Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Neuroinflammation ; 15(1): 164, 2018 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-29803222

RESUMO

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.


Assuntos
Trifosfato de Adenosina/farmacologia , Interleucina-1beta/metabolismo , Microglia/efeitos dos fármacos , Proteína Amiloide A Sérica/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Células Cultivadas , Interleucina-1beta/genética , Lipopolissacarídeos/farmacologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , RNA Mensageiro/metabolismo , Ratos , Fatores de Tempo
2.
Mediators Inflamm ; 2018: 2868702, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29576743

RESUMO

Several studies suggest that curcumin and related compounds possess antioxidant and anti-inflammatory properties including modulation of lipopolysaccharide- (LPS-) mediated signalling in macrophage cell models. We here investigated the effects of curcumin and the two structurally unrelated analogues GG6 and GG9 in primary human blood-derived macrophages as well as the signalling pathways involved. Macrophages differentiated from peripheral blood monocytes for 7 days were activated with LPS or selective Toll-like receptor agonists for 24 h. The effects of test compounds on cytokine production and immunophenotypes evaluated as CD80+/CCR2+ and CD206+/CD163+ subsets were examined by ELISA and flow cytometry. Signalling pathways were probed by Western blot. Curcumin (2.5-10 µM) failed to suppress LPS-induced inflammatory responses. While GG6 reduced LPS-induced IκB-α degradation and showed a trend towards reduced interleukin-1ß release, GG9 prevented the increase in proinflammatory CD80+ macrophage subset, downregulation of the anti-inflammatory CD206+/CD163+ subset, increase in p38 phosphorylation, and increase in cell-bound and secreted interleukin-1ß stimulated by LPS, at least in part through signalling pathways not involving Toll-like receptor 4 and nuclear factor-κB. Thus, the curcumin analogue GG9 attenuated the LPS-induced inflammatory response in human blood-derived macrophages and may therefore represent an attractive chemical template for macrophage pharmacological targeting.


Assuntos
Curcumina/análogos & derivados , Lipopolissacarídeos/farmacologia , Western Blotting , Células Cultivadas , Curcumina/química , Curcumina/farmacologia , Diarileptanoides , Humanos , Imunofenotipagem , Interleucina-1beta/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Immunology ; 151(1): 1-15, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28112808

RESUMO

Neurotrophic factors comprise a broad family of biomolecules - most of which are peptides or small proteins - that support the growth, survival and differentiation of both developing and mature neurons. The prototypical example and best-characterized neurotrophic factor is nerve growth factor (NGF), which is widely recognized as a target-derived factor responsible for the survival and maintenance of the phenotype of specific subsets of peripheral neurons and basal forebrain cholinergic nuclei during development and maturation. In addition to being active in a wide array of non-nervous system cells, NGF is also synthesized by a range of cell types not considered as classical targets for innervation by NGF-dependent neurons; these include cells of the immune-haematopoietic lineage and populations in the brain involved in neuroendocrine functions. NGF concentrations are elevated in numerous inflammatory and autoimmune states such as multiple sclerosis, chronic arthritis, systemic lupus erythematosus and mastocytosis, in conjunction with increased accumulation of mast cells. Intriguingly, NGF seems to be linked also with diabetic pathology and insulin homeostasis. Mast cells and NGF appear involved in neuroimmune interactions and tissue inflammation. As mast cells are capable of producing and responding to NGF, this suggests that alterations in mast cell behaviour could provoke maladaptive neuroimmune tissue responses, including those of an autoimmune nature. Moreover, NGF exerts a modulatory role on sensory nociceptive nerve physiology in the adult, which appears to correlate with hyperalgesic phenomena occurring in tissue inflammation. NGF can therefore be viewed as a multifactorial modulator of neuro-immune-endocrine functions.


Assuntos
Hiperalgesia/imunologia , Mastócitos/fisiologia , Fatores de Crescimento Neural/metabolismo , Neuroimunomodulação , Células Receptoras Sensoriais/fisiologia , Adulto , Animais , Doenças Autoimunes , Sistema Endócrino/imunologia , Regulação da Expressão Gênica no Desenvolvimento , Homeostase , Humanos , Inflamação , Insulina/metabolismo
4.
Pain Pract ; 17(4): 522-532, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28039964

RESUMO

Rheumatic and joint diseases, as exemplified by osteoarthritis and rheumatoid arthritis, are among the most widespread painful and disabling pathologies across the globe. Given the continuing rise in life expectancy, their prevalence is destined to grow. Osteoarthritis, a degenerative joint disease, is, in particular, on its way to becoming the fourth leading cause of disability worldwide by 2020, with the rising incidence of obesity in addition to age being important factors. It is estimated that 25% of osteoarthritic individuals are unable to perform daily activities. Accompanying osteoarthritis is rheumatoid arthritis, which is a chronic systemic disease that often causes pain and deformity. At least 50% of those affected are unable to remain gainfully employed within 10 years of disease onset. A growing body of evidence now points to inflammation, locally and more systemically, as a promoter of damage to joints and bones, as well as joint-related functional deficits. The pathogenesis underlying joint diseases remains unclear; however, it is currently believed that cross-talk between cartilage and subchondral bone-and loss of balance between these two structures in joint diseases-is a critical element. This view is amplified by the presence of mast cells, whose dysregulation is associated with alterations of junction structures (cartilage, bone, synovia, matrix, nerve endings, and blood vessels). In addition, persistent activation of mast cells facilitates the development of spinal neuroinflammation mediated through their interaction with microglia. Unfortunately, current treatment strategies for rheumatic and articular disease are symptomatic and do little to limit disease progression. Research now should be directed at therapeutic modalities that target osteoarticular structural elements and thereby delaying disease progression and joint replacement.


Assuntos
Cartilagem Articular/patologia , Artropatias/diagnóstico , Mastócitos/patologia , Artrite Reumatoide/diagnóstico , Artrite Reumatoide/epidemiologia , Artrite Reumatoide/imunologia , Cartilagem Articular/imunologia , Humanos , Inflamação/diagnóstico , Inflamação/epidemiologia , Inflamação/imunologia , Artropatias/epidemiologia , Artropatias/imunologia , Mastócitos/imunologia , Osteoartrite/diagnóstico , Osteoartrite/epidemiologia , Osteoartrite/imunologia
5.
J Neuroinflammation ; 12: 244, 2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-26714634

RESUMO

BACKGROUND: Toll-like receptor (TLR) activation on microglia and astrocytes are key elements in neuroinflammation which accompanies a number of neurological disorders. While TLR activation on glia is well-established to up-regulate pro-inflammatory mediator expression, much less is known about how ligand engagement of one TLR may affect expression of other TLRs on microglia and astrocytes. METHODS: In the present study, we evaluated the effects of agonists for TLR2 (zymosan), TLR3 (polyinosinic-polycytidylic acid (poly(I:C)), a synthetic analogue of double-stranded RNA) and TLR4 (lipopolysaccaride (LPS)) in influencing expression of their cognate receptor as well as that of the other TLRs in cultures of rat cortical purified microglia (>99.5 %) and nominally microglia-free astrocytes. Elimination of residual microglia (a common contaminant of astrocyte cultures) was achieved by incubation with the lysosomotropic agent L-leucyl-L-leucine methyl ester (L-LME). RESULTS: Flow cytometric analysis confirmed the purity (essentially 100 %) of the obtained microglia, and up to 5 % microglia contamination of astrocytes. L-LME treatment effectively removed microglia from the latter (real-time polymerase chain reaction). The three TLR ligands robustly up-regulated gene expression for pro-inflammatory markers (interleukin-1 and interleukin-6, tumor necrosis factor) in microglia and enriched, but not purified, astrocytes, confirming cellular functionality. LPS, zymosan and poly(I:C) all down-regulated TLR4 messenger RNA (mRNA) and up-regulated TLR2 mRNA at 6 and 24 h. In spite of their inability to elaborate pro-inflammatory mediator output, the nominally microglia-free astrocytes (>99 % purity) also showed similar behaviours to those of microglia, as well as changes in TLR3 gene expression. LPS interaction with TLR4 activates downstream mitogen-activated protein kinase and nuclear factor-κB signalling pathways and subsequently causes inflammatory mediator production. The effects of LPS on TLR2 mRNA in both cell populations were antagonized by a nuclear factor-κB inhibitor. CONCLUSIONS: TLR2 and TLR4 activation in particular, in concert with microglia and astrocytes, comprise key elements in the initiation and maintenance of neuropathic pain. The finding that both homologous (zymosan) and heterologous (LPS, poly(I:C)) TLR ligands are capable of regulating TLR2 gene expression, in particular, may have important implications in understanding the relative contributions of different TLRs in neurological disorders associated with neuroinflammation.


Assuntos
Astrócitos/metabolismo , Córtex Cerebral/metabolismo , Microglia/metabolismo , Receptores Toll-Like/biossíntese , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Técnicas de Cocultura , Regulação da Expressão Gênica , Ligantes , Microglia/efeitos dos fármacos , Poli I-C/metabolismo , Poli I-C/farmacologia , Ratos , Receptores Toll-Like/agonistas , Zimosan/metabolismo , Zimosan/farmacologia
6.
Inflammopharmacology ; 23(2-3): 127-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24862356

RESUMO

This is a reply to a recently published Commentary: "Palmitoylethanolamide: problems regarding micronization, ultra-micronization and additives" Inflammopharmacology DOI: 10.1007/s10787-014-0202-3 , written in relation to our review article: Skaper SD, Facci L, Fusco M, della Valle MF, Zusso M, Costa B, Giusti P (2014) "Palmitoylethanolamide, a naturally occurring disease-modifying agent in neuropathic pain" Inflammopharmacology 22:79-94 DOI: 10.1007/s10787-013-0191-7 . We believe that the Commentary by Kriek contains a number of erroneous statements and misinterpretations of the published scientific/medical literature which our reply shall elaborate on. Further, the writer of the Commentary has a direct connection to a company, JP Russell Science Ltd that sells palmitoylethanolamide. The take-home message of our review remains as originally stated: "Collectively, the findings presented here propose that palmitoylethanolamide merits further consideration as a disease-modifying agent for controlling inflammatory responses and related chronic and neuropathic pain".


Assuntos
Endocanabinoides/farmacologia , Endocanabinoides/uso terapêutico , Etanolaminas/farmacologia , Etanolaminas/uso terapêutico , Neuralgia/tratamento farmacológico , Ácidos Palmíticos/farmacologia , Ácidos Palmíticos/uso terapêutico , Animais , Humanos
7.
Immunology ; 141(3): 314-27, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24032675

RESUMO

Glia and microglia in particular elaborate pro-inflammatory molecules that play key roles in central nervous system (CNS) disorders from neuropathic pain and epilepsy to neurodegenerative diseases. Microglia respond also to pro-inflammatory signals released from other non-neuronal cells, mainly those of immune origin such as mast cells. The latter are found in most tissues, are CNS resident, and traverse the blood-spinal cord and blood-brain barriers when barrier compromise results from CNS pathology. Growing evidence of mast cell-glia communication opens new perspectives for the development of therapies targeting neuroinflammation by differentially modulating activation of non-neuronal cells that normally control neuronal sensitization - both peripherally and centrally. Mast cells and glia possess endogenous homeostatic mechanisms/molecules that can be up-regulated as a result of tissue damage or stimulation of inflammatory responses. Such molecules include the N-acylethanolamine family. One such member, N-palmitoylethanolamine is proposed to have a key role in maintenance of cellular homeostasis in the face of external stressors provoking, for example, inflammation. N-Palmitoylethanolamine has proven efficacious in mast-cell-mediated experimental models of acute and neurogenic inflammation. This review will provide an overview of recent progress relating to the pathobiology of neuroinflammation, the role of microglia, neuroimmune interactions involving mast cells and the possibility that mast cell-microglia cross-talk contributes to the exacerbation of acute symptoms of chronic neurodegenerative disease and accelerates disease progression, as well as promoting pain transmission pathways. We will conclude by considering the therapeutic potential of treating systemic inflammation or blockade of signalling pathways from the periphery to the brain in such settings.


Assuntos
Encéfalo/imunologia , Comunicação Celular , Encefalite/imunologia , Mastócitos/imunologia , Doenças Neurodegenerativas/imunologia , Neuroglia/imunologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Encefalite/metabolismo , Encefalite/patologia , Encefalite/terapia , Humanos , Mediadores da Inflamação/metabolismo , Mastócitos/metabolismo , Mastócitos/patologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Neuroglia/metabolismo , Neuroglia/patologia , Transdução de Sinais
8.
Gynecol Endocrinol ; 30(7): 472-7, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24811097

RESUMO

Inflammatory and neuroinflammatory processes are increasingly recognized as critical pathophysiologic steps in the development of multiple chronic diseases and in the etiology of persistent pain and depression. Mast cells are immune cells now viewed as cellular sensors in inflammation and immunity. When stimulated, mast cells release an array of mediators to orchestrate an inflammatory response. These mediators can directly initiate tissue responses on resident cells, and may also regulate the activity of other immune cells, including central microglia. New evidence supports the involvement of peripheral and central mast cells in the development of pain processes as well as in the transition from acute, to chronic and neuropathic pain. That behavioral and endocrine states can increase the number and activation of peripheral and brain mast cells suggests that mast cells represent the immune cells that peripherally and centrally coordinate inflammatory processes in neuropsychiatric diseases such as depression and anxiety which are associated with chronic pelvic pain. Given that increasing evidence supports the activated mast cell as a director of common inflammatory pathways/mechanisms contributing to chronic and neuropathic pelvic pain and comorbid neuropsychiatric diseases, mast cells may be considered a viable target for the multifactorial management of both pain and depression.


Assuntos
Transtorno Depressivo/patologia , Inflamação/patologia , Mastócitos/patologia , Dor Pélvica/patologia , Comorbidade , Transtorno Depressivo/metabolismo , Transtorno Depressivo/psicologia , Feminino , Humanos , Inflamação/metabolismo , Inflamação/psicologia , Mediadores da Inflamação/metabolismo , Mastócitos/metabolismo , Dor Pélvica/metabolismo , Dor Pélvica/psicologia
9.
Inflammopharmacology ; 22(2): 79-94, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24178954

RESUMO

Persistent pain affects nearly half of all people seeking medical care in the US alone, and accounts for at least $80 billion worth of lost productivity each year. Among all types of chronic pain, neuropathic pain stands out: this is pain resulting from damage or disease of the somatosensory nervous system, and remains largely untreatable. With few available treatment options, neuropathic pain represents an area of significant and growing unmet medical need. Current treatment of peripheral neuropathic pain involves several drug classes, including opioids, gabapentinoids, antidepressants, antiepileptic drugs, local anesthetics and capsaicin. Even so, less than half of patients achieve partial relief. This review discusses a novel approach to neuropathic pain management, based on knowledge of: the role of glia and mast cells in pain and neuroinflammation; the body's innate mechanisms to maintain cellular homeostasis when faced with external stressors provoking, for example, inflammation. The discovery that palmitoylethanolamide, a member of the N-acylethanolamine family which is produced from the lipid bilayer on-demand, is capable of exerting anti-allodynic and anti-hyperalgesic effects by down-modulating both microglial and mast cell activity has led to the application of this fatty acid amide in several clinical studies of neuropathic pain, with beneficial outcome and no indication of adverse effects at pharmacological doses. Collectively, the findings presented here propose that palmitoylethanolamide merits further consideration as a disease-modifying agent for controlling inflammatory responses and related chronic and neuropathic pain.


Assuntos
Endocanabinoides/farmacologia , Endocanabinoides/uso terapêutico , Etanolaminas/farmacologia , Etanolaminas/uso terapêutico , Neuralgia/tratamento farmacológico , Ácidos Palmíticos/farmacologia , Ácidos Palmíticos/uso terapêutico , Amidas , Animais , Humanos , Inflamação/tratamento farmacológico , Mastócitos/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Manejo da Dor/métodos
10.
FASEB J ; 26(8): 3103-17, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22516295

RESUMO

One of the more important recent advances in neuroscience research is the understanding that there is extensive communication between the immune system and the central nervous system (CNS). Proinflammatory cytokines play a key role in this communication. The emerging realization is that glia and microglia, in particular, (which are the brain's resident macrophages), constitute an important source of inflammatory mediators and may have fundamental roles in CNS disorders from neuropathic pain and epilepsy to neurodegenerative diseases. Microglia respond also to proinflammatory signals released from other non-neuronal cells, principally those of immune origin. Mast cells are of particular relevance in this context. These immunity-related cells, while resident in the CNS, are capable of migrating across the blood-spinal cord and blood-brain barriers in situations where the barrier is compromised as a result of CNS pathology. Emerging evidence suggests the possibility of mast cell-glia communications and opens exciting new perspectives for designing therapies to target neuroinflammation by differentially modulating the activation of non-neuronal cells normally controlling neuronal sensitization, both peripherally and centrally. This review aims to provide an overview of recent progress relating to the pathobiology of neuroinflammation, the role of microglia, neuroimmune interactions involving mast cells, in particular, and the possibility that mast cell-microglia crosstalk may contribute to the exacerbation of acute symptoms of chronic neurodegenerative disease and accelerate disease progression, as well as promote pain transmission pathways. We conclude by considering the therapeutic potential of treating systemic inflammation or blockade of signaling pathways from the periphery to the brain in such settings.


Assuntos
Mediadores da Inflamação/metabolismo , Inflamação/fisiopatologia , Mastócitos/fisiologia , Microglia/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Sistema Nervoso Central/patologia , Humanos , Imunidade Inata/fisiologia , Inflamação/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Neuroglia/fisiologia , Dor/fisiopatologia , Receptores Purinérgicos/fisiologia , Medula Espinal/fisiopatologia
11.
Neurochem Res ; 38(9): 1801-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23743620

RESUMO

Microglia can exacerbate central nervous system disorders, including stroke and chronic progressive neurodegenerative diseases such as Alzheimer disease. Mounting evidence points to ion channels expressed by microglia as contributing to these neuropathologies. The Chloride Intracellular Channel (CLIC) family represents a class of chloride intracellular channel proteins, most of which are localized to intracellular membranes. CLICs are unusual in that they possess both soluble and integral membrane forms. Amyloid ß-peptide (Aß) accumulation in plaques is a hallmark of familial Alzheimer disease. The truncated Aß25-35 species was shown previously to increase the expression of CLIC1 chloride conductance in cortical microglia and to provoke microglial neurotoxicity. However, the highly pathogenic and fibrillogenic full-length Aß1-42 species was not examined, nor was the potential role of CLIC1 in mediating microglial activation and neurotoxicity by other stimuli (e.g. ligands for the Toll-like receptors). In the present study, we utilized a two chamber Transwell™ cell culture system to allow separate treatment of microglia and neurons while examining the effect of pharmacological blockade of CLIC1 in protecting cortical neurons from toxicity caused by Aß1-42- and lipopolysaccaride-stimulated microglia. Presentation of Aß1-42 to the upper, microglia-containing chamber resulted in a progressive loss of neurons over 3 days. Neuronal cell injury was prevented by the CLIC1 ion channel blockers IAA-94 [(R(+)-[(6,7-dichloro-2-cyclopentyl-2,3-dihydro-2-methyl-1-oxo-1H-inden-5yl)-oxy] acetic acid)] and niflumic acid (2-{[3-(trifluoromethyl)phenyl]amino}nicotinic acid) when presented to the upper chamber only. Incubation of microglia with lipopolysaccharide plus interferon-γ led to neuronal cell injury which, however, was insensitive to inhibition by the CLIC1 channel blockers, suggesting a degree of selectivity in agents leading to CLIC1 activation.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Canais de Cloreto/fisiologia , Microglia/fisiologia , Fragmentos de Peptídeos/toxicidade , Animais , Células Cultivadas , Ratos
12.
FASEB J ; 24(2): 337-45, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19812374

RESUMO

Purine nucleotides are well established as extracellular signaling molecules. P2X receptors are ATP-gated cation channels that mediate fast excitatory transmission in diverse regions of the brain and spinal cord. Several P2X receptor subtypes, including P2X(7), have the unusual property of changing their ion selectivity during prolonged exposure to ATP, which results in progressive dilation of the channel pore and the development of permeability to molecules as large as 900 Da. The P2X(7) receptor was originally described in cells of hematopoietic origin, including macrophages, microglia, and certain lymphocytes, and mediates the influx of Ca(2+) and Na(+) ions, as well as the release of proinflammatory cytokines. P2X(7) receptors may affect neuronal cell death through their ability to regulate the processing and release of interleukin-1beta, a key mediator in neurodegeneration, chronic inflammation, and chronic pain. Activation of P2X(7) receptors provides an inflammatory stimulus, and P2X(7) receptor-deficient mice have substantially attenuated inflammatory responses, including models of neuropathic and chronic inflammatory pain. Moreover, P2X(7) receptor activity, by regulating the release of proinflammatory cytokines, may be involved in the pathophysiology of depression. The P2X(7) receptor may thus represent a critical communication link between the nervous and immune systems, while providing a target for therapeutic exploitation. This review discusses the current biology and cellular signaling pathways of P2X(7) receptor function, as well as insights into the role for this receptor in neurological/psychiatric diseases, outstanding questions, and the therapeutic potential of P2X(7) receptor antagonism.


Assuntos
Receptores Purinérgicos P2/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Trifosfato de Adenosina/fisiologia , Animais , Citocinas/fisiologia , Depressão/etiologia , Depressão/fisiopatologia , Humanos , Camundongos , Doenças do Sistema Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Dor/fisiopatologia , Ratos , Receptores Purinérgicos P2X7 , Transdução de Sinais/fisiologia
13.
Neurodegener Dis ; 8(1-2): 15-24, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20689247

RESUMO

BACKGROUND: Cleavage of the amyloid precursor protein (APP) by ß-site APP-cleaving enzyme and γ-secretase results in the generation of amyloid-ß (Aß) peptides that aggregate and deposit as senile plaques in brains of Alzheimer disease patients. Due to the fundamental role γ-secretase plays in the proteolysis of a number of proteins including Notch, pharmacological inhibition of γ-secretase has been associated with mechanism-based toxicities. Therefore, efforts have focussed on the modulation of γ-secretase activity to selectively decrease levels of Aß42 peptide while avoiding deleterious activity on Notch processing. OBJECTIVE: Here, we describe the in vitro and in vivo characterisation of a novel γ-secretase modulator, GSM-10h, and investigate the potential for shorter Aß peptides to induce neurotoxicity in rat primary cortical neurons. METHODS: The effect of GSM-10h on Aß levels was investigated in SH-SY5Y cells expressing mutant APP and in TASTPM mice expressing APP and presenilin-1 mutant transgenes. The effect of GSM-10h on Notch processing was also determined. RESULTS: In cells, GSM-10h decreased levels of Aß42 while concomitantly increasing levels of Aß38 in the absence of effects on Aß40 levels. In TASTPM mice, GSM-10h effectively lowered brain Aß42 and increased brain Aß38, with no effect on Notch signalling. Unlike Aß42, which causes neuronal cell death, neither Aß37 nor Aß38 were neurotoxic. CONCLUSIONS: These findings confirm GSM-10h exhibits the profile of a γ-secretase modulator. In addition, TASTPM mice are shown to be responsive to treatment with a γ-secretase modulator, thereby highlighting the utility of this bitransgenic mouse model in drug discovery efforts focussed on the development of γ-secretase modulators.


Assuntos
Acetatos/farmacologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/biossíntese , Fragmentos de Peptídeos/antagonistas & inibidores , Piperidinas/farmacologia , Presenilina-1/biossíntese , Precursor de Proteína beta-Amiloide/genética , Animais , Morte Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Presenilina-1/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transgenes/genética
14.
Prog Brain Res ; 266: 1-73, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34689857

RESUMO

Military personnel deployed in combat operations are highly prone to develop Parkinson's disease (PD) in later lives. PD largely involves dopaminergic pathways with hallmarks of increased alpha synuclein (ASNC), and phosphorylated tau (p-tau) in the cerebrospinal fluid (CSF) precipitating brain pathology. However, increased histaminergic nerve fibers in substantia nigra pars Compacta (SNpc), striatum (STr) and caudate putamen (CP) associated with upregulation of Histamine H3 receptors and downregulation of H4 receptors in human cases of PD is observed in postmortem cases. These findings indicate that modulation of histamine H3 and H4 receptors and/or histaminergic transmission may induce neuroprotection in PD induced brain pathology. In this review effects of a potent histaminergic H3 receptor inverse agonist BF-2549 or clobenpropit (CLBPT) partial histamine H4 agonist with H3 receptor antagonist, in association with monoclonal anti-histamine antibodies (AHmAb) in PD brain pathology is discussed based on our own observations. Our investigation shows that chronic administration of conventional or TiO2 nanowired BF 2649 (1mg/kg, i.p.) or CLBPT (1mg/kg, i.p.) once daily for 1 week together with nanowired delivery of HAmAb (25µL) significantly thwarted ASNC and p-tau levels in the SNpC and STr and reduced PD induced brain pathology. These observations are the first to show the involvement of histamine receptors in PD and opens new avenues for the development of novel drug strategies in clinical strategies for PD, not reported earlier.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Corpo Estriado , Histamina , Humanos , Imidazóis , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson/tratamento farmacológico , Receptores Histamínicos H4 , Tioureia/análogos & derivados
15.
Neurochem Res ; 35(4): 588-97, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19941067

RESUMO

Apoptosis-associated tyrosine kinase (AATYK) is up-regulated by phosphorylation in cultured cerebellar granule neurons (CGN) undergoing apoptosis upon switch to low KCl-containing medium. However, the underlying signaling pathways remain to be fully characterized. When CGN at culture day 7 were switched from 25 mM KCl (K25) to 5 mM (K5) medium, AATYK band migration on SDS-PAGE shifted to a more slowly migrating position expected for the hyperphosphorylated protein. The apoptosis-inducing agent C(2)-ceramide also caused a mobility shift of the AATYK protein. Exposing CGN (K25) to L-type voltage-dependent Ca(2+) channel antagonists shifted the AATYK band to the K5-induced position, while the Ca(2+) channel activator FPL-64176 had the contrary effect. FK-506, a calcineurin inhibitor caused AATYK hyperphosphorylation under high KCl conditions. CGN death in K5 medium is linked to inhibition of the PI 3-kinase/Akt survival pathway and concomitant activation of the pro-apoptotic downstream target glycogen synthase kinase-3 (GSK-3). GSK-3 inhibitors blocked the K5-induced mobility shift of AATYK. Moreover, CGN cultured from AATYK-deficient mice remained sensitive to death in K5 medium. Thus, AATYK activation may not be a physiologically relevant principal regulatory target of the GSK-3 death pathway in KCl-deprived CGN.


Assuntos
Apoptose , Neurônios/citologia , Proteínas Tirosina Quinases/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Western Blotting , Sinalização do Cálcio , Células Cultivadas , Cerebelo/citologia , Cerebelo/enzimologia , Grânulos Citoplasmáticos/enzimologia , Eletroforese em Gel de Poliacrilamida , Camundongos , Neurônios/enzimologia , Fosforilação , Potássio/metabolismo , Ratos
16.
J Orthop Translat ; 20: 14-24, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31908929

RESUMO

Functional restoration after spinal cord injury (SCI) is one of the most challenging tasks in neurological clinical practice. With a view to exploring effective neurorestorative methods in the acute, subacute, and chronic phases of SCI, "Clinical Therapeutic Guidelines of Neurorestoration for Spinal Cord Injury (China Version 2016)" was first â€‹proposed in 2016 by the Chinese Association of Neurorestoratology (CANR). Given the rapid advances in this field in recent years, the International Association of Neurorestoratology (IANR) and CANR formed and approved the "Clinical Neurorestorative Therapeutic Guidelines for Spinal Cord Injury (IANR/CANR version 2019)". These guidelines mainly introduce restoring damaged neurological structure and functions by varying neurorestorative strategies in acute, subacute, and chronic phases of SCI. These guidelines can provide a neurorestorative therapeutic standard or reference for clinicians and researchers in clinical practice to maximally restore functions of patients with SCI and improve their quality of life. THE TRANSLATIONAL POTENTIAL OF THIS ARTICLE: This guideline provided comprehensive management strategies for SCI, which contains the evaluation and diagnosis, pre-hospital first aid, treatments, rehabilitation training, and complications management. Nowadays, amounts of neurorestorative strategies have been demonstrated to be benefit in promoting the functional recovery and improving the quality of life for SCI patients by clinical trials. Also, the positive results of preclinical research provided lots of new neurorestorative strategies for SCI treatment. These promising neurorestorative strategies are worthy of translation in the future and can promote the advancement of SCI treatments.

17.
J Neurochem ; 109(6): 1680-90, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19457136

RESUMO

Ciliary neurotrophic factor (CNTF) is a multifunctional cytokine that can regulate the survival and differentiation of many types of developing and adult neurons. CNTF prevents the degeneration of motor neurons after axotomy and in mouse mutant progressive motor neuronopathy, which has encouraged trials of CNTF for human motor neuron disease. Given systemically, however, CNTF causes severe side effects, including cachexia and a marked immune response, which has limited its clinical application. The present work describes a novel approach for administering recombinant human CNTF (rhCNTF) while conserving neurotrophic activity and avoiding deleterious side effects. rhCNTF was fused to a protein transduction domain derived from the human immunodeficiency virus-1 TAT (transactivator) protein. The resulting fusion protein (TAT-CNTF) crosses the plasma membrane within minutes and displays a nuclear localization. TAT-CNTF was equipotent to rhCNTF in supporting the survival of cultured chicken embryo dorsal root ganglion neurons. Local or subcutaneous administration of TAT-CNTF, like rhCNTF rescued motor neurons from death in neonatal rats subjected to sciatic nerve transection. In contrast to subcutaneous rhCNTF, which caused a 20-30% decrease in body weight in neonatal rats between postnatal days 2 and 7 together with a considerable fat mobilization in brown adipose tissue, TAT-CNTF lacked such side effects. Together, these results indicate that rhCNTF fused with the protein transduction domain/TAT retains neurotrophic activity in the absence of CNTFs cytokine-like side effects and may be a promising candidate for the treatment of motor neuron and other neurodegenerative diseases.


Assuntos
Fator Neurotrófico Ciliar/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Neuropatia Ciática/tratamento farmacológico , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Animais , Animais Recém-Nascidos , Axotomia/métodos , Peso Corporal/efeitos dos fármacos , Contagem de Células/métodos , Células Cultivadas , Embrião de Galinha , Fator Neurotrófico Ciliar/metabolismo , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Gânglios Espinais/citologia , Proteínas de Fluorescência Verde/genética , Humanos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/uso terapêutico , Fator de Transcrição STAT3/metabolismo , Neuropatia Ciática/etiologia , Neuropatia Ciática/fisiopatologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/fisiologia , Transdução Genética/métodos , Produtos do Gene tat do Vírus da Imunodeficiência Humana/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/uso terapêutico
18.
J Neurochem ; 110(5): 1557-66, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19573019

RESUMO

Ciliary neurotrophic factor (CNTF) regulates the differentiation and survival of a wide spectrum of developing and adult neurons, including motor neuron loss after injury. We recently described a cell-penetrant recombinant human CNTF (rhCNTF) molecule, formed by fusion with the human immunodeficiency virus-1 transactivator of transcription (TAT) protein transduction domain (TAT-CNTF) that, upon subcutaneous administration, retains full neurotrophic activity without cytokine-like side-effects. Although the CNTF receptor is present in hypothalamic nuclei, which are involved in the control of energy, rhCNTF but not TAT-CNTF stimulates signal transducers and activators of transcription 3 phosphorylation in the rat hypothalamus after subcutaneous administration. This could be due limited TAT-CNTF distribution in the hypothalamus and/or altered intracellular signaling by the fusion protein. To explore these possibilities, we examined the effect of intracerebroventricular administration of TAT-CNTF in male adult rats. TAT-CNTF-induced weight loss, although the effect was smaller than that seen with either rhCNTF or leptin (which exerts CNTF-like effects via its receptor). In contrast to rhCNTF and leptin, TAT-CNTF neither induced morphological changes in adipose tissues nor increased uncoupling protein 1 expression in brown adipose tissue, a characteristic feature of rhCNTF and leptin. Acute intracerebroventricular administration of TAT-CNTF induced a less robust phosphorylation of signal transducers and activators of transcription 3 in the hypothalamus, compared with rhCNTF. The data show that fusion of a protein transduction domain may change rhCNTF CNS distribution, while further strengthening the utility of cell-penetrating peptide technology to neurotrophic factor biology beyond the neuroscience field.


Assuntos
Fator Neurotrófico Ciliar/administração & dosagem , Fator Neurotrófico Ciliar/antagonistas & inibidores , Produtos do Gene tat/metabolismo , Transdução Genética/métodos , Animais , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Produtos do Gene tat/administração & dosagem , Produtos do Gene tat/genética , Humanos , Hipotálamo/citologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Injeções Intraventriculares , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Estrutura Terciária de Proteína/genética , Ratos , Ratos Wistar , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
19.
Neurochem Res ; 34(12): 2243-50, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19557514

RESUMO

Alzheimer's disease is characterised by regional neuronal degeneration, synaptic loss, and the progressive deposition of the 4 kDa ß-amyloid peptide (Aß) in senile plaques and accumulation of tau protein as neurofibrillary tangles. Aß derives from the larger precursor molecule, amyloid precursor protein (APP) by proteolytic processing via ß- and γ-secretases. While APP expression is well documented in neurons and astrocytes, the case for oligodendrocytes is less clear. The latter cell type is reported to express different isoforms of APP, and we have confirmed this observation by immunocytochemistry in cultures of differentiated rat cortical oligodendrocytes. Moreover, by means of a sensitive electrochemiluminescent immunoassay employing Aß C-terminal specific antibodies, mature oligodendrocytes are shown to secrete the 40 and 42 amino acid Aß species (Aß40 and Aß42). Secretion of Aß peptides was reduced by incubating oligodendrocytes with α- and ß-secretase inhibitors, or a γ-secretase inhibitor. Disturbances of APP processing and/ or synthesis in oligodendrocytes may account for some myelin disorders observed in Alzheimer's disease and other senile dementias.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Oligodendroglia/metabolismo , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Fragmentos de Peptídeos/biossíntese , Ratos , Tiazinas/farmacologia
20.
Prog Brain Res ; 245: 119-144, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30961866

RESUMO

The mechanisms regulating differentiation of multipotent oligodendrocyte progenitor cells (OPCs) into mature oligodendrocytes (OLs) are critical to our understanding of myelination and remyelination. Following acute demyelination in the central nervous system, adult OPCs migrate to the injury site, differentiate into OLs and generate new myelin sheaths. A common feature of regenerative processes is the fact that remyelination efficiency declines with aging and, accounts for the observation that chronic demyelinating diseases like multiple sclerosis (MS) are characterized by an ineffective remyelination. Without doubt, impairment of OPC differentiation is an essential determinant of the aging effects in remyelination. However, spontaneous remyelination is limited in demyelinating diseases such as MS, owing in part to the failure of adult OPCs to differentiate into myelinating OLs. The inability to restore myelin after injury compromises axon integrity and renders them vulnerable to degeneration. Although the genes that regulate the proliferation and differentiation of OPCs during development have been intensively studied, relatively little is known about the molecular signals that regulate the function of adult OPCs after demyelination. Elucidating the mechanisms regulating OPC differentiation are key to identifying pharmacological targets for remyelination-enhancing therapy. This review will discuss OPC biology, myelination, and possible pharmacological targets for promoting the differentiation of OPCs as a strategy to enhance remyelination, including the potential for nanoscale delivery.


Assuntos
Diferenciação Celular , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/metabolismo , Células Precursoras de Oligodendrócitos , Remielinização , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Humanos , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Células Precursoras de Oligodendrócitos/fisiologia , Remielinização/efeitos dos fármacos , Remielinização/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA