1
|
Eberhard E, Burger L, Pastrana CL, Seyed-Allaei H, Giunta G, Gerland U. Force Generation by Enhanced Diffusion in Enzyme-Loaded Vesicles. NANO LETTERS 2025. [PMID: 40138661 DOI: 10.1021/acs.nanolett.5c00306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The diffusion coefficient of some metabolic enzymes increases with the concentration of their cognate substrate, a phenomenon known as enhanced diffusion. In the presence of substrate gradients, enhanced diffusion induces enzymatic drift, resulting in a nonhomogeneous enzyme distribution. Here, we study the effects of enhanced diffusion on enzyme-loaded vesicles placed in external substrate gradients using a combination of computer simulations and analytical modeling. We observe that the spatially inhomogeneous enzyme profiles generated by enhanced diffusion result in a pressure gradient across the vesicle, which leads to macroscopically observable effects, namely deformation and self-propulsion of the vesicle. Our analytical model allows us to characterize the dependence of the velocity of propulsion on experimentally tunable parameters. The effects predicted by our work provide an avenue for further validation of enhanced diffusion, and might be leveraged for the design of novel synthetic cargo transporters, such as targeted drug delivery systems.
Collapse
Affiliation(s)
- Eike Eberhard
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Ludwig Burger
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - César L Pastrana
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Hamid Seyed-Allaei
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Giovanni Giunta
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Ulrich Gerland
- Physics of Complex Biosystems, Department of Bioscience, School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
2
|
Yan L, Shi J, Zhu J. Cellular and molecular events in colorectal cancer: biological mechanisms, cell death pathways, drug resistance and signalling network interactions. Discov Oncol 2024; 15:294. [PMID: 39031216 PMCID: PMC11265098 DOI: 10.1007/s12672-024-01163-1] [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: 05/27/2024] [Accepted: 07/15/2024] [Indexed: 07/22/2024] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths worldwide, affecting millions each year. It emerges from the colon or rectum, parts of the digestive system, and is closely linked to both genetic and environmental factors. In CRC, genetic mutations such as APC, KRAS, and TP53, along with epigenetic changes like DNA methylation and histone modifications, play crucial roles in tumor development and treatment responses. This paper delves into the complex biological underpinnings of CRC, highlighting the pivotal roles of genetic alterations, cell death pathways, and the intricate network of signaling interactions that contribute to the disease's progression. It explores the dysregulation of apoptosis, autophagy, and other cell death mechanisms, underscoring the aberrant activation of these pathways in CRC. Additionally, the paper examines how mutations in key molecular pathways, including Wnt, EGFR/MAPK, and PI3K, fuel CRC development, and how these alterations can serve as both diagnostic and prognostic markers. The dual function of autophagy in CRC, acting as a tumor suppressor or promoter depending on the context, is also scrutinized. Through a comprehensive analysis of cellular and molecular events, this research aims to deepen our understanding of CRC and pave the way for more effective diagnostics, prognostics, and therapeutic strategies.
Collapse
Affiliation(s)
- Lei Yan
- Medical Department, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jia Shi
- Department of Obstetrics and Gynecology, The Central Hospital of Shaoyang Affiliated to University of South China, Shaoyang, China
| | - Jiazuo Zhu
- Department of Oncology, Xuancheng City Central Hospital, No. 117 Tong Road, Xuancheng, Anhui, China.
| |
Collapse
|
3
|
Lee PWT, Suwa T, Kobayashi M, Yang H, Koseki LR, Takeuchi S, Chow CCT, Yasuhara T, Harada H. Hypoxia- and Postirradiation reoxygenation-induced HMHA1/ARHGAP45 expression contributes to cancer cell invasion in a HIF-dependent manner. Br J Cancer 2024; 131:37-48. [PMID: 38740970 PMCID: PMC11231347 DOI: 10.1038/s41416-024-02691-x] [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: 07/19/2023] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Cancer cells in severely hypoxic regions have been reported to invade towards tumour blood vessels after surviving radiotherapy in a postirradiation reoxygenation- and hypoxia-inducible factor (HIF)-dependent manner and cause recurrence. However, how HIF induces invasiveness of irradiated and reoxygenated cancer cells remains unclear. METHODS Here, we identified human minor histocompatibility antigen 1 (HMHA1), which has been suggested to function in cytoskeleton dynamics and cellular motility, as a responsible factor and elucidated its mechanism of action using molecular and cellular biology techniques. RESULTS HMHA1 expression was found to be induced at the transcription initiation level in a HIF-dependent manner under hypoxia. Boyden chamber invasion assay revealed that the induction of HMHA1 expression is required for the increase in invasion of hypoxic cancer cells. Reoxygenation treatment after ionising radiation in vitro that mimics dynamic changes of a microenvironment in hypoxic regions of tumour tissues after radiation therapy further enhanced HMHA1 expression and invasive potential of HMHA1 wildtype cancer cells in ROS- and HIF-dependent manners, but not of HMHA1 knockout cells. CONCLUSION These results together provide insights into a potential molecular mechanism of the acquisition of invasiveness by hypoxic cancer cells after radiotherapy via the activation of the ROS/HIF/HMHA1 axis.
Collapse
Affiliation(s)
- Peter W T Lee
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Tatsuya Suwa
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Hui Yang
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Lina R Koseki
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Satoshi Takeuchi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Christalle C T Chow
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Takaaki Yasuhara
- Laboratory of Genome Stress Response, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
- Department of Late Effects Studies, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
4
|
Lin F, Long Y, Li M, Cai C, Wu Y, You X, Tian X, Zhou Q. Xihuang pills targeting the Warburg effect through inhibition of the Wnt/β-catenin pathway in prostate cancer. Heliyon 2024; 10:e32914. [PMID: 38994113 PMCID: PMC11237975 DOI: 10.1016/j.heliyon.2024.e32914] [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: 12/03/2023] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
Objective Prostate cancer, marked by a high incidence and mortality rate, presents a significant challenge, especially in the context of castration-resistant prostate cancer (CRPC) with limited treatment options due to drug resistance. This study aims to explore the anti-tumor effects of Xihuang Pills (XHP) on CRPC, focusing on metabolic reprogramming and the Wnt/β-catenin pathway. Methods In vitro and in vivo biofunctional assays were employed to assess the efficacy and mechanisms of XHP. Subcutaneous xenografts of PC3 in mice served as an in vivo model to evaluate XHP's anti-tumor activity. Tumor volume, weight, proliferation, and apoptosis were monitored. Various assays, including CCK8, TUNEL assay, QRT-PCR, and Western Blotting, were conducted to measure metabolic reprogramming, proliferation, apoptosis, and cell cycle in prostate cancer cells. RNA-seq analysis predicted XHP's impact on prostate cancer, validating the expression of Wnt/β-catenin-related proteins and mRNA. Additionally, 58 compounds in XHP were identified via LC-MS/MS, and molecular docking analysis connected these compounds to key genes. Results In vitro and in vivo experiments demonstrated that XHP significantly inhibited CRPC cell viability, induced apoptosis, and suppressed invasion and migration. mRNA sequencing revealed differentially expressed genes, with functional enrichment analysis indicating modulation of key biological processes. XHP treatment downregulated Wnt signaling pathway-related genes, including CCND2, PRKCG, and CCN4. Moreover, XHP effectively inhibited glucose uptake and lactate production, leading to reduced HIF-1α and glycolytic enzymes (GLUT1, HK2, PKM2), suggesting its potential in attenuating the Warburg effect. Molecular docking analysis suggested a plausible interaction between XHP's active compounds and Wnt1 protein, indicating a mechanism through which XHP modulates the Wnt/β-catenin pathway. Conclusion XHP demonstrated remarkable efficacy in suppressing the growth, proliferation, apoptosis, migration, and invasiveness of prostate tumors. The interaction between XHP's active constituents and Wnt1 was evident, leading to the inhibition of Wnt1 and downstream anti-carcinogenic factors, thereby influencing the β-catenin/HIF-1α-mediated glycolysis.
Collapse
Affiliation(s)
- Fengxia Lin
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
- Department of Cardiovascular, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Yan Long
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
- Graduate School of Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Mingyue Li
- Department of Pharmacy, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Changlong Cai
- Department of Urology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Yongrong Wu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xujun You
- Department of Andrology, Shenzhen Bao'an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong Province, China
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Qing Zhou
- Department of Andrology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, Hunan Province, China
| |
Collapse
|
5
|
Zhang X, Zheng P, Meng B, Zhuang H, Lu B, Yao J, Han F, Luo S. Histamine-related genes participate in the establishment of an immunosuppressive microenvironment and impact the immunotherapy response in hepatocellular carcinoma. Clin Exp Med 2024; 24:129. [PMID: 38884870 PMCID: PMC11182831 DOI: 10.1007/s10238-024-01399-9] [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: 05/06/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Chronic inflammation is pivotal in the pathogenesis of hepatocellular carcinoma (HCC). Histamine is a biologically active substance that amplifies the inflammatory and immune response and serves as a neurotransmitter. However, knowledge of histamine's role in HCC and its effects on immunotherapy remains lacking. We focused on histamine-related genes to investigate their potential role in HCC. The RNA-seq data and clinical information regarding HCC were obtained from The Cancer Genome Atlas (TCGA). After identifying the differentially expressed genes, we constructed a signature using the univariate Cox proportional hazard regression and least absolute shrinkage and selection operator (LASSO) analyses. The signature's predictive performance was evaluated using a receiver operating characteristic curve (ROC) analysis. Furthermore, drug sensitivity, immunotherapy effects, and enrichment analyses were conducted. Histamine-related gene expression in HCC was confirmed using quantitative real-time polymerase chain reaction (qRT-PCR). A histamine-related gene prognostic signature (HRGPS) was developed in TCGA. Time-dependent ROC and Kaplan-Meier survival analyses demonstrated the signature's strong predictive power. Importantly, patients in high-risk groups exhibited a higher frequency of TP53 mutations, elevated immune checkpoint-related gene expression, and increased infiltration of immunosuppressive cells-indicating a potentially favorable response to immunotherapy. In addition, drug sensitivity analysis revealed that the signature could effectively predict chemotherapy efficacy and sensitivity. qRT-PCR results validated histamine-related gene overexpression in HCC. Our findings demonstrate that inhibiting histamine-related genes and signaling pathways can impact the therapeutic effect of anti-PD-1/PD-L1. The precise predictive ability of our signature in determining the response to different therapeutic options highlights its potential clinical significance.
Collapse
Affiliation(s)
- Xianzhou Zhang
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Peng Zheng
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Bo Meng
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Hao Zhuang
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Bing Lu
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Jun Yao
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Feng Han
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Suxia Luo
- Department of Hepatic Biliary Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
6
|
Lee PWT, Koseki LR, Haitani T, Harada H, Kobayashi M. Hypoxia-Inducible Factor-Dependent and Independent Mechanisms Underlying Chemoresistance of Hypoxic Cancer Cells. Cancers (Basel) 2024; 16:1729. [PMID: 38730681 PMCID: PMC11083728 DOI: 10.3390/cancers16091729] [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: 04/18/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/13/2024] Open
Abstract
In hypoxic regions of malignant solid tumors, cancer cells acquire resistance to conventional therapies, such as chemotherapy and radiotherapy, causing poor prognosis in patients with cancer. It is widely recognized that some of the key genes behind this are hypoxia-inducible transcription factors, e.g., hypoxia-inducible factor 1 (HIF-1). Since HIF-1 activity is suppressed by two representative 2-oxoglutarate-dependent dioxygenases (2-OGDDs), PHDs (prolyl-4-hydroxylases), and FIH-1 (factor inhibiting hypoxia-inducible factor 1), the inactivation of 2-OGDD has been associated with cancer therapy resistance by the activation of HIF-1. Recent studies have also revealed the importance of hypoxia-responsive mechanisms independent of HIF-1 and its isoforms (collectively, HIFs). In this article, we collate the accumulated knowledge of HIF-1-dependent and independent mechanisms responsible for resistance of hypoxic cancer cells to anticancer drugs and briefly discuss the interplay between hypoxia responses, like EMT and UPR, and chemoresistance. In addition, we introduce a novel HIF-independent mechanism, which is epigenetically mediated by an acetylated histone reader protein, ATAD2, which we recently clarified.
Collapse
Affiliation(s)
- Peter Wai Tik Lee
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan (L.R.K.)
| | - Lina Rochelle Koseki
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan (L.R.K.)
| | - Takao Haitani
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan (L.R.K.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Harada
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan (L.R.K.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Minoru Kobayashi
- Laboratory of Cancer Cell Biology, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan (L.R.K.)
- Department of Genome Repair Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
7
|
Yang J, Wan S, Zhao M, Cai H, Gao Y, Wang H. Multi-omics Analysis Identifies Hypoxia Subtypes and S100A2 as an Immunosuppressive Factor in Cervical Cancer. Reprod Sci 2024; 31:107-121. [PMID: 37648942 DOI: 10.1007/s43032-023-01304-x] [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: 01/10/2023] [Accepted: 07/10/2023] [Indexed: 09/01/2023]
Abstract
Cervical cancer is a common gynecological oncology. Growing evidence indicates hypoxia plays an important role in tumor progression and immunity. However, no study has examined the hypoxia landscape in cervical cancer. In this study, using hierarchical clustering, we identified three hypoxia subtypes in cervical cancer samples from The Cancer Genome Atlas dataset according to formerly described hypoxia-related genes. The overall survival time, hypoxic features, genomics, and immunological characteristics of these subtypes existed distinct differences. We also created a hypoxia score by principle component analysis for dimension reduction. The hypoxiaScore was an effective prognostic biomarker validated by GSE44001 and was associated with immunotherapy response. Furthermore, combined with single-cell RNA-sequence (scRNA-seq) and experiments, S100A2 was identified as an immunosuppressive factor induced by hypoxia and regulated expression of PD-L1. S100A2 also served as an oncogene promoting the proliferation and migration of cervical cancer cells. These findings depicted a new hypoxia-based classification and identified S100A2 as a potential therapeutic target for cervical cancer, thereby advancing the understanding of immunotherapy resistance mechanisms and cervical cancer genetic markers.
Collapse
Affiliation(s)
- Junyuan Yang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Gynecology, Maternal and ChildHealth Hospital of Hubei Province, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Shimeng Wan
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Mengna Zhao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
| | - Hongbing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.
- Hubei Cancer Clinical Study Center, Wuhan, China.
| | - Yang Gao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.
- Hubei Cancer Clinical Study Center, Wuhan, China.
| | - Hua Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China.
- Hubei Cancer Clinical Study Center, Wuhan, China.
| |
Collapse
|
8
|
Zhang Q, Wang X, Zhao Y, Cheng Z, Fang D, Liu Y, Tian G, Li M, Luo Z. Nanointegrative In Situ Reprogramming of Tumor-Intrinsic Lipid Droplet Biogenesis for Low-Dose Radiation-Activated Ferroptosis Immunotherapy. ACS NANO 2023; 17:25419-25438. [PMID: 38055636 DOI: 10.1021/acsnano.3c08907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Low-dose radiotherapy (LDR) has shown significant implications for inflaming the immunosuppressive tumor microenvironment (TME). Surprisingly, we identify that FABP-dependent lipid droplet biogenesis in tumor cells is a key determinant of LDR-evoked cytotoxic and immunostimulatory effects and developed a nanointegrated strategy to promote the antitumor efficacy of LDR through cooperative ferroptosis immunotherapy. Specifically, TCPP-TK-PEG-PAMAM-FA, a nanoscale multicomponent functional polymer with self-assembly capability, was synthesized for cooperatively entrapping hafnium ions (Hf4+) and HIF-1α-inhibiting siRNAs (siHIF-1α). The TCPP@Hf-TK-PEG-PAMAM-FA@siHIF-1α nanoassemblies are specifically taken in by folate receptor-overexpressing tumor cells and activated by the elevated cellular ROS stress. siHIF-1α could readily inhibit the FABP3/7 expression in tumor cells via HIF-1α-FABP3/7 signaling and abolish lipid droplet biogenesis for enhancing the peroxidation susceptibility of membrane lipids, which synergizes with the elevated ROS stress in the context of Hf4+-enhanced radiation exposure and evokes pronounced ferroptotic damage in vital membrane structures. Interestingly, TCPP@Hf-TK-PEG-PAMAM-FA@siHIF-1α-enhanced ferroptotic biomembrane damage also facilitates the exposure of tumor-associated antigens (TAAs) to promote post-LDR immunotherapeutic effects, leading to robust tumor regression in vivo. This study offers a nanointegrative approach to boost the antitumor effects of LDR through the utilization of tumor-intrinsic lipid metabolism.
Collapse
Affiliation(s)
- Qiqi Zhang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yuanyuan Zhao
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Zhuo Cheng
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, P. R. China
| | - De Fang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Gan Tian
- Chongqing Institute of Advanced Pathology, Jinfeng Laboratory, Chongqing 401329, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
9
|
Miyabara H, Hirano R, Watanabe S, Soriano JCC, Watanabe H, Kuchimaru T, Kitada N, Kadonosono T, Maki SA, Kondoh G, Kizaka‐Kondoh S. In vivo optical imaging of tumor stromal cells with hypoxia-inducible factor activity. Cancer Sci 2023; 114:3935-3945. [PMID: 37482942 PMCID: PMC10551579 DOI: 10.1111/cas.15907] [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: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Tumors contain various stromal cells, such as immune cells, endothelial cells, and fibroblasts, which contribute to the development of a tumor-specific microenvironment characterized by hypoxia and inflammation, and are associated with malignant progression. In this study, we investigated the activity of intratumoral hypoxia-inducible factor (HIF), which functions as a master regulator of the cellular response to hypoxia and inflammation. We constructed the HIF activity-monitoring reporter gene hypoxia-response element-Venus-Akaluc (HVA) that expresses the green fluorescent protein Venus and modified firefly luciferase Akaluc in a HIF activity-dependent manner, and created transgenic mice harboring HVA transgene (HVA-Tg). In HVA-Tg, HIF-active cells can be visualized using AkaBLI, an ultra-sensitive in vivo bioluminescence imaging technology that produces an intense near-infrared light upon reaction of Akaluc with the D-luciferin analog AkaLumine-HCl. By orthotopic transplantation of E0771, a mouse triple negative breast cancer cell line without a reporter gene, into HVA-Tg, we succeeded in noninvasively monitoring bioluminescence signals from HIF-active stromal cells as early as 8 days after transplantation. The HIF-active stromal cells initially clustered locally and then spread throughout the tumors with growth. Immunohistochemistry and flow cytometry analyses revealed that CD11b+ F4/80+ macrophages were the predominant HIF-active stromal cells in E0771 tumors. These results indicate that HVA-Tg is a useful tool for spatiotemporal analysis of HIF-active tumor stromal cells, facilitating investigation of the roles of HIF-active tumor stromal cells in tumor growth and malignant progression.
Collapse
Affiliation(s)
- Hitomi Miyabara
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Ryuichiro Hirano
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Shigeaki Watanabe
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | | | - Hitomi Watanabe
- Institute for Life and Medical SciencesKyoto UniversityKyotoJapan
| | - Takahiro Kuchimaru
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
- Center for Molecular MedicineJichi Medical UniversityShimotsukeJapan
| | - Nobuo Kitada
- Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Tetsuya Kadonosono
- School of Life Science and TechnologyTokyo Institute of TechnologyYokohamaJapan
| | - Shojiro A. Maki
- Graduate School of Informatics and EngineeringThe University of Electro‐CommunicationsChofuJapan
| | - Gen Kondoh
- Institute for Life and Medical SciencesKyoto UniversityKyotoJapan
| | | |
Collapse
|
10
|
Nakamura N, Yoshida N, Suwa T. Three major reasons why transgenerational effects of radiation are difficult to detect in humans. Int J Radiat Biol 2023; 100:1297-1311. [PMID: 36880868 DOI: 10.1080/09553002.2023.2187478] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/18/2023] [Indexed: 03/08/2023]
Abstract
PURPOSE Ionizing radiation can induce mutations in germ cells in various organisms, including fruit flies and mice. However, currently, there is no clear evidence for the transgenerational effects of radiation in humans. This review is an effort to identify possible reasons for the lack of such observations. METHODS Literature search and narrative review. RESULTS 1) In both mice and humans, resting oocytes locate primarily in the cortical region of the ovary where the number of blood vessels is very low especially when young and extra-cellular material is rich, and this region is consequently hypoxic, which probably leads to immature oocytes being resistant to the cell killing and mutagenic effects of radiation. 2) In studies of spermatogonia, the mouse genes used for specific locus test (SLT) studies, which include coat color genes, were hypermutable when compared to many other genes. Recent studies which examined over 1000 segments of genomic DNA indicate that the induction rate of deletion mutation per segment was on the order of 10-6 per Gy, which is one order of magnitude lower than that obtained from the SLT data. Therefore, it appears possible that detecting any transgenerational effects of radiation following human male exposures will be difficult due to a lack of mutable marker genes. 3) Fetal malformations were examined in studies in humans, but the genetic component in such malformations is low, and abnormal fetuses are prone to undergo miscarriage which does not occur in mice, and which leads to difficulties in detecting transgenerational effects. CONCLUSION The lack of clear evidence for radiation effects in humans probably does not result from any problem in the methodologies used but may be due largely to biological properties. Currently, whole genome sequencing studies of exposed parents and offspring are planned, but ethical guidelines need to be followed to avoid discrimination, which had once happened to the atomic bomb survivors.
Collapse
Affiliation(s)
- Nori Nakamura
- Department of Molecular Biosciences, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Noriaki Yoshida
- Department of Clinical Studies, Radiation Effects Research Foundation, Hiroshima, Japan
| | - Tatsuya Suwa
- Department of Genome Dynamics, Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
11
|
Li R, Huang B, Tian H, Sun Z. Immune evasion in esophageal squamous cell cancer: From the perspective of tumor microenvironment. Front Oncol 2023; 12:1096717. [PMID: 36698392 PMCID: PMC9868934 DOI: 10.3389/fonc.2022.1096717] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Esophageal cancer (EC) is one of the most life-threatening malignancies worldwide. Esophageal squamous cell carcinoma (ESCC) is the dominant subtype, accounting for approximately 90% of new incident EC each year. Although multidisciplinary treatment strategies have advanced rapidly, patients with ESCC are often diagnosed at advanced stage and the long-term prognosis remains unsatisfactory. In recent decades, immunotherapy, such as immune checkpoint inhibitors (ICIs), tumor vaccines, and chimeric antigen receptor T-cell (CAR-T) therapy, has been successfully used in clinical practice as a novel therapy for treating tumors, bringing new hope to ESCC patients. However, only a small fraction of patients achieved clinical benefits due to primary or acquired resistance. Immune evasion plays a pivotal role in the initiation and progression of ESCC. Therefore, a thorough understanding of the mechanisms by which ESCC cells escape from anti-tumor immunity is necessary for a more effective multidisciplinary treatment strategy. It has been widely recognized that immune evasion is closely associated with the crosstalk between tumor cells and the tumor microenvironment (TME). TME is a dynamic complex and comprehensive system including not only cellular components but also non-cellular components, which influence hallmarks and fates of tumor cells from the outside. Novel immunotherapy targeting tumor-favorable TME represents a promising strategy to achieve better therapeutic responses for patients with ESCC. In this review, we provide an overview of immune evasion in ESCC, mainly focusing on the molecular mechanisms that underlie the role of TME in immune evasion of ESCC. In addition, we also discuss the challenges and opportunities of precision therapy for ESCC by targeting TME.
Collapse
|
12
|
Zou Y, Chen X, Cheng Z, Chen H, Wu J, Liu H, Ye Q. Evaluation of nitroreductase activity in nasopharyngeal carcinoma progression by an activatable two-photon fluorescent probe. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 281:121616. [PMID: 35841858 DOI: 10.1016/j.saa.2022.121616] [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: 01/20/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Nasopharyngeal carcinoma (NPC) originating from the epithelium cells is the most common malignant tumor of the head and neck. Small-molecule fluorescent probes for early diagnosis of NPC can effectively improve the 5-year survival rate of patients, which makes it become a research hotspot in recent years. Previous studies have suggested the expression levels of NTR in hypoxic tissues or cells and tumors increased relative to the normal state and were positively correlated with the degree of hypoxia. Regarding the mentioned above, we designed a two-photon fluorescent probe NaT-NTR for the detection of NTR in nasopharyngeal cell lines and tissues at different hypoxia levels. NaT-NTR showed high selectivity and sensitivity toward NTR in a complex physiological environment. Furthermore, imaging NTR in different cell lines revealed that the level of intracellular NTR might be positively correlated with the malignancy of nasopharyngeal carcinoma. More importantly, NaT-NTR was successfully applied to detect and image NTR in human nasopharyngeal carcinoma with a penetration depth of 100 µm. On this basis, NaT-NTR might be a powerful chemical tool for the early diagnosis of nasopharyngeal carcinoma.
Collapse
Affiliation(s)
- Yuxia Zou
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Xianghui Chen
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China
| | - Ziyi Cheng
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Hongwei Chen
- Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| | - Jinsheng Wu
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China
| | - Heng Liu
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou 571199, China; Key Laboratory of Emergency and Trauma, Ministry of Education, Key Laboratory of Hainan Trauma and Disaster Rescue, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China.
| | - Qing Ye
- Shengli Clinical Medical College of Fujian Medical University, Department of Otolaryngology, Head and Neck Surgery, Fujian Provincial Hospital, Fuzhou 350001, China.
| |
Collapse
|
13
|
Sarkar D, Chowdhury M, Das PK. Naphthalimide-Based Azo-Functionalized Supramolecular Vesicle in Hypoxia-Responsive Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:3480-3492. [PMID: 35261245 DOI: 10.1021/acs.langmuir.1c03334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Supramolecular materials that respond to external triggers are being extensively utilized in developing spatiotemporal control in biomedical applications ranging from drug delivery to diagnostics. The present article describes the development of self-assembled vesicles in 1:9 (v/v), tetrahydrofuran (THF)-water by naphthalimide-based azo moiety containing amphiphile (NI-Azo) where azo moiety would act as the stimuli-responsive junction. The self-assembly of NI-Azo took place through H-type of aggregation. Microscopic and spectroscopic analyses confirmed the formation of supramolecular vesicles with a dimension of 200-250 nm. Azo (-N═N-) moiety is known to get reduced to amine derivatives in the presence of the azoreductase enzyme, which is overexpressed in the hypoxic microenvironment. The absorbance intensity of this characteristic azo (-N═N-) moiety of NI-Azo (1:9 (v/v), THF-water) at 458 nm got diminished in the presence of both extracellular and intracellular bacterial azoreductase extracted from Escherichia coli bacteria. The same observation was noted in the presence of sodium dithionite (mimic of azoreductase), indicating that azoreductase/sodium dithionite induced azo bond cleavage of NI-Azo, which was confirmed by matrix-assisted laser desorption ionization time-of-flight spectrometric data of the corresponding aromatic amine fragments. The anticancer drug, curcumin, was encapsulated inside NI-Azo vesicles that successfully killed B16F10 cells (cancer cells) in CoCl2-induced hypoxic environment owing to the azoreductase-responsive release of drug. The cancer cell killing efficiency by curcumin-loaded NI-Azo vesicles in the hypoxic condition was 2.15-fold higher than that of the normoxic environment and 2.4-fold higher compared to that of native curcumin in the hypoxic condition. Notably, cancer cell killing efficiency of curcumin-loaded NI-Azo vesicles was 4.5- and 1.9-fold higher than that of noncancerous NIH3T3 cells in normoxic and hypoxic environments, respectively. Cell killing was found to be primarily through the early apoptotic pathway.
Collapse
Affiliation(s)
- Deblina Sarkar
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Monalisa Chowdhury
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| |
Collapse
|
14
|
Liu H, Wang X, Shen P, Ni Y, Han X. The basic functions of phosphoglycerate kinase 1 and its roles in cancer and other diseases. Eur J Pharmacol 2022; 920:174835. [DOI: 10.1016/j.ejphar.2022.174835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/15/2022] [Indexed: 01/17/2023]
|
15
|
Abe Y, Mukudai Y, Kurihara M, Houri A, Chikuda J, Yaso A, Kato K, Shimane T, Shirota T. Tumor protein D52 is upregulated in oral squamous carcinoma cells under hypoxia in a hypoxia-inducible-factor-independent manner and is involved in cell death resistance. Cell Biosci 2021; 11:122. [PMID: 34217360 PMCID: PMC8255020 DOI: 10.1186/s13578-021-00634-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/18/2021] [Indexed: 12/26/2022] Open
Abstract
Background Tumor protein D52 (TPD52) reportedly plays an important role in the proliferation and metastasis of various cancer cells, including oral squamous cell carcinoma (OSCC) cells, and is expressed strongly at the center of the tumor, where the microenvironment is hypoxic. Thus, the present study investigated the roles of TPD52 in the survival and death of OSCC cells under hypoxia, and the relationship with hypoxia-inducible factor (HIF). We examined the expression of TPD52 in OSCC cells under hypoxic conditions and analyzed the effects of HIF on the modulation of TPD52 expression. Finally, the combinational effects of TPD52 knockdown and HIF inhibition were investigated both in vitro and in vivo. Results The mRNA and protein levels of TPD52 increased in OSCC cells under hypoxia. However, the increase was independent of HIF transcription. Importantly, the observation was due to upregulation of mRNA stability by binding of mRNA to T-cell intercellular antigen (TIA) 1 and TIA-related protein (TIAR). Simultaneous knockdown of TPD52 and inhibition of HIF significantly reduced cell viability. In addition, the in vivo tumor-xenograft experiments showed that TPD52 acts as an autophagy inhibitor caused by a decrease in p62. Conclusions This study showed that the expression of TPD52 increases in OSCC cells under hypoxia in a HIF-independent manner and plays an important role in the proliferation and survival of the cells in concordance with HIF, suggesting that novel cancer therapeutics might be led by TPD52 suppression. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00634-0.
Collapse
Affiliation(s)
- Yuzo Abe
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Yoshiki Mukudai
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan.
| | - Mai Kurihara
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Asami Houri
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Junichiro Chikuda
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Atsutoshi Yaso
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Kosuke Kato
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Toshikazu Shimane
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| | - Tatsuo Shirota
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ota-ku, Tokyo, 145-8515, Japan
| |
Collapse
|
16
|
Chen X, Jin R, Jiang Q, Bi Q, He T, Song X, Barz M, Ai H, Shuai X, Nie Y. Delivery of siHIF-1α to Reconstruct Tumor Normoxic Microenvironment for Effective Chemotherapeutic and Photodynamic Anticancer Treatments. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100609. [PMID: 34032365 DOI: 10.1002/smll.202100609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/17/2021] [Indexed: 06/12/2023]
Abstract
The tumor hypoxic microenvironment not only induces genetic and epigenetic changes in tumor cells, immature vessels formation for oxygen demand, but also compromises the efficiency of therapeutic interventions. On the other hand, conventional therapeutic approaches which kill tumor cells or destroy tumor blood vessels to block nutrition and oxygen supply usually facilitate even harsher microenvironment. Thus, simultaneously relieving the strained response of tumor cells and blood vessels represents a promising strategy to reverse the adverse tumor hypoxic microenvironment. In the present study, an integrated amphiphilic system (RSCD) is designed based on Angiotensin II receptor blocker candesartan for siRNA delivery against the hypoxia-inducible factor-1 alpha (HIF-1α), aiming at both vascular and cellular "relaxation" to reconstruct a tumor normoxic microenvironment. Both in vitro and in vivo studies have confirmed that the hypoxia-inducible HIF-1α expression is down-regulated by 70% and vascular growth is inhibited by 60%. The "relaxation" therapy enables neovascularization with more complete and organized structures to obviously increase the oxygen level inside tumor, which results in a 50% growth inhibition. Moreover, reconstruction of tumor microenvironment enhances tumor-targeted drug delivery, and significantly improves the chemotherapeutic and photodynamic anticancer treatments.
Collapse
Affiliation(s)
- Xiaobing Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qian Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qunjie Bi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Ting He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xu Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Matthias Barz
- Leiden Academic Center for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
- Institute of Organic Chemistry, Johannes Gutenberg-University, Mainz Duesbergweg 10-14, 55099, Mainz, Germany
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
17
|
Dao NV, Ercole F, Urquhart MC, Kaminskas LM, Nowell CJ, Davis TP, Sloan EK, Whittaker MR, Quinn JF. Trisulfide linked cholesteryl PEG conjugate attenuates intracellular ROS and collagen-1 production in a breast cancer co-culture model. Biomater Sci 2021; 9:835-846. [PMID: 33231231 DOI: 10.1039/d0bm01544j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The progression of cancer has been closely-linked with augmentation of cellular reactive oxygen species (ROS) levels and ROS-associated changes in the tumour microenvironment (TME), including alterations to the extracellular matrix and associated low drug uptake. Herein we report the application of a co-culture model to simulate the ROS based cell-cell interactions in the TME using fibroblasts and breast cancer cells, and describe how novel reactive polymers can be used to modulate those interactions. Under the co-culture conditions, both cell types exhibited modifications in behaviour, including significant overproduction of ROS in the cancer cells, and elevation of the collagen-1 secretion and stained actin filament intensity in the fibroblasts. To examine the potential of using reactive antioxidant polymers to intercept ROS communication and thereby manipulate the TME, we employed H2S-releasing macromolecular conjugates which have been previously demonstrated to mitigate ROS production in HEK cells. The specific conjugate used, mPEG-SSS-cholesteryl (T), significantly reduced ROS levels in co-cultured cancer cells by approximately 50%. This reduction was significantly greater than that observed with the other positive antioxidant controls. Exposure to T was also found to downregulate levels of collagen-1 in the co-cultured fibroblasts, while exhibiting less impact on cells in mono-culture. This would suggest a possible downstream effect of ROS-mitigation by T on stromal-tumour cell signalling. Since fibroblast-derived collagens modulate crucial steps in tumorigenesis, this ROS-associated effect could potentially be harnessed to slow cancer progression. The model may also be beneficial for interrogating the impact of antioxidants on naturally enhanced ROS levels, rather than relying on the application of exogenous oxidants to simulate elevated ROS levels.
Collapse
Affiliation(s)
- Nam V Dao
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Department of Physical Chemistry and Physics, Hanoi University of Pharmacy, Hanoi 10000, Vietnam
| | - Francesca Ercole
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Matthew C Urquhart
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Lisa M Kaminskas
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas P Davis
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Erica K Sloan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia and Peter MacCallum Cancer Centre, Division of Surgery, Melbourne, VIC 3000, Australia
| | - Michael R Whittaker
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - John F Quinn
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Department of Chemical Engineering, Faculty of Engineering, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| |
Collapse
|
18
|
Energy Metabolism in Cancer: The Roles of STAT3 and STAT5 in the Regulation of Metabolism-Related Genes. Cancers (Basel) 2020; 12:cancers12010124. [PMID: 31947710 PMCID: PMC7016889 DOI: 10.3390/cancers12010124] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/03/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
A central characteristic of many types of cancer is altered energy metabolism processes such as enhanced glucose uptake and glycolysis and decreased oxidative metabolism. The regulation of energy metabolism is an elaborate process involving regulatory proteins such as HIF (pro-metastatic protein), which reduces oxidative metabolism, and some other proteins such as tumour suppressors that promote oxidative phosphorylation. In recent years, it has been demonstrated that signal transducer and activator of transcription (STAT) proteins play a pivotal role in metabolism regulation. STAT3 and STAT5 are essential regulators of cytokine- or growth factor-induced cell survival and proliferation, as well as the crosstalk between STAT signalling and oxidative metabolism. Several reports suggest that the constitutive activation of STAT proteins promotes glycolysis through the transcriptional activation of hypoxia-inducible factors and therefore, the alteration of mitochondrial activity. It seems that STAT proteins function as an integrative centre for different growth and survival signals for energy and respiratory metabolism. This review summarises the functions of STAT3 and STAT5 in the regulation of some metabolism-related genes and the importance of oxygen in the tumour microenvironment to regulate cell metabolism, particularly in the metabolic pathways that are involved in energy production in cancer cells.
Collapse
|
19
|
Guo X, Yuan Z, Xu Y, Wei M, Fang Z, Yuan WE. A fluorinated low-molecular-weight PEI/HIF-1α shRNA polyplex system for hemangioma therapy. Biomater Sci 2020; 8:2129-2142. [DOI: 10.1039/d0bm00171f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
RNAi technology targeting HIF-1α could benefit hemangioma therapy effectively and FPEI polyplexes which could inhibit the expression of HIF-1α at the translational level can provide a practicable strategy for clinical hemangioma treatment in the future.
Collapse
Affiliation(s)
- Xiaoshuang Guo
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Zihan Yuan
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Yang Xu
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Minyan Wei
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Zhiwei Fang
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| | - Wei-En Yuan
- Engineering Research Center of Cell & Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
| |
Collapse
|
20
|
Fluorine-18 click radiosynthesis and microPET/CT evaluation of a small peptide-a potential PET probe for carbonic anhydrase IX. Bioorg Med Chem 2019; 27:785-789. [DOI: 10.1016/j.bmc.2019.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/12/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022]
|
21
|
A feed-forward loop between nuclear translocation of CXCR4 and HIF-1α promotes renal cell carcinoma metastasis. Oncogene 2018; 38:881-895. [PMID: 30177838 PMCID: PMC6367212 DOI: 10.1038/s41388-018-0452-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 05/24/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) has been suggested to play a critical role in cancer metastasis. Some studies have described CXCR4 nuclear localization in metastatic lesions of renal cell carcinoma (RCC), which has been suggested to be correlated with cancer metastasis. However, the underlying mechanism and clinical significance of CXCR4 nuclear localization remains unknown. Here, we show that CXCR4 nuclear localization is more likely to occur in RCC tissues, especially in metastases, and is associated with poor prognosis. CXCR4 nuclear localization requires its nuclear localization sequence (NLS, residues 146-RPRK-149). After the mutation of NLS in CXCR4, CXCR4 nuclear localization in RCC cells is lost. Nuclear localization of CXCR4 promoted RCC tumorigenicity both in vitro and in vivo. Mechanistically, we found that CXCR4 and hypoxia-inducible factor-1α (HIF-1α) colocalized in RCC cells and interacted with each other. Moreover, CXCR4 nuclear localization promoted nuclear accumulation of HIF-1α, thereby promoting the expression of genes downstream of HIF-1α. Reciprocally, nuclear HIF-1α promoted CXCR4 transcription, thus forming a feed-forward loop. Subcellular CXCR4 and HIF-1α expression levels were independent adverse prognostic factors and could be combined with TNM stage to generate a predictive nomogram of the clinical outcome of patients with RCC. Therefore, our findings indicate that CXCR4 nuclear translocation plays a critical role in RCC metastasis and may serve as a prognostic biomarker and potential therapeutic target.
Collapse
|
22
|
Liu JN, Bu W, Shi J. Chemical Design and Synthesis of Functionalized Probes for Imaging and Treating Tumor Hypoxia. Chem Rev 2017; 117:6160-6224. [DOI: 10.1021/acs.chemrev.6b00525] [Citation(s) in RCA: 556] [Impact Index Per Article: 69.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jia-nan Liu
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Wenbo Bu
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
- Shanghai
Key Laboratory of Green Chemistry and Chemical Processes, School of
Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, P.R. China
| | - Jianlin Shi
- State
Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
| |
Collapse
|
23
|
Cao HY, Ding RL, Li M, Yang MN, Yang LL, Wu JB, Yang B, Wang J, Luo CL, Wen QL. Danshensu, a major water-soluble component of Salvia miltiorrhiza, enhances the radioresponse for Lewis Lung Carcinoma xenografts in mice. Oncol Lett 2016; 13:605-612. [PMID: 28356936 PMCID: PMC5351344 DOI: 10.3892/ol.2016.5508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 09/15/2016] [Indexed: 12/21/2022] Open
Abstract
The molecule 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoic acid (danshensu), a herbal preparation used in traditional Chinese medicine, has been found to possess potential antitumor and anti-angiogenesis effects. The aim of the present study was to investigate the efficacy of the combination of radiation therapy (RT) with danshensu in the treatment of Lewis lung carcinoma (LLC) xenografts, whilst exploring and evaluating the mechanism involved. In total, 8-week old female C57BL/6J mice were randomly assigned into 3 groups to receive: RT, RT + cisplatin and RT + danshensu, respectively, when LLC reached 100–150 mm3. Each group was divided into 7 subgroups according to the different irradiation doses that were administered. Tumor growth curves were created and the sensitization enhancement ratios of the drugs were calculated. The experiment was then repeated, and the 4 groups of tumor-bearing mice were treated with natural saline, danshensu, RT + danshensu and RT, respectively. The mice were sacrificed on day 7, and tumor tissue and blood were collected to determine microvessel density, the expression of proangiogenic factors, and the levels of blood thromboxane B2 and 6-keto-prostaglandin-F1α. Tumor hypoxia was also detected using in vivo fluorescence imaging. With respect to LLC xenografts, treatment with danshensu + RT significantly enhanced the effects of tumor growth inhibition (P<0.05). Furthermore, tumor vasculature was remodeled and microcirculation was improved, which significantly reduced tumor hypoxia (P<0.05). The present study demonstrated that danshensu significantly enhanced the radioresponse of LLC xenografts in mice. The mechanism involved may be associated with the alleviation of tumor cell hypoxia following treatment with danshensu + RT, caused by the improvement of tumor microcirculation and the remodeling of tumor vasculature.
Collapse
Affiliation(s)
- Hong-Ying Cao
- Department of Emergency, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Rui-Lin Ding
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Meng Li
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Mao-Nan Yang
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Ling-Lin Yang
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Jing-Bo Wu
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Bo Yang
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Jing Wang
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Cui-Lian Luo
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Qing-Lian Wen
- Department of Oncology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
24
|
Patel A, Sant S. Hypoxic tumor microenvironment: Opportunities to develop targeted therapies. Biotechnol Adv 2016; 34:803-812. [PMID: 27143654 PMCID: PMC4947437 DOI: 10.1016/j.biotechadv.2016.04.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/13/2016] [Accepted: 04/28/2016] [Indexed: 01/18/2023]
Abstract
In recent years, there has been great progress in the understanding of tumor biology and its surrounding microenvironment. Solid tumors create regions with low oxygen levels, generally termed as hypoxic regions. These hypoxic areas offer a tremendous opportunity to develop targeted therapies. Hypoxia is not a random by-product of the cellular milieu due to uncontrolled tumor growth; rather it is a constantly evolving participant in overall tumor growth and fate. This article reviews current trends and recent advances in drug therapies and delivery systems targeting hypoxia in the tumor microenvironment. In the first part, we give an account of important physicochemical changes and signaling pathways activated in the hypoxic microenvironment. This is then followed by various treatment strategies including hypoxia-sensitive signaling pathways and approaches to develop hypoxia-targeted drug delivery systems.
Collapse
Affiliation(s)
- Akhil Patel
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
25
|
Thambi T, Park JH, Lee DS. Hypoxia-responsive nanocarriers for cancer imaging and therapy: recent approaches and future perspectives. Chem Commun (Camb) 2016; 52:8492-500. [DOI: 10.1039/c6cc02972h] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This review focuses on hypoxia-responsive nanocarriers, which can be disintegrated by recognizing the hypoxic microenvironment of cancer cells, and their utilization in cancer imaging and therapy.
Collapse
Affiliation(s)
- Thavasyappan Thambi
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 16419
- Republic of Korea
| | - Jae Hyung Park
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 16419
- Republic of Korea
| | - Doo Sung Lee
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon 16419
- Republic of Korea
| |
Collapse
|
26
|
Van Hove B, Love AM, Ajikumar PK, De Mey M. Programming Biology: Expanding the Toolset for the Engineering of Transcription. Synth Biol (Oxf) 2016. [DOI: 10.1007/978-3-319-22708-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
27
|
Wen Z, Huang C, Xu Y, Xiao Y, Tang L, Dai J, Sun H, Chen B, Zhou M. α-Solanine inhibits vascular endothelial growth factor expression by down-regulating the ERK1/2-HIF-1α and STAT3 signaling pathways. Eur J Pharmacol 2015; 771:93-8. [PMID: 26688571 DOI: 10.1016/j.ejphar.2015.12.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/09/2015] [Accepted: 12/09/2015] [Indexed: 11/19/2022]
Abstract
In tumors, vascular endothelial growth factor (VEGF) contributes to angiogenesis, vascular permeability, and tumorigenesis. In our previous study, we found that α-solanine, which is widespread in solanaceae, has a strong anti-cancer effect under normoxia. However, it is unknown whether α-solanine has a similar effect under hypoxia. We used cobalt chloride (CoCl2) to mimic hypoxia in vitro. HIF-1α, which is almost undetectable under normoxia, was significantly increased. Simultaneously, another regulator of VEGF, STAT3, was also significantly activated by CoCl2. We utilized α-solanine in co-culture with CoCl2. α-solanine decreased the expression of VEGF and loss of E-cadherin. α-solanine also suppressed the activation of phospho-ERK1/2 (p-ERK1/2), HIF-1α, and STAT3 signaling. The results provide new evidence that α-solanine has a strong anti-cancer effect via the ERK1/2-HIF-1α and STAT3 signaling pathways and suggest that it may be a potential new drug.
Collapse
Affiliation(s)
- Zhengde Wen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaohao Huang
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaya Xu
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuwu Xiao
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Tang
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, China
| | - Juji Dai
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongwei Sun
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bicheng Chen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, China
| | - Mengtao Zhou
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
28
|
Zeng Y, Liu Y, Shang J, Ma J, Wang R, Deng L, Guo Y, Zhong F, Bai M, Zhang S, Wu D. Phosphorescence monitoring of hypoxic microenvironment in solid-tumors to evaluate chemotherapeutic effects using the hypoxia-sensitive iridium (III) coordination compound. PLoS One 2015; 10:e0121293. [PMID: 25786221 PMCID: PMC4365010 DOI: 10.1371/journal.pone.0121293] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/29/2015] [Indexed: 02/06/2023] Open
Abstract
Objectives To utilize phosphorescence to monitor hypoxic microenvironment in solid-tumors and investigate cancer chemotherapeutic effects in vivo. Methods A hypoxia-sensitive probe named BTP was used to monitor hypoxic microenvironment in solid-tumors. The low-dose metronomic treatment with cisplatin was used in anti-angiogenetic chemotherapeutic programs. The phosphorescence properties of BTP were detected by a spectrofluorometer. BTP cytotoxicity utilized cell necrosis and apoptosis, which were evaluated by trypan blue dye exclusion and Hoechst33342 plus propidium iodide assays. Tumor-bearing mouse models of colon adenocarcinoma were used for tumor imaging in vivo. Monitoring of the hypoxic microenvironment in tumors was performed with a Maestro 2 fluorescence imaging system. Tumor tissues in each group were harvested regularly and treated with pathological hematoxylin and eosin and immunohistochemical staining to confirm imaging results. Results BTP did not feature obvious cytotoxicity for cells, and tumor growth in low-dose metronomic cisplatin treated mice was significantly inhibited by chemotherapy. Hypoxic levels significantly increased due to cisplatin, as proven by the expression level of related proteins. Phosphorescence intensity in the tumors of mice in the cisplatin group was stronger and showed higher contrast than that in tumors of saline treated mice.
Collapse
Affiliation(s)
- Yun Zeng
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Yang Liu
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Jin Shang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Jingwen Ma
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Rong Wang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Lei Deng
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Youmin Guo
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
| | - Fan Zhong
- School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Mingfeng Bai
- Molecular Imaging Laboratory, Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Shaojuan Zhang
- Radiology Department, First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, P. R. China
- Molecular Imaging Laboratory, Department of Radiology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (SZ); (DW)
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering, Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, P. R. China
- * E-mail: (SZ); (DW)
| |
Collapse
|
29
|
Chen H, Feng J, Zhang Y, Shen A, Chen Y, Lin J, Lin W, Sferra TJ, Peng J. Pien Tze Huang Inhibits Hypoxia-Induced Angiogenesis via HIF-1 α /VEGF-A Pathway in Colorectal Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2015; 2015:454279. [PMID: 25649293 PMCID: PMC4310443 DOI: 10.1155/2015/454279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 12/12/2022]
Abstract
Hypoxia-induced angiogenesis plays an important role in the development and metastasis of solid tumors and is highly regulated by HIF-1α/VEGF-A pathway. Therefore, inhibiting tumor angiogenesis via suppression of HIF-1α/VEGF-A signaling represents a promising strategy for anticancer treatment. As a traditional Chinese medicine formula, Pien Tze Huang (PZH) has long been used as a folk remedy for cancer in China and Southeast Asia. Previously, we reported that PZH inhibits colorectal cancer (CRC) growth both in vivo and in vitro. To elucidate the antitumor mechanisms of PZH, in the present study we used human umbilical vein endothelial cells (HUVEC) and colorectal carcinoma HCT-8 cells to evaluate the effects of PZH on hypoxia-induced angiogenesis and investigated the underlying molecular mechanisms. We found that PZH could inhibit hypoxia-induced migration and tube formation of HUVEC cells in a dose-dependent manner, although the low concentrations of PZH had no effect on HUVEC viability. Moreover, PZH inhibited hypoxia-induced activation of HIF-1α signaling and the expression of VEGF-A and/or VEGFR2 in both HCT-8 and HUVEC cells. Collectively, our findings suggest that PZH can inhibit hypoxia-induced tumor angiogenesis via suppression of HIF-1α/VEGF-A pathway.
Collapse
Affiliation(s)
- Hongwei Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Jianyu Feng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Yuchen Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Youqin Chen
- Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Wei Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
| | - Thomas J. Sferra
- Rainbow Babies & Children's Hospital, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350122, China
- Postdoctoral Workstation, Zhangzhou Pien Tze Huang Pharmaceutical Co., Ltd., Shangjie, Zhangzhou, Fujian 363000, China
| |
Collapse
|
30
|
Li J, Zhang G, Wang X, Li XF. Is carbonic anhydrase IX a validated target for molecular imaging of cancer and hypoxia? Future Oncol 2015; 11:1531-41. [PMID: 25963430 PMCID: PMC4976829 DOI: 10.2217/fon.15.11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The presence of hypoxia is a general feature of most solid malignancies, and hypoxia is considered as one of major factors for anticancer therapy failure. Carbonic anhydrase IX (CAIX) has been reported to be an endogenous hypoxia marker, CAIX monoclonal antibodies, their segments and inhibitors are developed for CAIX imaging. However, growing evidence indicates that CAIX expression under hypoxia condition may be cancer cell lines or cancer-type dependent. Here we review the current literature on CAIX and discuss the advantage and limitation of CAIX as a target for tumor hypoxia imaging. Accordingly, CAIX would be unreliable as a universal target for cancer and tumor hypoxia visualization.
Collapse
Affiliation(s)
- Jianbo Li
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Guojian Zhang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| | - Xuemei Wang
- Department of Nuclear Medicine, Inner Mongolia Medical University Affiliated Hospital, Hohhot, Inner Mongolia, China
| | - Xiao-Feng Li
- Department of Diagnostic Radiology, University of Louisville, 530 S Jackson Street, CCB-C07, Louisville, KY 40202, USA
| |
Collapse
|
31
|
Liang J, Zhang Z, Liang L, Shen Y, Ouyang K. HIF-1α regulated tongue squamous cell carcinoma cell growth via regulating VEGF expression in a xenograft model. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:92. [PMID: 25405167 DOI: 10.3978/j.issn.2305-5839.2014.08.01] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/21/2014] [Indexed: 11/14/2022]
Abstract
OBJECTIVE We aimed to study the mechanism of hypoxia-inducible factor 1 alpha (HIF-1α) regulating the cell proliferation of tongue squamous cell carcinoma (TSCC) via vascular endothelial growth factor (VEGF). METHODS We used RNA interference (RNAi) technique, transfected chemically synthesized siRNA against HIF-1α into CAL-27 cells, and detected the expression of HIF-1α and VEGF by real time-PCR and Western blotting in order to find out if HIF-1α regulated the expression of VEGF. A xenograft experiment was carried out to observe the role of HIF-1α on the tumor growth of tongue squamous cell carcinoma. RESULTS HIF-1α and VEGF mRNA expression was significantly downregulated 36 and 48 h after transfection (P<0.05); the protein expression of HIF-1α and VEGF was also significantly suppressed by siRNA against HIF-1α. Furthermore, intratumoraly injection of HIF-1α targeting siRNA suppressed tumor growth in nude mice. CONCLUSIONS HIF-1α regulated VEGF expression, and they may contribute to TSCC cell tumor growth.
Collapse
Affiliation(s)
- Jun Liang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Zhaoqiang Zhang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Lizhong Liang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Yun Shen
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Kexiong Ouyang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| |
Collapse
|
32
|
Jiang H, Xie J, Xu G, Su Y, Li J, Zhu M, Wang M. Hypoxia regulates reactive oxygen species levels in SHG-44 glioma cells. Neural Regen Res 2014; 8:554-60. [PMID: 25206699 PMCID: PMC4146058 DOI: 10.3969/j.issn.1673-5374.2013.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/01/2012] [Indexed: 01/09/2023] Open
Abstract
In the present study, cultured human SHG-44 glioma cells were subjected to a hypoxic environment simulated using the CoCl2 method. Flow cytometry showed increased reactive oxygen species production in these cells. Real-time reverse transcription-PCR showed significantly increased hypoxia-inducible factor-1α mRNA expression in cells exposed to the hypoxic condition. The antioxidant N-acetylcysteine significantly inhibited reactive oxygen species production and reduced hypoxia-inducible factor-1α mRNA expression in normoxic and hypoxic groups, especially in the latter group. These findings indicate that hypoxia induces reactive oxygen species production and hypoxia-inducible factor-1α mRNA expression in human SHG-44 glioma cells, and that the antioxidant N-acetylcysteine can inhibit these changes.
Collapse
Affiliation(s)
- Haitao Jiang
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi Province, China
| | - Jiangtao Xie
- Xianyang Central Hospital, Xianyang 712000, Shaanxi Province, China
| | - Gaofeng Xu
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi Province, China
| | - Yongyong Su
- Xianyang Central Hospital, Xianyang 712000, Shaanxi Province, China
| | - Jinzheng Li
- Xianyang Central Hospital, Xianyang 712000, Shaanxi Province, China
| | - Mang Zhu
- Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Maode Wang
- First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710000, Shaanxi Province, China
| |
Collapse
|
33
|
Ginsenoside 20(S)-Rg3 targets HIF-1α to block hypoxia-induced epithelial-mesenchymal transition in ovarian cancer cells. PLoS One 2014; 9:e103887. [PMID: 25197976 PMCID: PMC4157750 DOI: 10.1371/journal.pone.0103887] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 07/04/2014] [Indexed: 01/21/2023] Open
Abstract
The prognosis of patients with ovarian cancer has remained poor mainly because of aggressive cancer progression. Since epithelial-mesenchymal transition (EMT) is an important mechanism mediating invasion and metastasis of cancer cells, targeting the EMT process with more efficacious and less toxic compounds to inhibit metastasis is of great therapeutic value for the treatment of ovarian cancer. We have found for the first time that the ginsenoside 20(S)-Rg3, a pharmacologically active component of the traditional Chinese herb Panax ginseng, potently blocks hypoxia-induced EMT of ovarian cancer cells in vitro and in vivo. Mechanistic studies confirm the mode of action of 20(S)-Rg3, which reduces the expression of hypoxia-inducible factor 1α (HIF-1α) by activating the ubiquitin-proteasome pathway to promote HIF-1α degradation. A decrease in HIF-1α in turn leads to up-regulation, via transcriptional suppression of Snail, of the epithelial cell-specific marker E-cadherin and down-regulation of the mesenchymal cell-specific marker vimentin under hypoxic conditions. Importantly, 20(S)-Rg3 effectively inhibits EMT in nude mouse xenograft models of ovarian cancer, promising a novel therapeutic agent for anticancer therapy.
Collapse
|
34
|
Radiolabeled probes targeting hypoxia-inducible factor-1-active tumor microenvironments. ScientificWorldJournal 2014; 2014:165461. [PMID: 25215311 PMCID: PMC4151590 DOI: 10.1155/2014/165461] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/06/2014] [Indexed: 11/17/2022] Open
Abstract
Because tumor cells grow rapidly and randomly, hypoxic regions arise from the lack of oxygen supply in solid tumors. Hypoxic regions in tumors are known to be resistant to chemotherapy and radiotherapy. Hypoxia-inducible factor-1 (HIF-1) expressed in hypoxic regions regulates the expression of genes related to tumor growth, angiogenesis, metastasis, and therapy resistance. Thus, imaging of HIF-1-active regions in tumors is of great interest. HIF-1 activity is regulated by the expression and degradation of its α subunit (HIF-1α), which is degraded in the proteasome under normoxic conditions, but escapes degradation under hypoxic conditions, allowing it to activate transcription of HIF-1-target genes. Therefore, to image HIF-1-active regions, HIF-1-dependent reporter systems and injectable probes that are degraded in a manner similar to HIF-1α have been recently developed and used in preclinical studies. However, no probe currently used in clinical practice directly assesses HIF-1 activity. Whether the accumulation of 18F-FDG or 18F-FMISO can be utilized as an index of HIF-1 activity has been investigated in clinical studies. In this review, the current status of HIF-1 imaging in preclinical and clinical studies is discussed.
Collapse
|
35
|
Song Z, Ren H, Gao S, Zhao X, Zhang H, Hao J. The clinical significance and regulation mechanism of hypoxia-inducible factor-1 and miR-191 expression in pancreatic cancer. Tumour Biol 2014; 35:11319-28. [PMID: 25119596 DOI: 10.1007/s13277-014-2452-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 08/05/2014] [Indexed: 12/17/2022] Open
Abstract
The aim of study was to discuss the correlation and regulatory mechanism of HIF-1 and miR-191 expression in pancreatic tumor. The association between the miR-191 and the clinicopathologic characteristics and the prognosis of pancreatic cancer was further explored. After hypoxic cultured for 6 and 12 h, qRT-PCR and Western blot were practiced to analyze the miR-191 and HIF-1 expression of MIA PaCa-2 and Aspac1 cells. We regulated the HIF-1 expression via plasmid and siRNA transfection to observe the alteration of HIF-1 and miR-191 expression. ChIP sequencing identified the binding sites of HIF-1 and miR-191. Dual luciferase assays were practiced to verify the binding sites. Immunohistochemical staining was practiced to analyze the expression of HIF-1, while qRT-PCR were done for investigating miR-191 in tumor tissues. Then, the association between the expression of them and the clinicopathologic characteristics and prognosis of pancreatic cancer were analyzed. After hypoxic cultured 12 h, the expression of HIF-1 protein, HIF-1mRNA and miR-191 of MIA PaCa-2 and AsPC-1 cells increased significantly (P < 0.05). After HIF-1 overexpressing plasmid transfected to the MIA PaCa-2 and AsPC-1 cells for 48 h, the expression of HIF-1 protein, HIF-1mRNA, and miR-191 upregulated significantly (P < 0.05). While after transfected the MIA PaCa-2 cells by HIF-1 siRNA, the significant decreasing of HIF-1 protein, HIF-1mRNA, and miR-191 expression were observed (P < 0.05). ChIP sequencing showed the protein synthesis of HIF-1 increased in hypoxia situation. Only the HRE5 (-1,169 bp, ChIP4) were significantly brighter in hypoxia in comparing with normoxic cells. In dual luciferase assays, after pGL3-miR-191 and HIF-1 overexpressing plasmid co-transfect the MIAPaCa-2 cells for 48 h, its relative expression of bioluminescence was higher than those co-transfected by mutant miR-191 vectors and HIF-1 overexpressing plasmid or by pGL3-miR-191 and HIF-1 empty plasmid. The expression of miR-191 closely associated with the tumor size, pTNM stage, lymph node metastasis, and perineural invasion (P < 0.05). Patients with higher expression of miR-191 were a risk factor for prognosis of pancreatic cancers. Expression of HIF-1 in pancreatic cancer cells increased under the condition of chronic hypoxia, which may bind to HRE2 in 5'flanking region of miR-191 and initiate transcription of miR-191. Expression of miR-191 was significantly higher in pancreatic tumor tissues. The expression of miR-191 closely associated with the tumor size, pTNM stage, lymph node metastasis and perineural invasion and poor prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Zhenguo Song
- Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,
| | | | | | | | | | | |
Collapse
|
36
|
Minocycline inhibits angiogenesis in vitro through the translational suppression of HIF-1α. Arch Biochem Biophys 2014; 545:74-82. [PMID: 24412777 DOI: 10.1016/j.abb.2013.12.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 12/09/2013] [Accepted: 12/30/2013] [Indexed: 01/04/2023]
Abstract
Minocycline was recently found to be effective against cancer. However, the precise molecular mechanisms of minocycline in cancer are poorly understood. Hypoxia-inducible factor-1 (HIF-1, a heterodimeric transcription factor composed of HIF-1α and β) activates the transcription of genes that are involved in angiogenesis in cancer. In this study, we found that minocycline significantly inhibits HIF-1α protein expression and suppresses HIF-1 transcriptional activity. The tube formation assay showed that minocycline has anti-angiogenic activity and suppresses hypoxia-induced vascular endothelial growth factor (VEGF) expression. The metabolic labeling assay showed that minocycline reduces HIF-1α protein translation and global protein synthesis. In addition, minocycline suppresses mTOR signaling and increases the phosphorylation of eIF2α, which is known to be related to the translational regulation of HIF-1α expression. These findings collectively indicate that minocycline is a potential inhibitor of HIF-1α and provide new insight into the discovery of drugs for cancer treatment.
Collapse
|
37
|
RETRACTED ARTICLE: The hypoxia-inducible factor-1 regulates the microRNA185 expression through binding to hypoxia response elements sequence 2. Med Oncol 2013; 30:756. [DOI: 10.1007/s12032-013-0756-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Accepted: 10/24/2013] [Indexed: 01/06/2023]
|
38
|
Pawlus MR, Hu CJ. Enhanceosomes as integrators of hypoxia inducible factor (HIF) and other transcription factors in the hypoxic transcriptional response. Cell Signal 2013; 25:1895-903. [PMID: 23707522 PMCID: PMC3700616 DOI: 10.1016/j.cellsig.2013.05.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/07/2013] [Indexed: 12/27/2022]
Abstract
Hypoxia is a prevalent attribute of the solid tumor microenvironment that promotes the expression of genes through posttranslational modifications and stabilization of alpha subunits (HIF1α and HIF2α) of hypoxia-inducible factors (HIFs). Despite significant similarities, HIF1 (HIF1α/ARNT) and HIF2 (HIF2α/ARNT) activate common as well as unique target genes and exhibit different functions in cancer biology. More surprisingly, accumulating data indicates that the HIF1- and/or HIF2-mediated hypoxia responses can be oncogenic as well as tumor suppressive. While the role of HIF in the hypoxia response is well established, recent data support the concept that HIF is necessary, but not sufficient for the hypoxic response. Other transcription factors that are activated by hypoxia are also required for the HIF-mediated hypoxia response. HIFs, other transcription factors, co-factors and RNA poll II recruited by HIF and other transcription factors form multifactorial enhanceosome complexes on the promoters of HIF target genes to activate hypoxia inducible genes. Importantly, HIF1 or HIF2 requires distinct partners in activating HIF1 or HIF2 target genes. Because HIF enhanceosome formation is required for the gene activation and distinct functions of HIF1 and HIF2 in tumor biology, disruption of the HIF1 or HIF2 specific enhanceosome complex may prove to be a beneficial strategy in tumor treatment in which tumor growth is specifically dependent upon HIF1 or HIF2 activity.
Collapse
Affiliation(s)
- Matthew R. Pawlus
- Molecular Biology Graduate Program University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cheng-Jun Hu
- Molecular Biology Graduate Program University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Department of Craniofacial Biology University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
39
|
Development of an Oxygen-Sensitive Degradable Peptide Probe for the Imaging of Hypoxia-Inducible Factor-1-Active Regions in Tumors. Mol Imaging Biol 2013; 15:713-21. [DOI: 10.1007/s11307-013-0647-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Parsons BL, Myers MB. KRAS mutant tumor subpopulations can subvert durable responses to personalized cancer treatments. Per Med 2013; 10:191-199. [PMID: 27867401 DOI: 10.2217/pme.13.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
KRAS mutations in colorectal and lung cancers predict failure to respond to therapies that target the EGFR. Significant percentages of patients with KRAS wild-type tumors also fail to respond to these therapies. Relapse occurs in patients with KRAS wild-type and mutant tumors, with moderately longer progression-free survival in patients with KRAS wild-type tumors. Colon and lung tumors frequently carry KRAS mutant tumor subpopulations not detected by DNA sequencing. This suggests detected and undetected KRAS mutant subpopulations in colon and lung tumors are undermining the efficacy of anti-EGFR therapies. Therefore, consideration should be given to combining therapies that target KRAS mutant cells with those that downregulate EGFR signaling. As tumors are frequently polyclonal in origin and comprised of distinct clonal populations carrying complementing genetic and/or epigenetic lesions, preclinical models that assess the efficacy of combination therapies in the context of heterogeneous tumor cell populations will be essential for progress in this area.
Collapse
Affiliation(s)
- Barbara L Parsons
- US FDA, National Center for Toxicological Research, Division of Genetic & Molecular Toxicology, HFT-120, 3900 NCTR Road, Jefferson, AR 72079, USA
| | - Meagan B Myers
- US FDA, National Center for Toxicological Research, Division of Genetic & Molecular Toxicology, HFT-120, 3900 NCTR Road, Jefferson, AR 72079, USA
| |
Collapse
|
41
|
Yeom CJ, Goto Y, Zhu Y, Hiraoka M, Harada H. Microenvironments and cellular characteristics in the micro tumor cords of malignant solid tumors. Int J Mol Sci 2012. [PMID: 23203043 PMCID: PMC3509559 DOI: 10.3390/ijms131113949] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Because of the accelerated proliferation of cancer cells and the limited distance that molecular oxygen can diffuse from functional tumor blood vessels, there appears to be a unique histology in malignant solid tumors, conglomerates of micro tumor cords. A functional blood vessel exists at the center of each tumor cord and is sequentially surrounded by well-oxygenated, oxygen-insufficient, and oxygen-depleted cancer cells in the shape of baumkuchen (layered). Cancer cells, by inducing the expression of various genes, adapt to the highly heterogeneous microenvironments in each layer. Accumulated evidence has suggested that not only tumor microenvironments but also cellular adaptive responses to them, influence the radioresistance of cancer cells. However, precisely how these factors affect one another and eventually influence the therapeutic effect of radiation therapy remains to be elucidated. Here, based on recent basic and clinical cancer research, we deduced extrinsic (oxygen concentration, glucose concentration, pH etc.) and intrinsic (transcriptional activity of hypoxia-inducible factor 1, metabolic pathways, cell cycle status, proliferative activity etc.) parameters in each layer of a tumor cord. In addition, we reviewed the latest information about the molecular mechanism linking these factors with both tumor radioresistance and tumor recurrence after radiation therapy.
Collapse
Affiliation(s)
- Chan Joo Yeom
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; E-Mails: (C.J.Y.); (Y.G.); (Y.Z.)
| | - Yoko Goto
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; E-Mails: (C.J.Y.); (Y.G.); (Y.Z.)
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; E-Mail:
| | - Yuxi Zhu
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; E-Mails: (C.J.Y.); (Y.G.); (Y.Z.)
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; E-Mail:
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, No.1 Friendship Road, Yuanjiagang, Yuzhong District, Chongqing 400016, China
| | - Masahiro Hiraoka
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; E-Mail:
| | - Hiroshi Harada
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; E-Mails: (C.J.Y.); (Y.G.); (Y.Z.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-75-753-9301; Fax: +81-75-753-9281
| |
Collapse
|
42
|
Omarova S, Charvet CD, Reem RE, Mast N, Zheng W, Huang S, Peachey NS, Pikuleva IA. Abnormal vascularization in mouse retina with dysregulated retinal cholesterol homeostasis. J Clin Invest 2012; 122:3012-23. [PMID: 22820291 DOI: 10.1172/jci63816] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 05/24/2012] [Indexed: 11/17/2022] Open
Abstract
Several lines of evidence suggest a link between age-related macular degeneration and retinal cholesterol maintenance. Cytochrome P450 27A1 (CYP27A1) is a ubiquitously expressed mitochondrial sterol 27-hydroxylase that plays an important role in the metabolism of cholesterol and cholesterol-related compounds. We conducted a comprehensive ophthalmic evaluation of mice lacking CYP27A1. We found that the loss of CYP27A1 led to dysregulation of retinal cholesterol homeostasis, including unexpected upregulation of retinal cholesterol biosynthesis. Cyp27a1-/- mice developed retinal lesions characterized by cholesterol deposition beneath the retinal pigment epithelium. Further, Cyp27a1-null mice showed pathological neovascularization, which likely arose from both the retina and the choroid, that led to the formation of retinal-choroidal anastomosis. Blood flow alterations and blood vessel leakage were noted in the areas of pathology. The Cyp27a1-/- retina was hypoxic and had activated Müller cells. We suggest a mechanism whereby abolished sterol 27-hydroxylase activity leads to vascular changes and identify Cyp27a1-/- mice as a model for one of the variants of type 3 retinal neovascularization occurring in some patients with age-related macular degeneration.
Collapse
Affiliation(s)
- Saida Omarova
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhu Y, Zhao T, Itasaka S, Zeng L, Yeom CJ, Hirota K, Suzuki K, Morinibu A, Shinomiya K, Ou G, Yoshimura M, Hiraoka M, Harada H. Involvement of decreased hypoxia-inducible factor 1 activity and resultant G1-S cell cycle transition in radioresistance of perinecrotic tumor cells. Oncogene 2012; 32:2058-68. [PMID: 22710721 PMCID: PMC3631307 DOI: 10.1038/onc.2012.223] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Cancer patients often suffer from local tumor recurrence after radiation therapy. Some intracellular and extracellular factors, such as activity of hypoxia-inducible factor 1 (HIF-1), cell cycle status and oxygen availability, have been suggested to affect DNA damage responses and eventual radioresistant characteristics of cancer cells. But when, where, and how these factors affect one another and induce cellular radioresistance is largely unknown. Here, we analyzed mechanistic and spatio-temporal relationships among them in highly heterogeneous tumor microenvironments. Experiments in vitro demonstrated that a decrease in the glucose concentration reduced the transcriptional activity of HIF-1 and expression of a downstream gene for the cell cycle regulator p27Kip1 even under hypoxic conditions. Then, the proportion of cells in the radioresistant S phase increased, whereas that in the radiosensitive G1 phase decreased, significantly. Immunohistochemical analyses showed that cancer cells in perinecrotic hypoxic regions, which should be under low-glucose conditions, expressed little HIF-1α, and therefore, were mainly in S phase and less damaged by radiation treatment. Continuous administration of glucagon, which increases the blood glucose concentration and so improves glucose availability in perinecrotic hypoxic regions, induced HIF-1α expression and increased radiation-induced DNA damage. Taken all together, these results indicate that cancer cells in perinecrotic regions, which would be under low-glucose and hypoxic conditions, obtain radioresistance by decreasing the level of both HIF-1 activity and p27Kip1 expression, and adjusting their cell cycle to the radioresistant S phase.
Collapse
Affiliation(s)
- Y Zhu
- Group of Radiation and Tumor Biology, Career-Path Promotion Unit for Young Life Scientists, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ueda M. Development of a Method for High-contrasted Nuclear Medical Imaging of Hypoxia-inducible Factor-1-active Tumor by Using a Pretargeting Approach. YAKUGAKU ZASSHI 2012; 132:595-600. [DOI: 10.1248/yakushi.132.595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masashi Ueda
- Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
45
|
Cancer cells that survive radiation therapy acquire HIF-1 activity and translocate towards tumour blood vessels. Nat Commun 2012; 3:783. [PMID: 22510688 PMCID: PMC3337987 DOI: 10.1038/ncomms1786] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Accepted: 03/14/2012] [Indexed: 01/16/2023] Open
Abstract
Tumour recurrence frequently occurs after radiotherapy, but the characteristics, intratumoural localization and post-irradiation behaviour of radioresistant cancer cells remain largely unknown. Here we develop a sophisticated strategy to track the post-irradiation fate of the cells, which exist in perinecrotic regions at the time of radiation. Although the perinecrotic tumour cells are originally hypoxia-inducible factor 1 (HIF-1)-negative, they acquire HIF-1 activity after surviving radiation, which triggers their translocation towards tumour blood vessels. HIF-1 inhibitors suppress the translocation and decrease the incidence of post-irradiation tumour recurrence. For the first time, our data unveil the HIF-1-dependent cellular dynamics during post-irradiation tumour recurrence and provide a rational basis for targeting HIF-1 after radiation therapy.
Collapse
|
46
|
Shigenaga A, Ogura K, Hirakawa H, Yamamoto J, Ebisuno K, Miyamoto L, Ishizawa K, Tsuchiya K, Otaka A. Development of a Reduction-Responsive Amino Acid that Induces Peptide Bond Cleavage in Hypoxic Cells. Chembiochem 2012; 13:968-71. [DOI: 10.1002/cbic.201200141] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Indexed: 02/04/2023]
|
47
|
Zeng W, Wan R, Zheng Y, Singh SR, Wei Y. Hypoxia, stem cells and bone tumor. Cancer Lett 2011; 313:129-36. [PMID: 21999934 PMCID: PMC3215823 DOI: 10.1016/j.canlet.2011.09.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 09/20/2011] [Accepted: 09/21/2011] [Indexed: 12/26/2022]
Abstract
Normal oxygen level is critical for niches that together with other components of the niche play vital role in regulating stem or tumor cells behavior. Hypoxia plays an important role in normal development and disease progression, including the growth of solid tumors. The hypoxia inducible factors (HIFs) are the key mediators of the cellular response to hypoxia. In this review, we focused on the role of HIFs on bone tumor formation. Further, we also emphasized how hypoxia, stem cells, and its niches regulate the bone tumorigenesis.
Collapse
Affiliation(s)
- Wen Zeng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Shanghai First People’s Hospital, Shanghai Jiaotong University, 200080, People’s Republic of China
| | - Rong Wan
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Yuehuan Zheng
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Shree Ram Singh
- Mouse Cancer Genetics Program, National Institutes of Health, National Cancer Institute at Frederick, Frederick, Maryland, 21702, USA
| | - Yiyong Wei
- Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
48
|
Vera-Ramirez L, Sanchez-Rovira P, Ramirez-Tortosa MC, Ramirez-Tortosa CL, Granados-Principal S, Lorente JA, Quiles JL. Free radicals in breast carcinogenesis, breast cancer progression and cancer stem cells. Biological bases to develop oxidative-based therapies. Crit Rev Oncol Hematol 2011; 80:347-68. [DOI: 10.1016/j.critrevonc.2011.01.004] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 12/15/2010] [Accepted: 01/11/2011] [Indexed: 12/30/2022] Open
|
49
|
Ueda M, Kudo T, Mutou Y, Umeda IO, Miyano A, Ogawa K, Ono M, Fujii H, Kizaka-Kondoh S, Hiraoka M, Saji H. Evaluation of [125I]IPOS as a molecular imaging probe for hypoxia-inducible factor-1-active regions in a tumor: comparison among single-photon emission computed tomography/X-ray computed tomography imaging, autoradiography, and immunohistochemistry. Cancer Sci 2011; 102:2090-6. [PMID: 21824221 DOI: 10.1111/j.1349-7006.2011.02057.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
To image hypoxia-inducible factor-1 (HIF-1)-active tumors, we previously developed a chimeric protein probe ([(123/125) I]IPOS) that is degraded in the same manner as HIF-1α under normoxic conditions. In the present study, we aim to show that the accumulation of radioiodinated POS reflects the expression of HIF-1. In vivo single-photon emission computed tomography (SPECT)/X-ray CT (CT) imaging, autoradiography, and double-fluorescent immunostaining for HIF-1α and pimonidazole (PIMO) were carried out 24 h after the injection of [(125) I]IPOS. Tumor metabolite analysis was also carried out. A tumor was clearly visualized by multi-pinhole, high-resolution SPECT/CT imaging with [(125) I]IPOS. The obtained images were in accordance with the corresponding autoradiograms and with the results of ex vivo biodistribution. A metabolite analysis revealed that 77% of the radioactivity was eluted in the macromolecular fraction, suggesting that the radioactivity mainly existed as [(125) I]IPOS in the tumors. Immunohistochemistry revealed that the HIF-1α-positive areas and PIMO-positive areas were not always identical, only some of the regions were positive for both markers. The areas showing [(125) I]IPOS accumulation were positively and significantly correlated with the HIF-1α-positive areas (R = 0.75, P < 0.0001). The correlation coefficient between [(125) I]IPOS-accumulated areas and HIF-1α-positive areas was significantly greater than that between the [(125) I]IPOS-accumulated areas and the PIMO-positive areas (P < 0.01). These findings indicate that [(125) I]IPOS accumulation reflects HIF-1 expression. Thus, [(123/125) I]IPOS can serve as a useful probe for the molecular imaging of HIF-1-active tumors.
Collapse
Affiliation(s)
- Masashi Ueda
- Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Hypoxia is a feature of most tumours, albeit with variable incidence and severity within a given patient population. It is a negative prognostic and predictive factor owing to its multiple contributions to chemoresistance, radioresistance, angiogenesis, vasculogenesis, invasiveness, metastasis, resistance to cell death, altered metabolism and genomic instability. Given its central role in tumour progression and resistance to therapy, tumour hypoxia might well be considered the best validated target that has yet to be exploited in oncology. However, despite an explosion of information on hypoxia, there are still major questions to be addressed if the long-standing goal of exploiting tumour hypoxia is to be realized. Here, we review the two main approaches, namely bioreductive prodrugs and inhibitors of molecular targets upon which hypoxic cell survival depends. We address the particular challenges and opportunities these overlapping strategies present, and discuss the central importance of emerging diagnostic tools for patient stratification in targeting hypoxia.
Collapse
Affiliation(s)
- William R Wilson
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand.
| | | |
Collapse
|