201
|
Wang SW, Liu SC, Sun HL, Huang TY, Chan CH, Yang CY, Yeh HI, Huang YL, Chou WY, Lin YM, Tang CH. CCL5/CCR5 axis induces vascular endothelial growth factor-mediated tumor angiogenesis in human osteosarcoma microenvironment. Carcinogenesis 2014; 36:104-14. [PMID: 25330803 DOI: 10.1093/carcin/bgu218] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chemokines modulate angiogenesis and metastasis that dictate cancer development in tumor microenvironment. Osteosarcoma is the most frequent bone tumor and is characterized by a high metastatic potential. Chemokine CCL5 (previously called RANTES) has been reported to facilitate tumor progression and metastasis. However, the crosstalk between chemokine CCL5 and vascular endothelial growth factor (VEGF) as well as tumor angiogenesis in human osteosarcoma microenvironment has not been well explored. In this study, we found that CCL5 increased VEGF expression and production in human osteosarcoma cells. The conditioned medium (CM) from CCL5-treated osteosarcoma cells significantly induced tube formation and migration of human endothelial progenitor cells. Pretreatment of cells with CCR5 antibody or transfection with CCR5 specific siRNA blocked CCL5-induced VEGF expression and angiogenesis. CCL5/CCR5 axis demonstrably activated protein kinase Cδ (PKCδ), c-Src and hypoxia-inducible factor-1 alpha (HIF-1α) signaling cascades to induce VEGF-dependent angiogenesis. Furthermore, knockdown of CCL5 suppressed VEGF expression and attenuated osteosarcoma CM-induced angiogenesis in vitro and in vivo. CCL5 knockdown dramatically abolished tumor growth and angiogenesis in the osteosarcoma xenograft animal model. Importantly, we demonstrated that the expression of CCL5 and VEGF were correlated with tumor stage according the immunohistochemistry analysis of human osteosarcoma tissues. Taken together, our findings provide evidence that CCL5/CCR5 axis promotes VEGF-dependent tumor angiogenesis in human osteosarcoma microenvironment through PKCδ/c-Src/HIF-1α signaling pathway. CCL5 may represent a potential therapeutic target against human osteosarcoma.
Collapse
Affiliation(s)
- Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Shih-Chia Liu
- Department of Orthopaedics, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Hui-Lung Sun
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio state University, Columbus, OH 43210, USA
| | - Te-Yang Huang
- Department of Orthopaedics, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Chia-Han Chan
- Department of Orthopaedics, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Chen-Yu Yang
- Department of Orthopaedics, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Hung-I Yeh
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung 413, Taiwan
| | - Wen-Yi Chou
- Department of Orthopedic Surgery, Kaohsiung Chang Gung Memorial Hospital Medical Center, Kaohsiung 833, Taiwan
| | - Yu-Min Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan, Department of Orthopedic Surgery, Taichung Veterans General Hospital, Taichung 407, Taiwan,
| | - Chih-Hsin Tang
- Department of Biotechnology, College of Health Science, Asia University, Taichung 413, Taiwan, Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan and Department of Pharmacology, School of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
202
|
Chen F, Zhuang M, Zhong C, Peng J, Wang X, Li J, Chen Z, Huang Y. Baicalein reverses hypoxia-induced 5-FU resistance in gastric cancer AGS cells through suppression of glycolysis and the PTEN/Akt/HIF-1α signaling pathway. Oncol Rep 2014; 33:457-63. [PMID: 25333894 DOI: 10.3892/or.2014.3550] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/25/2014] [Indexed: 01/25/2023] Open
Abstract
Cancer cells can survive under hypoxia by metabolic reprogramming to achieve a high level of glycolysis, which contributes to the development of chemoresistance. Therefore, inhibition of glycolysis would be a novel strategy for overcoming hypoxia‑induced drug resistance. Baicalein, a flavonoid derived from the root of Scutellaria baicalensis, has been reported to exert strong antitumor activity toward various types of cancer. In the present study, we evaluated the effect of baicalein on hypoxia-induced 5-fluorouracil (5-FU) resistance in gastric cancer AGS cells and investigated the possible molecular mechanisms. We found that baicalein increased the sensitivity of AGS cells to 5-FU treatment under hypoxia. In addition, the hypoxia-enhanced glycolytic flux and expression of several critical glycolysis-associated enzymes (HK2, LDH-A and PDK1) in the AGS cells were suppressed by baicalein. Furthermore, baicalein inhibited hypoxia-induced Akt phosphorylation by promoting PTEN accumulation, thereby attenuating hypoxia-inducible factor-1α (HIF-1α) expression in AGS cells. These results together suggest that inhibition of glycolysis via regulation of the PTEN/Akt/HIF-1α signaling pathway may be one of the mechanisms whereby baicalein reverses 5-FU resistance in cancer cells under hypoxia.
Collapse
Affiliation(s)
- Fenglin Chen
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Mingkai Zhuang
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Canmei Zhong
- College of Union Clinical Medicine, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine Biomedical Research Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350001, P.R. China
| | - Xiaozhong Wang
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Jianying Li
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhixin Chen
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yuehong Huang
- Department of Gastroenterology, Union Hospital of Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
203
|
Age-dependent metabolic dysregulation in cancer and Alzheimer's disease. Biogerontology 2014; 15:559-77. [PMID: 25305052 DOI: 10.1007/s10522-014-9534-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/27/2014] [Indexed: 01/12/2023]
Abstract
Age is the main risk factor for cancer and neurodegeneration; two radically divergent diseases. Yet selective pressure to meet cellular metabolic needs may provide a common mechanism linking these two disorders. The exclusive use of glycolysis, despite the presence of oxygen, is commonly referred to as aerobic glycolysis and is the primary metabolic pathway of cancer cells. Recent evidence suggests that aerobic glycolysis is also a key regulator of synaptic plasticity in the brain that may positively influence cognition. Elevated aerobic glycolysis is a contributing factor to the development of cancer as increased glycolytic flux plays an important role in the biosynthesis of macromolecules and promotes proliferation. In contrast, decreased aerobic glycolysis in the brain occurs with age and could lead to a loss of cell survival mechanisms that counter pathogenic processes underlying neurodegeneration. In this review we discuss the recent findings from epidemiological studies demonstrating an inverse comorbidity of cancer and Alzheimer's disease. We summarize evidence linking the two diseases through changes in metabolism over the course of normal aging. We discuss the key steps and regulatory mechanisms of aerobic glycolysis and mitochondrial oxidative phosphorylation which could be exploited for the development of novel therapies. In addition, we outline the regulation of aerobic glycolysis at the transcriptional level by HIF-1α and Pin1 and their roles in cancer and neurodegeneration. Finally, we provide a possible explanation for metabolic dysregulation that occurs with age, and how it may be a contributing factor to age-related diseases. Determining how metabolism becomes dysregulated over time could lead to the development of effective interventions for ensuring metabolic homeostasis and healthy aging.
Collapse
|
204
|
Zhang H, Sun L, Xiao X, Xie R, Liu C, Wang Y, Wei Y, Zhang H, Liu L. Krüppel-like factor 8 contributes to hypoxia-induced MDR in gastric cancer cells. Cancer Sci 2014; 105:1109-15. [PMID: 25040744 PMCID: PMC4462403 DOI: 10.1111/cas.12483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/31/2022] Open
Abstract
We previously reported that hypoxia-induced MDR in gastric cancer (GC) cells is hypoxia-inducible factor-1 (HIF-1)-dependent. However, the exact mechanisms are still unknown. Our previous study revealed that Krüppel-like factor 8 (KLF8), a novel transcription factor, was associated with malignant phenotype in GC cells. KLF8 is overexpressed in clear cell renal carcinoma lacking von Hippel-Lindau protein function, which resulted in HIF-1 stabilization. Given this association, we hypothesized that KLF8 contributed to hypoxia-induced MDR in GC cells. Initial experiments revealed that hypoxia could increase KLF8 and HIF-1α expressions in GC cells, and KLF8 levels in GC drug-resistant cell lines were higher than in parental cell lines. Subsequent experiments showed that in normoxia, exogenous KLF8 could promote the MDR phenotype; however, blocking KLF8 expression could effectively reverse the MDR phenotype induced by hypoxia. Overexpressed KLF8 increased resistance-associated gene MDR1 mRNA levels, Bcl-2 and P-gp protein levels, and decreased Bax and caspase-3 protein levels in GC cells, and knockout KLF8 reversed these effects. Dual luciferase reporter and ChIP assays showed that KLF8 could promote MDR1 transcriptional activity by combining with KLF8 binding sites located in the upstream of MDR1 transcriptional start site. These results suggest that KLF8 is involved in hypoxia-induced MDR through inhibiting apoptosis and increasing the drug release rate by directly regulating MDR1 transcription. This study aims to discuss whether KLF8 involves in hypoxia-induced multi-drug resistance and its mechanism. Our results showed that hypoxia could increase KLF8 expression in gastric cancer cells. Meanwhile, we found that KLF8 contributed to hypoxia-induced multi-drug resistance via regulating MDR1 directly. Through this research, we found a new target gene of KLF8 and further clarified the mechanism of multi-drug resistance happened in gastric cancer.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China; Department of Tuberculosis Control and Prevention, Xi'an Center for Disease Control and Prevention, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Fridman WH, Remark R, Goc J, Giraldo NA, Becht E, Hammond SA, Damotte D, Dieu-Nosjean MC, Sautès-Fridman C. The immune microenvironment: a major player in human cancers. Int Arch Allergy Immunol 2014; 164:13-26. [PMID: 24852691 DOI: 10.1159/000362332] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cancer is a major public health issue and figures among the leading causes of death in the world. Cancer development is a long process, involving the mutation, amplification or deletion of genes and chromosomal rearrangements. The transformed cells change morphologically, enlarge, become invasive and finally detach from the primary tumor to metastasize in other organs through the blood and/or lymph. During this process, the tumor cells interact with their microenvironment, which is complex and composed of stromal and immune cells that penetrate the tumor site via blood vessels and lymphoid capillaries. All subsets of immune cells can be found in tumors, but their respective density, functionality and organization vary from one type of tumor to another. Whereas inflammatory cells play a protumoral role, there is a large body of evidence of effector memory T cells controlling tumor invasion and metastasis. Thus, high densities of memory Th1/CD8 cytotoxic T cells in the primary tumors correlate with good prognosis in most tumor types. Tertiary lymphoid structures, which contain mature dendritic cells (DC) in a T cell zone, proliferating B cells and follicular DC, are found in the tumor stroma and they correlate with intratumoral Th1/CD8 T cell and B cell infiltration. Eventually, tumors undergo genetic and epigenetic modifications that allow them to escape being controlled by the immune system. This comprehensive review describes the immune contexture of human primary and metastatic tumors, how it impacts on patient outcomes and how it could be used as a predictive biomarker and guide immunotherapies.
Collapse
Affiliation(s)
- W H Fridman
- Cancer, Immune Control and Escape, UMRS1138, Cordeliers Research Center, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
206
|
Choi SH, Chung AR, Kang W, Park JY, Lee MS, Hwang SW, Kim DY, Kim SU, Ahn SH, Kim S, Han KH. Silencing of hypoxia-inducible factor-1β induces anti-tumor effects in hepatoma cell lines under tumor hypoxia. PLoS One 2014; 9:e103304. [PMID: 25068796 PMCID: PMC4113399 DOI: 10.1371/journal.pone.0103304] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 06/27/2014] [Indexed: 12/18/2022] Open
Abstract
Dimerization of hypoxia-inducible factor-1 beta (HIF-1β) [aryl hydrocarbon receptor nuclear translocator (ARNT)] with HIF-1α is involved in various aspects of cancer biology, including proliferation and survival under hypoxic conditions. We investigated the in vitro mechanism by which silencing of HIF-1β leads to the suppression of tumor cell growth and cellular functions. Various hepatocellular carcinoma (HCC) cell lines (Huh-7, Hep3B, and HepG2) were transfected with small interfering RNA (siRNA) against HIF-1β (siHIF-1β) and cultured under hypoxic conditions (1% O2 for 24 h). The expression levels of HIF-1β, HIF-1α, and growth factors were examined by immunoblotting. Tumor growth was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and tumor activity was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling, tumor cell invasion, and migration assays. Under hypoxic conditions, silencing of HIF-1β expression suppressed tumor cell growth and regulated the expression of tumor growth-related factors, such as vascular endothelial growth factor, epidermal growth factor, and hepatocyte growth factor. Suppression of tumor cell invasion and migration was also demonstrated in HIF-1β-silenced HCC cell lines. Silencing of HIF-1β expression may induce anti-tumor effects under hypoxic conditions in HCC cell lines.
Collapse
Affiliation(s)
- Sung Hoon Choi
- Brain Korea 21 plus project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Ae Ri Chung
- Brain Korea 21 plus project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Wonseok Kang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Mi Sol Lee
- Department of Premed, Yonsei University College of Medicine, Seoul, Korea
| | - Shin Won Hwang
- Department of Premed, Yonsei University College of Medicine, Seoul, Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Seung Up Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Sang Hoon Ahn
- Brain Korea 21 plus project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Seungtaek Kim
- Brain Korea 21 plus project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
| | - Kwang-Hyub Han
- Brain Korea 21 plus project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Yonsei Liver Cancer Special Clinic, Yonsei University Health System, Seoul, Korea
- Liver Cirrhosis Clinical Research Center, Yonsei University Health System, Seoul, Korea
- * E-mail:
| |
Collapse
|
207
|
Akimoto J, Nakayama M, Okano T. Temperature-responsive polymeric micelles for optimizing drug targeting to solid tumors. J Control Release 2014; 193:2-8. [PMID: 25037017 DOI: 10.1016/j.jconrel.2014.06.062] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/09/2014] [Accepted: 06/24/2014] [Indexed: 10/25/2022]
Abstract
Targeting to solid tumors is the most challenging issue in the drug delivery field. To obtain the ideal pharmacodynamics of administrated drugs, drug carriers must suppress drug release and interactions with non-target tissues while circulating in the bloodstream, yet actively release the incorporated drug and interact with target cells after delivery to the tumor tissue. To handle this situation, stimuli-responsive drug carriers are extremely useful, because carriers change their physicochemical properties to control the drug release rate and interaction with cells in response to the surrounding environmental conditions or applied physical signals. The current review focuses on the strategy and availability of temperature-responsive (TR) polymeric micelles as a next-generation drug carrier. In particular, we discuss the unique properties of TR polymeric micelles, such as temperature-triggered drug release and intracellular uptake system. In addition, we explore the methodology for integrating other targeting systems into TR micelles to pursue the ideal pharmacodynamics in conjunction with thermal therapy as a future prospective of the TR system.
Collapse
Affiliation(s)
- Jun Akimoto
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Masamichi Nakayama
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns), 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan.
| |
Collapse
|
208
|
Shuch B, Agochukwu N, Ricketts CJ, Vocke CD, Gautam R, Merino M, Linehan WM, Srinivasan R. Vascular Endothelial Growth Factor Receptor-Targeted Therapy in Succinate Dehydrogenase C Kidney Cancer. J Clin Oncol 2014; 34:e76-9. [PMID: 25024072 DOI: 10.1200/jco.2013.51.0214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
209
|
Peitzsch C, Perrin R, Hill RP, Dubrovska A, Kurth I. Hypoxia as a biomarker for radioresistant cancer stem cells. Int J Radiat Biol 2014; 90:636-52. [DOI: 10.3109/09553002.2014.916841] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
210
|
Vorrink SU, Sarsour EH, Olivier AK, Robertson LW, Goswami PC, Domann FE. PCB 126 perturbs hypoxia-induced HIF-1α activity and glucose consumption in human HepG2 cells. ACTA ACUST UNITED AC 2014; 66:377-82. [PMID: 24916446 DOI: 10.1016/j.etp.2014.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 05/08/2014] [Indexed: 12/23/2022]
Abstract
Aerobic organisms strongly depend on the availability of oxygen for respiration and countless other metabolic processes to maintain cellular homeostasis. Under certain conditions, the amount of available oxygen can be limited. To support survival in environments with limited oxygen supply, hypoxia-inducible factors (HIFs) reprogram vital components of cellular metabolism. HIF-1α is an important mediator of acute and adaptive responses to hypoxic stress. Interestingly, the heterodimeric partner required by HIF-1α to function as transcription factor, known as ARNT, is also an essential part of the aryl hydrocarbon receptor (AhR) transcription factor complex. Thus, via ARNT a crosstalk exists between these two pathways that might affect HIF-1α-mediated processes. In this study we sought to assess the effect of the AhR agonist PCB 126 on HIF-1α activity as well as on HIF-1α-regulated targets involved in cellular metabolism in human HepG2 cells. Our results show that PCB 126 reduced HIF-1α localization to the nucleus. Furthermore, in an in vivo setting, rats exposed to parenteral PCB 126 also displayed reduced hepatocyte nuclear localization of HIF-1α. Additionally, HepG2 cells exposed to PCB 126 displayed reduced hypoxia-regulated HRE-luciferase reporter gene expression as well as a reduction in glucose consumption in conditions of hypoxia. In summary, this study reveals that HIF-1α-regulated cellular metabolic processes are negatively affected by PCB 126 which might ultimately affect adaptive responses and cell survival in hypoxic environments.
Collapse
Affiliation(s)
- Sabine U Vorrink
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA
| | - Ehab H Sarsour
- Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA
| | - Alicia K Olivier
- Department of Pathology, The University of Iowa, Iowa City, IA, USA
| | - Larry W Robertson
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Occupational and Environmental Health, The University of Iowa, Iowa City, IA, USA
| | - Prabhat C Goswami
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA
| | - Frederick E Domann
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA; Department of Pathology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
211
|
Xu W, Kwon JH, Moon YH, Kim YB, Yu YS, Lee N, Choi KY, Kim YS, Park YK, Kim BW, Wang HJ. Influence of preoperative transcatheter arterial chemoembolization on gene expression in the HIF-1α pathway in patients with hepatocellular carcinoma. J Cancer Res Clin Oncol 2014; 140:1507-15. [PMID: 24853275 DOI: 10.1007/s00432-014-1713-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 05/13/2014] [Indexed: 01/20/2023]
Abstract
PURPOSE Although transcatheter arterial chemoembolization (TACE) is the most common treatment option in patients with hepatocellular carcinoma (HCC), its clinical benefits remain still controversial. Since TACE induces hypoxic necrosis in tumors, hypoxia-inducible factor 1α (HIF-1α) could critically affect biology in residual tumors after TACE treatment and subsequent prognosis. However, HIF-1α and its prognostic relevance in TACE have rarely been examined in human specimens. In the current study, we investigated the prognosis and expression of genes regulated by HIF-1α in HCC patients receiving preoperative TACE for the first time. METHODS In total, 35 patients with HCC (10 patients undergoing preoperative TACE) were retrospectively studied. The prognostic significance of TACE was analyzed using Kaplan-Meier and Cox regression models. Protein levels of HIF-1α and mRNA levels of HIF-1α-associated genes were examined using immunohistochemistry (IHC) and real-time RT-PCR, respectively. RESULTS Preoperative TACE was significantly associated with increased 2-year recurrence rate (80 vs. 36 %, P = 0.00402) and shorter disease-free survival (DFS) time (11.9 vs. 35.7 months, P = 0.0182). TACE was an independent prognostic factor for recurrence (P = 0.007) and poor DFS (P = 0.010) in a multivariate analysis. Immunohistochemical staining revealed in vivo activation of HIF-1α in human specimens treated with TACE. Notably, protein levels of HIF-1α were significantly increased in TACE tissues demonstrated by IHC. Transcriptional targets of HIF-1α showed mRNA expression patterns consistent with activation of HIF-1α in TACE tissues. CONCLUSIONS Our findings collectively demonstrate that preoperative TACE confers poor prognosis in HCC patients through activation of HIF-1α.
Collapse
Affiliation(s)
- Weiguang Xu
- Department of Surgery, Ajou University School of Medicine, Suwon, 443-721, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Vorrink SU, Domann FE. Regulatory crosstalk and interference between the xenobiotic and hypoxia sensing pathways at the AhR-ARNT-HIF1α signaling node. Chem Biol Interact 2014; 218:82-8. [PMID: 24824450 DOI: 10.1016/j.cbi.2014.05.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/29/2014] [Accepted: 05/04/2014] [Indexed: 11/17/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates many of the responses to toxic environmental chemicals such as TCDD or dioxin-like PCBs. To regulate gene expression, the AhR requires its binding partner, the aryl hydrocarbon receptor nuclear translocator (ARNT). ARNT is also required by the hypoxia-inducible factor-1α (HIF-1α), a crucial regulator of responses to conditions of reduced oxygen. The important role of ARNT in both the AhR and HIF-1α signaling pathways establishes a meaningful foundation for a possible crosstalk between these two vitally important signaling pathways. This crosstalk might lead to interference between the two signaling pathways and thus might play a role in the variety of cellular responses after exposure to AhR ligands and reduced oxygen availability. This review focuses on studies that have analyzed the effect of low oxygen environments and hypoxia-mimetic agents on AhR signaling and conversely, the effect of AhR ligands, with a special emphasis on PCBs, on HIF-1α signaling. We highlight studies that assess the role of ARNT, elucidate the mechanism of the crosstalk, and discuss the physiological implications for exposure to AhR-inducing compounds in the context of hypoxia.
Collapse
Affiliation(s)
- Sabine U Vorrink
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA
| | - Frederick E Domann
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, USA; Department of Radiation Oncology, The University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
213
|
Collier C, Petrovic S. Carbonic Anhydrase IX as a Target for Renal Cell Carcinoma Therapy. ACTA ACUST UNITED AC 2014. [DOI: 10.4081/nr.2011.e5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Renal cell carcinoma (RCC) is the third leading cause of death in genitourinary cancers, with a 126% increase in incidence over the past 60 years. The most common subtype of RCC, clear cell carcinoma, occurs in 7080% of RCC cases, most often caused by mutations in the Von-Hippel Lindau tumor suppressor gene. The VHL tumor suppressor protein acts to down-regulate Hypoxia Inducible Factor-1, a heterodimeric transcription factor which transactivates genes containing the Hypoxia Response Element (HRE) DNA sequences. Inactivation of this tumor suppressor leads to the stabilization of the HIF-1α subunit, driving the constituent expression of HRE-containing genes including several growth factors, and carbonic anhydrase IX. This carbonic anhydrase isoform is normally found in the gastrointestinal tract and absent in the kidney, however; it is over-expressed in clear cell RCC. Carbonic anhydrase is a ubiquitous enzyme, which maintains the acid-base balance of the cell, facilitates secretion of acid into the stomach and excretion of acid by the kidney, bone resorption, and production of cerebrospinal fluid. However, in clear cell RCC, the upregulation of CAIX has been shown to facilitate tumor progression and tumor cell invasiveness by decreasing the pH of the extracellular environment, leading to the degradation of the extracellular matrix. The specific expression pattern of CAIX makes it a useful biomarker in the diagnosis of clear cell RCC, and a strong candidate as a chemotherapeutic target. Current progress with this approach is discussed in this review.
Collapse
Affiliation(s)
- Cameron Collier
- Department of Cellular and Molecular Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Snezana Petrovic
- Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio
- Research Services, Veterans' Affairs Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
214
|
Yang J, Zhao JX, Wang Y, Chen G, Cheng WN, Luo X, Pei XT, Zhao L, Su Q, Zhou H. Effects of sodium pyruvate on ameliorating metabolic acidosis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 44:48-55. [PMID: 24697727 DOI: 10.3109/21691401.2014.901335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To examine the effects of sodium pyruvate (SP) on metabolic acidosis. METHODS For the in vivo experiments, we evaluated effects of SP on an ammonium chloride (NH4Cl)-induced hyperchloremic acidosis rat model. SP was infused at overall doses of 2, 4, and 6 mmol·kg(- 1) for the SP1, SP2, and SP3 groups, respectively. Treatment with sodium bicarbonate (SB) was used as a positive control (2 mmol·kg(- 1)), and treatment with normal saline (NS) was used as a volume control (2 mL·kg(- 1)). Blood was sampled from the ophthalmic venous plexus for pH, blood gases, electrolytes, glucose, creatinine (Cr), and urea analysis after injection. For the in vitro experiment, propionate was applied to induce intracellular acidosis in human endothelial cells. Intracellular pH (pHi) was fluorimetrically measured after the addition of SP. RESULTS In the in vivo study, the pH of SP1 group showed no significant difference compared with that of the NS group. The SP2 and SP3 groups had a higher pH than the NS group (P < 0.01). The SP3 group had a higher pH than the SB group (P < 0.05) and SP1 group (P < 0.05). Moreover, SP treatment ameliorated the abnormality of calcium and decreased the blood potassium levels. The SP3 group had higher glucose levels than SP1 group (P < 0.05). No significant differences were observed between all the groups in the plasma Cr and urea levels. In the in vitro study, the pHi increased immediately after the addition of SP. CONCLUSION The data suggest that intravascular treatment with SP represents a novel therapeutic strategy to ameliorate metabolic acidosis.
Collapse
Affiliation(s)
- Jing Yang
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Jing-Xiang Zhao
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Ying Wang
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Gan Chen
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Wei-Na Cheng
- b Department of Biological Engineering , College of Environment and Chemical Engineering, Yanshan University , Qinhuangdao , P. R. China
| | - Xin Luo
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Xue-Tao Pei
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Lian Zhao
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| | - Qin Su
- c The First Affiliated Hospital of General Hospital of PLA , Beijing , P. R. China
| | - Hong Zhou
- a Institute of Transfusion Medicine, Academy of Military Medical Sciences , Beijing , P.R. China
| |
Collapse
|
215
|
Park SH, Kim BR, Lee JH, Park ST, Lee SH, Dong SM, Rho SB. GABARBP down-regulates HIF-1α expression through the VEGFR-2 and PI3K/mTOR/4E-BP1 pathways. Cell Signal 2014; 26:1506-13. [PMID: 24686084 DOI: 10.1016/j.cellsig.2014.03.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/16/2014] [Indexed: 10/25/2022]
Abstract
Human γ-aminobutyrate type A (GABAA) receptor-binding protein (GABARBP), a tumor suppressor protein with apoptotic function, can be inhibited in response to angiogenesis through the PI3K/Akt signaling cascades. Here, we investigated whether GABARBP over-expression could regulate vascular endothelial growth factor (VEGF)/hypoxia-inducible factor-1α (HIF-1α) expression and angiogenic activity in a carcinoma model system. GABARBP dramatically inhibited VEGF-induced endothelial cell proliferation, migration, and tube formation, as well as VEGFR-2 phosphorylation in vitro. At the same time, GABARBP exposed potent anti-angiogenic activity and remarkably down-regulated the levels of VEGF and HIF-1α protein expression, key components for angiogenesis. In addressing its biological molecular mechanism, GABARBP was found to effectively inhibit the phosphorylation of down-stream PI3K components, such as PDK1, Akt, mTOR, TSC-2, p70S6K, and 4E-BP1 by directly binding with VEGFR-2. In contrast, p38/JNK phosphorylation was not suppressed by GABARBP. These findings disclose a novel function of GABARBP in suppressing VEGF and HIF-1α protein expression, which is important for tumor angiogenesis and tumor growth. Thus, our data strongly provides novel biological mechanistic insights into the regulatory function of GABARBP in ovarian tumor progression, and the important of pre-clinical certification of GABARBP as a potential angiogenesis agent targeting ovarian tumorigenesis.
Collapse
Affiliation(s)
- Sung Ho Park
- Department of Obstetrics and Gynecology, Hallym University, 948-1, Daerim 1-dong, Yeongdeungpo-gu, Seoul 150-950, Republic of Korea
| | - Boh-Ram Kim
- Research Institute, National Cancer Center, 323, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do 410-769, Republic of Korea
| | - Jeong Heon Lee
- Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju 561-712, Republic of Korea
| | - Sung Taek Park
- Department of Obstetrics and Gynecology, Hallym University, 948-1, Daerim 1-dong, Yeongdeungpo-gu, Seoul 150-950, Republic of Korea
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, 470, Samga-dong, Cheoin-gu, Yongin-si Gyeonggi-do 449-714, Republic of Korea
| | - Seung Myung Dong
- Research Institute, National Cancer Center, 323, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do 410-769, Republic of Korea
| | - Seung Bae Rho
- Research Institute, National Cancer Center, 323, Ilsan-ro, Ilsandong-gu, Goyang-si Gyeonggi-do 410-769, Republic of Korea.
| |
Collapse
|
216
|
Gulonolactone Addition to Human Hepatocellular Carcinoma Cells with Gene Transfer of Gulonolactone Oxidase Restores Ascorbate Biosynthesis and Reduces Hypoxia Inducible Factor 1. Biomedicines 2014; 2:98-109. [PMID: 28548062 PMCID: PMC5423483 DOI: 10.3390/biomedicines2010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/26/2014] [Accepted: 02/26/2014] [Indexed: 11/17/2022] Open
Abstract
Humans are unable to synthesise ascorbate (Vitamin C) due to the lack of a functional gulonolactone oxidase (Gulo), the enzyme that catalyses the final step in the biosynthesis pathway. Ascorbate is a vital micronutrient required for many biological functions, including as a cofactor for metalloenzymes that regulate the transcription factor hypoxia-inducible factor-1 (HIF-1), which governs cell survival under hypoxia. In most animals, ascorbate is made in liver cells. This study aimed to restore ascorbate synthesis to human hepatocellular carcinoma HepG2 cells and determine the effect of internally produced ascorbate on HIF-1 activation. HepG2 cells were gene-modified with a plasmid encoding the mouse Gulo cDNA, tested for genomic incorporation by PCR and ascorbate synthesis by high performance liquid chromatography. Levels of HIF-1 protein were measured using Western blotting. Gulo-modified HepG2 cells showed increased adherence compared to control HepG2 cells. A PCR-positive clone synthesised ascorbate when the Gulo substrate, l-gulono-1,4-lactone, was supplied. Intracellular ascorbate concentrations reached 5% of saturation levels (6 nmol/106 cells). Addition of ascorbate or gulonolactone reduced HIF-1 accumulation in the Gulo clone, but also in parental HepG2 cells. Our data confirm the requirement for a number of factors in addition to Gulo in the ascorbate biosynthesis pathway in human cells.
Collapse
|
217
|
Wittström E, Nordling M, Andréasson S. Genotype-phenotype correlations, and retinal function and structure in von Hippel-Lindau disease. Ophthalmic Genet 2014; 35:91-106. [PMID: 24555745 DOI: 10.3109/13816810.2014.886265] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
PURPOSE To investigate genotype-phenotype correlation and to analyze functional and structural changes in the retina of patients with von Hippel-Lindau (VHL) disease. METHODS Thirteen patients from four families (A, B, C and D) with known VHL disease and known mutations in the VHL gene were examined. All patients underwent clinical examination and optical coherence tomography (OCT). Full-field electroretinography (full-field ERG) was performed in twelve patients. RESULTS Family A, with deletion of exon 3 in the VHL gene, and family B, with the missense mutation p.R79P, exhibited type 1 VHL characterized by the absence of pheochromocytoma and a high incidence of central nervous system hemangioblastomas. One member of family B exhibited Goldenhar syndrome. A novel missense mutation (p.L198P) was identified in the VHL gene in the patient from family C. This p.L198P mutation caused a phenotype with early onset of a neuroendocrine tumor of the pancreas, bilateral pheochromocytomas, and optic nerve hemangioblastoma. Full-field ERG showed significantly prolonged implicit times of the b-wave and maximal combined a-wave in VHL patients, compared to controls. Examination of the retinal structure in all patients with VHL, using OCT, showed a significant decrease in retinal thickness in VHL patients without ocular hemangioblastomas, compared to controls. CONCLUSIONS Our findings support previously established genotype-phenotype correlations. However, we here describe an unusual phenotype with a novel missense mutation, p.L198P, and report the finding that VHL disease can be associated with Goldenhar syndrome. Electrophysiological and structural findings suggest that VHL disease is a progressive, neurodegenerative disease of the retina.
Collapse
|
218
|
Faloppi L, Scartozzi M, Bianconi M, Svegliati Baroni G, Toniutto P, Giampieri R, Del Prete M, De Minicis S, Bitetto D, Loretelli C, D'Anzeo M, Benedetti A, Cascinu S. The role of LDH serum levels in predicting global outcome in HCC patients treated with sorafenib: implications for clinical management. BMC Cancer 2014; 14:110. [PMID: 24552144 PMCID: PMC3930857 DOI: 10.1186/1471-2407-14-110] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022] Open
Abstract
Background In many tumour types serumlactate dehydrogenase (LDH) levels proved to represent an indirect marker of tumour hypoxia, neo-angiogenesis and worse prognosis. As we previously reported LDH is an important predictive factor in hepatocellular carcinoma (HCC) patients undergoing transarterial chemoembolization (TACE). Sorafenib represents the therapeutic stronghold in advanced HCC patients. As a tyrosine kinase inhibitor (TKI) mainly directed against the angiogenetic pathway, the correlation of sorafenib administration with markers of hypoxia could be an important tool in patients management. Aim of our analysis was to evaluate the role of LDH pre-treatment levels and its variation during treatment in HCC patients receiving sorafenib. Methods 78 patients were available for our analysis. For all patients LDH values were collected within one month before the start of treatment and after the end of therapy. For study purposes we divided our patients into two groups, according to LDH pre-treatment levels, cut-off levels was determined with ROC curve analysis. Patients were, also, classified according to the variation in LDH serum levels pre- and post-treatment (increased vs decreased). Results Patients proved homogeneous for all clinical characteristics analyzed. In patients with LDH values under the cut-off median progression free survival (PFS) was 6.7 months, whereas it was 1.9 months in patients above the cut-off (p = 0.0002). Accordingly median overall survival (OS) was 13.2 months and 4.9 months (p = 0.0006). In patients with decreased LDH values after treatment median PFS was 6.8 months, and median OS was 21.0 months, whereas PFS was 2.9 months and OS 8.6 months in patients with increased LDH levels (PFS: p = 0.0087; OS: p = 0.0035). Conclusions In our experience, LDH seemed able to predict clinical outcome in terms of PFS and OS for HCC patients treated with sorafenib. Given the correlation between LDH levels and tumour angiogenesis we can speculate that patients with high LDH pretreatment levels may be optimal candidates for other emerging therapeutic agents or strategies targeting different molecular pathways.
Collapse
Affiliation(s)
| | - Mario Scartozzi
- Department of Medical Oncology, Translational Oncology Unit, AOU Ospedali Riuniti, Università Politecnica delle Marche, Via Conca 71, 60126 Ancona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
Masson N, Ratcliffe PJ. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab 2014; 2:3. [PMID: 24491179 PMCID: PMC3938304 DOI: 10.1186/2049-3002-2-3] [Citation(s) in RCA: 227] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/22/2014] [Indexed: 12/31/2022] Open
Abstract
Both tumor hypoxia and dysregulated metabolism are classical features of cancer. Recent analyses have revealed complex interconnections between oncogenic activation, hypoxia signaling systems and metabolic pathways that are dysregulated in cancer. These studies have demonstrated that rather than responding simply to error signals arising from energy depletion or tumor hypoxia, metabolic and hypoxia signaling pathways are also directly connected to oncogenic signaling mechanisms at many points. This review will summarize current understanding of the role of hypoxia inducible factor (HIF) in these networks. It will also discuss the role of these interconnected pathways in generating the cancer phenotype; in particular, the implications of switching massive pathways that are physiologically 'hard-wired’ to oncogenic mechanisms driving cancer.
Collapse
Affiliation(s)
| | - Peter J Ratcliffe
- The Hypoxia Biology Laboratory, The Henry Wellcome Building for Molecular Physiology, The University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| |
Collapse
|
220
|
Kuiper C, Dachs GU, Munn D, Currie MJ, Robinson BA, Pearson JF, Vissers MCM. Increased Tumor Ascorbate is Associated with Extended Disease-Free Survival and Decreased Hypoxia-Inducible Factor-1 Activation in Human Colorectal Cancer. Front Oncol 2014; 4:10. [PMID: 24551593 PMCID: PMC3912592 DOI: 10.3389/fonc.2014.00010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/17/2014] [Indexed: 12/13/2022] Open
Abstract
Ascorbate is a co-factor for the hydroxylases that regulate the transcription factor hypoxia-inducible factor (HIF)-1, which provides cancer cells with a metabolic and survival advantage in the hypoxic environment of solid tumors. However, whether ascorbate affects tumor development is a highly debated issue. We aimed to determine whether tumor ascorbate was associated with HIF-1 activation and patient disease-free survival. In this study, we undertook a retrospective observational analysis of tissue-banked tumor and paired normal tissue from 49 colorectal cancer patients, measuring ascorbate levels, HIF-1α and its downstream gene products BNIP3, and vascular endothelial cell growth factor (VEGF). Patient survival was monitored for the first 6 years after surgery. We found that ascorbate levels were lower in tumor tissue compared to normal tissue (p < 0.001) but overall levels varied considerably. HIF-1α, VEGF, and BNIP3 were elevated in tumor samples (p < 0.01). There was an inverse relationship between tumor ascorbate content and HIF-1 pathway activation (p = 0.002) and tumor size (p = 0.018). Higher tumor ascorbate content was associated with significantly improved disease-free survival in the first 6 years after surgery (p = 0.006), with 141–1,094 additional disease-free days. This was independent of tumor grade and stage. Survival advantage was associated with the amount of ascorbate in the tumor, but not with the amount in adjacent normal tissue. Our results demonstrate that higher tumor ascorbate content is associated with decreased HIF-1 activation, most likely due to the co-factor activity of ascorbate for the regulatory HIF hydroxylases. Our findings support the need for future studies to determine whether raising tumor ascorbate is possible with clinical intervention and whether this results in modification of hydroxylase-dependent pathways in the tumor.
Collapse
Affiliation(s)
- Caroline Kuiper
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand
| | - Delwyn Munn
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand
| | - Margaret J Currie
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand
| | - Bridget A Robinson
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand ; Canterbury Regional Cancer and Blood Service, Canterbury District Health Board, Christchurch Hospital , Christchurch , New Zealand
| | - John F Pearson
- Biostatistics and Computational Biology Unit, Department of Public Health and General Practice, University of Otago Christchurch , Christchurch , New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago Christchurch , Christchurch , New Zealand
| |
Collapse
|
221
|
Shuch B, Linehan WM, Srinivasan R. Aerobic glycolysis: a novel target in kidney cancer. Expert Rev Anticancer Ther 2014; 13:711-9. [PMID: 23773105 DOI: 10.1586/era.13.57] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Renal cell carcinoma (RCC) is a heterogenous group of cancers that arise from the nephron. While there are distinct histologic subtypes associated with common genetic alterations, most forms of RCC are linked by a common pathway of dysregulated metabolism. Reliance on aerobic glycolysis, a feature of cancer first hypothesized by Warburg, is a common feature in sporadic and hereditary forms of kidney cancer. Two hereditary forms of RCC, succinate dehydrogenase (SDH) and hereditary leiomyomatosis and RCC (HLRCC), are characterized by mutations in Krebs cycle enzymes, rendering them dependent on glycolysis for energy requirements. The reliance on these pathways may make them vulnerable to novel metabolic strategies, including inhibition of glycolysis, glucose uptake and macromolecule biosynthesis.
Collapse
Affiliation(s)
- Brian Shuch
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
222
|
Monzon JG, Heng DYC. Management of metastatic kidney cancer in the era of personalized medicine. Crit Rev Clin Lab Sci 2014; 51:85-97. [PMID: 24450515 DOI: 10.3109/10408363.2013.869544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patients with localized renal cell cancer (RCC) are often cured following surgical resection. However, a significant proportion of patients will experience recurrence or present with metastatic disease at distant sites and may be deemed incurable. The worldwide incidence of RCC is rising, affecting more than 271,000 people and resulting in 116,000 deaths each year. Unfortunately, advanced RCC is typically resistant to classical chemotherapy and radiotherapy. Previously, non-specific immunotherapies such as interleukin-2 and interferon were used in hopes of improving cancer immunity, leading to rare but durable responses. However, enthusiasm for these immunotherapies has waned due to limited patient responses, their excessive toxicities, and the emergence of alternative targeted therapies that have resulted in improved clinical endpoints for patients with metastatic RCC (mRCC). Strides in targeted treatment can be attributed to an improved understanding of the molecular underpinnings that cause and drive the progression of renal cell cancers. More recently, interest in immunotherapies has resurfaced, as agents inhibiting specific checkpoints involved in cancer immune evasion have demonstrated promising activity in patients with mRCC. Here we review the novel targeted agents, biomarkers and immunotherapies that promise to change the clinical outcomes for patients with advanced RCC.
Collapse
Affiliation(s)
- Jose G Monzon
- Department of Medical Oncology, Tom Baker Cancer Center, University of Calgary , Calgary, AB , Canada
| | | |
Collapse
|
223
|
Radiotherapy and the tumor microenvironment: mutual influence and clinical implications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:147-65. [PMID: 24272358 DOI: 10.1007/978-1-4614-5915-6_7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ionizing radiation has been employed in targeted cancer treatments for more than a century because of its cytotoxic effects on cancer cells. However, the responsiveness to radiation and the behavior of tumors in vivo may differ dramatically from observed behaviors of isolated cancer cells in vitro. While not fully understood, these discrepancies are due to a complex constellation of extracellular and intercellular factors that are together termed the tumor microenvironment. Radiation may alter or affect the components of the adjacent tumor microenvironment in significant ways, often with consequences for cancer cells beyond the direct effects of the radiation itself. Moreover, different microenvironmental states, whether induced or at baseline, can modulate or even attenuate the effects of radiation, with consequences for therapeutic efficacy. This chapter describes this bidirectional relationship in detail, exploring the role and clinical implications of the tumor microenvironment with respect to therapeutic irradiation.
Collapse
|
224
|
Ma J, Zi Jiang Y, Shi H, Mi C, Li J, Xing Nan J, Wu X, Joon Lee J, Jin X. Cucurbitacin B inhibits the translational expression of hypoxia-inducible factor-1α. Eur J Pharmacol 2014; 723:46-54. [DOI: 10.1016/j.ejphar.2013.12.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 12/02/2013] [Accepted: 12/04/2013] [Indexed: 01/27/2023]
|
225
|
Hypoxia-Directed Drug Strategies to Target the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:111-45. [DOI: 10.1007/978-1-4614-5915-6_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
226
|
Park ST, Kim BR, Park SH, Lee JH, Lee EJ, Lee SH, Rho SB. Suppression of VEGF expression through interruption of the HIF‑1α and Akt signaling cascade modulates the anti‑angiogenic activity of DAPK in ovarian carcinoma cells. Oncol Rep 2013; 31:1021-9. [PMID: 24337450 DOI: 10.3892/or.2013.2928] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/02/2013] [Indexed: 11/05/2022] Open
Abstract
Death-associated protein kinase (DAPK) plays an important role in apoptosis regulation and has been shown to maintain antitumor and metastasis suppressor properties. In the present study, we investigated whether DAPK overexpression may mediate vascular endothelial growth factor (VEGF)/hypoxia-inducible factor-1α (HIF-1α) expression and angiogenic activity in the human carcinoma cell model system. VEGF plays a pivotal role in tumor angiogenesis and tumorigenesis. We found that DAPK significantly downregulated VEGF-induced endothelial cell proliferation, migration and tube formation as well as VEGF receptor-2 (VEGFR-2) phosphorylation in vitro. In addition, DAPK exhibited potent anti-angiogenic activity and clearly decreased the levels of VEGF and HIF-1α expression, a key regulator for angiogenesis. Notably, our results strongly indicated that DAPK can disturb VEGFR-2 transcriptional activity by inhibiting VEGFR-2 phosphorylation through the PI3K/Akt signaling cascade. Collectively, our study identified a novel function of DAPK in regulating cellular VEGF/HIF-1α activity during tumorigenesis, which may act together with its anti-angiogenic function to inhibit tumor progression.
Collapse
Affiliation(s)
- Sung Taek Park
- Department of Obstetrics and Gynecology, Hallym University, Seoul 150‑950, Republic of Korea
| | - Boh-Ram Kim
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi‑do 410‑769, Republic of Korea
| | - Sung Ho Park
- Department of Obstetrics and Gynecology, Hallym University, Seoul 150‑950, Republic of Korea
| | - Jeong Heon Lee
- Department of Obstetrics and Gynecology, Chonbuk National University Medical School, Jeonju 561‑712, Republic of Korea
| | - Eun-Ju Lee
- Department of Obstetrics and Gynecology, Chung‑Ang University School of Medicine/Chung‑Ang University Hospital, Seoul 156-755, Republic of Korea
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, Yongin-si, Gyeonggi-do 449-714, Republic of Korea
| | - Seung Bae Rho
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi‑do 410‑769, Republic of Korea
| |
Collapse
|
227
|
Glazer PM, Hegan DC, Lu Y, Czochor J, Scanlon SE. Hypoxia and DNA repair. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2013; 86:443-51. [PMID: 24348208 PMCID: PMC3848098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hypoxia is a characteristic feature of solid tumors and occurs very early in neoplastic development. Hypoxia transforms cell physiology in multiple ways, with profound changes in cell metabolism, cell growth, susceptibility to apoptosis, induction of angiogenesis, and increased motility. Over the past 20 years, our lab has determined that hypoxia also induces genetic instability. We have conducted a large series of experiments revealing that this instability occurs through the alteration of DNA repair pathways, including nucleotide excision repair, DNA mismatch repair, and homology dependent repair. Our work suggests that hypoxia, as a key component of solid tumors, can drive cancer progression through its impact on genomic integrity. However, the acquired changes in DNA repair that are induced by hypoxia may also render hypoxic cancer cells vulnerable to tailored strategies designed to exploit these changes.
Collapse
Affiliation(s)
- Peter M. Glazer
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut,To whom all correspondence should be
addressed: Peter M. Glazer, Department of Therapeutic Radiology, Yale
University, 333 Cedar St., New Haven, CT 06520-8040; Tele: 203-737-2788; Fax:
203-737-1467;
| | - Denise C. Hegan
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut
| | - Yuhong Lu
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut
| | - Jennifer Czochor
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut
| | - Susan E. Scanlon
- Department of Therapeutic Radiology, Yale School of
Medicine, New Haven, Connecticut,Department of Genetics, Yale School of Medicine, New
Haven, Connecticut
| |
Collapse
|
228
|
Ria R, Catacchio I, Berardi S, De Luisi A, Caivano A, Piccoli C, Ruggieri V, Frassanito MA, Ribatti D, Nico B, Annese T, Ruggieri S, Guarini A, Minoia C, Ditonno P, Angelucci E, Derudas D, Moschetta M, Dammacco F, Vacca A. HIF-1α of bone marrow endothelial cells implies relapse and drug resistance in patients with multiple myeloma and may act as a therapeutic target. Clin Cancer Res 2013; 20:847-58. [PMID: 24297864 DOI: 10.1158/1078-0432.ccr-13-1950] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To investigate the role of hypoxia-inducible factor-1α (HIF-1α) in angiogenesis and drug resistance of bone marrow endothelial cells of patients with multiple myeloma. EXPERIMENTAL DESIGN HIF-1α mRNA and protein were evaluated in patients with multiple myeloma endothelial cells (MMEC) at diagnosis, at relapse after bortezomib- or lenalidomide-based therapies or on refractory phase to these drugs, at remission; in endothelial cells of patients with monoclonal gammapathies of undetermined significance (MGUS; MGECs), and of those with benign anemia (controls). The effects of HIF-1α inhibition by siRNA or panobinostat (an indirect HIF-1α inhibitor) on the expression of HIF-1α proangiogenic targets, on MMEC angiogenic activities in vitro and in vivo, and on overcoming MMEC resistance to bortezomib and lenalidomide were studied. The overall survival of the patients was also observed. RESULTS Compared with the other endothelial cell types, only MMECs from 45% of relapsed/refractory patients showed a normoxic HIF-1α protein stabilization and activation that were induced by reactive oxygen species (ROS). The HIF-1α protein correlated with the expression of its proangiogenic targets. The HIF-1α inhibition by either siRNA or panobinostat impaired the MMECs angiogenesis-related functions both in vitro and in vivo and restored MMEC sensitivity to bortezomib and lenalidomide. Patients with MMECs expressing the HIF-1α protein had shorter overall survival. CONCLUSIONS The HIF-1α protein in MMECs may induce angiogenesis and resistance to bortezomib and lenalidomide and may be a plausible target for the antiangiogenic management of patients with well-defined relapsed/refractory multiple myeloma. It may also have prognostic significance.
Collapse
Affiliation(s)
- Roberto Ria
- Authors' Affiliations: Section of Internal Medicine, Department of Biomedical Sciences and Human Oncology; Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy, National Cancer Institute "Giovanni Paolo II", Bari, Italy; Department of Human Anatomy, Histology and Embryology, and Pathological Anatomy, University of Bari Medical School; Hematology Unit, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Oncologic Hospital; Laboratory of Preclinical and Translational Research, IRCCS Basilicata Cancer Reference Centre, Potenza; Department of Clinical and Experimental Medicine, University of Foggia Medical School, Foggia; Hematology Unit, Ospedale Di Venere, Carbonara di Bari, Bari; and Department of Haematology, Businco Hospital, Cagliari, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
229
|
Gupta B, Chiang L, Chae K, Lee DH. Phenethyl isothiocyanate inhibits hypoxia-induced accumulation of HIF-1α and VEGF expression in human glioma cells. Food Chem 2013; 141:1841-6. [DOI: 10.1016/j.foodchem.2013.05.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/16/2013] [Accepted: 05/02/2013] [Indexed: 01/11/2023]
|
230
|
Effects of tyrosine kinase inhibitors and CXCR4 antagonist on tumor growth and angiogenesis in rat glioma model: MRI and protein analysis study. Transl Oncol 2013; 6:660-9. [PMID: 24466368 DOI: 10.1593/tlo.13559] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/26/2013] [Accepted: 09/30/2013] [Indexed: 02/06/2023] Open
Abstract
The aim of the study was to determine the antiangiogenic efficacy of vatalanib, sunitinib, and AMD3100 in an animal model of human glioblastoma (GBM) by using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and tumor protein expression analysis. Orthotopic GBM-bearing animals were randomly assigned either to control group or vatalanib, sunitinib, and AMD3100 treatment groups. Following 2 weeks of drug treatment, tumor growth and vascular parameters were measured using DCE-MRI. Expression of different angiogenic factors in tumor extracts was measured using a membrane-based human antibody array kit. Tumor angiogenesis and invasion were determined by immunohistochemistry. DCE-MRI showed a significant increase in tumor size after vatalanib treatment. AMD3100-treated group showed a significant decrease in a number of vascular parameters determined by DCE-MRI. AMD3100 significantly decreased the expression of different angiogenic factors compared to sunitinib or vatalanib; however, there were no significant changes in vascular density among the groups. Sunitinib-treated animals showed significantly higher migration of the invasive cells, whereas in both vatalanib- and AMD3100-treated animals the invasive cell migration distance was significantly lower compared to that of control. Vatalanib and sunitinib resulted in suboptimal therapeutic effect, but AMD3100 treatment resulted in a significant reduction in tumor growth, permeability, interstitial space volume, and invasion of tumor cells in an animal model of GBM.
Collapse
|
231
|
Yan YE, Zhang J, Wang K, Xu Y, Ren K, Zhang BY, Shi M, Chen C, Shi Q, Tian C, Zhao G, Dong XP. Significant Reduction of the GLUT3 Level, but not GLUT1 Level, Was Observed in the Brain Tissues of Several Scrapie Experimental Animals and Scrapie-Infected Cell Lines. Mol Neurobiol 2013; 49:991-1004. [DOI: 10.1007/s12035-013-8574-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 10/21/2013] [Indexed: 11/30/2022]
|
232
|
Koperek O, Akin E, Asari R, Niederle B, Neuhold N. Expression of hypoxia-inducible factor 1 alpha in papillary thyroid carcinoma is associated with desmoplastic stromal reaction and lymph node metastasis. Virchows Arch 2013; 463:795-802. [DOI: 10.1007/s00428-013-1484-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/22/2013] [Accepted: 09/17/2013] [Indexed: 12/24/2022]
|
233
|
Welsh SJ, Dale AG, Lombardo CM, Valentine H, de la Fuente M, Schatzlein A, Neidle S. Inhibition of the hypoxia-inducible factor pathway by a G-quadruplex binding small molecule. Sci Rep 2013; 3:2799. [PMID: 24165797 PMCID: PMC3810677 DOI: 10.1038/srep02799] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/06/2013] [Indexed: 12/20/2022] Open
Abstract
The hypoxia-inducible transcription factor (HIF) co-ordinates the response of tumours to low oxygen by stimulating genes involved in metabolism and angiogenesis. HIF pathway activation is associated with decreased progression-free survival and increased mortality; compounds that target this pathway are potential agents for the treatment of a range of solid tumour malignancies. Renal cancers are likely to be particularly sensitive to inhibition of the HIF pathway since ~80% show constitutive activation of HIF. We have previously described the di-substituted naphthalene derivative, CL67, which binds to a G-quadruplex higher-order structure in the HIF promoter sequence in vitro. We show here that CL67 blocks HIF expression leading to inhibition of HIF-transactivation and down-regulation of downstream target genes and proteins in renal carcinoma cell lines and in a mouse xenograft model of renal cancer. This inhibition is independent of pathways that control HIF abundance through oxygen-dependant degradation and oxygen dependant HIF sub-unit expression.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Carcinogens/pharmacology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Cell Hypoxia/drug effects
- Female
- G-Quadruplexes/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Immunoenzyme Techniques
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Mice
- Mice, Nude
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Response Elements/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Small Molecule Libraries/pharmacology
- Transcriptional Activation/drug effects
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Sarah J. Welsh
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Aaron G. Dale
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Caterina M. Lombardo
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Helen Valentine
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Maria de la Fuente
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Andreas Schatzlein
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Stephen Neidle
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| |
Collapse
|
234
|
Goggins BJ, Chaney C, Radford-Smith GL, Horvat JC, Keely S. Hypoxia and Integrin-Mediated Epithelial Restitution during Mucosal Inflammation. Front Immunol 2013; 4:272. [PMID: 24062740 PMCID: PMC3769679 DOI: 10.3389/fimmu.2013.00272] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/27/2013] [Indexed: 12/27/2022] Open
Abstract
Epithelial damage and loss of intestinal barrier function are hallmark pathologies of the mucosal inflammation associated with conditions such as inflammatory bowel disease. In order to resolve inflammation and restore intestinal integrity the mucosa must rapidly and effectively repair the epithelial barrier. Epithelial wound healing is a highly complex and co-ordinated process and the factors involved in initiating intestinal epithelial healing are poorly defined. In order for restitution to be successful there must be a balance between epithelial cell migration, proliferation, and differentiation within and adjacent to the inflamed area. Endogenous, compensatory epithelial signaling pathways are activated by the changes in oxygen tensions that accompany inflammation. These signaling pathways induce the activation of key transcription factors, governing anti-apoptotic, and proliferative processes resulting in epithelial cell survival, proliferation, and differentiation at the site of mucosal inflammation. In this review, we will discuss the primary processes involved in epithelial restitution with a focus on the role of hypoxia-inducible factor and epithelial integrins as mediators of epithelial repair following inflammatory injury at the mucosal surface.
Collapse
Affiliation(s)
- Bridie J Goggins
- School of Biomedical Sciences and Pharmacy, University of Newcastle , Newcastle, NSW , Australia ; Hunter Medical Research Institute , New Lambton, NSW , Australia
| | | | | | | | | |
Collapse
|
235
|
Zeng KW, Li N, Dong X, Ma ZZ, Jiang Y, Jin HW, Tu PF. Sprengerinin C exerts anti-tumorigenic effects in hepatocellular carcinoma via inhibition of proliferation and angiogenesis and induction of apoptosis. Eur J Pharmacol 2013; 714:261-73. [DOI: 10.1016/j.ejphar.2013.04.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 04/15/2013] [Accepted: 04/18/2013] [Indexed: 11/30/2022]
|
236
|
Abstract
Breast cancer is now the most frequently diagnosed cancer and leading cause of cancer death in women worldwide. Strategies targeting the primary tumour have markedly improved, but systemic treatments to prevent metastasis are less effective; metastatic disease remains the underlying cause of death in the majority of patients with breast cancer who succumb to their disease. The long latency period between initial treatment and eventual recurrence in some patients suggests that a tumour may both alter and respond to the host systemic environment to facilitate and sustain disease progression. Results from studies in animal models suggest that specific subtypes of breast cancer may direct metastasis through recruitment and activation of haematopoietic cells. In this review, we focus on data implicating breast cancer as a systemic disease.
Collapse
Affiliation(s)
- A J Redig
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
237
|
Liu Y, Zhang JB, Qin Y, Wang W, Wei L, Teng Y, Guo L, Zhang B, Lin Z, Liu J, Ren ZG, Ye QH, Xie Y. PROX1 promotes hepatocellular carcinoma metastasis by way of up-regulating hypoxia-inducible factor 1α expression and protein stability. Hepatology 2013; 58:692-705. [PMID: 23505027 DOI: 10.1002/hep.26398] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 03/10/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is one of the most common cancers and the third leading cause of death from cancer worldwide. HCC has a very poor prognosis because of tumor invasiveness, frequent intrahepatic spread, and extrahepatic metastasis. The molecular mechanism of HCC invasiveness and metastasis is poorly understood. The homeobox protein PROX1 is required for hepatocyte migration during mouse embryonic liver development. In this study, we show that high PROX1 protein expression in primary HCC tissues is associated with significantly worse survival and early tumor recurrence in postoperative HCC patients. Knockdown of PROX1 expression in HCC cells inhibited cell migration and invasiveness in vitro and HCC metastasis in nude mice while overexpression of PROX1 in HCC cells promoted these processes. PROX1's pro-metastasis activity is most likely attributed to its up-regulation of hypoxia-inducible factor 1α (HIF-1α) transcription and stabilization of HIF-1α protein by recruiting histone deacetylase 1 (HDAC1) to prevent the acetylation of HIF-1α, which subsequently induces an epithelial-mesenchymal transition response in HCC cells. We further demonstrated the prognostic value of using the combination of PROX1 and HDAC1 levels to predict postoperative survival and early recurrence of HCC. CONCLUSION PROX1 is a critical factor that promotes HCC metastasis.
Collapse
Affiliation(s)
- Yanfeng Liu
- Key Laboratory of Medical Molecular Virology (MOE & MOH), Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Larsen AK, de Gramont A, Poindessous V, Bouygues A, Ayadi M, Mésange P. Functions and Clinical Implications of Autocrine VEGF Signaling in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2013. [DOI: 10.1007/s11888-013-0177-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
239
|
Role of vascular endothelial progenitor cells in construction of new vascular loop. Microvasc Res 2013; 90:1-11. [PMID: 23851047 DOI: 10.1016/j.mvr.2013.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 06/25/2013] [Accepted: 06/28/2013] [Indexed: 11/22/2022]
Abstract
Since bone marrow-derived endothelial progenitor cells (EPCs) have been detected in adult peripheral blood, the mode of vasculogenesis in the adult tissue has caught attention in field of vascularization research. To confirm the role of EPCs in construction of new vascular loop, we took the biomaterial scaffold implanted into adult rat as an experimental model to observe and examine the actions of the EPCs in neovascularization of the material by immunohistochemistry and transmission electron microscopy. Additionally, by establishing a chemotactic migration model for vascular endothelial cells (ECs) and EPCs, the migrations of ECs and EPCs were explored in simulations. The results of 20,000 simulations showed that the number of the vascular loops assisted by the EPCs was 2-5 times that of the vascular sprouts being naturally joined. Based on the results of experiments and simulations, we conclude that the EPCs are able to assist the angiogenic sprouts in joining under the condition of plenty of the EPCs being mobilized, which aggregate at sites close to sprout tips, forming a cell cord and differentiating to ECs in situ, and become vessel segments between neighboring sprouts. This suggests that there is a difference between the adult and embryo in the manner of vasculogenesis and that a small number of EPCs can play an important role to make the new blood vessels achieve rapid functionalization.
Collapse
|
240
|
Nagaraju GP, Ganji PN, Park W, Wen J, Mahaseth H, Landry J, Farris AB, Willingham F, Sullivan PS, Proia DA, El-Hariry I, Taliaferro-Smith L, Diaz R, El-Rayes BF. Antiangiogenic effects of ganetespib in colorectal cancer mediated through inhibition of HIF-1α and STAT-3. Angiogenesis 2013; 16:903-17. [PMID: 23838996 DOI: 10.1007/s10456-013-9364-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 06/24/2013] [Indexed: 11/28/2022]
Abstract
Hypoxia-inducible factors (HIFs) and STAT-3 play essential roles in angiogenesis. HIF-1α and STAT-3 are clients of the heat shock protein 90 (HSP90). We hypothesized that ganetespib, a potent HSP90 inhibitor, would disrupt angiogenesis in colorectal cancer (CRC) through inhibition of HIF-1α and STAT-3. CRC cell lines (HCT116 and HT29) were used in all the experiments. Egg CAM and HUVEC assays revealed decreased angiogenesis in ganetespib treated cell lines. Ganetespib inhibited matrigel plug vascularization and tumor growth of xenografts. Significant inhibition of PDGFA, FGF2, Ang-1, Ang-2, TGFβ1, VEGF, HIF-1α and STAT-3 expression was observed in both cell lines treated ganetespib. HIF-1α overexpression resulted in the increase VEGF and STAT-3 expression and this was inhibited by ganetespib. HIF-1α knockdown inhibited VEGF and STAT-3 expression. STAT-3 knockdown inhibited VEGF but not HIF-1α expression. HSP90, STAT-3 and VEGF expression was significantly higher in CRC compared to adjacent normal tissue. Significant downregulation of PDGFA, FGF2, Ang-1, Ang-2, TGFβ1, VEGF, STAT-3 and HIF-1α mRNA was observed in the post ganetespib treatment tumor samples from patients with rectal cancer. These results collectively suggest that inhibition of HSP90 is a promising antiangiogenic strategy in CRC. HSP90 angiogenic effects are mediated through HIF-1α and STAT-3.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Emory University, 1365 Clifton RD NE, Office 2080, Atlanta, GA, 30322, USA
| | - Purnachandra Nagaraju Ganji
- Department of Hematology and Medical Oncology, Emory University, 1365 Clifton RD NE, Office 2080, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
241
|
Liu KH, Huynh N, Patel O, Shulkes A, Baldwin G, He H. P21-activated kinase 1 promotes colorectal cancer survival by up-regulation of hypoxia-inducible factor-1α. Cancer Lett 2013; 340:22-9. [PMID: 23811286 DOI: 10.1016/j.canlet.2013.06.024] [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: 05/21/2013] [Revised: 06/18/2013] [Accepted: 06/20/2013] [Indexed: 12/16/2022]
Abstract
P21 activated kinase 1 (PAK1) enhances colorectal cancer (CRC) progression by stimulating Wnt/β-catenin and Ras oncogene, which promote CRC survival via stimulation of hypoxia-inducible factor 1α (HIF-1α). The aim of this study was to assess the mechanism involved in the stimulation by PAK1 of CRC survival. PAK1 promoted CRC cell survival by up-regulation of HIF-1α. PAK1 was over-expressed and hyper-activated in tumors of ApcΔ(14/+) mice, which was correlated with over-expression of HIF-1α and β-catenin. Inhibition of PAK1 decreased tumor growth and the expression of HIF-1α and β-catenin in tumors of ApcΔ(14/+) mice, and suppressed xenograft tumor survival in SCID mice. These findings indicate that PAK1 stimulates CRC survival by up-regulation of HIF-1α.
Collapse
Affiliation(s)
- Kevin H Liu
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia
| | | | | | | | | | | |
Collapse
|
242
|
Love RJ, Jones KS. Transient inhibition of connective tissue infiltration and collagen deposition into porous poly(lactic-co-glycolic acid) discs. J Biomed Mater Res A 2013; 101:3599-606. [PMID: 23766241 DOI: 10.1002/jbm.a.34648] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 01/12/2013] [Accepted: 02/11/2013] [Indexed: 11/11/2022]
Abstract
Connective tissue rapidly proliferates on and around biomaterials implanted in vivo, which impairs the function of the engineered tissues, biosensors, and devices. Glucocorticoids can be utilized to suppress tissue ingrowth, but can only be used for a limited time because they nonselectively arrest cell proliferation in the local environment. The present study examined use of a prolyl-4-hydroxylase inhibitor, 1,4-dihydrophenonthrolin-4-one-3-carboxylic acid (1,4-DPCA), to suppress connective tissue ingrowth in porous PLGA discs implanted in the peritoneal cavity for 28 days. The prolyl-4-hydroxylase inhibitor was found to be effective at inhibiting collagen deposition within and on the outer surface of the disc, and also limited connective tissue ingrowth, but not to the extent of glucocorticoid inhibition. Finally, it was discovered that 1,4-DPCA suppressed Scavenger Receptor A expression on a macrophage-like cell culture, which may account for the drug's ability to limit connective tissue ingrowth in vivo.
Collapse
Affiliation(s)
- Ryan J Love
- School of Biomedical Engineering, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
243
|
Díaz B, Yuen A, Iizuka S, Higashiyama S, Courtneidge SA. Notch increases the shedding of HB-EGF by ADAM12 to potentiate invadopodia formation in hypoxia. ACTA ACUST UNITED AC 2013; 201:279-92. [PMID: 23589494 PMCID: PMC3628517 DOI: 10.1083/jcb.201209151] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hypoxia increases the levels of ADAM12 in a Notch-dependent manner, leading to increased ectodomain shedding of HB-EGF and subsequent promotion of invadopodia formation. Notch regulates cell–cell contact-dependent signaling and is activated by hypoxia, a microenvironmental condition that promotes cellular invasion during both normal physiology and disease. The mechanisms by which hypoxia and Notch regulate cellular invasion are not fully elucidated. In this paper, we show that, in cancer cells, hypoxia increased the levels and activity of the ADAM12 metalloprotease in a Notch signaling–dependent manner, leading to increased ectodomain shedding of the epidermal growth factor (EGF) receptor (EGFR) ligand heparin-binding EGF-like growth factor. Released HB-EGF induced the formation of invadopodia, cellular structures that aid cancer cell invasion. Thus, we describe a signaling pathway that couples cell contact–dependent signaling with the paracrine activation of the EGFR, indicating cross talk between the Notch and EGFR pathways in promoting cancer cell invasion. This signaling pathway might regulate the coordinated acquisition of invasiveness by neighboring cells and mediate the communication between normoxic and hypoxic areas of tumors to facilitate cancer cell invasion.
Collapse
Affiliation(s)
- Begoña Díaz
- Cancer Center, Tumor Microenvironment Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
244
|
Electroacupuncture at Zusanli (ST36) accelerates intracerebral hemorrhage-induced angiogenesis in rats. Chin J Integr Med 2013; 19:367-73. [DOI: 10.1007/s11655-013-1458-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Indexed: 02/01/2023]
|
245
|
Yi T, Papadopoulos E, Hagner PR, Wagner G. Hypoxia-inducible factor-1α (HIF-1α) promotes cap-dependent translation of selective mRNAs through up-regulating initiation factor eIF4E1 in breast cancer cells under hypoxia conditions. J Biol Chem 2013; 288:18732-42. [PMID: 23667251 DOI: 10.1074/jbc.m113.471466] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Hypoxia promotes tumor evolution and metastasis, and hypoxia-inducible factor-1α (HIF-1α) is a key regulator of hypoxia-related cellular processes in cancer. The eIF4E translation initiation factors, eIF4E1, eIF4E2, and eIF4E3, are essential for translation initiation. However, whether and how HIF-1α affects cap-dependent translation through eIF4Es in hypoxic cancer cells has been unknown. Here, we report that HIF-1α promoted cap-dependent translation of selective mRNAs through up-regulation of eIF4E1 in hypoxic breast cancer cells. Hypoxia-promoted breast cancer tumorsphere growth was HIF-1α-dependent. We found that eIF4E1, not eIF4E2 or eIF4E3, is the dominant eIF4E family member in breast cancer cells under both normoxia and hypoxia conditions. eIF4E3 expression was largely sequestered in breast cancer cells at normoxia and hypoxia. Hypoxia up-regulated the expression of eIF4E1 and eIF4E2, but only eIF4E1 expression was HIF-1α-dependent. In hypoxic cancer cells, HIF-1α-up-regulated eIF4E1 enhanced cap-dependent translation of a subset of mRNAs encoding proteins important for breast cancer cell mammosphere growth. In searching for correlations, we discovered that human eIF4E1 promoter harbors multiple potential hypoxia response elements. Furthermore, using chromatin immunoprecipitation (ChIP) and luciferase and point mutation assays, we found that HIF-1α utilized hypoxia response elements in the human eIF4E1 proximal promoter region to activate eIF4E1 expression. Our study suggests that HIF-1α promotes cap-dependent translation of selective mRNAs through up-regulating eIF4E1, which contributes to tumorsphere growth of breast cancer cells at hypoxia. The data shown provide new insights into protein synthesis mechanisms in cancer cells at low oxygen levels.
Collapse
Affiliation(s)
- Tingfang Yi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
246
|
Crucet M, Wüst SJA, Spielmann P, Lüscher TF, Wenger RH, Matter CM. Hypoxia enhances lipid uptake in macrophages: role of the scavenger receptors Lox1, SRA, and CD36. Atherosclerosis 2013; 229:110-7. [PMID: 23706521 DOI: 10.1016/j.atherosclerosis.2013.04.034] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 04/11/2013] [Accepted: 04/26/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The core of advanced atherosclerotic plaques turns hypoxic as the arterial wall thickens and oxygen diffusion capacity becomes impaired. Macrophage-derived foam cells play a pivotal role in atherosclerotic plaque formation by expressing scavenger receptors that regulate lipid uptake. However, the role of hypoxia in scavenger receptor regulation remains incompletely understood. METHODS AND RESULTS Using RT-qPCR, flow cytometry and immunoblotting, we found that mRNA and protein expression levels of the scavenger receptor A (SRA) and the cluster of differentiation 36 (CD36) were upregulated by oxidized low-density lipoprotein (oxLDL), but decreased following exposure of macrophages to hypoxia. In contrast, lectin-like oxLDL receptor (Lox-1) mRNA and protein levels were upregulated under hypoxic conditions. Flow cytometry confirmed the increased lipid content in macrophages after exposure to 0.2% oxygen and the hypoxia-mimetic dimethyloxalylglycine (DMOG). Antibody-mediated blocking of Lox-1 receptor decreased the hypoxic induction of oxLDL uptake and lipid content. RNAi-mediated knock-down of hypoxia-inducible factor (HIF)-1α in macrophages attenuated the hypoxic induction of Lox-1. CONCLUSIONS Hypoxia increases lipid uptake into macrophages and differentially regulates the expression of oxLDL receptors. Lox-1 plays a major role in hypoxia-induced foam cell formation which is, at least in part, mediated by HIF-1α.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Atherosclerosis/metabolism
- Atherosclerosis/physiopathology
- Biological Transport/physiology
- CD36 Antigens/metabolism
- Carcinoma, Hepatocellular
- Cell Line, Tumor
- Cholesterol/metabolism
- Foam Cells/metabolism
- Gene Knockdown Techniques
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Lipid Metabolism/physiology
- Lipoproteins, LDL/metabolism
- Liver Neoplasms
- Macrophages/cytology
- Macrophages/metabolism
- Mice
- RNA, Messenger/metabolism
- Scavenger Receptors, Class A/genetics
- Scavenger Receptors, Class A/metabolism
- Scavenger Receptors, Class E/genetics
- Scavenger Receptors, Class E/immunology
- Scavenger Receptors, Class E/metabolism
Collapse
Affiliation(s)
- Margot Crucet
- Cellular Oxygen Physiology, Institute of Physiology, University of Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
247
|
Tamaki Y, Nakade Y, Yamauchi T, Makino Y, Yokohama S, Okada M, Aso K, Kanamori H, Ohashi T, Sato K, Nakao H, Haneda M, Yoneda M. Angiotensin II type 1 receptor antagonist prevents hepatic carcinoma in rats with nonalcoholic steatohepatitis. J Gastroenterol 2013; 48:491-503. [PMID: 22886508 DOI: 10.1007/s00535-012-0651-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 07/23/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Angiotensin II type 1 receptor blockers (ARBs) have been reported to attenuate hepatic fibrosis in non-alcoholic steatohepatitis (NASH). However, it is uncertain whether ARBs prevent hepatocarcinogenesis in NASH even after hepatic fibrosis has developed. METHODS Male Wistar rats were fed with a choline-deficient, L-amino acid-defined (CDAA) diet for 24 weeks, and then fed with the CDAA diet with telmisartan (2 mg/kg/day), a novel ARB, or vehicle for another 24 weeks. The liver histology and the expression of genes and proteins related to angiogenesis were investigated. RESULTS The 24-week CDAA diet induced liver cirrhosis. The 48-week CDAA diet exacerbated liver cirrhosis, and developed hepatocellular carcinoma (HCC) in 54.6 % of the rats concurrently with increases of hypoxia-inducible factor-1α (HIF-1α) protein and vascular endothelial growth factor (VEGF) mRNA, which are potent angiogenic factors in the liver. Telmisartan inhibited hepatic fibrosis and preneoplastic lesions and prevented the development of HCC along with inducing decreases in HIF-1α protein and VEGF mRNA. CONCLUSIONS These data indicated that telmisartan may prevent hepatocarcinogenesis through the inhibition of hepatic angiogenesis even after liver cirrhosis has been established.
Collapse
MESH Headings
- Amino Acids/administration & dosage
- Angiotensin II Type 1 Receptor Blockers/therapeutic use
- Animals
- Anticarcinogenic Agents/therapeutic use
- Benzimidazoles/therapeutic use
- Benzoates/therapeutic use
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/prevention & control
- Cell Transformation, Neoplastic/drug effects
- Choline Deficiency/complications
- Diet/adverse effects
- Drug Evaluation, Preclinical/methods
- Fatty Liver/complications
- Fatty Liver/drug therapy
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Gene Expression Regulation/drug effects
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Liver/blood supply
- Liver Cirrhosis/complications
- Liver Cirrhosis/drug therapy
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Neoplasms, Experimental/etiology
- Liver Neoplasms, Experimental/prevention & control
- Male
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/prevention & control
- Non-alcoholic Fatty Liver Disease
- Rats
- Rats, Wistar
- Telmisartan
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Yosui Tamaki
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical College, Asahikawa, Hokkaido, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
248
|
Park SI, Suh DS, Kim SJ, Choi KU, Yoon MS. Correlation between biological marker expression and F-fluorodeoxyglucose uptake in cervical cancer measured by positron emission tomography. ACTA ACUST UNITED AC 2013; 36:169-74. [PMID: 23548964 DOI: 10.1159/000349944] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND This study aimed to determine whether several biologic markers were associated with (18)fluorine-fluorodeoxyglucose ((18)F-FDG) uptake in patients with carcinoma of the cervix. PATIENTS AND METHODS 60 patients with International Federation of Gynecology and Obstetrics (FIGO) stages IA2 to IIB cervical cancer, who underwent (18)FDG positron emission tomography/computed tomography (PET/CT), were included in the current study. All patients underwent radical hysterectomy. Tumor sections were stained by immunohistochemistry for glucose transporter 1 (GLUT1), carbonic anhydrase-IX (CA-IX), vascular endothelial growth factor (VEGF), hexokinase type I (HK-I), hexokinase type II (HK-II), and cytoplasmic and nuclear hypoxia-inducible factor (HIF) 1α. RESULTS The expression of GLUT1 (p = 0.005), VEGF (p = 0.021), HK-II (p = 0.009), and cytoplasmic HIF1α (p = 0.024) was significantly associated with a higher median standardized uptake value (SUVmax). There was a positive correlation between (18)F-FDG uptake and GLUT1 (p = 0.008), CA-IX (p = 0.030), HK-II (p < 0.001) as well as cytoplasmic HIF1α (p = 0.016), whereas this relationship was not observed among the VEGF, HK-I and nuclear HIF1α. CONCLUSION The data presented in this study indicate that (18)F-FDG uptake is associated with the presence of GLUT1, VEGF, nuclear HK-II, and cytoplasmic HIF1α. There was also a significant correlation among the rate of expression of GLUT1, HK-II, cytoplasmic HIF1α, and CAIX in carcinomas of the cervix.
Collapse
Affiliation(s)
- Sang I Park
- Department of Obstetrics and Gynecology, Dongnam Institute of Radiological and Medical Sciences, Gijang-gun, Republic of Korea
| | | | | | | | | |
Collapse
|
249
|
A hypoxia-inducible factor (HIF)-3α splicing variant, HIF-3α4 impairs angiogenesis in hypervascular malignant meningiomas with epigenetically silenced HIF-3α4. Biochem Biophys Res Commun 2013; 433:139-44. [PMID: 23485455 DOI: 10.1016/j.bbrc.2013.02.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 02/14/2013] [Indexed: 12/23/2022]
Abstract
Hypoxia inducible factor is a dominant regulator of adaptive cellular responses to hypoxia and controls the expression of a large number of genes regulating angiogenesis as well as metabolism, cell survival, apoptosis, and other cellular functions in an oxygen level-dependent manner. When a neoplasm is able to induce angiogenesis, tumor progression occurs more rapidly because of the nutrients provided by the neovasculature. Meningioma is one of the most hypervascular brain tumors, making anti-angiogenic therapy an attractive novel therapy for these tumors. HIF-3α has been conventionally regarded as a dominant-negative regulator of HIF-1α, and although alternative HIF-3α splicing variants are extensively reported, their specific functions have not yet been determined. In this study, we found that the transcription of HIF-3α4 was silenced by the promoter DNA methylation in meningiomas, and inducible HIF-3α4 impaired angiogenesis, proliferation, and metabolism/oxidation in hypervascular meningiomas. Thus, HIF-3α4 could be a potential molecular target in meningiomas.
Collapse
|
250
|
Liu Z, Jia X, Duan Y, Xiao H, Sundqvist KG, Permert J, Wang F. Excess glucose induces hypoxia-inducible factor-1α in pancreatic cancer cells and stimulates glucose metabolism and cell migration. Cancer Biol Ther 2013; 14:428-35. [PMID: 23377827 DOI: 10.4161/cbt.23786] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pancreatic cancer patients frequently show hyperglycemia, but it is uncertain whether hyperglycemia stimulates pancreatic cancer cells. We have investigated whether excess glucose induces hypoxia-inducible factor-1α (HIF-1α) and stimulates glucose metabolism and cell migration in pancreatic cancer cells. We studied wild-type (wt) MiaPaCa2 pancreatic cancer cells and a MiaPaCa2 subline (namely si-MiaPaCa2) that had HIF-1α-specific small interfering RNA. Wt-MiaPaCa2 cells are known to be HIF-1α-positive in hypoxia and HIF-1α-negative in normoxia, whereas si-MiaPaCa2 cells are devoid of HIF-1α in both normoxia and hypoxia. We incubated these cells with different amounts of glucose and determined HIF-1α mRNA and protein by real-time polymerase chain reaction and western blotting. We determined glucose consumption, lactate production and intracellular hexokinase-II and ATP to assess glucose metabolisms and determined pyruvate dehydrogenase kinase-1, reactive oxygen species and fumarate to assess mitochondrial activities. Further, we studied cell migration using a Boyden chamber. Excess glucose (16.7-22.2mM) increased HIF-1α in hypoxic wt-MiaPaCa2 cells. HIF-1α expression increased ATP contents and inhibited mitochondrial activities. Extracellular glucose and hypoxia stimulated glucose metabolisms independent of HIF-1α. Excess glucose stimulated the migration of wt- and si-MiaPaCa2 cells in both normoxia and hypoxia. Thus, glucose stimulated cell migration independent of HIF-1α. Nevertheless, hypoxic wt-MiaPaCa2 cells showed greater migrating ability than their si-MiaPaCa2 counterparts. We conclude that (1) excess glucose increases HIF-1α and ATP in hypoxic wt-MiaPaCa2 cells, (2) extracellular glucose and hypoxia regulate glucose metabolisms independent of HIF-1α and (3) glucose stimulates cell migration by mechanisms that are both dependent on HIF-1α and independent of it.
Collapse
Affiliation(s)
- Zhiwen Liu
- Department of Clinical Immunology, Karolinska University Hospital, Huddinge, Sweden
| | | | | | | | | | | | | |
Collapse
|