the risk of disease progression in subjects with mild cognitive impairment (MCI). METHODS: We evaluated the supportive role of CSF Aß42, t-Tau, p-Tau levels, conventional brain MRI and visual assessment of FDG PET SPM t-maps in the early diagnosis of dementia and the evaluation of MCI progression. RESULTS: Diagnosis based on molecular biomarkers showed the best fit with the final diagnosis at a long follow-up. FDG PET SPM t-maps had the highest diagnostic accuracy in Alzheimer's disease and in the differential diagnosis of non-Alzheimer's disease dementias. The p-tau/Aß42 ratio was the only CSF biomarker providing a significant classification rate for Alzheimer's disease. An Alzheimer's disease-positive metabolic pattern as shown by FDG PET SPM in MCI was the best predictor of conversion to Alzheimer's disease. CONCLUSION: In this clinical setting, FDG PET SPM t-maps and the p-tau/Aß42 ratio improved clinical diagnostic accuracy, supporting the importance of these biomarkers in the emerging diagnostic criteria for Alzheimer's disease dementia. FDG PET using SPM t-maps had the highest predictive value by identifying hypometabolic patterns in different neurodegenerative dementias and normal brain metabolism in MCI, confirming its additional crucial exclusionary role.


Subject(s)
Alzheimer Disease/diagnosis , Biomarkers/cerebrospinal fluid , Aged , Amyloid beta-Peptides/cerebrospinal fluid , Cognitive Dysfunction/diagnosis , Diagnosis, Differential , Disease Progression , Female , Fluorodeoxyglucose F18/chemistry , Frontotemporal Lobar Degeneration/diagnosis , Humans , Lewy Body Disease/diagnosis , Magnetic Resonance Imaging , Male , Middle Aged , Peptide Fragments/cerebrospinal fluid , Positron-Emission Tomography , Predictive Value of Tests , Reproducibility of Results , tau Proteins/cerebrospinal fluid
14.
Arch Phys Med Rehabil ; 95(6): 1141-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24625546

ABSTRACT

OBJECTIVES: To assess the efficacy of high-frequency (20 Hz) brain stimulation on lower limb motor function in subjects with chronic (> 6 mo) subcortical stroke. DESIGN: Double-blind, placebo-controlled crossover study. SETTING: University hospital. PARTICIPANTS: Right-handed subjects (N=10) affected by a first-ever subcortical stroke in the territory of the middle cerebral artery were included in this study. INTERVENTIONS: Repetitive transcranial magnetic stimulation (rTMS) was delivered with the H-coil, specifically designed to target deeper and larger brains regions. Each subject received both real and sham rTMS in a random sequence. The 2 rTMS cycles (real or sham) were composed of 11 sessions each, administered over 3 weeks and separated by a 4-week washout period. MAIN OUTCOME MEASURES: Lower limb functions were assessed by the lower limb Fugl-Meyer scale, the 10-m walk test, and the 6-minute walk test before and 1 day after the end of each treatment period, as well as at a 4-week follow-up. RESULTS: Real rTMS treatment was associated with a significant improvement in lower limb motor function. This effect persisted over time (follow-up) and was significantly greater than that observed with sham stimulation. A significant increase in walking speed was also found after real rTMS, but this effect did not reach statistical significance in comparison with the sham stimulation. CONCLUSIONS: These data demonstrated that 3 weeks of high-frequency deep rTMS could induce long-term improvements in lower limb functions in the chronic poststroke period, lasting at least 1 month after the end of the treatment.


Subject(s)
Lower Extremity/physiopathology , Motor Skills/physiology , Stroke Rehabilitation , Stroke/diagnosis , Transcranial Magnetic Stimulation/methods , Analysis of Variance , Chronic Disease , Cross-Over Studies , Disability Evaluation , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Pilot Projects , Recovery of Function , Reference Values , Severity of Illness Index , Time Factors , Transcranial Magnetic Stimulation/instrumentation , Treatment Outcome , Walking/physiology
15.
Mediators Inflamm ; 2014: 805198, 2014.
Article in English | MEDLINE | ID: mdl-25165414

ABSTRACT

The extracellular concentration of adenosine in the brain increases dramatically during ischemia. Adenosine A(2A) receptor is expressed in neurons and glial cells and in inflammatory cells (lymphocytes and granulocytes). Recently, adenosine A(2A) receptor emerged as a potential therapeutic attractive target in ischemia. Ischemia is a multifactorial pathology characterized by different events evolving in the time. After ischemia the early massive increase of extracellular glutamate is followed by activation of resident immune cells, that is, microglia, and production or activation of inflammation mediators. Proinflammatory cytokines, which upregulate cell adhesion molecules, exert an important role in promoting recruitment of leukocytes that in turn promote expansion of the inflammatory response in ischemic tissue. Protracted neuroinflammation is now recognized as the predominant mechanism of secondary brain injury progression. A(2A) receptors present on central cells and on blood cells account for important effects depending on the time-related evolution of the pathological condition. Evidence suggests that A(2A) receptor antagonists provide early protection via centrally mediated control of excessive excitotoxicity, while A(2A) receptor agonists provide protracted protection by controlling massive blood cell infiltration in the hours and days after ischemia. Focus on inflammatory responses provides for adenosine A(2A) receptor agonists a wide therapeutic time-window of hours and even days after stroke.


Subject(s)
Brain Ischemia/immunology , Brain Ischemia/metabolism , Receptor, Adenosine A2A/metabolism , Animals , Cytokines/metabolism , Humans
16.
Glia ; 61(7): 1155-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23640798

ABSTRACT

In the developing and mature central nervous system, NG2 expressing cells comprise a population of cycling oligodendrocyte progenitor cells (OPCs) that differentiate into mature, myelinating oligodendrocytes (OLGs). OPCs are also characterized by high motility and respond to injury by migrating into the lesioned area to support remyelination. K(+) currents in OPCs are developmentally regulated during differentiation. However, the mechanisms regulating these currents at different stages of oligodendrocyte lineage are poorly understood. Here we show that, in cultured primary OPCs, the purinergic G-protein coupled receptor GPR17, that has recently emerged as a key player in oligodendrogliogenesis, crucially regulates K(+) currents. Specifically, receptor stimulation by its agonist UDP-glucose enhances delayed rectifier K(+) currents without affecting transient K(+) conductances. This effect was observed in a subpopulation of OPCs and immature pre-OLGs whereas it was absent in mature OLGs, in line with GPR17 expression, that peaks at intermediate phases of oligodendrocyte differentiation and is thereafter downregulated to allow terminal maturation. The effect of UDP-glucose on K(+) currents is concentration-dependent, blocked by the GPR17 antagonists MRS2179 and cangrelor, and sensitive to the K(+) channel blocker tetraethyl-ammonium, which also inhibits oligodendrocyte maturation. We propose that stimulation of K(+) currents is responsible for GPR17-induced oligodendrocyte differentiation. Moreover, we demonstrate, for the first time, that GPR17 activation stimulates OPC migration, suggesting an important role for this receptor after brain injury. Our data indicate that modulation of GPR17 may represent a strategy to potentiate the post-traumatic response of OPCs under demyelinating conditions, such as multiple sclerosis, stroke, and brain trauma.


Subject(s)
Cell Differentiation/drug effects , Cell Movement/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Uridine Diphosphate Glucose/pharmacology , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Animals, Newborn , Antigens/metabolism , Brain/cytology , Calcium/metabolism , Cells, Cultured , Glial Fibrillary Acidic Protein/metabolism , Membrane Potentials/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Proteoglycans/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Stem Cells , Tetraethylammonium/pharmacology , Tetrodotoxin/pharmacology
17.
Biomolecules ; 13(6)2023 05 27.
Article in English | MEDLINE | ID: mdl-37371474

ABSTRACT

In recent years, the use of multi-target compounds has become an increasingly pursued strategy to treat complex pathologies, including cerebral ischemia. Adenosine and its receptors (A1AR, A2AAR, A2BAR, A3AR) are known to play a crucial role in synaptic transmission either in normoxic or ischemic-like conditions. Previous data demonstrate that the selective antagonism of A2AAR or A2BAR delays anoxic depolarization (AD) appearance, an unequivocal sign of neuronal injury induced by a severe oxygen-glucose deprivation (OGD) insult in the hippocampus. Furthermore, the stimulation of A2AARs or A2BARs by respective selective agonists, CGS21680 and BAY60-6583, increases pre-synaptic neurotransmitter release, as shown by the decrease in paired-pulse facilitation (PPF) at Schaffer collateral-CA1 synapses. In the present research, we investigated the effect/s of the newly synthesized dual A2AAR/A2BAR antagonist, P626, in preventing A2AAR- and/or A2BAR-mediated effects by extracellular recordings of synaptic potentials in the CA1 rat hippocampal slices. We demonstrated that P626 prevented PPF reduction induced by CGS21680 or BAY60-6583 and delayed, in a concentration-dependent manner, AD appearance during a severe OGD. In conclusion, P626 may represent a putative neuroprotective compound for stroke treatment with the possible translational advantage of reducing side effects and bypassing differences in pharmacokinetics due to combined treatment.


Subject(s)
Adenosine , Hippocampus , Rats , Animals , Adenosine/pharmacology , Ischemia , Synaptic Transmission , Hypoxia , Oxygen/pharmacology , Neuronal Plasticity , Glucose/pharmacology
18.
J Cereb Blood Flow Metab ; 43(7): 1077-1088, 2023 07.
Article in English | MEDLINE | ID: mdl-36823998

ABSTRACT

Multicentre preclinical randomized controlled trials (pRCTs) are a valuable tool to improve experimental stroke research, but are challenging and therefore underused. A common challenge regards the standardization of procedures across centres. We here present the harmonization phase for the quantification of sensorimotor deficits by composite neuroscore, which was the primary outcome of two multicentre pRCTs assessing remote ischemic conditioning in rodent models of ischemic stroke. Ischemic stroke was induced by middle cerebral artery occlusion for 30, 45 or 60 min in mice and 50, 75 or 100 min in rats, allowing sufficient variability. Eleven animals per species were video recorded during neurobehavioural tasks and evaluated with neuroscore by eight independent raters, remotely and blindly. We aimed at reaching an intraclass correlation coefficient (ICC) ≥0.60 as satisfactory interrater agreement. After a first remote training we obtained ICC = 0.50 for mice and ICC = 0.49 for rats. Errors were identified in animal handling and test execution. After a second remote training, we reached the target interrater agreement for mice (ICC = 0.64) and rats (ICC = 0.69). In conclusion, a multi-step, online harmonization phase proved to be feasible, easy to implement and highly effective to align each centre's behavioral evaluations before project's interventional phase.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Rats , Mice , Animals , Infarction, Middle Cerebral Artery , Randomized Controlled Trials as Topic
19.
Br J Pharmacol ; 179(20): 4844-4856, 2022 10.
Article in English | MEDLINE | ID: mdl-35817954

ABSTRACT

BACKGROUND AND PURPOSE: Ischaemia is known to cause massive neuronal depolarization, termed anoxic depolarization (AD), due to energy failure and loss of membrane ion gradients. The neuromodulator adenosine accumulates extracellularly during ischaemia and activates four metabotropic receptors: A1 , A2A , A2B and A3 . Striatal medium spiny neurons (MSNs) express high levels of A2A receptors and are particularly vulnerable to ischaemic insults. A2A Receptor blockade reduces acute striatal post-ischaemic damage but the cellular mechanisms involved are still unknown. EXPERIMENTAL APPROACH: We performed patch-clamp recordings of MSNs in rat striatal slices subjected to oxygen and glucose deprivation (OGD) to investigate the effects of A2A receptor ligands or ion channel blockers on AD and OGD-induced ionic imbalance, measured as a positive shift in Erev of ramp currents. KEY RESULTS: Our data indicate that the A2A receptor antagonist SCH58261 (10 µM) significantly attenuated ionic imbalance and AD appearance in MSNs exposed to OGD. The K+ channel blocker Ba2+ (2 mM) or the Na+ channel blocker tetrodotoxin (1 µM) exacerbated and attenuated, respectively, OGD-induced changes. Spontaneous excitatory post-synaptic current (sEPSC) analysis in MSNs revealed that the A2A receptor agonist CGS21680 (1 µM) prevented OGD-induced decrease of sEPSCs within the first 5 min of the insult, an effect shared by the K+ channel blocker Ba2+ , indicating facilitated glutamate release. CONCLUSION AND IMPLICATIONS: Adenosine, released during striatal OGD, activates A2A receptors that may exacerbate OGD-induced damage through K+ channel inhibition. Our results could help to develop A2A receptor-selective therapeutic tools for the treatment of brain ischaemia.


Subject(s)
Glucose , Oxygen , Adenosine/pharmacology , Animals , Glucose/pharmacology , Glutamic Acid/pharmacology , Ion Channels , Ischemia , Ligands , Neurons , Oxygen/metabolism , Rats , Rats, Wistar , Receptor, Adenosine A2A/metabolism , Tetrodotoxin
SELECTION OF CITATIONS
SEARCH DETAIL