Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 176: 116844, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823279

ABSTRACT

In contemporary times, tumors have emerged as the primary cause of mortality in the global population. Ongoing research has shed light on the significance of neurotransmitters in the regulation of tumors. It has been established that neurotransmitters play a pivotal role in tumor cell angiogenesis by triggering the transformation of stromal cells into tumor cells, modulating receptors on tumor stem cells, and even inducing immunosuppression. These actions ultimately foster the proliferation and metastasis of tumor cells. Several major neurotransmitters have been found to exert modulatory effects on tumor cells, including the ability to restrict emergency hematopoiesis and bind to receptors on the postsynaptic membrane, thereby inhibiting malignant progression. The abnormal secretion of neurotransmitters is closely associated with tumor progression, suggesting that focusing on neurotransmitters may yield unexpected breakthroughs in tumor therapy. This article presents an analysis and outlook on the potential of targeting neurotransmitters in tumor therapy.


Subject(s)
Disease Progression , Neoplasms , Neurotransmitter Agents , Humans , Neurotransmitter Agents/metabolism , Neoplasms/pathology , Neoplasms/metabolism , Animals , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism
2.
Front Pharmacol ; 13: 1000377, 2022.
Article in English | MEDLINE | ID: mdl-36408240

ABSTRACT

Sodium cantharidate (SCA) is a derivative of cantharidin obtained by its reaction with alkali. Studies have shown that it inhibits the occurrence and progression of several cancers. However, therapeutic effects of SCA on breast cancer are less well studied. This study aimed to clarify the effect of SCA on breast cancer cells and its mechanism, and to provide a scientific basis for the clinical use of SCA for the treatment of breast cancer. The results of cell counting kit-8, colony formation assay, and 5-ethynyl-2'-deoxyuridine staining showed that SCA inhibited breast cancer cell proliferation. Wound-healing and transwell assays demonstrated that SCA inhibited the migration and invasion of breast cancer cells. Transmission electron microscopy revealed that SCA induced autophagy in breast cancer cells. RNA sequencing technology showed that SCA significantly regulated the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin (PI3K-Akt-mTOR) pathway, which was further verified using western blotting. The inducing effect of SCA on breast cancer autophagy was reversed by the mTOR activator MHY1485. In addition, subcutaneous xenograft experiments confirmed that SCA significantly inhibited tumor growth in vivo. Hematoxylin-eosin, TdT-mediated dUTP nick-end labeling, and immunohistochemical staining indicated that SCA induced tumor cell autophagy and apoptosis in nude mice without causing organ damage. In summary, we found that SCA promoted breast cancer cell apoptosis by inhibiting the PI3K-Akt-mTOR pathway and inducing autophagy.

3.
Article in English | MEDLINE | ID: mdl-36043773

ABSTRACT

The article has been withdrawn at the request of the authors of the journal Combinatorial Chemistry & High Throughput Screening.Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php. BENTHAM SCIENCE DISCLAIMER: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

4.
Phytomedicine ; 104: 154319, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35853302

ABSTRACT

BACKGROUND: Lung cancer has the highest mortality rate among all cancer types. In combination with multiple chemotherapeutic options, traditional Chinese medicine has proven indispensable for the comprehensive treatment of lung cancer. PURPOSE: To investigate the effects of Hedyotis diffusa on lung adenocarcinoma cell lines and a BALB/c nude mouse xenograft model, and determine whether HDI could induce ferroptosis in lung adenocarcinoma cells along with the underlying mechanism. METHODS: The anti-tumor activity of HDI was determined in vitro by cell counting kit-8, clonogenic, and transwell assays. Subsequently, electron microscopy, a lipid reactive oxygen species assay, ferrous ion staining, and a malondialdehyde assay were performed to determine the effect on ferroptosis in lung adenocarcinoma cells. The mechanism was then further investigated using small molecule inhibitors, siRNA, and plasmid overexpression in vitro. Finally, the effects of HDI were assessed in tumor-bearing BALB/c nude mice, and HE staining was performed to observe tissue damage after HDI treatment. RESULTS: In vitro experiments showed that HDI could inhibit the viability of lung adenocarcinoma cells and induce lung adenocarcinoma cells ferroptosis via mechanisms independent of GPX4 and PUFA-PLS pathways but closely associated with VDAC2/3. HDI regulated VDAC2/3 activity by promoting Bax via inhibiting Bcl2, thereby inducing ferroptosis in lung adenocarcinoma cells. Furthermore, in vivo experiments showed that HDI significantly inhibited the growth of subcutaneous tumors in BALB/c nude mice with less organ damage and toxicity, and significantly increased the expression of the ferroptosis-related indicators 4HNE, TFR, and HMOX1 in tumor tissue. CONCLUSION: HDI can significantly reduce the survival of lung adenocarcinoma cells in vitro, inhibit the growth of subcutaneously transplanted tumors in BALB/c nude mice in vivo, and induce ferroptosis in lung adenocarcinoma cells via Bcl2 inhibition to promote Bax regulation of VDAC2/3.


Subject(s)
Adenocarcinoma of Lung , Ferroptosis , Hedyotis , Lung Neoplasms , Adenocarcinoma of Lung/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/drug therapy , Mice , Mice, Nude , Mitochondrial Membrane Transport Proteins , Proto-Oncogene Proteins c-bcl-2 , Voltage-Dependent Anion Channel 2 , Voltage-Dependent Anion Channels , bcl-2-Associated X Protein
5.
Biosci Biotechnol Biochem ; 86(3): 305-312, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-34935885

ABSTRACT

Cerebral ischemia reperfusion (CIR) has become the leading cause of death and disability. Baicalein is a natural bioactive ingredient extracted from Scutellaria baicalensis Georgi and has neuroprotective activity. In our work, baicalein was found to reduce neurological deficits, brain water content, infarct area, and neuronal death of rats induced by middle cerebral artery occlusion/reperfusion. In vitro, oxygen-glucose deprivation/reperfusion induced inordinate ROS production and apoptosis that could be reversed by baicalein. Our study revealed for the first time that baicalein has the potential to bind and inhibit the activity of calpain 1, thereby inhibiting AIF nuclear translocation. These findings demonstrated that baicalein protected against CIR injury via inhibiting AIF nuclear translocation by inhibiting calpain 1 activity.


Subject(s)
Flavanones
6.
Front Pharmacol ; 12: 750031, 2021.
Article in English | MEDLINE | ID: mdl-34630120

ABSTRACT

Roughly one third of non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI)-sensitive mutated (EGFRm) tumors experience disease progression through central nervous system (CNS) metastases during treatment. Although EGFR-TKIs have been reported to be favored in some patients with EGFRm NSCLC CNS metastases, novel EGFR-TKIs with proven efficacy in CNS pathologies are clinically needed.To investigate whether almonertinib, a novel third-generation EGFR-TKI for NSCLC, can cross the blood-brain barrier (BBB) and deliver treatment for EGFR-mutant NSCLC brain metastases and spinal cord metastases, we constructed NSCLC brain metastasis and spinal cord metastasis models in vivo to observe the anti-tumor effects of almonertinib. Using ABCB1-MDCK and BCRP-MDCK monolayer cells as the in vitro study model, the effects of transport time and drug concentration on the apparent permeability coefficient of almonertinib and its active metabolite, HAS-719, were investigated. The results of this study show that almonertinib can significantly inhibit PC9 brain and spinal cord metastases. Pharmacokinetic studies in mice revealed that almonertinib has good BBB penetration ability, whereas the metabolite HAS-719 does not easily penetrate the BBB. Early clinical evidence of almonertinib activity in patients with EGFRm-advanced NSCLC and brain metastases has also been reported. In conclusion, almonertinib easily penetrates the BBB and inhibits advanced NSCLC brain and spinal cord metastases.

7.
Toxicol Appl Pharmacol ; 430: 115726, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34537213

ABSTRACT

Breast cancer is the leading cause of cancer-related death in women worldwide, and despite multiple chemotherapeutic approaches, effective treatment strategies for advanced metastatic breast cancer are still lacking. Metabolic reprogramming is essential for tumor cell growth and propagation, and most cancers, including breast cancer, are accompanied by abnormalities in energy metabolism. Here, we confirmed that sodium cantharidate inhibited cell viability using the Cell Counting Kit-8, clonogenic assay, and Transwell assay. The cell cycle and apoptosis assays indicated that sodium cantharidate induced apoptosis and cell cycle arrest in breast cancer cells. Additionally, proteomic assays, western blots, and metabolic assays revealed that sodium cantharidate converted the metabolic phenotype of breast cancer cells from glycolysis to oxidative phosphorylation. Furthermore, bioinformatics analysis identified possible roles for p53 with respect to the effects of sodium cantharidate on breast cancer cells. Western blot, docking, and phosphatase assays revealed that the regulation of p53 activity by sodium cantharidate was related to its inhibition of protein phosphatase 5 activity. Moreover, sodium cantharidate significantly inhibited tumor growth in tumor-bearing nude mice. In summary, our study provides evidence for the use of sodium cantharidate as an effective and new therapeutic candidate for the treatment of human breast cancer in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cantharidin/analogs & derivatives , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Nuclear Proteins/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cantharidin/pharmacology , Cell Cycle Checkpoints/drug effects , Female , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
8.
Front Pharmacol ; 12: 671328, 2021.
Article in English | MEDLINE | ID: mdl-34054543

ABSTRACT

The emergence of secondary resistance is the main failure cause of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) as a targeted therapy for non-small cell lung cancer (NSCLC). EGFR mutations of NSCLC cells can markedly increase glutamine transporter (SLC1A5) expression, thereby increasing glutamine metabolism. Glutamine metabolites can activate EGFR downstream signals, including mTOR, ERK1/2, STAT3, etc., which is an important cause for the decreased sensitivity of NSCLC to EGFR-TKIs. CCK8 and Annexin V/PI assays were conducted to detect the effects of Almonertinib and/or V9302 on the proliferation and apoptosis of NSCLC cells. Proteomics was used to determine the effect of Almonertinib on energy metabolism-related proteins in NSCLC. siRNA transfection was performed to study the effect of SLC1A5 down-regulation on cell proliferation. In addition, the effects of drugs on colony formation capacity were determined by colony formation assay. Immunofluorescence and Western blot were utilized to detect the apoptosis- and autophagy-related proteins expression. DAPI staining was utilized to detect the effect of drugs on the nucleus. Transmission electron microscope was used to observe the changes of submicroscopic structure such as autophagosomes and nucleus of cells. mCherry-GFP-LC3B tandem fluorescent protein was to used to detect the level of autophagy flux. Tumor-bearing nude mouse model was utilized to detect the effect of V9302 on the anti-tumor effect of Almonertinib in vivo. As a result, Almonertinib suppressed H1975 and A549 cell proliferation depended on its dosage and treatment duration, and it also induced apoptosis. A549 cells with wild-type EGFR had lower sensitivity to Almonertinib. The expression of SLC1A5 was up-regulated by stimulating with low concentration of Almonertinib in NSCLC cells. SLC1A5 was highly expressed in A549 cells with wild-type EGFR. Glutamine deletion or SLC1A5 inhibition/silencing inhibited the proliferation of NSCLC cells, and decreased cellular glutamine uptake. The combination of SLC1A5 inhibitor V9302 and Almonertinib had a synergistic inhibitory effect on the proliferation of NSCLC. V9302 enhanced the effect of Almonertinib in apoptosis-inducing in NSCLC cells. The combination of V9302 and Almonertinib might induce apoptosis by inhibiting autophagy.

9.
Photochem Photobiol ; 97(4): 795-804, 2021 07.
Article in English | MEDLINE | ID: mdl-33351957

ABSTRACT

Stem cell therapies have attracted a lot of attention in the fields of dermatological and esthetic medicine. The paracrine action of stem cells is deemed to play a crucial role in skin treatments. Many reports have demonstrated the beneficial effects of conditioned medium (CM) derived from ADSCs on skin photoaging. However, few reports have presented the application of exosome (Exo) derived from ADSCs in the treatment of photoaging. To clarify the effects of Exo, we collected Exo from the CM of ADSCs and the photoprotective effects of Exo, as well as those of the CM with and without Exo, were investigated by detecting the intracellular ROS, DNA damage and some photoaging-associated signal pathways on UVB-treated human dermal fibroblasts. The results showed that Exo had significant efficiency in preventing photoaging, and it could inhibit UVB-induced cellular DNA damage, overexpression of ROS and MMP-1 via regulating Nrf2 and MAPK/AP-1 pathway. In addition, Exo could effectively activate the TGF-ß/Smad pathway to elevate the expression of procollagen type I. However, these photoprotective effects were weakened when Exo was removed from the CM. Taken together, the results suggested that Exo, a key component of paracrine activity, played an important role in the treatment of photoaging.


Subject(s)
Exosomes , Skin Aging , Collagen Type I , Culture Media, Conditioned/pharmacology , Fibroblasts , Humans , Reactive Oxygen Species , Skin , Skin Diseases , Ultraviolet Rays
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(9): 1332-1339, 2020 Sep 30.
Article in Chinese | MEDLINE | ID: mdl-32990225

ABSTRACT

OBJECTIVE: To assess the changes in the effects of cantharides after alkaline processing on proliferation, migration, invasion, and apoptosis of human lung cancer A549 cells. METHODS: Human non-small cell lung cancer A549 cells were treated with cantharis extract (CTE) from raw cantharides and alkali processed cantharis extract (ACE). The proliferation of the cells was detected with CCK-8 assay, and the cell migration and invasion were assessed using wound healing assay and Transwell assay, respectively. The expressions of MMP1 and MMP2 in the cells were detected using Western blotting, the contents of IFN-γ, IL-1ß and TNF-α were measured with ELISA, and cell apoptosis was analyzed with annexinV/PI fluorescent staining. RESULTS: Both CTE and ACE significantly reduced the viability and inhibited the migration of A549 cells, and high-dose ACE produced a significantly stronger inhibitory effect on cell migration than high- dose CTE (P < 0.01). ACE showed more potent inhibitory effect than CTE on the invasion of A549 cells (P < 0.01). Both CTE and ACE inhibited the expressions of MMP1 and MMP2 and up-regulated the level of IFN-γ without significantly affecting the levels of IL-1ß and TNF-α. Annexin V/PI staining showed that both CTE and ACE caused apoptosis of A549 cells, but ACE had a stronger proapoptotic effect. CONCLUSIONS: Processing with sodium hydroxide can significantly improve the antitumor activity of cantharides, which inhibits the proliferation, migration and invasion of A549 cells possibly by down-regulating the expressions of MMP1 and MMP2, promoting apoptosis and increasing the level of IFN-γ.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Cantharidin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation , Humans , Neoplasm Invasiveness
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(6): 884-892, 2020 Jun 30.
Article in Chinese | MEDLINE | ID: mdl-32895203

ABSTRACT

OBJECTIVE: To observe the cell death pattern induced by gefitinib in non-small cell lung cancer A549 and H1975 cells and explore the possible mechanism in light of glycolysis. METHODS: The inhibitory effects of gefitinib at 20, 30, or 40 µmol/L in A549 cells and at 20, 40, or 80 µmol/L in H1975 cells were examined using MTT assay. The changes of lactic acid level in the cells were determined with a lactic acid kit, and the expression levels of glycolysis-related proteins (PKM2 and HK2) and the proteins in PI3K-Akt-mTOR signaling pathway were detected using Western blotting. 2-NBDG was used for detecting glucose uptake capacity of the cells, and ATP kit was used to detect the intracellular ATP level. The mitochondrial membrane potential of the cells was examined with the JC-1 kit, and cell apoptosis was analyzed with Annexin V-FITC/PI double staining. The relative expression levels of the apoptotic proteins Bax and Bcl-2 and the autophagy marker protein LC3B were detected with Western blotting. RESULTS: MTT assay showed that gefitinib inhibited the proliferation of A549 and H1975 cells in a time- and dose-dependent manner (P < 0.05). The IC50 of gefitinib at 24, 48 and 72 h was 48.6, 28.6 and 19.7 µmol/L in A549 cells and was 321.6, 49.1 and 14.6 µmol/L in H1975 cells, respectively. Gefitinib significantly lowered intracellular lactic acid level of the cells (P < 0.05) and down-regulated the expressions of PKM2 and HK2 proteins (P < 0.05) and PI3K-Akt-mTOR signaling pathway-associated proteins (P < 0.05). Gefitinib obviously inhibited glucose uptake and ATP levels in both A549 and H1975 cells (P < 0.05). Treatment with gefitinib induced obviously enhanced apoptosis in the cells, resulting in apoptosis rates of (10.77± 1.0)%, (14.5±0.4)%, (17.4±0.2)% and (32.1±0.6)% at 0, 20, 30 and 40 µmol/L in A549 cells (P < 0.05) and of (10.5±0.6)%, (13.2± 0.92)%, (18.9±0.98)% and (35.1±1.4)% at 0, 20, 40 and 80 µmol/L in H1975 cells, respectively (P < 0.05). The protein expression of Bax increased and that of Bcl-2 decreased following gefitinib treatment in the cells (P < 0.05). Gefitinib significantly increased autophagy in A549 and H1975 cells as shown by increased LC3B expressions following the treatment (P < 0.05). CONCLUSIONS: Gefitinib can inhibit the proliferation, induce apoptosis and increase autophagy in A549 and H1975 cells. Gefitinib induces apoptosis of the cells possibly by affecting glycolysis and PI3K-Akt-mTOR signaling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Apoptosis , Cell Line, Tumor , Cell Proliferation , Gefitinib , Glycolysis , Humans , Phosphatidylinositol 3-Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...