Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Sci Rep ; 14(1): 6769, 2024 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-38514720

RESUMO

Breast cancer is a serious threat to human health. The transforming growth factor-ß signaling pathway is an important pathway involved in the occurrence and development of cancer. The SMAD family genes are responsible for the TGF-ß signaling pathway. However, the mechanism by which genes of the SMAD family are involved in breast cancer is still unclear. Therefore, it is necessary to investigate the biological roles of the SMAD family genes in breast cancer. We downloaded the gene expression data, gene mutation data, and clinical pathological data of breast cancer patients from the UCSC Xena database. We used the Wilcox test to estimate the expression of genes of the SMAD family in cancers. And the biological functions of SMAD family genes using the DAVID website. The Pearson correlation method was used to explore the immune cell infiltration and drug response of SMAD family genes. We conducted in biological experiments vitro and vivo. In this study, we integrated the multi-omics data from TCGA breast cancer patients for analysis. The expression of genes of SMAD family was significantly dysregulated in patients with breast cancer. Except for SMAD6, the expression of other SMAD family genes was positively correlated. We also found that genes of the SMAD family were significantly enriched in the TGF-ß signaling pathway, Hippo signaling pathway, cell cycle, and cancer-related pathways. In addition, SMAD3, SMAD6, and SMAD7 were lowly expressed in stage II breast cancer, while SMAD4 and SMAD2 were lowly expressed in stage III cancer. Furthermore, the expression of genes of the SMAD family was significantly correlated with immune cell infiltration scores. Constructing a xenograft tumor mouse model, we found that SMAD3 knockdown significantly inhibited tumorigenesis. Finally, we analyzed the association between these genes and the IC50 value of drugs. Interestingly, patients with high expression of SMAD3 exhibited significant resistance to dasatinib and staurosporine, while high sensitivity to tamoxifen and auranofin. In addition, SMAD3 knockdown promoted the apoptosis of BT-549 cells and decreased cell activity, and BAY-1161909 and XK-469 increased drug efficacy. In conclusion, genes of the SMAD family play a crucial role in the development of breast cancer.


Assuntos
Neoplasias da Mama , Transativadores , Humanos , Animais , Camundongos , Feminino , Transativadores/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Transdução de Sinais , Proteína Smad4/genética , Proteína Smad4/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteína Smad3/genética , Proteína Smad3/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo
2.
Artigo em Inglês | MEDLINE | ID: mdl-37905205

RESUMO

Purpose: Programmed cell death ligand 1 (PDL1) has the predictive and prognostic value in a great deal of cancers. This study aims to explore the expression of PDL1 in stage III breast cancer (BC) and its correlation with clinical outcome. Methods: The protein expression of PDL1 in tumor tissues was determined by immunohistochemistry (IHC). The correlations between PDL1 and clinicopathological variables were performed by χ²-tests or Fisher's exact tests. The Cox proportional hazards model was used for univariate and multivariate analysis of the potential prognostic factors. Survival curves were estimated based on Kaplan-Meier analyses, and Log Rank test was used to contrast factors influencing the survival outcome. Results: On the basis of the semiquantitative scoring method for PDL1 expression, the patients were divided into low PDL1 expression group (109 cases) and high PDL1 expression group (107 cases). PDL1 expression was correlated with positive lymph nodes, positive axillary lymph nodes, postoperative radiotherapy, and CK5/6 expression (P < 0.05). The PDL1 expression in tumor tissues was discovered to be a potential prognostic risk factor with the disease-free survival (DFS) and overall survival (OS) for stage III BC. Moreover, patients with high PDL1 expression showed longer lifetime (DFS and OS) compared to those with low PDL1 expression in total patient population (P < 0.05). Moreover, the nomogram showed that the prediction line is in good agreement with the reference line for postoperative 1-, 3-, and 5-year lifetime. The DCA curve showed that the 3- and 5-year lifetime by nomogram had so much better divination of the clinical application than only by PDL1. Conclusion: PDL1 is a latent prognostic factor in stage III BC and is closely related to some clinicopathological features. PDL1 expression in tumor tissues is significantly associated with better lifetime rate in stage III BC.

3.
Immunopharmacol Immunotoxicol ; 45(6): 663-671, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37358084

RESUMO

OBJECTIVE: Breast cancer (BC) causes cancer-related death in women. Sufentanil is used for cancer pain and postoperative analgesia. This study aimed to explore the role of sufentanil in BC. METHODS: BC cells were treated with sufentanil, and cell viability was evaluated using the cell counting kit-8 (CCK-8) assay. Biological behaviors were analyzed using EDU assay, flow cytometry, transwell assay, western blotting, and ELISA. The levels of NF-κB pathway-related factors were examined using western blotting. A xenograft tumor model was established to assess the effects of sufentanil on tumor growth in vivo. RESULTS: Sufentanil at the concentration of 20, 40, 80, and 160 nM suppressed cell viability (IC50 = 39.84 in MDA-MB-231 cells, and IC50 = 47.46 in BT549 cells). Sufentanil inhibited the proliferation, invasion, epithelial-mesenchymal transition (EMT), and inflammation, but induced apoptosis of BC cells. Mechanically, sufentanil suppressed the activation of the NF-κB pathway. Rescue experiments showed that RANKL (NF-κB receptor agonist) abrogated the effects induced by sufentanil. Moreover, sufentanil inhibited tumor growth, inflammatory response, but promoted apoptosis via the NF-κB pathway in vivo. CONCLUSIONS: Sufentanil decelerated the progression of BC by regulating the NF-κB pathway, suggesting sufentanil may be used in BC therapy.


Sufentanil treatment inhibited BC cell proliferation, invasion, epithelial-mesenchymal transition (EMT), immune response, but induced apoptosis.Sufentanil suppressed the activation of the NF-κB pathway.RANKL (NF-κB receptor agonist) abrogated the effects induced by sufentanil.Sufentanil inhibited tumor growth, proliferation, immune response and promoted apoptosis via the NF-κB pathway.


Assuntos
Neoplasias da Mama , NF-kappa B , Humanos , Feminino , NF-kappa B/metabolismo , Neoplasias da Mama/patologia , Sufentanil/farmacologia , Transdução de Sinais , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Imunidade , Proliferação de Células , Movimento Celular , Apoptose
4.
Biotechnol Genet Eng Rev ; : 1-21, 2023 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-37066722

RESUMO

The score and prognostic value of necroptosis were analyzed in the TCGA and GSE120622 datasets. Necroptosis has the highest correlation with the immune microenvironment, and the high score in NSCLC correlates with poor prognosis. Differentially expressed genes between non-small cell lung cancer (NSCLC) and controls in both datasets were identified and subjected to construct co-expression networks, respectively. Black and blue modules were selected because of high correction with necroptosis. The intersected two module genes were mainly involved in immune and inflammatory response, cell cycle process and DNA replication. Nine marker genes of necroptosis were identified in these modules and considered as candidate genes. Based on candidate genes, we identified two clusters utilizing concordance clustering, additionally dividing NSCLC samples into high- and low-risk groups. There were significant differences in overall survival between two clusters and between high- and low-risk groups. Furthermore, PARP1 was found among the candidate genes to be the target gene of dexmedetomidine acting on necroptosis. Molecular experimental results found that PARP1 was highly expressed in the dexmedetomidine treated NSCLC compared with the NSCLC. Candidate genes associated with necroptosis may provide a powerful prognostic tool for precision oncology. Dexmedetomidine may target PARP1 to promote necroptosis and then affect NSCLC.

5.
Neuroscience ; 500: 12-25, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35803492

RESUMO

Hypoxic-ischemic brain damage (HIBD) usually induces chronic neurological disorder and even acute death, but effective neuroprotective strategy is still limited. Herein, we performed this study to clarify the mechanism of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) containing microRNA-93 (miR-93) in influencing this damage via regulation of the histone deacetylase 4 (HDAC4)/B-cell lymphoma-2 (Bcl-2) axis. Initially, differentially expressed Bcl-2 was identified in middle cerebral artery occlusion (MCAO), and the upstream regulatory miR-93 and its potential target HDAC4 were also predicted through bioinformatics analysis. HIBD was modeled in vitro by exposing hippocampal neurons to oxygen-glucose deprivation (OGD) and in vivo by MCAO in rats. EVs were isolated from the bone marrow MSCs of well-grown rats. Our experimental data validated that HDAC4 was highly expressed while miR-93 and Bcl-2 were poorly expressed in MCAO rats. Furthermore, HDAC4 overexpression, through inhibiting Bcl-2 via deacetylation, promoted the infarct volume and pathological changes in hippocampal tissues and neuron apoptosis, and impaired neurobehavioral ability of MCAO rats. Of note, miR-93 was found to target HDAC4. Importantly, MSC-derived EVs overexpressing miR-93 suppressed HDAC4 expression and subsequently impeded the apoptosis of OGD-exposed hippocampal neurons in vitro, and also ameliorated HIBD in vivo. Taken together, miR-93 delivered by MSC-derived EVs can ameliorate HIBD by suppressing hippocampal neuron apoptosis through targeting the HDAC4/Bcl-2 axis, a finding which may be of great significance in the treatment of HIBD.


Assuntos
Vesículas Extracelulares , Hipóxia-Isquemia Encefálica , Células-Tronco Mesenquimais , MicroRNAs , Animais , Apoptose , Vesículas Extracelulares/metabolismo , Glucose/metabolismo , Hipocampo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos
6.
Dis Markers ; 2022: 5113473, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845135

RESUMO

Background: Complications after colon surgery are a major obstacle to postoperative recovery. The purpose of this study was to investigate the effect of electroacupuncture (EA) at Zusanli (ST36) on gastrointestinal motility in rats after colonic anastomosis and the mechanism of transient receptor potential vanillin 1 (TRPV1) channel in regulating gastrointestinal motility, pain, and inflammation. Methods: The rats were randomly divided into six groups, including the control, model, EA, sham-EA, capsaicin, and capsaicin+EA groups, with preoperative capsaicin pretreatment and EA treatment at ST36 acupoint after surgery. Rats were treated using EA at ST36 or sham acupoints after surgery for 5 days. Capsaicin was intraperitoneally injected into rats 3 hours before surgery. Gastrointestinal motility was assessed by measuring the gastric residue, small intestinal propulsion in vivo, contractile tension, and frequency of isolated muscle strips in vitro. The mechanical withdrawal threshold (MWT) of abdominal incision skin and spontaneous nociceptive scores were observed and recorded in rats after colon anastomosis. The expressions of TRPV1, substance P (SP), neurokinin 1 (NK1) receptor, nuclear factor kappa-B (NF-κB), interleukin- (IL-) 6, L-1ß, and tumor necrosis factor- (TNF-) α were determined. Results: Compared with the model group, electroacupuncture at ST36 point could significantly reduce the residual rate of stomach in rats after operation and increase the propulsive force of the small intestine and the contraction tension of the isolated smooth muscle. Electroacupuncture also increased postoperative day 3 MWT values and decreased postoperative spontaneous nociception scores. In addition, electroacupuncture treatment downregulated the expressions of IL-6, IL-1ß, TNF-α, TRPV1, NF-κB, SP, and NK1 receptors in the colon tissue of rats after colonic anastomosis. Conclusions: Our study showed that electroacupuncture at ST36 acupoint could improve gastrointestinal motility in rats after colonic anastomosis and relieve intestinal inflammation and pain. The mechanism may be to inhibit the activation of NF-κB and SP/NK1 receptor signaling pathways by inhibiting TRPV1.


Assuntos
Eletroacupuntura , Animais , Capsaicina/farmacologia , Motilidade Gastrointestinal , Inflamação/terapia , NF-kappa B/metabolismo , Dor , Ratos , Fator de Necrose Tumoral alfa/metabolismo
7.
Radiol Case Rep ; 17(7): 2295-2298, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35570866

RESUMO

Hepatic metastasis from hepatoid adenocarcinoma of the stomach (HAS) is a rare malignant tumor with hepatocellular differentiation. For the hepatic tumor in middle-aged and elderly people, the image presence of hepatocellular carcinoma (HCC) and production of large amounts of alpha fetoprotein (AFP) and the presence of stomach tumor, that suggest the diagnosis of hepatic metastasis from HAS. Here, the authors report a case of hepatic metastasis from HAS. The characteristics of the disease were analyzed on the basis of clinical symptoms, MR imaging findings, laboratory examinations and pathological diagnosis results. The imaging features and differential diagnosis methods of the disease were summarized combined with literature review, aiming to improve the understanding and diagnostic ability of the disease.

8.
Cancer Cell Int ; 21(1): 401, 2021 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-34321010

RESUMO

BACKGROUNDS: As previously reported, midazolam anesthesia exerts tumor-suppressing effects in non-small cell lung cancer (NSCLC), but the regulating effects of this drug on cisplatin-resistance in NSCLC have not been studied. Thus, we designed this study to investigate this issue and preliminarily delineate the potential molecular mechanisms. METHODS: We performed MTT assay and trypan blue staining assay to measure cell proliferation and viability. Cell apoptosis was examined by FCM. qRT-PCR and immunoblotting were performed to determine the expression levels of genes. The targeting sites between genes were predicted by bioinformatics analysis and were validated by dual-luciferase reporter gene system assay. Mice tumor-bearing models were established and the tumorigenesis was evaluated by measuring tumor weight and volume. Immunohistochemistry (IHC) was used to examine the pro-proliferative Ki67 protein expressions in mice tumor tissues. RESULTS: The cisplatin-resistant NSCLC (CR-NSCLC) cells were treated with high-dose cisplatin (50 µg/ml) and low-dose midazolam (10 µg/ml), and the results showed that midazolam suppressed cell proliferation and viability, and promoted cell apoptosis in cisplatin-treated CR-NSCLC cells. In addition, midazolam enhanced cisplatin-sensitivity in CR-NSCLC cell via modulating the miR-194-5p/hook microtubule-tethering protein 3 (HOOK3) axis. Specifically, midazolam upregulated miR-194-5p, but downregulated HOOK3 in the CR-NSCLC cells, and further results validated that miR-194-5p bound to the 3' untranslated region (3'UTR) of HOOK3 mRNA for its inhibition. Also, midazolam downregulated HOOK3 in CR-NSCLC cells by upregulating miR-194-5p. Functional experiments validated that both miR-194-5p downregulation and HOOK3 upregulation abrogated the promoting effects of midazolam on cisplatin-sensitivity in CR-NSCLC cells. CONCLUSIONS: Taken together, this study found that midazolam anesthesia reduced cisplatin-resistance in CR-NSCLC cells by regulating the miR-194-5p/HOOK3 axis, implying that midazolam could be used as adjuvant drug for NSCLC treatment in clinical practices.

9.
Toxicol Appl Pharmacol ; 416: 115458, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33607128

RESUMO

Morphine is an opioid agonist and a nonselective mu, kappa and delta receptor agonist. It is a commonly used analgesic drug for the treatment of acute and chronic pain as well as cancer pain. Morphine is particularly important to address the problem of morphine tolerance. Tcf7l2, known as a risk gene for schizophrenia and autism, encodes a member of the LEF1/TCF transcription factor family. TCF7L2 is an important transcription factor that is upregulated in neuropathic pain models. However, the relationship between TCF7L2 and morphine tolerance has not been reported. In this study, we found that morphine tolerance led to the upregulation of TCF7L2 in the spinal cord, and also led to the upregulation of TCF7L2 expression in glial cells, which promoted inflammation related signal, and activated TLR4 / NF-κB/NLRP3 pathway. In addition, TCF7L2 regulated microglial cell activation induced by chronic morphine treatment. Mechanically, we found that TCF7L2 transcriptionally regulated TLR4 expression, and the depletion of TCF7L2 alleviated morphine tolerance induced by chronic morphine treatment, and further alleviated pain hypersensitivity induced by chronic morphine treatment. We therefore suggested that TCF7L2 regulates the activation of TLR4/ NF-κB/NLRP3 pathway in microglia, and is involved in the formation of morphine tolerance. Our results provide a new idea for the regulation mechanism of morphine tolerance.


Assuntos
Analgésicos Opioides/toxicidade , Tolerância a Medicamentos , Hiperalgesia/induzido quimicamente , Microglia/efeitos dos fármacos , Morfina/toxicidade , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Dor Nociceptiva/prevenção & controle , Receptor 4 Toll-Like/metabolismo , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Dor Nociceptiva/metabolismo , Dor Nociceptiva/fisiopatologia , Limiar da Dor/efeitos dos fármacos , Transdução de Sinais , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Receptor 4 Toll-Like/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Regulação para Cima
10.
Transl Androl Urol ; 10(11): 4219-4230, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34984187

RESUMO

BACKGROUND: Bladder cancer is one of the most common malignant tumors among humans and has a high mortality. Clinically, lidocaine is the most commonly used local anesthetic, which can inhibit the proliferation of bladder cancer cells; however, its downstream specific molecular mechanisms are unclear. METHODS: The SwissTarget and TargetNet databases were used to analyze the target of lidocaine. The online public cancer transcriptome database UALCAN was used to analyze the up-regulated genes in The Cancer Genome Atlas Urothelial Bladder Carcinoma (TCGA-BLCA) data collection, and the intersection of the 2 was used to obtain the core target. The only target, isoprenylcysteine carboxylmethyltransferase (ICMT), was obtained by combining the correlation between the target and the clinical information of bladder cancer and the Kaplan-Meier (K-M) survival curve. Then, UMUC3 and T24 cells were selected as research vectors in vitro. Cell proliferation, cell cycle, and apoptosis were detected by cell counting kit-8, colony formation, flow cytometry, and western blotting. RESULTS: Network pharmacology analysis showed that ICMT might be one of the targets of lidocaine, and the expression level of ICMT was closely related to the clinical phenotype of bladder cancer. Lidocaine treatment (4 and 8 mM) significantly inhibited the proliferation of UMUC3 and T24 cells, promoted apoptosis, and significantly inhibited the mass and volume of xenograft tumors. In vitro experiments showed that ICMT promoted the proliferation of UMUC3 and T24 cells. Lidocaine inhibited the expression of ICMT in UMUC3 and T24 cells in a concentration and time-dependent manner, and inhibited cell proliferation by down-regulating ICMT expression. CONCLUSIONS: Lidocaine exerts anti-tumor effect by down-regulating the expression of ICMT in bladder cancer.

11.
Life Sci ; 264: 118635, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33131746

RESUMO

AIMS: Cancer pain seriously affects the life quality of patients. Morphine is commonly used for cancer pain, but tolerance development limits its clinical administration. Central immune signaling is important in the development of cancer pain and morphine tolerance. Cannabinoid receptor 2 (CB2) inhibits cancer pain and morphine tolerance by regulating central immune signaling. In the present study, we investigated the mechanisms of central immune signaling involved in morphine tolerance inhibition by the CB2 agonist AM1241 in cancer pain treatment. MAIN METHODS: Rats were implanted with tumor cells and divided into 4 groups: Vehicle (PBS), 0.07 µg AM1241, 0.03 µg AM1241, and AM630 (10 µg) + AM1241 (0.07 µg). All groups received morphine (20 µg/day, i.t.) for 8 days. AM630 (CB2 antagonist) was intrathecally injected 30 min before AM1241, and AM1241 was intrathecally injected 30 min before morphine. The spinal cord (SC) and dorsal root ganglion (DRG) were collected to determine the expression of Toll-like receptor 4 (TLR4), the p38 mitogen-activated protein kinase (MAPK), microglial markers, interleukin (IL)-1ß, and tumor necrosis factor (TNF)-α. KEY FINDINGS: The expression of TLR4, p38 MAPK, microglial markers, IL-1ß, and TNF-α was significantly higher in AM1241-pretreated groups than in the vehicle group (P < 0.05). No difference in microglial markers, IL-1ß, and TNF-α expression was detected in the AM630 + AM1241 group compared with the vehicle group. SIGNIFICANCE: Our results suggest that in a cancer pain-morphine tolerance model, an i.t. non-analgesic dose of AM1241 induces microglial activation and IL-1ß TNF-α upregulation in SC and DRG via the CB2 receptor pathway.


Assuntos
Dor do Câncer/tratamento farmacológico , Tolerância a Medicamentos , Morfina/farmacologia , Limiar da Dor/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Animais , Dor do Câncer/metabolismo , Canabinoides/farmacologia , Cateterismo , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Hiperalgesia/fisiopatologia , Sistema Imunitário , Interleucina-1beta/metabolismo , Masculino , Ratos , Ratos Wistar , Transdução de Sinais , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Front Genet ; 11: 602035, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33362860

RESUMO

There are associations between DNA methylation and the expression of long non-coding RNA (lncRNA), also known as lncRNA expression quantitative trait methylations (lnc-eQTMs). Lnc-eQTMs may induce a wide range of carcinogenesis pathways. However, lnc-eQTMs have not been globally identified and studied, and their roles in lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC) are largely unknown. In the present study, we identified some differential methylation sites located in genes of long intergenic non-coding RNAs (lincRNAs) and other types of lncRNAs in LUAD and LUSC. An integrated pipeline was established to construct two global cancer-specific regulatory networks of lnc-eQTMs in LUAD and LUSC. The associations between eQTMs showed common and specific features between LUAD and LUSC. Some lnc-eQTMs were also related with survival in LUAD- and LUSC-specific regulatory networks. Lnc-eQTMs were associated with cancer-related functions, such as lung epithelium development and vasculogenesis by functional analysis. Drug repurposing analysis revealed that these lnc-eQTMs may mediate the effects of some anesthesia-related drugs in LUAD and LUSC. In summary, the present study elucidates the roles of lnc-eQTMs in LUAD and LUSC, which could improve our understanding of lung cancer pathogenesis and facilitate treatment.

13.
J Pain Res ; 13: 2617-2627, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33116804

RESUMO

BACKGROUND: Sustained morphine treatment for cancer pain has been limited due to analgesic tolerance. Opioid receptor internalization and desensitization mediated by downregulation of mu-opioid receptor (MOR) expression have been confirmed as one of the mechanisms of chronic morphine tolerance. In addition to the opiate system, the α2-adrenergic system is involved in the development of morphine tolerance. Several studies reported that co-administration of α2-adrenoceptor agonist dexmedetomidine inhibits morphine tolerance in normal or neuropathic pain animals. However, the effect of dexmedetomidine on morphine tolerance has not been studied in cancer pain. Therefore, we investigated the effect of intrathecal injection of dexmedetomidine on the development of morphine tolerance in cancer pain and on the expression of MOR in the spinal cord of morphine-tolerant cancer pain rats. METHODS: The model was established using a rat's right hind paw injection of Walker 256 cancer cells. Subcutaneous morphine (10mg/kg) was administrated twice daily for 7 days; meanwhile, the rats received intrathecal α2-adrenoceptor agonist dexmedetomidine (10µ/kg) or antagonist MK-467 (0.25mg/kg) in test groups. Rats receiving drug vehicle served as the control group. Antinociception was detected by von Frey filaments and hot-plate tests. The expression of MOR in the spinal cord was examined through real-time reverse transcription polymerase chain reaction and Western blotting. The data were analyzed via analysis of variance followed by Student t-test with Bonferroni correction. RESULTS: Seven-day chronic morphine administration elicited notable analgesic tolerance in the rats with cancer pain. Co-administration of α2-adrenoceptor agonist dexmedetomidine enhanced morphine analgesia and attenuated morphine tolerance, which could be blocked by α2-adrenoceptor antagonist MK-467. Furthermore, pre-treatment of dexmedetomidine significantly upregulated MOR protein expression without a notable change in MOR mRNA expression in the spinal cord. CONCLUSION: Our findings suggest that intrathecal injection of dexmedetomidine enhanced morphine analgesia and attenuated morphine tolerance in cancer pain, potentially by upregulating MOR expression in the spinal cord. The α2-adrenoceptor agonist may provide a more versatile analgesia option for morphine treatment for cancer pain.

14.
Anesth Analg ; 131(4): 1270-1280, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32925348

RESUMO

BACKGROUND: Propofol is a common sedative-hypnotic drug traditionally used for inducing and maintaining general anesthesia. Recent studies have drawn attention to the nonanesthetic effects of propofol, but the potential mechanism by which propofol suppresses non-small-cell lung cancer (NSCLC) progression has not been fully elucidated. METHODS: For the in vitro experiments, we used propofol (0, 2, 5, and 10 µg/mL) to treat A549 cells for 1, 4, and 12 hours and Cell Counting Kit-8 (CCK-8) to detect proliferation. Apoptosis was measured with flow cytometry. We also transfected A549 cells with an microribonucleic acid-21 (miR-21) mimic or negative control ribonucleic acid (RNA) duplex and phosphatase and tensin homolog, deleted on chromosome 10 (PTEN) small interfering ribonucleic acid (siRNA) or negative control. PTEN, phosphorylated protein kinase B (pAKT), and protein kinase B (AKT) expression were detected using Western blotting, whereas miR-21 expression was examined by real-time polymerase chain reaction (RT-PCR). In vivo, nude mice were given injections of A549 cells to grow xenograft tumors; 8 days later, the mice were intraperitoneally injected with propofol (35 mg/kg) or soybean oil. Tumors were then collected from mice and analyzed by immunohistochemistry and Western blotting. RESULTS: Propofol inhibited growth (1 hour, P = .001; 4 hours, P ≤ .0001; 12 hours, P = .0004) and miR-21 expression (P ≤ .0001) and induced apoptosis (1 hour, P = .0022; 4 hours, P = .0005; 12 hours, P ≤ .0001) in A549 cells in a time and concentration-dependent manner. MiR-21 mimic and PTEN siRNA transfection antagonized the suppressive effects of propofol on A549 cells by decreasing PTEN protein expression (mean differences [MD] [95% confidence interval {CI}], -0.51 [-0.86 to 0.16], P = .0058; MD [95% CI], 0.81 [0.07-1.55], P = .0349, respectively), resulting in an increase in pAKT levels (MD [95% CI] = -0.82 [-1.46 to -0.18], P = .0133) following propofol exposure. In vivo, propofol treatment reduced NSCLC tumor growth (MD [95% CI] = -109.47 [-167.03 to -51.91], P ≤ .0001) and promoted apoptosis (MD [95% CI] = 38.53 [11.69-65.36], P = .0093). CONCLUSIONS: Our study indicated that propofol inhibited A549 cell growth, accelerated apoptosis via the miR-21/PTEN/AKT pathway in vitro, suppressed NSCLC tumor cell growth, and promoted apoptosis in vivo. Our findings provide new implications for propofol in cancer therapy and indicate that propofol is extremely advantageous in surgical treatment.


Assuntos
Anestésicos Intravenosos/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Propofol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células A549 , Anestésicos Intravenosos/uso terapêutico , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/efeitos dos fármacos , PTEN Fosfo-Hidrolase/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Ann Transl Med ; 8(16): 1040, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32953840

RESUMO

[This corrects the article DOI: 10.21037/atm.2020.04.28.].

16.
Ann Transl Med ; 8(8): 531, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32411754

RESUMO

BACKGROUND: Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, has been reported to increase the malignancy of breast cancer cells in vitro and stimulate tumor growth in mice. Transmembrane protease serine 2 (TMPRSS2) demonstrates proteolytic activity, resulting in degradation of the extracellular matrix (ECM). This study investigated whether and how TMPRSS2 regulates migration of DEX-treated breast cancer cells. METHODS: Breast cancer cell lines MCF-7 and MDA-MB-231 were treated with DEX and scratch assay was performed. Expressions of TMPRSS2, α2-adrenergic receptor, phospho-STAT3Tyr705, Rab11, and ECM components were assessed using real-time polymerase chain reaction (real-time PCR), Western blotting, and immunofluorescence staining. ELISA and ultracentrifugation were used to quantify secreted exosomal proteins. Knockdown assay was used to inhibit the expression of TMPRSS2 and Rab11. RESULTS: DEX significantly increased the migration of MCF-7 and MDA-MB-231, which was accompanied by the upregulation and colocalization of TMPRSS2 and α2-adrenergic receptor. Nuclear phospho-STAT3Tyr705 was increased dramatically following DEX treatment, and TMPRSS2 upregulation was significantly suppressed by the STAT3 inhibitor WP1066. Meanwhile, TMPRSS2 knockdown decreased DEX-induced cellular migration. TMPRSS2 and Rab11 were significantly detected in the media and the isolated exosomes from DEX-treated cells, and their colocalization was also revealed. Rab11 knockdown prevented exosomal TMPRSS2 from increasing in DEX-treated cells. In normal cultured MDA-MB-231, migration was increased by Rab11-positive exosomes isolated from DEX-treated MCF-7. Moreover, transmission electron microscopy showed that Rab11-positive exosomes enriched more components than Rab11-negative exosomes. Additionally, a reduction in ECM components fibronectin, collagen IV, matrix metallopeptidase 16, and Tenascin C was detected after DEX treatment, but was prohibited when TMPRSS2 or Rab11 were knocked down. CONCLUSIONS: This study provides evidence that DEX upregulates TMPRSS2 expression via the activation of α2-adrenergic receptor/STAT3 signaling and promotes TMPRSS2 secretion in exosomes through Rab11, thus resulting in degradation of the ECM, which is responsible for DEX-induced migration of breast cancer cells.

17.
Br J Anaesth ; 123(6): 777-794, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31668347

RESUMO

BACKGROUND: Dexmedetomidine (DEX) is a highly selective alpha2 adrenoceptor agonist with broad pharmacological effects, including sedation, analgesia, anxiolysis, and sympathetic tone inhibition. Here we report a systematic review and meta-analysis of its effects on stress, inflammation, and immunity in surgical patients during the perioperative period. METHODS: We searched MEDLINE, METSTR, Embase, and Web of Science for clinical studies or trials to analyse the effects of DEX on perioperative stress, inflammation, and immune function. RESULTS: Sixty-seven studies (including randomised controlled trials and eight cohort studies) with 4842 patients were assessed, of which 2454 patients were in DEX groups and 2388 patients were in control (without DEX) groups. DEX infusion during the perioperative period inhibited release of epinephrine, norepinephrine, and cortisol; decreased blood glucose, interleukin (IL)-6, tumour necrosis factor-α, and C-reactive protein; and increased interleukin-10 in surgical patients. In addition, the numbers of natural killer cells, B cells, and CD4+ T cells, and the ratios of CD4+:CD8+ and Th1:Th2 were significantly increased; CD8+ T-cells were decreased in the DEX group when compared with the control group. CONCLUSIONS: DEX, an anaesthesia adjuvant, can attenuate perioperative stress and inflammation, and protect the immune function of surgical patients, all of which may contribute to decreased postoperative complications and improved clinical outcomes.


Assuntos
Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Dexmedetomidina/farmacologia , Inflamação/tratamento farmacológico , Complicações Intraoperatórias/tratamento farmacológico , Complicações Pós-Operatórias/tratamento farmacológico , Estresse Fisiológico/efeitos dos fármacos , Humanos , Imunidade , Período Pré-Operatório
18.
J Pain Res ; 12: 2231-2238, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31413621

RESUMO

BACKGROUND: Reports of postoperative pain treatment after uniportal video-assisted thoracoscopic surgery are limited. Thoracic paravertebral block and serratus anterior plane block have been described recently in pain management after thoracic surgery. A comparison between these two blocks for postoperative analgesia after uniportal video-assisted thoracoscopic surgery has not been previously reported. The aim of this study was to compare the analgesic benefits of serratus plane block and thoracic paravertebral block after uniportal video-assisted thoracoscopic surgery and examined the two block types for noninferiority. METHODS: From December 2015 to May 2018, a total of 636 relevant records of patients who underwent uniportal video-assisted thoracoscopic surgery under general anaesthesia alone or with the addition of serratus plane block or thoracic paravertebral block performed preoperatively were identified. A propensity-matched analysis incorporating preoperative variables was used to compare the efficacy of postoperative analgesia in three groups. RESULTS: Overall, 123 patients were identified for analysis. Propensity score matching resulted in 41 patients in each group. The visual analogue scale scores were significantly lower in the serratus plane block group and the thoracic paravertebral block group than in the control group at the 1st, 2nd, 4th, and 6th postoperative hours. Cumulative opioid consumption was significantly lower in the serratus plane and thoracic paravertebral block groups than in the control group at 6 hrs (18.3±3.1 mg, 18.7±3.9 mg vs 21.5±4.4 mg; P=0.001) and 24 hrs (43.4±7.3 mg, 42.5±7.7 mg vs 49.3±8.8 mg; P<0.001) postoperatively. The serratus plane block group was noninferior to the thoracic paravertebral block group on pain score and opioid consumption. CONCLUSION: The addition of single-injection serratus plane or thoracic paravertebral block is associated with early analgesic benefits in patients undergoing uniportal video-assisted thoracoscopic surgery, including a reduction in the postoperative opioid consumption and pain scores. Serratus plane block is as effective as thoracic paravertebral block for reducing postoperative pain.

19.
Ann Surg Oncol ; 26(3): 884-893, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30565043

RESUMO

BACKGROUND: The programmed cell death-1/programmed cell death-ligand 1 (PD-1/PD-L1) pathway has been shown to be involved in trauma-induced immunosuppression and to influence CD4+ T cell differentiation. MicroRNA (miR)-21 is a critical player in immune responses. However, it remains largely unknown whether miR-21 is regulated by PD-1 and influences CD4+ T-cell lineage choice after gastric cancer resection. METHODS: In the present study, we analyzed the percentages of T-helper (Th)-17/regulatory T (Treg) cells and PD-1/PD-L1 expression on peripheral blood mononuclear cells (PBMCs) during the perioperative period. We also detected the secretion of interleukin (IL)-17 and transforming growth factor (TGF)-ß1 using enzyme-linked immunosorbent assays (ELISAs). Furthermore, PBMCs isolated from patients were transfected with or without adenovirus-short hairpin-PD-1 (Ad-sh-PD1), pre-miR-21 or adenovirus-green fluorescent protein (Ad-GFP), and the percentages of Th17/Treg cells and related transcription factors were measured. RESULTS: In patients who underwent gastric cancer resection, the number of Th17 cells decreased, whereas the number of Treg cells increased, accompanied by an increased expression of PD-1/PD-L1. In addition, the expression of RORγt and IL-17 decreased, whereas the expression of Foxp3 and TGF-ß1 increased. In vitro, silencing PD-1 via Ad-sh-PD1 promoted the expression of miR-21 and increased the percentage of Th17 cells, but decreased the percentage of Treg cells. The overexpression of miR-21 increased the percentage of Th17 cells but decreased the percentage of Treg cells. CONCLUSIONS: Our study demonstrated that gastric cancer resection altered the balance of Th17/Treg cells and increased PD-1/PD-L1 expression. In the in vitro experiments, the transfection of Ad-sh-PD1 ameliorated Th17/Treg cell imbalance partially by increasing the expression of miR-21.


Assuntos
Antígeno B7-H1/metabolismo , Gastrectomia/mortalidade , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Gástricas/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Biomarcadores Tumorais/metabolismo , Feminino , Seguimentos , Humanos , Interleucina-17/metabolismo , Masculino , Pessoa de Meia-Idade , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Prognóstico , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Taxa de Sobrevida , Fator de Crescimento Transformador beta1/metabolismo
20.
J Pain ; 19(10): 1113-1129, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29729431

RESUMO

Morphine is a potent opioid analgesic used to alleviate moderate or severe pain, but the development of drug tolerance and dependence limits its use in pain management. Previous studies showed that cannabinoid type 2 (CB2) receptor ligands may modulate opioid effects. However, there is no report of the effect of CB2 receptor agonist on acute morphine tolerance and physical dependence. We therefore investigated the effect of a CB2 receptor agonist (AM1241) on morphine-induced morphine tolerance and physical dependence in mice. Repeated coadministration of AM1241 (1 or 3mg/kg intraperitoneally) and morphine (10mg/kg subcutaneously) for 7days increased the mechanical paw withdrawal threshold in mice as measured by the von Frey filament test, and 3mg/kg AM1241 in combination with morphine increased the thermal paw withdrawal latency as measured by the hot-plate test. Combination with 3mg/kg AM1241 and morphine increased acute morphine antinociception. Coadministration of 1 or 3mg/kg AM1241 and morphine reduced acute morphine tolerance, and 3mg/kg AM1241 reduced chronic morphine tolerance. Coadministration of 1 or 3mg/kg AM1241 and morphine reduced naloxone-precipitated withdrawal jumping, but not diarrhea. Coadministration of AM1241 and morphine did not inhibit spontaneous locomotor activity. Pretreatment with 3mg/kg AM1241 decreased the chronic morphine-induced Iba1 expression in spinal cord. Coadministration of AM1241 (3 mg/kg) reduced the production of interleukin-1ß, tumor necrosis factor-α, and interleukin-6 induced by long-term and acute morphine treatment. Our findings suggest that the coadministration of the CB2 receptor agonist and morphine could increase morphine antinociception and reduce morphine tolerance and physical dependence in mice. PERSPECTIVE: The combination of a CB2 agonist and morphine may provide a new strategy for better treatment of acute and chronic pain and prevention of opioid tolerance and dependence. This finding may also provide a clue for the treatment of opioid tolerance and dependence in clinics.


Assuntos
Analgésicos Opioides/farmacologia , Tolerância a Medicamentos , Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Animais , Canabinoides/farmacologia , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dependência de Morfina , Limiar da Dor/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA