Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.004
Filtrar
1.
J Ethnopharmacol ; 336: 118716, 2025 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-39179055

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Panax notoginseng flowers, which are the buds of the traditional Chinese medicinal herb Sanqi, are widely used in China for their cough-ameliorating properties, with demonstrated therapeutic effects in the treatment of both acute and chronic coughs. However, both the antitussive mechanism and active compound basis of P. notoginseng flowers remain poorly understood. AIM OF THE STUDY: We investigated the antitussive effects of P. notoginseng flowers, identified the bioactive constituents responsible for alleviating cough symptoms, and elucidated the underlying pharmacological mechanisms. MATERIALS AND METHODS: We analyzed the major chemical constituents of aqueous extracts of P. notoginseng flowers using liquid chromatography-mass spectrometry and quantitatively analyzed the key component, 20S-ginsenoside Rh2, using high-performance liquid chromatography. Using a cough reflex model in healthy mice and an ovalbumin-induced, highly sensitive guinea pig cough model, we verified the suppressive effects of P. notoginseng flowers and their saponin constituents on coughing. Furthermore, we explored the mechanisms of action of the key ion channels, NaV1.7 and TRPV1, using whole-cell patch-clamp techniques and molecular docking. Finally, the therapeutic mechanisms of P. notoginseng flowers on pathological cough were revealed using hematoxylin and eosin staining, immunohistochemistry, and western blotting. RESULTS: The active components of P. notoginseng flowers were primarily protopanaxadiol-type saponins, among which 20S-ginsenoside Rh2 had the highest content (51.46 mg/g). In the mouse model, P. notoginseng flowers exhibited antitussive effects comparable to those of pentoxyverine citrate. Although its main saponin component, 20S-ginsenoside Rh2, showed slightly weaker effects, it still demonstrated concentration-dependent inhibition of channel activity. The whole-cell patch-clamp technique and virtual molecular docking showed that Rh2 might exert its effects by directly binding to the NaV1.7 and TRPV1 channels. In the guinea pig model, P. notoginseng flowers and their saponin components not only reduced cough frequency and prolonged the latency period before cough onset, but also significantly inhibited tracheal and pulmonary inflammation and the overexpression of TRPV1. CONCLUSIONS: 20S-Ginsenoside Rh2, the major bioactive saponin in P. notoginseng flowers, exhibits potent antitussive effects. The potential mechanism of action of 20S-Ginsenoside Rh2 in the treatment of cough may involve inhibiting NaV1.7 and TRPV1 channel currents through direct binding to core protein active sites and downregulating TRPV1 expression.


Asunto(s)
Antitusígenos , Tos , Regulación hacia Abajo , Flores , Ginsenósidos , Canal de Sodio Activado por Voltaje NAV1.7 , Panax notoginseng , Canales Catiónicos TRPV , Animales , Canales Catiónicos TRPV/metabolismo , Cobayas , Flores/química , Tos/tratamiento farmacológico , Ginsenósidos/farmacología , Antitusígenos/farmacología , Masculino , Ratones , Panax notoginseng/química , Regulación hacia Abajo/efectos de los fármacos , Humanos , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Células HEK293 , Simulación del Acoplamiento Molecular , Cricetulus , Modelos Animales de Enfermedad , Células CHO , Saponinas/farmacología , Ovalbúmina
2.
J Cell Mol Med ; 28(20): e70138, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39431551

RESUMEN

Myeloproliferative neoplasms (MPNs) are characterized by an increased production of blood cells due to the acquisition of mutations such as JAK2V617F. TGF-ß, whose secretion is increased in MPN patients, is known to negatively regulate haematopoietic stem cell (HSC) proliferation. Using an isogenic JAK2V617F or JAK2 wild-type UT-7 cell line we observed that JAK2V617F cells resist to TGF-ß antiproliferative activity. Although TGF-ß receptors and SMAD2/3 expressions are similar in both cell types, TGF-ß-induced phosphorylation of SMAD2/3 is reduced in UT-7 JAK2V617F cells compared with JAK2 WT cells. We confirmed that JAK2V617F mutated cells are resistant to the antiproliferative effect of TGF-ß in a competitive assay as we observed a positive selection of JAK2V617F cells when exposed to TGF-ß. Using cell lines, CD34-positive cells from MPN patients and bone marrow cells from JAK2V617F knock-in mice we identified a down regulation of the SHP-1 phosphatase, which is required for the regulation of HSC quiescence by TGF-ß. The transduction of SHP-1 cDNA (but not a phosphatase inactive cDNA) restores the antiproliferative effect of TGF-ß in JAK2V617F mutated cells. Finally, SC-1, a known agonist of SHP-1, antagonized the selection of JAK2V617F mutated cells in the presence of TGF-ß. In conclusion, we show a JAK2-dependent down regulation of SHP-1 in MPN patients' cells which is related to their resistance to the antiproliferative effect of TGF-ß. This may participate in the clonal selection of cancer cells in MPNs.


Asunto(s)
Proliferación Celular , Janus Quinasa 2 , Trastornos Mieloproliferativos , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Factor de Crecimiento Transformador beta , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Humanos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Ratones , Línea Celular Tumoral , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Regulación hacia Abajo/genética , Regulación hacia Abajo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteína Smad2/metabolismo , Proteína Smad2/genética , Mutación/genética
4.
Braz J Biol ; 84: e286928, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39383417

RESUMEN

Early postnatal administration of antibiotics has been linked to lasting effects on brain development and behavior. Research conducted on animals that are free from germs has demonstrated that the impact of microbiome colonization on the regulation of the hypothalamic-pituitary-adrenal (HPA) axis and neuroendocrine pathways is substantial, which play a crucial role in stress management. Nevertheless, it is still uncertain if the exposure to antibiotics in rat dams (F0-generation) before weaning is associated with neurobehavioral changes in rat offspring (F1-generation) during adulthood. In order to investigate the effects, we perturbed the intestinal microbiota of rat dams (F0 generation) by administering cefixime (CEF), an antibiotic commonly used for obstetric purposes, at clinically relevant doses (1 mg/kg, 2.5 mg/kg or 5 mg/kg). Anxiety-like behaviors in adult offspring was evaluated through the utilization of elevated plus maze (EPM) and open field paradigm (OFP) following a six-week interval from birth (PND42). Subsequent to behavioral assessments, the rats were euthanized, and their brains and blood was collected for biochemical analysis. Plasma corticosterone concentration was used to assess HPA activity, whereas the quantitative real-time polymerase chain reaction (PCR) was employed to determine the transcription levels of the glucocorticoid receptor (GR) Nr3c1. The offspring of F1 that were administered antibiotics before being weaned spent less time in the EPM open arm. The alterations were accompanied by increased levels of corticosterone in the bloodstream. The gene expression study revealed a decrease in the levels of mRNA transcription of Nr3c1. This research emphasizes the possible long-term effects of antibiotic exposure before weaning on the development of anxiety in offspring upon adulthood.


Asunto(s)
Antibacterianos , Ansiedad , Regulación hacia Abajo , Sistema Hipotálamo-Hipofisario , Sistema Hipófiso-Suprarrenal , Ratas Wistar , Receptores de Glucocorticoides , Animales , Receptores de Glucocorticoides/metabolismo , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Femenino , Antibacterianos/farmacología , Masculino , Regulación hacia Abajo/efectos de los fármacos , Ratas , Corticosterona/sangre , Embarazo , Efectos Tardíos de la Exposición Prenatal , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Prueba de Campo Abierto/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Animales Recién Nacidos
5.
Mol Biol Rep ; 51(1): 1054, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39412689

RESUMEN

Aluminum chloride (Al) is associated with Alzheimer's disease (AD) and reproductive disorders. But the relationship between gonadotropin-releasing hormone (GnRH) and c-Fos levels, the end product of MAP-kinase signaling, in AD is unknown, so we aimed to investigate this relationship. We exposed rats to Al dissolved in drinking water (10 and 50 mg/kg) for two and four weeks. The control group received only drinking water. At the end, the blood sample was collected under deep anesthesia and the brain was dissected on ice, and the testicular tissue was fixed in formalin. Amyloid beta (ßA) plaques in brain regions and the number of CA1 neurons were evaluated by Congo red staining and cresyl violet staining. Activation of neuronal nitric oxide synthase (nNOS) was studied using NADPH-diaphorase. The levels of c-Fos and testosterone receptors in the target area were examined immunohistochemically. Brain GnRH levels were determined by blotting, and serum levels of gonadotropins and steroids were measured by enzyme-linked immunosorbent assay (ELISA). All data were analyzed using analysis of variance (ANOVA) at α = 0.05 level. The accumulation of ßA plaque was observed along with a decrease in the number of CA1 pyramidal neurons and a significant decrease in the levels of c-Fos and GnRH in the brains of rats receiving Al, which was aligned with a significant decrease in serum levels of testosterone and LH. This study, for the first time, showed a link between dementia and a concomitant decrease in brain GnRH and c-Fos levels.


Asunto(s)
Cloruro de Aluminio , Enfermedad de Alzheimer , Hormona Liberadora de Gonadotropina , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-fos , Animales , Hormona Liberadora de Gonadotropina/metabolismo , Masculino , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Cloruro de Aluminio/efectos adversos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Memoria/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ratas Wistar , Aprendizaje/efectos de los fármacos , Modelos Animales de Enfermedad , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/metabolismo , Placa Amiloide/metabolismo , Placa Amiloide/patología
6.
Life Sci ; 357: 123085, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39362584

RESUMEN

AIMS: Alzheimer's disease (AD) is characterized by ß-amyloid (Aß) aggregation and neuroinflammation, leading to progressive synaptic loss and cognitive decline. Recent evidence suggests that Galectin-3 (Gal-3) plays a critical role in Aß pathogenesis. However, strategies to simultaneously target Gal-3 and Aß are currently insufficient. This study evaluates the therapeutic efficacy of (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one (D30), in reducing Gal-3 and Aß pathogenesis. MATERIALS AND METHODS: We applied exogenous oligomeric Aß and used 5 × FAD mice to assess the impact of Aß on Gal-3 deposition, microglial activation, and cognitive function. Thy1-EGFP mice were employed to observe dendritic spines. Comprehensive evaluations of D30's effects included behavioral studies, transcriptomic analysis, Western blotting, and immunofluorescent staining. The interaction between D30 and Gal-3 was examined using fluorescence resonance energy transfer (FRET) and microscale thermophoresis (MST). KEY FINDINGS: D30 effectively reduced Aß monomer production by inhibiting Amyloid Precursor Protein (APP) and presenilin 1 (PS1) expression, and decreased Aß aggregation. Treatment with D30 improved cognitive functions, reversed dendritic spine loss, and increased PSD95 expression in 5 × FAD mice. Additionally, D30 significantly lowered Gal-3 levels in both plasma and hippocampal tissues. D30 binds to Gal-3 and disrupts the interaction between Gal-3 and TREM2, as confirmed by FRET and MST. SIGNIFICANCE: Our findings underscore the interaction between Gal-3 and Aß in AD and its role in systemic inflammation using the 5 × FAD mouse model. Being able to target and regulate Gal-3 together with Aß is crucial for preventing neuroinflammation and protecting synapses, D30 emerged as a novel compound with promising potential for AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Enfermedades Neuroinflamatorias , Animales , Ratones , Péptidos beta-Amiloides/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ratones Transgénicos , Masculino , Galectina 3/metabolismo , Galectina 3/genética , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Microglía/efectos de los fármacos , Microglía/metabolismo , Galectinas/metabolismo
7.
Signal Transduct Target Ther ; 9(1): 293, 2024 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-39438476

RESUMEN

Clinical evidence supports the notion that T cell exhaustion and terminal differentiation pose challenges to the persistence and effectiveness of chimeric antigen receptor-T (CAR-T) cells. MEK1/2 inhibitors (MEKIs), widely used in cancer treatment due to their ability to inhibit aberrant MAPK signaling, have shown potential synergistic effects when combined with immunotherapy. However, the impact and mechanisms of MEKIs on CAR-T cells remain uncertain and controversial. To address this, we conducted a comprehensive investigation to determine whether MEKIs enhance or impair the efficacy of CAR-T cells. Our findings revealed that MEKIs attenuated CAR-T cell exhaustion and terminal differentiation induced by tonic signaling and antigen stimulation, thereby improving CAR-T cell efficacy against hematological and solid tumors. Remarkably, these effects were independent of the specific scFvs and costimulatory domains utilized in CARs. Mechanistically, analysis of bulk and single-cell transcriptional profiles demonstrates that the effect of MEK inhibition was related to diminish anabolic metabolism and downregulation of c-Fos and JunB. Additionally, the overexpression of c-Fos or JunB in CAR-T cells counteracted the effects of MEK inhibition. Furthermore, our Cut-and-Tag assay revealed that MEK inhibition downregulated the JunB-driven gene profiles associated with exhaustion, differentiation, anergy, glycolysis, and apoptosis. In summary, our research unveil the critical role of the MAPK-c-Fos-JunB axis in driving CAR-T cell exhaustion and terminal differentiation. These mechanistic insights significantly broaden the potential application of MEKIs to enhance the effectiveness of CAR-T therapy.


Asunto(s)
Diferenciación Celular , Proteínas Proto-Oncogénicas c-fos , Humanos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Ratones , Regulación hacia Abajo/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Inmunoterapia Adoptiva , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Agotamiento de Células T , Factores de Transcripción
8.
Int J Mol Sci ; 25(19)2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39408627

RESUMEN

Cardiac remodeling, a critical process that can lead to heart failure, is primarily characterized by cardiac hypertrophy. Studies have shown that transgenic mice with Gαq receptor blockade exhibit reduced hypertrophy under induced pressure overload. GQ262, a novel Gαq/11 inhibitor, has demonstrated good biocompatibility and specific inhibitory effects on Gαq/11 compared to other inhibitors. However, its role in cardiac remodeling remains unclear. This study aims to explore the anti-cardiac remodeling effects and mechanisms of GQ262 both in vitro and in vivo, providing data and theoretical support for its potential use in treating cardiac remodeling diseases. Cardiac hypertrophy was induced in mice via transverse aortic constriction (TAC) for 4 weeks and in H9C2 cells through phenylephrine (PE) induction, confirmed with WGA and H&E staining. We found that GQ262 improved cardiac function, inhibited the protein and mRNA expression of hypertrophy markers, and reduced the levels of apoptosis and fibrosis. Furthermore, GQ262 inhibited the Akt/mTOR signaling pathway activation induced by TAC or PE, with its therapeutic effects disappearing upon the addition of the Akt inhibitor ARQ092. These findings reveal that GQ262 inhibits cardiomyocyte hypertrophy and apoptosis through the Akt/mTOR signaling pathway, thereby reducing fibrosis levels and mitigating cardiac remodeling.


Asunto(s)
Cardiomegalia , Miocitos Cardíacos , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Remodelación Ventricular , Animales , Ratones , Ratas , Apoptosis/efectos de los fármacos , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/tratamiento farmacológico , Línea Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Fibrosis , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Fenilefrina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Remodelación Ventricular/efectos de los fármacos
9.
Int J Mol Sci ; 25(19)2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39409130

RESUMEN

Thyroid hormone binds to specific nuclear receptors, regulating the expression of target genes, with major effects on cardiac function. Triiodothyronine (T3) increases the expression of key proteins related to calcium homeostasis, such as the sarcoplasmic reticulum calcium ATPase pump, but the detailed mechanism of gene regulation by T3 in cardiac voltage-gated calcium (Cav1.2) channels remains incompletely explored. Furthermore, the effects of T3 on Cav1.2 auxiliary subunits have not been investigated. We conducted quantitative reverse transcriptase polymerase chain reaction, Western blot, and immunofluorescence experiments in H9c2 cells derived from rat ventricular tissue, examining the effects of T3 on the expression of α1c, the principal subunit of Cav1.2 channels, and Cavß4, an auxiliary Cav1.2 subunit that regulates gene expression. The translocation of phosphorylated cyclic adenosine monophosphate response element-binding protein (pCREB) by T3 was also examined. We found that T3 has opposite effects on these channel proteins, upregulating α1c and downregulating Cavß4, and that it increases the nuclear translocation of pCREB while decreasing the translocation of Cavß4. Finally, we found that overexpression of Cavß4 represses the mRNA expression of α1c, suggesting that T3 upregulates the expression of the α1c subunit in response to a decrease in Cavß4 subunit expression.


Asunto(s)
Canales de Calcio Tipo L , Miocitos Cardíacos , Animales , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/genética , Ratas , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Triyodotironina/farmacología , Triyodotironina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Hormonas Tiroideas/metabolismo , Línea Celular , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Subunidades de Proteína/metabolismo , Subunidades de Proteína/genética
10.
Life Sci ; 357: 123111, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39369843

RESUMEN

AIMS: The incidence of recurrent gliomas is high, exerting low survival rates and poor prognoses. Transcription factor AP-2α has been reported to regulate the progression of primary glioblastoma (GBM). However, the function of AP-2α in recurrent gliomas is largely unclear. METHODS: The expression of AP-2α and O6-methylguanine DNA-methyltransferase (MGMT) was detected in recurrent glioma tissues and cell lines by Western blots, the regulation mechanisms between AP-2α/MGMT promoter and RA/AP-2α promoter were studied by luciferase reporter assays, EMSA, and chIP assays. The effects of AP-2α and TMZ/RA treatment on cell viability in vitro and in vivo were investigated by MTT assays, γH2AX staining, comet assays and intracranial injection. KEY FINDINGS: AP-2α expression negatively correlates with the expression of MGMT in glioma samples. AP-2α could directly bind with the promoter of the MGMT gene, suppresses transcriptional levels of MGMT and downregulate MGMT expression in TMZ-resistant U87MG-R and T98G cells, but TMZ treatment decreases AP-2α expression and increases MGMT expression. The extended TMZ treatment and increased TMZ concentrations reversed these effects. Moreover, AP-2α overexpression combines with TMZ to decrease cell viability, concurrently with improved DNA damage marker γH2AX. Furthermore, retinoic acid (RA) activates RAR/RXR heterodimers, which bind to RA-responsive elements (RAREs) of the AP-2α promoter, and activates AP-2α expression in recurrent glioma cells. Finally, in intracranial relapsed glioma mouse model, both RA and TMZ could retard tumor development and prolong the mouse survival. SIGNIFICANCE: AP-2α activation by gene overexpression or RA treatment reveals the suppressive effects on glioma relapse, providing a novel therapeutic strategy against malignant refractory gliomas.


Asunto(s)
Antineoplásicos Alquilantes , Neoplasias Encefálicas , Daño del ADN , Metilasas de Modificación del ADN , Enzimas Reparadoras del ADN , Regulación hacia Abajo , Resistencia a Antineoplásicos , Glioblastoma , Temozolomida , Factor de Transcripción AP-2 , Proteínas Supresoras de Tumor , Animales , Femenino , Humanos , Ratones , Persona de Mediana Edad , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/metabolismo , Regiones Promotoras Genéticas , Temozolomida/farmacología , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Int J Mol Sci ; 25(19)2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39408695

RESUMEN

Porcine reproductive and respiratory syndrome (PRRS), caused by PRRS virus (PRRSV) infection, has been a serious threat to the pork industry worldwide and continues to bring significant economic loss. Current vaccination strategies offer limited protection against PRRSV transmission, highlighting the urgent need for novel antiviral approaches. In the present study, we reported for the first time that betulonic acid (BA), a widely available pentacyclic triterpenoids throughout the plant kingdom, exhibited potent inhibition on PRRSV infections in both Marc-145 cells and primary porcine alveolar macrophages (PAMs), with IC50 values ranging from 3.3 µM to 3.7 µM against three different type-2 PRRSV strains. Mechanistically, we showed that PRRSV replication relies on energy supply from cellular ATP production, and BA inhibits PRRSV infection by reducing cellular ATP production. Our findings indicate that controlling host ATP production could be a potential strategy to combat PRRSV infections, and that BA might be a promising therapeutic agent against PRRSV epidemics.


Asunto(s)
Adenosina Trifosfato , Antivirales , Macrófagos Alveolares , Virus del Síndrome Respiratorio y Reproductivo Porcino , Replicación Viral , Virus del Síndrome Respiratorio y Reproductivo Porcino/efectos de los fármacos , Virus del Síndrome Respiratorio y Reproductivo Porcino/fisiología , Animales , Replicación Viral/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Porcinos , Macrófagos Alveolares/virología , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Antivirales/farmacología , Línea Celular , Ácido Oleanólico/farmacología , Síndrome Respiratorio y de la Reproducción Porcina/virología , Síndrome Respiratorio y de la Reproducción Porcina/metabolismo , Chlorocebus aethiops , Regulación hacia Abajo/efectos de los fármacos
12.
Arthritis Res Ther ; 26(1): 173, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39350214

RESUMEN

BACKGROUND: CXC chemokine CXCL12 is involved in the pathological development of rheumatoid arthritis (RA) through abnormal migration of peripheral immune cells in the joint. Although low dose methotrexate (MTX) is clinically used to treat RA patients, CXCL12 signaling responses to MTX-mediated treatments is still not well understood. METHODS: In this study, we examined the expression of CXCR4 (cognatic receptor for CXCL12) in peripheral T cells from RA patients and arthritis mice models received from low dose MTX therapies. The effects of low dose MTX on CXCR4 were further determined via both in vitro CD3+ T cells and Cxcr4 conditional knockout (CKO) arthritis mice models. RESULTS: Our clinical data shows that low dose MTX treatment was clinically associated with down-regulated expression of chemokine receptor CXCR4 on patient peripheral T cells. In vitro, low dose MTX significantly decreased cell transmigration through down-regulated CXCR4's expression in CD3+ T cells. Consistently, CD3+ T cells treated with low dose MTX demonstrated an increased genomic hypermethylation across the promoter region of Cxcr4 gene. Furthermore, our preclinical studies showed that low dose MTX-mediated downregulation of CXCR4 significantly improved the pathological development in mouse arthritis models. Conditional disruption of the Cxcr4 gene in peripheral immune cells potentially alleviated inflammation of joints and lung tissue in the arthritis mice, though genetic modification itself overall did not change their clinical scores of arthritis, except for a significant improvement on day 45 in CXCR4 CKO arthritis mice models during the recovery phase. CONCLUSION: Our findings suggest that the effect of low dose MTX treatment could serve to eliminate inflammation in RA patients through impairment of immune cell transmigration mediated by CXCR4.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Regulación hacia Abajo , Metotrexato , Ratones Noqueados , Receptores CXCR4 , Linfocitos T , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/genética , Animales , Metotrexato/farmacología , Regulación hacia Abajo/efectos de los fármacos , Humanos , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ratones , Antirreumáticos/farmacología , Masculino , Femenino , Persona de Mediana Edad , Movimiento Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inmunología , Artritis Experimental/genética , Artritis Experimental/metabolismo , Artritis Experimental/patología
13.
Mediators Inflamm ; 2024: 6618927, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39421730

RESUMEN

Background: Baicalein has been used to treat inflammation-related diseases; nevertheless, its specific mechanism of action is unclear. Therefore, we examined the protective effects of baicalein on lipopolysaccharide-induced damage to AR42J pancreatic acinar cells (PACs) and determined its mechanism of action for protection. Methods: An in vitro cell model of acute pancreatitis (AP) was established using lipopolysaccharide (LPS) (1 mg/L)-induced PACs (AR42J), and the relative survival rate was determined using the 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide (MTT) technique. Flow cytometry was applied to evaluate the apoptotic rates of AR42J PACs. The RNA and protein expression of miR-224-5p, poly ADP-ribose polymerase-1 (PARP1), nuclear transcription factor-κB65 (NF-κB65), phospho-kappa B alpha(p-IκB-α), interleukin(IL)-18R, NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), gasdermin D (GSDMD), apoptosis-associated speck-like protein containing a CARD (ASC), and caspase-1 was detected based on the WB and RT-PCR assays. IL-1ß, IL-6, IL-18, and TNF-α expression levels in AR42J cells were measured via ELISA method. The cell morphology was examined using the AO/EB method. Results: The experiment confirmed a significant increase in the activity of AR42J cells treated with various doses of baicalein. Moreover, IL-1ß, IL-6, TNF-α, and IL-18 expression levels in AR42J cells were dramatically reduced (P < 0.05), while miR-224-5p level was obviously enhanced. The protein and gene expression of PARP1, NF-κB65, p-IκB-α, IL-18R, GSDMD, ASC, NLRP3, and caspase-1 was obviously decreased (P < 0.05). Apoptosis in AR42J cells was significantly reduced with significant improvement in cell morphology. Conclusion: Baicalein may significantly alleviate LPS-induced AR42J PAC damage by inhibiting the inflammatory response and pyroptosis. Its mode of action might be linked to higher miR-224-5p expression, which inhibits the PARP1/NF-κB and NLPR3/ASC/caspase-1/GSDMD pathways.


Asunto(s)
Flavanonas , Inflamación , Lipopolisacáridos , MicroARNs , Poli(ADP-Ribosa) Polimerasa-1 , Piroptosis , Piroptosis/efectos de los fármacos , Flavanonas/farmacología , Flavanonas/uso terapéutico , Animales , MicroARNs/metabolismo , MicroARNs/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Ratas , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Línea Celular , Apoptosis/efectos de los fármacos , Células Acinares/metabolismo , Células Acinares/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , FN-kappa B/metabolismo
14.
Neurosciences (Riyadh) ; 29(4): 276-283, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39379083

RESUMEN

OBJECTIVES: To determine the effects of all-trans-retinoic acid (ATRA) on the post-stroke inflammatory response and elucidate the underlying molecular mechanisms. METHODS: This animal experiment was conducted at Central Laboratory, the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China during 2020-2022. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1.5 h, and treated with ATRA at 2 and 24 h after reperfusion. Neurological deficit scores on behavioral tests, and cerebral infarct volume, microglial polarization, and the expression levels of inflammatory cytokines and proteins associated with TLR4/NF-κB signaling were assessed. RESULTS: The ATRA administration reduced cerebral infarct volume and ameliorated neurological deficit scores in MCAO rats. Additionally, ATRA relieved cerebral edema and downregulated the secretion of proinflammatory cytokines after stroke. Finally, ATRA attenuated the polarization of the microglia toward the M1 phenotype and promoted the activation of the beneficial M2 phenotype; the underlying mechanism potentially involved the suppression of the TLR4/NF-κB signaling pathway. CONCLUSION: The ATRA treatment promoted functional recovery in an experimental model of ischemic stroke by attenuating neural inflammation. ATRA potentially modulated microglia-mediated neuroinflammation via the downregulation of the TLR4/NF-κB signaling pathway, which makes it a candidate treatment for post-stroke neuroinflammation.


Asunto(s)
Regulación hacia Abajo , Infarto de la Arteria Cerebral Media , FN-kappa B , Fármacos Neuroprotectores , Ratas Sprague-Dawley , Transducción de Señal , Receptor Toll-Like 4 , Tretinoina , Animales , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/efectos de los fármacos , Tretinoina/farmacología , Tretinoina/uso terapéutico , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal/efectos de los fármacos , FN-kappa B/metabolismo , FN-kappa B/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Masculino , Ratas , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Microglía/metabolismo
15.
J Tradit Chin Med ; 44(5): 944-953, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39380225

RESUMEN

OBJECTIVE: To determine the molecular mechanisms underlying the neuroprotective effects of Naochuxue prescription (,NCXP) in rats with intracerebral hemorrhage (ICH). METHODS: Sprague-Dawley rats were injected with collagenase to generate ICH models, which were then randomly divided into six groups, including control, sham, model, and three intervention groups. The intervention groups received different doses of NCXP (0.13, 0.26, and 0.52 g/kg) daily for 10 d. High-performance liquid chromatography (HPLC) was used to analyze the chemical characteristics of NCXP. The neurobehavioral outcomes of the rats were evaluated using neurological deficit scores (Zea Longa 5) and the corner turn test. Pathomorphological changes in perihematomal tissues after ICH were observed using hematoxylin and eosin staining. Immunohistochemistry (IHC) was used to detect the inflammation expression of interleukin 6 (IL-6) and toll-like receptor 4 (TLR4). High mobility group box-1 (HMGB1), Beclin1, microtubule-associated protein 1 light chain 3 beta (LC3), and sequestosome 1 (p62) were detected using real-time quantitative polymerase chain reaction and Western blotting in perihematomal tissues. RESULTS: HPLC showed that the NCXP had good stability. Rats with ICH had severe neurological function deficits compared to the control group. IHC results showed that NCXP significantly downregulated the expression of the inflammatory proteins IL-6 and TLR4. ICH rats treated with NCXP showed less neurological injury than the model group, accompanied by a significantly decreased expression of HMGB1, Beclin1, and LC3 and an increased expression of p62. CONCLUSIONS: The neuroprotective effect of NCXP alleviated inflammation and autophagy possibly by downregulating HMGB1 expression. However, further research on the signaling pathways is required to verify this hypothesis.


Asunto(s)
Autofagia , Hemorragia Cerebral , Medicamentos Herbarios Chinos , Proteína HMGB1 , Fármacos Neuroprotectores , Ratas Sprague-Dawley , Animales , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/genética , Ratas , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/farmacología , Autofagia/efectos de los fármacos , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Masculino , Medicamentos Herbarios Chinos/administración & dosificación , Medicamentos Herbarios Chinos/farmacología , Humanos , Regulación hacia Abajo/efectos de los fármacos , Interleucina-6/genética , Interleucina-6/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Beclina-1/genética , Beclina-1/metabolismo
16.
FASEB J ; 38(19): e70095, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39373984

RESUMEN

Bergenin is the main active ingredient of Bergenia purpurascens, a medicinal plant which has long been used to treat a variety of Th17 cell-related diseases in China, such as allergic airway inflammation and colitis. This study aimed to uncover the underlying mechanisms by which bergenin impedes Th17 cell response in view of cellular metabolism. In vitro, bergenin treatment reduced the frequency of Th17 cells generated from naïve CD4+ T cells of mice. Mechanistically, bergenin preferentially restrained fatty acid synthesis (FAS) but not other metabolic pathways in differentiating Th17 cells, and exogenous addition of either palmitic acid (PA) or oleic acid (OA) and combination with acetyl-CoA carboxylase 1 (ACC1) activator citric acid dampened the inhibition of bergenin on Th17 cell differentiation. Bergenin inhibited FAS through downregulating the expression of SREBP1 via restriction of histone H3K27 acetylation in the SREBP1 promoter, and SREBP1 overexpression weakened the inhibition of bergenin on Th17 differentiation. Furthermore, bergenin was shown to directly interact with SIRT1 and result in activation of SIRT1. Either combination with SIRT1 inhibitor EX527 or point mutation plasmid of SIRT1 diminished the inhibitory effect of bergenin on FAS and Th17 cell differentiation. Finally, the inhibitory effect of bergenin on Th17 cell response and SIRT1 dependence were verified in mice with dextran sulfate sodium-induced colitis. In short, bergenin repressed Th17 cell response by downregulating FAS via activation of SIRT1, which might find therapeutic use in Th17 cell-related diseases.


Asunto(s)
Benzopiranos , Diferenciación Celular , Ácidos Grasos , Células Th17 , Animales , Células Th17/efectos de los fármacos , Células Th17/metabolismo , Ratones , Diferenciación Celular/efectos de los fármacos , Ácidos Grasos/metabolismo , Benzopiranos/farmacología , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis/inducido químicamente , Ratones Endogámicos C57BL , Sirtuina 1/metabolismo , Sirtuina 1/genética , Saxifragaceae/química , Regulación hacia Abajo/efectos de los fármacos , Masculino
17.
Hum Exp Toxicol ; 43: 9603271241269021, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39441175

RESUMEN

BACKGROUND: Propofol, a commonly utilized anesthetic, has been shown to induce neurotoxicity in developing neurons. A previous study showed that microRNA (miR)-138-5p was dysregulated in hippocampus tissue of mice administrated with propofol. The current study aimed to investigate the functions of miR-138-5p and its target gene in propofol-induced neurotoxicity. METHODS: SH-SY5Y neuronal cells were treated with increasing doses of propofol for indicated time to identify the optimal concentration and treatment time. MiR-138-5p and SIRT1 expression in SH-SY5Y neuronal cells stimulated with propofol were measured by RT-qPCR. Western blotting was performed to quantify protein levels of SIRT1 and autophagy markers. After interference of miR-138-5p and/or SIRT1 expression, the toxicity of SH-SY5Y neuronal cells was evaluated by cell counting kit-8 (CCK-8) assays and flow cytometry. The formation of autophagosomes was estimated by monodansylcadaverine staining. RESULTS: Propofol induced neurotoxicity in a dose- or time-dependent manner. Propofol upregulated miR-138-5p while downregulating SIRT1 in SH-SY5Y neuronal cells. The propofol-stimulated neurotoxicity and autophagy was inhibited by miR-138-5p knockdown. Moreover, miR-138-5p bound to SIRT1 3'untranslated region. SIRT1 overexpression increased cell viability while inhibiting apoptosis and autophagy in the context of propofol. SIRT1 downregulation reversed the ameliorative effect of miR-138-5p inhibition on propofol-induced neurotoxicity and autophagy. CONCLUSION: Downregulation of miR-138-5p alleviates propofol-induced neurotoxicity and autophagy via upregulation of SIRT1.


Asunto(s)
Autofagia , Regulación hacia Abajo , MicroARNs , Propofol , Sirtuina 1 , Propofol/toxicidad , Propofol/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Autofagia/efectos de los fármacos , Humanos , Regulación hacia Abajo/efectos de los fármacos , Línea Celular Tumoral , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Supervivencia Celular/efectos de los fármacos
18.
PLoS One ; 19(10): e0310293, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39480787

RESUMEN

BACKGROUND: Osteoarthritis (OA) is featured as cartilage loss, joint pain and loss of labor, which the inflammatory reaction may play critical roles. Ononin is an isoflavone isolating from medicinal plants and has anti-inflammatory effects. Our study investigated the anti-inflammation response of ononin on OA. METHODS: Anterior cruciate ligament transection (ACLT)-induced OA operation was used to establish research model, then treated with ononin for 8 weeks. The condition of joint injury was assessed using pathological staining. The concentration of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) in serum were measured by Elisa kit. The expression of collagen II and matrix metalloproteinase 13 (MMP-13) proteins to assess cartilage metabolism level by immunohistochemistry and Western blot. We detected the expression of proteins involved in the MAPK and NF-κB signaling pathways. Finally, we used molecular docking to assess the affinity of ononin for the target proteins ERK1/2, JNK1/2, p38 and p65. RESULTS: Our results confirmed that ononin ameliorated cartilage impairment through histopathological analysis by improving the morphological structures and cartilage tidal lines and decreasing Osteoarthritis Research Society International (OARSI) scores in OA rats. Moreover, ononin inhibited the secretion of above factors in OA rats. Furthermore, ononin has been shown to improve cartilage content levels in OA rats. In addition, ononin inhibited the reactivity of MAPK and NF-κB pathways in OA rats. And molecular docking indicated the ligand molecules could stably bind to the proteins of above receptors. CONCLUSION: Our results demonstrated that ononin may ameliorate cartilage damage and inflammatory response in OA rats by downgrading MAPK and NF-κB pathways, thus identifying ononin as a potential novel drug to treat OA.


Asunto(s)
FN-kappa B , Osteoartritis , Animales , Osteoartritis/metabolismo , Osteoartritis/tratamiento farmacológico , Osteoartritis/patología , Ratas , FN-kappa B/metabolismo , Masculino , Modelos Animales de Enfermedad , Ratas Sprague-Dawley , Regulación hacia Abajo/efectos de los fármacos , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Isoflavonas/química , Simulación del Acoplamiento Molecular , Transducción de Señal/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo
19.
Pestic Biochem Physiol ; 205: 106118, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39477579

RESUMEN

Etofenprox is a pyrethroid insecticide that acts on the nervous system of insects. Due to its low toxicity to aquatic animals, it is permitted for use in controlling insect pests in rice fields. The brown planthopper (BPH), Nilaparvata lugens, a significant piercing-sucking pest feeding on rice exclusively, secretes various salivary components when feeding. Salivary proteins are essential for BPH feeding, but their response to etofenprox is not well understood. The application of etofenprox down-regulated the expression of 21 salivary protein genes, among which 9 genes (NlShpa, Salivap 3, CA, NlSEF1, Nl12, NlHSC70-3, NlSP1, NlG14, and NlDNAJB9) showed significant differences. Most differentially expressed genes are found important for BPH physiological processes, except Nl12. Here we found that silencing Nl12 impeded ovary development, thereby inhibiting oocyte formation. The potential explanation was that Nl12 was highly expressed in both salivary gland and ovary, and the ovary development abnormality may be due to the direct effect from expression reduction in ovary and/or indirect influence from expression reduction in salivary gland. Altogether, our findings provide a new insight into the mechanism of action of etofenprox on insect pests and explain part of the reason why etofenprox does not stimulate reproduction in BPH.


Asunto(s)
Regulación hacia Abajo , Fertilidad , Hemípteros , Insecticidas , Piretrinas , Proteínas y Péptidos Salivales , Animales , Hemípteros/efectos de los fármacos , Hemípteros/genética , Piretrinas/farmacología , Piretrinas/toxicidad , Fertilidad/efectos de los fármacos , Insecticidas/farmacología , Insecticidas/toxicidad , Regulación hacia Abajo/efectos de los fármacos , Proteínas y Péptidos Salivales/genética , Proteínas y Péptidos Salivales/metabolismo , Femenino , Proteínas de Insectos/genética , Proteínas de Insectos/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo
20.
Int J Mol Med ; 54(6)2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39364750

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a particularly aggressive form of cancer with high mortality. In the present study, a novel 8­hydroxyquinoline derivative (91b1) was investigated for its anticancer activities in ESCC along with its associated mechanisms. The in vitro cytotoxic effect of 91b1 were evaluated across five ESCC cell lines using MTS assay, with cisplatin serving as a comparative standard. Changes in gene expression profile were identified by cDNA microarray and further validated by qualitative PCR and immunostaining. Additionally, protein levels of the most notably downregulated target in archival ESCC samples were also studied. 91b1 demonstrated comparable anticancer effect with cisplatin. Notably, chemokine ligand 5 (Ccl5) was identified as the most substantially downregulated gene, with its suppression at both mRNA and protein expression in ESCC cells, exhibiting a dose­dependent manner. The recombinant human protein of CCL5 enhanced the invasion of ESCC cells using the Transwell assay. The upregulation of CCL5 protein was also detected in 50% of ESCC cell lines. CCL5 was also overexpressed in 76.9% of ESCC specimens. The overall results indicated that 91b1 could effectively induce anticancer effect on ESCC cells through downregulating CCL5 expression with suppression of tumor invasion. Overall, these findings suggested that 91b1 effectively inhibited ESCC cell proliferation and tumor invasion by downregulating CCL5 expression, highlighting its potential as a therapeutic agent for ESCC treatment.


Asunto(s)
Quimiocina CCL5 , Regulación hacia Abajo , Neoplasias Esofágicas , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/genética , Línea Celular Tumoral , Quimiocina CCL5/metabolismo , Quimiocina CCL5/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Invasividad Neoplásica , Oxiquinolina/farmacología , Oxiquinolina/química , Oxiquinolina/análogos & derivados , Antineoplásicos/farmacología , Antineoplásicos/química , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/metabolismo , Carcinoma de Células Escamosas de Esófago/genética , Proliferación Celular/efectos de los fármacos , Masculino , Femenino , Movimiento Celular/efectos de los fármacos , Cisplatino/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...