Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunity ; 57(3): 528-540.e6, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38417442

RESUMO

RNA splicing is involved in cancer initiation and progression, but how it influences host antitumor immunity in the metabolically abnormal tumor microenvironment (TME) remains unclear. Here, we demonstrate that lactate modulates Foxp3-dependent RNA splicing to maintain the phenotypic and functional status of tumor-infiltrating regulatory T (Treg) cells via CTLA-4. RNA splicing in Treg cells was correlated with the Treg cell signatures in the TME. Ubiquitin-specific peptidase 39 (USP39), a component of the RNA splicing machinery, maintained RNA-splicing-mediated CTLA-4 expression to control Treg cell function. Mechanistically, lactate promoted USP39-mediated RNA splicing to facilitate CTLA-4 expression in a Foxp3-dependent manner. Moreover, the efficiency of CTLA-4 RNA splicing was increased in tumor-infiltrating Treg cells from patients with colorectal cancer. These findings highlight the immunological relevance of RNA splicing in Treg cells and provide important insights into the environmental mechanism governing CTLA-4 expression in Treg cells.


Assuntos
Neoplasias , Linfócitos T Reguladores , Humanos , Antígeno CTLA-4 , Fatores de Transcrição Forkhead/genética , Ácido Láctico/metabolismo , Linfócitos do Interstício Tumoral , Neoplasias/genética , Neoplasias/metabolismo , Microambiente Tumoral , Proteases Específicas de Ubiquitina/metabolismo
2.
Cell Rep ; 42(12): 113518, 2023 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-38041812

RESUMO

The dysfunction and clonal constriction of tumor-infiltrating CD8+ T cells are accompanied by alterations in cellular metabolism; however, how the cell-intrinsic metabolic pathway specifies intratumoral CD8+ T cell features remains elusive. Here, we show that cell-autonomous generation of nicotinamide adenine dinucleotide (NAD+) via the kynurenine pathway (KP) contributes to the maintenance of intratumoral CD8+ T cell metabolic and functional fitness. De novo NAD+ synthesis is involved in CD8+ T cell metabolism and antitumor function. KP-derived NAD+ promotes PTEN deacetylation, thereby facilitating PTEN degradation and preventing PTEN-dependent metabolic defects. Importantly, impaired cell-autonomous NAD+ synthesis limits CD8+ T cell responses in human colorectal cancer samples. Our results reveal that KP-derived NAD+ regulates the CD8+ T cell metabolic and functional state by restricting PTEN activity and suggest that modulation of de novo NAD+ synthesis could restore CD8+ T cell metabolic fitness and antitumor function.


Assuntos
Linfócitos T CD8-Positivos , NAD , Humanos , NAD/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Cinurenina/metabolismo , Redes e Vias Metabólicas
3.
Cell Metab ; 35(12): 2107-2118.e6, 2023 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-37863051

RESUMO

Fructose consumption is associated with tumor growth and metastasis in mice, yet its impact on antitumor immune responses remains unclear. Here, we show that dietary fructose modulates adipocyte metabolism to enhance antitumor CD8+ T cell immune responses and control tumor growth. Transcriptional profiling of tumor-infiltrating CD8+ T cells reveals that dietary fructose mediates attenuated transition of CD8+ T cells to terminal exhaustion, leading to a superior antitumor efficacy. High-fructose feeding initiates adipocyte-derived leptin production in an mTORC1-dependent manner, thereby triggering leptin-boosted antitumor CD8+ T cell responses. Importantly, high plasma leptin levels are correlated with elevated plasma fructose concentrations and improved antitumor CD8+ T cell responses in patients with lung cancer. Our study characterizes a critical role for dietary fructose in shaping adipocyte metabolism to prime antitumor CD8+ T cell responses and highlights that the fructose-leptin axis may be harnessed for cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Humanos , Camundongos , Animais , Leptina/metabolismo , Neoplasias/metabolismo , Imunoterapia , Ativação Linfocitária
4.
J Clin Invest ; 133(23)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-37788092

RESUMO

The functional integrity of Tregs is interwoven with cellular metabolism; however, the mechanisms governing Treg metabolic programs remain elusive. Here, we identified that the deubiquitinase USP47 inhibited c-Myc translation mediated by the RNA N6-methyladenosine (m6A) reader YTHDF1 to maintain Treg metabolic and functional homeostasis. USP47 positively correlated with the tumor-infiltrating Treg signature in samples from patients with colorectal cancer and gastric cancer. USP47 ablation compromised Treg homeostasis and function in vivo, resulting in the development of inflammatory disorders, and boosted antitumor immune responses. USP47 deficiency in Tregs triggered the accumulation of the c-Myc protein and in turn exacerbated hyperglycolysis. Mechanistically, USP47 prevented YTHDF1 ubiquitination to attenuate the association of YTHDF1 with translation initiation machinery, thereby decreasing m6A-based c-Myc translation efficiency. Our findings reveal that USP47 directs m6A-dependent metabolic programs to orchestrate Treg homeostasis and suggest novel approaches for selective immune modulation in cancer and autoimmune diseases by targeting of USP47.


Assuntos
Doenças Autoimunes , Neoplasias , Humanos , Homeostase , Linfócitos T Reguladores , Ubiquitinação
5.
J Clin Invest ; 133(7)2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36821379

RESUMO

Activation of STING signaling in DCs promotes antitumor immunity. Aerobic glycolysis is a metabolic hallmark of activated DCs, but how the glycolytic pathway intersects with STING signaling in tumor-infiltrating DCs remains elusive. Here, we show that glycolysis drives STING signaling to facilitate DC-mediated antitumor immune responses. Tumor-infiltrating DCs exhibited elevated glycolysis, and blockade of glycolysis by DC-specific Ldha/Ldhb double deletion resulted in defective antitumor immunity. Mechanistically, glycolysis augmented ATP production to boost STING activation and STING-dependent DC antitumor functions. Moreover, DC-intrinsic STING activation accelerated HIF-1α-mediated glycolysis and established a positive feedback loop. Importantly, glycolysis facilitated STING-dependent DC activity in tissue samples from patients with non-small cell lung cancer. Our results provide mechanistic insight into how the crosstalk of glycolytic metabolism and STING signaling enhances DC antitumor activity and can be harnessed to improve cancer therapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Transdução de Sinais , Glicólise , Células Dendríticas
6.
Br J Radiol ; 96(1143): 20220802, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36350061

RESUMO

OBJECTIVES: To investigate the association of pre-diabetes(i.e., the early stages of glucometabolic disturbance) and Type 2 diabetes mellitus (T2DM) with intracranial plaque characteristics in patients with acute ischemic stroke using three-dimensional high-resolution MR imaging. METHODS: One hundred and forty-three symptomatic patients with acute ischemic stroke attribute to intracranial atherosclerotic plaque were prospectively enrolled. All participants were further divided into three groups: normal glucose metabolism(non-diabetes) group(n = 41), pre-diabetes group(n = 45), and T2DM group(n = 57) according to glucometabolic status. Culprit plaque characteristics (such as plaque burden, normalized wall index and enhancement ratio), total plaque number, and global plaque enhancement score were analyzed and compared among the three glucometabolic groups. The association between pre-diabetes and T2DM with intracranial plaque characteristics was assessed by logistic regression and multivariate linear regression. RESULTS: Plaque number was higher in patients with pre-diabetes and T2DM compared with those with non-diabetes(3.71 ± 1.83 and 3.75 ± 1.71 vs 2.24 ± 1.46, p = 0.006). Multivariate logistic regression showed a significant association of multiple intracranial plaques with pre-diabetes(OR 3.524, 95% CI 1.082 ~ 11.479, p = 0.037), T2DM(OR 3.760, 95% CI 1.098 ~ 12.872, p = 0.035) and luminal stenotic rate. Both pre-diabetes and T2DM were significantly associated with culprit plaque enhancement ratio(ß = 0.527 and ß = 0.536; respectively; p < 0.001) and global plaque enhancement score(ß = 0.264 and ß = 0.373; respectively; p < 0.05). CONCLUSIONS: Patients with pre-diabetes and T2DM had similar intracranial atherosclerotic plaque vulnerability, as demonstrated by multiple plaques, increased culprit plaque enhancement ratio and global plaque enhancement score. ADVANCES IN KNOWLEDGE: Pre-diabetes might be a risk factor for intracranial plaque vulnerability. It is necessary to monitor a slight increase in blood glucose in non-diabetes patients with acute ischemic stroke.


Assuntos
Diabetes Mellitus Tipo 2 , Arteriosclerose Intracraniana , AVC Isquêmico , Placa Aterosclerótica , Estado Pré-Diabético , Acidente Vascular Cerebral , Humanos , Diabetes Mellitus Tipo 2/complicações , Estado Pré-Diabético/complicações , Placa Aterosclerótica/complicações , Placa Aterosclerótica/diagnóstico por imagem , Arteriosclerose Intracraniana/complicações , Arteriosclerose Intracraniana/diagnóstico por imagem , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/etiologia , Imageamento por Ressonância Magnética/métodos
7.
Front Pharmacol ; 13: 971541, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36313320

RESUMO

As the incidence of rare diseases increases each year, the total number of rare disease patients worldwide is nearly 400 million. Orphan medications are drugs used to treat rare diseases. Orphan drugs, however, are rare and patients often struggle to utilize them and expensive medications during treatment. Orphan drugs have been the focus of new drug research and development for both domestic and international pharmaceutical companies as a result of the substantial investment being made in the field of rare diseases. Clinical breakthroughs have been made in every field, from traditional antibodies and small molecule drugs to gene therapy, stem cell therapy and small nucleic acid drugs. We here review the therapeutic means of rare diseases and drug development of rare diseases to show the progress of treatment of rare diseases in order to provide a reference for clinical use and new drug development of rare diseases in China.

8.
Int Immunopharmacol ; 109: 108920, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35691275

RESUMO

Dexmedetomidine, a highly selective α2-adrenoceptor agonist, has been recently reported to alleviate systemic inflammatory response induced by lipopolysaccharide (LPS), in addition to its sedative, analgesic, bradycardic and hypotensive properties. This study aimed to illustrate the molecular mechanisms underlying dexmedetomidine-induced anti-inflammation. In the LPS-pretreated mice, subcutaneous injection of dexmedetomidine reduced the spleen weight as well as serum and spleen expression of proinflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and IL-1ß, and increased serum and spleen expression of IL-10, a known anti-inflammatory cytokine. In addition, dexmedetomidine-attenuated proinflammatory cytokine reduction was entirely inhibited by selective α7 nicotinic acetylcholine receptor (nAChR) antagonist methyllycaconitine but not α2-adrenoceptor antagonist yohimbine. Dexmedetomidine also increased macrophageal IL-10 expression in the presence and absence of LPS, which was also attenuated by methyllycaconitine but not yohimbine. Furthermore, the stimulatory effect of dexmedetomidine on the expression of IL-10 was also reduced by the α7 nAChR gene silencer siRNA/α7 nAChR. Lastly, pretreatment with the IL-10 neutralizing antibody reversed dexmedetomidine-supressed expression of proinflammatory cytokines. Our findings illustrate that dexmedetomidine-induced anti-inflammation is through macrophageal expression of IL-10 following activation of α7 nAchRs but not α2-adrenoceptors.


Assuntos
Dexmedetomidina , Lipopolissacarídeos , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Citocinas/metabolismo , Dexmedetomidina/farmacologia , Dexmedetomidina/uso terapêutico , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Receptores Adrenérgicos/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
9.
J Clin Invest ; 132(7)2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35143421

RESUMO

The functional integrity of CD8+ T cells is tightly coupled to metabolic reprogramming, but how oxidative stress directs CD8+ T cell metabolic fitness in the tumor microenvironment (TME) remains elusive. Here, we report that SUMO-specific protease 7 (SENP7) senses oxidative stress to maintain the CD8+ T cell metabolic state and antitumor functions. SENP7-deficient CD8+ T cells exhibited decreased glycolysis and oxidative phosphorylation, resulting in attenuated proliferation in vitro and dampened antitumor functions in vivo. Mechanistically, CD8+ T cell-derived ROS triggered cytosolic SENP7-mediated PTEN deSUMOylation, thereby promoting PTEN degradation and preventing PTEN-dependent metabolic defects. Importantly, lowering T cell-intrinsic ROS restricted SENP7 cytosolic translocation and repressed CD8+ T cell metabolic and functional activity in human colorectal cancer samples. Our findings reveal that SENP7, as an oxidative stress sensor, sustains CD8+ T cell metabolic fitness and effector functions and unveil an oxidative stress-sensing machinery in tumor-infiltrating CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Linfócitos T CD8-Positivos/metabolismo , Endopeptidases/metabolismo , Humanos , Neoplasias/metabolismo , Estresse Oxidativo , Microambiente Tumoral
10.
Int J Med Sci ; 19(1): 47-64, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34975298

RESUMO

Background: Clear cell renal cell carcinoma (ccRCC) is a cell metabolic disease with high metastasis rate and poor prognosis. Our previous studies demonstrate that glucose-6-phosphate dehydrogenase (G6PD), the first and rate-limiting enzyme of the pentose phosphate pathway, is highly expressed in ccRCC and predicts poor outcomes of ccRCC patients. The aims of this study were to confirm the oncogenic role of G6PD in ccRCC and unravels novel mechanisms involving Cyclin E1 and MMP9 in G6PD-mediated ccRCC progression. Methods: Real-time RT-PCR, Western blot and immunohistochemistry were used to determine the expression patterns of G6PD, Cyclin E1 and MMP9 in ccRCC. TCGA dataset mining was used to identify Cyclin E1 and MMP9 correlations with G6PD expression, relationships between clinicopathological characteristics of ccRCC and the genes of interest, as well as the prognosis of ccRCC patients. The role of G6PD in ccRCC progression and the regulatory effect of G6PD on Cyclin E1 and MMP9 expression were investigated by using a series of cytological function assays in vitro. To verify this mechanism in vivo, xenografted mice models were established. Results: G6PD, Cyclin E1 and MMP9 were overexpressed and positively correlated in ccRCC, and they were associated with poor prognosis of ccRCC patients. Moreover, G6PD changed cell cycle dynamics, facilitated cells proliferation, promoted migration in vitro, and enhanced ccRCC development in vivo, more likely through enhancing Cyclin E1 and MMP9 expression. Conclusion: These findings present G6PD, Cyclin E1 and MMP9, which contribute to ccRCC progression, as novel biomarkers and potential therapeutic targets for ccRCC treatment.


Assuntos
Carcinoma de Células Renais/genética , Ciclina E/genética , Regulação Neoplásica da Expressão Gênica , Glucosefosfato Desidrogenase/fisiologia , Neoplasias Renais/genética , Metaloproteinase 9 da Matriz/genética , Proteínas Oncogênicas/genética , Regulação para Cima , Animais , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Ciclina E/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Oncogênicas/metabolismo
11.
Front Pharmacol ; 12: 784329, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867417

RESUMO

Morroniside, a secoiridoid glycoside from Cornus officinalis, is a class of small molecule non-peptide glucagon-like peptide-1 receptor (GLP-1R) agonists and possess many important biomedical functions. Our previous studies reported that GLP-1R agonist exenatide promoted M2 polarization and the expression of cell-specific anti-inflammatory factor interleukin-10 in neuropathological pain model. In this study, we proved that morroniside not only induced M2 polarization and stimulated interleukin-10 expression specifically in cortical primary microglia by p38ß mitogen-activated protein kinases pathway but also protected nerve cells against H2O2-induced cell oxidative damage and prohibited ischemic injury by reducing infarct size, which is at least in part mediated by enhanced expression of microglial interleukin-10. In the cortical penumbra area in middle cerebral artery occlusion (MCAO) mice. In general, our results indicated that GLP-1R agonist morroniside might play a neuroprotective effect by inducing M2 polarization, and cyclic-AMP/protein kinase A/p38ß pathway might mediate morroniside-induced expression of interleukin-10 protein in M2 microglia.

12.
Biochem Pharmacol ; 192: 114727, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34390739

RESUMO

Thalidomide is an antiinflammatory, antiangiogenic and immunomodulatory agent which has been used for the treatment of erythema nodosum leprosum and multiple myeloma. It has also been employed in treating complex regional pain syndromes. The current study aimed to reveal the molecular mechanisms underlying thalidomide-induced pain antihypersensitive effects in neuropathic pain. Thalidomide gavage, but not its more potent analogs lenalidomide and pomalidomide, inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain rats induced by tight ligation of spinal nerves, with ED50 values of 44.9 and 23.5 mg/kg, and Emax values of 74% and 84% MPE respectively. Intrathecal injection of thalidomide also inhibited mechanical allodynia and thermal hyperalgesia in neuropathic pain. Treatment with thalidomide, lenalidomide and pomalidomide reduced peripheral nerve injury-induced proinflammatory cytokines (TNFα, IL-1ß and IL-6) in the ipsilateral spinal cords of neuropathic rats and LPS-treated primary microglial cells. In contrast, treatment with thalidomide, but not lenalidomide or pomalidomide, stimulated spinal expressions of IL-10 and ß-endorphin in neuropathic rats. Particularly, thalidomide specifically stimulated IL-10 and ß-endorphin expressions in microglia but not astrocytes or neurons. Furthermore, pretreatment with the IL-10 antibody blocked upregulation of ß-endorphin in neuropathic rats and cultured microglial cells, whereas it did not restore thalidomide-induced downregulation of proinflammatory cytokine expression. Importantly, pretreatment with intrathecal injection of the microglial metabolic inhibitor minocycline, IL-10 antibody, ß-endorphin antiserum, and preferred or selective µ-opioid receptor antagonist naloxone or CTAP entirely blocked thalidomide gavage-induced mechanical antiallodynia. Our results demonstrate that thalidomide, but not lenalidomide or pomalidomide, alleviates neuropathic pain, which is mediated by upregulation of spinal microglial IL-10/ß-endorphin expression, rather than downregulation of TNFα expression.


Assuntos
Interleucina-10/biossíntese , Microglia/metabolismo , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Talidomida/uso terapêutico , beta-Endorfina/biossíntese , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Interleucina-10/agonistas , Masculino , Microglia/efeitos dos fármacos , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Talidomida/farmacologia , beta-Endorfina/agonistas
13.
Biochem Pharmacol ; 190: 114600, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33992630

RESUMO

Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, attenuates inflammation. The present study aimed to study the mechanisms underlying cynandione A-induced antiinflammation. Treatment with cynandione A and the specific α7 nicotinic acetylcholine receptor (α7 nAChR) agonist PHA-543613 remarkably reduced overexpression of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1ß in lipopolysaccharide (LPS)-treated RAW264.7 cells and primary peritoneal macrophages, and endotoxemic mice. Both cynandione A and PHA-543613 also stimulated IL-10 expression in naïve and LPS-treated macrophages and endotoxemic mice. Cynandione A- and PHA-543613-inhibited proinflammatory cytokine expression was completely blocked by the α7 nAChR antagonist methyllycaconitine and the IL-10 antibody. The stimulatory effect of cynandione A and PHA-543613 on IL-10 expression were suppressed by methyllycaconitine and knockdown of α7 nAChRs using siRNA/α7 nAChR. Cynandione A significantly stimulated STAT3 phosphorylation, which was attenuated by methyllycaconitine and the IL-10 neutralizing antibody. The STAT3 activation inhibitor NSC74859 also blocked cynandione A-inhibited proinflammatory cytokine expression. Taken together, our results, for the first time, demonstrate that cynandione A and PHA-543613 inhibit inflammation through macrophageal α7 nAChR activation and subsequent IL-10 expression.


Assuntos
Compostos de Bifenilo/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Interleucina-10/agonistas , Macrófagos/efeitos dos fármacos , Quinuclidinas/farmacologia , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Animais , Compostos de Bifenilo/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Células Cultivadas , Cynanchum , Relação Dose-Resposta a Droga , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-10/biossíntese , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinuclidinas/uso terapêutico , Células RAW 264.7 , Receptor Nicotínico de Acetilcolina alfa7/metabolismo
14.
Pharmacol Res Perspect ; 9(2): e00727, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33710781

RESUMO

D-Amino acid oxidase (DAAO) specifically catalyzes the oxidative deamination of neutral and polar D-amino acids and finally yields byproducts of hydrogen peroxide. Our previous work demonstrated that the spinal astroglial DAAO/hydrogen peroxide (H2 O2 ) pathway was involved in the process of pain and morphine antinociceptive tolerance. This study aimed to report mouse strain specificity of DAAO inhibitors on antinociception and explore its possible mechanism. DAAO inhibitors benzoic acid, CBIO, and SUN significantly inhibited formalin-induced tonic pain in Balb/c and Swiss mice, but had no antinociceptive effect in C57 mice. In contrast, morphine and gabapentin inhibited formalin-induced tonic pain by the same degrees among Swiss, Balb/c and C57 mice. Therefore, mouse strain difference in antinociceptive effects was DAAO inhibitors specific. In addition, intrathecal injection of D-serine greatly increased spinal H2 O2 levels by 80.0% and 56.9% in Swiss and Balb/c mice respectively, but reduced spinal H2 O2 levels by 29.0% in C57 mice. However, there was no remarkable difference in spinal DAAO activities among Swiss, Balb/c and C57 mice. The spinal expression of glutathione (GSH) and glutathione peroxidase (GPx) activity in C57 mice were significantly higher than Swiss and Balb/c mice. Furthermore, the specific GPx inhibitor D-penicillamine distinctly restored SUN antinociception in C57 mice. Our results reported that DAAO inhibitors produced antinociception in a strain-dependent manner in mice and the strain specificity might be associated with the difference in spinal GSH and GPx activity.


Assuntos
Analgésicos/administração & dosagem , Variação Biológica da População , D-Aminoácido Oxidase/antagonistas & inibidores , Nociceptividade/efeitos dos fármacos , Analgésicos/farmacocinética , Animais , D-Aminoácido Oxidase/metabolismo , Glutationa/análise , Glutationa/metabolismo , Glutationa Peroxidase/análise , Glutationa Peroxidase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
15.
Cancer Cell Int ; 20(1): 565, 2020 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-33292264

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

16.
Cancer Cell Int ; 20: 483, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33041664

RESUMO

BACKGROUND: Glucose 6-phosphate dehydrogenase (G6PD) serves key roles in cancer cell metabolic reprogramming, and has been reported to be involved in certain carcinogenesis. Previous results from our laboratory demonstrated that overexpressed G6PD was a potential prognostic biomarker in clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer. G6PD could stimulate ccRCC growth and invasion through facilitating reactive oxygen species (ROS)-phosphorylated signal transducer and activator of transcription 3 (pSTAT3) activation and ROS-MAPK-MMP2 axis pathway, respectively. However, the reasons for ectopic G6PD overexpression and the proliferation repressive effect of G6PD inhibition in ccRCC are still unclear. METHODS: The impact of ROS accumulation on NF-κB signaling pathway and G6PD expression was determined by real-time RT-PCR and Western blot in ccRCC cells following treatment with ROS stimulator or scavenger. The regulatory function of NF-κB signaling pathway in G6PD transcription was analyzed by real-time RT-PCR, Western blot, luciferase and ChIP assay in ccRCC cells following treatment with NF-κB signaling activator/inhibitor or lentivirus infection. ChIP and Co-IP assay was performed to demonstrate protein-DNA and protein-protein interaction of NF-κB and pSTAT3, respectively. MTS assay, human tissue detection and xenograft model were conducted to characterize the association between NF-κB, pSTAT3, G6PD expression level and proliferation functions. RESULTS: ROS-stimulated NF-κB and pSTAT3 signaling over-activation could activate each other, and exhibit cross-talks in G6PD aberrant transcriptional regulation. The underlying mechanism was that NF-κB signaling pathway facilitated G6PD transcription via direct DNA-protein interaction with p65 instead of p50. p65 and pSTAT3 formed a p65/pSTAT3 complex, occupied the pSTAT3-binding site on G6PD promoter, and contributed to ccRCC proliferation following facilitated G6PD overexpression. G6PD, pSTAT3, and p65 were highly expressed and positively correlated with each other in ccRCC tissues, confirming that NF-κB and pSTAT3 synergistically promote G6PD overexpression. Moreover, G6PD inhibitor exhibited tumor-suppressor activities in ccRCC and attenuated the growth of ccRCC cells both in vitro and in vivo. CONCLUSION: ROS-stimulated aberrations of NF-κB and pSTAT3 signaling pathway synergistically drive G6PD transcription through forming a p65/pSTAT3 complex. Moreover, G6PD activity inhibition may be a promising therapeutic strategy for ccRCC treatment.

17.
Int J Oncol ; 57(1): 197-212, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32319593

RESUMO

Glucose­6­phosphate dehydrogenase (G6PD) is crucial rate­limiting enzyme of the pentose phosphate pathway (PPP). G6PD dysregulation has been reported in various types of human cancer, and the role of G6PD in cancer progression was demonstrated in numerous studies. A previous study from our laboratory described the prognostic significance of G6PD in clear cell renal cell carcinoma (ccRCC), and demonstrated its proliferative role through positive feedback regulation of the phosphorylated form of signal transducer and activator of transcription 3. However, the role of G6PD in ccRCC invasion remains unclear. In the present study, reverse transcription­quantitative (RT­q) PCR, western blotting, enzyme activity assay, transwell assay and immunohistochemistry analysis in cell model, xenograft mice model and human specimen studies were performed to evaluate the role of G6PD in ccRCC invasion. The results from the present study demonstrated that G6PD may promote ccRCC cell invasive ability by increasing matrix metalloproteinase 2 (MMP2) mRNA and protein expression both in vitro and in vivo. In addition, a positive correlation between G6PD and MMP2 expression was demonstrated by RT­qPCR and western blotting in twenty pairs of ccRCC tumor specimens and matched adjacent normal tissues. Furthermore, G6PD promoted reactive oxygen species (ROS) generation and activated the MAPK signaling pathway in ccRCC cells. In addition, ROS significantly promoted the MAPK signaling pathway activation, which in turn contributed to MMP2 overexpression in ccRCC cells. In conclusion, the present study demonstrated that G6PD may facilitate ccRCC cell invasive ability by enhancing MMP2 expression through ROS­MAPK axis pathway.


Assuntos
Carcinoma de Células Renais/patologia , Glucosefosfato Desidrogenase/metabolismo , Neoplasias Renais/patologia , Metaloproteinase 2 da Matriz/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glucosefosfato Desidrogenase/genética , Humanos , Rim/patologia , Sistema de Sinalização das MAP Quinases/genética , Invasividade Neoplásica/patologia , Via de Pentose Fosfato/genética , Espécies Reativas de Oxigênio/metabolismo , Organismos Livres de Patógenos Específicos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Biochem Biophys Res Commun ; 525(4): 1087-1094, 2020 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-32184015

RESUMO

Lemairamin (also known as wgx-50), is isolated from the pericarps of the Zanthoxylum plants. As an agonist of α7 nicotinic acetylcholine receptors (α7nAChRs), it can reduce neuroinflammation in Alzheimer's disease. This study evaluated its antinociceptive effects in pain hypersensitivity and explored the underlying mechanisms. The data showed that subcutaneous lemairamin injection dose-dependently inhibited formalin-induced tonic pain but not acute nociception in mice and rats, while intrathecal lemairamin injection also dose-dependently produced mechanical antiallodynia in the ipsilateral hindpaws of neuropathic and bone cancer pain rats without affecting mechanical thresholds in the contralateral hindpaws. Multiple bi-daily lemairamin injections for 7 days did not induce mechanical antiallodynic tolerance in neuropathic rats. Moreover, the antinociceptive effects of lemairamin in formalin-induced tonic pain and mechanical antiallodynia in neuropathic pain were suppressed by the α7nAChR antagonist methyllycaconitine. In an α7nAChR antagonist-reversible manner, intrathecal lemairamin also stimulated spinal expression of IL-10 and ß-endorphin, while lemairamin treatment induced IL-10 and ß-endorphin expression in primary spinal microglial cells. In addition, intrathecal injection of a microglial activation inhibitor minocycline, anti-IL-10 antibody, anti-ß-endorphin antiserum or µ-opioid receptor-preferred antagonist naloxone was all able to block lemairamin-induced mechanical antiallodynia in neuropathic pain. These data demonstrated that lemairamin could produce antinociception in pain hypersensitivity through the spinal IL-10/ß-endorphin pathway following α7nAChR activation.


Assuntos
Acrilamidas/farmacologia , Analgésicos/farmacologia , Dor do Câncer/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Microglia/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Aconitina/análogos & derivados , Aconitina/farmacologia , Acrilamidas/administração & dosagem , Acrilamidas/uso terapêutico , Analgésicos/administração & dosagem , Analgésicos/uso terapêutico , Animais , Feminino , Formaldeído , Hiperalgesia/genética , Hiperalgesia/metabolismo , Injeções Espinhais , Interleucina-10/genética , Interleucina-10/metabolismo , Masculino , Camundongos , Microglia/metabolismo , Minociclina/administração & dosagem , Naloxona/administração & dosagem , Ratos , Ratos Wistar , Medula Espinal/metabolismo , Zanthoxylum/química , Zanthoxylum/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/genética , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , beta-Endorfina/genética , beta-Endorfina/metabolismo
19.
Front Pharmacol ; 11: 614450, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584292

RESUMO

Cynandione A, an acetophenone isolated from Cynanchum Wilfordii Radix, exhibits antineuropathic pain effect. This study further explored the target molecule and signaling mechanisms underlying cynandione-A-induced antineuropathic pain. Intrathecal injection of cynandione A significantly attenuated mechanical allodynia in neuropathic rats and substantially increased spinal expression of IL-10 and ß-endorphin but not dynorphin A. Cynandione A treatment also enhanced expression of IL-10 and ß-endorphin but not α7 nicotinic acetylcholine receptors (nAChRs) in cultured microglia. The IL-10 antibody attenuated cynandione-A-induced spinal or microglial gene expression of ß-endorphin and mechanical allodynia, whereas the ß-endorphin antiserum blocked cynandione-A-induced mechanical antiallodynia but not spinal or microglial IL-10 gene expression. The α7 nAChR antagonist methyllycaconitine significantly reduced cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and ß-endorphin. Furthermore, cynandione A stimulated microglial phosphorylation of PKA, p38, and CREB in an α7-nAChR-dependent manner, and treatment with their inhibitors attenuated cynandione-A-induced mechanical antiallodynia and spinal or microglial expression of IL-10 and ß-endorphin. In addition, cynandione A stimulated spinal phosphorylation of the transcription factor STAT3, which was inhibited by methyllycaconitine, the PKA activation inhibitor or IL-10 antibody. The STAT3 inhibitor NSC74859 also abolished cynandione-A-induced mechanical antiallodynia and spinal expression of ß-endorphin. These findings suggest that cynandione A suppresses neuropathic pain through α7-nAChR-dependent IL-10/ß-endorphin signaling pathway in spinal microglia.

20.
Br J Pharmacol ; 176(17): 3336-3349, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31309542

RESUMO

BACKGROUND AND PURPOSE: d-Amino acid oxidase (DAAO) is a flavine adenine dinucleotide-containing flavoenzyme and specifically catalyses oxidative deamination of d-amino acids. This study aimed to explore the association between increased cerebral DAAO expression or enzymic activity and the development of cerebral ischaemia. EXPERIMENTAL APPROACH: A mouse model of transient (90 min) middle cerebral artery occlusion (MCAO) was established, and western blotting, enzymic activity assay, and fluorescent immunostaining techniques were used. KEY RESULTS: The expression and enzymic activity of DAAO increased over time in the cortical peri-infarct area of the mice subjected to transient MCAO. The DAAO was specifically expressed in astrocytes, and its double immunostaining with the astrocytic intracellular marker, glial fibrillary acidic protein, in the cortical peri-infarct area was up-regulated following ischaemic insult, with peak increase on Day 5 after MCAO. Single intravenous injection of the specific and potent DAAO inhibitor Compound SUN reduced the cerebral DAAO enzymic activity and attenuated neuronal infarction and neurobehavioural deficits with optimal improvement apparent immediately after the MCAO procedure. The neuroprotective effect was dose dependent, with ED50 values of 3.9-4.5 mg·kg-1 . Intracerebroventricular injection of the DAAO gene silencer siRNA/DAAO significantly reduced cerebral DAAO expression and attenuated MCAO-induced neuronal infarction and behavioural deficits. CONCLUSIONS AND IMPLICATIONS: Our results, for the first time, demonstrated that increased cerebral astrocytic DAAO expression and enzymic activity were causally associated with the development of neuronal destruction following ischaemic insults, suggesting that targeting cerebral DAAO could be a potential approach for treatment of neurological conditions following cerebral ischaemia.


Assuntos
Isquemia Encefálica/metabolismo , D-Aminoácido Oxidase/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Animais , Isquemia Encefálica/induzido quimicamente , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/induzido quimicamente , Injeções Intraventriculares , Masculino , Camundongos , Pentobarbital/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...