1
|
Zhang YQ, Zhang W, Kong XT, Hai WX, Guo R, Zhang M, Zhang SL, Li B. The therapeutic effect of a novel GAPDH inhibitor in mouse model of breast cancer and efficacy monitoring by molecular imaging. Cancer Cell Int 2024; 24:188. [PMID: 38811918 PMCID: PMC11138053 DOI: 10.1186/s12935-024-03361-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND Breast cancer is a serious threat to women's health with high morbidity and mortality. The development of more effective therapies for the treatment of breast cancer is strongly warranted. Growing evidence suggests that targeting glucose metabolism may be a promising cancer treatment strategy. We previously identified a new glyceraldehyde-3-phosphate dehydrogenase (GAPDH) inhibitor, DC-5163, which shows great potential in inhibiting tumor growth. Here, we evaluated the anticancer potential of DC-5163 in breast cancer cells. METHODS The effects of DC-5163 on breast cancer cells were investigated in vitro and in vivo. Seahorse, glucose uptake, lactate production, and cellular ATP content assays were performed to examine the impact of DC-5163 on cellular glycolysis. Cell viability, colony-forming ability, cell cycle, and apoptosis were assessed by CCK8 assay, colony formation assay, flow cytometry, and immunoblotting respectively. The anticancer activity of DC-5163 in vivo was evaluated in a mouse breast cancer xenograft model. RESULTS DC-5163 suppressed aerobic glycolysis and reduced energy supply of breast cancer cells, thereby inhibiting breast cancer cell growth, inducing cell cycle arrest in the G0/G1 phase, and increasing apoptosis. The therapeutic efficacy was assessed using a breast cancer xenograft mouse model. DC-5163 treatment markedly suppressed tumor growth in vivo without inducing evident systemic toxicity. Micro-PET/CT scans revealed a notable reduction in tumor 18F-FDG and 18F-FLT uptake in the DC-5163 treatment group compared to the DMSO control group. CONCLUSIONS Our results suggest that DC-5163 is a promising GAPDH inhibitor for suppressing breast cancer growth without obvious side effects. 18F-FDG and 18F-FLT PET/CT can noninvasively assess the levels of glycolysis and proliferation in tumors following treatment with DC-5163.
Collapse
Affiliation(s)
- Yun-Qi Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Wei Zhang
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Xiang-Tai Kong
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China
| | - Wang-Xi Hai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Su-Lin Zhang
- Drug Discovery and Design Canter, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, China.
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, China.
- Collaborative Innovation Center for Molecular Imaging of Precision Medicine, Shanxi Medical University, Taiyuan, 030000, China.
| |
Collapse
|
2
|
Chen H, Zhang LF, Zhang L, Miao Y, Xi Y, Liu MF, Zhang M, Li B. CircANKRD17 promotes glycolysis by inhibiting miR-143 in breast cancer cells. J Cell Physiol 2023; 238:2765-2777. [PMID: 37812578 DOI: 10.1002/jcp.31128] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023]
Abstract
Glucose metabolic reprogramming, known as the Warburg effect, is one of the metabolic hallmarks of tumor cells. Cancer cells preferentially metabolize glucose by glycolysis rather than mitochondrial oxidative phosphorylation regardless of oxygen availability, but the regulatory mechanism underlying this switch has been incompletely understood. Here, we report that the circular RNA circ ankyrin repeat domain 17 (ANKRD17) functions as a key regulator for glycolysis to promote cell growth, migration, invasion, and cell-cycle progression in breast cancer (BC) cells. We further show that circANKRD17 acts to accelerate glycolysis in BC cells by acting as a sponge for miR-143 and in turn overrides the repressive effect of miR-143, a well-documented glycolytic repressor, on hexokinase 2 in BC cells, thus resulting in enhanced glycolysis in BC cells. These data suggest the circANKRD17-miR-143 cascade as a novel mechanism in controlling glucose metabolic reprogramming in BC cells and suggest circANKRD17 as a promising therapeutic target to interrupt cancerous glycolysis.
Collapse
Affiliation(s)
- Hong Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| | - Ling-Fei Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- State Key Laboratory of Molecular Biology, State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lu Zhang
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| | - Yun Xi
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| | - Mo-Fang Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- State Key Laboratory of Molecular Biology, State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Min Zhang
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| | - Biao Li
- Department of Nuclear Medicine, Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, Shanghai, China
| |
Collapse
|
3
|
Liu J, Wang H, Zeng D, Xiong J, Luo J, Chen X, Chen T, Xi Q, Sun J, Ren X, Zhang Y. The novel importance of miR-143 in obesity regulation. Int J Obes (Lond) 2023; 47:100-108. [PMID: 36528726 DOI: 10.1038/s41366-022-01245-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
Obesity and substantially increased risk of metabolic diseases have become a global epidemic. microRNAs have attracted a great deal of attention as a potential therapeutic target for obesity. MiR-143 has been known to specifically promote adipocyte differentiation by downregulating extracellular signal-regulated kinase 5. Our latest study found that miR-143 knockout is against diet-induced obesity by promoting brown adipose tissue thermogenesis and inhibiting white adipose tissue adipogenesis. Moreover, LPS- or IL-6-induced inhibition of miR-143 expression in brown adipocytes promotes thermogenesis by targeting adenylate cyclase 9. In this review, we will summarize the expression and functions of miR-143 in different tissues, the influence of obesity on miR-143 in various tissues, the important role of adipose-derived miR-143 in the development of obesity, the role of miR-143 in immune cells and thermoregulation and discuss the potential significance and application prospects of miR-143 in obesity management.
Collapse
Affiliation(s)
- Jie Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Huan Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dewei Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiali Xiong
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.,Jiangxi Province Key Laboratory of Animal Nutrition, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaohui Ren
- Ocean College of Hebei Agricultural University, Qinhuangdao, 066003, China.
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
4
|
Schreier A, Zappasodi R, Serganova I, Brown KA, Demaria S, Andreopoulou E. Facts and Perspectives: Implications of tumor glycolysis on immunotherapy response in triple negative breast cancer. Front Oncol 2023; 12:1061789. [PMID: 36703796 PMCID: PMC9872136 DOI: 10.3389/fonc.2022.1061789] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/17/2022] [Indexed: 01/11/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive disease that is difficult to treat and portends a poor prognosis in many patients. Recent efforts to implement immune checkpoint inhibitors into the treatment landscape of TNBC have led to improved outcomes in a subset of patients both in the early stage and metastatic settings. However, a large portion of patients with TNBC remain resistant to immune checkpoint inhibitors and have limited treatment options beyond cytotoxic chemotherapy. The interplay between the anti-tumor immune response and tumor metabolism contributes to immunotherapy response in the preclinical setting, and likely in the clinical setting as well. Specifically, tumor glycolysis and lactate production influence the tumor immune microenvironment through creation of metabolic competition with infiltrating immune cells, which impacts response to immune checkpoint blockade. In this review, we will focus on how glucose metabolism within TNBC tumors influences the response to immune checkpoint blockade and potential ways of harnessing this information to improve clinical outcomes.
Collapse
Affiliation(s)
- Ashley Schreier
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, United States,Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| | - Inna Serganova
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States,Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Sandra Demaria
- Department of Radiation Oncology and Department of Pathology, Weill Cornell Medicine, New York, NY, United States
| | - Eleni Andreopoulou
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York Presbyterian Hospital, New York, NY, United States,*Correspondence: Eleni Andreopoulou,
| |
Collapse
|
5
|
Alshahrani SH, Ibrahim YS, Jalil AT, Altoum AA, Achmad H, Zabibah RS, Gabr GA, Ramírez-Coronel AA, Alameri AA, Qasim QA, Karampoor S, Mirzaei R. Metabolic reprogramming by miRNAs in the tumor microenvironment: Focused on immunometabolism. Front Oncol 2022; 12:1042196. [PMID: 36483029 PMCID: PMC9723351 DOI: 10.3389/fonc.2022.1042196] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/24/2022] [Indexed: 01/15/2023] Open
Abstract
MicroRNAs (miRNAs) are emerging as a significant modulator of immunity, and their abnormal expression/activity has been linked to numerous human disorders, such as cancer. It is now known that miRNAs potentially modulate the production of several metabolic processes in tumor-associated immune cells and indirectly via different metabolic enzymes that affect tumor-associated signaling cascades. For instance, Let-7 has been identified as a crucial modulator for the long-lasting survival of CD8+ T cells (naive phenotypes) in cancer by altering their metabolism. Furthermore, in T cells, it has been found that enhancer of zeste homolog 2 (EZH2) expression is controlled via glycolytic metabolism through miRNAs in patients with ovarian cancer. On the other hand, immunometabolism has shown us that cellular metabolic reactions and processes not only generate ATP and biosynthetic intermediates but also modulate the immune system and inflammatory processes. Based on recent studies, new and encouraging approaches to cancer involving the modification of miRNAs in immune cell metabolism are currently being investigated, providing insight into promising targets for therapeutic strategies based on the pivotal role of immunometabolism in cancer. Throughout this overview, we explore and describe the significance of miRNAs in cancer and immune cell metabolism.
Collapse
Affiliation(s)
- Shadia Hamoud Alshahrani
- Medical Surgical Nursing Department, King Khalid University, Almahala, Khamis Mushate, Saudi Arabia
| | - Yousif Saleh Ibrahim
- Department of Medical Laboratory Techniques, Al-maarif University College, Ramadi, Al-Anbar, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, Iraq
| | - Abdelgadir Alamin Altoum
- Department of Medical Laboratory Sciences, College of Health Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Harun Achmad
- Department of Pediatric Dentistry, Faculty of Dentistry, Hasanuddin University, Makassar, Indonesia
| | - Rahman S. Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Gamal A. Gabr
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
- Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center, Giza, Egypt
| | - Andrés Alexis Ramírez-Coronel
- Health and Behavior Research Group (HBR), Catholic University of Cuenca, Cuenca, Ecuador
- Laboratory of Psychometry and Ethology, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, Universidad CES, Medellin, Colombia
| | | | | | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Oligo-Fucoidan supplementation enhances the effect of Olaparib on preventing metastasis and recurrence of triple-negative breast cancer in mice. J Biomed Sci 2022; 29:70. [PMID: 36109724 PMCID: PMC9479298 DOI: 10.1186/s12929-022-00855-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/08/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Seaweed polysaccharides have been recommended as anticancer supplements and for boosting human health; however, their benefits in the treatment of triple-negative breast cancers (TNBCs) and improving immune surveillance remain unclear. Olaparib is a first-in-class poly (ADP-ribose) polymerase inhibitor. Oligo-Fucoidan, a low-molecular-weight sulfated polysaccharide purified from brown seaweed (Laminaria japonica), exhibits significant bioactivities that may aid in disease management. METHODS Macrophage polarity, clonogenic assays, cancer stemness properties, cancer cell trajectory, glucose metabolism, the TNBC 4T1 cells and a 4T1 syngeneic mouse model were used to inspect the therapeutic effects of olaparib and Oligo-Fucoidan supplementation on TNBC aggressiveness and microenvironment. RESULTS Olaparib treatment increased sub-G1 cell death and G2/M arrest in TNBC cells, and these effects were enhanced when Oligo-Fucoidan was added to treat the TNBC cells. The levels of Rad51 and programmed death-ligand 1 (PD-L1) and the activation of epidermal growth factor receptor (EGFR) and adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) facilitate drug resistance and TNBC metastasis. However, the combination of olaparib and Oligo-Fucoidan synergistically reduced Rad51 and PD-L1 levels, as well as the activity of EGFR and AMPK; consistently, TNBC cytotoxicity and stemness were inhibited. Oligo-Fucoidan plus olaparib better inhibited the formation of TNBC stem cell mammospheroids with decreased subpopulations of CD44high/CD24low and EpCAMhigh cells than monotherapy. Importantly, Oligo-Fucoidan plus olaparib repressed the oncogenic interleukin-6 (IL-6)/p-EGFR/PD-L1 pathway, glucose uptake and lactate production. Oligo-Fucoidan induced immunoactive and antitumoral M1 macrophages and attenuated the side effects of olaparib, such as the promotion on immunosuppressive and protumoral M2 macrophages. Furthermore, olaparib plus Oligo-Fucoidan dramatically suppressed M2 macrophage invasiveness and repolarized M2 to the M0-like (F4/80high) and M1-like (CD80high and CD86high) phenotypes. In addition, olaparib- and Oligo-Fucoidan-pretreated TNBC cells resulted in the polarization of M0 macrophages into CD80(+) M1 but not CD163(+) M2 macrophages. Importantly, olaparib supplemented with oral administration of Oligo-Fucoidan in mice inhibited postsurgical TNBC recurrence and metastasis with increased cytotoxic T cells in the lymphatic system and decreased regulatory T cells and M2 macrophages in tumors. CONCLUSION Olaparib supplemented with natural compound Oligo-Fucoidan is a novel therapeutic strategy for reprogramming cancer stemness, metabolism and the microenvironment to prevent local postsurgical recurrence and distant metastasis. The combination therapy may advance therapeutic efficacy that prevent metastasis, chemoresistance and mortality in TNBC patients.
Collapse
|
7
|
Xi Y, Li T, Xi Y, Zeng X, Miao Y, Guo R, Zhang M, Li B. Combination treatment with hENT1 and miR-143 reverses gemcitabine resistance in triple-negative breast cancer. Cancer Cell Int 2022; 22:271. [PMID: 36050724 PMCID: PMC9438150 DOI: 10.1186/s12935-022-02681-0] [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: 10/14/2021] [Accepted: 08/11/2022] [Indexed: 12/24/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer and is susceptible to develop gemcitabine (GEM) resistance. Decreased expression of human equilibrative nucleoside transporter 1 (hENT1) accompanied by compensatory increase of glycolysis is strongly associated with GEM resistance in TNBC. In this study, we investigated the treatment feasibility of combined hENT1 upregulation and miR-143-mediated inhibition of glycolysis for reversing GEM resistance in TNBC. Methods Experiments were performed in vitro and in vivo to compare the efficacy of GEM therapies. In this study, we established stable drug-resistant cell line, GEM-R cells, from parental cells (MDA-MB-231) through exposure to GEM following a stepwise incremental dosing strategy. Then GEM-R cells were transfected by lentiviral plasmids and GEM-R cells overexpressing hENT1 (GEM-R-hENT1) were established. The viability and apoptosis of wild-type (MDA-MB-231), GEM-R, and GEM-R-hENT1 cells treated with GEM or GEM + miR-143 were analyzed by CCK8 assay and flow cytometry. The RNA expression and protein expression were measured by RT-PCR and western blotting respectively. GEM uptake was determined by multiple reaction monitoring (MRM) analysis. Glycolysis was measured by glucose assay and 18F-FDG uptake. The antitumor effect was assessed in vivo in a tumor xenograft model by evaluating toxicity, tumor volume, and maximum standardized uptake value in 18F-FDG PET. Immunohistochemistry and fluorescence photography were taken in tumor samples. Pairwise comparisons were performed using Student’s t-test. Results Our results represented that overexpression of hENT1 reversed GEM resistance in GEM-R cells by showing lower IC50 and higher rate of apoptosis. MiR-143 suppressed glycolysis in GEM-R cells and enhanced the effect of reversing GEM resistance in GEM-R-hENT1 cells. The therapeutic efficacy was validated using a xenograft mouse model. Combination treatment decreased tumor growth rate and maximum standardized uptake value in 18F-FDG PET more effectively. Conclusions Combined therapy of exogenous upregulation of hENT1 expression and miR-143 mimic administration was effective in reversing GEM resistance, providing a promising strategy for treating GEM-resistant TNBC.
Collapse
Affiliation(s)
- Yue Xi
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China
| | - Ting Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China
| | - Yun Xi
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China
| | - Xinyi Zeng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Miao
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China
| | - Rui Guo
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China
| | - Min Zhang
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China.
| | - Biao Li
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China. .,Collaboration Innovation Center for Molecular Imaging of Precision Medicine, Ruijin Center, Shanghai, 200025, China.
| |
Collapse
|
8
|
Chen X, Luo J, Yang L, Guo Y, Fan Y, Liu J, Sun J, Zhang Y, Jiang Q, Chen T, Xi Q. miR-143-Mediated Responses to Betaine Supplement Repress Lipogenesis and Hepatic Gluconeogenesis by Targeting MAT1a and MAPK11. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:7981-7992. [PMID: 35734958 DOI: 10.1021/acs.jafc.2c02940] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liver as the central organ is responsible for lipogenesis, gluconeogenesis and one-carbon metabolism. Methyl donors (e.g., betaine) modulate metabolic homeostasis and gene regulation through one-carbon metabolism. MiR-143 regulates DNA methylation by targeting DNMT3A, thereby suggesting that this miRNA participates in one-carbon metabolic pathways. However, the effect and mechanism that regulate glucose and lipid metabolism via the methyl group metabolism pathway remain elusive. In this study, we found that a betaine supplement and miR-143 KO significantly promoted lipolysis and glucose utilization and repressed lipogenesis and gluconeogenesis through enhancing energy consumption and thermogenesis, repressing GPNMB and targeting MAPK11, respectively. We further explored the relationship between miR-143 and a methyl donor (betaine) and the miR-143-mediated responses to the betaine supplement regulating the mechanism of the glucose and lipid metabolism. The results showed that betaine significantly down-regulated the expression of miR-143 that subsequently increased SAM levels in the liver by targeting MAT1a. In brief, the regulations of glucose and lipid metabolism are related to the miR-143-regulation of one-carbon units, and the relationship between betaine and miR-143 in the methionine cycle is a typical yin-yang type of regulation. Thus, betaine and miR-143 function together as key regulators and biomarkers for preventing and diagnosing metabolic diseases such as fatty liver disease, obesity, and diabetes.
Collapse
Affiliation(s)
- Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Lekai Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yue Guo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yaotian Fan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Jie Liu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, No. 483 Wushan Road, Guangzhou, 510642 China
| |
Collapse
|
9
|
Orlandella FM, Auletta L, Greco A, Zannetti A, Salvatore G. Preclinical Imaging Evaluation of miRNAs' Delivery and Effects in Breast Cancer Mouse Models: A Systematic Review. Cancers (Basel) 2021; 13:6020. [PMID: 34885130 PMCID: PMC8656589 DOI: 10.3390/cancers13236020] [Citation(s) in RCA: 4] [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: 10/13/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We have conducted a systematic review focusing on the advancements in preclinical molecular imaging to study the delivery and therapeutic efficacy of miRNAs in mouse models of breast cancer. METHODS A systematic review of English articles published in peer-reviewed journals using PubMed, EMBASE, BIOSIS™ and Scopus was performed. Search terms included breast cancer, mouse, mice, microRNA(s) and miRNA(s). RESULTS From a total of 2073 records, our final data extraction was from 114 manuscripts. The most frequently used murine genetic background was Balb/C (46.7%). The most frequently used model was the IV metastatic model (46.8%), which was obtained via intravenous injection (68.9%) in the tail vein. Bioluminescence was the most used frequently used tool (64%), and was used as a surrogate for tumor growth for efficacy treatment or for the evaluation of tumorigenicity in miRNA-transfected cells (29.9%); for tracking, evaluation of engraftment and for response to therapy in metastatic models (50.6%). CONCLUSIONS This review provides a systematic and focused analysis of all the information available and related to the imaging protocols with which to test miRNA therapy in an in vivo mice model of breast cancer, and has the purpose of providing an important tool to suggest the best preclinical imaging protocol based on available evidence.
Collapse
Affiliation(s)
| | - Luigi Auletta
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, 80145 Naples, Italy; (L.A.); (A.Z.)
| | - Adelaide Greco
- InterDepartmental Center of Veterinary Radiology, University of Naples Federico II, 80131 Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council, IBB-CNR, 80145 Naples, Italy; (L.A.); (A.Z.)
| | - Giuliana Salvatore
- IRCCS SDN, 80143 Naples, Italy;
- Department of Motor Sciences and Wellness, University of Naples Parthenope, 80133 Naples, Italy
- CEINGE-Biotecnologie Avanzate S.C.A.R.L., 80145 Naples, Italy
| |
Collapse
|
10
|
Haque S, Cook K, Sahay G, Sun C. RNA-Based Therapeutics: Current Developments in Targeted Molecular Therapy of Triple-Negative Breast Cancer. Pharmaceutics 2021; 13:pharmaceutics13101694. [PMID: 34683988 PMCID: PMC8537780 DOI: 10.3390/pharmaceutics13101694] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly heterogeneous and aggressive cancer that has the highest mortality rate out of all breast cancer subtypes. Conventional clinical treatments targeting ER, PR, and HER2 receptors have been unsuccessful in the treatment of TNBC, which has led to various research efforts in developing new strategies to treat TNBC. Targeted molecular therapy of TNBC utilizes knowledge of key molecular signatures of TNBC that can be effectively modulated to produce a positive therapeutic response. Correspondingly, RNA-based therapeutics represent a novel tool in oncology with their ability to alter intrinsic cancer pathways that contribute to poor patient prognosis. Current RNA-based therapeutics exist as two major areas of investigation-RNA interference (RNAi) and RNA nanotherapy, where RNAi utilizes principles of gene silencing, and RNA nanotherapy utilizes RNA-derived nanoparticles to deliver chemotherapeutics to target cells. RNAi can be further classified as therapeutics utilizing either small interfering RNA (siRNA) or microRNA (miRNA). As the broader field of gene therapy has advanced significantly in recent years, so too have efforts in the development of effective RNA-based therapeutic strategies for treating aggressive cancers, including TNBC. This review will summarize key advances in targeted molecular therapy of TNBC, describing current trends in treatment using RNAi, combination therapies, and recent efforts in RNA immunotherapy, utilizing messenger RNA (mRNA) in the development of cancer vaccines.
Collapse
Affiliation(s)
- Sakib Haque
- College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (S.H.); (G.S.)
| | - Kiri Cook
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Gaurav Sahay
- College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (S.H.); (G.S.)
| | - Conroy Sun
- College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; (S.H.); (G.S.)
- Department of Radiation Medicine, Oregon Health & Science University, Portland, OR 97239, USA;
- Correspondence: ; Tel.: +1-503-346-4699
| |
Collapse
|
11
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
12
|
Amelioration of cyclophosphamide-induced myelosuppression during treatment to rats with breast cancer through low-intensity pulsed ultrasound. Biosci Rep 2020; 40:226432. [PMID: 32936241 PMCID: PMC7517537 DOI: 10.1042/bsr20201350] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/02/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Abstract
To investigate the alleviating effects of low-intensity pulsed ultrasound (LIPUS) on myelosuppression of Sprague–Dawley rats with breast cancer induced by cyclophosphamide (CTX). Breast cancer in rats was triggered by intragastric gavage with 7,12-dimethylbenz[a]anthracene (150 mg/kg). Then, the rats with breast cancer were randomly allocated to the LIPUS group (n=50) and the control group (n=50). The LIPUS group was injected intraperitoneally with CTX (50 mg/kg) for 4 consecutive days and underwent LIPUS treatment at femoral metaphysis 20 min per day from the first day of injection for 7 consecutive days. The control group was injected with CTX (50 mg/kg) and treated with LIPUS without energy output. Blood, enzyme-linked immunosorbent assay (ELISA), real-time quantitative polymerase chain reaction, Hematoxylin and Eosin (H&E) staining, and scanning electron microscopy were applied to detect the changes. The results indicated that LIPUS significantly promoted the proliferation of bone marrow nucleated cells, white blood cells (WBCs), IgA, IgG, and IgM in the peripheral blood (P<0.05) without the damage to liver and kidney function simultaneously. The mechanisms may result from the LIPUS alleviation effect on bone marrow hematopoietic function through regulating cytokines such as LIPUS can increase the expression of granulocyte colony-stimulating factor (G-CSF), stem cell factor, transforming growth factor-β, and intercellular cell adhesion molecule-1, meanwhile LIPUS will decrease the expression of interleukin-6, tumor necrosis factor-α, and vascular cell adhesion molecule-1. LIPUS has potential to be a new adjuvant therapy method in clinic for ameliorating chemotherapy-induced myelosuppression.
Collapse
|
13
|
Samec M, Liskova A, Koklesova L, Samuel SM, Zhai K, Buhrmann C, Varghese E, Abotaleb M, Qaradakhi T, Zulli A, Kello M, Mojzis J, Zubor P, Kwon TK, Shakibaei M, Büsselberg D, Sarria GR, Golubnitschaja O, Kubatka P. Flavonoids against the Warburg phenotype-concepts of predictive, preventive and personalised medicine to cut the Gordian knot of cancer cell metabolism. EPMA J 2020; 11:377-398. [PMID: 32843908 PMCID: PMC7429635 DOI: 10.1007/s13167-020-00217-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 06/30/2020] [Indexed: 01/10/2023]
Abstract
The Warburg effect is characterised by increased glucose uptake and lactate secretion in cancer cells resulting from metabolic transformation in tumour tissue. The corresponding molecular pathways switch from oxidative phosphorylation to aerobic glycolysis, due to changes in glucose degradation mechanisms known as the 'Warburg reprogramming' of cancer cells. Key glycolytic enzymes, glucose transporters and transcription factors involved in the Warburg transformation are frequently dysregulated during carcinogenesis considered as promising diagnostic and prognostic markers as well as treatment targets. Flavonoids are molecules with pleiotropic activities. The metabolism-regulating anticancer effects of flavonoids are broadly demonstrated in preclinical studies. Flavonoids modulate key pathways involved in the Warburg phenotype including but not limited to PKM2, HK2, GLUT1 and HIF-1. The corresponding molecular mechanisms and clinical relevance of 'anti-Warburg' effects of flavonoids are discussed in this review article. The most prominent examples are provided for the potential application of targeted 'anti-Warburg' measures in cancer management. Individualised profiling and patient stratification are presented as powerful tools for implementing targeted 'anti-Warburg' measures in the context of predictive, preventive and personalised medicine.
Collapse
Affiliation(s)
- Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Constanze Buhrmann
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011 Australia
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3011 Australia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, P. J. Šafarik University, 040 11 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, P. J. Šafarik University, 040 11 Košice, Slovakia
| | - Pavol Zubor
- Department of Gynecologic Oncology, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- OBGY Health & Care, Ltd., 01001 Zilina, Slovak Republic
| | - Taeg Kyu Kwon
- Department of Immunology and School of Medicine, Keimyung University, Dalseo-Gu, Daegu, 426 01 South Korea
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumour Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine in Qatar, Education City, Qatar Foundation, 24144, Doha, Qatar
| | - Gustavo R. Sarria
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Olga Golubnitschaja
- Predictive, Preventive Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
14
|
Otoukesh B, Abbasi M, Gorgani HOL, Farahini H, Moghtadaei M, Boddouhi B, Kaghazian P, Hosseinzadeh S, Alaee A. MicroRNAs signatures, bioinformatics analysis of miRNAs, miRNA mimics and antagonists, and miRNA therapeutics in osteosarcoma. Cancer Cell Int 2020; 20:254. [PMID: 32565738 PMCID: PMC7302353 DOI: 10.1186/s12935-020-01342-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) involved in key signaling pathways and aggressive phenotypes of osteosarcoma (OS) was discussed, including PI3K/AKT/MTOR, MTOR AND RAF-1 signaling, tumor suppressor P53- linked miRNAs, NOTCH- related miRNAs, miRNA -15/16 cluster, apoptosis related miRNAs, invasion-metastasis-related miRNAs, and 14Q32-associated miRNAs cluster. Herrin, we discussed insights into the targeted therapies including miRNAs (i.e., tumor-suppressive miRNAs and oncomiRNAs). Using bioinformatics tools, the interaction network of all OS-associated miRNAs and their targets was also depicted.
Collapse
Affiliation(s)
- Babak Otoukesh
- Orthopedic Surgery Fellowship in Département Hospitalo-Universitaire MAMUTH « Maladies musculo-squelettiques et innovations thérapeutiques » , Université Pierre et Marie-Curie, Sorbonne Université, Paris, France.,Department of Orthopedic Surgery, Bone and Joint Reconstruction Research Center, Iran University of Medical Science, Postal code : 1445613131 Tehran, Iran
| | - Mehdi Abbasi
- Brain Mapping Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib-O-Lah Gorgani
- Department of Orthopedic Surgery, Bone and Joint Reconstruction Research Center, Iran University of Medical Science, Postal code : 1445613131 Tehran, Iran
| | - Hossein Farahini
- Department of Orthopedic Surgery, Bone and Joint Reconstruction Research Center, Iran University of Medical Science, Postal code : 1445613131 Tehran, Iran
| | - Mehdi Moghtadaei
- Department of Orthopedic Surgery, Bone and Joint Reconstruction Research Center, Iran University of Medical Science, Postal code : 1445613131 Tehran, Iran
| | - Bahram Boddouhi
- Department of Orthopedic Surgery, Bone and Joint Reconstruction Research Center, Iran University of Medical Science, Postal code : 1445613131 Tehran, Iran
| | - Peyman Kaghazian
- Department of Orthopedic and Traumatology, Universitätsklinikum Bonn, Bonn, Germany
| | - Shayan Hosseinzadeh
- Department of Orthopedic Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA USA
| | - Atefe Alaee
- Department of Information Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Xia M, Feng S, Chen Z, Wen G, Zu X, Zhong J. Non-coding RNAs: Key regulators of aerobic glycolysis in breast cancer. Life Sci 2020; 250:117579. [PMID: 32209425 DOI: 10.1016/j.lfs.2020.117579] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/04/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022]
Abstract
Although extensive research progress has been made in breast cancer in recent years, yet the morbidity and mortality rates of breast cancer are rising, making it the major disease that endangers women's health. Energy metabolism reprogramming is featured by a state termed "aerobic glycolysis" or the Warburg effect that glycolysis is preferred even under aerobic conditions in neoplastic diseases. Widely acknowledged as an emerging hallmark in cancers, this metabolic switch shows a sophisticated role in the pathogenesis of breast cancer. The regulating effect of non-coding RNAs (ncRNAs) composed of microRNAs, long non-coding RNAs and circular RNAs is closely related to the glycolysis in breast cancer. Therefore, understand the mechanisms of ncRNAs of aerobic glycolysis in breast cancer may provide new strategy for the disease.
Collapse
Affiliation(s)
- Min Xia
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Department of Metabolism and Endocrinology, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Shujun Feng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, University of South China
| | - Zuyao Chen
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Gebo Wen
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Department of Metabolism and Endocrinology, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China
| | - Xuyu Zu
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China.
| | - Jing Zhong
- Institute of Clinical Medicine, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China; Cancer Research Institute, the First Affiliated Hospital of University of South China, Hengyang, Hunan 421001, PR China.
| |
Collapse
|
16
|
Hu GF, Wang C, Hu GX, Wu G, Zhang C, Zhu W, Chen C, Gu Y, Zhang H, Yang Z. AZD3463, an IGF-1R inhibitor, suppresses breast cancer metastasis to bone via modulation of the PI3K-Akt pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:336. [PMID: 32355780 PMCID: PMC7186597 DOI: 10.21037/atm.2020.02.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The bone-derived insulin-like growth factor I (IGF-1) and its receptor IGF-1R play a crucial role in promoting the survival and proliferation of cancer cells, and have thus been considered as prime targets for the development of novel antitumor therapeutics. Methods By using the MDA-MB-231BO cell line, which is the osteotropic metastatic variant of the human breast adenocarcinoma cell line MDA-MB-231, and an in vivo model of breast cancer metastasis to bone, the current study evaluated the effect of AZD3463, an IGF-1R inhibitor, used alone or in combination with zoledronic acid (ZA), on the regulation of IGF-1R associated signal pathway and treatment of bone metastases (BM). Cell proliferation and invasion were measured by methyl thiazolyl tetrazolium (MTT) and Transwell assay respectively. Apoptotic cell number was detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL). Results AZD3463 was shown to alleviate IGF-1R phosphorylation promoted by IGF-1 treatment in MDA-MB-231BO cells in a dose-dependent manner. In both the cells and the mouse model, 5 nM of AZD3463 stimulated cell apoptosis and suppressed proliferation on a level similar to that of 100 µM of ZA. Remarkably, the combined use of AZD3463 and ZA exhibited a synergistic effect and greater antitumor activity compared to when they were employed individually. Mechanistic investigations indicated that the apoptosis-inducing activity of AZD3463 could be associated to its role in the activation of the phosphoinositide 3-kinase (PI3K)-Akt signaling pathway. Conclusions These findings suggested that AZD3463 could serve as a promising therapeutic molecule for treating BM in breast cancer patients, particularly when applied in conjunction with ZA or other antitumor agents.
Collapse
Affiliation(s)
- Guang-Fu Hu
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Cheng Wang
- Department of Breast Surgery, Huangpu Branch, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Guang-Xia Hu
- Department of Pathology, Binzhong People's Hospital, Binzhong 256600, China
| | - Ge Wu
- Department of Pathology, Binzhong People's Hospital, Binzhong 256600, China
| | - Chengjiao Zhang
- Department of Psychological Measurement, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 20030, China
| | - Wei Zhu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Cong Chen
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Yutong Gu
- Department of Orthopaedic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China.,Department of Orthopaedic Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Hongwei Zhang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Zi'ang Yang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Bae SW, Berlth F, Jeong KY, Suh YS, Kong SH, Lee HJ, Kim WH, Chung JK, Yang HK. Establishment of a [ 18F]-FDG-PET/MRI Imaging Protocol for Gastric Cancer PDX as a Preclinical Research Tool. J Gastric Cancer 2020; 20:60-71. [PMID: 32269845 PMCID: PMC7105419 DOI: 10.5230/jgc.2020.20.e7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/29/2019] [Accepted: 01/13/2020] [Indexed: 12/27/2022] Open
Abstract
Purpose The utility of 18-fluordesoxyglucose positron emission tomography ([18F]-FDG-PET) combined with computer tomography or magnetic resonance imaging (MRI) in gastric cancer remains controversial and a rationale for patient selection is desired. This study aims to establish a preclinical patient-derived xenograft (PDX) based [18F]-FDG-PET/MRI protocol for gastric cancer and compare different PDX models regarding tumor growth and FDG uptake. Materials and Methods Female BALB/c nu/nu mice were implanted orthotopically and subcutaneously with gastric cancer PDX. [18F]-FDG-PET/MRI scanning protocol evaluation included different tumor sizes, FDG doses, scanning intervals, and organ-specific uptake. FDG avidity of similar PDX cases were compared between ortho- and heterotopic tumor implantation methods. Microscopic and immunohistochemical investigations were performed to confirm tumor growth and correlate the glycolysis markers glucose transporter 1 (GLUT1) and hexokinase 2 (HK2) with FDG uptake. Results Organ-specific uptake analysis showed specific FDG avidity of the tumor tissue. Standard scanning protocol was determined to include 150 μCi FDG injection dose and scanning after one hour. Comparison of heterotopic and orthotopic implanted mice revealed a long growth interval for orthotopic models with a high uptake in similar PDX tissues. The H-score of GLUT1 and HK2 expression in tumor cells correlated with the measured maximal standardized uptake value values (GLUT1: Pearson r=0.743, P=0.009; HK2: Pearson r=0.605, P=0.049). Conclusions This preclinical gastric cancer PDX based [18F]-FDG-PET/MRI protocol reveals tumor specific FDG uptake and shows correlation to glucose metabolic proteins. Our findings provide a PET/MRI PDX model that can be applicable for translational gastric cancer research.
Collapse
Affiliation(s)
- Seong-Woo Bae
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Felix Berlth
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Seoul National University Hospital, Seoul, Korea.,Department of General, Visceral and Transplant Surgery, University of Mainz, Mainz, Germany
| | - Kyoung-Yun Jeong
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Hyuk-Joon Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Seoul National University Hospital, Seoul, Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - June-Key Chung
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea
| | - Han-Kwang Yang
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.,Department of Surgery, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
18
|
Jiang Q, Yuan Y, Gong Y, Luo X, Su X, Hu X, Zhu W. Therapeutic delivery of microRNA-143 by cationic lipoplexes for non-small cell lung cancer treatment in vivo. J Cancer Res Clin Oncol 2019; 145:2951-2967. [PMID: 31654121 DOI: 10.1007/s00432-019-03051-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths worldwide and new improvements are urgently needed. Several miRNA-targeted therapeutics have reached clinical development. MicroRNA-143 (miR-143) was found to significantly suppress the migration and invasion of NSCLC. It might be of great potential for NSCLC treatment. However, the therapeutic effect of miR-143 against NSCLC in vivo has not been explored until now. METHODS The cationic liposome/pVAX-miR-143 complex (CL-pVAX-miR-143) was prepared and its biodistribution was assessed. The tumor suppression effects of CL-pVAX-miR-143 were evaluated in early-stage and advanced experimental lung cancer metastasis mice models by systemic delivery, respectively, and also in subcutaneous tumor models by intratumoral injection. The toxicity of CL-pVAX-miR-143 was assessed by H&E analysis and biochemical measurements. The preliminary mechanism of CL-pVAX-miR-143 on tumor suppression was explored by immunochemistry and western blotting. RESULTS The assays on the stability and safety of CL-pVAX-miR-143 showed that it mainly accumulated in the lung after systemic administration. The intratumoral delivery of CL-pVAX-miR-143 effectively inhibited A549 subcutaneous tumor growth. Notably, systemic delivery of CL-pVAX-miR-143 significantly inhibited tumor metastasis and prolonged survival dose dependently in early-stage experimental lung cancer metastasis models. More importantly, same results were shown in advanced mice models with metastasis. CL-pVAX-miR-143 treatment did not induce obvious acute toxicity. The preliminary mechanism on inhibiting tumor metastasis might be induced by targeting CD44v3. CONCLUSIONS Our results suggested that CL-pVAX-miR-143 might be a promising strategy for clinical treatment of non-small cell lung cancer, especially for advanced NSCLC with metastasis.
Collapse
Affiliation(s)
- Qianqian Jiang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Yue Yuan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Yi Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Xinmei Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Xueting Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China
| | - Wen Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/Collaborative Innovation Center of Biotherapy, No. 1, Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
19
|
18F-FDG PET imaging for monitoring the early anti-tumor effect of albendazole on triple-negative breast cancer. Breast Cancer 2019; 27:372-380. [DOI: 10.1007/s12282-019-01027-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/21/2019] [Indexed: 01/01/2023]
|
20
|
Mesgarzadeh AH, Aali M, Farhadi F, Noorolyai S, Baghbani E, Mohammadnejad F, Baradaran B. Transfection of microRNA-143 mimic could inhibit migration of HN-5 cells through down-regulating of metastatic genes. Gene 2019; 716:144033. [DOI: 10.1016/j.gene.2019.144033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 11/17/2022]
|
21
|
Long non-coding RNA 520 is a negative prognostic biomarker and exhibits pro-oncogenic function in nasopharyngeal carcinoma carcinogenesis through regulation of miR-26b-3p/USP39 axis. Gene 2019; 707:44-52. [DOI: 10.1016/j.gene.2019.02.093] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
|
22
|
Miao Y, Zhang LF, Zhang M, Guo R, Liu MF, Li B. Therapeutic Delivery of miR-143 Targeting Tumor Metabolism in Poorly Differentiated Thyroid Cancer Xenografts and Efficacy Evaluation Using 18F-FDG MicroPET-CT. Hum Gene Ther 2019; 30:882-892. [PMID: 30848162 DOI: 10.1089/hum.2018.160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Poorly differentiated thyroid carcinoma cells tend to be more aggressive and show enhanced glucose uptake which could be exploited for anti-cancer strategy. Previously, we identified hexokinase 2 (HK2) as a direct target of miR-143. In our current study, the effects of miR-143 on glucose metabolism and tumor biological behavior were investigated in FTC-133 cells which is a poorly differentiated thyroid carcinoma (PDTC). Additionally, tumor-bearing mice xenografts of PDTC were constructed, with encapsulated miR-143 agomir being administered intravenously. 18F-FDG microPET-CT scanning was used for the evaluation of therapeutic efficacy. The tumor-restrained effect of miR-143 was demonstrated in PDTC. Furthermore, microPET/CT imaging exhibited a reduction of 18F-FDG uptake in tumors, corresponding to the downregulated expression of HK2 in tissues. In summary, our results suggest that miR-143 can be an alternative treatment for PDTC and the specific assessment of therapeutic response to miR-143 can be achieved by 18F-FDG microPET/CT in advanced thyroid carcinoma xenografts.
Collapse
Affiliation(s)
- Ying Miao
- 1Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling-Fei Zhang
- 2Center for RNA Research, State Key Laboratory of Molecular Biology, University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China
- 3Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Zhang
- 1Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Guo
- 1Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mo-Fang Liu
- 2Center for RNA Research, State Key Laboratory of Molecular Biology, University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China
- 3Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- 4School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Biao Li
- 1Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
23
|
The miR-186-3p/EREG axis orchestrates tamoxifen resistance and aerobic glycolysis in breast cancer cells. Oncogene 2019; 38:5551-5565. [PMID: 30967627 DOI: 10.1038/s41388-019-0817-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/10/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023]
Abstract
Tamoxifen resistance is one of the major challenges for its medical uses in estrogen receptor (ER)-positive breast cancer. Aerobic glycolysis, an anomalous characteristic of glucose metabolism in cancer cells, has been shown to associate with the resistance to chemotherapeutic agents. It remains, however, largely unclear whether and how tamoxifen resistance contributes to aerobic glycolysis in breast cancer. Here, we report that tamoxifen resistance is associated with enhanced glycolysis in ER-positive breast cancer cells. We demonstrate that EREG, an agonist of EGFR, has an important role in enhancing glycolysis via activating EGFR signaling and its downstream glycolytic genes in tamoxifen-resistant breast cancer cells. We further show that EREG is a direct target of miR-186-3p and that downregulation of miR-186-3p by tamoxifen results in EREG upregulation in tamoxifen-resistant breast cancer cells. Importantly, systemic delivery of cholesterol-modified agomiR-186-3p to mice bearing tamoxifen-resistant breast tumors effectively attenuates both tumor growth and [18F]-fluoro-deoxyglucose ([18F]-FDG) uptake. Together, our results reveal a novel molecular mechanism of resistance to hormone therapies in which the miR-186-3p/EREG axis orchestrates tamoxifen resistance and aerobic glycolysis in ER-positive breast cancer, suggesting targeting miR-186-3p as a promising strategy for therapeutic intervention in endocrine-resistant breast tumors.
Collapse
|
24
|
LncRNA LINC00460 promotes tumor growth of human lung adenocarcinoma by targeting miR-302c-5p/FOXA1 axis. Gene 2019; 685:76-84. [DOI: 10.1016/j.gene.2018.10.058] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/19/2018] [Accepted: 10/20/2018] [Indexed: 12/18/2022]
|
25
|
Long non-coding RNA 319 facilitates nasopharyngeal carcinoma carcinogenesis through regulation of miR-1207-5p/KLF12 axis. Gene 2019; 680:51-58. [DOI: 10.1016/j.gene.2018.09.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/18/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
|
26
|
Zhang T, Zhang Z, Li F, Ping Y, Qin G, Zhang C, Zhang Y. miR-143 Regulates Memory T Cell Differentiation by Reprogramming T Cell Metabolism. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2165-2175. [PMID: 30150287 DOI: 10.4049/jimmunol.1800230] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/28/2018] [Indexed: 12/31/2022]
Abstract
MicroRNAs are an important regulator for T cell immune response. In this study, we aimed to identify microRNAs with the potential to regulate T cell differentiation. The influence of miR-143 on differentiation and function of CD8+ T cells from healthy donors were detected, and it was found that miR-143 overexpression could significantly increase the differentiation of central memory T (Tcm) CD8+ cells, decrease cell apoptosis, and increase proinflammatory cytokine secretion. Furthermore, the specific killing of HER2-CAR T cells against esophageal cancer cell line TE-7 was enhanced by miR-143 overexpression. Glucose transporter 1 (Glut-1) was identified as the critical target gene of miR-143 in the role of T cell regulation. By inhibition Glut-1, miR-143 inhibited glucose uptake and glycolysis in T cell to regulated T cell differentiation. Tcm cell populations were also suppressed in parallel with the downregulation of miR-143 in tumor tissues from 13 patients with esophagus cancer. IDO and its metabolite kynurenine in the tumor microenvironment were screened as an upstream regulator of miR-143. IDO small interfering RNA significantly increased the expression of miR-143 and Tcm cell population. In conclusion, our results show that miR-143 enhanced antitumor effects of T cell by promoting memory T cell differentiation and metabolism reprogramming through Glut-1. Our findings will encourage the development of new strategies targeting miR-143 in both cancer cells and T cells.
Collapse
Affiliation(s)
- Tengfei Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Zhen Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Feng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Guohui Qin
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chaoqi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China;
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan 450052, China; and
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan 450052, China
| |
Collapse
|
27
|
Guda MR, Asuthkar S, Labak CM, Tsung AJ, Alexandrov I, Mackenzie MJ, Prasad DVR, Velpula KK. Targeting PDK4 inhibits breast cancer metabolism. Am J Cancer Res 2018; 8:1725-1738. [PMID: 30323966 PMCID: PMC6176187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023] Open
Abstract
Dysregulated metabolism in the form of aerobic glycolysis occurs in many cancers including breast carcinoma. Here, we report PDK4 (pyruvate dehydrogenase kinase 4) as key enzyme implicated in the control of glucose metabolism and mitochondrial respiration is relatively highly expressed in breast cancers, and its expression correlates with poor patient outcomes. Silencing of PDK4 and ectopic expression of miR-211 attenuates PDK4 expression in breast cancer cells. Interestingly, low miR-211 expression is significantly associated with shorter overall survival and reveals an inverse correlation between expression of miR-211 and PDK4. We have found that depletion of PDK4 by miR-211 shows an oxidative phosphorylation-dominant phenotype consisting of the reduction of glucose with increased expression of PDH and key enzymes of the TCA cycle. miR-211 expression causes alteration of mitochondrial membrane potential and induces mitochondrial apoptosis as observed via IPAD assay. Further, by inhibiting PDK4 expression, miR-211 promotes a phenotype shift towards a pro-glycolytic state evidenced by decreased extracellular acidification rate (ECAR); increased oxygen consumption rate (OCR); and increased spare respiratory capacity in breast cancer cell lines. Taken together this data establishes a molecular connection between PDK4 and miR-211 and suggests that targeting miR-211 to inhibit PDK4 could represent a novel therapeutic strategy in breast cancers.
Collapse
Affiliation(s)
- Maheedhara R Guda
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Swapna Asuthkar
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Collin M Labak
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
| | - Andrew J Tsung
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Illinois Neurological InstitutePeoria, IL, USA
| | | | | | | | - Kiran K Velpula
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Neurosurgery, University of Illinois College of Medicine at PeoriaPeoria, IL, USA
- Department of Microbiology, Yogi Vemana UniversityKadapa, India
| |
Collapse
|
28
|
Zhang ZW, Chen JJ, Xia SH, Zhao H, Yang JB, Zhang H, He B, Jiao J, Zhan BT, Sun CC. Long intergenic non-protein coding RNA 319 aggravates lung adenocarcinoma carcinogenesis by modulating miR-450b-5p/EZH2. Gene 2018; 650:60-67. [DOI: 10.1016/j.gene.2018.01.096] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 01/11/2023]
|
29
|
Sun L, Zhai R, Zhang L, Zhao S. MicroRNA-149 suppresses the proliferation and increases the sensitivity of ovarian cancer cells to cisplatin by targeting X-linked inhibitor of apoptosis. Oncol Lett 2018; 15:7328-7334. [PMID: 29731888 DOI: 10.3892/ol.2018.8240] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022] Open
Abstract
Currently, ovarian cancer is identified as one of the leading causes of cancer-associated mortality in females. Despite numerous efforts that were made on developing novel treatments for ovarian cancer, the survival rate remains unsatisfactory. Considering the important regulatory role of miRNAs in different types of cancer, the present study aims to identify a novel therapeutic target for treatment of ovarian cancer. The expression of miR-149 was detected using reverse transcription-quantitative polymerase chain reaction in cancerous and normal cells. Furthermore, the effects of miR-149 on ovarian cancer cell activities were investigated using MTT assay, colony formation, flow cytometry and western blotting analysis. In the present study, it was revealed that microRNA (miR)-149 was significantly downregulated in ovarian cancer tissues and cell lines, and that the miR-149 expression was correlated with the patient prognosis. In addition, it was observed that forced expression of miR-149 increased the sensitivity of ovarian cancer cell to cisplatin. Based on bioinformatics analysis and luciferase assay, X-linked inhibitor of apoptosis (XIAP) was identified as a direct target gene of miR-149 in ovarian cancer cells. It was also demonstrated that XIAP expression was upregulated in the ovarian cancer tissues and cell lines, while it was negatively correlated with miR-149 in these tissues and cells. Furthermore, results revealed that ectopic expression of XIAP was able to abolish the miR-149-enhanced cell sensitivity to cisplatin. In conclusion, the present study revealed that miR-149 functioned as a tumor suppressor in the progression of ovarian cancer, increasing the sensitivity of ovarian cancer cells to cisplatin treatment.
Collapse
Affiliation(s)
- Lin Sun
- Department of Gynecology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China.,Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Ruixia Zhai
- Department of Obstetrics, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Li Zhang
- Department of Gynecology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272100, P.R. China
| | - Shuping Zhao
- Department of Gynecology, Qingdao Women and Children's Hospital of Qingdao University, Qingdao, Shandong 266034, P.R. China
| |
Collapse
|
30
|
Chen G, Sun W, Hua X, Zeng W, Yang L. Long non-coding RNA FOXD2-AS1 aggravates nasopharyngeal carcinoma carcinogenesis by modulating miR-363-5p/S100A1 pathway. Gene 2018; 645:76-84. [DOI: 10.1016/j.gene.2017.12.026] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/06/2017] [Accepted: 12/13/2017] [Indexed: 12/29/2022]
|
31
|
Kong YG, Cui M, Chen SM, Xu Y, Xu Y, Tao ZZ. LncRNA-LINC00460 facilitates nasopharyngeal carcinoma tumorigenesis through sponging miR-149-5p to up-regulate IL6. Gene 2017; 639:77-84. [PMID: 28987345 DOI: 10.1016/j.gene.2017.10.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/30/2017] [Accepted: 10/04/2017] [Indexed: 01/17/2023]
Abstract
Long non-coding RNAs (lncRNAs) have played crucial roles in various cancers, including nasopharyngeal carcinoma (NPC). In our study, we focused on the biological function and clinical significance of lncRNA LINC00460 in NPC. It was indicated that LINC00460 was markedly increased in NPC tissues and cells compared to their corresponding controls. Silencing LINC00460 was able to suppress NPC cell growth in vitro while overexpressing LINC00460 reversed this process. Moreover, in vivo tumor xenografts were established using CNE-1/SUNE-1 cells to detect the function of LINC00460 in NSCLC tumorigenesis. Rescue assay was performed to further confirm that LINC00460 contributed to the progression of NPC through regulating miR-149-5p/IL6 signal pathway. In conclusion, we have uncovered that LINC00460 could be regarded as a novel prognostic biomarker and therapeutic target in NPC diagnosis and treatment.
Collapse
Affiliation(s)
- Yong-Gang Kong
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Min Cui
- Department of Orthopaedic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shi-Ming Chen
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Yong Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China
| | - Ze-Zhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, People's Republic of China.
| |
Collapse
|
32
|
Xu P, Li Y, Yang S, Li M, Li C. Positron Emission Tomographic Imaging Elucidates the Complex Relationship Between Glucose Uptake and Tissue Blood Flow Mechanism in Squamous Cell Oral Cancer Patients. Med Sci Monit 2017; 23:4533-4540. [PMID: 28934176 PMCID: PMC5621790 DOI: 10.12659/msm.903974] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Through the clinical use of positron emission tomography, we aimed to elucidate the complex relationship between glucose uptake and squamous cell oral cancer (ScOC) growth, along with its mechanism with respect to tissue blood flow (tBF). MATERIAL AND METHODS We retrospectively reviewed a total of 69 newly diagnosed ScOC patients by Fluorine-18 fluorodeoxyglucose (18F-FDG) positron emission tomography (PET). Maximum and mean standard uptake values (SUV↑ and SUV) were recorded to assess glucose uptake. Multi-shot spin-echo echo-planar imaging-based pseudo-continuous arterial spin labeling (pcASL) technique at 3.0 T MRI was used to obtain tBF values in ScOC (tBF-ScOC). Patients were divided according to T-stage and location. Pearson's correlation coefficients were calculated between both SUV and tBF-ScOC for significant correlations. RESULTS Forty-one (59.4%) patients had oropharynx and the other 28 (40.6%) patients had laryngopharynx. Significant positive correlations were detected between SUV↑, SUV, tBF-ScOC and non-advanced T-stage (T1a, T1b, T2 and T3), while a negative correlation was observed in the advanced T-stage (T4a and T4b). CONCLUSIONS Using PET imaging, we established the relationship between glucose uptake and ScOC growth on the basis of the division of T-stage and tumor location of ScOC, thereby elucidating the underlying mechanism. Our findings provide insights important to the diagnosis, treatment, and care of ScOC patients.
Collapse
Affiliation(s)
- Ping Xu
- Department of Stomatology, Chengdu Military General Hospital, Chengdu, Sichuan, China (mainland)
| | - Yan Li
- Department of Stomatology, Chengdu Military General Hospital, Chengdu, Sichuan, China (mainland)
| | - Shuyong Yang
- Department of Stomatology, Chengdu Military General Hospital, Chengdu, Sichuan, China (mainland)
| | - Mingzhe Li
- Department of Stomatology, Chengdu Military General Hospital, Chengdu, Sichuan, China (mainland)
| | - Chenjun Li
- Department of Stomatology, Chengdu Military General Hospital, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
33
|
Hong H, Tao T, Chen S, Liang C, Qiu Y, Zhou Y, Zhang R. MicroRNA-143 promotes cardiac ischemia-mediated mitochondrial impairment by the inhibition of protein kinase Cepsilon. Basic Res Cardiol 2017; 112:60. [PMID: 28887629 DOI: 10.1007/s00395-017-0649-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
The cardioprotection of protein kinase Cepsilon (PKCε) against myocardial infarction (MI) mediated by its anti-apoptotic property and underlying mechanism of targeted regulation by microRNA (miRNA) are not established. MI-induced injury, PKCε expression, and targeted regulation of miRNA-143 (miR-143) to PKCε have been evaluated using animal MI and cellular hypoxic models conjugated with series of state-of-art molecular techniques. The results demonstrated that PKCε significantly downregulated along with increased infarcted area and apoptotic and necrotic damage in MI model, and the targeted relationship and potential binding profile were established between miR-143 and PKCε. Both in vivo and in vitro ischemic tests showed that miR-143 induced apoptosis and necrosis, which was reversed by antagomiR-143 or AMO-143. The upregulation of miR-143 by transfection of miR-143 in vitro also induced cell loss, and this effect of miR-143 was completely reversed by co-transfection of miR-143 with AMO-143. The identically deleterious action of miR-143 on mitochondrial membrane potential and ATP synthesis was also observed in both animal MI and cellular hypoxic models, as well as miR-143 overexpressed models and converted by either antagomiR or AMO. Importantly, overexpression of miR-143 downregulated PKCε in all tested models and this downregulation was reversed in the presence of antagomiR or AMO. The direct targeted regulation of miR-143 on PKCε was confirmed by luciferase reporter and miRNA-masking tests. In conclusion, MI-mediated upregulation of miR-143 inhibits PKCε expression and consequently interference with the cardioprotection of PKCε to mitochondrial, and leads to mitochondrial membrane potential dissipation and myocardial death eventually.
Collapse
Affiliation(s)
- Hong Hong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ting Tao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Si Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chaoqi Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yue Qiu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yuhong Zhou
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Rong Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
34
|
Zhang LF, Jiang S, Liu MF. MicroRNA regulation and analytical methods in cancer cell metabolism. Cell Mol Life Sci 2017; 74:2929-2941. [PMID: 28321489 PMCID: PMC11107497 DOI: 10.1007/s00018-017-2508-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 02/28/2017] [Accepted: 03/14/2017] [Indexed: 12/19/2022]
Abstract
The reprogramming of glucose metabolism from oxidative to glycolytic metabolism, known as the Warburg effect, is an anomalous characteristic of cancer cell metabolism. Recent studies have revealed a subset of microRNAs (miRNAs) that play critical roles in regulating the reprogramming of glucose metabolism in cancer cells. These miRNAs regulate cellular glucose metabolism by directly targeting multiple metabolic genes, including those encoding key glycolytic enzymes. In the first part of this review, we summarized the recent knowledge of miRNA regulation in the reprogramming of glucose metabolism in cancer cells and discussed the potential utilization of the key miRNA regulators as metabolic targets for developing new antitumor agents. Then, we summarized recent advances in methods and techniques for studying miRNA regulation in cancer cell metabolism.
Collapse
Affiliation(s)
- Ling-Fei Zhang
- Center for RNA Research, State Key Laboratory of Molecular Biology, University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China
- Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Shuai Jiang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Mo-Fang Liu
- Center for RNA Research, State Key Laboratory of Molecular Biology, University of Chinese Academy of Sciences, CAS Center for Excellence in Molecular Cell Science, Shanghai, China.
- Shanghai Key Laboratory of Molecular Andrology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 200031, China.
| |
Collapse
|
35
|
Dong X, Lv B, Li Y, Cheng Q, Su C, Yin G. MiR-143 regulates the proliferation and migration of osteosarcoma cells through targeting MAPK7. Arch Biochem Biophys 2017; 630:47-53. [PMID: 28734729 DOI: 10.1016/j.abb.2017.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 07/04/2017] [Accepted: 07/18/2017] [Indexed: 12/12/2022]
Abstract
Accumulating documents have been suggested that microRNA-143 (miR-143) function as a tumor suppressor, involved in many biological processes including tumor initiation and progression. However, the biological function and molecular mechanism of miR-143 in Osteosarcoma (OS) still remains to be further investigated. Despite many efforts have been made, the prognosis of OS is still unsatisfied. Thus, exploring the underlying mechanism of OS and finding new treatment targets is essential for improving the survival rate of OS patients. In our study, we determined the level of miR-143 in clinical OS tissues and cells, and explored its function and underlying mechanisms in the tumorigenesis of OS. Our findings revealed that miR-143 expression was significantly downregulated in OS tissues and cell lines. Gain-of-function assays indicated that forced expression of miR-143 in OS cells inhibited cell proliferation and migration/invasion. Bioinformatics and luciferase reporter assays confirmed that MAPK7 was targets gene of miR-143. The results of the present study indicated that miR-143 could be a potential target for treating OS.
Collapse
Affiliation(s)
- Xiancheng Dong
- Nanjing Medical University, Nanjing, PR China; Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, PR China
| | - Bin Lv
- Nanjing Medical University, Nanjing, PR China
| | - Yusong Li
- Department of Orthopedics, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, PR China
| | - Qinghua Cheng
- Department of Orthopedics, People's Hospital of Lishui District in Nanjing City, Nanjing, PR China
| | - Chuan Su
- Nanjing Medical University, Nanjing, PR China; Department of Pathogen Biology & Immunology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, PR China.
| | - Guoyong Yin
- Nanjing Medical University, Nanjing, PR China; Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
36
|
García-Vazquez R, Ruiz-García E, Meneses García A, Astudillo-de la Vega H, Lara-Medina F, Alvarado-Miranda A, Maldonado-Martínez H, González-Barrios JA, Campos-Parra AD, Rodríguez Cuevas S, Marchat LA, López-Camarillo C. A microRNA signature associated with pathological complete response to novel neoadjuvant therapy regimen in triple-negative breast cancer. Tumour Biol 2017; 39:1010428317702899. [PMID: 28621239 DOI: 10.1177/1010428317702899] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neoadjuvant chemotherapy aims to improve the outcome of breast cancer patients, but only few would benefit from this treatment. Pathological complete response has been proposed as a surrogate marker for the prediction of long-term clinical benefits; however, 50%-85% patients have an unfavorable pathological complete response to chemotherapy. MicroRNAs are known biomarkers of breast cancer progression; nevertheless, their potential to identify patients with pathological complete response remains poorly understood. Here, we investigated whether a microRNA profile could be associated with pathological complete response in triple-negative breast cancer patients receiving 5-fluorouracil, adriamycin, cyclophosphamide-cisplatin/paclitaxel as a novel neoadjuvant chemotherapy. In the discovery cohort, the expression of 754 microRNAs was examined in tumors from 10 triple-negative breast cancer patients who achieved pathological complete response and 8 without pathological complete response using TaqMan Low-Density Arrays. Unsupervised hierarchical cluster analysis identified 11 microRNAs with significant differences between responder and no-responder patients (fold change ≥ 1.5; p < 0.05). The differential expression of miR-30a, miR-9-3p, miR-770, and miR-143-5p was validated in an independent group of 17 patients with or without pathological complete response. Moreover, Kaplan-Meier analysis showed that expression of these four microRNAs was associated with an increased disease-free survival. Gene ontology classification of predicted microRNA targets indicated that numerous genes are involved in pathways related to chemoresistance, such as vascular endothelial growth factor, focal adhesion kinase, WNT, ERbB, phosphoinositide 3-kinase, and AKT signaling. In summary, we identified a novel microRNA expression signature associated with pathological complete response in breast cancer. We propose that the four validated microRNAs could be used as molecular biomarkers of clinical response in triple-negative breast cancer patients with pathological complete response to neoadjuvant therapy.
Collapse
Affiliation(s)
- Raúl García-Vazquez
- 1 Programas en Biomedicina Molecular y Biotecnología, Instituto Politécnico Nacional, Ciudad de México, México
| | - Erika Ruiz-García
- 2 Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Abelardo Meneses García
- 2 Laboratorio de Medicina Traslacional, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Horacio Astudillo-de la Vega
- 3 Laboratorio de Investigación Traslacional en Cáncer y Terapia Celular, Hospital de Oncología, Centro Médico Siglo XXI, Ciudad de México, México
| | - Fernando Lara-Medina
- 4 Unidad de Cáncer de Mama, Instituto Nacional de Cancerología, Ciudad de México, México
| | | | | | - Juan A González-Barrios
- 6 Laboratorio de Medicina Genómica, Hospital Regional 1 de Octubre ISSSTE, Ciudad de México, México
| | - Alma D Campos-Parra
- 7 Laboratorio de Genómica, Instituto Nacional de Cancerología, Ciudad de México, México
| | | | - Laurence A Marchat
- 1 Programas en Biomedicina Molecular y Biotecnología, Instituto Politécnico Nacional, Ciudad de México, México
| | - César López-Camarillo
- 9 Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Ciudad de México, México
| |
Collapse
|
37
|
Sun X, Wang M, Wang M, Yu X, Guo J, Sun T, Li X, Yao L, Dong H, Xu Y. Birth order and multiple sclerosis. Acta Neurol Scand 1982; 10:428. [PMID: 32296646 PMCID: PMC7136496 DOI: 10.3389/fonc.2020.00428] [Citation(s) in RCA: 140] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/10/2020] [Indexed: 01/16/2023]
Abstract
Metabolic reprogramming is an emerging hallmark of cancer cells, in which cancer cells exhibit distinct metabolic phenotypes to fuel their proliferation and progression. The significant advancements made in the area of metabolic reprogramming make possible new strategies for overcoming malignant cancer, including triple-negative breast cancer. Triple-negative breast cancer (TNBC) is associated with high histologic grade, aggressive phenotype, and poor prognosis. Even though triple-negative breast cancer patients benefit from standard chemotherapy, they still face high recurrence rates and are more likely to develop resistance to chemotherapeutic drugs. Therefore, there is an urgent need to explore vulnerabilities of triple-negative breast cancer and develop novel therapeutic drugs to improve clinical outcomes for triple-negative breast cancer patients. Metabolic reprogramming may provide promising therapeutic targets for the treatment of triple-negative breast cancer. In this paper, we primarily discuss how triple-negative breast cancer cells reprogram their metabolic phenotype and that of stromal cells in the microenvironment to survive under nutrient-poor conditions. Considering that metastasis and chemoresistance are the main contributors to mortality in triple-negative breast cancer patients, we also focus on the role of metabolic adaption in mediating metastasis and chemoresistance of triple-negative breast cancer tumors.
Collapse
|