Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Purinergic Signal ; 19(2): 441-450, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36400869

RESUMEN

Cancer pain is the most prevalent symptom experienced by cancer patients. It substantially impacts a patient's long-term physical and emotional health, making it a pressing issue that must be addressed. Purinergic receptor P2X7 (P2X7R) is a widely distributed and potent non-selective ATP-gated ion channel that regulates tumor proliferation, chronic pain, and the formation of inflammatory lesions in the central nervous system. P2X7R plays an essential role in cancer pain and complications related to cancer pain including depression and opioid tolerance. This review focuses on the structure and distribution of P2X7R, its role in diverse tissues in cancer pain, and the application of P2X7R antagonists in the treatment of cancer pain to propose new ideas for cancer pain management.


Asunto(s)
Dolor en Cáncer , Neoplasias , Humanos , Receptores Purinérgicos P2X7 , Analgésicos Opioides , Antagonistas del Receptor Purinérgico P2X/farmacología , Tolerancia a Medicamentos , Neoplasias/complicaciones , Neoplasias/patología
2.
Mol Pain ; 18: 17448069211060962, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35057643

RESUMEN

BACKGROUND: The transition from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype presents a novel therapeutic strategy for chronic pain. OBJECTIVE: We investigated the role of microglia polarization in cancer-induced bone pain (CIBP), as well as the role of the P2X7 receptor in modulating M1 to M2 polarization. METHODS: Walker-256 breast cancer cells were administered into tibias of female rats to induce bone cancer-associated cancer. RESULTS: During bone cancer development, the P2X7 receptor and M1 microglia markers were upregulated. In contrast, inhibition of the P2X7 receptor by BBG, a blood-brain barrier-permeable P2X7R-specific antagonist, alleviated the pain and promoted microglia polarization toward the M2 phenotype, while suppressing the M1 phenotype in vivo and in vitro. CONCLUSION: P2X7 receptor-mediated spinal microglia polarization is involved in alleviation of CIBP. Therefore, P2X7R is a potential option for CIBP treatment.


Asunto(s)
Microglía , Neoplasias , Animales , Femenino , Microglía/fisiología , Dolor , Fenotipo , Ratas , Receptores Purinérgicos P2X7
3.
BMC Med Educ ; 21(1): 60, 2021 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-33461550

RESUMEN

BACKGROUND: Simulation training is an essential criterion for medical staff. The majority of residents are trained in operating room crisis management (ORCM), but only a few pre-clinical anesthesia undergraduate students are trained. Anesthesia methodology and technology were studied by the anesthesia undergraduate students in theory, but they were not able to practically resolve all clinical problems scientifically and reasonably. Consequently, there is a need to apply their competencies and bring together their technology knowledge practically. The crisis management of operating room emergencies was a method of choice applied and used over time. Here, we designed the scenarios for comprehensive crisis management to train anesthesia undergraduate students. We tried to establish or identify the problems which occurred during attempts to implement these scenarios. METHODS: Anesthesia undergraduate students initially examined the basic theory, fundamental practice techniques, and case studies before the simulation training program. Subsequently, they participated in comprehensive ORCM training. Training outcomes were evaluated through different viewpoints: understanding the subject, crisis management, nontechnical skills, and a user experience evaluation. RESULTS: Anesthesia undergraduate students performed significantly better with completion of ORCM, indicated by higher scores in all four tests (P < 0.001), as well as clinical crisis management (P = 0.0016) and nontechnical skills (P = 0.0002). Following the simulation, the students described the experience as helpful in "combining theoretical knowledge with clinical practice", helpful with memorization, and in "promoting understanding of the subject," while "learning clinical logic authentically" and "inspiring learning interests." CONCLUSIONS: This research indicates that ORCM could be implemented as a useful learning tool for pre-clinical anesthesia undergraduate students. The ORCM could be an excellent training method to help improve students' professional competence in crisis management and nontechnical skills, integrating the knowledge and technology of the field of anesthesiology.


Asunto(s)
Anestesiología , Hipotensión , Entrenamiento Simulado , Anestesiología/educación , Competencia Clínica , Humanos , Quirófanos , Estudiantes
4.
BMC Anesthesiol ; 19(1): 236, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31856760

RESUMEN

BACKGROUND: Perioperative immune function plays an important role in the prognosis of patients. Several studies have indicated that low-dose opioid receptor blockers can improve immune function. METHODS: Sixty-nine patients undergoing video-assisted thoracoscopic resection of the lung cancer were randomly assigned to either the naloxone group (n = 35) or the non-naloxone group (n = 34) for postoperative analgesia during the first 48 h after the operation. Both groups received sufentanil and palonosetron via postoperative analgesia pump, while 0.05 µg·kg- 1·h- 1 naloxone was added in naloxone group. The primary outcomes were the level of opioid growth factor (OGF) and immune function assessed by natural killer cells and CD4+/CD8+ T-cell ratio. Second outcomes were assessed by the intensity of postoperative pain, postoperative rescue analgesia dose, postoperative nausea and vomiting (PONV). RESULTS: The level of OGF in the naloxone group increased significantly at 24 h (p<0.001) and 48 h after the operation (P < 0.01). The natural killer cells (P < 0.05) and CD4+/CD8+ T-cell ratio (P < 0.01) in the naloxone group increased significantly at 48 h after the operation. The rest VAS scores were better with naloxone at 12 and 24 h after operation(P < 0.05), and the coughing VAS scores were better with naloxone at 48 h after the operation(P < 0.05). The consumption of postoperative rescue analgesics in the naloxone group was lower (0.00(0.00-0.00) vs 25.00(0.00-62.50)), P < 0.05). Postoperative nausea scores at 24 h after operation decreased in naloxone group(0.00 (0.00-0.00) vs 1.00 (0.00-2.00), P < 0.01). CONCLUSION: Infusion of 0.05 µg·kg- 1·h- 1 naloxone for patients undergoing sufentanil-controlled analgesia for postoperative pain can significantly increase the level of OGF, natural killer cells, and CD4+/CD8+ T-cell ratio compared with non-naloxone group, and postoperative pain intensity, request for rescue analgesics, and opioid-related side effects can also be reduced. TRIAL REGISTRATION: The trial was registered at the Chinese Clinical Trial Registry on January 26, 2019 (ChiCTR1900021043).


Asunto(s)
Neoplasias Pulmonares/cirugía , Naloxona/administración & dosificación , Sufentanilo/administración & dosificación , Cirugía Torácica Asistida por Video/métodos , Analgesia Controlada por el Paciente/métodos , Analgésicos Opioides/administración & dosificación , Relación CD4-CD8 , Femenino , Humanos , Sistema Inmunológico/efectos de los fármacos , Células Asesinas Naturales/inmunología , Masculino , Persona de Mediana Edad , Dolor Postoperatorio/tratamiento farmacológico , Proyectos Piloto , Náusea y Vómito Posoperatorios/epidemiología
5.
Cell Physiol Biochem ; 44(3): 1051-1063, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29179207

RESUMEN

BACKGROUND/AIMS: Cancer stem cells (CSCs) are considered to be responsible for tumor relapse and metastasis, which serve as a potential therapeutic target for cancer. Aspirin has been shown to reduce cancer risk and mortality, particularly in colorectal cancer. However, the CSCs-suppressing effect of aspirin and its relevant mechanisms in colorectal cancer remain unclear. METHODS: CCK8 assay was employed to detect the cell viability. Sphere formation assay, colony formation assay, and ALDH1 assay were performed to identify the effects of aspirin on CSC properties. Western blotting was performed to detect the expression of the stemness factors. Xenograft model was employed to identify the anti-cancer effects of aspirin in vivo. Unpaired Student t test, ANOVA test and Kruskal-Wallis test were used for the statistical comparisons. RESULTS: Aspirin attenuated colonosphere formation and decreased the ALDH1 positive cell population of colorectal cancer cells. Aspirin inhibited xenograft tumor growth and reduced tumor cells stemness in nude mice. Consistently, aspirin decreased the protein expression of stemness-related transcription factors, including c-Myc, OCT4 and NANOG. Suppression of NANOG blocked the effect of aspirin on sphere formation. Conversely, ectopic expression of NANOG rescued the aspirin-repressed sphere formation, suggesting that NANOG is a key downstream target. Moreover, we found that aspirin repressed NANOG expression in protein level by decreasing its stability. CONCLUSION: We have provided new evidence that aspirin attenuates CSC properties through down-regulation of NANOG, suggesting aspirin as a promising therapeutic agent for colorectal cancer treatment.


Asunto(s)
Aspirina/toxicidad , Proliferación Celular/efectos de los fármacos , Proteína Homeótica Nanog/metabolismo , Animales , Aspirina/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína Homeótica Nanog/antagonistas & inhibidores , Proteína Homeótica Nanog/genética , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Estabilidad Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción SOXB1/metabolismo , Trasplante Heterólogo
6.
J Biol Chem ; 290(4): 1994-2006, 2015 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-25492872

RESUMEN

Novel therapeutic regimens for tissue renewal incorporate mesenchymal stem cells (MSCs) as they differentiate into a variety of cell types and are a stem cell type that is easy to harvest and to expand in vitro. However, surface chemokine receptors, such as CXCR4, which are involved in the mobilization of MSCs, are expressed only on the surface of a small proportion of MSCs, and the lack of CXCR4 expression may underlie the low efficiency of homing of MSCs toward tissue damage, which results in a poor curative effect. Here, a rat CXCR4 expressing lentiviral vector was constructed and introduced into MSCs freshly prepared from rat bone marrow. The influence of CXCR4 expression on migration, proliferation, differentiation, and paracrine effects of MSCs was examined in vitro. The in vivo properties of CXCR4-MSCs were also investigated in a model of acute lung injury in rats induced by lipopolysaccharide. Expression of CXCR4 in MSCs significantly enhanced the chemotactic and paracrine characteristics of the cells in vitro but did not affect self-renewal or differentiation into alveolar and vascular endothelial cells. In vivo, CXCR4 improved MSC homing and colonization of damaged lung tissue, and furthermore, the transplanted CXCR4-MSCs suppressed the development of acute lung injury in part by modulating levels of inflammatory molecules and the neutrophil count. These results indicated that efficient mobilization of MSCs to sites of tissue injury may be due to CXCR4, and therefore, increased expression of CXCR4 may improve their therapeutic potential in the treatment of diseases where tissue damage develops.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Células Madre Mesenquimatosas/citología , Receptores CXCR4/metabolismo , Lesión Pulmonar Aguda/metabolismo , Animales , Médula Ósea/metabolismo , Líquido del Lavado Bronquioalveolar , Diferenciación Celular , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Quimiotaxis , Proteínas Fluorescentes Verdes/metabolismo , Inflamación , Lentivirus , Lipopolisacáridos/química , Masculino , Ratas , Transducción de Señal
8.
J Pharmacol Sci ; 124(1): 54-65, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24441773

RESUMEN

The therapeutic potential of adult neural stem cells (NSCs)-derived from bone marrow (BM) has been recently described in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis; however, the beneficial effects are modest due to their marginal anti-inflammatory capacity. To overcome this weakness and endow BM-NSC therapy with profound anti-inflammatory capacity, in this study we pretreated EAE mice with osthole, a natural coumarin with a broad spectrum of pharmacological activities, including anti-inflammation, immunomodulation, and neuroprotection, before NSC-application and continued throughout the study. We found that osthole conferred a potent anti-inflammatory capacity to this BM-NSC therapy, thus more profoundly suppressing ongoing EA and exhibiting significant advantages over conventional NSC-therapy as follows: 1) Enhanced anti-inflammatory effect, thus improving survival environment for engrafted BM-NSCs and protecting myelin sheaths from further demyelination; 2)Drove transplanted (exogenous) BM-NSCs to differentiate into more oligodendrocytes and neurons but inhibited differentiation into astrocytes, thus promoting remyelination and axonal growth, and reducing astrogliosis; and 3) augmented CNS neurotrophic support thus promoted resident (endogenous) repair of myelin/axonal damage. These effects make the BM-NSCs-based therapy a more promising approach to enhance remyelination and neuronal repopulation, thus more effectively promoting anatomic and functional recovery from neurological deficits.


Asunto(s)
Trasplante de Médula Ósea , Cumarinas/farmacología , Cumarinas/uso terapéutico , Encefalomielitis Autoinmune Experimental/terapia , Esclerosis Múltiple/terapia , Células-Madre Neurales/trasplante , Trasplante de Células Madre , Animales , Antiinflamatorios , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Enfermedades Desmielinizantes/prevención & control , Modelos Animales de Enfermedad , Femenino , Supervivencia de Injerto/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vaina de Mielina/patología , Células-Madre Neurales/citología , Neuronas , Fármacos Neuroprotectores , Oligodendroglía
9.
Stem Cell Res Ther ; 15(1): 211, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39020426

RESUMEN

Pain is a very common and complex medical problem that has a serious impact on individuals' physical and mental health as well as society. Non-steroidal anti-inflammatory drugs and opioids are currently the main drugs used for pain management, but they are not effective in controlling all types of pain, and their long-term use can cause adverse effects that significantly impair patients' quality of life. Mesenchymal stem cells (MSCs) have shown great potential in pain treatment. However, limitations such as the low proliferation rate of MSCs in vitro and low survival rate in vivo restrict their analgesic efficacy and clinical translation. In recent years, researchers have explored various innovative approaches to improve the therapeutic effectiveness of MSCs in pain treatment. This article reviews the latest research progress of MSCs in pain treatment, with a focus on methods to enhance the analgesic efficacy of MSCs, including engineering strategies to optimize the in vitro culture environment of MSCs and to improve the in vivo delivery efficiency of MSCs. We also discuss the unresolved issues to be explored in future MSCs and pain research and the challenges faced by the clinical translation of MSC therapy, aiming to promote the optimization and clinical translation of MSC-based analgesia therapy.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Manejo del Dolor , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Manejo del Dolor/métodos , Animales , Analgesia/métodos , Dolor/tratamiento farmacológico
10.
Eur J Pharmacol ; 967: 176380, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38311279

RESUMEN

Severe acute pancreatitis-associated acute lung injury (SAP-ALI) remains a significant challenge for healthcare practitioners because of its high morbidity and mortality; therefore, there is an urgent need for an effective treatment. Mesenchymal stem cells (MSCs) have shown significant potential in the treatment of a variety of refractory diseases, including lung diseases. This study aimed to investigate the protective effects of MSCs against SAP-ALI and its underlying mechanisms. Our results suggest that MSCs mitigate pathological injury, hemorrhage, edema, inflammatory response in lung tissue, and lipopolysaccharide (LPS)-induced cell damage in RLE-6TN cells (a rat alveolar epithelial cell line). The results also showed that MSCs, similar to the effects of ferrostatin-1 (ferroptosis inhibitor), suppressed the ferroptosis response, which was manifested as down-regulated Fe2+, malondialdehyde, and reactive oxygen species (ROS) levels, and up-regulated glutathione peroxidase 4 (GPX4) and glutathione (GSH) levels in vivo and in vitro. The activation of ferroptosis by erastin (a ferroptosis agonist) reversed the protective effect of MSCs against SAP-ALI. Furthermore, MSCs activated the nuclear factor erythroid 2 associated factor 2 (Nrf2) transcription factor, and blocking the Nrf2 signaling pathway with ML385 abolished the inhibitory effect of MSCs on ferroptosis in vitro. Collectively, these results suggest that MSCs have therapeutic effects against SAP-ALI. The specific mechanism involves inhibition of ferroptosis by activating the Nrf2 transcription factor.


Asunto(s)
Lesión Pulmonar Aguda , Ferroptosis , Células Madre Mesenquimatosas , Pancreatitis , Animales , Ratas , Enfermedad Aguda , Lesión Pulmonar Aguda/etiología , Antioxidantes/farmacología , Glutatión , Factor 2 Relacionado con NF-E2 , Pancreatitis/complicaciones
11.
Biol Pharm Bull ; 36(12): 1950-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24432380

RESUMEN

Accumulation of ß-amyloid peptide (Aß) in the brain plays an important role in the pathogenesis of Alzheimer's disease (AD). Previous studies have demonstrated the neuroprotective role of osthole against oxygen and glucose deprivation in cortical neurons. However, the effects of osthole on Aß-induced neurotoxicity in neural cells have rarely been reported. The current study was designed to investigate the protective effects of osthole on a cell model of AD insulted by exogenous Aß25-35 and the potential mechanism(s). In this study, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, immunofluorescence analysis, apoptosis assay, reverse transcription polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) techniques were used in primary cortical neurons and SH-SY5Y cells. Our data showed that osthole reduced intracellular Aß levels in neural cells, which was associated with decreased BACE1 protein; osthole reversed exogenous Aß25-35-induced cell viability loss, apoptosis, and synapsin-1 reduction, which was related to the reestablishment of phosphorylation of cyclic AMP response element-binding protein (CREB). The collective evidence indicates that osthole possesses the ability to protect cortical neurons and SH-SY5Y cells against Aß injury, and the underlying mechanism may be attributed to the enhancement of CREB phosphorylation.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Cumarinas/farmacología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/genética , Sinapsinas/metabolismo , Proteína X Asociada a bcl-2/genética
12.
Int J Mol Sci ; 14(9): 18657-69, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-24025424

RESUMEN

Arctigenin (Arc) has been shown to act on scopolamine-induced memory deficit mice and to provide a neuroprotective effect on cultured cortical neurons from glutamate-induced neurodegeneration through mechanisms not completely defined. Here, we investigated the neuroprotective effect of Arc on H89-induced cell damage and its potential mechanisms in mouse cortical neurons and human SH-SY5Y neuroblastoma cells. We found that Arc prevented cell viability loss induced by H89 in human SH-SY5Y cells. Moreover, Arc reduced intracellular beta amyloid (Aß) production induced by H89 in neurons and human SH-SY5Y cells, and Arc also inhibited the presenilin 1(PS1) protein level in neurons. In addition, neural apoptosis in both types of cells, inhibition of neurite outgrowth in human SH-SY5Y cells and reduction of synaptic marker synaptophysin (SYN) expression in neurons were also observed after H89 exposure. All these effects induced by H89 were markedly reversed by Arc treatment. Arc also significantly attenuated downregulation of the phosphorylation of CREB (p-CREB) induced by H89, which may contribute to the neuroprotective effects of Arc. These results demonstrated that Arc exerted the ability to protect neurons and SH-SY5Y cells against H89-induced cell injury via upregulation of p-CREB.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Furanos/farmacología , Lignanos/farmacología , Neuroblastoma/metabolismo , Fármacos Neuroprotectores/farmacología , Animales , Células Cultivadas , Humanos , Ratones , Fosforilación/efectos de los fármacos , Células Tumorales Cultivadas
13.
J Breast Cancer ; 26(3): 268-285, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37387350

RESUMEN

PURPOSE: Invasive ductal carcinoma (IDC) accounts for 90% of triple-negative breast cancer (TNBC). IDC is mainly derived from the breast ductal epithelium which is innervated by the 4th to 6th thoracic sympathetic nerves. However, little is known about the contribution of the interactions between sympathetic nerves and breast cancer cells to the malignant progression of TNBC. METHODS: The expression levels of the ß2-adrenergic receptor (ß2-AR, encoded by ADRB2 gene), nerve growth factor (NGF), and tropomyosin receptor kinase A (TrkA) were determined using immunohistochemistry (IHC). NGF expression levels in the serum were compared by enzyme-linked immunosorbent assay (ELISA). Cell proliferation was assessed using the Cell Counting Kit-8 assay. The ß2-AR, NGF, p-ERK, and p-CERB expression levels were determined using western blotting. TNBC cells and neuronal cells of the dorsal root ganglion (DRG) in 2-day-old Sprague Dawley rats were co-cultured. Using norepinephrine (NE), NGF, and ß2-AR, NGF/TrkA blocker pretreatments, the axon growth of each group of DRG neuron cells was detected by immunofluorescence analysis. RESULTS: The sympathetic adrenergic neurotransmitter NE activated the ERK signaling pathway in TNBC cells. NE/ß2-AR signaling promotes NGF secretion. NGF further facilitates the malignant progression of TNBC by increasing sympathetic neurogenesis. In the co-culture assay, the sympathetic adrenergic NE/ß2-AR signal pathway also enhanced NGF secretion. NGF binds TrkA in DRG neurons and promotes axonal growth. CONCLUSION: These results suggest that NE/ß2-AR pathway promotes cell proliferation and NGF production in triple-negative breast cancer.

14.
J Pain Res ; 16: 1767-1778, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37273275

RESUMEN

Postoperative pain is one of the most prevalent complications following surgery, and more than 47% of surgical patients endure postoperative discomfort worldwide. In Africa, due to resource shortages and other issues, postoperative pain is substantially more common when compared to developed countries. Severe postoperative pain has many negative effects, including possibly death, which can burden both individuals and society as a whole. Therefore, effectively controlling postoperative pain is becoming increasingly important. To enhance the effectiveness of future pain management, a thorough analysis of the current reasons for inadequate postoperative pain management is necessary. In this article, the present situations of occurring postoperative pain, children's postoperative pain, and pain management in Africa are reviewed, based on relevant and recent literature. In particular, the reasons for inadequate postoperative pain management in Africa are detailed in this article from five perspectives: the inadequate assessment of postoperative pain, the knowledge gap among medical professionals, the patients' misconceptions, the scarcity of resources, and the lack of medications. Additionally, we offer appropriate solutions following various factors.

15.
Front Pharmacol ; 14: 1189486, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37251314

RESUMEN

Background: Severe acute pancreatitis (SAP) is a severe form of acute pancreatitis with the potential to cause life-threatening complications. Patients with acute SAP require surgical intervention and are admitted to the intensive care unit for non-invasive ventilation. Dexmedetomidine (Dex) is currently used by intensive care clinicians and anaesthesiologists as an adjunctive sedative. Therefore, the clinical availability of Dex makes it easier to implement in SAP treatment than developing new drugs. Methods: Randomly dividing thirty rats into sham-operated (Sham), SAP, and Dex groups. The severity of pancreatic tissue injury in each rat was assessed by Hematoxylin and eosin (HE) staining. Serum amylase activity and inflammatory factor levels were measured using commercially available kits. The expressions of necroptosis-related proteins, myeloperoxidase (MPO), CD68, and 4-hydroxy-trans-2-nonenal (HNE) were detected using immunohistochemistry (IHC). Transferase-mediated dUTP nick-end labeling (TUNEL) staining was utilized to identify pancreatic acinar cell apoptosis. The subcellular organelle structure of pancreatic acinar cells was observed using transmission electron microscopy. The regulatory effect of Dex on the gene expression profile of SAP rat pancreas tissue was investigated using RNA sequencing. We screened for differentially expressed genes (DEGs). Quantitative real-time PCR (qRT-PCR) measured critical DEG mRNA expression in rat pancreatic tissues. Results: Dex attenuated SAP-induced pancreatic injury, infiltration of neutrophils and macrophages, and oxidative stress. Dex inhibited the expression of necroptosis-associated proteins RIPK1, RIPK3, and MLKL and alleviated apoptosis in acinar cells. Dex also mitigated the structural damage caused by SAP to mitochondria and endoplasmic reticulum. Dex inhibited SAP-induced 473 DEGs, as determined by RNA sequencing. Dex may regulate SAP-induced inflammatory response and tissue damage by inhibiting the toll-like receptor/nuclear factor κB (TLR/NF-κB) signaling pathway and neutrophil extracellular trap formation. Conclusion: This study elucidated the remarkable effect of Dex against SAP and investigated the potential mechanism of action, providing an experimental base for the future clinical application of Dex in the treatment of SAP.

16.
Biomolecules ; 12(11)2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36358919

RESUMEN

Autophagy is a critical cellular adaptive response in tumor formation. Nutritional deficiency and hypoxia exacerbate autophagic flux in established malignancies, promoting tumor cell proliferation, migration, metastasis, and resistance to therapeutic interventions. Pro-survival autophagy inhibition may be a promising treatment option for advanced cancer. Furthermore, excessive or persistent autophagy is cytotoxic, resulting in tumor cell death. Targeted autophagy activation has also shown significant promise in the fight against tumor drug resistance. Several research groups have examined the ability of natural products (NPs) such as alkaloids, terpenoids, polyphenols, and anthraquinones to serve as autophagy inhibitors or activators. The data support the capacity of NPs that promote lethal autophagy or inhibit pro-survival autophagy from being employed against tumor drug resistance. This paper discusses the potential applications of NPs that regulate autophagy in the fight against tumor drug resistance, some limitations of the current studies, and future research needs and priorities.


Asunto(s)
Antineoplásicos , Productos Biológicos , Neoplasias , Humanos , Productos Biológicos/farmacología , Productos Biológicos/uso terapéutico , Autofagia , Resistencia a Antineoplásicos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/metabolismo , Línea Celular Tumoral
17.
Pain Physician ; 25(8): E1199-E1210, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36375190

RESUMEN

BACKGROUND: Bone cancer pain (BCP) is the most severe and intractable type of cancer pain. Emerging evidence has demonstrated that activated microglia in the spinal cord could release a series of neurotoxic substances to stimulate neurons and form neuronal sensitization. The P2X7 receptor (P2X7R) is a nonselective ATP-gated ion channel predominantly present in microglia in the spinal cord as the key modulator of microglial activity. However, the specific effect and underlying molecular mechanism of P2X7R in BCP have not yet been elucidated. OBJECTIVES: This study aimed at investigating whether P2X7R-induced BCP by regulating microglial activity through NLRP3/IL-1beta signaling involvement in BCP. STUDY DESIGN: Controlled animal study. METHODS: A BCP animal model was established by injecting Walker-256 breast cancer cells into the tibia of female rats. Fifty percent paw withdrawal thresholds (50% PWTs), number of spontaneous flinches (NSF), and limb use scores were used to evaluate behavior in rats. P2X7R inhibitor brilliant blue G (BBG) was used to assess the role of P2X7R in BCP-induced NLRP3 inflammasome activation. Western blot, RT-PCR, and immunofluorescence were used for quantitative comparison. In vitro, BV2 cells were treated with lipopolysaccharide (LPS) and BzATP, in the presence or absence of P2X7 siRNA, with nigericin (an agonist of the NLRP3 inflammasome) to further study the mechanism of P2X7R regulate NLRP3/IL-1beta signaling. RESULTS: The inhibition of spinal P2X7R with BBG could effectively inhibit BCP due to suppressing the expression of NF-kappaB p-p65, NLRP3 inflammasome formation, and downstream pain factors IL-1beta. Furthermore, P2X7 siRNA could reduce microglial activity, the nuclear translocation of NF-kappaB, and the synthesis of NLRP3 and IL-1beta in BV2 cells. In addition, nigericin partially reversed the ameliorating effect of P2X7 siRNA on BV2 cells induced by LPS and BzATP. LIMITATIONS: BBG could relieve BCP but not improve the destruction of bone, which may be related to the specificity of inoculated cells. Further mechanisms should be investigated. CONCLUSION: These findings suggest that targeting the microglial P2X7R activated NLRP3/IL-1beta signaling pathway could serve as a potential strategy for BCP treatment.


Asunto(s)
Dolor en Cáncer , Neoplasias , Receptores Purinérgicos P2X7 , Animales , Femenino , Ratas , Dolor en Cáncer/tratamiento farmacológico , Dolor en Cáncer/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Microglía/metabolismo , FN-kappa B/metabolismo , Nigericina/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Purinérgicos P2X7/metabolismo , ARN Interferente Pequeño/metabolismo , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología
18.
Bioengineering (Basel) ; 9(12)2022 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-36550973

RESUMEN

The leading cause of many respiratory diseases is an ongoing and progressive inflammatory response. Traditionally, inflammatory lung diseases were studied primarily through animal models, cell cultures, and organoids. These technologies have certain limitations, despite their great contributions to the study of respiratory diseases. Precision-cut lung slices (PCLS) are thin, uniform tissue slices made from human or animal lung tissue and are widely used extensively both nationally and internationally as an in vitro organotypic model. Human lung slices bridge the gap between in vivo and in vitro models, and they can replicate the living lung environment well while preserving the lungs' basic structures, such as their primitive cells and trachea. However, there is no perfect model that can completely replace the structure of the human lung, and there is still a long way to go in the research of lung slice technology. This review details and analyzes the strengths and weaknesses of precision lung slices as an in vitro model for exploring respiratory diseases associated with inflammation, as well as recent advances in this field.

19.
Signal Transduct Target Ther ; 7(1): 97, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35361747

RESUMEN

Aberrant RNA splicing produces alternative isoforms of genes to facilitate tumor progression, yet how this process is regulated by oncogenic signal remains largely unknown. Here, we unveil that non-canonical activation of nuclear AURKA promotes an oncogenic RNA splicing of tumor suppressor RBM4 directed by m6A reader YTHDC1 in lung cancer. Nuclear translocation of AURKA is a prerequisite for RNA aberrant splicing, specifically triggering RBM4 splicing from the full isoform (RBM4-FL) to the short isoform (RBM4-S) in a kinase-independent manner. RBM4-S functions as a tumor promoter by abolishing RBM4-FL-mediated inhibition of the activity of the SRSF1-mTORC1 signaling pathway. Mechanistically, AURKA disrupts the binding of SRSF3 to YTHDC1, resulting in the inhibition of RBM4-FL production induced by the m6A-YTHDC1-SRSF3 complex. In turn, AURKA recruits hnRNP K to YTHDC1, leading to an m6A-YTHDC1-hnRNP K-dependent exon skipping to produce RBM4-S. Importantly, the small molecules that block AURKA nuclear translocation, reverse the oncogenic splicing of RBM4 and significantly suppress lung tumor progression. Together, our study unveils a previously unappreciated role of nuclear AURKA in m6A reader YTHDC1-dependent oncogenic RNA splicing switch, providing a novel therapeutic route to target nuclear oncogenic events.


Asunto(s)
Empalme Alternativo , Aurora Quinasa A , Proteínas del Tejido Nervioso , Factores de Empalme de ARN , Proteínas de Unión al ARN , Aurora Quinasa A/genética , Aurora Quinasa A/metabolismo , Núcleo Celular/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Empalme del ARN , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
20.
J Ethnopharmacol ; 264: 113246, 2021 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-32781257

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Cyperus rotundus L. (Cyperaceae) is a widespread herbal in China and widely used in Traditional Chinese Medicine for multiple effects such as anti-arthritic, anti-genotoxic, anti-mutagenic, anti-bacterial effects, and analgesic. α-Cyperone is an active compound in Cyperus rotundus and has analgesic effects, but the exact molecular mechanisms require further investigations. MATERIALS AND METHODS: Tumor-derived DNA isolated from Lewis cell lines was transfected into microglia, and analyzed for stimulator of interferon genes (STING) effects. The downstream protein, such as interferon regulatory factor 3 (IRF3) and p65 nuclear factor-κB (NF-κB) were treated with STING siRNA and 5,6-dimethyllxanthenone-4-acetic acid (DMXAA) in microglia. The α-Cyperone effect on microglia was also investigated. RESULTS: Tumor-derived DNA activate microglia by upregulation of STING and downstream proteins. STING siRNA was reduced to its downstream expression and neuroinflammation inhibition was caused by tumor-derived DNA. However, DMXAA reversed the STING siRNA effect and increased neuroinflammation. α-Cyperone takes inhibitory effects on tumor-derived DNA that trigger microglia by STING pathway. CONCLUSIONS: α-Cyperone inhibition by tumor-derived DNA activated microglial to neuroinflammation in STING signaling pathway.


Asunto(s)
ADN de Neoplasias/antagonistas & inhibidores , ADN de Neoplasias/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Microglía/efectos de los fármacos , Naftalenos/farmacología , Animales , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Carcinoma Pulmonar de Lewis/genética , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Ratones , Microglía/fisiología , Naftalenos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA