Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
BMC Plant Biol ; 22(1): 390, 2022 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-35922748

RESUMEN

BACKGROUND: Soil salinization leads to a significant decline in crop yield and quality, including licorice, an important medicinal cash crop. Studies have proofed that the application of exogenous silicon can significantly improve the ability of licorice to resist salt stress, however, few studies concentrated on the effects of foliar silicon application on the morphology, physiological characteristics, and anatomical structure of licorice leaves under salt stress. In this study, the effects of Si (K2SiO3) on the structural and physiological characteristics of Glycyrrhiza uralensis Fisch. and G. inflata Bat. leaves under different salt concentrations (medium- and high-salt) were studied. RESULTS: Compared with the control (without salt), the plant height, total dry weight, leaf area, leaf number, relative water content, xylem area, phloem area, ratio of palisade to spongy tissue, gas exchange parameters, and photosynthetic pigment content of both licorice varieties were significantly reduced under high-salt (12S) conditions. However, the thickness of the leaf, palisade tissue, and spongy tissue increased significantly. Applying Si to the leaf surface increased the area of the vascular bundle, xylem, and parenchyma of the leaf's main vein, promoted water transportation, enhanced the relative leaf water content, and reduced the decomposition of photosynthetic pigments. These changes extended the area of photosynthesis and promoted the production and transportation of organic matter. G. uralensis had a better response to Si application than did G. inflata. CONCLUSIONS: In conclusion, foliar application of Si can improve water absorption, enhance photosynthesis, improve photosynthetic capacity and transpiration efficiency, promote growth and yield, and alleviate the adverse effects of salt stress on the leaf structure of the two kinds of licorice investigated.


Asunto(s)
Glycyrrhiza , Hojas de la Planta , Silicio , Glycyrrhiza/efectos de los fármacos , Glycyrrhiza/fisiología , Glycyrrhiza uralensis/efectos de los fármacos , Glycyrrhiza uralensis/fisiología , Hojas de la Planta/efectos de los fármacos , Hojas de la Planta/fisiología , Estrés Salino , Silicio/farmacología , Agua/metabolismo
2.
ACS Omega ; 9(20): 21727-21750, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38799345

RESUMEN

Coal seam spontaneous combustion fire is not only one of the main forms of the five major mine disasters, but also the main cause of secondary disasters such as mine gas and coal dust explosions. In recent years, with the advancement of mechanization, automation, and intelligent mine construction, spontaneous coal fires in mines have presented a series of new characteristics, and the prevention and control of spontaneous coal fires are also facing new challenges. On the basis of literature research, this paper summarizes and discusses the basic theory of coal spontaneous combustion, monitoring and early warning methods, and prevention and control technology, summarizes the development process of coal spontaneous combustion theory, reviews the research progress of coal spontaneous combustion monitoring and early warning methods and prevention and control technologies, and discusses the future development direction. In terms of the basic theory of spontaneous combustion of coal, from the initial hypothesis of spontaneous combustion of multielement coal to the unified understanding of coal-oxygen composite theory, a complete set of theoretical systems have been established, and a lot of macro and micro studies have been carried out and analyzed from multiple perspectives. In terms of coal spontaneous combustion monitoring and early warning, from the initial single indicator gas early warning to the construction of gas index system, the hierarchical early warning system is studied, and gradually tends to be perfect. With the development of automation and intelligence technology, the monitoring of coal spontaneous combustion disasters has also formed a new monitoring technology with beam tube monitoring as the traditional method, distributed optical fiber, wireless AD hoc network temperature measurement, and a coal spontaneous combustion multiparameter wireless monitoring system. In terms of fire prevention and control, the traditional "prevention" and "treatment" have changed to the "prevention-control-extinction" technical system based on hierarchical early warning, and the focus has gradually shifted to "prevention", and a large number of antifire materials have been developed, including blocking materials and fire-fighting materials. However, the precise inhibition and control of coal spontaneous combustion disasters, the evolution model of coal spontaneous combustion under the conditions of multifactor coupling in the field, the reliability and stability of intelligent monitoring system, the noncontact detection method of fire source, and the collaborative adaptation of multiple prevention and control techniques are not yet clear. In the future development, the mechanism of spontaneous coal combustion and its evolution process and other basic theories should be deeply studied. On the basis of the mechanism optimization early warning method of spontaneous coal combustion process, flame retardant and fire prevention materials should be targeted and developed. On the basis of the spatiotemporal evolution of spontaneous coal combustion, monitoring and monitoring system equipment with high speed, high precision, and high stability should be developed, which should accelerate the realization of accurate dynamic sensing and intelligent early warning of coal spontaneous combustion, and form an active hierarchical collaborative prevention and control system based on the trinity of "prevention-control-extinction" of coal spontaneous combustion. The conclusions and prospects of this paper can be used for reference in the future research direction, and have a certain role in promoting the exchange of research results of coal science and technology workers.

3.
Sci Rep ; 14(1): 2367, 2024 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-38287125

RESUMEN

Multiple primary cancer (MPC) denotes individuals with two or more malignant tumors occurring simultaneously or successively. Herein, a total of 11,000 pancancer patients in TCGA database (1993-2013) were divided into MPC or non-MPC groups based on their history of other malignant tumors. The incidence of MPC has risen to 8.5-13.1% since 2000. Elderly individuals, males, early-stage cancer patients, and African Americans and Caucasians are identified as independent risk factors (p < 0.0001). Non-MPC patients exhibit significantly longer overall survival (OS) and disease-free survival (DFS) (p = 0.0038 and p = 0.0014). Age (p < 0.001) and tumor staging at initial diagnosis (p < 0.001) contribute to this difference. In our center, MPC was identified in 380 out of 801 tumor events based on SEER criteria. The peak occurrence of secondary primary was about 1-5 years after the first primary tumor, with a second small peak around 10-15 years. Multiple tumors commonly occur in the same organ (e.g., breast and lung), constituting 12.6%. Certain cancer types, notably skin cutaneous melanoma (SKCM), exhibit significantly higher tumor mutational burden (TMB) in the MPC group (17.31 vs. 6.55 mutations/MB, p < 0.001), with high TMB associated with improved survival (p < 0.001). High TMB in MPC may serve as a predictor for potential immunotherapy application.


Asunto(s)
Melanoma , Neoplasias Primarias Múltiples , Neoplasias Cutáneas , Masculino , Humanos , Anciano , Melanoma/patología , Neoplasias Cutáneas/patología , Estadificación de Neoplasias , Genómica , Neoplasias Primarias Múltiples/epidemiología , Mutación , Biomarcadores de Tumor
4.
Environ Sci Pollut Res Int ; 30(3): 6435-6453, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35997881

RESUMEN

LongWangGou coal mine auxiliary inclined shaft heading face has a large section, fast heading speed, and large dust production. The problems of high concentration and wide distribution of dust in the roadway air are very prominent. To improve the efficiency of roadway ventilation and dust removal, long-compression and short suction (after this referred to as LCSS) are adopted. The dust concentration and airflow field distribution in the roadway are numerically simulated by FLUENT. The change of the position between the pressure duct and the exhaust duct and the different pressure-extraction ratios affect the distribution of airflow velocity, the formation of vortex zone, and the distribution of dust concentration in the roadway. When Ly=25 m, Lc=5.0 m, W=1.2, a wind curtain is formed at 5 m in front of the heading face to prevent dust from spreading deeper into the roadway.


Asunto(s)
Minas de Carbón , Succión , Polvo/análisis , Simulación por Computador , Carbón Mineral/análisis
5.
J Gastrointest Oncol ; 14(6): 2617-2626, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38196522

RESUMEN

Background: There are limited treatment options available for patients with metastatic colorectal cancer (mCRC). About 95% of CRC patients have mismatch repair proficient/microsatellite stable (pMMR/MSS) tumors are virtually unresponsive to programmed cell death protein 1 (PD-1) antibody treatment. This report shows that a patient with pMMR/MSS mCRC achieved significant response and the longest progression-free survival (PFS) of 28 months currently reported from tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor receptor (VEGFR) family (VEGFR-1,2,3) (fruquintinib) plus anti-PD-1 immunotherapy in the third line, providing a new and promising treatment option for some MSS mCRC patients. Case Description: This case details a 65-year-old male with CRC who was diagnosed with pT4aN2bM0, IIIC, and pMMR/MSS after curative surgery in August 2018. Subsequently, he received adjuvant chemotherapy [FOLFOX (folinic acid, fluorouracil, and oxaliplatin) for 5 cycles], first-line treatment (pelvic radiation plus capecitabine), and second-line treatment [TOMIRI (raltitrexed and irinotecan) plus cetuximab for 2 cycles]. Lung, liver, and pelvic cavity metastases worsened in October 2019. He began receiving the fruquintinib plus PD-1 inhibitor (FP) regimen as third-line treatment and after 3 cycles, the size of the lung lesions was significantly reduced and evaluated as partial response (PR), whereas the liver and pelvic cavity lesions remained stable. As of December 2021, he had received a total of 33 courses of FP regimen. In February 2022, liver metastases progressed. In brief, he achieved a long PFS of 28 months and an overall survival (OS) of 40 months from the third-line treatment. Additionally, the patient only experienced mild proteinuria after the combined treatment and tolerated well. Conclusions: Fruquintinib combined with immunotherapy could exert good therapeutic effects with safety in MSS mCRC patients. And patients with lung metastasis may be the principal beneficiaries.

6.
Sci Rep ; 12(1): 5089, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35332196

RESUMEN

Silicon (Si) effectively alleviates the effects of salt stress in plants and can enhance salt tolerance in liquorice. However, the mechanisms by which Si improved salt tolerance in liquorice and the effects of foliar application of Si on different liquorice species under salt stress are not fully understood. We investigated the effects of foliar application of Si on the growth, physiological and biochemical characteristics, and ion balance of two liquorice species, Glycyrrhiza uralensis and G. inflata. High salt stress resulted in the accumulation of a large amount of Na+, decreased photosynthetic pigment concentrations, perturbed ion homeostasis, and eventually inhibited both liquorice species growth. These effects were more pronounced in G. uralensis, as G. inflata is more salt tolerant than G. uralensis. Foliar application of Si effectively reduced the decomposition of photosynthetic pigments and improved gas exchange parameters, thereby promoting photosynthesis. It also effectively inhibited lipid peroxidation and leaf electrolyte leakage and enhanced osmotic adjustment of the plants. Furthermore, Si application increased the K+ concentration and reduced Na+ absorption, transport, and accumulation in the plants. The protective effects of Si were more pronounced in G. uralensis than in G. inflata. In conclusion, Si reduces Na+ absorption, improves ion balance, and alleviates the negative effects of salt stress in the two liquorice species studied, but the effect is species dependent. These findings may help to develop novel strategies for protecting liquorice plants against salt stress and provide a theoretical basis for the evaluation of salt tolerance and the scientific cultivation of liquorice.


Asunto(s)
Glycyrrhiza , Silicio , Homeostasis , Iones/farmacología , Hojas de la Planta , Estrés Salino , Tolerancia a la Sal , Silicio/farmacología , Sodio/farmacología
7.
PeerJ ; 10: e12928, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35190786

RESUMEN

Long-term and widespread cotton production in Xinjiang, China, has resulted in significant soil degradation, thereby leading to continuous cropping obstacles; cotton stalk biochar (CSB) addition may be an effective countermeasure to this issue, with effects that are felt immediately by root systems in direct contact with the soil. In this study, we assess the effects of different CSB application rates on soil nutrient contents, root morphology, and root physiology in two soil types commonly used for cotton production in the region. Compared with CK (no CSB addition), a 1% CSB addition increased total nitrogen (TN), available phosphorus (AP), and organic matter (OM) by 13.3%, 7.2%, and 50% in grey desert soil, respectively , and 36.5%, 19.9%, and 176.4%, respectively, in aeolian sandy soil. A 3% CSB addition increased TN, AP, and OM by 38.8%, 23.8%, and 208.1%, respectively, in grey desert soil, and 36%, 13%, and 183.2%, respectively, in aeolian sandy soil. Compared with the aeolian sandy soil, a 1% CSB addition increased TN, OM, and AP by 95%, 94.8%, and 33.3%, respectively, in the grey desert soil , while in the same soil 3% CSB addition increased TN, OM, and AP by 108%, 21.1%, and 73.9%, respectively. In the grey desert soil, compared with CK, a 1% CSB application increased the root length (RL) (34%), specific root length (SRL) (27.9%), and root volume (RV) (32.6%) during the bud stage, increased glutamine synthetase (GS) (13.9%) and nitrate reductase (NR) activities (237%), decreased the RV (34%) and average root diameter (ARD) (36.2%) during the harvesting stage. A 3% CSB addition increased the RL (44%), SRL (20%), and RV (41.2%) during the bud stage and decreased the RV (29%) and ARD (27%) during the harvesting stage. In the aeolian sandy soil, 1% CSB increased the RL (38.3%), SRL (73.7%), and RV (17%), while a 3% caused a greater increase in the RL (55%), SRL (89%), RV (28%), soluble sugar content (128%), and underground biomass (33.8%). Compared with the grey desert soil, a 1% CSB addition increased the RL (48.6%), SRL (58%), and RV (18.6%) in the aeolian sandy soil, while a 3% further increased the RL (54.8%), SRL (84.2%), RV (21.9%), and soluble sugar content (233%). The mechanisms by which CSB addition improves the two soils differ: root morphology changed from coarse and short to fine and long in the grey desert soil, and from fine and long to longer in the aeolian sandy soil. Overall, a 3% CSB addition may be a promising and sustainable strategy for maintaining cotton productivity in aeolian sandy soil in the Xinjiang region.


Asunto(s)
Carbón Orgánico , Suelo , Biomasa , Arena , China , Nitrógeno/análisis , Fósforo/análisis
8.
Oncoimmunology ; 11(1): 2131084, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36268178

RESUMEN

Myeloid-derived suppressor cells (MDSCs) have been demonstrated to suppress antitumor immunity and induce resistance to PD-1/PD-L1 blockade immunotherapy in gastric and colon cancer patients. Herein, we found that MDSCs accumulate in mice bearing syngeneic gastric cancer and colon cancer. Death receptor 5 (DR5), a receptor of TNF-related apoptosis-inducing ligand (TRAIL), was highly expressed on MDSCs and cancer cells; targeting DR5 using agonistic anti-DR5 antibody (MD5-1) specifically depleted MDSCs and induced enrichment of CD8+ T lymphocytes in tumors and exhibited stronger tumor inhibition efficacy in immune-competent mice than in T-cell-deficient nude mice. Importantly, the combination of MD5-1 and anti-PD-L1 antibody showed synergistic antitumor effects in gastric and colon tumor-bearing mice, resulting in significantly suppressed tumor growth and extended mice survival, whereas single-agent treatment had limited effect. Moreover, the combination therapy induced sustained memory immunity in mice that exhibited complete tumor regression. The enhanced antitumor effect was associated with increased intratumoral CD8+ T-cell infiltration and activation, and a more vigorous tumor-inhibiting microenvironment. In summary, our findings highlight the therapeutic potential of combining PD-L1 blockade therapy with agonistic anti-DR5 antibody that targets MDSCs in gastric and colon cancers.


Asunto(s)
Neoplasias del Colon , Células Supresoras de Origen Mieloide , Ratones , Animales , Células Supresoras de Origen Mieloide/patología , Receptor de Muerte Celular Programada 1 , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ratones Desnudos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Microambiente Tumoral , Neoplasias del Colon/tratamiento farmacológico
9.
Front Oncol ; 12: 841977, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371995

RESUMEN

Background: Programmed death receptor-1 (PD-1) blockade shows little benefit in patients with microsatellite-stable colorectal cancer (MSS-CRC). Fruquintinib is a China-made anti-angiogenic drug which is approved for the third line therapy in mCRC. This study investigates the effect of the combination of fruquintinib and PD-1 blockade on MSS-CRC and its relative mechanisms. Methods: The mouse allograft tumor models that represent MSS and microsatellite instability (MSI) CRC were established using murine CT26 and MC38 colon cancer cells, respectively, to assess the treatment efficacy. The percentages of immune cells were detected in the peripheral blood, spleen and tumor tissues in the tumor-bearing mice by flow cytometry analysis. Angiogenesis in tumor tissues was detected by immunofluorescence. The safety of drug treatment was evaluated by histopathology analysis in murine main organs. The efficacy of the combination of fruquintinib and sintilimab were verified in the treatment of MSS-CRC patients. Results: Our results showed that the combination of fruquintinib and sintilimab exhibited the strongest inhibition of tumor growth and achieved the longest survival time in mice bearing MC38 or CT26 xenograft tumors, compared to fruquintinib and sintilimab alone. Mechanistically, the combination of fruquintinib and sintilimab reduced angiogenesis, reprogramed the vascular structure, enhanced the infiltration of CD8+T cells (p<0.05), CD8+TNFα+ (p<0.05) T cells and CD8+IFNγ+ (p<0.05) T cells and reduced the ratios of MDSCs and macrophages in mice. There was no obvious toxicity observed in the main organs of the tumor-bearing mice with the combined treatment. Moreover, the treatment using the combination of fruquintinib and sintilimab achieved effective response in five patients with refractory advanced MSS CRC. Conclusion: Our results show that the combination of fruquintinib and sintilimab greatly inhibits CRC growth by altering tumor immune microenvironment. This study provides the rational for using the combination of fruquintinib and anti-PD-1 antibody for the treatment of advanced CRC.

10.
J Oncol ; 2022: 4233782, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35368895

RESUMEN

Background: Human epidermal growth factor receptor 2 (HER2) is a member of the large ErbB family and an important oncogene in many solid tumors. Pyrotinib has been approved for the treatment of HER2-positive, recurrent, or metastatic breast cancer. However, there are very few clinical studies on pyrotinib in other HER2-positive solid tumors. Therefore, more evidence of clinical research is impendently needed to shepherd pyrotinib-based therapy in HER2-positive nonbreast advanced solid tumors. Patients and Methods. We performed a retrospective analysis of HER2-positive nonbreast advanced solid tumors patients with HER2 amplification or mutations who were administered with pyrotinib-based therapy in Henan Cancer Hospital between July 1, 2019, and December 2, 2021. In our research, 25 eligible patients were included with 16 patients with lung cancer, 6 patients with gastric cancer, 2 patients with colorectal cancer, and 1 patient with cholangiocarcinoma. Progression-free survival (PFS) is our main research end point. Results: The median PFS was 188 days (95% CI: 83-not reached (NR)), and overall survival (OS) was 250 days (95% CI: 188-NR), respectively. 16 patients with lung cancer had a median PFS of 204 days (95% CI: 55-NR) and 6 patients with gastric cancer had PFS of 142 days (95% CI: 83-NR), respectively. The median OS was 366 days (95% CI: 248-NR) in patients with lung cancer and 179 days (95% CI: 90-NR) in patients with gastric cancer. The median PFS and OS of patients receiving >3 line treatment were lower than those receiving ≤3 line treatment (PFS: 188 days vs 204 days, p = 0.92; OS: 188 days vs 366 days, p = 0.43). All 25 patients can be evaluated. The objective response rate (ORR) was 24%, and the disease control rate (DCR) was 68%. Lung cancer ORR was 25%, and gastric cancer ORR was 16.7%. In addition, the DCR of lung cancer was 62.5% and that of gastric cancer was 66.7%. In addition, the ORR and DCR of patients receiving treatment ≤3 lines were higher than those receiving treatment >3 lines (ORR: 35.7% vs 9.1%, p = 0.18; DCR: 71.4% vs 63.6%, p > 0.99). The most common treatment-related adverse events (TRAEs) were diarrhea (84%), but only 3 patients (12%) reported grade 3 diarrhea with good control. Conclusion: These results show that in HER2-positive nonbreast advanced solid tumors, the treatment based on pyrotinib regimen has good antitumor activity and acceptable safety. This retrospective study aims to promote larger clinical studies to further clarify the efficacy and safety of pyrotinib in the treatment of nonbreast solid tumors.

11.
Front Oncol ; 12: 924149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35719979

RESUMEN

Background: Unresectable advanced or recurrent gastric cancer patients have a poor prognosis. PD-1 monotherapy regimen and PD-1 combined chemotherapy regimen have become the standard third- and first-line treatment for advanced gastric cancer, respectively. However, the status of immune checkpoint inhibitors in the second-line treatment for advanced gastric cancer has not been established. The combination of chemotherapy and anti-PD-1 antibody has been demonstrated to have a synergistic effect. In this study, we aimed to evaluate the efficacy and safety of sintilimab combined with nab-paclitaxel in the second-line treatment for advanced gastric cancer (GC)/gastroesophageal junction (GEJ) cancer patients. Patients and Methods: We retrospectively analyzed patients with advanced GC/GEJ cancer that progressed after first-line systemic therapies with sintilimab combined with nab-paclitaxel from April 1, 2019 to December 31, 2021. The primary endpoint was progression-free survival (PFS). The secondary endpoints included objective response rate (ORR), disease control rate (DCR), and safety. Results: Thirty-nine patients were enrolled and eligible for response assessment. Complete response (CR) was not observed, 15 patients achieved partial response (PR), 16 patients had stable disease (SD) and 9 patients had progressive disease (PD). The ORR and DCR were 15 (38.5%) and 31 (79.5%), respectively. Median PFS was 5.4 months (95%CI: 3.072-7.728). PFSs between different subgroups were analyzed. The results showed that gender, age, Human epidermal growth factor receptors 2 (HER2) status, PD-L1 expression, primary tumor site and chemotherapy cycles had no significant effect on PFS. Most of the adverse events (AEs) were of grade 1-2 and manageable. The common treatment-related adverse events of grade 3 or 4 included anemia (12.8%), neutropenia (12.8%), leukopenia (10.3%), hand-foot syndrome (7.7%), thrombocytopenia (7.7%). The potential immune-related adverse events (irAEs) were grade 1 pneumonia (1 pts [2.6%]) and grade 4 hepatitis (1 pts [2.6%]). There were no treatment-related deaths. Conclusion: These results indicate that sintilimab combined with nab-paclitaxel exhibits good anti-tumor activity and an acceptable safety profile as a second-line treatment for advanced or metastatic gastric cancer. These results warrant further investigation and evaluation to identify patients who can benefit more from the combined treatment strategy.

12.
Cancer Lett ; 521: 119-129, 2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34464671

RESUMEN

Immune checkpoint inhibitors (ICIs), as a subverter of immunotherapy in oncology, are changing all aspects of therapy for malignant tumors, especially their remarkable effects on melanoma and non-small cell lung cancer (NSCLC). For colorectal cancer (CRC), only a small number of patients with high immunogenicity (microsatellite instability-high/mismatch-repair deficient (MSI-H/dMMR)) benefit greatly from ICIs treatment, and most CRC patients with low immunogenicity (microsatellite instability-low/mismatch-repair proficient (MSI-L/pMMR)) do not. Currently, numerous clinical trials are ongoing to improve CRC patients' response to ICIs immunotherapy through better patient selection and novel combination strategies. Thus, this review discusses the current status and latest progress of ICIs treatment in CRC. We expect that these studies can change the pattern of CRC immunotherapy in the future.

13.
Cell Death Dis ; 12(4): 338, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33795638

RESUMEN

Utidelone (UTD1), a novel microtubule stabilizing agent, is an epothilone B analogue which was produced by genetic engineering. UTD1 has exhibited broad antitumor activity in multiple solid tumors. However, its activity and mechanism in colorectal cancer (CRC) remain to be studied. In this study, UTD1 dramatically inhibited CRC cell proliferation (with 0.38 µg/ml, 0.77 µg/ml IC50 in RKO and HCT116, respectively) in vitro. Immunofluorescence staining showed that UTD1 induced the formation of microtubule bundling and asters in RKO cells. Flow cytometry analysis demonstrated that UTD1 induced cell cycle to arrest in G2/M phase, subsequent apoptosis. Significantly, UTD1 exhibited stronger effect on inducing apoptosis than paclitaxel and 5-FU, especially in HCT15 cells which is ABCB1 high-expression. UTD1 exposure cleaved caspase-3 and poly ADP-ribose polymerase (PARP), decreased mitochondrial membrane potential, released cytochrome c, increased the production of active oxygen and activated c-Jun N-terminal kinase (JNK), suggesting ROS/JNK pathway was involved in this process. Moreover, UTD1 inhibited tumor growth and was more effective and safer compared with paclitaxel and 5-FU in RKO xenograft in nude mice. Taken together, our findings first indicate that UDT1 inhibits tumor growth in CRC xenograft model and may be a promising agent for CRC treatment.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Epotilonas/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Desnudos , Paclitaxel/farmacología
14.
Front Immunol ; 12: 619209, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790893

RESUMEN

Cancer-associated fibroblasts (CAFs) has been recognized as one cause of tumor resistance to immune checkpoint blockade therapy, but the underlying mechanisms still remain elusive. In the present study, a bone marrow-derived CAF (BMF) -rich tumor model is successfully established by subcutaneously mixed inoculation of BMFs and tumor cells into mice and the BMF-mixed tumor xenografts are demonstrated to be resistant to anti-PD-L1 antibody immunotherapy compared to the mere tumor xenografts. In vitro assays via the co-culture system of BMFs and tumor cells indicate that the co-cultured BMFs are induced to overexpress PD-L1, while there is no such a phenomenon in the co-cultured cancer cells. The further knock-out of PD-L1 in BMFs rescues the sensitivity of BMF-mixed tumor xenografts to PD-L1 blockade therapy. Mechanistically, via the microarray assay, we identify that the upregulation of PD-L1 in BMFs stimulated by cancer cells is medicated by the activation of the Wnt/ß-catenin signaling pathway in BMFs. Moreover, the administration of Wnt/ß-catenin signaling inhibitors, including XAV-939 and Wnt-C59, distinctly inhibits the upregulation of PD-L1 expression in the co-cultured BMFs. The further combination administration of XAV-939 significantly potentiates the therapeutic outcome of PD-L1 blockade therapy in BMF-mixed tumors. In summary, our study demonstrates that Wnt inhibition augments PD-L1 blockade efficacy by overcoming BMF-mediated immunotherapy resistance.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Fibroblastos Asociados al Cáncer/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Puntos de Control Inmunológico/farmacología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Animales , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Expresión Génica , Genes Reporteros , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunohistoquímica , Ratones , Miofibroblastos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Onco Targets Ther ; 13: 10567-10580, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33116635

RESUMEN

BACKGROUND: Murine bone marrow-derived myofibroblasts (BMFs) have previously been shown to promote gastric cancer growth. However, whether BMFs promote gastric cancer cell metastasis remains largely unknown. METHODS: Wound healing assay, Transwell invasion and migration assay and 3D organotypic co-culture systems were conducted to study the effects of BMFs on invasion and migration of gastric cancer cells and the invasion and migration ability of gastric cancer stem cell-like cells (CSC-LCs) induced by BMFs. We employed two animal model to study the role of BMFs on the in vivo metastasis of gastric cancer cells and the metastatic ability of gastric BMF-induced CSC-LCs. A human gastric cancer tissue microarray and TCGA gastric cancer database were analysed to study the relationship between the expression of IL-6 and TGF-ß1 and clinicopathological characteristics and survival in gastric cancer. RESULTS: We found that BMFs promoted the in vitro migration and invasion of gastric cancer cells. BMFs promoted liver, lung, subcutaneous, and splenic metastases of MKN28 cells in the spleen injection liver metastasis model and co-injection of caudal vein (IOCV) mouse model. BMFs reprogrammed non-gastric cancer stem cell (CSC) to CSC-LCs and enhanced CSC-LC migration and metastasis. BMF-derived IL-6 and gastric cancer cell-secreted TGF-ß1 mediated the interaction between BMFs and gastric cancer cells, promoting tumour metastasis. BMFs enhanced the expressions of STAT3 and p-STAT3 in co-cultured gastric cancer cells. A combination of Napabucasin and Galunisertib exhibited the strongest inhibition of cell migration compared to when administered alone. Gastric cancer tissue array and TCGA database indicated that the overexpression of IL-6 and TGF-ß1 was associated with gastric cancer metastasis. CONCLUSION: Our results demonstrated that BMFs promote gastric cancer metastasis through the activation of the TGF-ß1 and IL-6/STAT3 signalling pathways. Targeting the inhibition of these interactions may be a potent therapeutic strategy for addressing gastric cancer metastasis.

16.
Sci Rep ; 7(1): 8660, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28819126

RESUMEN

To investigate the role of TGF-ß and IL-6 in myofibroblasts (MFs) - lung cancer cell interactions, lung cancer cells (Lewis and CTM-167 cell lines) were stimulated by IL-6, MF-conditioned medium (MF-CM) or MFs, with or without TGF-ß signaling inhibitor - SB431542 and/or JAK2/STAT3 inhibitor - JSI-124. MFs were stimulated by TGF-ß, cancer cell-CM or cancer cells, with or without SB431542 and JSI-124. Cell proliferation, the levels of cytokines, expression of mRNA and protein were determined. Mice bearing xenograft tumors were intraperitoneally treated with SB431542 or JSI-124 and monitored for up to 45 days. In co-culture systems, MFs secreted high levels of IL-6, while cancer cells produced high levels of TGF-ß. Recombinant IL-6 and MF-CM activated STAT3 and upregulated TGF-ß in cancer cells. In contrast, cancer cell-CM or TGF-ß stimulated MFs to produce IL-6. Blockade of JAK2/STAT3 and TGF-ß signaling by specific inhibitors significantly inhibited cell proliferation in vitro and tumor growth in vivo of lung cancer cells. Our study demontrated that the TGF-ß and IL-6/JAK2/STAT3 signaling pathways form a positive feedback signaling loop that mediated the interactions between MFs and lung cancer cells. Targeted inhibiton of this signaling loop could be a new approach for lung cancer prevention and therapy.


Asunto(s)
Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antineoplásicos/farmacología , Biomarcadores , Línea Celular Tumoral , Proliferación Celular , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Terapia Molecular Dirigida , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncogene ; 35(41): 5388-5399, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27109105

RESUMEN

Bone marrow-derived cells have important roles in cancer development and progression. Our previous studies demonstrated that murine bone marrow-derived myofibroblasts (BMFs) enhanced tumor growth. In this study, we investigated the mechanisms of BMF actions. We found that co-injection of BMFs with gastric cancer cells markedly promoted tumorigenesis. Co-cultured BMFs or BMF-conditioned medium (BMF-CM) induced the formation of spheres, which expressed stem cell signatures and exhibited features of self-renewal, epithelial-to-mesenchymal transition and tumor initiation. Furthermore, CD44+ fractions in spheres were able to initiate tumorigenesis and re-establish tumors in serially passaged xenografts. In co-culture systems, BMFs secreted high levels of murine interleukin-6 (IL-6) and hepatocyte growth factor (HGF), whereas cancer cells produced high level of transformation growth factor-ß1 (TGF-ß1). BMF-CM and IL-6 activated BMFs to produce mHGF, which activated signal transducer and activator of transcription 3 (STAT3) and upregulated TGF-ß1 in human cancer cells. In return, cancer cell-CM stimulated BMFs to produce IL-6, which was inhibited by anti-TGF-ß1 neutralizing antibody. Blockade of HGF/Met, Janus kinase 2 (JAK2)/STAT3 and TGF-ß1 signaling by specific inhibitors inhibited BMF-induced sphere formation. STAT3 knockdown in cancer cells also inhibited BMF-induced sphere formation and tumorigenesis. Moreover, TGF-ß1 overexpression in cancer cells was co-related with IL-6 and HGF overexpression in stromal cells in human gastric cancer tissues. Our results show that BMF-derived IL-6/HGF and cancer cell-derived TGF-ß1 mediate the interactions between BMFs and gastric cancer cells, which regulate cancer stemness and promote tumorigenesis. Targeting inhibition of the interactions between BMFs and cancer cells may be a new strategy for cancer therapy.


Asunto(s)
Carcinogénesis/genética , Factor de Crecimiento de Hepatocito/genética , Interleucina-6/genética , Neoplasias Gástricas/genética , Factor de Crecimiento Transformador beta1/genética , Animales , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Línea Celular Tumoral , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Transición Epitelial-Mesenquimal/genética , Humanos , Janus Quinasa 2/genética , Ratones , Miofibroblastos/metabolismo , Miofibroblastos/patología , Células Madre Neoplásicas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
18.
Cancer Lett ; 343(1): 80-9, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24145153

RESUMEN

Bone marrow-derived myofibroblasts (BMFs) have been shown to promote tumor growth. Here, we found that BMFs or BMF conditioned medium (BMF-CM) induced cancer stem cell-like sphere formation of colon cancer cells. The co-cultured BMFs, but not co-cultured cancer cells, expressed higher levels of IL-6 than BMFs or cancer cells cultured alone. Anti-mouse IL-6 neutralizing antibody, JAK2 inhibitors and STAT3 knockdown in mouse cancer cells reduced BMF- and BMF-CM-induced sphere formation of colon cancer cells. When co-injected, BMFs significantly enhanced tumorigenesis of colon cancer cells in mice. Our results demonstrate that BMFs promote tumorigenesis via the activation of the IL-6/JAK2/STAT3 pathway.


Asunto(s)
Células de la Médula Ósea/citología , Neoplasias del Colon/patología , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Miofibroblastos/citología , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica , Técnicas de Cocultivo , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA