Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(2): 783-795, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36610792

RESUMEN

The number of genetic variations in the SARS-CoV-2 genome has been increasing primarily due to continuous viral mutations. Here, we report that the human APOBEC3A (A3A) cytidine deaminase plays a critical role in the induction of C-to-U substitutions in the SARS-CoV-2 genome. Bioinformatic analysis of the chronological genetic changes in a sequence database indicated that the largest UC-to-UU mutation signature, consistent with APOBEC-recognized nucleotide motifs, was predominant in single-stranded RNA regions of the viral genome. In SARS-CoV-2-infected cells, exogenous expression of A3A but not expression of other APOBEC proteins induced UC-to-UU mutations in viral RNA (vRNA). Additionally, the mutated C bases were often located at the tips in bulge or loop regions in the vRNA secondary structure. Interestingly, A3A mRNA expression was drastically increased by interferons (IFNs) and tumour necrosis factor-α (TNF-α) in epithelial cells derived from the respiratory system, a site of efficient SARS-CoV-2 replication. Moreover, the UC-to-UU mutation rate was increased in SARS-CoV-2 produced from lung epithelial cells treated with IFN-ß and TNF-α, but not from CRISPR/Cas9-based A3A knockout cells. Collectively, these findings demonstrate that A3A is a primary host factor that drives mutations in the SARS-CoV-2 RNA genome via RNA editing.


Asunto(s)
Citidina Desaminasa , Mutación , SARS-CoV-2 , Humanos , COVID-19/metabolismo , COVID-19/virología , Citidina Desaminasa/metabolismo , Genoma Viral , ARN Viral/genética , SARS-CoV-2/genética , Factor de Necrosis Tumoral alfa/genética
2.
J Biol Chem ; 298(3): 101724, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35157849

RESUMEN

ORF8 is an accessory protein encoded by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Consensus regarding the biological functions of ORF8 is lacking, largely because the fundamental characteristics of this protein in cells have not been determined. To clarify these features, we herein established an ORF8 expression system in 293T cells. Using this system, approximately 41% of the ORF8 expressed in 293T cells were secreted extracellularly as a glycoprotein homodimer with inter/intramolecular disulfide bonds. Intracellular ORF8 was sensitive to the glycosidase Endo H, whereas the secreted portion was Endo-H-resistant, suggesting that secretion occurs via a conventional pathway. Additionally, immunoblotting analysis showed that the total amounts of the major histocompatibility complex class Ι (MHC-I), angiotensin-converting enzyme 2 (ACE2), and SARS-CoV-2 spike (CoV-2 S) proteins coexpressed in cells were not changed by the increased ORF8 expression, although FACS analysis revealed that the expression of the cell surface MHC-I protein, but not that of ACE2 and CoV-2 S proteins, was reduced by ORF8 expression. Finally, we demonstrate by RNA-seq analysis that ORF8 had no significant stimulatory effects in human primary monocyte-derived macrophages (MDMs). Taken together, our results provide fundamental evidence that the ORF8 glycoprotein acts as a secreted homodimer, and its functions are likely associated with the intracellular transport and/or extracellular signaling in SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Glicoproteínas , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Proteínas Virales , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Glicoproteínas/metabolismo , Humanos , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Proteínas Virales/metabolismo
3.
J Nat Prod ; 86(11): 2487-2495, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-37874155

RESUMEN

The emergence of drug-resistant viruses is a serious concern in current chemotherapy for human immunodeficiency virus type-1 (HIV-1) infectious diseases. Hence, antiviral drugs aiming at targets that are different from those of approved drugs are still required, and the RNase H activity of HIV-1 reverse transcriptase is a suitable target. In this study, a search of a series of natural compounds was performed to identify the RNase H inhibitors. Three compounds were found to block the RNase H enzymatic activity. A laccaic acid skeleton was observed in all three natural compounds. A hydroxy phenyl group is connected to an anthraquinone backbone in the skeleton. An acetamido-ethyl, amino-carboxy-ethyl, and amino-ethyl are bound to the phenyl in laccaic acids A, C, and E, respectively. Laccaic acid C showed a 50% inhibitory concentration at 8.1 µM. Laccaic acid C also showed inhibitory activity in a cell-based viral proliferation assay. Binding structures of these three laccaic acids were determined by X-ray crystallographic analysis using a recombinant protein composed of the HIV-1 RNase H domain. Two divalent metal ions were located at the catalytic center in which one carbonyl and two hydroxy groups on the anthraquinone backbone chelated two metal ions. Molecular dynamics simulations were performed to examine the stabilities of the binding structures. Laccaic acid C showed the strongest binding to the catalytic site. These findings will be helpful for the design of potent inhibitors with modification of laccaic acids to enhance the binding affinity.


Asunto(s)
Infecciones por VIH , Ribonucleasa H , Humanos , Ribonucleasa H/metabolismo , Iones , Antraquinonas/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Inhibidores de la Transcriptasa Inversa/química
4.
J Cell Physiol ; 236(10): 6777-6792, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33665818

RESUMEN

Decreased expression of brain-derived neurotrophic factor (BDNF) is involved in the pathology of depressive disorders. Astrocytes produce BDNF following antidepressant treatment or stimulation of adrenergic receptors. Connexin43 (Cx43) is mainly expressed in central nervous system astrocytes and its expression is downregulated in patients with major depression. How changes in Cx43 expression affect astrocyte function, including BDNF production, is poorly understood. The current study examined the effect of Cx43 knockdown on BDNF expression in cultured cortical astrocytes after stimulation of adrenergic receptors. The expression of Cx43 in rat primary cultured cortical astrocytes was downregulated with RNA interference. Levels of messenger RNAs (mRNAs) or proteins were measured by real-time PCR and western blotting, respectively. Knockdown of Cx43 potentiated noradrenaline (NA)-induced expression of BDNF mRNA in cultured astrocytes. NA treatment induced proBDNF protein expression in astrocytes transfected with small interfering RNA (siRNA) targeting Cx43, but not with control siRNA. This potentiation was mediated by the Src tyrosine kinase-extracellular signal-regulated kinase (ERK) pathway through stimulation of adrenergic α1 and ß receptors. Furthermore, the Gq/11 protein-Src-ERK pathway and the G-protein coupled receptor kinase 2-Src-ERK pathway were involved in α1 and ß adrenergic receptor-mediated potentiation of BDNF mRNA expression, respectively. The current studies demonstrate a novel mechanism of BDNF expression in cortical astrocytes mediated by Cx43, in which downregulation of Cx43 increases, through adrenergic receptors, the expression of BDNF. The current findings indicate a potentially novel mechanism of action of antidepressants, via regulation of astrocytic Cx43 expression and subsequent BDNF expression.


Asunto(s)
Astrocitos/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/efectos de los fármacos , Conexina 43/metabolismo , Norepinefrina/farmacología , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Conexina 43/genética , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Cultivo Primario de Células , Interferencia de ARN , Ratas Wistar , Receptores Adrenérgicos alfa 1/efectos de los fármacos , Receptores Adrenérgicos alfa 1/metabolismo , Receptores Adrenérgicos beta/efectos de los fármacos , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal , Familia-src Quinasas/metabolismo
5.
J Neurochem ; 158(4): 849-864, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33118159

RESUMEN

Lysophosphatidic acid (LPA), a brain membrane-derived lipid mediator, plays important roles including neural development, function, and behavior. In the present study, the effects of LPA on astrocyte-derived synaptogenesis factor thrombospondins (TSPs) production were examined by real-time PCR and western blotting, and the mechanism underlying this event was examined by pharmacological approaches in primary cultured rat cortical astrocytes. Treatment of astrocytes with LPA increased TSP-1 mRNA, and TSP-2 mRNA, but not TSP-4 mRNA expression. TSP-1 protein expression and release were also increased by LPA. LPA-induced TSP-1 production were inhibited by AM966 a LPA1 receptor antagonist, and Ki16425, LPA1/3 receptors antagonist, but not by H2L5146303, LPA2 receptor antagonist. Pertussis toxin, Gi/o inhibitor, but not YM-254890, Gq inhibitor, and NF499, Gs inhibitor, inhibited LPA-induced TSP-1 production, indicating that LPA increases TSP-1 production through Gi/o-coupled LPA1 and LPA3 receptors. LPA treatment increased phosphorylation of extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), and c-Jun N-terminal kinase (JNK). LPA-induced TSP-1 mRNA expression was inhibited by U0126, MAPK/ERK kinase (MEK) inhibitor, but not SB202190, p38 MAPK inhibitor, or SP600125, JNK inhibitor. However, LPA-induced TSP-1 protein expression was diminished with inhibition of all three MAPKs, indicating that these signaling molecules are involved in TSP-1 protein production. Treatment with antidepressants, which bind to astrocytic LPA1 receptors, increased TSP-1 mRNA and protein production. The current findings show that LPA/LPA1/3 receptors signaling increases TSP-1 production in astrocytes, which could be important in the pathogenesis of affective disorders and could potentially be a target for the treatment of affective disorders.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/metabolismo , Lisofosfolípidos/farmacología , Trombospondina 1/biosíntesis , Animales , Astrocitos/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Femenino , Proteínas Quinasas JNK Activadas por Mitógenos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/genética , Embarazo , Cultivo Primario de Células , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Wistar , Receptores del Ácido Lisofosfatídico/antagonistas & inhibidores , Trombospondinas/biosíntesis
6.
Neurochem Res ; 46(9): 2285-2296, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34081246

RESUMEN

It has been demonstrated that peripheral inflammation induces cognitive dysfunction. Several histone deacetylase (HDAC) inhibitors ameliorate cognitive dysfunction in animal models of not only peripheral inflammation but also Alzheimer's disease. However, it is not clear which HDAC expressed in the central nervous system or peripheral tissues is involved in the therapeutic effect of HDAC inhibition on cognitive dysfunction. Hence, the present study investigated the effect of peripheral HDAC inhibition on peripheral inflammation-induced cognitive dysfunction. Suberoylanilide hydroxamic acid (SAHA), a pan-HDAC inhibitor that is mainly distributed in peripheral tissues after intraperitoneal administration, was found to prevent peripheral inflammation-induced cognitive dysfunction. Moreover, pretreatment with SAHA dramatically increased mRNA expression of interleukin-10, an anti-inflammatory cytokine, in peripheral and central tissues and attenuated peripheral inflammation-induced microglial activation in the CA3 region of the hippocampus. Minocycline, a macrophage/microglia inhibitor, also ameliorated cognitive dysfunction. Furthermore, as a result of treatment with liposomal clodronate, depletion of peripheral macrophages partially ameliorated the peripheral inflammation-evoked cognitive dysfunction. Taken together, these findings demonstrate that inhibition of peripheral HDAC plays a critical role in preventing cognitive dysfunction induced by peripheral inflammation via the regulation of anti-inflammatory cytokine production and the inhibition of microglial functions in the hippocampus. Thus, these findings could provide support for inhibition of peripheral HDAC as a novel therapeutic strategy for inflammation-induced cognitive dysfunction.


Asunto(s)
Disfunción Cognitiva/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Microglía/efectos de los fármacos , Vorinostat/uso terapéutico , Animales , Disfunción Cognitiva/etiología , Disfunción Cognitiva/metabolismo , Citocinas/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Inflamación/inducido químicamente , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos , Masculino , Ratones , Microglía/metabolismo
7.
Biochem Biophys Res Commun ; 533(4): 971-975, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33008602

RESUMEN

Previous studies have reported that continuous infusion with substance P (SP) into rat dorsal striatum ameliorated both mechanical allodynia in both formalin-evoked transient inflammatory pain and neuropathic pain models. However, a role of striatal SP in persistent inflammatory pain has not been demonstrated. The current study examined the effect of continuous infusion of SP into the rat dorsal striatum by reverse microdialysis on persistent inflammatory pain induced by complete Freund's adjuvant (CFA). Intraplantar injection of CFA evoked both mechanical allodynia and paw edema 3 and 7 days post-injection. The continuous infusion of SP ameliorated the CFA-evoked mechanical allodynia, but not paw edema, 3 days after the CFA injection. This antinociceptive effect of SP was partially inhibited by co-infusion with the neurokinin-1 (NK1) receptor antagonist CP96345. Conversely, at 7 days both CFA-evoked mechanical allodynia and paw edema were not affected by SP treatment. To clarify why the effect of SP treatment on CFA-induced pain changed, we evaluated NK1 receptor protein levels at both time points. The NK1 receptor protein level was decreased at 7, but not 3, days post CFA injection. These data suggest that persistent inflammatory pain can downregulate the striatal NK1 receptor. The current study demonstrates that striatal SP-NK1 receptor pathway can exert antinociceptive effect only on the third days of inflammatory pain phase defined as an acute but not the 7 days defined as a subacute.


Asunto(s)
Inflamación/fisiopatología , Dolor/tratamiento farmacológico , Sustancia P/administración & dosificación , Enfermedad Aguda , Analgésicos/administración & dosificación , Analgésicos/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Edema/tratamiento farmacológico , Adyuvante de Freund/antagonistas & inhibidores , Adyuvante de Freund/toxicidad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Inflamación/etiología , Infusiones Parenterales , Masculino , Dolor/etiología , Dolor/fisiopatología , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de Neuroquinina-1/metabolismo , Sustancia P/metabolismo
8.
J Neurochem ; 150(6): 738-758, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31273787

RESUMEN

Intrathecal treatment with recombinant high-mobility group box-1 (rHMGB1) in naïve mice leads to a persistent and significantly decreased hind paw withdrawal threshold to mechanical stimuli, suggesting that spinal HMGB1 evokes abnormal pain processing. By contrast, repeated intrathecal treatment with anti-HMGB1 antibody significantly reverses hind paw mechano-hypersensitivity in mice with a partial sciatic nerve ligation (PSNL). By contrast, the cellular mechanism by which spinal HMGB1 induces neuropathic pain has yet to be fully elaborated. The current study tested the hypothesis that spinal HMGB1 could induce mechanical hypersensitivity through the activation of specific receptor in glial cells. Intrathecal pretreatment with toll-like receptor (TLR) 4 inhibitors, but not TLR5, receptor for advanced glycation end-products and C-X-C chemokine receptor type 4 inhibitors, prevented rHMGB1-evoked mechanical hypersensitivity. Activation of spinal astrocytes appears to be crucial for the mechanism of action of rHMGB1 in naïve mice, as intrathecal pretreatment with astrocytic inhibitors prevented the rHMGB1-induced mechanical hypersensitivity. Interleukin-1ß (IL-1ß) was up-regulated within activated astrocytes and block of TLR4 prevented the upregulation of IL-1ß. Interleukin-1ß appears to be secreted by activated astrocytes, as IL-1ß neutralizing antibody prevented rHMGB1-induced mechanical hypersensitivity. Furthermore, intrathecal pretreatment with either MK801 or gabapentin prevented the rHMGB1-induced mechanical hypersensitivity, suggesting roles for spinal glutamate and the N-methyl-d-aspartate receptor in the mediation of rHMGB1-induced mechanical hypersensitivity. Thus, the current findings suggest that spinal HMGB1 upregulates IL-1ß in spinal astrocytes through a TLR4-dependent pathway and increases glutamatergic nociceptive transduction. These spinal mechanisms could be key steps that maintain neuropathic pain.


Asunto(s)
Astrocitos/metabolismo , Proteína HMGB1/metabolismo , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Interleucina-1beta/metabolismo , Masculino , Ratones , Médula Espinal/metabolismo , Regulación hacia Arriba
9.
Brain Behav Immun ; 78: 116-130, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30682503

RESUMEN

The orphan nuclear receptors REV-ERBα and REV-ERBß (REV-ERBs) are crucial in the regulation of inflammatory-related gene transcription in astroglioma cells, but their role in nociceptive transduction has yet to be elaborated. Spinal dorsal horn astrocytes contribute to the maintenance of chronic pain. Treatment of cultured spinal astrocytes with specific REV-ERBs agonists SR9009 or GSK4112 significantly prevented lipopolysaccharide (LPS)-induced mRNA upregulation of pronociceptive molecules interleukin-1ß (IL-1ß) mRNA, interleukin-6 (IL-6) mRNA and matrix metalloprotease-9 (MMP-9) mRNA, but not CCL2 mRNA expression. Treatment with SR9009 also blocked tumor necrosis factor-induced IL-1ß mRNA, IL-6 mRNA and MMP-9 mRNA. In addition, treatment with SR9009 significantly blocked LPS-induced upregulation of IL-1ß protein, IL-6 protein and MMP-9 activity. The inhibitory effects of SR9009 on LPS-induced expression of pronociceptive molecules were blocked by knockdown of REV-ERBs expression with short interference RNA, confirming that SR9009 exerts its effect through REV-ERBs. Intrathecal LPS treatment in male mice induces hind paw mechanical hypersensitivity, and upregulation of IL-1ß mRNA, IL-6 mRNA and glial fibrillary acidic protein (GFAP) expression in spinal dorsal horn. Intrathecal pretreatment of SR9009 prevented the onset of LPS-induced mechanical hypersensitivity, cytokine expression and GFAP expression. Intrathecal injection of SR9009 also ameliorated mechanical hypersensitivity during the maintenance phase of complete Freund's adjuvant-induced inflammatory pain and partial sciatic nerve ligation-, paclitaxel-, and streptozotocin-induced neuropathy in mice. The current findings suggest that spinal astrocytic REV-ERBs could be critical in the regulation of nociceptive transduction through downregulation of pronociceptive molecule expression. Thus, spinal REV-ERBs could be an effective therapeutic target in the treatment of chronic pain.


Asunto(s)
Astrocitos/metabolismo , Nociceptores/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Animales , Citocinas/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicina/análogos & derivados , Glicina/farmacología , Hiperalgesia/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Neuralgia/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/fisiología , Cultivo Primario de Células , Pirrolidinas/farmacología , Ratas , Ratas Wistar , Receptores Citoplasmáticos y Nucleares/metabolismo , Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Tiofenos/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Biol Pharm Bull ; 42(6): 857-866, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31155584

RESUMEN

Chronic pain, including inflammatory, neuropathic pain, is a serious clinical issue. There are increasing numbers of patients with chronic pain due to the growing number of elderly and it is estimated that about 25% of the global population will develop chronic pain. Chronic pain patients are refractory to medications used to treat acute pain such as opioids and non-steroidal anti-inflammatory drugs. Furthermore, the complexity and diversity of chronic pain mechanisms hinder the development of new analgesics. Thus, a better understanding of the mechanism of chronic pain is needed, which would facilitate the development of novel analgesics based on novel mechanisms. With this goal, connexins (Cxs) could be targeted for the development of new analgesics. Connexins are proteins with 20 subtypes, and function as channels, gap junctions between cells, and hemichannels that sample the extracellular space and release molecules such as neurotransmitters. Furthermore, Cxs could have functions independent of channel activity. Recent studies have shown that Cxs could be crucial in the induction and maintenance of chronic pain, and modulation of the activity or the expression of Cxs ameliorates nociceptive hypersensitivity in multiple chronic pain models. This review will cite novel findings on the role of of Cxs in the nociceptive transduction pathway under the chronic pain state and antinociceptive effects of various molecules modulating activity or expression of Cxs. Also, the potential of Cx modulation as a therapeutic strategy for intractable chronic pain will be discussed.


Asunto(s)
Analgésicos/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Conexinas/metabolismo , Animales , Dolor Crónico/metabolismo , Humanos
11.
Glia ; 66(2): 428-444, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29105869

RESUMEN

Connexin43 (Cx43), involved in intercellular signaling, is expressed in spinal dorsal horn astrocytes and crucial in the maintenance of neuropathic pain. Downregulation of spinal astrocytic Cx43 in mice enhances glutamatergic neurotransmission by decreasing glutamate transporter GLT-1 expression, resulting in cutaneous hypersensitivity. Decreased expression of astrocytic Cx43 could lead to altered expression of other nociceptive molecules. Transfection of Cx43-targeting siRNA in cultured spinal astrocytes increased expression of the pronociceptive cytokine interleukin-6 (IL-6) and the prostaglandin synthesizing enzyme cyclooxygenase-2 (COX-2). Increased expression of IL-6 and COX-2 was due to decreased Cx43 expression rather than due to diminished Cx43 channel function. In mice, downregulation of spinal Cx43 expression by intrathecal treatment with Cx43-targeting siRNA increased IL-6 and COX-2 expression and induced hind paw mechanical hypersensitivity. Cx43 siRNA-induced mechanical hypersensitivity was attenuated by intrathecal treatment with anti-IL-6 neutralizing antibody and intraperitoneal treatment of selective COX-2 inhibitor celecoxib, demonstrating that these molecules play a role in nociceptive processing following Cx43 downregulation. Restoring spinal Cx43 by intrathecal injection of an adenovirus vector expressing Cx43 in mice with a partial sciatic nerve ligation reduced spinal IL-6 and COX-2 expression. Suppression of glycogen synthase kinase-3ß (GSK-3ß), a serine/threonine protein kinase, prevented upregulation of IL-6 and COX-2 expression induced by Cx43 downregulation in both cultured astrocytes and in mouse spinal dorsal horn. Inhibition of spinal GSK-3ß also ameliorated Cx43 siRNA-induced mechanical hypersensitivity. The current findings indicate that downregulation of spinal astrocytic Cx43 leads to changes in spinal expression of pronociceptive molecules underlying the maintenance of pain following nerve injury.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Ciclooxigenasa 2/biosíntesis , Hiperalgesia/metabolismo , Interleucina-6/biosíntesis , Médula Espinal/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Conexina 43/antagonistas & inhibidores , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Femenino , Hiperalgesia/inducido químicamente , Inyecciones Espinales , Interleucina-6/antagonistas & inhibidores , Masculino , Ratones , Embarazo , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Wistar , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
12.
Biol Pharm Bull ; 40(10): 1759-1766, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28966248

RESUMEN

A significant reduction of glial cell line-derived neurotrophic factor (GDNF) has been identified in the pathophysiology of neurodegenerative and neuropsychiatric disorders. Thus, clarification of the mechanism of GDNF production, and modulating brain GDNF levels could be a novel therapeutic approach. A previous study demonstrated that antidepressant amitriptyline-induced GDNF production was significantly inhibited by pertussis toxin (PTX), a Gi/o protein inhibitor in astrocytes, the main source of GDNF in the brain. However, it is not known whether direct activation of Gi/o protein might induce GDNF expression, and what mechanisms might be involved after Gi/o protein activation. The current study investigated Gi/o protein-initiated GDNF production in rat cortical astrocytes using activators that directly activate Gi/o protein, mastoparan and compound48/80. Treatment of astrocytes with either mastoparan or compound48/80 increased GDNF mRNA expression at 3 and 6 h, and GDNF protein release at 24 h. Treatment of astrocyte with either mastoparan or compound48/80 increased brain-derived neurotrophic factor (BDNF) mRNA expression as well as GDNF. Mastoparan and compound48/80-induced GDNF mRNA expression were significantly inhibited by not only PTX, but also fibroblast growth factor receptor (FGFR) inhibitors, and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor. In fact, both FGFR substrate2α (FRS2α) and ERK phosphorylation were increased by treatment with either mastoparan or compound48/80, and these were significantly blocked by PTX. Thus, direct, receptor-independent Gi/o protein activation increases GDNF production through FGFR/ERK signaling pathway. The current results indicate a critical role of Gi/o signaling in the regulation of GDNF expression in astrocytes.


Asunto(s)
Astrocitos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Astrocitos/efectos de los fármacos , Células Cultivadas , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Péptidos y Proteínas de Señalización Intercelular , Péptidos/farmacología , Ratas Wistar , Venenos de Avispas/farmacología , p-Metoxi-N-metilfenetilamina/farmacología
13.
Mol Cell Neurosci ; 72: 72-83, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26808220

RESUMEN

Disturbances of circadian rhythm and dysregulation of clock gene expression are involved in the induction of various neurological disorder states, including chronic pain. However, the relationship between the CNS circadian-clock gene system and nociception remains poorly defined. Significant circadian oscillations of Period (Per1, Per2), Bmal1 and Cryptochrome 1 (Cry1) mRNA expression have been observed in the lumbar spinal dorsal horn of naïve mice. The current study examined the expression of clock genes in the lumbar spinal dorsal horn of mice with neuropathic pain due to a partial sciatic nerve ligation (PSNL). Seven days after PSNL, the mice displayed a robust unilateral hind paw mechanical hypersensitivity. The normal circadian oscillations of Per1, Per2 and Cry1, but not Bmal1, mRNA expression were significantly suppressed in the ipsilateral lumbar spinal dorsal horn of PSNL mice 7days following surgery. The circadian expression of PER1 protein, in particular, was also significantly suppressed in the ipsilateral spinal dorsal horn of PSNL mice. Double-labeling immunohistochemistry revealed downregulation of PER1 in neurons and astrocytes, but not microglia. Knockdown of Per1 expression by intrathecal treatment with Per1 siRNA also induced mechanical hypersensitivity, phosphorylation of c-jun N-terminal kinase (JNK) and the upregulation of chemokine (C-C motif) ligand 2 (CCL2) production in the lumbar spinal dorsal horn. Per1 siRNA-induced mechanical hypersensitivity was attenuated with intrathecal treatment of either the JNK inhibitor SP600125 or the selective CCL2 receptor (CCR2) antagonist RS504393, indicating that these intracellular messengers are crucial in mediating the mechanical hypersensitivity following the downregulation of PER1 expression. These results suggest that the downregulation of the spinal dorsal horn clock genes such as Per1 expressed could be crucial in the induction of neuropathic pain following peripheral nerve injury. Modulating clock gene Per1 expression could be a novel therapeutic strategy in alleviating neuropathic pain.


Asunto(s)
Quimiocina CCL2/metabolismo , Regulación hacia Abajo , Hiperalgesia/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Circadianas Period/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Animales , Astrocitos/metabolismo , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/genética , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/genética , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos ICR , Neuronas/metabolismo , Proteínas Circadianas Period/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Asta Dorsal de la Médula Espinal/citología , Asta Dorsal de la Médula Espinal/fisiología
14.
J Biol Chem ; 290(22): 13678-91, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25869129

RESUMEN

Further elaborating the mechanism of antidepressants, beyond modulation of monoaminergic neurotransmission, this study sought to elucidate the mechanism of amitriptyline-induced production of glial cell line-derived neurotrophic factor (GDNF) in astroglial cells. Previous studies demonstrated that an amitriptyline-evoked matrix metalloproteinase (MMP)/FGF receptor (FGFR)/FGFR substrate 2α (FRS2α)/ERK cascade is crucial for GDNF production, but how amitriptyline triggers this cascade remains unknown. MMP is activated by intracellular mediators such as G proteins, and this study sought to clarify the involvement of G protein signaling in amitriptyline-evoked GDNF production in rat C6 astroglial cells (C6 cells), primary cultured rat astrocytes, and normal human astrocytes. Amitriptyline-evoked GDNF mRNA expression and release were inhibited by pertussis toxin (PTX), a Gα(i/o) inhibitor, but not by NF449, a Gα(s) inhibitor, or YM-254890, a Gαq inhibitor. The activation of the GDNF production cascade (FGFR/FRS2α/ERK) was also inhibited by PTX. Deletion of Gα(ο1) and Gα(i3) by RNAi demonstrated that these G proteins play important roles in amitriptyline signaling. G protein activation was directly analyzed by electrical impedance-based biosensors (CellKey(TM) assay), using a label-free (without use of fluorescent proteins/probes or radioisotopes) and real time approach. Amitriptyline increased impedance, indicating Gα(i/o) activation that was suppressed by PTX treatment. The impedance evoked by amitriptyline was not affected by inhibitors of the GDNF production cascade. Furthermore, FGF2 treatment did not elicit any effect on impedance, indicating that amitriptyline targets PTX-sensitive Gα(i/o) upstream of the MMP/FGFR/FRS2α/ERK cascade. These results suggest novel targeting for the development of antidepressants.


Asunto(s)
Amitriptilina/farmacología , Antidepresivos Tricíclicos/farmacología , Astrocitos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Toxina del Pertussis/química , Animales , Astrocitos/citología , Técnicas Biosensibles , Línea Celular , Trastorno Depresivo Mayor/tratamiento farmacológico , Impedancia Eléctrica , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células HEK293 , Humanos , Masculino , Fosforilación , ARN/metabolismo , Interferencia de ARN , Ratas , Receptores Opioides/metabolismo , Transducción de Señal
15.
J Neurochem ; 136(4): 837-850, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26578177

RESUMEN

High-mobility group box-1 (HMGB1) has been shown to be critical in the modulation of nociceptive transduction following a peripheral neuropathy. However, the precise role of peripherally expressed HMGB1 in neuropathic pain has yet to be fully elaborated. Following a partial sciatic nerve ligation (PSNL) in mice, a persistent ipsilateral up-regulation of HMGB1 was observed from 3 to 21 days after PSNL, in paralleled with a robust ipsilateral hind paw mechanical hypersensitivity. Increased HMGB1 was detected in both infiltrating macrophages and proliferating Schwann cells in the ipsilateral nerve 14 days following PSNL. Repeated perineural treatment with anti-HMGB1 antibody significantly ameliorated PSNL-induced mechanical hypersensitivity. Several pronociceptive molecules, including matrix metalloprotease-9 (MMP-9), tumor necrosis factor-α, interleukin-1ß (IL-1ß), and cyclooxygenase-2, were up-regulated in injured sciatic nerve 14 days following PSNL. Repeated perineural treatment with an anti-HMGB1 antibody significantly suppressed expression of MMP-9, but not other pronociceptive molecules. Perineural treatment with a selective MMP-9 inhibitor ameliorated PSNL-induced mechanical hypersensitivity. The current findings demonstrate that the maintenance of the neuropathic state following an injured nerve is dependent on the up-regulation of HMGB1 and MMP-9. Thus, blocking HMGB1 function in sciatic nerve could be a potent therapeutic strategy for the treatment of neuropathic pain. Increased peripheral high-mobility group box-1 (HMGB1) is involved in the modulation of nociceptive transduction following a peripheral neuropathy. Following nerve injury in mice, increased HMGB1 is detected in both infiltrating macrophages and proliferating Schwann cells in the ipsilateral nerve. Repeated perineural treatment with anti-HMGB1 antibody significantly ameliorates nerve injury-induced mechanical hypersensitivity, and suppresses expression of matrix metalloprotease-9 (MMP-9). The findings demonstrate that the maintenance of the neuropathic state following an injury nerve is dependent on the up-regulation of HMGB1 and MMP-9.

16.
Biochem Biophys Res Commun ; 469(2): 151-7, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26616049

RESUMEN

Under physiological conditions, astrocytes maintain homeostasis in the CNS. Following inflammation and injury to the CNS, however, activated astrocytes produce neurotoxic molecules such as cytokines and chemokines, amplifying the initial molecular-cellular events evoked by inflammation and injury. Nuclear receptors REV-ERBα and REV-ERBß (REV-ERBs) are crucial in the regulation of inflammation- and metabolism-related gene transcription. The current study sought to elucidate a role of REV-ERBs in rat C6 astroglial cells on the expression of inflammatory molecules following stimulation with the neuroinflammatory cytokine tumor necrosis factor (TNF). Stimulation of C6 cells with TNF (10 ng/ml) significantly increased the mRNA expression of CCL2, interleukin-6 (IL-6), inducible nitric oxide synthase (iNOS), and matrix metalloprotease (MMP)-9, but not fibroblast growth factor-2 (FGF-2), cyclooxygenase-2 (COX-2) and MMP-2. Treatment with either REV-ERB agonists GSK4112 or SR9009 significantly blocked TNF-induced upregulation of CCL2 mRNA and MMP-9 mRNA, but not IL-6 mRNA and iNOS mRNA expression. Furthermore, treatment with RGFP966, a selective histone deacetylase 3 (HDAC3) inhibitor, potently reversed the inhibitory effects of GSK4112 on TNF-induced expression of MMP-9 mRNA, but not CCL2 mRNA. Expression of Rev-erbs mRNA in C6 astroglial cells, primary cultured rat cortical and spinal astrocytes was confirmed by reverse transcription polymerase chain reaction. Together, the findings demonstrate an anti-inflammatory effect, downregulating of MMP-9 and CCL2 transcription, of astroglial REV-ERBs activation through HDAC3-dependent and HDAC3-independent mechanisms.


Asunto(s)
Línea Celular/inmunología , Citocinas/inmunología , Factores Inmunológicos/inmunología , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/inmunología , Receptores Citoplasmáticos y Nucleares/inmunología , Proteínas Represoras/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Ratas
17.
Biochem Biophys Res Commun ; 479(4): 907-912, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27678293

RESUMEN

Neurotrophic/growth factors derived from glial cells, especially astrocytes, have been implicated in mood disorders and the pharmacological effects of antidepressant drugs. Previous studies demonstrated that the release of glial cell line-derived neurotrophic factor (GDNF) induced by the tricyclic antidepressant amitriptyline was significantly inhibited by a broad-spectrum matrix metalloproteinase (MMP) inhibitor in rat C6 astroglial cells (C6 cells). However, it is unknown whether amitriptyline affects MMP enzymatic activity or expression, and the MMP subtype has yet to be identified. The current study measured the effect of antidepressants on MMP activity with gelatin zymography, an in vitro assay for enzymatic activity, in C6 cells and primary cultured rat astrocytes (primary astrocytes). Treatment with amitriptyline increased zymographic MMP-9 activity without changing MMP-9 mRNA expression in C6 cells. Several different classes of antidepressants significantly increased zymographic MMP-9 activity in C6 cells and primary astrocytes, whereas antipsychotic drugs without antidepressant pharmacological activity did not. The amitriptyline-induced expression of GDNF mRNA was completely blocked by selective inhibition of MMP-9 in C6 cells. Treatment of C6 cells and primary astrocytes with exogenous recombinant MMP-9 increased GDNF mRNA expression, similar to that observed with amitriptyline. Inhibiting MMP-3 blocked amitriptyline-induced zymographic MMP-9 activation in C6 cells and primary astrocytes, indicating that MMP-3 is necessary for MMP-9 activity. The current study suggests that MMP-9 activation is indispensable in the amitriptyline-induced expression of GDNF mRNA in astrocytes and further supports a role of astrocytic neurotrophic/growth factors in the pharmacological effect of antidepressants.


Asunto(s)
Antidepresivos/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Amitriptilina/farmacología , Animales , Línea Celular , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
18.
J Neurochem ; 135(1): 27-37, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26183033

RESUMEN

Recently, we demonstrated that several antidepressants including amitriptyline increased fibroblast growth factor 2 (FGF2) mRNA expression slowly over 24 h through de novo protein synthesis in rat primary cultured astrocytes. This study defined the signaling cascade that mediates amitriptyline-induced FGF2 production in rat primary cultured astrocytes. Amitriptyline treatment significantly increased early growth response 1 (EGR1), a transcription factor known to regulate FGF2 expression. Knockdown of EGR1 using siRNA blocked amitriptyline-evoked FGF2 mRNA expression. Treatment with several different classes of antidepressants leads to expression of EGR1 mRNA as well as FGF2 mRNA. These results confirm that EGR1 production is likely to be indispensable for the amitriptyline-evoked FGF2 mRNA expression. Signal transduction inhibitors were used to elaborate the cellular signaling cascade that leads to EGR1-mediated FGF2 expression following amitriptyline treatment. Amitriptyline-evoked EGR1-mediated FGF2 mRNA expression was blocked by mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK)1/2 inhibitor. Furthermore, extracellular signal-regulated kinase/EGR1-mediated FGF2 mRNA expression evoked by amitriptyline was blocked by inhibitors of the FGF receptor, epidermal growth factor receptor (EGFR), and matrix metalloproteinase. Taken together, these results demonstrate that amitriptyline increases FGF2 mRNA expression through a matrix metalloproteinase/receptor tyrosine kinases (RTK) (FGF receptor and EGFR)/extracellular signal-regulated kinase/EGR1 signaling pathway in rat primary cultured astrocytes. Recent studies suggest that fibroblast growth factor 2 (FGF2) is involved in the antidepressant effect in the model of depression. The production of ERK-dependent early growth response 1 (EGR1) is a crucial part of the amitriptyline-induced FGF2 expression signaling cascade in rat primary cultured astrocytes. The findings elaborate an astrocytic mechanism that could be used to develop antidepressants.


Asunto(s)
Amitriptilina/farmacología , Antidepresivos Tricíclicos/farmacología , Astrocitos/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , ARN Mensajero/metabolismo , Animales , Astrocitos/metabolismo , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratas , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
19.
Biochem Biophys Res Commun ; 464(4): 1202-1208, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26212436

RESUMEN

Astrocytic gap junctions formed by connexin 43 (Cx43) are crucial for intercellular communication between spinal cord astrocytes. Various neurological disorders are associated with dysfunctional Cx43-gap junctions. However, the mechanism modulating Cx43-gap junctions in spinal astrocytes under pathological conditions is not entirely clear. A previous study showed that treatment of spinal astrocytes in culture with pro-inflammatory cytokines tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ) decreased both Cx43 expression and gap junction intercellular communication (GJIC) via a c-jun N-terminal kinase (JNK)-dependent pathway. The current study further elaborates the intracellular mechanism that decreases Cx43 under an inflammatory condition. Cycloheximide chase analysis revealed that TNF-α (10 ng/ml) alone or in combination with IFN-γ (5 ng/ml) accelerated the degradation of Cx43 protein in cultured spinal astrocytes. The reduction of both Cx43 expression and GJIC induced by a mixture of TNF-α and IFN-γ were blocked by pretreatment with proteasome inhibitors MG132 (0.5 µM) and epoxomicin (25 nM), a mixture of TNF-α and IFN-γ significantly increased proteasome activity and Cx43 ubiquitination. In addition, TNF-α and IFN-γ-induced activation of ubiquitin-proteasome systems was prevented by SP600125, a JNK inhibitor. Together, these results indicate that a JNK-dependent ubiquitin-proteasome system is induced under an inflammatory condition that disrupts astrocytic gap junction expression and function, leading to astrocytic dysfunction and the maintenance of the neuroinflammatory state.


Asunto(s)
Astrocitos/inmunología , Conexina 43/inmunología , Citocinas/inmunología , Uniones Comunicantes/inmunología , Médula Espinal/inmunología , Complejos de Ubiquitina-Proteína Ligasa/inmunología , Animales , Animales Recién Nacidos , Astrocitos/citología , Células Cultivadas , Regulación hacia Abajo/inmunología , Factores Inmunológicos/inmunología , Ratas , Ratas Wistar , Médula Espinal/citología , Ubiquitinación
20.
J Neurosci Res ; 93(4): 584-91, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25400139

RESUMEN

Connexin36 (Cx36), a component of neuronal gap junctions, is crucial for interneuronal communication and regulation. Gap junction dysfunction underlies neurological disorders, including chronic pain. Following a peripheral nerve injury, Cx36 expression in the ipsilateral spinal dorsal horn was markedly decreased over time, which paralleled the time course of hind paw tactile allodynia. Intrathecal (i.t.) injection of Cx36 siRNA (1 and 5 pg) significantly reduced the expression of Cx36 protein in the lumbar spinal cord, peaking 3 days after the injection, which corresponded with the onset of hind paw tactile allodynia. It is possible that some of the tactile allodynia resulting from Cx36 downregulation could be mediated through excitatory neuromodulators, such as glutamate and substance P. The Cx36 knockdown-evoked tactile allodynia was significantly attenuated by i.t. treatment with the N-methyl-D-aspartate glutamate receptor antagonist MK-801 but not the substance P receptor antagonist CP96345. Immunohistochemistry showed that Cx36 was colocalized with glycine transporter-2, a marker for inhibitory glycinergic spinal interneurons, but not with glutamate decarboxylase 67, a marker for inhibitory GABAergic spinal interneurons. The results indicate that spinal inhibition through glycinergic interneurons is reduced, leading to increased glutamatergic neurotransmission, as a result of Cx36 downregulation. The current data suggest that gap junction dysfunction underlies neuropathic pain and further suggest a novel target for the development of analgesics.


Asunto(s)
Conexinas/metabolismo , Regulación hacia Abajo/fisiología , Hiperalgesia/etiología , Células del Asta Posterior/metabolismo , Neuropatía Ciática/complicaciones , Neuropatía Ciática/patología , Análisis de Varianza , Animales , Compuestos de Bifenilo/farmacología , Conexinas/genética , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Glutamato Descarboxilasa/metabolismo , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Interneuronas/efectos de los fármacos , Interneuronas/metabolismo , Masculino , Ratones , Fármacos Neuroprotectores/farmacología , Umbral del Dolor/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Proteína delta-6 de Union Comunicante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA