1
|
Wang P, Zhang H, Guo K, Liu C, Chen S, Pu B, Chen S, Feng T, Jiao H, Gao C. Rapamycin inhibits B16 melanoma cell viability invitro and invivo by inducing autophagy and inhibiting the mTOR/p70‑S6k pathway. Oncol Lett 2024; 27:140. [PMID: 38385108 PMCID: PMC10877231 DOI: 10.3892/ol.2024.14273] [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: 09/15/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
Rapamycin is an immunosuppressant that has been shown to prevent tumor growth following organ transplantation. However, its exact mode of antitumor action remains unknown. The present study used the B16-F10 (B16) murine melanoma model to explore the antitumor mechanism of rapamycin, and it was revealed that rapamycin reduced B16 cell viability in vitro and in vivo. In addition, in vitro and in vivo, the results of western blotting showed that rapamycin reduced Bcl2 expression, and enhanced the protein expression levels of cleaved caspase 3 and Bax, indicating that it can induce the apoptosis of B16 melanoma cells. Furthermore, the results of cell cycle analysis and western blotting showed that rapamycin induced B16 cell cycle arrest in the G1 phase, based on the reduction in the protein expression levels of CDK1, cyclin D1 and CDK4, as well as the increase in the percentage of cells in G1 phase. Rapamycin also significantly increased the number of autophagosomes in B16 melanoma cells, as determined by transmission electron microscopy. Furthermore, the results of RT-qPCR and western blotting showed that rapamycin upregulated the protein expression levels of microtubule-associated protein light chain 3 (LC3) and Beclin-1, while downregulating the expression of p62 in vitro and in vivo, thus indicating that rapamycin could trigger cellular autophagy. The present study revealed that rapamycin in combination with chloroquine (CQ) further increased LC3 expression compared with that in the CQ group, suggesting that rapamycin induced an increase in autophagy in B16 cells. Furthermore, the results of western blotting showed that rapamycin blocked the phosphorylation of p70 ribosomal S6 kinase (p70-S6k) and mammalian target of rapamycin (mTOR) proteins in vitro and in vivo, thus suggesting that rapamycin may exert its antitumor effect by inhibiting the phosphorylation of the mTOR/p70-S6k pathway. In conclusion, rapamycin may inhibit tumor growth by inducing cellular G1 phase arrest and apoptosis. In addition, rapamycin may exert its antitumor effects by inducing the autophagy of B16 melanoma cells in vitro and in vivo, and the mTOR/p70-S6k signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Haifang Zhang
- Hainan Institute for Drug Control, Haikou, Hainan 570216, P.R. China
| | - Kaikai Guo
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Chun Liu
- Hainan Institute for Drug Control, Haikou, Hainan 570216, P.R. China
| | - Shimin Chen
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Baopeng Pu
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Sirun Chen
- Hainan Medical University Press, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Tong Feng
- School of Pharmacy, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Hanyi Jiao
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| | - Chang Gao
- Department of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, Hainan 570100, P.R. China
| |
Collapse
|
2
|
Shademan B, Karamad V, Nourazarian A, Masjedi S, Isazadeh A, Sogutlu F, Avcı CB. MicroRNAs as Targets for Cancer Diagnosis: Interests and Limitations. Adv Pharm Bull 2023; 13:435-445. [PMID: 37646065 PMCID: PMC10460809 DOI: 10.34172/apb.2023.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 04/02/2022] [Accepted: 07/01/2022] [Indexed: 09/01/2023] Open
Abstract
MicroRNAs are small RNAs with ability to attach to the large number of RNA that regulate gene expression on post-transcriptional level via inhibition or degradation of specific mRNAs. MiRNAs in cells are the primary regulators of functions such as cell growth, differentiation, and apoptosis and considerably influence cell function. The expression levels of microRNAs change in human diseases, including cancer. These changes highlight their essential role in cancer pathogenesis. Ubiquitous irregular expression profiles of miRNAs have been detected in various human cancers using genome-wide identification techniques, which are emerging as novel diagnostic and prognostic cancer biomarkers of high specificity and sensitivity. The measurable miRNAs with enhanced stability in blood, tissues, and other body fluids provide a comprehensive source of miRNA-dependent biomarkers for human cancers. The leading role of miRNAs as potential biomarkers in human cancers is discussed in this article. In addition, the interests and difficulties of miRNAs as biomarkers have been explored.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Sepideh Masjedi
- Department of Cellular and Molecular Biology Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatma Sogutlu
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| |
Collapse
|
3
|
Shademan B, Masjedi S, Karamad V, Isazadeh A, Sogutlu F, Rad MHS, Nourazarian A. CRISPR Technology in Cancer Diagnosis and Treatment: Opportunities and Challenges. Biochem Genet 2022; 60:1446-1470. [PMID: 35092559 DOI: 10.1007/s10528-022-10193-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022]
Abstract
A novel gene editing tool, the Cas system, associated with the CRISPR system, is emerging as a potential method for genome modification. This simple method, based on the adaptive immune defense system of prokaryotes, has been developed and used in human cancer research. These technologies have tremendous therapeutic potential, especially in gene therapy, where a patient-specific mutation is genetically corrected to cure diseases that cannot be cured with conventional treatments. However, translating CRISPR/Cas9 into the clinic will be challenging, as we still need to improve the efficiency, specificity, and application of the technology. In this review, we will explain how CRISPR-Cas9 technology can treat cancer at the molecular level, focusing on ordination and the epigenome. We will also focus on the promise and shortcomings of this system to ensure its application in the treatment and prevention of cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Sepideh Masjedi
- Department of Cellular and Molecular Biology Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Isazadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatma Sogutlu
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | | | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
4
|
Li M, Wang C, Yu Z, Lan Q, Xu S, Ye Z, Li R, Ying L, Zhang X, Zhou Z. MgIG exerts therapeutic effects on crizotinib-induced hepatotoxicity by limiting ROS-mediated autophagy and pyroptosis. J Cell Mol Med 2022; 26:4492-4505. [PMID: 35855570 PMCID: PMC9357634 DOI: 10.1111/jcmm.17474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/05/2022] [Accepted: 06/21/2022] [Indexed: 01/11/2023] Open
Abstract
Crizotinib (CRIZO) has been widely employed to treat non‐small‐cell lung cancer. However, hepatic inflammatory injury is the major toxicity of CRIZO, which limits its clinical application, and the underlying mechanism of CRIZO‐induced hepatotoxicity has not been fully explored. Herein, we used cell counting kit‐8 assay and flow cytometry to detect CRIZO‐induced cytotoxicity on human hepatocytes (HL‐7702). CRIZO significantly reduced the survival rate of hepatocytes in a dose‐dependent manner. Furthermore, the reactive oxygen species (ROS) assay kit showed that CRIZO treatment strongly increased the level of ROS. In addition, CRIZO treatment caused the appearance of balloon‐like bubbles and autophagosomes in HL‐7702 cells. Subsequently, Western blotting, quantitative real‐time PCR and ELISA assays revealed that ROS‐mediated pyroptosis and autophagy contributed to CRIZO‐induced hepatic injury. Based on the role of ROS in CRIZO‐induced hepatotoxicity, magnesium isoglycyrrhizinate (MgIG) was used as an intervention drug. MgIG activated the Nrf2/HO‐1 signalling pathway and reduced ROS level. Additionally, MgIG suppressed hepatic inflammation by inhibiting NF‐κB activity, thereby reducing CRIZO‐induced hepatotoxicity. In conclusion, CRIZO promoted autophagy activation and pyroptosis via the accumulation of ROS in HL‐7702 cells. MgIG exerts therapeutic effects on CRIZO‐induced hepatotoxicity by decreasing the level of ROS.
Collapse
Affiliation(s)
- Min Li
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenxiang Wang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheng Yu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qin Lan
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shaolin Xu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhongjiang Ye
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongqi Li
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Ying
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiuhua Zhang
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ziye Zhou
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Paskeh MDA, Entezari M, Clark C, Zabolian A, Ranjbar E, Farahani MV, Saleki H, Sharifzadeh SO, Far FB, Ashrafizadeh M, Samarghandian S, Khan H, Ghavami S, Zarrabi A, Łos MJ. Targeted regulation of autophagy using nanoparticles: New insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166326. [DOI: 10.1016/j.bbadis.2021.166326] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/31/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022]
|
6
|
Wu HT, Li CL, Fang ZX, Chen WJ, Lin WT, Liu J. Induced Cell Cycle Arrest in Triple-Negative Breast Cancer by Combined Treatment of Itraconazole and Rapamycin. Front Pharmacol 2022; 13:873131. [PMID: 35517785 PMCID: PMC9062109 DOI: 10.3389/fphar.2022.873131] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the aggressive molecular type of breast carcinoma, with a high metastasis/relapse incidence and cancer-related death rate, due to lack of specific therapeutic targets in the clinic. Exploring potential therapeutic targets or developing novel therapeutic strategies are the focus of intense research to improve the survival and life quality of patients with TNBC. The current study focused on drugs targeting the mTOR signaling pathway by investigating the potential utilization of itraconazole (ITZ) combined with rapamycin in the treatment of TNBC. CCK-8, colony formation and transwell assays were conducted to evaluate the effect of ITZ with rapamycin in combination on MDA-MB-231 and BT-549 TNBC cells. Synergistic inhibition was found in terms of proliferation and motility of TNBC cells. However, apoptosis was not enhanced by the combined treatment of ITZ and rapamycin. Flow cytometry analysis showed that ITZ and/or rapamycin arrested cells in G0/G1 phase and prevented G1/S phase transition. Reduced cyclin D1 protein levels were consistent with G0/G1 phase arrest, especially when resulting from the combination of ITZ with rapamycin. In conclusion, the combination of ITZ with rapamycin is a promising therapeutic strategy for patients with TNBC through synergistically arresting cells in the G0/G1 phase of the cell cycle, rather than inducing apoptosis.
Collapse
Affiliation(s)
- Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Chun-Lan Li
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Physiology/Changjiang Scholar’s Laboratory, Shantou University Medical College, Shantou, China
| | - Ze-Xuan Fang
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Physiology/Changjiang Scholar’s Laboratory, Shantou University Medical College, Shantou, China
| | - Wen-Jia Chen
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Physiology/Changjiang Scholar’s Laboratory, Shantou University Medical College, Shantou, China
| | - Wen-Ting Lin
- Department of Pathology, Shantou University Medical College, Shantou, China
| | - Jing Liu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou, China
- Department of Physiology/Changjiang Scholar’s Laboratory, Shantou University Medical College, Shantou, China
| |
Collapse
|
7
|
Li CH, Liao CC. The Metabolism Reprogramming of microRNA Let-7-Mediated Glycolysis Contributes to Autophagy and Tumor Progression. Int J Mol Sci 2021; 23:113. [PMID: 35008539 PMCID: PMC8745176 DOI: 10.3390/ijms23010113] [Citation(s) in RCA: 11] [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: 10/30/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
Cancer is usually a result of abnormal glucose uptake and imbalanced nutrient metabolization. The dysregulation of glucose metabolism, which controls the processes of glycolysis, gives rise to various physiological defects. Autophagy is one of the metabolic-related cellular functions and involves not only energy regeneration but also tumorigenesis. The dysregulation of autophagy impacts on the imbalance of metabolic homeostasis and leads to a variety of disorders. In particular, the microRNA (miRNA) Let-7 has been identified as related to glycolysis procedures such as tissue repair, stem cell-derived cardiomyocytes, and tumoral metastasis. In many cancers, the expression of glycolysis-related enzymes is correlated with Let-7, in which multiple enzymes are related to the regulation of the autophagy process. However, much recent research has not comprehensively investigated how Let-7 participates in glycolytic reprogramming or its links to autophagic regulations, mainly in tumor progression. Through an integrated literature review and omics-related profiling correlation, this review provides the possible linkage of the Let-7 network between glycolysis and autophagy, and its role in tumor progression.
Collapse
Affiliation(s)
- Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Chiao-Chun Liao
- Department of Tropical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Public Health and Department of Social Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
8
|
Concha S, Rey-Jurado E, Poli MC, Hoyos-Bachiloglu R, Borzutzky A. Refractory systemic juvenile idiopathic arthritis successfully treated with rapamycin. Rheumatology (Oxford) 2021; 60:e250-e251. [PMID: 33547778 DOI: 10.1093/rheumatology/keab114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/22/2020] [Accepted: 01/28/2021] [Indexed: 11/12/2022] Open
Affiliation(s)
- Sara Concha
- Department of Infectious Diseases and Pediatric Immunology, School of Medicine, Pontificia Universidad Católica de Chile
| | - Emma Rey-Jurado
- Facultad de Medicina Clínica Alemana de Santiago, Universidad del Desarrollo.,Program of Immunogenetics and Translational Immunology, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - M Cecilia Poli
- Facultad de Medicina Clínica Alemana de Santiago, Universidad del Desarrollo.,Program of Immunogenetics and Translational Immunology, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana-Universidad del Desarrollo, Santiago, Chile.,Texas Children's Hospital, Section of Pediatric Immunology, Allergy, and Retrovirology, Houston, TX, USA
| | - Rodrigo Hoyos-Bachiloglu
- Department of Infectious Diseases and Pediatric Immunology, School of Medicine, Pontificia Universidad Católica de Chile
| | - Arturo Borzutzky
- Department of Infectious Diseases and Pediatric Immunology, School of Medicine, Pontificia Universidad Católica de Chile.,Millennium Institute on Immunology and Immunotherapy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
9
|
Shademan B, Karamad V, Nourazarian A, Avcı CB. CAR T Cells: Cancer Cell Surface Receptors Are the Target for Cancer Therapy. Adv Pharm Bull 2021; 12:476-489. [PMID: 35935042 PMCID: PMC9348524 DOI: 10.34172/apb.2022.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/12/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
Immunotherapy has become a prominent strategy for the treatment of cancer. A method that improves the immune system's ability to attack a tumor (Enhances antigen binding). Targeted killing of malignant cells by adoptive transfer of chimeric antigen receptor (CAR) T cells is a promising immunotherapy technique in the treatment of cancers. For this purpose, the patient's immune cells, with genetic engineering aid, are loaded with chimeric receptors that have particular antigen binding and activate cytotoxic T lymphocytes. That increases the effectiveness of immune cells and destroying cancer cells. This review discusses the basic structure and function of CAR-T cells and how antigenic targets are identified to treat different cancers and address the disadvantages of this treatment for cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| |
Collapse
|
10
|
Wu T, Yao Y, Sun R, Wang H, Zhang J, Yin X, Zhou Q, Huangfu C. Arterial instillation of rapamycin in treatment of rabbit hepatic xenograft tumors and its effects on VEGF, iNOS, HIF-1α, Bcl-2, Bax expression and microvessel density. Sci Prog 2021; 104:368504211026417. [PMID: 34392719 PMCID: PMC10364938 DOI: 10.1177/00368504211026417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma is one of the leading causes of malignant tumor related death word wide with poor prognosis. Chemotherapy and TACE are main treatment methods for advanced stage cases. Rapamycin, a macrolide compound that initially used to coat coronary stents, can inhibit the growth of a variety of cancer cells especially hepatocellular carcinoma. Twenty-four healthy adult New Zealand white rabbits underwent CT-guided puncture to prepare a model of VX2 liver xenograft tumor. The rabbits were randomly divided into four groups with six in each group and received the following treatments: APR-TACE1: arterial perfusion of high-dose rapamycin combined with TACE; APR-TACE2: arterial perfusion of low-dose rapamycin combined with TACE; TACE: TACE alone; and IVR-TACE: intravenous injection of rapamycin combined with TACE. Two weeks after TACE treatment, the rabbits received CT scan and DSA angiography examination, and then killed by air embolism. The non-necrotic region and surrounding tissues were obtained from the peripheral tumor for iNOS, HIF-1α, VEGF, Bcl-2, and Bax protein expression analysis. Protein expression of iNOS, HIF-1α, VEGF, and Bcl-2 in APR-TACE1 were significantly lower than those in groups APR-TACE2, TACE, and IVR-TACE (p < 0.05). iNOS, HIF-1α, and VEGF in APR-TACE2 were lower than those in TACE (p < 0.05). iNOS and VEGF in APR-TACE2 were significantly lower than those in IVR-TACE (p < 0.05). iNOS in IVR-TACE was significantly lower than that in TACE (p < 0.05). The expression levels of Bcl-2 and Bax were statistically significant between APR-TACE2 and TACE (p < 0.05). The MVD of the tumor tissue in APR-TACE1 was lower than that of groups APR-TACE2, TACE, IVR-TACE with statistical difference (p < 0.05). However, MVD of APR-TACE2 was lower than that of groups TACE, IVR-TACE with significant statistical difference (p < 0.05). Arterial instillation of rapamycin+TACE in treatment of rabbit hepatic xenograft tumors can reduce tumor neovascularization and inhibit iNOS, HIF-1α, VEGF, Bcl-2, and Bax protein expression.
Collapse
Affiliation(s)
- Tao Wu
- Department of Radiology Intervention, The First affiliated Hospital of Henan University of Traditional Chinese Medicine (TMC), Zhengzhou, Henan, P.R. China
- Department of Radiology Intervention, The First Affiliated Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Yihui Yao
- Department of Radiology Intervention, The First Affiliated Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Ruimin Sun
- Department of Radiology Intervention, The First affiliated Hospital of Henan University of Traditional Chinese Medicine (TMC), Zhengzhou, Henan, P.R. China
| | - Huili Wang
- Department of Radiology Intervention, The First affiliated Hospital of Henan University of Traditional Chinese Medicine (TMC), Zhengzhou, Henan, P.R. China
| | - Junna Zhang
- Department of Pathology, The First Affiliated Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Xiaoxiang Yin
- Department of Radiology Intervention, The First Affiliated Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Qing Zhou
- Department of Radiology Intervention, The First Affiliated Hospital of Henan University, Kaifeng, Henan, P.R. China
| | - Chaoshen Huangfu
- College of Basic Medicine, Henan University, Kaifeng, Henan, P.R. China
| |
Collapse
|
11
|
He W, Cao X, Rong K, Chen X, Han S, Qin A. Combination of AZD3463 and DZNep Prevents Bone Metastasis of Breast Cancer by Suppressing Akt Signaling. Front Pharmacol 2021; 12:652071. [PMID: 34122074 PMCID: PMC8193724 DOI: 10.3389/fphar.2021.652071] [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: 01/11/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
Osteolysis resulting from osteoclast overactivation is one of the severe complications of breast cancer metastasis to the bone. Previous studies reported that the anti-cancer agent DZNep induces cancer cell apoptosis by activating Akt signaling. However, the effect of DZNep on breast cancer bone metastasis is unknown. We previously found that DZNep enhances osteoclast differentiation by activating Akt. Therefore, we explored the use of the anti-cancer agent AZD3463 (an Akt inhibitor) along with DZNep, as AZD3463 can act as an anti-cancer agent and can also potentially ameliorate bone erosion. We evaluated osteoclast and breast cancer cell phenotypes and Akt signaling in vitro by treating cells with DZNep and AZD3463. Furthermore, we developed a breast cancer bone metastasis animal model in mouse tibiae to further determine their combined effects in vivo. Treatment of osteoclast precursor cells with DZNep alone increased osteoclast differentiation, bone resorption, and expression of osteoclast-specific genes. These effects were ameliorated by AZD3463. The combination of DZNep and AZD3463 inhibited breast cancer cell proliferation, colony formation, migration, and invasion. Finally, intraperitoneal injection of DZNep and AZD3463 ameliorated tumor progression and protected against bone loss. In summary, DZNep combined with AZD3463 prevented skeletal complications and inhibited breast cancer progression by suppressing Akt signaling.
Collapse
Affiliation(s)
- Wenxin He
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiankun Cao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojun Chen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuai Han
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - An Qin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
The Dual Role of Autophagy in Cancer Development and a Therapeutic Strategy for Cancer by Targeting Autophagy. Int J Mol Sci 2020; 22:ijms22010179. [PMID: 33375363 PMCID: PMC7795059 DOI: 10.3390/ijms22010179] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a delicate intracellular degradation process that occurs due to diverse stressful conditions, including the accumulation of damaged proteins and organelles as well as nutrient deprivation. The mechanism of autophagy is initiated by the creation of autophagosomes, which capture and encapsulate abnormal components. Afterward, autophagosomes assemble with lysosomes to recycle or remove degradative cargo. The regulation of autophagy has bipolar roles in cancer suppression and promotion in diverse cancers. Furthermore, autophagy modulates the features of tumorigenesis, cancer metastasis, cancer stem cells, and drug resistance against anticancer agents. Some autophagy regulators are used to modulate autophagy for anticancer therapy but the dual roles of autophagy limit their application in anticancer therapy, and present as the main reason for therapy failure. In this review, we summarize the mechanisms of autophagy, tumorigenesis, metastasis, cancer stem cells, and resistance against anticancer agents. Finally, we discuss whether targeting autophagy is a promising and effective therapeutic strategy in anticancer therapy.
Collapse
|