1
|
Han X, Zhou C, Luo X, Pang H, Han C, Tang L, Yang Z, Nong Y, Lu C. Tumor Targeting with Apatinib-loaded Nanoparticles and Sonodynamic Combined Therapy. Curr Mol Med 2024; 24:648-666. [PMID: 37312441 DOI: 10.2174/1566524023666230613140341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION This study implies the enhancement of apatinib killing effect in 4T1 tumor cells through constructing drug-loaded nanoparticles apatinib/Ce6@ZIF- 8@Membranes (aCZM) to enhance tumor therapeutic targeting and reduce toxic side following sonodynamic therapy (SDT). METHODS apatinib/Ce6@ZIF-8 (aCZ) were synthesized by in situ encapsulation, and aCZM were constructed by encapsulating the nanoparticles with extracted breast cancer 4T1 cell membranes. aCZM were characterized and tested for the stability by electron microscopy, and the membrane proteins on the nanoparticles' surface were assessed using SDS-PAGE gel electrophoresis. The cell viability of 4T1 cells following treatment with aCZM was tested using cell counting kit-8 (CCK-8). The uptake of nanoparticles was detected by laser confocal microscopy and flow cytometry, and the SDT-mediated production of reactive oxygen species (ROS) was verified by singlet oxygen sensor green (SOSG), electron spin resonance (ESR), and DCFH-DA fluorescent probes. The CCK-8 assay and flow cytometry using Calcein/PI were used to assess the antitumoral effect of aCZM nanoparticles under SDT. The biosafety of aCZM was further verified in vitro and in vivo using the hemolysis assay, routine blood test and H&E staining of vital organs in Balb/c mice. RESULTS aCZM with an average particle size of about 210.26 nm were successfully synthesized. The results of the SDS-PAGE gel electrophoresis experiment showed that aCZM have a band similar to that of pure cell membrane proteins. The CCK-8 assay demonstrated the absence of effects on cell viability at a low concentration range, and the relative cell survival rate reached more than 95%. Laser confocal microscopy and flow cytometry analysis showed that aCZM treated group has the strongest fluorescence and the highest cellular uptake of nanoparticles. SOSG, ESR, and DCFH-DA fluorescent probes all indicated that the aCZM + SDT treated group has the highest ROS production. The CCK-8 assay also showed that when the ultrasound intensity was fixed at 0.5 W/cm2, the relative cell survival rates in the medium concentration group (10 μg/ml) (5.54 ± 1.26%) and the high concentration group (20 μg/ml) (2.14 ± 1.63%) were significantly lower than those in the low concentration group (5 μg/ml) (53.40 ± 4.25%). Moreover, there was a concentration and intensity dependence associated with the cellkilling effect. The mortality rate of the aCZM in the ultrasound group (44.95 ± 3.03%) was significantly higher than that of the non-ultrasound (17.00 ± 2.26%) group and aCZ + SDT group (24.85 ± 3.08%) (P<0.0001). The live and dead cells' staining (Calcein/PI) also supported this result. Finally, in vitro hemolysis test at 4 and 24 hours showed that the hemolysis rate of the highest concentration group was less than 1%. The blood routine, biochemistry, and H&E staining results of major organs in Balb/c mice undergoing nano-treatments showed no obvious functional abnormalities and tissue damage in 30 days. CONCLUSION In this study, a multifunctional bionic drug delivery nanoparticles (aCZM) system with good biosafety and compatibility in response to acoustic dynamics was successfully constructed and characterized. This system enhanced apatinib killing effect on tumor cells and reduced toxic side effects under SDT.
Collapse
Affiliation(s)
- Xiao Han
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caifu Zhou
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xiaoling Luo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hongbing Pang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Libo Tang
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Ziye Yang
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yingdan Nong
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Chunmiao Lu
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
2
|
Pan Y, Wei M, Gong T. Ultrasound microbubble-mediated delivery of ANLN silencing-repressed EZH2 expression alleviates cervical cancer progression. Tissue Cell 2022; 77:101843. [PMID: 35679682 DOI: 10.1016/j.tice.2022.101843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
Abstract
Ultrasound-targeted microbubble destruction (UTMD) is a new gene therapy method that uses ultrasound and microbubbles carrying target genes to achieve gene transfection. However, whether UTMD-mediated ANLN silencing transfection helps to restrain the growth of cervical cancer (CC) is obscure. ANLN level in tumor tissues, adjacent tissues, and cells was tested using the database, qRT-PCR, and western blot. The optimal concentration of SF6 was determined by MTT assay. Mechanical index (MI) was selected by flow cytometry. After transfection with liposome or UTMD-mediated liposome, cell function experiments, qRT-PCR, and western blot were employed to assess CC cell biological behaviors and EZH2 level. Epithelial-mesenchymal transition (EMT)-related marker and apoptosis-related marker expressions were examined utilizing qRT-PCR and western blot. 10% SF6 and MI of 0.28 were selected for subsequent tests. ANLN was highly expressed in CC and cells. The transfection efficiency of the UTMD-siANLN group was higher than that of the L-siANLN group. Moreover, the repression of UTMD-siANLN on CC cell malignant phenotypes was stronger than L-siANLN. UTMD-siANLN attenuated EZH2 expression in CC cells. The modulatory role of UTMD-siANLN on EMT- and apoptosis-related markers was reversed by EZH2 overexpression. UTMD can improve the efficiency of siANLN transfection into CC cells to induce suppression of CC cell malignant phenotypes, which may become a new target of gene therapy for CC.
Collapse
Affiliation(s)
- Yanyan Pan
- Ultrasonic Diagnosis Center, Northwest Women's and Children's Hospital, Xi'an 710061, China
| | - Min Wei
- Department of Ultrasonic, Shanghai Rici Women's and Children's Hospital, Shanghai 200040, China
| | - Ting Gong
- Ultrasonic Diagnosis Center, Northwest Women's and Children's Hospital, Xi'an 710061, China.
| |
Collapse
|
3
|
Dong L, Li N, Wei X, Wang Y, Chang L, Wu H, Song L, Guo K, Chang Y, Yin Y, Pan M, Shen Y, Wang F. A Gambogic Acid-Loaded Delivery System Mediated by Ultrasound-Targeted Microbubble Destruction: A Promising Therapy Method for Malignant Cerebral Glioma. Int J Nanomedicine 2022; 17:2001-2017. [PMID: 35535034 PMCID: PMC9078874 DOI: 10.2147/ijn.s344940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/19/2022] [Indexed: 12/12/2022] Open
Abstract
Background Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Lei Dong
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Xixi Wei
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yongling Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Liansheng Chang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Hongwei Wu
- Department of Chemistry, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Liujiang Song
- Department of Ophthalmology, Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517, USA
| | - Kang Guo
- Department of Oncology, The Third affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yuqiao Chang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Yaling Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
| | - Min Pan
- Department of Ultrasound, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518034, People’s Republic of China
- Min Pan, Department of Ultrasound, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, No. 6001 Beihuan Avenue, Shenzhen, 518034, People’s Republic of China, Email
| | - Yuanyuan Shen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518060, People’s Republic of China
| | - Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453003, People’s Republic of China
- Correspondence: Feng Wang, Henan Key Laboratory of Medical Tissue Regeneration, School of Basic Medical Sciences, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, Henan, 453002, People’s Republic of China, Email
| |
Collapse
|
4
|
Tang X, Hao N, Zhou Y, Liu Y. Ultrasound targeted microbubble destruction-mediated SOCS3 attenuates biological characteristics and epithelial-mesenchymal transition (EMT) of breast cancer stem cells. Bioengineered 2022; 13:3896-3910. [PMID: 35109743 PMCID: PMC8973955 DOI: 10.1080/21655979.2022.2031384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
SOCS3 is low-expressed in breast cancer and may be a potential target. Ultrasound targeted microbubble destruction (UTMD) improved the efficiency of gene transfection. We explored the effects of UTMD-mediated transfection of SOCS3 on the biological characteristics and epithelial-mesenchymal transition (EMT) of breast cancer stem cells (BCSCs). The expression of SOCS3 in breast cancer (BC) and its association with prognosis were evaluated by GEPIA and The Cancer Genome Atlas (TCGA) websites. BCSCs were sorted by flow cytometry and immunomagnetic bead method, followed by sphere formation, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and xenograft assays to test their effects in vitro and in vivo. The levels of SOCS3, EMT- and STAT3 pathway-related genes were determined by RT-qPCR and Western blot, respectively. The effects of liposome and UTMD on BCSCs and mice were compared by the gain-of-function experiments. Low expression of SOCS3 was associated with poor prognosis of BC patients, and found in BC and BCSCs. BCSCs were successfully sorted, with high viability and tumorigenicity. UTMD increased the transfection rate of SOCS3. Moreover, UTMD- and liposome-mediated SOCS3 reduced cell viability, proliferation, migration and invasion, blocked cell cycle, inhibited sphere formation in BCSCs, and retarded tumor growth in mice. Mechanistically, overexpressed SOCS3 inhibited the expressions of EMT-related genes and the activation of STAT3 pathway in BCSCs and mice. The regulatory effects of UTMD-mediated SOCS3 on the above-mentioned biological characteristics were better than liposome-mediated SOCS3. UTMD-mediated SOCS3 has a better therapeutic effect in BC, providing new experimental evidence for the treatment of BC.
Collapse
Affiliation(s)
- Xiaojiang Tang
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Na Hao
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuhui Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yang Liu
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
5
|
Bifunctional alginate/chitosan stabilized perfluorohexane nanodroplets as smart vehicles for ultrasound and pH responsive delivery of anticancer agents. Int J Biol Macromol 2021; 191:1068-1078. [PMID: 34600955 DOI: 10.1016/j.ijbiomac.2021.09.166] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 09/07/2021] [Accepted: 09/24/2021] [Indexed: 12/30/2022]
Abstract
The combination of ultrasound and chemotherapy has been proposed as a promising strategy to achieve a better anticancer therapeutic efficacy. Here we present a facile strategy to construct novel bifunctional nanodroplets as smart vehicles for ultrasound and pH responsive delivery of anticancer agents. PFH is used as core and chitosan/alginate complexes are used as the stable shells of the nanodroplets. The effects of alginate/chitosan ratio, and the amount of surfactant as well as PFH on the size, size distribution, and encapsulation efficiency of nanodroplets are systematically investigated with the optimized formulation identified. The release of the encapsulated doxorubicin hydrochloride can be triggered by changing the pH value of the surrounding environment and the exposure to ultrasound. The nanodroplets also show strong ultrasound contrast via droplet-to-bubble transition as demonstrated by B-mode ultrasound imaging. The hemolytic activity and cytotoxicity are further studied, revealing the biocompatibility of the nanodroplets. The in vivo antitumor results demonstrate that the prepared droplets show excellent antitumor therapeutic efficacy and outstanding tumor-targeting ability. The proposed alginate/chitosan stabilized PFH nanodroplets represent an important advance in fabricating multifunctional therapeutic materials with great promises in the applications of combined antitumor therapies.
Collapse
|
6
|
Cheng L, Zhang D, Yan W. Ultrasound‑targeted microbubble destruction‑mediated overexpression of Sirtuin 3 inhibits the progression of ovarian cancer. Oncol Rep 2021; 46:220. [PMID: 34396428 PMCID: PMC8377464 DOI: 10.3892/or.2021.8171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Ultrasound-targeted microbubble destruction (UTMD) has recently been developed as a promising noninvasive tool for organ- and tissue-specific gene or drug delivery. The aim of the present study was to explore the role of UTMD-mediated Sirtuin 3 (SIRT3) overexpression in the malignant behaviors of human ovarian cancer (HOC) cells. Reverse transcription-quantitative PCR was performed to detect SIRT3 mRNA expression levels in normal human ovarian epithelial cells and HOC cell lines; low SIRT3 expression was found in HOC cell lines, and the SKOV3 cell line was used in the following experiments. The SIRT3-microbubble (MB) was prepared, and the effects of ultrasound-treated SIRT3-MB on biological processes of SKOV3 cells were determined. The proliferation, migration, invasion and apoptosis of SKOV3 cells were measured after SIRT3 upregulation by UTMD. Xenograft tumors in nude mice were induced to observe tumor growth in vivo. Upregulation of SIRT3 inhibited the malignant behaviors of SKOV3 cells, whereas UTMD-mediated SIRT3 upregulation further inhibited proliferation, epithelial-mesenchymal transition, invasion and migration, and induced apoptosis of SKOV3 cells, and it also inhibited tumor formation and growth in vivo. Moreover, the present study identified hypoxia inducible factor-1α (HIF-1α) as a target of SIRT3. The present study provided evidence that UTMD-mediated overexpression of SIRT3 may suppress HOC progression through the inhibition of HIF-1α.
Collapse
Affiliation(s)
- Li Cheng
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Dongmei Zhang
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Wei Yan
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
7
|
Chen X, Cheng P, Hu C. LncRNA FEZF1-AS1 accelerates the migration and invasion of laryngeal squamous cell carcinoma cells through miR-4497 targeting GBX2. Eur Arch Otorhinolaryngol 2021; 278:1523-1535. [PMID: 33550476 DOI: 10.1007/s00405-021-06636-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/20/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND MiR-4497 has been previously proved to exert an anti-cancer role in laryngeal squamous cell carcinoma (LSCC) by negatively regulating gastrulation brain homeobox 2 (GBX2). However, the mechanism of miR-4497 in LSCC has yet to be fully elucidated. This study intended to investigate the role of FEZF1-AS1 in the migration and invasion of LSCC cells and clarified its mechanism through miR-4497 and GBX2. METHODS qPCR evaluated the expression of FEZF1-AS1, miR-4497 and GBX2 in LSCC tissues and cells, compared with controls. Western blotting analyzed GBX2, E-cadherin, N-cadherin and Vimentin. CCK8, wound healing and transwell assays assessed the viability, migration and invasion of TU686 and UM-SCC-17A cells. Luciferase reporter assay affirmed the interplay of miR-4497 with FEZF1-AS1 or GBX2 and Pearson's correlation analysis explored the association between each two genes in both tumor and non-tumor tissues. RESULTS FEZF1-AS1 was highly expressed in LSCC tissues and cells. Silence or elevation of FEZF1-AS1 inhibited or promoted the migration and invasion of TU686 and UM-SCC-17A cells. FEZF1-AS1 targeted and negatively modulated miR-4497. Inhibition of miR-4497 markedly restored the FEZF1-AS1 silence-repressed cell viability of TU686 and UM-SCC-17A cells. Further, FEZF1-AS1 could positively regulate GBX2 via negative regulation of miR-4497. In these two cells, GBX2 deficiency reversed the promoting impacts of miR-4497 repression on migration and invasion. CONCLUSION Taken together, FEZF1-AS1, heightened in LSCC tissues and cells, promotes cell migration and invasion of LSCC cells via targeting miR-4497 that inhibits GBX2. The finding may offer new options for the treatment of this cancer.
Collapse
Affiliation(s)
- Xudong Chen
- Department of Otolaryngology, First Hospital of Ningbo City, Ningbo, Zhejiang, 315000, People's Republic of China.
| | - Peng Cheng
- Department of Otolaryngology, First Hospital of Ningbo City, Ningbo, Zhejiang, 315000, People's Republic of China
| | - Cihao Hu
- Department of Otolaryngology, First Hospital of Ningbo City, Ningbo, Zhejiang, 315000, People's Republic of China
| |
Collapse
|
8
|
Distinct roles for the hypoxia-inducible transcription factors HIF-1α and HIF-2α in human osteoclast formation and function. Sci Rep 2020; 10:21072. [PMID: 33273561 PMCID: PMC7713367 DOI: 10.1038/s41598-020-78003-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/19/2020] [Indexed: 01/04/2023] Open
Abstract
Bone homeostasis is maintained by a balance between osteoblast-mediated bone formation and osteoclast-driven bone resorption. Hypoxia modulates this relationship partially via direct and indirect effects of the hypoxia-inducible factor-1 alpha (HIF-1α) transcription factor on osteoclast formation and bone resorption. Little data is available on the role(s) of the HIF-2α isoform of HIF in osteoclast biology. Here we describe induction of HIF-1α and HIF-2α during the differentiation of human CD14+ monocytes into osteoclasts. Knockdown of HIF-1α did not affect osteoclast differentiation but prevented the increase in bone resorption that occurs under hypoxic conditions. HIF-2α knockdown did not affect bone resorption but moderately inhibited osteoclast formation. Growth of osteoclasts in 3D gels reversed the effect of HIF-2α knockdown; HIF-2α siRNA increasing osteoclast formation in 3D. Glycolysis is the main HIF-regulated pathway that drives bone resorption. HIF knockdown only affected glucose uptake and bone resorption in hypoxic conditions. Inhibition of glycolysis with 2-deoxy-d-glucose (2-DG) reduced osteoclast formation and activity under both basal and hypoxic conditions, emphasising the importance of glycolytic metabolism in osteoclast biology. In summary, HIF-1α and HIF-2α play different but overlapping roles in osteoclast biology, highlighting the importance of the HIF pathway as a potential therapeutic target in osteolytic disease.
Collapse
|
9
|
Guo C, Wang C, Zhang J, Chen X. Treatment for Liver Tumor Using Combined Transarterial Embolization and Interaarterial Transfecting HIF-1α shRNA in a Rabbit VX2 Model. Onco Targets Ther 2020; 13:8511-8519. [PMID: 32904418 PMCID: PMC7455770 DOI: 10.2147/ott.s262434] [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: 05/21/2020] [Accepted: 07/31/2020] [Indexed: 11/23/2022] Open
Abstract
Background Hypoxia-inducible factor-1α (HIF-1α) has been selected as therapeutic gene in gene therapy. The aim of this study was to explore the treatment effect of combined transarterial embolization using microsphere treatment (MD) and intraarterial transfecting HIF-1α shRNA on hepatocellular carcinoma (HCC). Materials and Methods Rabbit skin fibroblast was transfected with HIF-1α shRNA to evaluate the knocking down efficiency. Sixteen rabbit VX2 liver tumor models were randomly divided into four groups: the control group without any treatment, the MD group, the shRNA group (HIF-1α shRNA transfection by transcatheter intraarterial infusion), and the shRNA+MD group. The necrotic score, mitotic count and expression of HIF-1α, vascular endothelial growth factor (VEGF), CD34 and periodic acid-Schiff (PAS) stain were evaluated at the 14th and 28th day after treatment. The expression of HIF-1α and VEGF of VX2 tumors was also evaluated by real-time polymerase chain reaction on the 28th day. Results The expression of HIF-1α-mRNA was lower in HIF-1α shRNA group than the control (p < 0.01). The tumor size was smaller in the shRNA + MD group than the shRNA group and the MD group (p < 0.05) on the 28th day. The growth rate of tumors in the shRNA + MD group was also lower than in other groups. The gene and protein expressions of both HIF-1α and VEGF in the shRNA + MD group were lower than the MD group, shRNA group and control group on the 28th day (p < 0.05). The necrotic score was higher in the shRNA + MD group than the MD group and control group (p < 0.05). The mitotic count and PAS-positive cells in shRNA + MD group were lower and CD34 was higher than the other three groups (p < 0.05). Conclusion Compared to therapy with MD or HIF-1α shRNA with transcatheter intraarterial transfection alone, the combined treatment has a better effect on HCC.
Collapse
Affiliation(s)
- Chuangen Guo
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, People's Republic of China
| | - Cheng Wang
- Department of Radiology, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu Province 210029, People's Republic of China
| | - Jingfeng Zhang
- Department of Radiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province 310003, People's Republic of China.,Department of Radiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang Province 315010, People's Republic of China
| | - Xiao Chen
- Department of Radiology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province 210029, People's Republic of China
| |
Collapse
|