1
|
Cao R, Feng Z, Mo J, Wu J, Li J, Li W, Wang Z, Ma Q, Wu Z, Zhou C. Pharmacological inhibition of SREBP1 suppresses pancreatic cancer growth via inducing GPX4-mediated ferroptosis. Cell Signal 2024; 124:111381. [PMID: 39243918 DOI: 10.1016/j.cellsig.2024.111381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/18/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Pancreatic cancer (PC) is highly malignancy with poor survival. Ferroptosis offers a novel therapeutic target for cancer treatment and glutathione peroxidase 4 (GPX4) shields tumor cells from ferroptosis damage. Although Sterol regulatory element-binding protein 1 (SREBP1) has been implicated in the development of pancreatic cancer, its underlying mechanisms remain unclear. This research aims to explore the role of SREBP1 in ferroptosis by using its inhibitor Fatostatin. In this study, Fatostatin was found to inhibit the proliferation and clonogenicity of pancreatic cancer cell lines. This was accompanied by a reduction in intracellular lipid synthesis, increased iron accumulation, elevated levels of reactive oxygen species (ROS), and accumulation of malondialdehyde (MDA). The JASPAR database shows that there is a binding site of the SREBP1 on the promoter region of GPX4. What's more, it was verified that SREBP1 can transcriptionally regulate GPX4 by CHIP. In vivo experiments further revealed that Fatostatin could suppress the growth of subcutaneous tumors in nude mice. In conclusion, our study suggests that Fatostatin may inhibit pancreatic cancer cell proliferation by inducing ferroptosis through the SREBP1/GPX4 pathway. These findings shed light on the therapeutic potential of Fatostatin and lay the groundwork for future investigations into its mechanism of action in pancreatic cancer.
Collapse
Affiliation(s)
- Ruiqi Cao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhengyuan Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jiaoxing Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China; Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
2
|
Zhao G, Zhang X, Meng L, Dong K, Shang S, Jiang T, Liu Z, Gao H. Single-cell RNA-sequencing reveals a unique landscape of the tumor microenvironment in obesity-associated breast cancer. Oncogene 2024; 43:3277-3290. [PMID: 39300255 PMCID: PMC11534694 DOI: 10.1038/s41388-024-03161-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
As two diseases with rapidly increasing incidence, the molecular linkages between obesity and breast cancer (BC) are intriguing. Overall, obesity may be a negative prognostic factor for BC. Single-cell RNA-sequencing (scRNA-seq) was performed on tumor tissues from 6 obese and non-obese BC patients. With 48,033 cells analyzed, we found heterogeneous tumor epithelium and microenvironment in these obese and lean BC patients. Interestingly, the obesity-associated epithelial cells exhibited specific expression signatures which linked tumor growth and hormone metabolism in BC. Notably, one population of obesity-specific macrophage up-regulated the nuclear receptor subfamily 1 group H member 3 (NR1H3), which acted a transcription factor and regulated FABP4 expression through its interaction with the DNA of SREBP1, and further increased the proliferation of tumor cells in BC. Using single-cell signatures, our study illustrate cell diversity and transcriptomic differences in tumors from obese and non-obese BC patients, and sheds light on potential molecular link between lipid metabolism and BC.
Collapse
Affiliation(s)
- Guanghui Zhao
- Peking University People's Hospital, Qingdao; Women and Children's Hospital, QINGDAO UNIVERSITY, Qingdao, 266111, China
| | - Xiaodong Zhang
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Liying Meng
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Ke Dong
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Shipeng Shang
- School of Basic Medicine, Qingdao University, Qingdao, 266035, China
| | - Tengfei Jiang
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Ziqian Liu
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Haidong Gao
- Department of Breast Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China.
- Oncology Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China.
| |
Collapse
|
3
|
Alshareef NS, AlSedairy SA, Al-Harbi LN, Alshammari GM, Yahya MA. Carthamus tinctorius L. (Safflower) Flower Extract Attenuates Hepatic Injury and Steatosis in a Rat Model of Type 2 Diabetes Mellitus via Nrf2-Dependent Hypoglycemic, Antioxidant, and Hypolipidemic Effects. Antioxidants (Basel) 2024; 13:1098. [PMID: 39334757 PMCID: PMC11428842 DOI: 10.3390/antiox13091098] [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: 07/15/2024] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to examine the hepatic and anti-steatotic protective effects of methanolic extract from Carthamus tinctorius (safflower) flowers (SFFE), using a rat model of type 2 diabetes mellitus (T2DM), and to examine the molecular mechanisms underlying these effects. Adult male Wistar rats were used for this study. First, T2DM was induced in some rats by feeding them a high-fat diet (HFD) for 4 weeks, followed by a single dose of streptozotocin (STZ) (35 mg/kg, i.p.). Experimental groups included the following five groups (n = 8 in each): control, control + SFFE, T2DM, T2DM + SFFE, and T2DM + SFFE + brusatol (an Nrf2 inhibitor, 2 mg/kg, i.p.). SFFE was administered at a concentration of 300 mg/kg, and all experiments concluded after 8 weeks. Treatments with SFFE significantly reduced fasting blood glucose levels, free fatty acids (FFAs), cholesterol, triglycerides, and low-density lipoprotein cholesterol in both the control and T2DM rats, but they failed to reduce fasting insulin levels in these groups. SFFE treatments also improved the liver structure and reduced hepatocyte vacuolization and hepatic levels of triglycerides and cholesterol in T2DM rats, in addition to increasing the hepatic mRNA levels of keap1 and the cytoplasmic levels and nuclear activities of Nrf2 in both the control and T2DM rats. SFFE also stimulated the expression levels of PPARα and CPT-1 but reduced the malondialdehyde (MDA), mRNA levels of SREBP1, fatty acid synthase, and acetyl CoA carboxylase in both the control and T2DM rats; meanwhile, it reduced hepatic mRNA and the nuclear activities of NF-κB and increased levels of glutathione, superoxide dismutase, and heme oxygenase-1 in the livers of both groups of treated rats. Furthermore, SFFE suppressed the levels of caspase-3, Bax, tumor necrosis factor-α, and interleukin-6 in the T2DM rats. Treatment with brusatol prevented all of these effects of SFFE. In conclusion, SFFE suppresses liver damage and hepatic steatosis in T2DM through Nrf2-dependent hypoglycemic, antioxidant, anti-inflammatory, and hypolipidemic effects.
Collapse
Affiliation(s)
| | | | - Laila Naif Al-Harbi
- Department of Food Science and Nutrition, College of Food Science and Agriculture, King Saud University, Riyadh 11451, Saudi Arabia; (N.S.A.); (S.A.A.); (G.M.A.); (M.A.Y.)
| | | | | |
Collapse
|
4
|
Jin H, Meng R, Li CS, Kim SH, Chai OH, Lee YH, Park BH, Lee JS, Kim SM. HN1-mediated activation of lipogenesis through Akt-SREBP signaling promotes hepatocellular carcinoma cell proliferation and metastasis. Cancer Gene Ther 2024:10.1038/s41417-024-00827-y. [PMID: 39251779 DOI: 10.1038/s41417-024-00827-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths worldwide, with more than 800,000 deaths each year, and its 5-year survival rate is less than 12%. The role of the HN1 gene in HCC has remained elusive, despite its upregulation in various cancer types. In our investigation, we identified HN1's heightened expression in HCC tissues, which, upon overexpression, fosters cell proliferation, migration, and invasion, unveiling its role as an oncogene in HCC. In addition, silencing HN1 diminished the viability and metastasis of HCC cells, whereas HN1 overexpression stimulated their growth and invasion. Gene expression profiling revealed HN1 silencing downregulated 379 genes and upregulated 130 genes, and suppressive proteins associated with the lipogenic signaling pathway networks. Notably, suppressing HN1 markedly decreased the expression levels of SREBP1 and SREBP2, whereas elevating HN1 had the converse effect. This dual modulation of HN1 affected lipid formation, hindering it upon HN1 silencing and promoting it upon HN1 overexpression. Moreover, HN1 triggers the Akt pathway, fostering tumorigenesis via SREBP1-mediated lipogenesis and silencing HN1 effectively curbed HCC tumor growth in mouse xenograft models by deactivating SREBP-1, emphasizing the potential of HN1 as a therapeutic target, impacting both external and internal factors, it holds promise as an effective therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Hua Jin
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Ruoyu Meng
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, China
| | - Cong Shan Li
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Seong-Hun Kim
- Division of Gastroenterology, Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Ok Hee Chai
- Department of Anatomy, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea
| | - Young-Hoon Lee
- Department of Oral Anatomy, School of Dentistry, Jeonbuk National University, Jeonju, 54907, Republic of Korea
| | - Byung-Hyun Park
- Graduate School of Medical Science and Engineering, Korean Advanced Institute of Science and Technology, Daejon, 34141, Republic of Korea
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, 77045, TX, USA
| | - Soo Mi Kim
- Department of Physiology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju, 54907, Republic of Korea.
| |
Collapse
|
5
|
Zhou Y, Zhou R, Wang N, Zhao T, Qiu P, Gao C, Chang M, Lin N, Zhang X, Li JZ, Wang Q. Inhibition of STRA6 suppresses NSCLC growth via blocking STAT3/SREBP-1c axis-mediated lipogenesis. Mol Cell Biochem 2024:10.1007/s11010-024-05085-y. [PMID: 39168951 DOI: 10.1007/s11010-024-05085-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/01/2024] [Indexed: 08/23/2024]
Abstract
Dysregulation in lipid metabolism is among the most prominent metabolic alterations in cancer. Stimulated by retinoic acid 6 (STRA6), a vitamin A transporter has shown to be involved in the pathogenesis of cancers. Nevertheless, the function of STRA6 in non-small cell lung cancer (NSCLC) progression remains undefined. We obtained cancer and adjacent tissues from NSCLC patients and conducted functional experiments on STRA6 on NSCLC cell lines and mice. High STRA6 expression is correlated with poor prognosis in patients with NSCLC. Results from in vitro and in vivo animal studies showed that STRA6 knockdown suppressed the proliferation, migration, and invasion of NSCLC cells in vitro and tumor growth in vivo through regulation of lipid synthesis. Mechanistically, STRA6 activated a Janus kinase 2/signal transducer and activator of transcription 3 (JAK2-STAT3) signaling cascade which inducing the expression of STAT3 target gene. By inducing the expression of the target gene of STAT3, sterol regulatory element binding protein 1 (SREBP-1), STRA6 promotes SREBP-1-mediated adipogenesis and provides energy for NSCLC cell growth. Our study uncovers a novel STRA6/STAT3/SREBP-1 regulatory axis that enhances NSCLC metastasis by reprogramming of lipid metabolism. These results demonstrate the potential use of STRA6 as a biomarker for diagnosing NSCLC, which may therefore potentially serve as a therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Thoracic Surgery, First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Rong Zhou
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Ning Wang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Tingfeng Zhao
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Pan Qiu
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Chenzi Gao
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Meijia Chang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China
| | - Ning Lin
- NHC Contraceptives Adverse Reaction Surveillance Center, Jiangsu Health Development Research Center, Nanjing, 210036, Jiangsu, China
| | - Xu Zhang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| | - John Zhong Li
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| | - Qian Wang
- Jiangsu Province Key Lab of Human Functional Genomics, Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
6
|
Topkan E, Senyurek S, Kılic Durankus N, Ozturk D, Selek U. Novel Somay's GLUCAR Index Efficiently Predicts Survival Outcomes in Locally Advanced Pancreas Cancer Patients Receiving Definitive Chemoradiotherapy: A Propensity-Score-Matched Cohort Analysis. J Pers Med 2024; 14:746. [PMID: 39064000 PMCID: PMC11278407 DOI: 10.3390/jpm14070746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Propensity score matching (PSM) was used to investigate the prognostic value of a novel GLUCAR index [Glucose × (C-reactive protein ÷ albumin)] in unresectable locally advanced pancreatic cancer (LA-NPC) patients who received definitive concurrent chemoradiotherapy (CCRT). METHODS The PSM analysis comprised 142 LA-PAC patients subjected to definitive CCRT. Receiver operating characteristic (ROC) curve analysis was utilized to identify relevant pre-CCRT cutoffs that could effectively stratify survival results. The primary and secondary objectives were the correlations between the pre-CCRT GLUCAR measures and overall survival (OS) and progression-free survival (PFS). RESULTS The ROC analysis revealed significance at 43.3 for PFS [area under the curve (AUC): 85.1%; sensitivity: 76.8%; specificity: 74.2%; J-index: 0.510)] and 42.8 for OS (AUC: 81.8%; sensitivity: 74.2%; specificity: 71.7%; J-index: 0.459). Given that these cutoff points were close, the standard cutoff point, 42.8, was selected for further analysis. Comparative survival analyses showed that pre-CCRT GLUCAR ≥ 42.8 (n = 71) measures were associated with significantly shorter median PFS (4.7 vs. 15.8 months; p < 0.001) and OS (10.1 vs. 25.4 months; p < 0.001) durations compared to GLUCAR < 42.8 measures (n = 71). The multivariate analysis results confirmed the independent significance of the GLUCAR index on PFS (p < 0.001) and OS (p < 0.001) outcomes. CONCLUSIONS Elevated pre-CCRT GLUCAR levels are robustly and independently linked to significantly poorer PFS and OS outcomes in unresectable LA-PAC patients treated with definitive CCRT.
Collapse
Affiliation(s)
- Erkan Topkan
- Department of Radiation Oncology, Faculty of Medicine, Baskent University, Adana 01120, Turkey
| | - Sukran Senyurek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| | - Nulifer Kılic Durankus
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| | - Duriye Ozturk
- Department of Radiation Oncology, Faculty of Medicine, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Turkey;
| | - Ugur Selek
- Department of Radiation Oncology, Koc University School of Medicine, Istanbul 34450, Turkey; (S.S.); (N.K.D.); (U.S.)
| |
Collapse
|
7
|
Wei Y, Qin Z, Liao X, Zhou X, Huang H, Lan C, Qin W, Zhu G, Su H, Peng T. Pancreatic cancer mortality trends attributable to high fasting blood sugar over the period 1990-2019 and projections up to 2040. Front Endocrinol (Lausanne) 2024; 15:1302436. [PMID: 39036051 PMCID: PMC11257875 DOI: 10.3389/fendo.2024.1302436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 06/13/2024] [Indexed: 07/23/2024] Open
Abstract
Background Pancreatic cancer (PC) is a prevalent malignancy within the digestive system, with diabetes recognized as one of its well-established risk factors. Methods Data on PC mortality attributed to high fasting blood sugar were retrieved from the Global Burden of Disease (GBD) study 2019 online database. To assess the temporal trends of PC burden attributable to high fasting plasma glucose (HFPG), estimated annual percentage changes (EAPCs) for age-standardized death rates (ASDRs) between 1990 and 2019 were determined using a generalized linear model. Furthermore, a Bayesian age-period-cohort (BAPC) model using the integrated nested Laplacian approximation algorithm was employed to project the disease burden over the next 20 years. Results Globally, the crude death number of PC attributable to HFPG almost tripled (from 13,065.7 in 1990 to 48,358.5 in 2019) from 1990 to 2019, and the ASDR increased from 0.36/100,000 to 0.61/100,000 with an EAPC of 2.04 (95% CI 1.91-2.16). The population aged ≥70 years accounted for nearly 60% of total deaths in 2019 and experienced a more significant increase, with the death number increasing approximately fourfold and the ASDR increasing annually by 2.65%. In regions with different sociodemographic indexes (SDIs), the highest disease burden was observed in the high-SDI region, whereas more pronounced increasing trends in ASDR were observed in the low to middle-SDI, low-SDI, and middle-SDI regions. Additionally, a significantly negative association was found between EAPCs and ASDRs of PC attributable to HFPG from 1990 to 2019. Moreover, the BAPC model predicts that ASDR and age-standardized disability-adjusted life-years (DALYs) rate for PC attributed to HFPG was projected to increase obviously for men and women from 2019 to 2040. Conclusions The burden of PC attributed to HFPG has increased globally over the past three decades, with the elderly population and high-SDI regions carrying a relatively greater disease burden, but more adverse trends observed in low-SDI areas. Furthermore, the burden is projected to continue increasing over the next 20 years. Hence, more tailored prevention methodologies should be established to mitigate this increasing trend.
Collapse
Affiliation(s)
- Yongguang Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Zedong Qin
- Departments of Oncology, Xichang People’s Hospital, Xichang, China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Huasheng Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Chenlu Lan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Wei Qin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Guangzhi Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Hao Su
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment (Guangxi Medical University), Ministry of Education, Nanning, China
| |
Collapse
|
8
|
Wang Q, Gong M, Liu R, Mo J, Bai R, An R, Wang X, Han L, Wang Z, Ma Q, Wu Z, Zhou C. Huaier enhances the tumor-killing effect and reverses gemcitabine-induced stemness by suppressing FoxM1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155656. [PMID: 38723529 DOI: 10.1016/j.phymed.2024.155656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 03/03/2024] [Accepted: 04/18/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Gemcitabine is the first-line chemotherapy drug that can easily cause chemotherapy resistance. Huaier is a traditional Chinese medicine and shows an antitumor effect in pancreatic cancer, but whether it can enhance the gemcitabine chemotherapeutic response and the potential mechanism remain unknown. PURPOSE This study was performed to explore the effect of Huaier in promoting the tumor-killing effect of gemcitabine and elucidate the possible mechanism in pancreatic cancer. METHODS Cell Counting Kit-8 assays and colony formation assays were used to detect proliferation after different treatments. Protein coimmunoprecipitation was applied to demonstrate protein interactions. Nuclear protein extraction and immunofluorescence were used to confirm the intracellular localization of the proteins. Western blotting was performed to detect cell proliferation-related protein expression or cancer stem cell-associated protein expression. Sphere formation assays and flow cytometry were used to assess the stemness of pancreatic cancer cells. The in vivo xenograft model was used to confirm the inhibitory effect under physiological conditions, and immunohistochemistry was used to detect protein expression. RESULTS Huaier suppressed the proliferation and stem cell-like properties of pancreatic cancer cells. We found that Huaier suppressed the expression of forkhead box protein M1 (FoxM1). In addition, Huaier inhibited FoxM1 function by blocking its nuclear translocation. Treatment with Huaier reversed the stemness induced by gemcitabine in a FoxM1-dependent manner. Furthermore, we verified the above results by an in vivo study, which reached the same conclusion as those in vitro. CONCLUSION Overall, this study illustrates that Huaier augments the tumor-killing effect of gemcitabine through suppressing the stemness induced by gemcitabine in a FoxM1-dependent way. These results indicate that Huaier can be applied to overcome gemcitabine resistance.
Collapse
Affiliation(s)
- Qiqi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Mengyuan Gong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Rujuan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Jiantao Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Ruiping Bai
- Department of Anesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Rui An
- Department of Anesthesiology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Xueni Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Liang Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China.
| | - Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China; Pancreatic Disease Center of Xi'an Jiaotong University, Xi'an, PR China.
| |
Collapse
|
9
|
Zare I, Zirak Hassan Kiadeh S, Varol A, Ören Varol T, Varol M, Sezen S, Zarepour A, Mostafavi E, Zahed Nasab S, Rahi A, Khosravi A, Zarrabi A. Glycosylated nanoplatforms: From glycosylation strategies to implications and opportunities for cancer theranostics. J Control Release 2024; 371:158-178. [PMID: 38782062 DOI: 10.1016/j.jconrel.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/12/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Glycosylated nanoplatforms have emerged as promising tools in the field of cancer theranostics, integrating both therapeutic and diagnostic functionalities. These nanoscale platforms are composed of different materials such as lipids, polymers, carbons, and metals that can be modified with glycosyl moieties to enhance their targeting capabilities towards cancer cells. This review provides an overview of different modification strategies employed to introduce glycosylation onto nanoplatforms, including chemical conjugation, enzymatic methods, and bio-orthogonal reactions. Furthermore, the potential applications of glycosylated nanoplatforms in cancer theranostics are discussed, focusing on their roles in drug delivery, imaging, and combination therapy. The ability of these nanoplatforms to selectively target cancer cells through specific interactions with overexpressed glycan receptors is highlighted, emphasizing their potential for enhancing efficacy and reducing the side effects compared to conventional therapies. In addition, the incorporation of diagnostic components onto the glycosylated nanoplatforms provided the capability of simultaneous imaging and therapy and facilitated the real-time monitoring of treatment response. Finally, challenges and future perspectives in the development and translation of glycosylated nanoplatforms for clinical applications are addressed, including scalability, biocompatibility, and regulatory considerations. Overall, this review underscores the significant progress made in the field of glycosylated nanoplatforms and their potential to revolutionize cancer theranostics.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz 7178795844, Iran
| | - Shahrzad Zirak Hassan Kiadeh
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, P.O. Box 14395-1561, Tehran, Iran
| | - Ayşegül Varol
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Tuğba Ören Varol
- Department of Chemistry, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla TR48000, Turkiye
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla TR48000, Turkiye
| | - Serap Sezen
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, 34956 Istanbul, Turkiye; Nanotechnology Research and Application Center, Sabanci University, Tuzla, 34956 Istanbul, Turkiye
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shima Zahed Nasab
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, P.O. Box 14395-1561, Tehran, Iran
| | - Amid Rahi
- Pathology and Stem cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
10
|
He Y, Qi S, Chen L, Zhu J, Liang L, Chen X, Zhang H, Zhuo L, Zhao S, Liu S, Xie T. The roles and mechanisms of SREBP1 in cancer development and drug response. Genes Dis 2024; 11:100987. [PMID: 38560498 PMCID: PMC10978545 DOI: 10.1016/j.gendis.2023.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 04/04/2024] Open
Abstract
Cancer occurrence and development are closely related to increased lipid production and glucose consumption. Lipids are the basic component of the cell membrane and play a significant role in cancer cell processes such as cell-to-cell recognition, signal transduction, and energy supply, which are vital for cancer cell rapid proliferation, invasion, and metastasis. Sterol regulatory element-binding transcription factor 1 (SREBP1) is a key transcription factor regulating the expression of genes related to cholesterol biosynthesis, lipid homeostasis, and fatty acid synthesis. In addition, SREBP1 and its upstream or downstream target genes are implicated in various metabolic diseases, particularly cancer. However, no review of SREBP1 in cancer biology has yet been published. Herein, we summarized the roles and mechanisms of SREBP1 biological processes in cancer cells, including SREBP1 modification, lipid metabolism and reprogramming, glucose and mitochondrial metabolism, immunity, and tumor microenvironment, epithelial-mesenchymal transition, cell cycle, apoptosis, and ferroptosis. Additionally, we discussed the potential role of SREBP1 in cancer prognosis, drug response such as drug sensitivity to chemotherapy and radiotherapy, and the potential drugs targeting SREBP1 and its corresponding pathway, elucidating the potential clinical application based on SREBP1 and its corresponding signal pathway.
Collapse
Affiliation(s)
- Ying He
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shasha Qi
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lu Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jinyu Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Linda Liang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Xudong Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Hao Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lvjia Zhuo
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shujuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shuiping Liu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
11
|
Su F, Koeberle A. Regulation and targeting of SREBP-1 in hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:673-708. [PMID: 38036934 PMCID: PMC11156753 DOI: 10.1007/s10555-023-10156-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
Hepatocellular carcinoma (HCC) is an increasing burden on global public health and is associated with enhanced lipogenesis, fatty acid uptake, and lipid metabolic reprogramming. De novo lipogenesis is under the control of the transcription factor sterol regulatory element-binding protein 1 (SREBP-1) and essentially contributes to HCC progression. Here, we summarize the current knowledge on the regulation of SREBP-1 isoforms in HCC based on cellular, animal, and clinical data. Specifically, we (i) address the overarching mechanisms for regulating SREBP-1 transcription, proteolytic processing, nuclear stability, and transactivation and (ii) critically discuss their impact on HCC, taking into account (iii) insights from pharmacological approaches. Emphasis is placed on cross-talk with the phosphatidylinositol-3-kinase (PI3K)-protein kinase B (Akt)-mechanistic target of rapamycin (mTOR) axis, AMP-activated protein kinase (AMPK), protein kinase A (PKA), and other kinases that directly phosphorylate SREBP-1; transcription factors, such as liver X receptor (LXR), peroxisome proliferator-activated receptors (PPARs), proliferator-activated receptor γ co-activator 1 (PGC-1), signal transducers and activators of transcription (STATs), and Myc; epigenetic mechanisms; post-translational modifications of SREBP-1; and SREBP-1-regulatory metabolites such as oxysterols and polyunsaturated fatty acids. By carefully scrutinizing the role of SREBP-1 in HCC development, progression, metastasis, and therapy resistance, we shed light on the potential of SREBP-1-targeting strategies in HCC prevention and treatment.
Collapse
Affiliation(s)
- Fengting Su
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria
| | - Andreas Koeberle
- Michael Popp Institute and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, 6020, Innsbruck, Austria.
| |
Collapse
|
12
|
Li F, Wan X, Li Z, Zhou L. High glucose inhibits autophagy and promotes the proliferation and metastasis of colorectal cancer through the PI3K/AKT/mTOR pathway. Cancer Med 2024; 13:e7382. [PMID: 38872380 PMCID: PMC11176572 DOI: 10.1002/cam4.7382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/16/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) ranks among the most prevalent malignancies worldwide, characterized by its complex etiology and slow research progress. Diabetes, as an independent risk factor for CRC, has been widely certified. Consequently, this study centers on elucidating the intricacies of CRC cells initiation and progression within a high-glucose environment. METHODS A battery of assays was employed to assess the proliferation and metastasis of CRC cells cultured under varying glucose concentrations. Optimal glucose levels conducive to cells' proliferation and migration were identified. Western blot analyses were conducted to evaluate alterations in apoptosis, autophagy, and EMT-related proteins in CRC cells under high-glucose conditions. The expression of PI3K/AKT/mTOR pathway-associated proteins was assessed using western blot. The effect of high glucose on xenograft growth was investigated in vivo by MC38 cells, and changes in inflammatory factors (IL-4, IL-13, TNF-α, IL-5, and IL-12) were measured via serum ELISA. RESULTS Our experiments demonstrated that elevated glucose concentrations promoted both the proliferation and migration of CRC cells; the most favorable glucose dose is 20 mM. Western blot analyses revealed a decrease in apoptotic proteins, such as Bim, Bax, and caspase-3 with increasing glucose levels. Concurrently, the expression of EMT-related proteins, including N-cadherin, vimentin, ZEB1, and MMP9, increased. High-glucose cultured cells exhibited elevated levels of PI3K/AKT/mTOR pathway proteins. In the xenograft model, tumor cells stimulated by high glucose exhibited accelerated growth, larger tumor volumes, and heightened KI67 expression of immunohistochemistry. ELISA experiments revealed higher expression of IL-4 and IL-13 and lower expression of TNF-α and IL-5 in the serum of high-glucose-stimulated mice. CONCLUSION The most favorable dose and time for tumor cells proliferation and migration is 20 mM, 48 h. High glucose fosters CRC cell proliferation and migration while suppressing autophagy through the activation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Feng Li
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xing Wan
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhigui Li
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Liming Zhou
- Department of Pharmacology, West China School of Basic Science and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Wang T, Zhang M, Gong X, Chen W, Peng Y, Liao C, Xu H, Li Q, Shen G, Ren H, Zhu Y, Zhang B, Mao J, Wei L, Chen Y, Yang X. Inhibition of Nogo-B reduces the progression of pancreatic cancer by regulation NF-κB/GLUT1 and SREBP1 pathways. iScience 2024; 27:109741. [PMID: 38706871 PMCID: PMC11068639 DOI: 10.1016/j.isci.2024.109741] [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: 10/10/2023] [Revised: 01/22/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
Pancreatic cancer (PC) is a lethal disease and associated with metabolism dysregulation. Nogo-B is related to multiple metabolic related diseases and types of cancers. However, the role of Nogo-B in PC remains unknown. In vitro, we showed that cell viability and migration was largely reduced in Nogo-B knockout or knockdown cells, while enhanced by Nogo-B overexpression. Consistently, orthotopic tumor and metastasis was reduced in global Nogo knockout mice. Furthermore, we indicated that glucose enhanced cell proliferation was associated to the elevation expression of Nogo-B and nuclear factor κB (NF-κB). While, NF-κB, glucose transporter type 1 (GLUT1) and sterol regulatory element-binding protein 1 (SREBP1) expression was reduced in Nogo-B deficiency cells. In addition, we showed that GLUT1 and SREBP1 was downstream target of NF-κB. Therefore, we demonstrated that Nogo deficiency inhibited PC progression is regulated by the NF-κB/GLUT1 and SREBP1 pathways, and suggested that Nogo-B may be a target for PC therapy.
Collapse
Affiliation(s)
- Tianxiang Wang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Min Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Xinyu Gong
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Wanjing Chen
- Department of General Surgery, The Second Affiliated Hospital, Anhui Medical University, Hefei 230000, China
| | - Ying Peng
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Hongmei Xu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Qingshan Li
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Guodong Shen
- Department of Geriatrics, The First Affiliated Hospital of University of Science and Technology of China, Gerontology Institute of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Huirong Ren
- Department of Geriatrics, The First Affiliated Hospital of University of Science and Technology of China, Gerontology Institute of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230000, China
| | - Yaxin Zhu
- Institute for International Health Professions Education and Research, China Medical University, Shenyang 110000, China
| | - Baotong Zhang
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen 518000, China
| | - Jiali Mao
- Department of Anesthesiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230000, China
| | - Lingling Wei
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Xiaoxiao Yang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, College of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| |
Collapse
|
14
|
Liu M, Zhang Z, Chen Y, Feng T, Zhou Q, Tian X. Circadian clock and lipid metabolism disorders: a potential therapeutic strategy for cancer. Front Endocrinol (Lausanne) 2023; 14:1292011. [PMID: 38189049 PMCID: PMC10770836 DOI: 10.3389/fendo.2023.1292011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Recent research has emphasized the interaction between the circadian clock and lipid metabolism, particularly in relation to tumors. This review aims to explore how the circadian clock regulates lipid metabolism and its impact on carcinogenesis. Specifically, targeting key enzymes involved in fatty acid synthesis (SREBP, ACLY, ACC, FASN, and SCD) has been identified as a potential strategy for cancer therapy. By disrupting these enzymes, it may be possible to inhibit tumor growth by interfering with lipid metabolism. Transcription factors, like SREBP play a significant role in regulating fatty acid synthesis which is influenced by circadian clock genes such as BMAL1, REV-ERB and DEC. This suggests a strong connection between fatty acid synthesis and the circadian clock. Therefore, successful combination therapy should target fatty acid synthesis in addition to considering the timing and duration of drug use. Ultimately, personalized chronotherapy can enhance drug efficacy in cancer treatment and achieve treatment goals.
Collapse
Affiliation(s)
- Mengsi Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Zhen Zhang
- Department of Oncology, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, China
| | - Yating Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Ting Feng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xuefei Tian
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
- Hunan Key Laboratory of Traditional Chinese Medicine Prescription and Syndromes Translational Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine, Changsha, China
- Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention and Treatment, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
15
|
Liu XR, Liu F, Li ZW, Lv Q, Shu XP, Li LS, Tong Y, Zhang W, Peng D. The Use of Metformin and Postoperative Insulin Pump Were Predictive Factors for Outcomes of Diabetic Colorectal Cancer Patients after Surgery. Nutr Cancer 2023; 75:1926-1933. [PMID: 37870939 DOI: 10.1080/01635581.2023.2272815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/24/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE This present study aims to explore the influence of metformin and postoperative insulin pump use on colorectal cancer (CRC) patients with type II diabetes mellitus (T2DM) who received surgery in terms of short-term and long-term outcomes. METHODS 613 CRC patients who had comorbid T2DM and received surgery at a single clinical center from Jan, 2011 to Dec, 2021 were included in this study. Univariate and multivariate logistic regression analyses were used to find predictive factors for overall complications and major complications. Cox regression analyses was used to find prognostic factors for overall survival (OS) and disease-free survival (DFS). All statistical analysis was performed using SPSS (version 22.0) software. The Kaplan-Meier curve was used to show the OS and DFS between the insulin pump group and the no insulin pump group. RESULTS Multivariate logistic regression analysis reported that lower body mass index (BMI) (p < 0.01, OR = 0.922, 95% CI = 0.870-0.977) and metformin use (p = 0.03, OR = 0.643, 95% CI = 0.431-0.959) were independent protective factors for overall complications, and insulin pump after surgery (p < 0.01, OR = 3.991, 95% CI = 2.434-6.544) was an independent risk factor for overall complications. As for major complications, metformin use (p = 0.042, OR = 0.274, 95% CI = 0.079-0.956) and insulin pump after surgery (p = 0.03, OR = 2.892, 95% CI = 1.107-7.552) remained independent protective factors and independent risk factors, respectively. Moreover, in Cox regression analyses, age (OS: p < 0.01, HR = 1.032, 95% CI = 1.008-1.057; DFS: p < 0.01, HR = 1.030, 95% CI = 1.008-1.052), tumor stage (OS: p < 0.01, HR = 1.709, 95% CI = 1.244-2.346; DFS: p < 0.01, HR = 1.696, 95% CI = 1.276-2.254), and Insulin pump after surgery (OS: p < 0.01, HR = 2.923, 95% CI = 1.887-4.527; DFS: p < 0.01, HR = 2.671, 95% CI = 1.779-4.009) were independent prognostic factors for both OS and DFS. After comparing the OS and DFS between the insulin pump group and the no insulin pump group, patients who received postoperative insulin pump had worse OS and DFS in all tumor node metastasis (TNM) stages (p < 0.01). CONCLUSION Diabetic CRC patients who used metformin had a lower risk of postoperative complications. However, there was no difference from patients not using metformin in terms of survival. Furthermore, patients receiving postoperative insulin pump had more postoperative complications and worse survival in all TNM stages.
Collapse
Affiliation(s)
- Xu-Rui Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fei Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-Wei Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Quan Lv
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin-Peng Shu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lian-Shuo Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Tong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Liu M, Li C, Qu J, Sun S, Zhao Z, Wang W, Lv W, Zhang Y, Cai Y, Zhao F, Wu F, Zhang S, Zhao X. Baicalein enhances immune response in TNBC by inhibiting leptin expression of adipocytes. Cancer Sci 2023; 114:3834-3847. [PMID: 37489486 PMCID: PMC10551602 DOI: 10.1111/cas.15916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a special pathological type of breast cancer (BC) with poor prognosis. Obesity is shown to be involved in TNBC tumor progression. The interaction between obesity and BC has generated great attention in recent years, however, the mechanism is still unclear. Here, we showed that leptin secreted by adipocytes upregulated PD-L1 expression in TNBC through the p-STAT3 signaling pathway and that baicalein inhibited PD-L1 expression in tumor microenvironment by suppressing leptin transcription of adipocytes. Collectively, our findings suggest that leptin may be the key factor participating in obesity-related tumor progression and that baicalein can break through the dilemma to boost the anti-tumor immune response.
Collapse
Affiliation(s)
- Mengjie Liu
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Chaofan Li
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Jingkun Qu
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Shiyu Sun
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Zitong Zhao
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Weiwei Wang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Wei Lv
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yu Zhang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yifan Cai
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Fang Zhao
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Fei Wu
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Shuqun Zhang
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Xixi Zhao
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
- Department of Radiation OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
17
|
Wu J, Tang L, Zheng F, Chen X, Li L. A review of the last decade: pancreatic cancer and type 2 diabetes. Arch Physiol Biochem 2023:1-9. [PMID: 37646618 DOI: 10.1080/13813455.2023.2252204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023]
Abstract
Pancreatic cancer (PC) is a prevalent gastrointestinal tumour known for its high degree of malignancy, resulting in a mere 10% five-year survival rate for most patients. Over the past decade, a growing body of research has shed light on the intricate bidirectional association between PC and Type 2 diabetes (T2DM). The collection of PC- and T2DM-related articles is derived from two comprehensive databases, namely WOS (Web of Science Core Collection) and CNKI (China National Knowledge Infrastructure). This article discusses the last 10 years of research trends in PC and T2DM and explores their potential regulatory relationship as well as related medications.
Collapse
Affiliation(s)
- Jiaqi Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Liang Tang
- Department of General Medicine, Zhuzhou Central Hospital, Zhuzhou, China
| | - Feng Zheng
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Xun Chen
- Department of the Trauma center, Zhuzhou Central Hospital, Zhuzhou, China
- Department of hepatobiliary surgery, Zhuzhou Central Hospital, Zhuzhou, China
| | - Lei Li
- Department of Pathology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Ashrafizadeh M, Zhang W, Zou R, Sethi G, Klionsky DJ, Zhang X. A bioinformatics analysis, pre-clinical and clinical conception of autophagy in pancreatic cancer: Complexity and simplicity in crosstalk. Pharmacol Res 2023; 194:106822. [PMID: 37336429 DOI: 10.1016/j.phrs.2023.106822] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Pancreatic cancer (PC) is a serious gastrointestinal tract disease for which the 5-year survival rate is less than 10%, even in developed countries such as the USA. The genomic profile alterations and dysregulated biological mechanisms commonly occur in PC. Macroautophagy/autophagy is a cell death process that is maintained at a basal level in physiological conditions, whereas its level often changes during tumorigenesis. The function of autophagy in human cancers is dual and can be oncogenic and onco-suppressor. Autophagy is a potent controller of tumorigenesis in PC. The supportive autophagy in PC escalates the growth rate of PC cells and its suppression can mediate cell death. Autophagy also determines the metastasis of PC cells, and it can control the EMT in affecting migration. Moreover, starvation and hypoxia can stimulate glycolysis, and glycolysis induction can be mediated by autophagy in enhancing tumorigenesis in PC. Furthermore, protective autophagy stimulates drug resistance and gemcitabine resistance in PC cells, and its inhibition can enhance radiosensitivity. Autophagy can degrade MHC-I to mediate immune evasion and also regulates polarization of macrophages in the tumor microenvironment. Modulation of autophagy activity is provided by silibinin, ursolic acid, chrysin and huaier in the treatment of PC. Non-coding RNAs are also controllers of autophagy in PC and its inhibition can improve therapy response in patients. Moreover, mitophagy shows dysregulation in PC, which can enhance the proliferation of PC cells. Therefore, a bioinformatics analysis demonstrates the dysregulation of autophagy-related proteins and genes in PC as biomarkers.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Xianbin Zhang
- Department of General Surgery and Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
19
|
Murillo Carrasco AG, Giovanini G, Ramos AF, Chammas R, Bustos SO. Insights from a Computational-Based Approach for Analyzing Autophagy Genes across Human Cancers. Genes (Basel) 2023; 14:1550. [PMID: 37628602 PMCID: PMC10454514 DOI: 10.3390/genes14081550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
In the last decade, there has been a boost in autophagy reports due to its role in cancer progression and its association with tumor resistance to treatment. Despite this, many questions remain to be elucidated and explored among the different tumors. Here, we used omics-based cancer datasets to identify autophagy genes as prognostic markers in cancer. We then combined these findings with independent studies to further characterize the clinical significance of these genes in cancer. Our observations highlight the importance of innovative approaches to analyze tumor heterogeneity, potentially affecting the expression of autophagy-related genes with either pro-tumoral or anti-tumoral functions. In silico analysis allowed for identifying three genes (TBC1D12, KERA, and TUBA3D) not previously described as associated with autophagy pathways in cancer. While autophagy-related genes were rarely mutated across human cancers, the expression profiles of these genes allowed the clustering of different cancers into three independent groups. We have also analyzed datasets highlighting the effects of drugs or regulatory RNAs on autophagy. Altogether, these data provide a comprehensive list of targets to further the understanding of autophagy mechanisms in cancer and investigate possible therapeutic targets.
Collapse
Affiliation(s)
- Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Guilherme Giovanini
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, Brazil; (G.G.); (A.F.R.)
| | - Alexandre Ferreira Ramos
- Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, Av. Arlindo Béttio, 1000, São Paulo 03828-000, Brazil; (G.G.); (A.F.R.)
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Silvina Odete Bustos
- Center for Translational Research in Oncology (LIM24), Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), São Paulo 01246-000, Brazil; (A.G.M.C.); (S.O.B.)
- Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
20
|
Simanurak O, Pekthong D, Somran J, Wangteeraprasert A, Srikummool M, Kaewpaeng N, Parhira S, Srisawang P. Enhanced apoptosis of HCT116 colon cancer cells treated with extracts from Calotropis gigantea stem bark by starvation. Heliyon 2023; 9:e18013. [PMID: 37483695 PMCID: PMC10362240 DOI: 10.1016/j.heliyon.2023.e18013] [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: 02/14/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/25/2023] Open
Abstract
Calotropis gigantea stem bark extract, particularly the dichloromethane fraction (CGDCM), demonstrated the most potent antiproliferative effects on hepatocellular carcinoma HepG2 and colorectal HCT116 cells. The current study focused on enhancing the effectiveness of cancer treatment with CGDCM at concentrations close to the IC50 in HCT116 cells by reducing their nutrient supply. CGDCM (2, 4, and 8 μg/mL) treatment for 24 h under glucose conditions of 4.5 g/L without fetal bovine serum (FBS) supplementation or serum starvation (G+/F-), glucose 0 g/L with 10% FBS or glucose starvation (G-/F+), and glucose 0 g/L with 0% FBS or complete starvation (G-/F-) induced a greater antiproliferative effect in HCT116 cells than therapy in complete medium with glucose 4.5 g/L and 10% FBS (G+/F+). Nonetheless, the anticancer effect of CGDCM at 4 μg/mL under (G-/F-) showed the highest activity compared to other starvation conditions. The three starvation conditions showed a significant reduction in cell viability compared to the control (G+/F+) medium group, while the inhibitory effect on cell viability did not differ significantly among the three starvation conditions. CGDCM at 4 μg/mL in (G-/F-) medium triggered apoptosis by dissipating the mitochondrial membrane potential and arresting cells in the G2/M phase. This investigation demonstrated that a decrease in intracellular ATP and fatty acid levels was associated with enhanced apoptosis by treatment with CGDCM at 4 μg/mL under (G-/F-) conditions. In addition, under (G-/F-), CGDCM at 4 μg/mL increased levels of reactive oxygen species (ROS) and was suggested to primarily trigger apoptosis in HCT116 cells. Thus, C. gigantea extracts may be useful for the future development of alternative, effective cancer treatment regimens.
Collapse
Affiliation(s)
- Orakot Simanurak
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Dumrongsak Pekthong
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Julintorn Somran
- Department of Pathology, Faculty of Medicine, Naresuan University, Phitsanulok, 65000, Thailand
| | | | - Metawee Srikummool
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| | - Naphat Kaewpaeng
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Supawadee Parhira
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Environmental Health and Toxicology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok, 65000, Thailand
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Piyarat Srisawang
- Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence for Innovation in Chemistry, Naresuan University, Phitsanulok 65000, Thailand
- Center of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
21
|
Chu X, Zhong L, Dan W, Wang X, Zhang Z, Liu Z, Lu Y, Shao X, Zhou Z, Chen S, Liu B. DNMT3A R882H mutation promotes acute leukemic cell survival by regulating glycolysis through the NRF2/NQO1 axis. Cell Signal 2023; 105:110626. [PMID: 36758683 DOI: 10.1016/j.cellsig.2023.110626] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
BACKGROUND Studies have confirmed that acute myeloid leukemia (AML) cells with DNA methyltransferase 3A Arg882His (DNMT3A R882H) mutation show an increased proliferation capability. However, the associated mechanism is still unclear. Glycolysis is involved in regulating malignant proliferation of cancer cell. Hence, we analyzed whether the DNMT3A R882H mutation interferes with glycolysis and thereby influences AML cell proliferation. METHODS We generated AML cell line carrying a DNMT3A-R882H mutation and compared it with the wild type (DNMT3A-WT) with regard to glycolysis regulation. Moreover, we analyzed the cell line's proliferation and apoptosis by a CCK-8 assay, western blotting, and flow cytometry. The role of NRF2/NQO1 signaling in regulating glycolysis was investigated by NRF2-knockdown and Brusatol (specific inhibitor of NRF2) treatment. RESULTS DNMT3A R882H cells had a higher glucose transport capacity compared to WT cells and their viability could be reduced by glucose deprivation. Moreover, daunorubicin had a slight inhibitory effect on glycolysis while glycolysis inhibition re-sensitized mutant cells to daunorubicin. Obviously, DNMT3A R882H mutation activated the NRF2/NQO1 pathway and enhanced the glycolytic activity in mutant cells. CONCLUSION Taken together, these results suggest a novel mechanism by which a DNMT3A R882H mutation promotes glycolysis via activation of NRF2/NQO1 pathway. A parallel glycolysis inhibition adds to the anticancer effects of daunorubicin which might lead to a novel therapeutic approach for the treatment of AML patients carrying a DNMT3A R882H mutation.
Collapse
Affiliation(s)
- Xuan Chu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Wenran Dan
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xiao Wang
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhonghui Zhang
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhenyan Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Yang Lu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xin Shao
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Ziwei Zhou
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Shuyu Chen
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Beizhong Liu
- Central Laboratory of Yong-Chuan Hospital, Chongqing Medical University, Chongqing 402160, China; Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
22
|
Zhu YH, Zheng JH, Jia QY, Duan ZH, Yao HF, Yang J, Sun YW, Jiang SH, Liu DJ, Huo YM. Immunosuppression, immune escape, and immunotherapy in pancreatic cancer: focused on the tumor microenvironment. Cell Oncol (Dordr) 2023; 46:17-48. [PMID: 36367669 DOI: 10.1007/s13402-022-00741-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by poor treatment response and low survival time. The current clinical treatment for advanced PDAC is still not effective. In recent years, the research and application of immunotherapy have developed rapidly and achieved substantial results in many malignant tumors. However, the translational application in PDAC is still far from satisfactory and needs to be developed urgently. To carry out the study of immunotherapy, it is necessary to fully decipher the immune characteristics of PDAC. This review summarizes the recent progress of the tumor microenvironment (TME) of PDAC and highlights its link with immunotherapy. We describe the molecular cues and corresponding intervention methods, collate several promising targets and progress worthy of further study, and put forward the importance of integrated immunotherapy to provide ideas for future research of TME and immunotherapy of PDAC.
Collapse
Affiliation(s)
- Yu-Heng Zhu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jia-Hao Zheng
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Qin-Yuan Jia
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Zong-Hao Duan
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Hong-Fei Yao
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Jian Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China
| | - Yong-Wei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 800 Dongchuan Road, 200240, People's Republic of China.
| | - De-Jun Liu
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, People's Republic of China.
| |
Collapse
|
23
|
Alshammari GM, Abdelhalim MA, Al-Ayed MS, Al-Harbi LN, Yahya MA. Concomitant Sub-Chronic Administration of Small-Size Gold Nanoparticles Aggravates Doxorubicin-Induced Liver Oxidative and Inflammatory Damage, Hyperlipidemia, and Hepatic Steatosis. Molecules 2023; 28:796. [PMID: 36677854 PMCID: PMC9863023 DOI: 10.3390/molecules28020796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023] Open
Abstract
This study examined the effect of gold nanoparticles (AuNPs) on doxorubicin (DOX)-induced liver damage and steatosis in rats and tested its effect mechanism. Wistar male rats were divided into four groups (each of eight rats) as control, AuNPs (50 µL of 10 nm), DOX (15 mg/kg; 3 mg/kg/week), and DOX + AuNPs-treated rats. DOX is known to induce fasting hyperglycemia and hyperinsulinemia in treated rats. Individual treatment of both DOX and AuNPs also promoted liver damage, increased circulatory levels of ALT and AST, and stimulated serum and liver levels of TGs, CHOL, LDL-c, and FFAs. They also stimulated MDA, TNF-α, and IL-6, reduced GSH, SOD, HO-1, and CAT, upregulated mRNA levels of Bax and caspases-3 and -8 and downregulated mRNA levels of Bcl2 in the livers of rats. However, while DOX alone reduced hepatic levels of PPARα, both AuNPs and DOX stimulated mRNA levels of SREBP1, reduced the mRNA, cytoplasmic and nuclear levels of Nrf2, and increased mRNA, cytoplasmic, and nuclear levels of NF-κB. The liver damage and the alterations in all these parameters were significantly more profound when both AuNPs and DOX were administered together. In conclusion, AuNPs exaggerate liver damage, hyperlipidemia, and hepatic steatosis in DOX-treated rats by activating SREBP1 and NF-κB and suppressing the Nrf2/antioxidant axis.
Collapse
Affiliation(s)
- Ghedeir M. Alshammari
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Anwar Abdelhalim
- Department of Physics and Astronomy, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed S. Al-Ayed
- Department of Physics and Astronomy, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Laila Naif Al-Harbi
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Abdo Yahya
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
24
|
Tan M, Lin X, Chen H, Ye W, Yi J, Li C, Liu J, Su J. Sterol regulatory element binding transcription factor 1 promotes proliferation and migration in head and neck squamous cell carcinoma. PeerJ 2023; 11:e15203. [PMID: 37090107 PMCID: PMC10117388 DOI: 10.7717/peerj.15203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/17/2023] [Indexed: 04/25/2023] Open
Abstract
Background Sterol-regulatory element-binding protein 1 (SREBP1) is a transcription factor involved in lipid metabolism that is encoded by sterol regulatory element binding transcription factor 1(SREBF1). SREBP1 overexpression is associated with the progression of several human tumors; however, the role of SREBP1 in head and neck squamous cell carcinoma (HNSC) remains unclear. Methods SREBF1 expression in pan-cancer was analyzed using the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) data, and the association between SREBF1 expression and clinical characteristics of HNSC patients was examined using the UALCAN database. Enrichment analysis of SREBF1-related genes was performed using the Cluster Profiler R package. TCGA database was used to investigate the relationship between immune cell infiltration and SREBF1 expression. CCK-8, flow cytometry, and wound healing assays were performed to investigate the effect of SREBF1 knockdown on the proliferation and migration of HNSC cells. Results SREBF1 was significantly upregulated in several tumor tissues, including HNSC, and SREBF1 overexpression was positively correlated with sample type, cancer stage, tumor grade, and lymph node stage in HNSC patients. Gene enrichment analysis revealed that SREBF1 is associated with DNA replication and homologous recombination. SREBF1 upregulation was positively correlated with the infiltration of cytotoxic cells, B cells, T cells, T helper cells, and NK CD56 bright cells in HNSC. Knockdown of SREBF1 inhibited the proliferation and migration of HNSC cells (Hep2 and TU212) and induced apoptosis by downregulating the expression of steroidogenic acute regulatory protein-related lipid transfer 4 (STARD4). Conclusions SREBF1 may promote HNSC proliferation, migration and inhibit apoptosis by upregulating STARD4 and affecting the level of immune cell infiltration.
Collapse
|
25
|
Li L, Yu XJ, Gao L, Cheng L, Sun B, Wang G. Diabetic Ferroptosis and Pancreatic Cancer: Foe or Friend? Antioxid Redox Signal 2022; 37:1206-1221. [PMID: 35996983 DOI: 10.1089/ars.2022.0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Pancreatic cancer and diabetes have a reciprocal causation relationship. As a potential risk factor, diabetes increases morbidity and promotes pancreatic cancer progression. The main mechanisms include islet dysfunction-induced systemic metabolic disorder, pancreatic stellate cell activation, and immunosuppression. Ferroptosis is regarded as regulated cell death, which participates in chemotherapy resistance and is refractory to radiation therapy and immunotherapy. Diabetes-induced ferroptosis causes many complications, but the underlying mechanism of diabetes-related ferroptosis in pancreatic cancer has not been discussed. Recent Advances: Ferroptosis alleviates pancreatic intraepithelial neoplasia (PanIN) progression by activating chronic inflammation. The specific drugs that cause ferroptosis achieve tumor suppression by inducing lipid peroxidation. Ferroptosis plays pro and con roles in cancer. Both the ferroptosis inhibitor and inducer exhibit antitumor effects through killing cancer cells or directly affecting tumor growth. Diabetes-induced ferroptosis contributes to tumor cell death by different components, including tumor cells, fibroblasts, immune cells, and adipocytes. A better understanding of its role in modulating the tumor microenvironment will reveal diabetes-associated ferroptotic features in cancer development, which can be used to figure out possible treatment strategies for cancer patients with hyperglycemia. Critical Issues: We demonstrate the potential roles of diabetes-related ferroptosis in pancreatic cancer progression and discuss ferroptosis-related antitumor effects and therapeutics for pancreatic cancer treatment. Future Directions: Further studies are required to highlight mechanisms of diabetes-mediated ferroptosis in pancreatic cancer tumorigenesis and progression. The antitumor effects of ferroptosis regulators combined with chemotherapy, targeted therapy, or immunotherapy in diabetic patients should be investigated. We hope that pancreatic cancer patients with diabetes will benefit from ferroptosis-related therapies. Antioxid. Redox Signal. 37, 1206-1221.
Collapse
Affiliation(s)
- Le Li
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xing-Jia Yu
- Department of Centric Operating Room, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lei Gao
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Cheng
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gang Wang
- Department of Pancreatic and Biliary Surgery and The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
26
|
Differentially Infiltrated Identification of Novel Diagnostic Biomarkers Associated with Immune Infiltration in Nasopharyngeal Carcinoma. DISEASE MARKERS 2022; 2022:3934704. [DOI: 10.1155/2022/3934704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/19/2022] [Accepted: 10/29/2022] [Indexed: 11/19/2022]
Abstract
Background. The prognostic value of tumor-infiltrating immune cells has been widely studied in nasopharyngeal carcinoma (NPC). However, the role of tumor-infiltrating immune cells in the diagnosis of NPC has not been fully elucidated. Thus, tumor-infiltrating immune cell-related biomarkers in the diagnosis of NPC patients were explored in the current study. Methods. Gene expression profiles of NPC patients were downloaded from the Gene Expression Omnibus (GEO) database. Differentially infiltrating immune cells (DDICs) between NPC and control samples were analyzed by the CIBERSORT algorithm. Weighted gene coexpression network analysis (WGCNA) was performed to screen hub genes significantly correlated with DDIC. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of hub genes were performed with R package clusterProfiler. The diagnostic value of hub genes was evaluated by receiver operating characteristic (ROC) curves. RT-qPCR was conducted to validate the expression patterns of diagnostic markers in NPC and adjacent control tissues. The correlations between diagnostic markers and immunomodulators were analyzed using the TISIDB. The protein-protein interaction (PPI) network based on immunomodulators significantly associated with diagnostic biomarkers was constructed and visualized by STRING. The functional enrichment analysis of genes in the PPI network was analyzed by the WebGestalt online tool. Results. The abundances of memory B cells, plasma cells, follicular helper T cells, activated NK cells, M0 macrophages, M1 macrophages, M2 macrophages, resting mast cells, and activated mast cells were significantly different between NPC and control samples. Dark orange was identified as the hub module, with a total of 371 genes associated with memory B cells, plasma cells, and M0 and M1 macrophages defined as hub genes, which were enriched into immune-related biological processes and pathways. FCER2, KHDRBS2, and IGSF9 were considered diagnostic biomarkers with areas under ROC curves as 0.985, 0.978, and 0.975, respectively. Moreover, real-time reverse transcriptase-polymerase chain reaction (RT-qPCR) suggested that the expression patterns of FCER2, KHDRBS2, and IGSF9 were consistent with the results in GEO datasets. TISIDB analysis revealed that FCER2, KHDRBS2, and IGSF9 had a strong association with 8 immunoinhibitors (BTLA, CD160, CD96, LAG3, PDCD1, TIGIT, CD244, and TGFB1) and 11 immunostimulators (CD27, CD28, CD40LG, CD48, ICOS, KLRC1, KLRK1, TMIGD2, TNFRSF13C, CXCR4, and C10 or f54). The PPI network implied that these 19 immunomodulators had interactions with other 50 genes. WebGestalt analysis demonstrated that 69 genes in the PPI network were enriched into cytokine-cytokine receptor interaction, NF-kappa B signaling pathway, and pathways in cancer. Conclusion. Our study identified novel diagnostic biomarkers and revealed potential immune-related mechanisms in NPC. These findings enlighten the investigation of the molecular mechanisms of tumor-infiltrating immune cells regulating NPC.
Collapse
|
27
|
Shiau JP, Chuang YT, Tang JY, Yang KH, Chang FR, Hou MF, Yen CY, Chang HW. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants (Basel) 2022; 11:1845. [PMID: 36139919 PMCID: PMC9495789 DOI: 10.3390/antiox11091845] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 01/10/2023] Open
Abstract
Oxidative stress and AKT serine-threonine kinase (AKT) are responsible for regulating several cell functions of cancer cells. Several natural products modulate both oxidative stress and AKT for anticancer effects. However, the impact of natural product-modulating oxidative stress and AKT on cell functions lacks systemic understanding. Notably, the contribution of regulating cell functions by AKT downstream effectors is not yet well integrated. This review explores the role of oxidative stress and AKT pathway (AKT/AKT effectors) on ten cell functions, including apoptosis, autophagy, endoplasmic reticulum stress, mitochondrial morphogenesis, ferroptosis, necroptosis, DNA damage response, senescence, migration, and cell-cycle progression. The impact of oxidative stress and AKT are connected to these cell functions through cell function mediators. Moreover, the AKT effectors related to cell functions are integrated. Based on this rationale, natural products with the modulating abilities for oxidative stress and AKT pathway exhibit the potential to regulate these cell functions, but some were rarely reported, particularly for AKT effectors. This review sheds light on understanding the roles of oxidative stress and AKT pathway in regulating cell functions, providing future directions for natural products in cancer treatment.
Collapse
Affiliation(s)
- Jun-Ping Shiau
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
| | - Ya-Ting Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaoshiung Medical University, Kaohsiung 80708, Taiwan
| | - Kun-Han Yang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ming-Feng Hou
- Division of Breast Oncology and Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan or
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
28
|
Li C, Sun S, Tu Y, Zhang H, Yao F, Liao S, Sun S, Li Z, Wang Z. High Glucose Accelerates Tumor Progression by Regulating MEDAG-Mediated Autophagy Levels in Breast Cancer. Int J Biol Sci 2022; 18:4289-4300. [PMID: 35864962 PMCID: PMC9295059 DOI: 10.7150/ijbs.70002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/23/2022] [Indexed: 12/11/2022] Open
Abstract
Recent studies have shown that diabetes is a major risk factor for breast cancer (BC), but the mechanism is incompletely understood. Mesenteric estrogen-dependent adipogenesis (MEDAG) plays a significant role in both glucose uptake and BC development. However, the relationship between MEDAG and BC under high glucose (HG) conditions remains unclear. In our study, MEDAG expression was higher in BC tissue from diabetic patients than in BC tissue from nondiabetic patients. HG promoted BC progression in vitro and in vivo by upregulating MEDAG expression. Furthermore, MEDAG deficiency increased the autophagosome number and autophagic flux. Moreover, inhibition of autophagy partially reversed MEDAG knockdown (MEDAGKD)-induced suppression of tumorigenic biological behaviors and epithelial-mesenchymal transition (EMT) progression. Finally, MEDAG significantly suppressed AMPK phosphorylation. Additionally, the AMPK inhibitor Compound C markedly reduced autophagosome accumulation and antitumor effects in MEDAGKD cells. Treatment with the AMPK activator AICAR exhibited similar effects in MEDAG-overexpressing (MEDAGOE) cells. In conclusion, the MEDAG-AMPK-autophagy axis is vital to BC progression in diabetic patients. Our findings provide a novel treatment target for BC in patients with diabetes.
Collapse
Affiliation(s)
- Chenyuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Si Sun
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Yi Tu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Hanpu Zhang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Feng Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shichong Liao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Zhiyu Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, P. R. China
| |
Collapse
|
29
|
Zhao Q, Lin X, Wang G. Targeting SREBP-1-Mediated Lipogenesis as Potential Strategies for Cancer. Front Oncol 2022; 12:952371. [PMID: 35912181 PMCID: PMC9330218 DOI: 10.3389/fonc.2022.952371] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Sterol regulatory element binding protein-1 (SREBP-1), a transcription factor with a basic helix–loop–helix leucine zipper, has two isoforms, SREBP-1a and SREBP-1c, derived from the same gene for regulating the genes of lipogenesis, including acetyl-CoA carboxylase, fatty acid synthase, and stearoyl-CoA desaturase. Importantly, SREBP-1 participates in metabolic reprogramming of various cancers and has been a biomarker for the prognosis or drug efficacy for the patients with cancer. In this review, we first introduced the structure, activation, and key upstream signaling pathway of SREBP-1. Then, the potential targets and molecular mechanisms of SREBP-1-regulated lipogenesis in various types of cancer, such as colorectal, prostate, breast, and hepatocellular cancer, were summarized. We also discussed potential therapies targeting the SREBP-1-regulated pathway by small molecules, natural products, or the extracts of herbs against tumor progression. This review could provide new insights in understanding advanced findings about SREBP-1-mediated lipogenesis in cancer and its potential as a target for cancer therapeutics.
Collapse
Affiliation(s)
- Qiushi Zhao
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Xingyu Lin
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Xingyu Lin, ; Guan Wang,
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
- *Correspondence: Xingyu Lin, ; Guan Wang,
| |
Collapse
|
30
|
Lou Q, Zhang M, Yang Y, Gao Y. Low-dose arsenic trioxide enhances membrane-GLUT1 expression and glucose uptake via AKT activation to support L-02 cell aberrant proliferation. Toxicology 2022; 475:153237. [PMID: 35714947 DOI: 10.1016/j.tox.2022.153237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 10/18/2022]
Abstract
Long term low dose exposure of arsenic has been reported to lead various cells proliferation and malignant transformation. GLUT1, as the key transporter of glucose, has been reported to have association with rapid proliferation of various cells or tumor cells. In our study, we found that low dose exposure to arsenic trioxide (0.1μmol/L As2O3) could induce an increase in glucose uptake and promote cell viability and DNA synthesis. And, 2-DG, a non-metabolized glucose analog, significantly decreased the glucose uptake and cell proliferation of 0.1μmol/L As2O3 treated L-02 cells. However, 4 mmol/L 2-DG was co-utilized with equal dose glucose had no significant effect on the cell proliferation of 0.1μmol/L As2O3 treated L-02 cells. Further studies showed that exposure to 0.1μmol/L As2O3 could promote the expression of GLUT1 on plasma membrane. Inhibition of GLUT1 expression by 5μmol/L BAY-876 significantly decreased the abilities of glucose uptake and cell proliferation in As2O3-treated L-02 cells. Moreover, 0.1μmol/L As2O3 induced the AKT activation indicated by increased the phospho-AKT (Ser473 and Thr308). Knockdown AKT by shRNA or inhibited AKT activation by LY294002 was followed by significantly decreased glucose uptake, GLUT1 plasma membrane expression and cell proliferation in As2O3-treated L-02 cells. All in all, these results demonstrated that arsenic trioxide-induced AKT activation contributed to the cells proliferation through upregulating expression of GLUT1 on plasma membrane that enhanced glucose uptake.
Collapse
Affiliation(s)
- Qun Lou
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Meichen Zhang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin Medical University, Harbin 150081, Heilongjiang Province, China.
| |
Collapse
|
31
|
Wang WM, Shen H, Liu ZN, Chen YY, Hou LJ, Ding Y. Interaction between tumor microenvironment, autophagy, and epithelial-mesenchymal transition in tumor progression. Cancer Treat Res Commun 2022; 32:100592. [PMID: 35728404 DOI: 10.1016/j.ctarc.2022.100592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
Tumor microenvironment (TME) is the ecosystem surrounding a tumor to influence tumor cells' growth, metastasis and immunological battlefield, in which the tumor systems fight against the body system. TME has been considered as the essential link between the tumorigenesis and development of neoplasm. Both nutrients intake and tumor progression to malignancy require the participation of components in TME. Epithelial-mesenchymal transition (EMT) is a key step in the metastasis of tumor cells. Cells that lost polarity and acquired migration ability are prone to metastasize. Autophagy is an important self-protective mechanism in tumor cells and a necessity for the tumor cells to respond to harmful stress. Protective autophagy benefits tumor cells while abnormal autophagy leads to cell injury or death. EMT and autophagy are directly regulated by TME. To date, there are numerous studies on TME, autophagy and EMT separately, but few on their complex interrelationships. This review aims to comprehensively analyze the existing mechanisms and convincing evidence so far to seek novel therapeutic strategies and research directions.
Collapse
Affiliation(s)
- Wen-Ming Wang
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Hua Shen
- Department of Mathematics and Statistics, University of Calgary, Alberta T2N 1N4, Canada
| | - Zi-Ning Liu
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Yuan-Yuan Chen
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Li-Jun Hou
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong, 261053, China.
| | - Yi Ding
- Laboratory of Pathophysiology, Weifang Medical University, Weifang, Shandong, 261053, China; Key Laboratory of Applied Pharmacology, Weifang Medical University, Weifang, Shandong, 261053, China.
| |
Collapse
|
32
|
Lai HC, Kuo YW, Huang YH, Chan SM, Cheng KI, Wu ZF. Pancreatic Cancer and Microenvironments: Implications of Anesthesia. Cancers (Basel) 2022; 14:cancers14112684. [PMID: 35681664 PMCID: PMC9179559 DOI: 10.3390/cancers14112684] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Pancreatic cancer is a lethal malignant neoplasm with less than 10% 5-year relative survival after the initial diagnosis. Several factors may be related to the poor prognosis of pancreatic cancer, including the rapid tumor progression, increased metastatic propensity, insignificant symptoms, shortage of early diagnostic biomarkers, and its tendency toward resistance to both chemotherapy and radiotherapy. Pancreatic neoplastic cells interact intimately with a complicated microenvironment that can foster drug resistance, metastasis, or relapse in pancreatic cancer. In addition, evidence shows that perioperative factors, including surgical manipulation, anesthetics, or analgesics, might alter the tumor microenvironment and cancer progression. This review outlines the up-to-date knowledge of anesthesia implications in the pancreatic microenvironment and provides future anesthetic strategies for improving pancreatic cancer survival. Abstract Pancreatic malignancy is a lethal neoplasm, as well as one of the leading causes of cancer-associated mortality, having a 5-year overall survival rate of less than 10%. The average life expectancy of patients with advanced pancreatic cancer does not exceed six months. Although surgical excision is a favorable modality for long-term survival of pancreatic neoplasm, metastasis is initially identified in nearly 80% of the patients by the time of diagnosis, making the development of therapeutic policy for pancreatic cancer extremely daunting. Emerging evidence shows that pancreatic neoplastic cells interact intimately with a complicated microenvironment that can foster drug resistance, metastasis, or relapse in pancreatic cancer. As a result, the necessity of gaining further insight should be focused on the pancreatic microenvironment contributing to cancer progression. Numerous evidence reveals that perioperative factors, including surgical manipulation and anesthetics (e.g., propofol, volatile anesthetics, local anesthetics, epidural anesthesia/analgesia, midazolam), analgesics (e.g., opioids, non-steroidal anti-inflammatory drugs, tramadol), and anesthetic adjuvants (such as ketamine and dexmedetomidine), might alter the tumor microenvironment and cancer progression by affecting perioperative inflammatory or immune responses during cancer surgery. Therefore, the anesthesiologist plays an important role in perioperative management and may affect surgical outcomes. However, the literature on the impact of anesthesia on the pancreatic cancer microenvironment and progression is limited. This review summarizes the current knowledge of the implications of anesthesia in the pancreatic microenvironment and provides future anesthetic strategies for improving pancreatic cancer survival rates.
Collapse
Affiliation(s)
- Hou-Chuan Lai
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei 114, Taiwan; (H.-C.L.); (Y.-H.H.); (S.-M.C.)
| | - Yi-Wei Kuo
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-W.K.); (K.-I.C.)
| | - Yi-Hsuan Huang
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei 114, Taiwan; (H.-C.L.); (Y.-H.H.); (S.-M.C.)
| | - Shun-Ming Chan
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei 114, Taiwan; (H.-C.L.); (Y.-H.H.); (S.-M.C.)
| | - Kuang-I Cheng
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-W.K.); (K.-I.C.)
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Zhi-Fu Wu
- Department of Anesthesiology, Tri-Service General Hospital and National Defense Medical Center, Taipei 114, Taiwan; (H.-C.L.); (Y.-H.H.); (S.-M.C.)
- Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-W.K.); (K.-I.C.)
- Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Regional Anesthesia and Pain Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Correspondence:
| |
Collapse
|
33
|
Ding Y, Wang L, Li H, Miao F, Zhang Z, Hu C, Yu W, Tang Q, Shao G. Application of lipid nanovesicle drug delivery system in cancer immunotherapy. J Nanobiotechnology 2022; 20:214. [PMID: 35524277 PMCID: PMC9073823 DOI: 10.1186/s12951-022-01429-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has gradually emerged as the most promising anticancer therapy. In addition to conventional anti-PD-1/PD-L1 therapy, anti-CTLA-4 therapy, CAR-T therapy, etc., immunotherapy can also be induced by stimulating the maturation of immune cells or inhibiting negative immune cells, regulating the tumor immune microenvironment and cancer vaccines. Lipid nanovesicle drug delivery system includes liposomes, cell membrane vesicles, bacterial outer membrane vesicles, extracellular vesicles and hybrid vesicles. Lipid nanovesicles can be used as functional vesicles for cancer immunotherapy, and can also be used as drug carriers to deliver immunotherapy drugs to the tumor site for cancer immunotherapy. Here, we review recent advances in five kinds of lipid nanovesicles in cancer immunotherapy and assess the clinical application prospects of various lipid nanovesicles, hoping to provide valuable information for clinical translation in the future.
Collapse
Affiliation(s)
- Yinan Ding
- Medical School of Southeast University, Nanjing, 210009, China
| | - Luhong Wang
- Medical School of Southeast University, Nanjing, 210009, China
| | - Han Li
- Department of Tuberculosis, the Second Affiliated Hospital of Southeast University (the Second Hospital of Nanjing), Nanjing, 210009, China
| | - Fengqin Miao
- Medical School of Southeast University, Nanjing, 210009, China
| | - Zhiyuan Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Chunmei Hu
- Department of Tuberculosis, the Second Affiliated Hospital of Southeast University (the Second Hospital of Nanjing), Nanjing, 210009, China
| | - Weiping Yu
- Medical School of Southeast University, Nanjing, 210009, China.
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, 210009, China.
| | - Guoliang Shao
- Department of Interventional Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
34
|
Cancer cell's internal and external warriors: Autophagosomes and exosomes. Life Sci 2022; 300:120552. [PMID: 35452638 DOI: 10.1016/j.lfs.2022.120552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/09/2022] [Accepted: 04/10/2022] [Indexed: 12/24/2022]
Abstract
"That survival instinct, that will to live, that need to get back to life again, is more powerful than any consideration of taste, decency, politeness, manners, civility, anything. It's such a powerful force." This quote by famous director Danny Boyle is a perfect analogy to describe the cancer cell's inexhaustible drive to persist against all odds. In order to adapt to a hostile environment, the cancer cells rely on multiple mechanisms including immune escape, epithelial to mesenchymal transition, angiogenesis, extravasation, autophagy, exosome release among others. Cancer cells depute their internal and external warriors, autophagosomes and exosomes, to dwell in the belligerent tumor microenvironment. It is quite reasonable for a cancer cell, striving to survive, to invest in pathways that will provide the maximum advantage. Autophagy is an important cellular degradation pathway, while the exosome pathway provides an alternative cargo disposal mechanism to maintain the homeostasis and cell survival. While autophagic degradation provides the essential nutrients to rapidly dividing cells, exosomal secretion ensures that the tumor microenvironment is attuned to accommodate the swiftly expanding tumor mass. Studies have revealed that exosomes secreted by cancer cells can modulate autophagy in recipient cells, while autophagy can influence the biogenesis of exosomes. Autophagy and exosome crosstalk is extremely complex and it is only beginning to be recognized and documented. This review is focused on discussing the roles of autophagy and exosomes in the cancer cell's adaptation to the tumor microenvironment and how the two pathways are coordinately regulated to facilitate cancer cell survival.
Collapse
|
35
|
Hsu YS, Wu PJ, Jeng YM, Hu CM, Lee WH. Differential effects of glucose and N-acetylglucosamine on genome instability. Am J Cancer Res 2022; 12:1556-1576. [PMID: 35530290 PMCID: PMC9077085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/19/2022] [Indexed: 06/14/2023] Open
Abstract
Aberrant sugar metabolism is linked to an increased risk of pancreatic cancer. Previously, we found that high glucose induces genome instability and de novo oncogenic KRAS mutation preferentially in pancreatic cells through dysregulation of O-GlcNAcylation. Increasing O-GlcNAcylation by extrinsically supplying N-acetyl-D-glucosamine (GlcNAc) causes genome instability in all kinds of cell types regardless of pancreatic origin. Since many people consume excessive amount of sugar (glucose, fructose, and sucrose) in daily life, whether high sugar consumption directly causes genome instability in animals remains to be elucidated. In this communication, we show that excess sugar in the daily drink increases DNA damage and protein O-GlcNAcylation preferentially in pancreatic tissue but not in other kinds of tissue of mice. The effect of high sugar on the pancreatic tissue may be attributed to the intrinsic ratio of GFAT and PFK activity, a limiting factor that dictates UDP-GlcNAc levels. On the other hand, GlcNAc universally induces DNA damage in all six organs examined. Either inhibiting O-GlcNAcylation or supplementing dNTP pool diminishes the induced DNA damage in these organs, indicating that the mechanism of action is similar to that of high glucose treatment in pancreatic cells. Taken together, these results suggest the potential hazards of high sugar drinks and high glucosamine intake to genomic instability and possibly cancer initiation.
Collapse
Affiliation(s)
- Yuan-Sheng Hsu
- Graduate Institute of Biomedical Science, China Medical UniversityTaichung 40402, Taiwan
- Genomics Research Center, Academia SinicaTaipei 11529, Taiwan
| | - Pei-Jung Wu
- Genomics Research Center, Academia SinicaTaipei 11529, Taiwan
| | - Yung-Ming Jeng
- Department of Pathology, National Taiwan University Hospital, Graduate Institute of Pathology, College of Medicine, National Taiwan UniversityTaipei 10041, Taiwan
| | - Chun-Mei Hu
- Graduate Institute of Biomedical Science, China Medical UniversityTaichung 40402, Taiwan
- Genomics Research Center, Academia SinicaTaipei 11529, Taiwan
| | - Wen-Hwa Lee
- Genomics Research Center, Academia SinicaTaipei 11529, Taiwan
- Drug Development Center, China Medical UniversityTaichung 40402, Taiwan
- Department of Biological Chemistry, University of CaliforniaIrvine, California 92697, USA
| |
Collapse
|
36
|
Shen J, Dong J, Shao F, Zhao J, Gong L, Wang H, Chen W, Zhang Y, Cai Y. Graphene oxide induces autophagy and apoptosis via ROS-dependent AMPK/mTOR/ULK-1 pathway in colorectal cancer cells. Nanomedicine (Lond) 2022; 17:591-605. [PMID: 35394351 DOI: 10.2217/nnm-2022-0030] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To investigate the anticancer effects and action mechanism of graphene oxide (GO) in colorectal cancer (CRC). Materials & methods: Anticancer effects and mechanisms of GO in CRC were investigated both in vivo and in vitro. Results: GO significantly inhibited tumor growth both in vitro and in vivo. GO was able to enter HCT116 cells through endocytosis. GO treatment resulted in cytotoxicity, reactive oxygen species (ROS) production, apoptosis, autophagy and activation of the AMPK/mTOR/ULK1 signal pathway. However, ROS scavenger N-acetylcysteine (NAC) attenuated the above effects and restored the effects of GO on protein expressions related to apoptosis, autophagy and AMPK/mTOR/ULK1 signal pathways. Conclusion: GO exerts anticancer effects against CRC via ROS-dependent AMPK/mTOR/ULK-1 pathway-related autophagy and apoptosis.
Collapse
Affiliation(s)
- Jiamen Shen
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Jiatian Dong
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Feng Shao
- Key Laboratory of Thin Film & Microfabrication Technology (Ministry of Education), School of Electronics, Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jiaying Zhao
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Lifeng Gong
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Huipeng Wang
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Wenjie Chen
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| | - Yafei Zhang
- Key Laboratory of Thin Film & Microfabrication Technology (Ministry of Education), School of Electronics, Information & Electrical Engineering, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yuankun Cai
- Department of General Surgery, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
37
|
Zhang L, Zhang W, Zhang X, Min Y, Zhao Y, Wang B, Li W, Mao S, Min W. High‑glucose microenvironment promotes perineural invasion of pancreatic cancer via activation of hypoxia inducible factor 1α. Oncol Rep 2022; 47:64. [PMID: 35088883 PMCID: PMC8848475 DOI: 10.3892/or.2022.8275] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/29/2021] [Indexed: 11/26/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal diseases, with a 5-year survival rate of <9%. Perineural invasion (PNI) is a common pathological hallmark of PC and is correlated with a poor prognosis in this disease. Hyperglycemia has been shown to promote the invasion and migration of PC cells; however, the effect of hyperglycemia on the PNI of PC and its underlying mechanism remains unclear. In the present study, Western blotting was utilized to detect the expression of hypoxia inducible factor 1α (HIF1α) and nerve growth factor (NGF). Transwell and wound-healing assays were performed to detect the influence of hyperglycemia on the invasion and migration ability of PC cells. An in vitro PC-dorsal root ganglion (DRG) co-culture system and an in vivo PNI sciatic nerve-infiltrating tumor model were used to evaluate the severity of PNI in hyperglycemic conditions. In the results, hyperglycemia promoted the invasion/migration ability and elevated the expression of NGF in PC by upregulating HIF1α. Moreover, in vitro short-term hyperglycemia caused little damage on the DRG axons and accelerated both the PNI of the PC and the outgrowth of the DRGs by increasing the expression of NGF via activation of HIF1α. Indeed, in vivo long-term hyperglycemia promoted the infiltration and growth of PC, and then disrupted the function of the sciatic nerve in a HIF1α-dependent manner. In conclusion, a high-glucose microenvironment promotes PNI of PC via activation of HIF1α.
Collapse
Affiliation(s)
- Lun Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wunai Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xin Zhang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yihe Min
- Chongqing Three Gorges Medical College, Chongqing 404100, P.R. China
| | - Yang Zhao
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Baofeng Wang
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Wei Li
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shuai Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Weili Min
- Department of Oncology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
38
|
Sebestyén A, Dankó T, Sztankovics D, Moldvai D, Raffay R, Cervi C, Krencz I, Zsiros V, Jeney A, Petővári G. The role of metabolic ecosystem in cancer progression — metabolic plasticity and mTOR hyperactivity in tumor tissues. Cancer Metastasis Rev 2022; 40:989-1033. [PMID: 35029792 PMCID: PMC8825419 DOI: 10.1007/s10555-021-10006-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022]
Abstract
Despite advancements in cancer management, tumor relapse and metastasis are associated with poor outcomes in many cancers. Over the past decade, oncogene-driven carcinogenesis, dysregulated cellular signaling networks, dynamic changes in the tissue microenvironment, epithelial-mesenchymal transitions, protein expression within regulatory pathways, and their part in tumor progression are described in several studies. However, the complexity of metabolic enzyme expression is considerably under evaluated. Alterations in cellular metabolism determine the individual phenotype and behavior of cells, which is a well-recognized hallmark of cancer progression, especially in the adaptation mechanisms underlying therapy resistance. In metabolic symbiosis, cells compete, communicate, and even feed each other, supervised by tumor cells. Metabolic reprogramming forms a unique fingerprint for each tumor tissue, depending on the cellular content and genetic, epigenetic, and microenvironmental alterations of the developing cancer. Based on its sensing and effector functions, the mechanistic target of rapamycin (mTOR) kinase is considered the master regulator of metabolic adaptation. Moreover, mTOR kinase hyperactivity is associated with poor prognosis in various tumor types. In situ metabolic phenotyping in recent studies highlights the importance of metabolic plasticity, mTOR hyperactivity, and their role in tumor progression. In this review, we update recent developments in metabolic phenotyping of the cancer ecosystem, metabolic symbiosis, and plasticity which could provide new research directions in tumor biology. In addition, we suggest pathomorphological and analytical studies relating to metabolic alterations, mTOR activity, and their associations which are necessary to improve understanding of tumor heterogeneity and expand the therapeutic management of cancer.
Collapse
|
39
|
Li X, Liu Z, Xia C, Yan K, Fang Z, Fan Y. SETD8 stabilized by USP17 epigenetically activates SREBP1 pathway to drive lipogenesis and oncogenesis of ccRCC. Cancer Lett 2021; 527:150-163. [PMID: 34942305 DOI: 10.1016/j.canlet.2021.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023]
Abstract
Recently, epigenetic modifications, including DNA methylation, histone modification and noncoding RNA (ncRNA)-associated gene silencing, have received increasing attention from the scientific community. Many studies have demonstrated that epigenetic regulation can render dynamic alterations in the transcriptional potential of a cell, which then affects the cell's biological function. The initiation and development of clear cell renal cell carcinoma (ccRCC), the most common subtype of renal cell cancer (RCC), is also closely related to genomic alterations by epigenetic modification. For ccRCC, lipid accumulation is one of the most typical characteristics. In other words, dysregulation of lipid uptake and synthesis occurs in ccRCC, which inversely promotes cancer proliferation and progression. However, the link among epigenetic alterations, lipid biosynthesis and renal cancer progression remains unclear. SETD8 is a histone methyltransferase and plays pivotal roles in cell cycle regulation and oncogenesis of various cancers, but its role in RCC is not well understood. In this study, we discovered that SETD8 was significantly overexpressed in RCC tumors, which was positively related to lipid storage and correlated with advanced tumor grade and stage and poor patient prognosis. Depletion of SETD8 by siRNAs or inhibitor UNC0379 diminished fatty acid (FA) de novo synthesis, cell proliferation and metastasis in ccRCC cells. Mechanistically, SETD8, which was posttranslationally stabilized by USP17, could transcriptionally modulate sterol regulatory element-binding protein 1 (SREBP1), a key transcription factor in fatty acid biosynthesis and lipogenesis, by monomethylating the 20th lysine of the H4 histone, elevating lipid biosynthesis and accumulation in RCC and further promoting cancer progression and metastasis. Taken together, the USP17/SETD8/SREBP1 signaling pathway plays a pivotal role in promoting RCC progression. SETD8 might be a novel biomarker and potential therapeutic target for treating RCC.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji Nan 250012, Shandong, China; Key Laboratory of Cardio-vascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health, China.
| | - Zhengfang Liu
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji Nan 250012, Shandong, China.
| | - Chuanyou Xia
- The First Affiliated Hospital of Shandong First Medical University/Shandong Provincial Qian-Fo-Shan Hospital, China.
| | - Keqiang Yan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji Nan 250012, Shandong, China.
| | - Zhiqing Fang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji Nan 250012, Shandong, China.
| | - Yidong Fan
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji Nan 250012, Shandong, China.
| |
Collapse
|
40
|
Xu C, Zhang L, Wang D, Jiang S, Cao D, Zhao Z, Huang M, Jin J. Lipidomics reveals that sustained SREBP-1-dependent lipogenesis is a key mediator of gefitinib-acquired resistance in EGFR-mutant lung cancer. Cell Death Discov 2021; 7:353. [PMID: 34775471 PMCID: PMC8590692 DOI: 10.1038/s41420-021-00744-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/17/2021] [Accepted: 10/14/2021] [Indexed: 12/24/2022] Open
Abstract
Patients with EGFR mutations in non-small cell lung cancer (NSCLC) have been greatly benefited from gefitinib, however, the therapeutic has failed due to the presence of acquired resistance. In this study, we show that gefitinib significantly induces downregulation of Sterol Regulator Element Binding (SREBP1) in therapy-sensitive cells. However, this was not observed in EGFR mutant NSCLC cells with acquired resistance. Lipidomics analysis showed that gefitinib could differently change the proportion of saturated phospholipids and unsaturated phospholipids in gefitinib-sensitive and acquired-resistant cells. Besides, levels of ROS and MDA were increased upon SREBP1 inhibition and even more upon gefitinib treatment. Importantly, inhibition of SREBP1 sensitizes EGFR-mutant therapy-resistant NSCLC to gefitinib both in vitro and in vivo models. These data suggest that sustained de novo lipogenesis through the maintenance of active SRBEP-1 is a key feature of acquired resistance to gefitinib in EGFR mutant lung cancer. Taken together, targeting SREBP1-induced lipogenesis is a promising approach to overcome acquired resistance to gefitinib in EGFR-mutant lung cancer.
Collapse
Affiliation(s)
- Chuncao Xu
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Lei Zhang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Daifei Wang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shiqin Jiang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Di Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhongxiang Zhao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Jin
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
41
|
Hu Y, Wang K, Ye C. "Four-in-One" Nanozyme and Natural Enzyme Symbiotic System of Cu 2-x Se-GOx for Cervical Cancer Therapy. Chemistry 2021; 28:e202102885. [PMID: 34773414 DOI: 10.1002/chem.202102885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Indexed: 12/19/2022]
Abstract
Cervical cancer, as a common malignant tumor of the reproductive system, seriously threatens women's life and health, and is difficult to be cured by traditional treatments, such as surgery, chemotherapy and radiotherapy. Fortunately, tumor microenvironment (TME)-activated catalytic therapy with high efficiency and reduced off-target toxicity has emerged as a novel treatment model. Herein, we designed a "four-in-one" nanozyme and natural enzyme symbiotic system of Cu2-x Se-GOx for TME-triggered cascaded catalytic enhanced cancer treatment. In response to unique TME, Cu2-x Se with catalase activity could effectively catalyze over-expressed H2 O2 in cancer cells into O2 . Subsequently, the glucose oxidase (GOx) could deplete intracellular glucose with the assistance of O2 ; this not only achieves starvation therapy, but also regenerates H2 O2 to boost the generation of highly cytotoxic . OH due to the peroxidase activity of Cu2-x Se. Moreover, although the free-radical scavenger glutathione (GSH) is overexpressed in tumor cells, Cu2-x Se with glutathione oxidase activity could effectively consume GSH for enhanced ROS production. Thus, the "four-in-one" nanozyme@natural enzyme symbiotic system of Cu2-x Se-GOx could induce significant ROS accumulation at the tumor regions, thus providing a potential approach for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Yubo Hu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University Changchun, Jilin, 130000, P. R. China
| | - Ke Wang
- Department of Gynaecology and Obstetrics, China-Japan Union Hospital of Jilin University Changchun, Jilin, 130000, P. R. China
| | - Cong Ye
- Department of Gynaecology and Obstetrics, China-Japan Union Hospital of Jilin University Changchun, Jilin, 130000, P. R. China
| |
Collapse
|
42
|
Li J, Chen X, Kang R, Zeh H, Klionsky DJ, Tang D. Regulation and function of autophagy in pancreatic cancer. Autophagy 2021; 17:3275-3296. [PMID: 33161807 PMCID: PMC8632104 DOI: 10.1080/15548627.2020.1847462] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/12/2022] Open
Abstract
Oncogenic KRAS mutation-driven pancreatic ductal adenocarcinoma is currently the fourth-leading cause of cancer-related deaths in the United States. Macroautophagy (hereafter "autophagy") is one of the lysosome-dependent degradation systems that can remove abnormal proteins, damaged organelles, or invading pathogens by activating dynamic membrane structures (e.g., phagophores, autophagosomes, and autolysosomes). Impaired autophagy (including excessive activation and defects) is a pathological feature of human diseases, including pancreatic cancer. However, dysfunctional autophagy has many types and plays a complex role in pancreatic tumor biology, depending on various factors, such as tumor stage, microenvironment, immunometabolic state, and death signals. As a modulator connecting various cellular events, pharmacological targeting of nonselective autophagy may lead to both good and bad therapeutic effects. In contrast, targeting selective autophagy could reduce potential side effects of the drugs used. In this review, we describe the advances and challenges of autophagy in the development and therapy of pancreatic cancer.Abbreviations: AMPK: AMP-activated protein kinase; CQ: chloroquine; csc: cancer stem cells; DAMP: danger/damage-associated molecular pattern; EMT: epithelial-mesenchymal transition; lncRNA: long noncoding RNA; MIR: microRNA; PanIN: pancreatic intraepithelial neoplasia; PDAC: pancreatic ductal adenocarcinoma; PtdIns3K: phosphatidylinositol 3-kinase; SNARE: soluble NSF attachment protein receptor; UPS: ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Xin Chen
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Herbert Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel J. Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
43
|
Roda N, Blandano G, Pelicci PG. Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance. Cancers (Basel) 2021; 13:cancers13174471. [PMID: 34503281 PMCID: PMC8431382 DOI: 10.3390/cancers13174471] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The interactions between cancer cells and the surrounding blood vessels and peripheral nerves are critical in all the phases of tumor development. Accordingly, therapies that specifically target vessels and nerves represent promising anticancer approaches. The first aim of this review is to document the importance of blood vessels and peripheral nerves in both cancer onset and local or distant growth of tumoral cells. We then focus on the state-of-the-art therapies that limit cancer progression through the impairment of blood vessels and peripheral nerves. The mentioned literature is helpful for the scientific community to appreciate the recent advances in these two fundamental components of tumors. Abstract Cancer cells continuously interact with the tumor microenvironment (TME), a heterogeneous milieu that surrounds the tumor mass and impinges on its phenotype. Among the components of the TME, blood vessels and peripheral nerves have been extensively studied in recent years for their prominent role in tumor development from tumor initiation. Cancer cells were shown to actively promote their own vascularization and innervation through the processes of angiogenesis and axonogenesis. Indeed, sprouting vessels and axons deliver several factors needed by cancer cells to survive and proliferate, including nutrients, oxygen, and growth signals, to the expanding tumor mass. Nerves and vessels are also fundamental for the process of metastatic spreading, as they provide both the pro-metastatic signals to the tumor and the scaffold through which cancer cells can reach distant organs. Not surprisingly, continuously growing attention is devoted to the development of therapies specifically targeting these structures, with promising initial results. In this review, we summarize the latest evidence that supports the importance of blood vessels and peripheral nerves in cancer pathogenesis, therapy resistance, and innovative treatments.
Collapse
Affiliation(s)
- Niccolò Roda
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (N.R.); (G.B.)
| | - Giada Blandano
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (N.R.); (G.B.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (N.R.); (G.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Correspondence:
| |
Collapse
|
44
|
Roy A, Sahoo J, Kamalanathan S, Naik D, Mohan P, Kalayarasan R. Diabetes and pancreatic cancer: Exploring the two-way traffic. World J Gastroenterol 2021; 27:4939-4962. [PMID: 34497428 PMCID: PMC8384733 DOI: 10.3748/wjg.v27.i30.4939] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is often associated with a poor prognosis. Long-standing diabetes mellitus is considered as an important risk factor for its development. This risk can be modified by the use of certain antidiabetic medications. On the other hand, new-onset diabetes can signal towards an underlying PC in the elderly population. Recently, several attempts have been made to develop an effective clinical tool for PC screening using a combination of history of new-onset diabetes and several other clinical and biochemical markers. On the contrary, diabetes affects the survival after treatment for PC. We describe this intimate and complex two-way relationship of diabetes and PC in this review by exploring the underlying pathogenesis.
Collapse
Affiliation(s)
- Ayan Roy
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences, Jodhpur 342005, India
| | - Jayaprakash Sahoo
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Sadishkumar Kamalanathan
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Dukhabandhu Naik
- Department of Endocrinology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Pazhanivel Mohan
- Department of Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| | - Raja Kalayarasan
- Department of Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry 605006, India
| |
Collapse
|
45
|
Ninfole E, Pinto C, Benedetti A, Marzioni M, Maroni L. Role of autophagy in cholangiocarcinoma: Pathophysiology and implications for therapy. World J Clin Cases 2021; 9:6234-6243. [PMID: 34434990 PMCID: PMC8362566 DOI: 10.12998/wjcc.v9.i22.6234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/26/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumour of the biliary system that originates from the neoplastic transformation of cholangiocytes. CCA is characterized by late diagnosis and poor outcome, with surgery considered as the last option for management. Autophagy is a physiological lysosomal degradation process, essential for cellular homeostasis and ubiquitous in all eukaryotic cells. Several studies have reported a potential involvement of autophagy in cancer, but it remains unclear whether activation of this process represents a survival mechanism of cancer cells. In the present review, we examine the autophagic process and summarize the current knowledge about the involvement of autophagy in the progression of cancer. The link between autophagy and chemoresistance and the use of autophagic markers in diagnosis are also considered in detail. Preliminary evidence shows that the combination of autophagy modulators (activators or inhibitors) with conventional chemotherapeutic agents offers a possible treatment option against signalling pathways that are hyperactivated or altered in CCA. In vitro evidence suggests that combination of chemotherapy agents, such as cisplatin, under activation or inhibition of autophagic processes, in two different CCA cell lines, may improve chemosensitivity and reduce cell survival, respectively. A deeper understanding of these pathways, in both cancer and non-cancer cells, could unveil possible therapeutic targets to treat CCA patients.
Collapse
Affiliation(s)
- Elisabetta Ninfole
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Claudio Pinto
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Antonio Benedetti
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Marco Marzioni
- Department of Gastroenterology, Università Politecnica delle Marche, Ancona 60126, Italy
| | - Luca Maroni
- Department of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona 60126, Italy
| |
Collapse
|
46
|
Yang M, Chen X, Zhang J, Xiong E, Wang Q, Fang W, Li L, Fei F, Gong A. ME2 Promotes Proneural-Mesenchymal Transition and Lipogenesis in Glioblastoma. Front Oncol 2021; 11:715593. [PMID: 34381734 PMCID: PMC8351415 DOI: 10.3389/fonc.2021.715593] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/20/2022] Open
Abstract
Malic enzyme 2 (ME2) catalyzes the formation of pyruvate from malic acid and is abnormally expressed in some tumors. However, the exact effects of ME2 on proneural–mesenchymal transition (PMT) and lipogenesis in glioblastoma multiforme (GBM) remain unexplored. Here, we found that ME2 expression was significantly higher in GBM than in normal brain tissues and negatively correlated with overall survival of patients with GBM. Furthermore, we demonstrated that ME2 was positively correlated with mesenchymal features in GBM and promoted proliferation, migration, and invasion of glioma cells. Moreover, ME2 upregulated the expression of mesenchymal markers (N-cadherin, vimentin, YKL40, and MET), whereas it inhibited the expression of proneural maker OLIG2, indicating that ME2 might promote PMT in GBM. We also found that ME2 inhibited the production of mitochondrial reactive oxygen species and AMPK phosphorylation, resulting in SREBP-1 maturation and nuclear localization and enhancing the ACSS2 lipogenesis pathway. Taken together, these results suggest that ME2 promotes PMT and is linked with reprogramming of lipogenesis via AMPK–SREBP-1–ACSS2 signaling in GBM. Therefore, ME2 has potential as a new classification marker in GBM and could provide a new approach to glioma treatment.
Collapse
Affiliation(s)
- Mengting Yang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xi Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Junyao Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ermeng Xiong
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Qianqian Wang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Wenjing Fang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fei Fei
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
47
|
Wang TB, Geng M, Jin H, Tang AG, Sun H, Zhou LZ, Chen BH, Shen G, Sun Q. SREBP1 site 1 protease inhibitor PF-429242 suppresses renal cell carcinoma cell growth. Cell Death Dis 2021; 12:717. [PMID: 34285190 PMCID: PMC8292369 DOI: 10.1038/s41419-021-03999-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022]
Abstract
Renal cell carcinoma (RCC) cells have increased lipogenesis and cholesterol synthesis. Sterol regulatory element-binding protein-1 (SREBP1) is cleaved by site 1 protease (S1P) to release the transcriptionally active amino-terminal domain. PF-429242 is a potent and competitive S1P inhibitor. We here tested its activity in RCC cells. In established and primary human RCC cells, PF-429242 potently inhibited cell proliferation, migration, and invasion. The S1P inhibitor provoked apoptosis activation in RCC cells. Furthermore, shRNA-mediated S1P silencing or CRISPR/Cas9-induced S1P knockout led to RCC cell growth inhibition and apoptosis activation. Conversely, ectopic overexpression of SREBP1 or S1P augmented RCC cell proliferation and migration. Daily i.v. injection of a single dose of PF-429242 robustly inhibited RCC xenograft growth in severe combined immunodeficiency mice. Additionally, intratumoral injection of S1P shRNA lentivirus inhibited RCC xenograft growth in mice. SREBP1, S1P, and its target gene low density lipoprotein receptor (LDLR) were significantly elevated in human RCC tissues. These results suggest that targeting S1P by PF-429242 inhibited RCC cell growth in vitro and in vivo.
Collapse
Affiliation(s)
- Tong-Bing Wang
- Department of Urology, People's Hospital of Yangzhong City, Yangzhong, China
| | - Mei Geng
- Department of Oncology, Rui Jin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Jin
- The Child Health Care Department, Suzhou Ninth People's Hospital, Suzhou, China
| | - Ai-Guo Tang
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hao Sun
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Liu-Zheng Zhou
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Bin-Hai Chen
- Department of Urology, The affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Gang Shen
- Department of Urology, DUSHU Lake Hospital Affiliated to Soochow University, Suzhou, China.
| | - Qiang Sun
- Department of Nephrology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| |
Collapse
|
48
|
Wang Y, Qin C, Yang G, Zhao B, Wang W. The role of autophagy in pancreatic cancer progression. Biochim Biophys Acta Rev Cancer 2021; 1876:188592. [PMID: 34274449 DOI: 10.1016/j.bbcan.2021.188592] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/03/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022]
Abstract
Patients with pancreatic cancer have an abysmal survival rate. The poor prognosis of pancreatic cancer is due to the difficulty of making an early diagnosis, high rate of metastasis, and frequent chemoresistance. In recent years, as a self-regulatory procedure within cells, the effect and mechanism of autophagy have been explored. Dysregulated autophagy serves as a double-edged sword in cancer development in which autophagy inhibits cancer initiation but promotes cancer progression. After tumor formation, activation of autophagy can induce epithelial-mesenchymal transition, regulate metabolism, specifically glutamine usage and the glycolytic process, and mediate drug resistance in pancreatic cancer. Multiple genes, RNA molecules, proteins, and certain drugs exert antitumor effects by inhibiting autophagy-mediated drug resistance. Several clinical trials have combined autophagy inhibitors with chemotherapeutic drugs in pancreatic cancer treatment, some of which have shown promising results. In conclusion, autophagy plays a vital role in pancreatic cancer progression and deserves further study.
Collapse
Affiliation(s)
- Yuanyang Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China.
| | - Cheng Qin
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Gang Yang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Bangbo Zhao
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China
| | - Weibin Wang
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, PR China.
| |
Collapse
|
49
|
Supabphol S, Seubwai W, Wongkham S, Saengboonmee C. High glucose: an emerging association between diabetes mellitus and cancer progression. J Mol Med (Berl) 2021; 99:1175-1193. [PMID: 34036430 DOI: 10.1007/s00109-021-02096-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/16/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The association of cancer and diabetes mellitus (DM) has been studied for decades. Hyperglycemia and the imbalance of hormones are factors that contribute to the molecular link between DM and carcinogenesis and cancer progression. Hyperglycemia alone or in combination with hyperinsulinemia are key factors that promote cancer aggressiveness. Many preclinical studies suggest that high glucose induces abnormal energy metabolism and aggressive cancer via several mechanisms. As evidenced by clinical studies, hyperglycemia is associated with poor clinical outcomes in patients who have comorbid DM. The prognoses of cancer patients with DM are improved when their plasma glucose levels are controlled. This suggests that high glucose level maybe be involved in the molecular mechanism that causes the link between DM and cancer and may also be useful for prognosis of cancer progression. This review comprehensively summarizes the evidence from recent pre-clinical and clinical studies of the impact of hyperglycemia on cancer advancement as well as the underlying molecular mechanism for this impact. Awareness among clinicians of the association between hyperglycemia or DM and cancer progression may improve cancer treatment outcome in patients who have DM.
Collapse
Affiliation(s)
- Suangson Supabphol
- The Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Wunchana Seubwai
- Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sopit Wongkham
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand.,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Charupong Saengboonmee
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Center for Translational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
50
|
Wang Q, Hu R, Li W, Tai Y, Gu W, Das BC, Yang F, Ji J, Wang C, Zhou J. BF175 inhibits endometrial carcinoma through SREBP-regulated metabolic pathways in vitro. Mol Cell Endocrinol 2021; 523:111135. [PMID: 33359761 DOI: 10.1016/j.mce.2020.111135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023]
Abstract
Elevated lipogenesis is an important metabolic hallmark of rapidly proliferating tumor such as endometrial carcinoma (EC). The sterol regulatory element-binding protein 1 (SREBP1) is a master regulator of lipogenesis and involved in EC proliferation. BF175 is a novel chemical inhibitor of SREBP pathway, and has shown potent anti-lipogenic effects. However, the effect of BF175 on EC cells are yet to be determined. In the present study, we found that BF175 decreased cell viability, colony formation and migratory capacity, inducing autophagy and mitochondrial related apoptosis in EC cell line AN3CA. Z-VAD-FMK partially attenuated the effect of BF175 on AN3CA. In addition, BF175 significantly downregulated SREBPs and their downstream genes. The levels of free fatty acids and total cholesterol were also inhibited. Microarray analysis suggested BF175 treatment obviously affected lipid metabolic pathways in EC. Taken together, we validated BF175 exhibited anti-tumor activity by targeting SREBP-dependent lipogenesis and inducing apoptosis which mitochondrial pathway involved in, suggesting that it's potential as a novel therapeutic reagent for EC.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ruofan Hu
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihua Li
- Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yanhong Tai
- Department of Pathology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Weiting Gu
- Departments of Cancer Biology, Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bhaskar C Das
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fajun Yang
- Departments of Medicine and Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Junyuan Ji
- Department of Molecular and Cellular Medicine, Colleage of Medicine, Texas A&M University Health Science Center, College Station, TX, USA
| | - Chenguang Wang
- Departments of Cancer Biology, Stem Cell Biology and Regenerative Medicine, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jie Zhou
- Department of Endocrinology and Metabolism, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|