1
|
Fu C, Li H, Li N, Miao X, Xie M, Du W, Zhang LM. Conjugating an anticancer drug onto thiolated hyaluronic acid by acid liable hydrazone linkage for its gelation and dual stimuli-response release. Carbohydr Polym 2015; 128:163-170. [PMID: 26005152 DOI: 10.1016/j.carbpol.2015.04.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/02/2015] [Accepted: 04/04/2015] [Indexed: 01/17/2023] [Imported: 12/03/2023]
Abstract
A prodrug gelation strategy was developed for the sustained and dual stimuli-response release of doxorubicin hydrochloride (DOX·HCl), a commonly used anticancer drug. For this purpose, the chemical conjugation of DOX·HCl onto thiolated hyaluronic acid (HA) was carried out by an acid liable hydrazone linkage and verified by (1)H NMR analyses. When exposed to the air, such a polysaccharide conjugate showed unique self-gelation ability in aqueous solution. The gelation time and extent depended mainly on the content of thiol groups on thiolated HA. The resultant hydrogel exhibited a dominant elastic response and a thixotropic property. In particular, it could release sustainably conjugated DOX·HCl in dual pH- and reduction-responsive modes. The cumulative drug release was found to be significantly accelerated under the conditions mimicking the intracellular environments of cancer cells. The in vitro cytotoxicity assays for the human nasopharyngeal carcinoma CNE2 cells treated with various release media confirmed the effectiveness of this conjugate hydrogel for cancer cell inhibition.
Collapse
|
|
10 |
35 |
2
|
Wang H, Huang F, Zhang Z, Wang P, Luo Y, Li H, Li N, Wang J, Zhou J, Wang Y, Li S. Feedback Activation of SGK3 and AKT Contributes to Rapamycin Resistance by Reactivating mTORC1/4EBP1 Axis via TSC2 in Breast Cancer. Int J Biol Sci 2019; 15:929-941. [PMID: 31182914 PMCID: PMC6535796 DOI: 10.7150/ijbs.32489] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/14/2019] [Indexed: 12/20/2022] [Imported: 02/03/2025] Open
Abstract
The mTORC1 inhibitors, such as rapamycin and its analogs, show limited antitumor activity in clinic, reasons for which have not been clearly elucidated. Here, we undertook an effort to uncover the mechanisms underlying the limited efficacy of rapamycin, and found that the transit suppression of 4EBP1 phosphorylation led to cap-dependent translation and cell proliferation in breast cancer cells. AKT only partially contributed to 4EBP1 re-phosphorylation. By taking advantage of mass spectrometry-based phosphoproteomic analysis, we identified SGK3 as a potent kinase involved in 4EBP1 re-phosphorylation. SGK3 deletion inhibited 4EBP1 phosphorylation and cap-dependent translation. Importantly, 4EBP1 phosphorylation was positively correlated with SGK3 activity in 67 clinical breast cancer specimens. Moreover, SGK3 deletion in combination with AKT inhibition almost blocked the 4EBP1 re-phosphorylation that was induced by rapamycin and profoundly enhanced rapamycin-induced growth inhibition in vitro and in an MCF7 breast cancer mouse xenograft model in vivo. Mechanistically, the feedback activation of SGK3 by rapamycin was dependent on hVps34 and mTORC2, and reactivated mTORC1/4EBP1 axis by phosphorylating TSC2. Collectively, our study reveals a critical role of SGK3 in mediating rapamycin resistance, and provides a rationale for targeting SGK3 to improve mTOR-targeted therapies.
Collapse
|
research-article |
6 |
22 |
3
|
Weng H, Bejjanki NK, Zhang J, Miao X, Zhong Y, Li H, Xie H, Wang S, Li Q, Xie M. TAT peptide-modified cisplatin-loaded iron oxide nanoparticles for reversing cisplatin-resistant nasopharyngeal carcinoma. Biochem Biophys Res Commun 2019; 511:597-603. [PMID: 30826059 DOI: 10.1016/j.bbrc.2019.02.117] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 02/21/2019] [Indexed: 02/07/2023] [Imported: 02/03/2025]
Abstract
As chemo-radiotherapy continues to increase the lifespan of patients with nasopharyngeal carcinoma (NPC), adverse reaction and drug resistance remain two major problems when using cisplatin (CDDP). In this study, we took the lead in designing a dual-mechanism anti-cancer system modified with cell-penetrating peptide on the surface of superparamagnetic iron oxide nanoparticles (SPION) to enhance CDDP delivery efficacy to NPC cells, especially CDDP resistant NPC cells. The combinatorial delivery of CDDP and iron oxide nanoparticles showed an unexpected effect on reversal of CDDP resistance due to the Fenton reaction with an average decrease in the half maximal inhibitory concentration (IC 50) of 85% and 94% in HNE-1/DDP and CNE-2/DDP resistant cells respectively compared to CDDP alone. On this basis, modification with TAT peptide (YGRKKRRQRRR) significantly improved tumor intracellular uptake, devoting to better curative effects and minimized side effects by reducing CDDP therapeutic doses. Furthermore, we specifically labelled CDDP with fluorescence for detection of intracellular nanoparticles uptake and mechanism research through drug tracing. This novel compound provides a promising therapy for reducing chemotherapy side effects and reversing CDDP-resistant nasopharyngeal carcinoma.
Collapse
|
|
6 |
22 |
4
|
Qiu L, Li H, Fu S, Chen X, Lu L. Surface markers of liver cancer stem cells and innovative targeted-therapy strategies for HCC. Oncol Lett 2018; 15:2039-2048. [PMID: 29434903 PMCID: PMC5776936 DOI: 10.3892/ol.2017.7568] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 11/02/2017] [Indexed: 12/20/2022] [Imported: 02/03/2025] Open
Abstract
Liver cancer stem cells (LCSCs) have important roles in the occurrence, development, recurrence, therapy resistance and metastasis of hepatocellular carcinoma (HCC). Therefore, intensive studies are undergoing to identify the mechanisms by which LCSCs contribute to HCC invasion and metastasis, and to design more efficient treatments for this disease. With continuous efforts in LCSC research over the years, therapies targeting LCSCs are thought to have great potential for the clinical treatment and prognosis of liver cancer. Novel LCSC surface markers are continuously discovered and several have been used in targeted therapies to reduce HCC recurrence, metastasis, and drug resistance following tumor resection. The present review describes the surface markers characterizing LCSCs and the recent progress in therapies targeting these markers, including antibodies and polypeptides.
Collapse
|
research-article |
7 |
19 |
5
|
Wang H, Wu R, Yu L, Wu F, Li S, Zhao Y, Li H, Luo G, Wang J, Zhou J. SGEF is overexpressed in prostate cancer and contributes to prostate cancer progression. Oncol Rep 2012; 28:1468-1474. [PMID: 22824926 DOI: 10.3892/or.2012.1917] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Accepted: 05/30/2012] [Indexed: 11/05/2022] [Imported: 02/03/2025] Open
Abstract
The purpose of this study was to investigate the potential roles of the SH3-containing guanine nucleotide exchange factor (SGEF) in human prostate cancer. Experimental data showed that SGEF was overexpressed in human prostate cancer cells and specimens. The reduction of SGEF expression through an SGEF-targeting siRNA in androgen-independent C4-2 and C4-2B cells suppressed both anchorage-dependent and anchorage-independent growth. In addition, the androgen receptor (AR) antagonist bicalutamide further strengthened this inhibitory effect due to the suppression of the elevated AR transactivation after knockdown of SGEF. Collectively, our results provide the first demonstration that SGEF is a novel promoter of human prostate cancer progression and development.
Collapse
|
|
13 |
11 |
6
|
Zhao Y, Li H, Wu R, Li S, Wang P, Wang H, Wang J, Zhou J. Antitumor Effects of Oncolytic Adenovirus-Carrying siRNA Targeting Potential Oncogene EphA3. PLoS One 2015; 10:e0126726. [PMID: 25978371 PMCID: PMC4433270 DOI: 10.1371/journal.pone.0126726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 04/07/2015] [Indexed: 11/18/2022] [Imported: 02/03/2025] Open
Abstract
Conditionally replicating adenoviruses (CRAds) armed with antitumor transgenes hold promise for cancer treatment. In previous studies, we showed that the 1504-siRNA targeting potential oncogene EphA3 was an efficient therapeutic transgene and that the telomerase reverse transcriptase promoter (TERTp) driving the CRAd was a more advanced generation of CRAd. Therefore, we combined Ad-TERTp-E1A-1504 by inserting 1504-siRNA into the CRAd to study its antitumor effects and mechanism of action, using Ad-TERTp-E1A-NC and nonreplicating adenovirus carrying 1504-siRNA as controls. Cell viability assays and ED50 studies of growth inhibition confirmed that Ad-TERTp-E1A-1504 has 3.5- and 1,400-fold greater ability to kill EphA3- and TERT-expressing tumor cells compared to Ad-TERTp-E1A-NC and Ad-ΔE1A-1504, respectively. Also, Ad-TERTp-E1A-1504 had little effect on cells that modestly expressed EphA3 and TERT such as 2BS. The antitumor efficacy of Ad-TERTp-E1A-1504 was also validated in vivo. Furthermore, the virus yield of Ad-TERTp-E1A-1504 in C4-2B was ~1,000 times greater than that in 2BS. No obvious differences were observed between Ad-TERTp-E1A-1504 and Ad-TERTp-E1A-NC. Both acridine orange staining and Beclin1 protein measurements indicated that autophagy with Ad-TERTp-E1A-1504 at 5 and 10 MOI was higher than that of Ad-TERTp-E1A-NC. Finally, the classical negatively regulated autophagy signaling pathway, PI3K/AKT/mTOR, was suppressed (reduced phosphorylated form) in contrast to NC, and that this was mediated by 1504-siRNA. Thus, Ad- TERTp-E1A-1504 does not harm normal cells but has dual inhibiting and killing effects on TERT- and EphA3-positive tumor cells, and this effect is mediated by the AKT/mTOR signaling pathway via induction of autophagy. These data may offer a foundation for novel antitumor therapies targeting this mechanism.
Collapse
|
research-article |
10 |
10 |
7
|
Li H, Fu C, Miao X, Li Q, Zhang J, Yang H, Liu T, Chen X, Xie M. Multifunctional magnetic co-delivery system coated with polymer mPEG-PLL-FA for nasopharyngeal cancer targeted therapy and MR imaging. J Biomater Appl 2017; 31:1169-1181. [PMID: 28185478 DOI: 10.1177/0885328217692964] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] [Imported: 12/03/2023]
Abstract
The gene and drug co-delivery system has become one of the primary strategies to overcome cancers. Here, we designed a multifunctional magnetic co-delivery system for nasopharyngeal carcinoma-targeted therapy and MR imaging. Aldehyde sodium alginate (ASA) was used to decorate the oxide iron and load cisplain through coordinate bond to form a core complex. The polymer shell poly(l-lysine)-methoxy-polyethylene glycol-folate was used to coat the core complex through electric interaction to give this nano-medicine a target ability. And this polymer could also give the nano-medicine abilities to adhere and protect DNA, and enhance its solubleness in water. After being transfected with this nano-medicine, the plasmids which contain cancer suppressor gene TFPI2 could enter and express in HNE-1 cells. It caused a higher death and apoptosis rate, inhibited nasopharyngeal carcinoma cells' migration and cloning by the synergic effect together with cisplain. Besides, clear images of this nano-medicine could be got under T2 MR imaging. This magnetic co-delivery system demonstrates a potential as a powerful multifunctional vector for drug delivery and gene vector applications in nasopharyngeal carcinoma.
Collapse
|
|
8 |
10 |
8
|
Feng S, Liu J, Hailiang L, Wen J, Zhao Y, Li X, Lu G, Gao P, Zeng X. Amplification of RAD54B promotes progression of hepatocellular carcinoma via activating the Wnt/β-catenin signaling. Transl Oncol 2021; 14:101124. [PMID: 34049150 PMCID: PMC8167290 DOI: 10.1016/j.tranon.2021.101124] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/24/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] [Imported: 08/30/2023] Open
Abstract
Liver cancer was reported to be the sixth most frequently diagnosed cancer, and hepatocellular carcinoma (HCC) accounts for 75%-85% of primary liver cancer. Nevertheless, the concrete molecular mechanisms of HCC progression remain obscure, which is essential to elucidate. The expression profile of RAD54B in HCC was measured using qPCR and western blotting. Moreover, the levels of RAD54B in paraffin-embedded samples were evaluated using immunohistochemistry (IHC). The effect of RAD54B on HCC progression was testified by in vitro experiments, and in vivo orthotopic xenograft tumor experiments. The mechanisms of RAD54B promoting HCC progression were investigated through molecular and function experiments. Herein, RAD54B are dramatically upregulated in HCC tissues and cell lines both on mRNA and protein levels, and RAD54B can servers as an independent prognostic parameter of 5-year overall survival and 5-year disease-free survival for patients with HCC. Moreover, up-regulation of RAD54B dramatically increases the capacity for in vitro cell viability and motility, and in vivo intrahepatic metastasis of HCC cells. Mechanistically, RAD54B promotes the HCC progression through modulating the wnt/β-catenin signaling. Notably, blocking the wnt/β-catenin signaling axis can counteract the activating effects of RAD54B on motility of HCC cells. Besides, further analysis illustrates that DNA amplification is one of the mechanisms leading to mRNA overexpression of RAD54B in HCC. Our findings indicate that RAD54B might be a promising potential prognostic marker and a candidate therapeutic target to therapy HCC.
Collapse
|
research-article |
4 |
9 |
9
|
Wang H, Li S, Li H, Li C, Guan K, Luo G, Yu L, Wu R, Zhang X, Wang J, Zhou J. SGEF enhances EGFR stability through delayed EGFR trafficking from early to late endosomes. Carcinogenesis 2013; 34:1976-1983. [PMID: 23661635 DOI: 10.1093/carcin/bgt157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] [Imported: 02/03/2025] Open
Abstract
Previously, we demonstrated an elevated SH3-containing guanine nucleotide exchange factor (SGEF) expression in clinical specimens with prostate cancer and implicated the role of SGEF in prostate tumorigenesis. However, the molecular mechanism behind the SGEF regulation of prostate cancer development is not known. In this study, we show that SGEF expression delays epidermal growth factor receptor (EGFR) degradation in prostate cancer cells and is independent from its guanine nucleotide exchange factor (GEF) function. We further show that the delayed degradation is due to a delay in EGFR trafficking from early to late endosomes and not to a decrease in EGFR ubiquitination. Finally, we show that depletion of SGEF significantly inhibits epidermal growth factor-induced EGFR signaling cascade and cell migration in the prostate cancer cells. We report for the first time an SGEF function for RhoG that excludes GEF and the ability of SGEF to enhance EGFR stability and signaling by delaying its lysosomal sorting and degradation. This could be one mechanism by which SGEF contributes to prostate cancer progression.
Collapse
|
|
12 |
8 |
10
|
Wang H, Li S, Li H, Wang P, Huang F, Zhao Y, Yu L, Luo G, Zhang X, Wang J, Zhou J. Grb2 interacts with SGEF and antagonizes the ability of SGEF to enhance EGF-induced ERK1/2 activation. Mol Cell Biochem 2014; 389:239-247. [PMID: 24399467 DOI: 10.1007/s11010-013-1945-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 12/18/2013] [Indexed: 02/02/2023] [Imported: 02/03/2025]
Abstract
Previously, we demonstrated that SGEF enhances EGFR stability; however, SGEF-mediated downstream signaling of EGFR is not well understood. Here, we show that SGEF enhances EGF-induced ERK1/2 activation independent of its guanine nucleotide exchange (GEF) activity. We further show that SGEF interacts with Grb2, a critical downstream transducer of EGFR. Surprisingly, we found that interaction of Grb2 to SGEF antagonizes the ability of SGEF to enhance EGF-induced ERK1/2 activation. Taken together, this study reports a novel function of SGEF that excludes GEF and also provides important insights into the complex role of Grb2 in EGFR signal transduction.
Collapse
|
|
11 |
6 |
11
|
Chen J, Li HL, Li BB, Li W, Ma D, Li YH, Liu T. Serum- and glucocorticoid-inducible kinase 3 is a potential oncogene in nasopharyngeal carcinoma. Braz J Otorhinolaryngol 2019; 85:705-715. [PMID: 30108027 PMCID: PMC9443024 DOI: 10.1016/j.bjorl.2018.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 05/03/2018] [Accepted: 05/30/2018] [Indexed: 01/06/2023] [Imported: 02/03/2025] Open
Abstract
INTRODUCTION Serum- and glucocorticoid-inducible kinase 3, a serine/threonine kinase that functions downstream of the PI3K signaling pathway, plays a critical role in neoplastic processes. It is expressed by various tumors and contributes to carcinogenesis. OBJECTIVE The objective was to investigate serum- and glucocorticoid-inducible kinase 3 expression in nasopharyngeal carcinoma, to study the anti-tumor effects of serum- and glucocorticoid-inducible kinase 3 shRNA by inhibiting its expression in nasopharyngeal carcinoma cells and to discuss the potential implications of our findings. METHODS Serum- and glucocorticoid-inducible kinase 3 protein expression in nasopharyngeal carcinoma cell lines (CNE-1, CNE-2, HNE-1, HONE-1, and SUNE-1) and the human immortalized nasopharyngeal epithelium cell line NP69 were assayed by western blotting. Serum- and glucocorticoid-inducible kinase 3 expression in 42 paraffin-embedded nasopharyngeal carcinoma tissues were performed by immunohistochemistry. MTT assay, flow cytometry, and scratch tests were performed after CNE-2 cells were transfected with the best serum- and glucocorticoid-inducible kinase 3 shRNA plasmid selected by western blotting using lipofectamine to study its effect on cell proliferation, apoptosis, and migration. RESULTS Serum- and glucocorticoid-inducible kinase 3 was overexpressed in human nasopharyngeal carcinoma tissues and cells. Serum- and glucocorticoid-inducible kinase 3 expression decreased markedly after CNE-2 cells were transfected with the serum- and glucocorticoid-inducible kinase 3 shRNA, leading to strong inhibition of cell proliferation and migration. In addition, the apoptosis rate increased in CNE-2 cells after serum- and glucocorticoid-inducible kinase 3 knockdown. CONCLUSION Serum- and glucocorticoid-inducible kinase 3 expression was more frequently observed as the nasopharyngeal epithelium progresses from normal tissue to carcinoma. This suggests that serum- and glucocorticoid-inducible kinase 3 contributes to the multistep process of NPC carcinogenesis. Serum- and glucocorticoid-inducible kinase 3 represents a target for nasopharyngeal carcinoma therapy, and a basis exists for the further investigation of this adjuvant treatment modality for nasopharyngeal carcinoma.
Collapse
|
research-article |
6 |
5 |
12
|
Xiao F, Liu J, Zheng Y, Quan Z, Sun W, Fan Y, Luo C, Li H, Wu X. The targeted inhibition of prostate cancer by iron-based nanoparticles based on bioinformatics. J Biomater Appl 2021; 36:3-14. [PMID: 33283584 PMCID: PMC8217887 DOI: 10.1177/0885328220975249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] [Imported: 08/30/2023]
Abstract
Prostate cancer is an epithelial malignant tumor of the prostate, and it is one of the malignant tumors with a high incidence of urogenital system in men. The local treatment of prostate cancer is mainly radical resection and radical radiotherapy, but they are not applicable to advanced prostate cancer. Systemic therapy mainly includes targeted therapy and immunotherapy which could cause many complications, and will affect the prognosis and quality of life of patients. It is urgent to find new treatments for prostate cancer. Bioinformatics offers hope for us to find reliable therapeutic targets. Bioinformatics can use the tumor informations in database and analyze them to screen out the best differentially expressed genes. Using the selected differentially expressed genes as targets, a gene interference plasmid was designed, and the constructed plasmid was used for targeted gene therapy. There are some problems about gene therapy that need to be solved, such as how to transfer genes to target cells is also an important challenge. Due to their large molecular weight and hydrophilic nature, they cannot enter cells through passive diffusion mechanisms. Here we synthesized a DNA carrier used surface modified iron based nanoparticles, and used it to load plasmid including ShRNA which can inhibit the expression of oncogene SLC4A4 selected by bioinformatics' method. After that we use this iron based nanoparticles/plasmid DNA nanocomposite to treat prostate cancer cells in vitro and in vivo. The target gene SLC4A4 we had selected using bioinformatics had a strong effect on the proliferation of prostate cells; Our nanocomposite could inhibit the expression of SLC4A4 effectively, it had strong inhibitory effects on prostate cancer cells both in vivo and in vitro, and can be used as a potential method for prostate cancer treatment.
Collapse
|
research-article |
4 |
4 |
13
|
Wang H, Li S, Li H, Li C, Guan K, Luo G, Yu L, Wu R, Zhang X, Wang J, Zhou J. SGEF enhances EGFR stability through delayed EGFR trafficking from early to late endosomes. Carcinogenesis 2013; 34:1976-1983. [DOI: doi.org/10.1093/carcin/bgt157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023] Open
|
|
12 |
|
14
|
Liang Z, Li X, Chen X, Zhou J, Li Y, Peng J, Lin Z, Liu G, Zeng X, Li C, Hang L, Li H. Fe/MOF based platform for NIR laser induced efficient PDT/PTT of cancer. Front Bioeng Biotechnol 2023; 11:1156079. [PMID: 37064235 PMCID: PMC10098195 DOI: 10.3389/fbioe.2023.1156079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/15/2023] [Indexed: 04/18/2023] [Imported: 08/30/2023] Open
Abstract
Introduction: Photodynamic therapy (PDT) and photothermal therapy (PTT) are widely used in the treatment of tumors. However, their application in the treatment of clinical tumors is limited by the complexity and irreversible hypoxia environment generated by tumor tissues. To overcome this limitation, a nanoparticle composed of indocyanine green (ICG) and Fe-MOF-5 was developed. Methods: We prepared F-I@FM5 and measured its morphology, particle size, and stability. Its enzyme like ability and optical effect was verified. Then we used MTT, staining and flow cytometry to evaluated the anti-tumor effect on EMT-6 cells in vitro. Finally, the anti-tumor effect in vivo has been studied on EMT-6 tumor bearing mice. Results: For the composite nanoparticle, we confirmed that Fe-MOF-5 has the best nanozyme activity. In addition, it has excellent photothermal conversion efficiency and generates reactive oxygen species (ROS) under near-infrared light irradiation (808 nm). The composite nanoparticle showed good tumor inhibition effect in vitro and in vivo, which was superior to the free ICG or Fe-MOF-5 alone. Besides, there was no obvious cytotoxicity in major organs within the effective therapeutic concentration. Discussion: Fe-MOF-5 has the function of simulating catalase, which can promote the decomposition of excessive H2O2 in the tumor microenvironment and produce oxygen to improve the hypoxic environment. The improvement of tumor hypoxia can enhance the efficacy of PDT and PTT. This research not only provides an efficient and stable anti-tumor nano platform, but also has broad application prospects in the field of tumor therapy, and provides a new idea for the application of MOF as an important carrier material in the field of photodynamic therapy.
Collapse
|
research-article |
2 |
|
15
|
Hang L, Li H, Zhang T, Men D, Zhang C, Gao P, Zhang Q. Au@Prussian Blue Hybrid Nanomaterial Synergy with a Chemotherapeutic Drug for Tumor Diagnosis and Chemodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:39493-39502. [DOI: doi.org/10.1021/acsami.9b13470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023]
|
|
6 |
|
16
|
Odda AH, Li H, Kumar N, Ullah N, Khan MI, Wang G, Liang K, Liu T, Pan YY, Xu AW. Polydopamine Coated PB-MnO 2 Nanoparticles as an Oxygen Generator Nanosystem for Imaging-Guided Single-NIR-Laser Triggered Synergistic Photodynamic/Photothermal Therapy. Bioconjug Chem 2020; 31:1474-1485. [DOI: doi.org/10.1021/acs.bioconjchem.0c00165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023]
|
|
5 |
|
17
|
Chen J, Li HL, Li BB, Li W, Ma D, Li YH, Liu T. Serum- and glucocorticoid-inducible kinase 3 is a potential oncogene in nasopharyngeal carcinoma. Braz J Otorhinolaryngol 2019; 85:705-715. [DOI: doi.org/10.1016/j.bjorl.2018.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023] Open
|
|
6 |
|
18
|
Chen J, Li HL, Li BB, Li W, Ma D, Li YH, Liu T. Serum- and glucocorticoid-inducible kinase 3 is a potential oncogene in nasopharyngeal carcinoma. Braz J Otorhinolaryngol 2019; 85:705-715. [DOI: https:/doi.org/10.1016/j.bjorl.2018.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023] Open
|
|
6 |
|
19
|
WANG HONGTAO, WU RUIQIN, YU LAN, WU FEIMA, LI SHANHU, ZHAO YALI, LI HAILIANG, LUO GUOLAN, WANG JIAN, ZHOU JIANGUANG. SGEF is overexpressed in prostate cancer and contributes to prostate cancer progression. Oncol Rep 2012; 28:1468-1474. [DOI: doi.org/10.3892/or.2012.1917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2023] [Imported: 12/03/2023] Open
|
|
13 |
|
20
|
Nie J, Zhang H, Li X, Qin J, Zhou J, Lu Y, Yang N, Li Y, Li H, Li C. Pachymic acid promotes ferroptosis and inhibits gastric cancer progression by suppressing the PDGFRB-mediated PI3K/Akt pathway. Heliyon 2024; 10:e38800. [PMID: 39512319 PMCID: PMC11541463 DOI: 10.1016/j.heliyon.2024.e38800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] [Imported: 02/22/2025] Open
Abstract
Gastric cancer (GC) is a common malignant tumour with high incidence and mortality rates worldwide. Despite current treatment modalities, including surgical resection and chemotherapy, challenges such as postoperative recurrence, metastasis and drug resistance persist. Therefore, investigating the feasibility and mechanism of traditional Chinese medicine in treating gastric cancer is crucial for discovering new anti-gastric cancer drugs or adjuvant therapies. Pachymic acid (PA) is a natural triterpenoid found in the traditional Chinese medicinal herb Poria cocos (PC) (Schw. Wolf). Recent studies have reported its inhibitory effects on various cancer cells, including liver, cervical, breast and gastric cancer. Our in vitro and in vivo experiments confirmed that PA inhibits the proliferation, migration and invasion of gastric cancer cells. The treatment of gastric cancer cells with various death inhibitors revealed that PA may suppress gastric cancer progression by inducing ferroptosis. Malondialdehyde, Fe2+, reactive oxygen species and glutathione assays were performed to validate the effects of PA on ferroptosis in gastric cancer. High-throughput sequencing combined with analysis of the TCGA database identified PDGFRB as a potential downstream target of PA. In vivo experiments indicated that the PDGFRB overexpression could counteract the antitumour effects of PA, while ferroptosis induced by the PI3K/Akt signalling pathway may play a key role in this process. This study provides initial evidence that PA, through its interaction with PDGFRB, alters the PI3K/Akt signalling pathway, leading to ferroptosis in gastric cancer cells, thus manifesting its antitumour properties. This discovery holds promise for the development of novel therapeutic strategies for gastric cancer patients.
Collapse
|
research-article |
1 |
|
21
|
Zhang H, Nie J, Bao Z, Shi Y, Gong J, Li H. FOXC1 promotes EMT and colorectal cancer progression by attracting M2 macrophages via the TGF-β/Smad2/3/snail pathway. Cell Signal 2025; 130:111680. [PMID: 39978609 DOI: 10.1016/j.cellsig.2025.111680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] [Imported: 02/22/2025]
Abstract
Colorectal cancer is a highly prevalent and deadly malignancy worldwide. Current treatment strategies, including surgery, chemotherapy, and targeted therapy, still face limitations due to recurrence and metastasis. By conducting a weighted gene coexpression network analysis on gene expression data from The Cancer Genome Atlas, we pinpointed critical genes linked to M2 macrophages and tumor metastasis. Among these, FOXC1 emerged as a significant prognostic indicator within our predictive model. Clinical sample analysis further confirmed that FOXC1 is upregulated in colorectal cancer tissues and associated with an unfavorable patient outcome. Both in vivo and in vitro experimental results revealed that FOXC1 promotes CRC cell migration, invasion and proliferation by regulating the expression of Snail and TGF-β/Smad2/3 pathways, thereby facilitating the epithelial-mesenchymal transition process. Additionally, FOXC1 recruits M2 macrophages to the tumor microenvironment by regulating CXCL2 expression through Snail. The TGF-β factor secreted by M2 macrophages further activates the TGF-β/Smad2/3 pathway, forming a positive feedback loop. In these processes, FOXC1 plays a critical regulatory role. In summary, this study highlights the critical significance of FOXC1 in CRC progression and indicates its viability as a therapeutic target, offering a novel theoretical foundation for the development of future CRC treatment strategies.
Collapse
|
|
1 |
|