Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
J Pain ; : 104504, 2024 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-38442838

RESUMO

The dorsal spinal cord is crucial for the transmission and modulation of multiple somatosensory modalities, such as itch, pain, and touch. Despite being essential for the well-being and survival of an individual, itch and pain, in their chronic forms, have increasingly been recognized as clinical problems. Although considerable progress has been made in our understanding of the neurochemical processing of nociceptive and chemical itch sensations, the neural substrate that is crucial for mechanical itch processing is still unclear. Here, using genetic and functional manipulation, we identified a population of spinal neurons expressing neuromedin U receptor 2 (Nmur2+) as critical elements for mechanical itch. We found that spinal Nmur2+ neurons are predominantly excitatory neurons, and are enriched in the superficial laminae of the dorsal horn. Pharmacogenetic activation of cervical spinal Nmur2+ neurons evoked scratching behavior. Conversely, the ablation of these neurons using a caspase-3-based method decreased von Frey filament-induced scratching behavior without affecting responses to other somatosensory modalities. Similarly, suppressing the excitability of cervical spinal Nmur2+ neurons via the overexpression of functional Kir2.1 potassium channels reduced scratching in response to innocuous mechanical stimuli, but not to pruritogen application. At the lumbar level, pharmacogenetic activation of these neurons evoked licking and lifting behaviors. However, ablating these neurons did not affect the behavior associated with acute pain. Thus, these results revealed the crucial role of spinal Nmur2+ neurons in mechanical itch. Our study provides important insights into the neural basis of mechanical itch, paving the way for developing novel therapies for chronic itch. PERSPECTIVE: Excitatory Nmur2+ neurons in the superficial dorsal spinal cord are essential for mechanical but not chemical itch information processing. These spinal Nmur2+ neurons represent a potential cellular target for future therapeutic interventions against chronic itch. Spinal and supraspinal Nmur2+ neurons may play different roles in pain signal processing.

3.
J Neurosci ; 44(1)2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37952941

RESUMO

Peripheral sensitization is one of the primary mechanisms underlying the pathogenesis of chronic pain. However, candidate molecules involved in peripheral sensitization remain incompletely understood. We have shown that store-operated calcium channels (SOCs) are expressed in the dorsal root ganglion (DRG) neurons. Whether SOCs contribute to peripheral sensitization associated with chronic inflammatory pain is elusive. Here we report that global or conditional deletion of Orai1 attenuates Complete Freund's adjuvant (CFA)-induced pain hypersensitivity in both male and female mice. To further establish the role of Orai1 in inflammatory pain, we performed calcium imaging and patch-clamp recordings in wild-type (WT) and Orai1 knockout (KO) DRG neurons. We found that SOC function was significantly enhanced in WT but not in Orai1 KO DRG neurons from CFA- and carrageenan-injected mice. Interestingly, the Orai1 protein level in L3/4 DRGs was not altered under inflammatory conditions. To understand how Orai1 is modulated under inflammatory pain conditions, prostaglandin E2 (PGE2) was used to sensitize DRG neurons. PGE2-induced increase in neuronal excitability and pain hypersensitivity was significantly reduced in Orai1 KO mice. PGE2-induced potentiation of SOC entry (SOCE) was observed in WT, but not in Orai1 KO DRG neurons. This effect was attenuated by a PGE2 receptor 1 (EP1) antagonist and mimicked by an EP1 agonist. Inhibition of Gq/11, PKC, or ERK abolished PGE2-induced SOCE increase, indicating PGE2-induced SOCE enhancement is mediated by EP1-mediated downstream cascade. These findings demonstrate that Orai1 plays an important role in peripheral sensitization. Our study also provides new insight into molecular mechanisms underlying PGE2-induced modulation of inflammatory pain.Significance Statement Store-operated calcium channel (SOC) Orai1 is expressed and functional in dorsal root ganglion (DRG) neurons. Whether Orai1 contributes to peripheral sensitization is unclear. The present study demonstrates that Orai1-mediated SOC function is enhanced in DRG neurons under inflammatory conditions. Global and conditional deletion of Orai1 attenuates complete Freund's adjuvant (CFA)-induced pain hypersensitivity. We also demonstrate that prostaglandin E2 (PGE2) potentiates SOC function in DRG neurons through EP1-mediated signaling pathway. Importantly, we have found that Orai1 deficiency diminishes PGE2-induced SOC function increase and reduces PGE2-induced increase in neuronal excitability and pain hypersensitivity. These findings suggest that Orai1 plays an important role in peripheral sensitization associated with inflammatory pain. Our study reveals a novel mechanism underlying PGE2/EP1-induced peripheral sensitization. Orai1 may serve as a potential target for pathological pain.


Assuntos
Cálcio , Dinoprostona , Animais , Feminino , Masculino , Camundongos , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Dinoprostona/farmacologia , Dinoprostona/metabolismo , Adjuvante de Freund/toxicidade , Adjuvante de Freund/metabolismo , Gânglios Espinais/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Dor
4.
Bioact Mater ; 14: 120-133, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35310342

RESUMO

Accurate drug delivery to the lesion has been deliberated for several decades, but one important phenomenon is usually neglected that the immune system can prevent smooth transportation of nanomedicine. Although injection would reduce first-pass effect, macrophages in the blood can still recognize and phagocytose nanomedicine. Here we show that a lubricated nanocontainer, which is prepared based on polyelectrolytes and mesoporous silica nanoparticles, can accurately target muscarinic bioreceptor while escaping from the identification of macrophages. Through in vitro and in vivo studies, this nanocontainer, combining both immune escape and bioreceptor targeting, has greatly improved the drug bioavailability. Additionally, this nanocontainer shows good biocompatibility, and the targeted heart tissues and other important metabolic organs, such as liver and kidney, keep physiological structures and functions without the detection of side effects. Furthermore, the mechanism of immune escape for the developed nanocontainer has been investigated by lubrication test and molecular simulation. We anticipate that our study will establish a new perspective on the achievement of immune escape-based targeted drug delivery, which can provide a fundamental approach for the design of related biomaterials.

5.
Pain ; 163(4): 652-664, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-34252911

RESUMO

ABSTRACT: Group I metabotropic glutamate receptors (group I mGluRs) have been implicated in several central nervous system diseases including chronic pain. It is known that activation of group I mGluRs results in the production of inositol triphosphate (IP3) and diacylglycerol that leads to activation of extracellular signal-regulated kinases (ERKs) and an increase in neuronal excitability, but how group I mGluRs mediate this process remains unclear. We previously reported that Orai1 is responsible for store-operated calcium entry and plays a key role in central sensitization. However, how Orai1 is activated under physiological conditions is unknown. Here, we tested the hypothesis that group I mGluRs recruit Orai1 as part of its downstream signaling pathway in dorsal horn neurons. We demonstrate that neurotransmitter glutamate induces STIM1 puncta formation, which is not mediated by N-Methyl-D-aspartate (NMDA) or α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Glutamate-induced Ca2+ entry in the presence of NMDA or AMPA receptor antagonists is eliminated in Orai1-deficient neurons. Dihydroxyphenylglycine (DHPG) (an agonist of group I mGluRs)-induced Ca2+ entry is abolished by Orai1 deficiency, but not affected by knocking down of transient receptor potential cation channel 1 (TRPC1) or TRPC3. Dihydroxyphenylglycine-induced activation of ERKs and modulation of neuronal excitability are abolished in cultured Orai1-deficient neurons. Moreover, DHPG-induced nociceptive behavior is markedly reduced in Orai1-deficient mice. Our findings reveal previously unknown functional coupling between Orai1 and group I mGluRs and shed light on the mechanism underlying group I mGluRs-mediated neuronal plasticity.


Assuntos
N-Metilaspartato , Receptores de Glutamato Metabotrópico , Animais , Cálcio/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Ácido Glutâmico/metabolismo , Camundongos , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacologia , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Células do Corno Posterior/metabolismo , Receptores de AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transdução de Sinais
6.
Elife ; 92020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32519950

RESUMO

Mu-opioid receptors (MORs) are crucial for analgesia by both exogenous and endogenous opioids. However, the distinct mechanisms underlying these two types of opioid analgesia remain largely unknown. Here, we demonstrate that analgesic effects of exogenous and endogenous opioids on inflammatory pain are mediated by MORs expressed in distinct subpopulations of neurons in mice. We found that the exogenous opioid-induced analgesia of inflammatory pain is mediated by MORs in Vglut2+ glutamatergic but not GABAergic neurons. In contrast, analgesia by endogenous opioids is mediated by MORs in GABAergic rather than Vglut2+ glutamatergic neurons. Furthermore, MORs expressed at the spinal level is mainly involved in the analgesic effect of morphine in acute pain, but not in endogenous opioid analgesia during chronic inflammatory pain. Thus, our study revealed distinct mechanisms underlying analgesia by exogenous and endogenous opioids, and laid the foundation for further dissecting the circuit mechanism underlying opioid analgesia.


Assuntos
Analgésicos Opioides/uso terapêutico , Inflamação/complicações , Neurônios/metabolismo , Dor/tratamento farmacológico , Dor/etiologia , Receptores Opioides mu/metabolismo , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/farmacologia , Receptores Opioides mu/genética , Tamoxifeno/farmacologia , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
7.
Int Immunopharmacol ; 75: 105813, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31404889

RESUMO

Our previous studies demonstrated that oral administration of madecassoside could markedly attenuate collagen-induced arthritis in rats, a rodent model of rheumatoid arthritis. As the autonomic nervous system is critically involved in the modulation of peripheral inflammation and immune response, the present study aims to explore the possible involvement of adrenergic and cholinergic nerves in the effect of madecassoside on rheumatoid arthritis. Arthritis was induced by chicken collagen in rats, and madecassoside was orally administered daily for two weeks from day 14 after the primary immunization. The antagonists of adrenoceptor and cholinergic receptors were co-administered with madecassoside, respectively. Unilateral cervical vagotomy was performed four days before the arthritis induction. The results showed that madecassoside (30 mg/kg) treatment markedly ameliorated arthritis symptoms in rats, mainly evidenced by the reduction of paw swelling and arthritis index scores. Co-administration of madecassoside with atropine (an antagonist of the muscarinic acetylcholine receptor) or hexamethonium (an antagonist of the nicotinic acetylcholine receptor) markedly diminished the therapeutic effects of madecassoside in arthritis. However, co-administration with phentolamine (an antagonist of the α-adrenoceptor) or propranolol (an antagonist of the ß-adrenoceptor) did not alter the effect of madecassoside on arthritis. Furthermore, unilateral cervical vagotomy significantly reduced the anti-arthritis efficacy of madecassoside, including the amelioration of clinical symptoms, as well as the inhibition of the production of pro-inflammatory cytokines except T lymphocytes-related cytokines. These findings suggest that madecassoside exerts inhibitory effects on collagen-induced arthritis through, at least partially, the peripheral cholinergic system.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Triterpenos/uso terapêutico , Nervo Vago , Administração Oral , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Citocinas/sangue , Citocinas/imunologia , Feminino , Articulação do Joelho/efeitos dos fármacos , Articulação do Joelho/patologia , Ratos Wistar , Vagotomia
8.
J Neurosci ; 39(33): 6472-6481, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31217328

RESUMO

Dopamine (DA) neurons in the VTA play essential roles in adaptive motivated behavior, which requires rapid discrimination between positive and negative motivational signature. However, the precise functional DA circuitry processing reward and aversive information remain elusive. Here, we report that the encoding of reward and aversion by the DA system in the NAc is tightly associated with its anatomical location. By recording the dynamics of DA release with genetically encoded fluorescent DA sensor using in vivo fiber photometry in freely moving male mice, we found that the DA-sensor signal in the dorsomedial NAc shell and dorsolateral NAc shell were increased during rewarding events and decreased during aversive noxious events. In contrast, the release of DA in the ventromedial NAc shell was increased by both rewarding and aversive stimuli, whereas the DA-sensor signal in the central ventromedial NAc shell and ventrolateral NAc shell showed complex dynamics. Furthermore, the activity of DA fibers in different subregions of NAc measured with calcium sensor largely recapitulated the changes of DA-sensor signal in response to rewarding and aversive stimuli. In addition, correlation analysis showed that the response magnitude of DA-sensor or fibers significantly changed along the DV axis of the NAc. These results revealed the distinct role of the mesolimbic DA system in different subregions of NAc in encoding value and salience.SIGNIFICANCE STATEMENT Adaptive motivated behavior requires rapid discrimination between favorable and harmful events and is dynamically modulated by dopamine (DA) neurons in the VTA. However, the precise relationship between distinct DA circuitry and reward/aversion signal encoding is not well understood. Here, by recording the dynamics of DA release and the activity of DA fibers in each subregion of the NAc using in vivo fiber photometry in freely moving animals, we found that the DA system in the dorsomedial/dorsolateral, ventromedial, and ventrolateral NAc shell plays different roles in encoding value and salience. These results extend our knowledge about how the mesolimbic DA system process motivational information at the circuitry level.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Motivação/fisiologia , Vias Neurais/fisiologia , Núcleo Accumbens/fisiologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Recompensa
9.
Front Pharmacol ; 9: 675, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29997506

RESUMO

Sinomenine has long been used for the treatment of rheumatoid arthritis in China. However, its anti-inflammatory mechanism is still debatable because the in vitro minimal effective concentration (≥250 µM) is hardly reached in either synovium or serum after oral administration at a therapeutic dose. Recent findings suggest that the α7 nicotinic acetylcholine receptor (α7nAChR) might mediate the inhibitory effect of sinomenine on macrophage activation, which attracts us to explore the anti-arthritis mechanism of sinomenine by taking neuroendocrine-inflammation axis into consideration. Here, we showed that orally administered sinomenine ameliorated the systemic inflammation of collagen-induced arthritis (CIA) rats, which was significantly diminished by either vagotomy or the antagonists of nicotinic acetylcholine receptors (especially the antagonist of α7nAChR), but not by the antagonists of muscarinic receptor. Sinomenine might bind to α7nAChR through interacting with the residues Tyr184 and Tyr191 in the pocket. In addition, the generation of vasoactive intestinal polypeptide (VIP) from the gut of CIA rats and cultured neuron-like cells was selectively enhanced by sinomenine through the activation of α7nAChR-PI3K/Akt/mTOR pathway. The elevated levels of VIP in the serum and small intestine of rats were negatively correlated with the scores of joint destruction. The crucial role of VIP in the anti-arthritic effect of sinomenine was confirmed by using VIP hybrid, a non-specific antagonist of VIP receptor. Taken together, intestine-sourced VIP mediates the anti-arthritic effect of sinomenine, which is generated by the activation of α7nAChR.

10.
J Neuroinflammation ; 15(1): 6, 2018 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-29306322

RESUMO

BACKGROUND: Previous studies have demonstrated that oral administration of curcumin exhibited an anti-arthritic effect despite its poor bioavailability. The present study aimed to explore whether the gut-brain axis is involved in the therapeutic effect of curcumin. METHODS: The collagen-induced arthritis (CIA) rat model was induced by immunization with an emulsion of collagen II and complete Freund's adjuvant. Sympathetic and parasympathetic tones were measured by electrocardiographic recordings. Unilateral cervical vagotomy (VGX) was performed before the induction of CIA. The ChAT, AChE activities, and serum cytokine levels were determined by ELISA. The expression of the high-affinity choline transporter 1 (CHT1), ChAT, and vesicular acetylcholine transporter (VAChT) were determined by real-time PCR and immunohistochemical staining. The neuronal excitability of the vagus nerve was determined by whole-cell patch clamp recording. RESULTS: Oral administration of curcumin restored the imbalance between the sympathetic and parasympathetic tones in CIA rats and increased ChAT activity and expression of ChAT and VAChT in the gut, brain, and synovium. Additionally, VGX eliminated the effects of curcumin on arthritis and ACh biosynthesis and transport. Electrophysiological data showed that curcumin markedly increased neuronal excitability of the vagus nerve. Furthermore, selective α7 nAChR antagonists abolished the effects of curcumin on CIA. CONCLUSIONS: Our results demonstrate that curcumin attenuates CIA through the "gut-brain axis" by modulating the function of the cholinergic system. These findings provide a novel approach for mechanistic studies of anti-arthritic compounds with low oral absorption and bioavailability.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Encéfalo/metabolismo , Curcumina/uso terapêutico , Trato Gastrointestinal/metabolismo , Acetilcolina/antagonistas & inibidores , Acetilcolina/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Artrite Experimental/patologia , Encéfalo/efeitos dos fármacos , Células Cultivadas , Colina O-Acetiltransferase , Curcumina/farmacologia , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Antagonistas Nicotínicos/farmacologia , Gânglio Nodoso/efeitos dos fármacos , Gânglio Nodoso/metabolismo , Distribuição Aleatória , Ratos , Ratos Wistar , Vagotomia/tendências , Nervo Vago/cirurgia
11.
J Neurosci ; 38(4): 887-900, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29229703

RESUMO

Pathological pain is a common and debilitating condition that is often poorly managed. Central sensitization is an important mechanism underlying pathological pain. However, candidate molecules involved in central sensitization remain unclear. Store-operated calcium channels (SOCs) mediate important calcium signals in nonexcitable and excitable cells. SOCs have been implicated in a wide variety of human pathophysiological conditions, including immunodeficiency, occlusive vascular diseases, and cancer. However, the role of SOCs in CNS disorders has been relatively unexplored. Orai1, a key component of SOCs, is expressed in the human and rodent spinal cord dorsal horn, but its functional significance in dorsal horn neurons is poorly understood. Here we sought to explore a potential role of Orai1 in the modulation of neuronal excitability and A-type potassium channels involved in pain plasticity. Using both male and female Orai1 knock-out mice, we found that activation of Orai1 increased neuronal excitability and reduced A-type potassium channels via the protein kinase C-extracellular signal-regulated protein kinase (PKC-ERK) pathway in dorsal horn neurons. Orai1 deficiency significantly decreased acute pain induced by noxious stimuli, nearly eliminated the second phase of formalin-induced nociceptive response, markedly attenuated carrageenan-induced ipsilateral pain hypersensitivity and abolished carrageenan-induced contralateral mechanical allodynia. Consistently, carrageenan-induced increase in neuronal excitability was abolished in the dorsal horn from Orai1 mutant mice. These findings uncover a novel signaling pathway involved in the pain process and central sensitization. Our study also reveals a novel link among Orai1, ERK, A-type potassium channels, and neuronal excitability.SIGNIFICANCE STATEMENT Orai1 is a key component of store-operated calcium channels (SOCs) in many cell types. It has been implicated in such pathological conditions as immunodeficiency, autoimmunity, and cancer. However, the role of Orai1 in CNS disorders remains poorly understood. The functional significance of Orai1 in neurons is elusive. Here we demonstrate that activation of Orai1 modulates neuronal excitability and Kv4-containing A-type potassium channels via the protein kinase C-extracellular signal-regulated protein kinase (PKC-ERK) pathway. Genetic knock-out of Orai1 nearly eliminates the second phase of formalin-induced pain and markedly attenuates carrageenan-induced pain hypersensitivity and neuronal excitability. These findings reveal a novel link between Orai1 and neuronal excitability and advance our understanding of central sensitization.


Assuntos
Sensibilização do Sistema Nervoso Central/fisiologia , Proteína ORAI1/metabolismo , Células do Corno Posterior/metabolismo , Animais , Feminino , Hiperalgesia/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Knockout , Dor/metabolismo , Proteína Quinase C/metabolismo , Canais de Potássio Shal/metabolismo
12.
J Biochem Mol Toxicol ; 31(8)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28346732

RESUMO

Higher expression of growth differentiation factor-9 (GDF-9) in keloids compared with hypertrophic scars and normal skin tissues has been reported recently. The present study was performed to investigate the role of GDF-9 in keloid pathogenesis, and to elucidate its implication for asiaticoside in the keloid management. The data showed that GDF-9 could enhance the proliferation, migration, and invasion of keloid fibroblasts (KFs), while it only slightly elevated collagen expression, indicating that the effect of GDF-9 was opposite to that of TGF-ß1. The bioactivity difference between GDF-9 and TGF-ß1 could be explained by the different phosphorylated sites on the downstream Smad2/3. Moreover, asiaticoside could inhibit GDF-9-induced activation of MAPKs and Smad pathway in KFs. In conclusion, GDF-9 enhanced the invasive growth of KFs, which was achieved by phosphorylation of Smad 2/3 at the linker region through activation of MAPKs pathway. Asiaticoside hindered the invasive growth of KFs by inhibiting the GDF-9/MAPK/Smad pathway.


Assuntos
Proliferação de Células/efeitos dos fármacos , Fibroblastos/metabolismo , Fator 9 de Diferenciação de Crescimento/metabolismo , Queloide/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Triterpenos/farmacologia , Adolescente , Adulto , Movimento Celular/efeitos dos fármacos , Feminino , Fibroblastos/patologia , Humanos , Queloide/patologia , Fosforilação/efeitos dos fármacos , Fator de Crescimento Transformador beta1/metabolismo
13.
J Cell Mol Med ; 21(9): 2172-2183, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28332288

RESUMO

Tetrandrine, a bisbenzylisoquinoline alkaloid constituent of the root of Stephania tetrandra S. Moore, was previously shown to suppress the differentiation of T helper 17 (Th17) cells and consequently ameliorate the collagen-induced arthritis (CIA) in mice by activating the aryl hydrocarbon receptor (AhR), but its underlying mechanism is incompletely understood. Here, we investigated how tetrandrine suppressed Th17 cell differentiation through the AhR pathway. The naïve CD4+ T cells were stimulated with anti-CD3/CD28 for 72 hrs in the presence or absence of tetrandrine under the Th17-polarizing condition. Tetrandrine inhibited the phosphorylation of signal transducer and activator of transcription-3 (STAT3) and boosted the phosphorylation of STAT5, while it did not alter the expression levels of phospho-Janus kinase-1 (p-JAK1), p-JAK2, p-JAK3, and suppressor of cytokine signalling-3 (SOCS3). The tetrandrine-mediated inhibition of the Th17 cell differentiation could be diminished by the activator of STAT3 and the inhibitor of STAT5. Meanwhile, the effect of tetrandrine on the either STAT3 or STAT5 phosphorylation was almost completely reversed by the AhR antagonist CH223191 and the AhR knockdown. In CIA mice, tetrandrine decreased p-STAT3 levels and increased p-STAT5 levels, which could also be reversed by the AhR antagonist resveratrol administration. Furthermore, tetrandrine promoted the AhR binding to the STAT5, but not to the STAT3. The tetrandrine-induced inhibition of the STAT3 phosphorylation was diminished by the inhibitor of STAT5. Taken together, tetrandrine suppressed Th17 cell differentiation by reciprocally modulating the activities of STAT3 and STAT5 in an AhR-dependent manner.


Assuntos
Benzilisoquinolinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/agonistas , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Células Th17/citologia , Animais , Janus Quinases/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fosforilação/efeitos dos fármacos , Receptores de Hidrocarboneto Arílico/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Células Th17/efeitos dos fármacos
14.
Lab Invest ; 96(10): 1076-86, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27617398

RESUMO

Sinomenine (SIN), an anti-arthritis drug, has previously been proven to exert immunomodulatory activity in rats by inducing intestinal regulatory T-cells (Treg cells). Here, we assessed the effect of SIN on the generation and function of Treg cells in autoimmune arthritis, and the underlying mechanisms in view of aryl hydrocarbon receptor (AhR). The proportions of Treg cells and IL-17-producing T-cells (Th17 cells) differentiated from naive T-cells were analyzed by flow cytometric analysis. The AhR agonistic effect of SIN was tested by analyzing the activation of downstream signaling pathways and target genes. The dependence of intestinal Treg cell induction and arthritis alleviation by SIN on AhR activation was confirmed in a mouse collagen-induced arthritis (CIA) model. SIN promoted the differentiation and function of intestinal Treg cells in vitro. It induced the expression and activity of AhR target gene, promoted AhR/Hsp90 dissociation and AhR nuclear translocation, induced XRE reporter activity, and facilitated AhR/XRE binding in vitro, displaying the potential to be an agonist of AhR. In CIA mice, SIN induced the generation of intestinal Treg cells, and facilitated the immunosuppressive function of these Treg cells as shown by an adoptive transfer test. In addition, the induction of intestinal Treg cells and the anti-arthritic effect of SIN in CIA mice could be largely diminished by the AhR antagonist resveratrol. SIN attenuates arthritis by promoting the generation and function of Treg cells in an AhR-dependent manner.


Assuntos
Antirreumáticos/uso terapêutico , Artrite/tratamento farmacológico , Morfinanos/uso terapêutico , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Antirreumáticos/farmacologia , Artrite/imunologia , Artrite/metabolismo , Diferenciação Celular/efeitos dos fármacos , Citocromo P-450 CYP1A1/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Masculino , Camundongos Endogâmicos DBA , Morfinanos/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Linfócitos T Reguladores/metabolismo
15.
Acta Pharmacol Sin ; 37(6): 794-804, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27133302

RESUMO

AIM: Paeoniflorin has shown to attenuate bleomycin-induced pulmonary fibrosis (PF) in mice. Because the epithelial-mesenchymal transition (EMT) in type 2 lung endothelial cells contributes to excessive fibroblasts and myofibroblasts during multiple fibrosis of tissues, we investigated the effects of paeoniflorin on TGF-ß mediated pulmonary EMT in bleomycin-induced PF mice. METHODS: PF was induced in mice by intratracheal instillation of bleomycin (5 mg/kg). The mice were orally treated with paeoniflorin or prednisone for 21 d. After the mice were sacrificed, lung tissues were collected for analysis. An in vitro EMT model was established in alveolar epithelial cells (A549 cells) incubated with TGF-ß1 (2 ng/mL). EMT identification and the expression of related proteins were performed using immunohistochemistry, transwell assay, ELISA, Western blot and RT-qPCR. RESULTS: In PF mice, paeoniflorin (50, 100 mg·kg(-1)·d(-1)) or prednisone (6 mg·kg(-1)·d(-1)) significantly decreased the expression of FSP-1 and α-SMA, and increased the expression of E-cadherin in lung tissues. In A549 cells, TGF-ß1 stimulation induced EMT, as shown by the changes in cell morphology, the increased cell migration, and the increased vimentin and α-SMA expression as well as type I and type III collagen levels, and by the decreased E-cadherin expression. In contrast, effects of paeoniflorin on EMT disappeared when the A549 cells were pretreated with TGF-ß1 for 24 h. TGF-ß1 stimulation markedly increased the expression of Snail and activated Smad2/3, Akt, ERK, JNK and p38 MAPK in A549 cells. Co-incubation with paeoniflorin (1-30 µmol/L) dose-dependently attenuated TGF-ß1-induced expression of Snail and activation of Smad2/3, but slightly affected TGF-ß1-induced activation of Akt, ERK, JNK and p38 MAPK. Moreover, paeoniflorin markedly increased Smad7 level, and decreased ALK5 level in A549 cells. CONCLUSION: Paeoniflorin suppresses the early stages of TGF-ß mediated EMT in alveolar epithelial cells, likely by decreasing the expression of the transcription factors Snail via a Smad-dependent pathway involving the up-regulation of Smad7.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glucosídeos/uso terapêutico , Pulmão/efeitos dos fármacos , Monoterpenos/uso terapêutico , Fibrose Pulmonar/tratamento farmacológico , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Células A549 , Animais , Anti-Inflamatórios não Esteroides/química , Bleomicina , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Glucosídeos/química , Humanos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Monoterpenos/química , Paeonia/química , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos
16.
Int J Biochem Cell Biol ; 75: 63-73, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27032495

RESUMO

OBJECTIVE: Norisoboldine (NOR), an isoquinoline alkaloid with very poor oral bioavailability, was previously proven to have a unique anti-arthritis activity in rats by inducing intestinal Treg cells. Herein, we explored its underlying mechanism in view of aryl hydrocarbon receptor (AhR). METHODS: The differentiation of regulatory T cells (Treg cells) and IL-17-producing T cells (Th17 cells) from naïve T cells was analyzed in vitro. The key role of AhR was ascertained using siRNA transfection. AhR agonistic effect was verified based on the activation of downstream signaling pathway and target genes. The correlation between AhR activation and Treg cell induction as well as pathological changes of joints was confirmed in collagen-induced arthritis (CIA) mouse model. RESULTS: NOR promoted intestinal Treg cell differentiation and function in an AhR-dependent manner. It acted as an AhR agonist, as evidenced by induction of CYP1A1 expression and activity, promotion of AhR/Hsp90 dissociation and AhR nuclear translocation, induction of XRE reporter activity, and facilitation of AhR/XRE binding. In CIA mice, NOR exerted substantial anti-arthritic effect through enhancing AhR activation in intestinal tissues and inducing intestinal Treg cell generation, which could be largely abolished by resveratrol (a antagonist of AhR). An adoptive transfer of Treg cells from NOR-treated mice could successfully alleviate arthritis symptoms in CIA mice. CONCLUSION: Oral NOR induces the generation of intestinal Treg cells by the activation of AhR, and thereby exerts anti-arthritic effect.


Assuntos
Alcaloides/metabolismo , Alcaloides/farmacologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Intestinos/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Alcaloides/uso terapêutico , Animais , Artrite Reumatoide/metabolismo , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteínas de Choque Térmico HSP90/metabolismo , Intestinos/efeitos dos fármacos , Ligantes , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th17/citologia , Células Th17/efeitos dos fármacos
17.
Biochem Pharmacol ; 101: 87-99, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26640276

RESUMO

Tetrandrine is an alkaloid constituent of the root of Stephania tetrandra S. Moore. The long-term clinical uses of tetrandrine for treatments of rheumatalgia and arthralgia as well as the inhibition of rat adjuvant-induced arthritis imply that tetrandrine may have therapeutic potential in rheumatoid arthritis (RA). Here, we explored its anti-RA mechanism in collagen-induced arthritis (CIA) in relation to the balance between T helper (Th) 17 cells and regulatory T (Treg) cells. DBA/1 mice were immunized with chicken type II collagen and were orally administered tetrandrine for 14 consecutive days. Then, the mice were sacrificed, their joints were removed for histological analysis, and spleens and mesenteric lymph nodes (MLNs) were removed to examine the Th17 and Treg cells. Tetrandrine markedly alleviated the severity of arthritis, reduced the serum levels of pro-inflammatory cytokines, and restored the Th17/Treg balance, as demonstrated by the serum levels of their related cytokines (IL-17 and IL-10) and the proportion of each cell type. Tetrandrine inhibited Th17 cell differentiation and induced Treg cell differentiation in vitro . Notably, aryl hydrocarbon receptor (AhR) was proven to play a crucial role in tetrandrine-mediated T cell differentiation. The correlation between AhR activation, regulation of Th17/Treg and amelioration of arthritis by tetrandrine was verified in the CIA mice. Moreover, tetrandrine might be a ligand of AhR because it facilitated the expression of the AhR target gene cytochrome P450 1A1 (CYP1A1) and the activation of its downstream signaling pathways. Taken together, tetrandrine exerts its anti-arthritis efficacy by restoring Th17/Treg balance via AhR.


Assuntos
Antirreumáticos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Benzilisoquinolinas/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Antirreumáticos/farmacologia , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/agonistas , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzilisoquinolinas/farmacologia , Linhagem Celular , Citocromo P-450 CYP1A1/química , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Articulações/efeitos dos fármacos , Articulações/imunologia , Articulações/metabolismo , Articulações/patologia , Extremidade Inferior , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Linfopoese/efeitos dos fármacos , Masculino , Mesentério , Camundongos Endogâmicos DBA , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Organismos Livres de Patógenos Específicos , Baço/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Baço/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/patologia , Regulação para Cima/efeitos dos fármacos
18.
Int Immunopharmacol ; 29(2): 583-590, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26456500

RESUMO

Endotoxin can stimulate inflammatory cytokine release from monocytes/macrophages and result in septic shock. Glycyrrhetinic acid (GA), the main bioactive component of licorice, possesses substantial anti-inflammatory activity. Here, we explored effect of 11-deoxy-18α-glycyrrhetinic acid-30-ethyl ester (DGAEE), a newly synthesized derivative of GA, on septic shock. DGAEE and its main metabolite 11-deoxy-18α-glycyrrhetinic acid (DGA) significantly alleviated septic shock as evidenced by improvements of survival rates, lung histopathological changes and wet/dry ratio in lipopolysaccharide (LPS)/D-galactosamine-stimulated mice, and decreased blood pressure in LPS/D-galactosamine-stimulated rats. The two compounds decreased serum levels of NO, TNF-α, IL-6, IL-1ß, and increased the level of IL-10 more potently in mice. In LPS-stimulated RAW 264.7 cells, DGA but not DGAEE showed marked regulation of NO, TNF-α, IL-6 and IL-10 levels, suggesting that DGAEE display anti-shock effect by DGA rather than itself. Moreover, the neutralizing antibody against IL-10 markedly prohibited the inhibitory effect of DGA on the production of cytokines from RAW 264.7 cells, and AS101 (an inhibitor of IL-10 biosynthesis) almost completely reversed the anti-shock effect of DGA in mice. In addition, DGA did not affect activation of NF-κB-p65 and p38 MAPK as well as IκBα degradation, but moderately reduced activation of ERK and JNK, and markedly increased phosphorylation of GSK3ß in LPS-stimulated RAW 264.7 cells. LY294002 (an inhibitor of GSK3ß phosphorylation) and LiCl (an inhibitor of GSK3ß activity) diminished and potentiated increase of IL-10 levels by DGA, respectively. In conclusion, DGAEE alleviates septic shock through DGA in an IL-10-dependent manner, and the mechanism is related to inactivation of GSK3ß.


Assuntos
Ácido Glicirretínico/análogos & derivados , Ácido Glicirretínico/farmacologia , Interleucina-10/metabolismo , Choque Séptico/prevenção & controle , Animais , Anticorpos Neutralizantes , Linhagem Celular , Cromonas/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Ácido Glicirretínico/química , Inflamação/metabolismo , Inflamação/prevenção & controle , Interleucina-10/genética , Macrófagos/efeitos dos fármacos , Camundongos , Estrutura Molecular , Morfolinas/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos
19.
Zhong Yao Cai ; 38(2): 333-8, 2015 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-26415412

RESUMO

OBJECTIVE: To explore the changes of intestinal mucosal immunity in collagen-induced arthritis rats and the impact of madecassoside on these changes. METHODS: Collagen-induced arthritis was established in female Wistar rats. Treatment group was orally administrated madecassoside once daily for consecutive 21 days, while blank control and model groups were orally administered saline at the same volume. The concentrations of sIgA in small intestine content and IFN-γ in small intestinal tissue homogenate were determined by ELISA. The proportions of CD4+ T and CD8+ T in the epithelium and laminar propria of small intestine were detected by flow cytometry, and the ratios of CD4+/CD8+ were calculated. The relative expressions of CD80, CD86, IL-6, IL-12 and Foxp3 mRNA in the small intestine were determined by real-time PCR. RESULTS: Compared with blank control rats, the concentrations of sIgA in small intestine content and IFN-γ in small intestinal tissue homogenate from model rats were increased, the ratios of CD4+/CD8+ in the epithelium and laminar propria of small intestine were higher and the relative expressions of CD80, CD86, IL-6 and IL-12 mRNA in the small intestine were increased. Madecassoside treatment decreased the concentrations of sIgA in small intestine content and IFN-γ in small intestinal tissue, downregulated the ratios of CD4+/CD8+ in the epithelium and laminar propria and decreased the relative expressions of CD80, CD86, IL-6 and IL-12 mRNA, while upregulated the relative expression of Foxp3 mRNA in the small intestine. CONCLUSION: The intestinal mucosal immune response is enhanced in collagen-induced arthritis rats, the antigen presenting cells are activated abnormally and the immune tolerance is disturbed. Madecassoside treatment can downregulate the intestinal mucosal immune response and benefit for the induction and maintenance of intestinal immune tolerance.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Experimental/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Triterpenos/farmacologia , Administração Oral , Animais , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Relação CD4-CD8 , Feminino , Fatores de Transcrição Forkhead/metabolismo , Tolerância Imunológica , Imunoglobulina A Secretora/imunologia , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-6/imunologia , Mucosa Intestinal/imunologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/imunologia , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
20.
Biochem Pharmacol ; 96(4): 323-36, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26074264

RESUMO

Arctigenin, the main effective constituent of Arctium lappa L. fruit, has previously been proven to dramatically attenuate dextran sulfate sodium (DSS)-induced colitis in mice, a frequently used animal model of inflammatory bowel disease (IBD). As Th1 and Th17 cells play a crucial role in the pathogenesis of IBD, the present study addressed whether and how arctigenin exerted anti-colitis efficacy by interfering with the differentiation and activation of Th1/Th17 cells. In vitro, arctigenin was shown to markedly inhibit the differentiation of Th17 cells from naïve T cells, and moderately inhibit the differentiation of Th1 cells, which was accompanied by lowered phosphorylation of STAT3 and STAT4, respectively. In contrast, arctigenin was lack of marked effect on the differentiation of either Th2 or regulatory T cells. Furthermore, arctigenin was shown to suppress the mammalian target of rapamycin complex 1 (mTORC1) pathway in T cells as demonstrated by down-regulated phosphorylation of the downstream target genes p70S6K and RPS6, and it functioned independent of two well-known upstream kinases PI3K/AKT and ERK. Arctigenin was also able to inhibit the activity of mTORC1 by dissociating raptor from mTOR. Interestingly, the inhibitory effect of arctigenin on T cell differentiation disappeared under a status of mTORC1 overactivation via knockdown of tuberous sclerosis complex 2 (TSC2, a negative regulator of mTORC1) or pretreatment of leucine (an agonist of mTOR). In DSS-induced mice, the inhibition of Th1/Th17 responses and anti-colitis effect of arctigenin were abrogated by leucine treatment. In conclusion, arctigenin ameliorates colitis through down-regulating the differentiation of Th1 and Th17 cells via mTORC1 pathway.


Assuntos
Anti-Inflamatórios/uso terapêutico , Colite/tratamento farmacológico , Furanos/uso terapêutico , Lignanas/uso terapêutico , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Colite/induzido quimicamente , Colite/patologia , Sulfato de Dextrana , Humanos , Células Jurkat , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Endogâmicos C57BL , Transdução de Sinais , Células Th1/patologia , Células Th17/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA