Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 12(7)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38964787

ABSTRACT

BACKGROUND: Chimeric antigen receptor natural killer (CAR-NK) therapy holds great promise for treating hematologic tumors, but its efficacy in solid tumors is limited owing to the lack of suitable targets and poor infiltration of engineered NK cells. Here, we explore whether immunogenic cell death (ICD) marker ERp57 translocated from endoplasmic reticulum to cell surface after drug treatment could be used as a target for CAR-NK therapy. METHODS: To target ERp57, a VHH phage display library was used for screening ERp57-targeted nanobodies (Nbs). A candidate Nb with high binding affinity to both human and mouse ERp57 was used for constructing CAR-NK cells. Various in vitro and in vivo studies were performed to assess the antitumor efficacy of the constructed CAR-NK cells. RESULTS: We demonstrate that the translocation of ERp57 can not only be induced by low-dose oxaliplatin (OXP) treatment but also is spontaneously expressed on the surface of various types of tumor cell lines. Our results show that G6-CAR-NK92 cells can effectively kill various tumor cell lines in vitro on which ERp57 is induced or intrinsically expressed, and also exhibit potent antitumor effects in cancer cell-derived xenograft and patient-derived xenograft mouse models. Additionally, the antitumor activity of G6-CAR-NK92 cells is synergistically enhanced by the low-dose ICD-inducible drug OXP. CONCLUSION: Collectively, our findings suggest that ERp57 can be leveraged as a new tumor antigen for CAR-NK targeting, and the resultant CAR-NK cells have the potential to be applied as a broad-spectrum immune cell therapy for various cancers by combining with ICD inducer drugs.


Subject(s)
Immunogenic Cell Death , Killer Cells, Natural , Oxaliplatin , Protein Disulfide-Isomerases , Humans , Animals , Mice , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Immunogenic Cell Death/drug effects , Protein Disulfide-Isomerases/metabolism , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Xenograft Model Antitumor Assays , Neoplasms/immunology , Neoplasms/drug therapy , Neoplasms/therapy , Female
2.
J Cell Mol Med ; 27(19): 2906-2921, 2023 10.
Article in English | MEDLINE | ID: mdl-37471521

ABSTRACT

Numerous studies have shown the positive correlation between high levels of Pi and tumour progression. A critical goal of macrophage-based cancer therapeutics is to reduce anti-inflammatory macrophages (M2) and increase proinflammatory antitumour macrophages (M1). This study aimed to investigate the relationship between macrophage polarization and low-Pi stress. First, the spatial populations of M2 and M1 macrophages in 22 HCC patient specimens were quantified and correlated with the local Pi concentration. The levels of M2 and M1 macrophage markers expressed in the peritumour area were higher than the intratumour levels, and the expression of M2 markers was positively correlated with Pi concentration. Next, monocytes differentiated from THP-1 cells were polarized against different Pi concentrations to investigate the activation or silencing of the expression of p65, IκB-α and STAT3 as well as their phosphorylation. Results showed that low-Pi stress irreversibly repolarizes tumour-associated macrophages (TAMs) towards the M1 phenotype by silencing stat6 and activating p65. Moreover, HepG-2 and SMCC-7721 cells were cultured in conditioned medium to investigate the innate anticancer immune effects on tumour progression. Both cancer cell lines showed reduced proliferation, migration and invasion, as epithelial-mesenchymal transition (EMT) was inactivated. In vivo therapeutic effect on the innate and adaptive immune processes was validated in a subcutaneous liver cancer model by the intratumoural injection of sevelamer. Tumour growth was significantly inhibited by the partial deprivation of intratumoural Pi as the tumour microenvironment under low-Pi stress is more immunostimulatory. The anticancer immune response, activated by low-Pi stress, suggests a new macrophage-based immunotherapeutic modality.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Tumor-Associated Macrophages/metabolism , Macrophages/metabolism , Monocytes/metabolism , Cell Line, Tumor , Tumor Microenvironment
3.
Biochem Pharmacol ; 213: 115593, 2023 07.
Article in English | MEDLINE | ID: mdl-37196682

ABSTRACT

Sorafenib is a tyrosine kinase inhibitor for the treatment of advanced-stage HCC; however, clinical trials of sorafenib failed to demonstrate long-term survival benefits due to drug resistance. Low Pi stress has been shown to inhibit tumor growth and the expression of multidrug resistance-associated proteins. In this study, we investigated the sensitivity of HCC to sorafenib under conditions of low Pi stress. As a result, we found that low Pi stress facilitated sorafenib-mediated suppression of migration and invasion of HepG-2 and Hepa1-6 cells by decreasing the phosphorylation or expression of AKT, Erk and MMP-9. Angiogenesis was inhibited due to decreased expression of PDGFR under low Pi stress. Low Pi stress also decreased the viability of sorafenib-resistant cells by directly regulating the expression of AKT, HIF-1a and P62. In vivo drug sensitivity analysis in the four animal models showed a similar tendency that low Pi stress enhances sorafenib sensitivity in both the normal and drug-resistant models. Altogether, low Pi stress enhances the sensitivity of hepatocellular carcinoma to sorafenib and expands the indications for sevelamer.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Sorafenib/pharmacology , Sorafenib/therapeutic use , Carcinoma, Hepatocellular/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Liver Neoplasms/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Cell Line, Tumor , Mice, Inbred Strains , Drug Resistance, Neoplasm , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
4.
J Thorac Dis ; 15(4): 2012-2021, 2023 Apr 28.
Article in English | MEDLINE | ID: mdl-37197556

ABSTRACT

Background: Orthotopic models of lung cancer have been widely utilized, and the purpose of this study was to demonstrate the viability of our proposed modified modeling approach. Methods: A total of 50 female BALB/c mice were implanted with 1×1×1 mm fragments of a tumor sample into the left lung lobe. After 2 months of observation, the mice were humanely euthanized through CO2 inhalation. The macroscopic specimens were photographed, and the most representative neoplastic lesions were collected for histological analysis. Small-animal positron emission tomography/computed tomography (PET/CT) scans were conducted on 6 randomly selected mice. Results: Local tumor formation, ipsilateral thoracic tissue infiltration, the contralateral chest wall, right lung metastases, and distant kidney metastases were observed in these models. Overall, the tumor development and metastasis rates were 60.86% (28/46) and 57.14% (16/28), respectively. The 3 mice that had a small-animal PET/CT scan developed a local tumor, but no distant metastases were observed. Conclusions: This modified method was deemed reliable, reproducible, minimally invasive, straightforward, and comprehensible; it might serve as the foundation for developing patient-derived orthotopic xenografts of lung cancer.

5.
Biomed Pharmacother ; 158: 114094, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36502755

ABSTRACT

As an emerging tumor therapy, ideal oncolytic viruses preferentially replicate in malignant cells, reverse the immunosuppressive tumor microenvironment, and eventually can be eliminated by the patient. It is of great significance for cancer treatment to discover new excellent oncolytic viruses. Here, we found that WNV live attenuated vaccine WNV-poly(A) could be developed as a novel ideal oncolytic agent against several types of cancers. Mechanistically, due to its high sensitivity to type Ι interferon (IFN-Ι), WNV-poly(A) could specifically kill tumor cells rather than normal cells. At the same time, WNV-poly(A) could activate Dendritic cells (DCs) and trigger tumor antigen specific response mediated by CD8 + T cell, which contributed to inhibit the propagation of original and distal tumor cells. Like intratumoral injection, intravenous injection with WNV-poly(A) also markedly delays Huh7 hepatic carcinoma (HCC) transplanted tumor progression. Most importantly, in addition to an array of mouse xenograft tumor models, WNV-poly(A) also has a significant inhibitory effect on many different types of patient-derived tumor tissues and HCC patient-derived xenograft (PDX) tumor models. Our studies reveal that WNV-poly(A) is a potent and excellent oncolytic agent against many types of tumors and may have a role in metastatic and recurrent tumors.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Oncolytic Viruses , Animals , Mice , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Immunity , Liver Neoplasms/therapy , Neoplasm Recurrence, Local , Oncolytic Viruses/metabolism , Tumor Microenvironment , Virus Replication
6.
Sci Rep ; 10(1): 4681, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32170125

ABSTRACT

Triple-negative breast cancer (TNBC) has a more aggressive phenotype and higher metastasis and recurrence rates than other breast cancer subtypes. TNBC currently lacks a transplantation model that is suitable for clinical simulations of the tumor microenvironment. Intraductal injection of tumor cells into the mammary duct could mimic the occurrence and development of breast cancer. Herein, we injected 4T1 cells into the mammary ducts of BALB/C mice to build a preclinical model of TNBC and optimized the related construction method to observe the occurrence and spontaneous metastasis of tumors. We compared the effects of different cell numbers on tumorigenesis rates, times to tumorigenesis, and metastases to determine the optimal number of cells for modelling. We demonstrated that 4T1-MIND model mice injected with 20,000 cells revealed a suitable tumor formation rate and time, thus indicating a potential treatment time window after distant metastasis. We also injected 20,000 cells directly into the breast fat pad or breast duct for parallel comparison. The results still showed that the 4T1-MIND model provides sufficient treatment time for lung metastases in mice and that it is a more reliable model for early tumor development. The 4T1-MIND model requires continuous improvement and optimization. A suitable and optimized model for translational research and studies on the microenvironment in TNBC should be developed.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/pathology , Triple Negative Breast Neoplasms/pathology , Animals , Biopsy , Female , Immunohistochemistry , Isografts , Mice , Models, Biological , Neoplasm Metastasis , Neoplasm Staging , Organ Specificity , Translational Research, Biomedical
7.
Hum Reprod ; 32(2): 290-298, 2017 02.
Article in English | MEDLINE | ID: mdl-28031325

ABSTRACT

STUDY QUESTION: Is diethylstilbestrol (DES), a prototypical endocrine-disrupting chemical (EDC), able to induce physiological changes in human spermatozoa and affect progesterone actions? SUMMARY ANSWER: DES promoted Ca2+ flux into human spermatozoa by activating the cation channel of sperm (CatSper) and suppressed progesterone-induced Ca2+ signaling, tyrosine phosphorylation and sperm functions. WHAT IS KNOWN ALREADY: DES significantly impairs the male reproductive system both in fetal and postnatal exposure. Although various EDCs affect human spermatozoa in a non-genomic manner, the effect of DES on human spermatozoa remains unknown. STUDY DESIGN, SIZE, DURATION: Sperm samples from normozoospermic donors were exposed in vitro to a range of DES concentrations with or without progesterone at 37°C in a 5% CO2 incubator to mimic the putative exposure to this toxicant in seminal plasma and the female reproductive tract fluids. The incubation time varied according to the experimental protocols. All experiments were repeated at least five times using different individual sperm samples. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human sperm intracellular calcium concentrations ([Ca2+]i) were monitored with a multimode plate reader following sperm loading with Ca2+ indicator Fluo-4 AM, and the whole-cell patch-clamp technique was performed to record CatSper and alkalinization-activated sperm K+ channel (KSper) currents. Sperm viability and motility parameters were assessed by an eosin-nigrosin staining kit and a computer-assisted semen analysis system, respectively. The ability of sperm to penetrate into viscous media was examined by penetration into 1% methylcellulose. The sperm acrosome reaction was measured using chlortetracycline staining. The level of tyrosine phosphorylation was determined by western blot assay. MAIN RESULTS AND THE ROLE OF CHANCE: DES exposure rapidly increased human sperm [Ca2+]i dose dependently and even at an environmentally relevant concentration (100 pM). The elevation of [Ca2+]i was derived from extracellular Ca2+ influx and mainly mediated by CatSper. Although DES did not affect sperm viability, motility, penetration into viscous media, tyrosine phosphorylation or the acrosome reaction, it suppressed progesterone-stimulated Ca2+ signaling and tyrosine phosphorylation. Consequently, DES (1-100 µM) significantly inhibited progesterone-induced human sperm penetration into viscous media and acrosome reaction. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Although DES has been shown to disturb progesterone actions on human spermatozoa, this study was performed in vitro, and caution must be taken when extrapolating the results in practical applications. WIDER IMPLICATIONS OF THE FINDINGS: The present study revealed that DES interfered with progesterone-stimulated Ca2+ signaling and tyrosine phosphorylation, ultimately inhibited progesterone-induced human sperm functions and, thereby, might impair sperm fertility. The non-genomic manner in which DES disturbs progesterone actions may be a potential mechanism for some estrogenic endocrine disruptors to affect human sperm function. STUDY FUNDING/COMPETING INTERESTS: National Natural Science Foundation of China (No. 31400996); Natural Science Foundation of Jiangxi, China (No. 20161BAB204167 and No. 20142BAB215050); open project of National Population and Family Planning Key Laboratory of Contraceptives and Devices Research (No. 2016KF07) to T. Luo; National Natural Science Foundation of China (No. 81300539) to L.P. Zheng. The authors have no conflicts of interest to declare.


Subject(s)
Calcium Channels/metabolism , Diethylstilbestrol/pharmacology , Estrogens, Non-Steroidal/pharmacology , Progesterone/pharmacology , Spermatozoa/drug effects , Acrosome Reaction/drug effects , Calcium/metabolism , Calcium Signaling/drug effects , Dose-Response Relationship, Drug , Humans , Male , Phosphorylation/drug effects , Sperm Motility/drug effects , Spermatozoa/metabolism
8.
Reprod Toxicol ; 60: 69-75, 2016 04.
Article in English | MEDLINE | ID: mdl-26867864

ABSTRACT

Matrine, a bioactive alkaloid widely used in Chinese medicine, inhibits mouse sperm functions in vitro. In this study, we investigated the reproductive toxicity of matrine to male mice in vivo. C57BL/6J mice were administered with daily doses of 0, 1, 10 and 50mg/kg matrine by intraperitoneal injection for 30 days. The results showed that matrine did not affect testis size, testis weight, sperm count and sperm viability, but it significantly inhibited total motility, progressive motility, linear velocity, capacitation and the progesterone-induced acrosome reaction of mouse sperm. Furthermore, the intracellular Ca(2+) concentration ([Ca(2+)]i), a key regulator of sperm function, was reduced in sperm of matrine-exposed mice. The current and gene expression of the sperm specific Ca(2+) channel, CatSper, which modulates Ca(2+) influx in sperm, were decreased in testes of matrine-exposed mice. These results indicate that matrine inhibits mouse sperm functions by a [Ca(2+)]i-related mechanism via CatSper channel.


Subject(s)
Alkaloids/toxicity , Calcium/metabolism , Quinolizines/toxicity , Spermatozoa/drug effects , Animals , Calcium Channels/genetics , Calcium Channels/physiology , Male , Mice, Inbred C57BL , Sperm Capacitation/drug effects , Sperm Count , Sperm Motility/drug effects , Spermatozoa/metabolism , Spermatozoa/physiology , Testis/anatomy & histology , Testis/drug effects , Matrines
9.
Cell Physiol Biochem ; 35(1): 374-85, 2015.
Article in English | MEDLINE | ID: mdl-25591778

ABSTRACT

BACKGROUND: Matrine is a bioactive alkaloid that has a variety of pharmacological effects and is widely used in Chinese medicine. However, its effects on male reproduction are not well known. In this study, we aimed to investigate the in vitro toxicity of matrine on mature mouse sperm. METHODS: Mouse cauda epididymal sperm were exposed to matrine (10-200 µM) in vitro. The viability, motility, capacitation, acrosome reaction and fertilization ability of the mouse sperm were examined. Furthermore, the intracellular calcium concentration ([Ca(2+)]i), calcium (Catsper) and potassium (Ksper) currents, and phosphorylation of extracellular signal regulated kinases 1/2 (p-ERK1/2) of the sperm were analyzed. RESULTS: After exposure to 100 µM or more of matrine, mouse cauda epididymal sperm exhibited a significant reduction in total motility, progressive motility, linear velocity and acrosome reaction rate induced by Ca(2+) ionophore A23187. As a result, the fertilization ability of mouse sperm was remarkably decreased by matrine. Our data further demonstrated that matrine significantly reduced sperm [Ca(2+)]i and [Ca(2+)]i-related p-ERK1/2; however, both the CatSper and KSper currents, which are thought to interactively regulate Ca(2+) influx in sperm, were not affected by matrine. CONCLUSION: Our findings indicate that matrine inhibits mouse sperm function by reducing sperm [Ca(2+)]i and suppressing the phosphorylation of ERK1/2.


Subject(s)
Alkaloids/pharmacology , Calcium/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Quinolizines/pharmacology , Spermatozoa/drug effects , Acrosome Reaction/drug effects , Aniline Compounds/chemistry , Animals , Calcimycin/pharmacology , Female , Fertilization in Vitro/drug effects , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Patch-Clamp Techniques , Phosphorylation/drug effects , Potassium/metabolism , Sperm Motility/drug effects , Spermatozoa/chemistry , Spermatozoa/metabolism , Xanthenes/chemistry , Matrines
10.
Reprod Toxicol ; 51: 14-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25463531

ABSTRACT

Emodin, a bioactive anthraquinone widely used in Chinese traditional medicine, disrupts mouse testicular gene expression in vivo. In this study, we investigated the toxicity of emodin to human sperm in vitro. Different doses of emodin (25, 50, 100, 200 and 400µM) were applied to ejaculated human sperm. The results indicated that 100, 200 and 400µM emodin significantly inhibited the total motility, progressive motility and linear velocity of human sperm. In addition, sperm's ability to penetrate viscous medium together with progesterone induced capacitation and acrosome reaction was also adversely affected by emodin. In contrast, emodin did not affect sperm viability. Furthermore, intracellular Ca(2+) concentration ([Ca(2+)]i) and tyrosine phosphorylation, which serve as key regulators of sperm function, were dose-dependently reduced by emodin (50-400µM). These results suggest that emodin inhibits human sperm functions by reducing sperm [Ca(2+)]i and suppressing tyrosine phosphorylation in vitro.


Subject(s)
Emodin/toxicity , Spermatozoa/drug effects , Calcium/metabolism , Humans , Male , Phosphorylation , Progesterone , Sperm Capacitation/drug effects , Sperm Motility/drug effects , Spermatozoa/metabolism , Spermatozoa/physiology , Tyrosine/metabolism
11.
Inflammation ; 38(1): 400-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25348861

ABSTRACT

Uterine cervical carcinoma (UCC) is one of the most common malignant tumors in females, and UCC has a close relationship with chronic cervicitis. As the endogenous "braking signal," lipoxins can regulate anti-inflammation and the resolution of inflammation. We investigated the effect of lipoxin A4 (LXA4) on the proliferation, apoptosis, and migration in lipopolysaccharide (LPS)-stimulated Hela cells. We demonstrated that LXA4 could significantly suppress p53, cyclin D1 expression, and migration of LPS-stimulated Hela cells via nuclear factor-κB (NF-κB) pathway, and these effects could be blocked by Boc-2, the specific inhibitor of FPR2/ALX (the receptor of LXA4). We presented evidence for a novel role of LXA4 on the proliferation and migration in LPS-stimulated Hela cells, and FPR2/ALX was involved in the procedures. These results showed that LXA4 could be a possible candidate for UCC therapy, and blocking the activation of NF-κB would be an effective drug target.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Lipopolysaccharides/toxicity , Lipoxins/pharmacology , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Movement/physiology , Cell Proliferation/physiology , Dose-Response Relationship, Drug , Female , HeLa Cells , Humans , NF-kappa B/metabolism , Signal Transduction/physiology
12.
Med Sci Monit Basic Res ; 19: 228-36, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23982457

ABSTRACT

BACKGROUND: Plumbagin, a quinonoid constituent isolated from the root of Plumbago zeylanica L., has been proven to possess anti-tumor activity both in vitro and in vivo. However, its anti-tumor properties for human tongue carcinoma have not been reported. This study aimed to investigate the inhibitory effect and the underlying mechanism of plumbagin on the growth of human tongue carcinoma cells. MATERIAL AND METHODS: Cell proliferation ability was detected by EdU incorporation assay and colony formation assay. Cell-cycle distribution was determined by flow cytometric analysis using propidium iodide (PI) staining. Cellular apoptosis was then evaluated by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. Western blotting was applied to assay the expression of Bax and Bcl-2. RESULTS: Plumbagin inhibited the growth and proliferation of Tca8113 cells in vitro in a concentration- and time-dependent manner. The cell cycles of plumbagin-treated Tca8113 cells were arrested at the G2/M phase. Cells treated with plumbagin presented the characteristic morphological changes of apoptosis. The ratio of Bax/Bcl-2 was raised by plumbagin in a concentration-dependent manner. CONCLUSIONS: These results indicate that plumbagin induces the apoptosis of Tca8113 cells through mitochondria-mediated pathway.


Subject(s)
Apoptosis/drug effects , Mitochondria/metabolism , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Deoxyuridine/analogs & derivatives , Deoxyuridine/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mitochondria/drug effects , Mitosis/drug effects , Time Factors , Tumor Stem Cell Assay , bcl-2-Associated X Protein/metabolism
13.
Acta Pharmacol Sin ; 33(2): 242-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22231395

ABSTRACT

AIM: To investigate the effects and underlying mechanisms of plumbagin, a naphthoquinone derived from medicinal plant Plumbago zeylanica, on human gastric cancer (GC) cells. METHODS: Human gastric cancer cell lines SGC-7901, MKN-28, and AGS were used. The cell viability was examined using CCK-8 viability assay. Cell proliferation rate was determined using both clonogenic assay and EdU incorporation assay. Apoptosis was detected via Annexin V/propidium iodide double-labeled flow cytometry. Western blotting was used to assess the expression of both NF-κB-regulated gene products and TNF-α-induced activation of p65, IκBα, and IKK. The intracellular location of NF-κB p65 was detected using confocal microscopy. RESULTS: Plumbagin (2.5-40 µmol/L) concentration-dependently reduced the viability of the GC cells. The IC(50) value of plumbagin in SGC-7901, MKN-28, and AGS cells was 19.12, 13.64, and 10.12 µmol/L, respectively. The compound (5-20 µmol/L) concentration-dependently induced apoptosis of SGC-7901 cells, and potentiated the sensitivity of SGC-7901 cells to chemotherapeutic agents TNF-αand cisplatin. The compound (10 µmol/L) downregulated the expression of NF-κB-regulated gene products, including IAP1, XIAP, Bcl-2, Bcl-xL, tumor factor (TF), and VEGF. In addition to inhibition of NF-κB p65 nuclear translocation, the compound also suppressed TNF-α-induced phosphorylation of p65 and IKK, and the degradation of IκBα. CONCLUSION: Plumbagin inhibits cell growth and potentiates apoptosis in human GC cells through the NF-κB pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , NF-kappa B/immunology , Naphthoquinones/pharmacology , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , I-kappa B Kinase/immunology , Plumbaginaceae/chemistry , Stomach Neoplasms/immunology , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...