51
|
Aier I, Varadwaj PK. Understanding the Mechanism of Cell Death in Gemcitabine Resistant Pancreatic Ductal Adenocarcinoma: A Systems Biology Approach. Curr Genomics 2019; 20:483-490. [PMID: 32655287 PMCID: PMC7327974 DOI: 10.2174/1389202920666191025102726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/11/2019] [Accepted: 10/11/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Gemcitabine is the standard chemotherapeutic drug administered in advanced Pancreatic Ductal Adenocarcinoma (PDAC). However, due to drug resistance in PDAC patients, this treatment has become less effective. Over the years, clinical trials for the quest of finding novel compounds that can be used in combination with gemcitabine have met very little success. OBJECTIVE To predict the driving factors behind pancreatic ductal adenocarcinoma, and to understand the effect of these components in the progression of the disease and their contribution to cell growth and proliferation. METHODS With the help of systems biology approaches and using gene expression data, which is generally found in abundance, dysregulated elements in key signalling pathways were predicted. Prominent dysregulated elements were integrated into a model to simulate and study the effect of gemcitabine-induced hypoxia. RESULTS In this study, several transcription factors in the form of key drivers of cancer-related genes were predicted with the help of CARNIVAL, and the effect of gemcitabine-induced hypoxia on the apoptosis pathway was shown to have an effect on the downstream elements of two primary pathway models; EGF/VEGF and TNF signalling pathway. CONCLUSION It was observed that EGF/VEGF signalling pathway played a major role in inducing drug resistance through cell growth, proliferation, and avoiding cell death. Targeting the major upstream components of this pathway could potentially lead to successful treatment.
Collapse
Affiliation(s)
- Imlimaong Aier
- Department of Bioinformatics and Applied Science, Indian Institute of Information Technology, Allahabad, 20015, India
| | - Pritish K. Varadwaj
- Department of Bioinformatics and Applied Science, Indian Institute of Information Technology, Allahabad, 20015, India
| |
Collapse
|
52
|
Zhang Z, Ji S, Zhang B, Liu J, Qin Y, Xu J, Yu X. Role of angiogenesis in pancreatic cancer biology and therapy. Biomed Pharmacother 2018; 108:1135-1140. [PMID: 30372814 DOI: 10.1016/j.biopha.2018.09.136] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/23/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis, and there is a close parallel between disease mortality and incidence. Malignancy is often diagnosed at an advanced stage due to the lack of early symptoms. For the majority of advanced or metastatic pancreatic cancer patients, therapeutic options are limited. Although several new chemotherapeutic regimens have been developed, the overall response rate remains low. Invasive tumour growth and distant metastasis require angiogenesis, a hallmark of cancer, and angiogenic inhibition is a valuable option for cancer therapy. Some anti-angiogenic drugs have been developed for cancer treatment. This review will focus on the role of angiogenesis and anti-angiogenic treatment strategies as well as combination therapy in pancreatic cancer. Translational information from recent molecular biology and animal studies is also summarized. Finally, the dosing schedule for bevacizumab with other chemotherapeutic protocols for pancreatic cancer treatment is discussed.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
53
|
Tamtaji OR, Mirhosseini N, Reiter RJ, Behnamfar M, Asemi Z. Melatonin and pancreatic cancer: Current knowledge and future perspectives. J Cell Physiol 2018; 234:5372-5378. [PMID: 30229898 DOI: 10.1002/jcp.27372] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer has a high mortality rate due to the absence of early symptoms and subsequent late diagnosis; additionally, pancreatic cancer has a high resistance to radio- and chemotherapy. Multiple inflammatory pathways are involved in the pathophysiology of pancreatic cancer. Melatonin an indoleamine produced in the pineal gland mediated and receptor-independent action is the pancreas and other where has both receptors. Melatonin is a potent antioxidant and tissue protector against inflammation and oxidative stress. In vivo and in vitro studies have shown that melatonin supplementation is an appropriate therapeutic approach for pancreatic cancer. Melatonin may be an effective apoptosis inducer in cancer cells through regulation of a large number of molecular pathways including oxidative stress, heat shock proteins, and vascular endothelial growth factor. Limited clinical studies, however, have evaluated the role of melatonin in pancreatic cancer. This review summarizes what is known regarding the effects of melatonin on pancreatic cancer and the mechanisms involved.
Collapse
Affiliation(s)
- Omid Reza Tamtaji
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science, Center, San Antonio, Texas
| | - Morteza Behnamfar
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
54
|
Adebambo OA, Shea D, Fry RC. Cadmium disrupts signaling of the hypoxia-inducible (HIF) and transforming growth factor (TGF-β) pathways in placental JEG-3 trophoblast cells via reactive oxygen species. Toxicol Appl Pharmacol 2018; 342:108-115. [DOI: 10.1016/j.taap.2018.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 12/16/2022]
|
55
|
Longo V, Tamma R, Brunetti O, Pisconti S, Argentiero A, Silvestris N, Ribatti D. Mast cells and angiogenesis in pancreatic ductal adenocarcinoma. Clin Exp Med 2018; 18:319-323. [PMID: 29492715 DOI: 10.1007/s10238-018-0493-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 11/15/2017] [Indexed: 12/23/2022]
Abstract
Mast cells are recognized as critical components of the tumor stromal microenvironment in several solid and hematological malignancies, promoting angiogenesis and tumor growth. A correlation between mast cells infiltration, angiogenesis and tumor progression has been reported for pancreatic ductal adenocarcinoma as well. Mast cells contribute to the aggressiveness of the pancreatic ductal carcinoma enhancing the expression of several pro-angiogenic factors such as vascular endothelial growth factor, fibroblast growth factor-2, platelet-derived growth factor and angiopoietin-1 as well as stimulating the pancreatic cancer cells proliferation by IL-13 and tryptase. The disruption of this pro-angiogenic and proliferative stimulation by inhibiting the mast cells migration and degranulation is under investigation as a potential therapeutic approach in pancreatic ductal adenocarcinoma patients. This review will summarize the literature concerning the mast cells infiltration in the pancreatic ductal adenocarcinoma analyzing its role in angiogenesis and tumor progression.
Collapse
Affiliation(s)
- Vito Longo
- Department of Medical Oncology, Hospital of Taranto, Taranto, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit and Scientific Directorate, Cancer Institute "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124, Bari, Italy
| | | | - Antonella Argentiero
- Medical Oncology Unit and Scientific Directorate, Cancer Institute "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit and Scientific Directorate, Cancer Institute "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124, Bari, Italy.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
56
|
Pharmacologic ascorbate (P-AscH -) suppresses hypoxia-inducible Factor-1α (HIF-1α) in pancreatic adenocarcinoma. Clin Exp Metastasis 2018; 35:37-51. [PMID: 29396728 DOI: 10.1007/s10585-018-9876-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/31/2018] [Indexed: 12/24/2022]
Abstract
HIF-1α is a transcriptional regulator that functions in the adaptation of cells to hypoxic conditions; it strongly impacts the prognosis of patients with cancer. High-dose, intravenous, pharmacological ascorbate (P-AscH-), induces cytotoxicity and oxidative stress selectively in cancer cells by acting as a pro-drug for the delivery of hydrogen peroxide (H2O2); early clinical data suggest improved survival and inhibition of metastasis in patients being actively treated with P-AscH-. Previous studies have demonstrated that activation of HIF-1α is necessary for P-AscH- sensitivity. We hypothesized that pancreatic cancer (PDAC) progression and metastasis could be be targeted by P-AscH- via H2O2-mediated inhibition of HIF-1α stabilization. Our study demonstrates an oxygen- and prolyl hydroxylase-independent regulation of HIF-1α by P-AscH-. Additionally, P-AscH- decreased VEGF secretion in a dose-dependent manner that was reversible with catalase, consistent with an H2O2-mediated mechanism. Pharmacological and genetic manipulations of HIF-1α did not alter P-AscH--induced cytotoxicity. In vivo, P-AscH- inhibited tumor growth and VEGF expression. We conclude that P-AscH- suppresses the levels of HIF-1α protein in hypoxic conditions through a post-translational mechanism. These findings suggest potential new therapies specifically designed to inhibit the mechanisms that drive metastases as a part of PDAC treatment.
Collapse
|
57
|
Abstract
OBJECTIVES N-myc downstream-regulated gene-1 (NDRG1) is a hypoxia-inducible and differentiation-related protein and candidate biomarker in pancreatic cancer. As NDRG1 expression is lost in high-grade tumors, the effects of the differentiating histone deacetylase inhibitor trichostatin A (TSA) were examined in human pancreatic cancer cell lines representing different tumor grades. METHODS PANC-1 (poorly differentiated) and Capan-1 (moderately to well-differentiated) cells were treated with TSA. Effects were assessed in vitro by microscopic analysis, colorimetric assays, cell counts, real-time polymerase chain reaction, and Western blotting. RESULTS Treatment of PANC-1 cells over 4 days with 0.5 μM TSA restored cellular differentiation, inhibited proliferation, and enhanced p21 protein expression. Trichostatin A upregulated NDRG1 mRNA and protein levels under normoxia from day 1 and by 6-fold by day 4 (P < 0.01 at all time points). After 24 hours under hypoxia, NDRG1 expression was further increased in differentiated cells (P < 0.01). Favorable changes were identified in the expression of other hypoxia-regulated genes. CONCLUSIONS Histone deacetylase inhibitors offer a potential novel epidrug approach for pancreatic cancer by reversing the undifferentiated phenotype and allowing patients to overcome resistance and better respond to conventional cytotoxic treatments.
Collapse
|
58
|
Schofield HK, Tandon M, Park MJ, Halbrook CJ, Ramakrishnan SK, Kim EC, Shi J, Omary MB, Shah YM, Esni F, Pasca di Magliano M. Pancreatic HIF2α Stabilization Leads to Chronic Pancreatitis and Predisposes to Mucinous Cystic Neoplasm. Cell Mol Gastroenterol Hepatol 2017; 5:169-185.e2. [PMID: 29693047 PMCID: PMC5904051 DOI: 10.1016/j.jcmgh.2017.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Tissue hypoxia controls cell differentiation in the embryonic pancreas, and promotes tumor growth in pancreatic cancer. The cellular response to hypoxia is controlled by the hypoxia-inducible factor (HIF) proteins, including HIF2α. Previous studies of HIF action in the pancreas have relied on loss-of-function mouse models, and the effects of HIF2α expression in the pancreas have remained undefined. METHODS We developed several transgenic mouse models based on the expression of an oxygen-stable form of HIF2α, or indirect stabilization of HIF proteins though deletion of von Hippel-Lindau, thus preventing HIF degradation. Furthermore, we crossed both sets of animals into mice expressing oncogenic KrasG12D in the pancreas. RESULTS We show that HIF2α is not expressed in the normal human pancreas, however, it is up-regulated in human chronic pancreatitis. Deletion of von Hippel-Lindau or stabilization of HIF2α in mouse pancreata led to the development of chronic pancreatitis. Importantly, pancreatic HIF1α stabilization did not disrupt the pancreatic parenchyma, indicating that the chronic pancreatitis phenotype is specific to HIF2α. In the presence of oncogenic Kras, HIF2α stabilization drove the formation of cysts resembling mucinous cystic neoplasm (MCN) in humans. Mechanistically, we show that the pancreatitis phenotype is linked to expression of multiple inflammatory cytokines and activation of the unfolded protein response. Conversely, MCN formation is linked to activation of Wnt signaling, a feature of human MCN. CONCLUSIONS We show that pancreatic HIF2α stabilization disrupts pancreatic homeostasis, leading to chronic pancreatitis, and, in the context of oncogenic Kras, MCN formation. These findings provide new mouse models of both chronic pancreatitis and MCN, as well as illustrate the importance of hypoxia signaling in the pancreas.
Collapse
Affiliation(s)
- Heather K. Schofield
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Medical Scientist Training Program, University of Michigan, Ann Arbor, Michigan
| | - Manuj Tandon
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Min-Jung Park
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Christopher J. Halbrook
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Sadeesh K. Ramakrishnan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Esther C. Kim
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Jiaqi Shi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - M. Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Farzad Esni
- Department of Surgery, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Department of Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
59
|
Xu XL, Yang YR, Mo XF, Wei JL, Zhang XJ, You QD. Design, synthesis, and evaluation of benzofuran derivatives as novel anti-pancreatic carcinoma agents via interfering the hypoxia environment by targeting HIF-1α pathway. Eur J Med Chem 2017; 137:45-62. [PMID: 28554092 DOI: 10.1016/j.ejmech.2017.05.042] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/16/2017] [Accepted: 05/19/2017] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common type of pancreatic cancer, and has still been the medicinal mystery. New drugs and treatment strategies are urgently needed. In this study, 32 benzofuran derivatives are designed, synthesized and evaluated as potential agents against the pancreatic cancer. Among them, compound 9o with the best physicochemical and pharmacokinetic properties exhibited excellent cytotoxicity against many tumor cell lines. In vivo study showed that compound 9o dramatically suppressed the tumor growth of nude mice. Furthermore, compound 9o could affect the hypoxia environment through Hif-1α/VEGF pathway, resulting in the anti-angiogenic activity. These studies indicated that compound 9o was a promising candidate for the treatment of PDAC, deserving further studies.
Collapse
Affiliation(s)
- Xiao-Li Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying-Rui Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Fei Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jin-Lian Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Jin Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Organic Chemistry, School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
60
|
Yamada Y, Kinoshita I, Kohashi K, Yamamoto H, Kuma Y, Ito T, Koda K, Kisanuki A, Kurosawa M, Yoshimura M, Furue M, Oda Y. HIF-1α, MDM2, CDK4, and p16 expression in ischemic fasciitis, focusing on its ischemic condition. Virchows Arch 2017; 471:117-122. [PMID: 28477272 DOI: 10.1007/s00428-017-2122-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/02/2017] [Accepted: 04/04/2017] [Indexed: 11/28/2022]
Abstract
Ischemic fasciitis is a benign myofibroblastic lesion, occurring in the sacral region or proximal thigh of elderly or bedridden individuals. The pathogenesis of ischemic fasciitis is thought to be based on ischemic condition; however, it has never been demonstrated. In this study, we examined the expression of ischemia-associated proteins in ischemic fasciitis by immunohistochemical and genetic methods. Specifically, this study aimed to reveal the expression of HIF-1α, MDM2, CDK4, p16, and gene amplification of MDM2 gene. Seven cases of ischemic fasciitis from among the soft-tissue tumors registered at our institution were retrieved. Histopathological findings were as follows: poorly demarcated nodular masses, a proliferation of spindle-shaped fibroblastic or myofibroblastic cells with oval nuclei and eosinophilic or pale cytoplasm, zonal fibrinous deposition, pseudocystic degeneration, granulation-like proliferation of capillary vessels, ganglion-like cells, myxoid or hyalinized stroma, and chronic inflammatory infiltration. Immunohistochemically, the spindle cells were positive for HIF-1α (7/7 cases), MDM2 (4/7 cases), CDK4 (4/7 cases), p16 (7/7 cases), p53 (2/7 case), cyclin D1 (7/7 cases), and alpha-smooth muscle actin (6/7 cases). Neither MDM2 gene amplification nor USP6 gene split signal was detected in any case. Overexpression of the above proteins may be associated with the pathogenic mechanism of ischemic fasciitis. It is noted that the immunohistochemical positivity of MDM2, CDK4, and p16 do not necessarily indicate malignant neoplasm such as dedifferentiated liposarcoma.
Collapse
Affiliation(s)
- Yuichi Yamada
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Izumi Kinoshita
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Kenichi Kohashi
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Yuki Kuma
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Takamichi Ito
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Kenji Koda
- Department of Pathology, Fujieda Municipal General Hospital, 4-1-11 Surugadai, Fujieda-shi, Shizuoka-ken, 426-8677, Japan
| | - Atsushi Kisanuki
- Department of Pathology, Nichinan Prefectural Miyazaki Hospital, 1-9-5 Kiyama, Nichinan-shi, Miyazaki-ken, 887-0013, Japan
| | - Manabu Kurosawa
- Department of Pathology, Nagahama City Hospital, 313 Ooinui-cho, Nagahama-shi, Shiga-ken, 526-8580, Japan
| | - Michiko Yoshimura
- Department of Pathology, Belland Hospital, 500-3 Higashiyama, Naka-ku, Sakai-shi, Osaka, 599-8247, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka-shi, Fukuoka-ken, 812-8582, Japan.
| |
Collapse
|
61
|
Min-Wen JC, Yan-Jiang BC, Mishra S, Dai X, Magae J, Shyh-Chang N, Kumar AP, Sethi G. Molecular Targets of Ascochlorin and Its Derivatives for Cancer Therapy. STRESS AND INFLAMMATION IN DISORDERS 2017; 108:199-225. [DOI: 10.1016/bs.apcsb.2017.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
62
|
Wu Y, Meitzler JL, Antony S, Juhasz A, Lu J, Jiang G, Liu H, Hollingshead M, Haines DC, Butcher D, Panter MS, Roy K, Doroshow JH. Dual oxidase 2 and pancreatic adenocarcinoma: IFN-γ-mediated dual oxidase 2 overexpression results in H2O2-induced, ERK-associated up-regulation of HIF-1α and VEGF-A. Oncotarget 2016; 7:68412-68433. [PMID: 27637085 PMCID: PMC5340089 DOI: 10.18632/oncotarget.12032] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 09/01/2016] [Indexed: 12/18/2022] Open
Abstract
Several NADPH oxidase family members, including dual oxidase 2 [DUOX2], are expressed in human tumors, particularly gastrointestinal cancers associated with long-standing chronic inflammation. We found previously that exposure of pancreatic ductal adenocarcinoma cells to the pro-inflammatory cytokine IFN-γ increased DUOX2 expression (but not other NADPH oxidases) leading to long-lived H2O2 production. To elucidate the pathophysiology of DUOX2-mediated H2O2 formation in the pancreas further, we demonstrate here that IFN-γ-treated BxPC-3 and CFPAC-1 pancreatic cancer cells (known to increase DUOX2 expression) produce significant levels of intracellular oxidants and extracellular H2O2 which correlate with concomitant up-regulation of VEGF-A and HIF-1α transcription. These changes are not observed in the PANC-1 line that does not increase DUOX2 expression following IFN-γ treatment. DUOX2 knockdown with short interfering RNA significantly decreased IFN-γ-induced VEGF-A or HIF-1α up-regulation, as did treatment of pancreatic cancer cells with the NADPH oxidase inhibitor diphenylene iodonium, the multifunctional reduced thiol N-acetylcysteine, and the polyethylene glycol-modified form of the hydrogen peroxide detoxifying enzyme catalase. Increased DUOX2-related VEGF-A expression appears to result from reactive oxygen-mediated activation of ERK signaling that is responsible for AP-1-related transcriptional effects on the VEGF-A promoter. To clarify the relevance of these observations in vivo, we demonstrate that many human pre-malignant pancreatic intraepithelial neoplasms and frank pancreatic cancers express substantial levels of DUOX protein compared to histologically normal pancreatic tissues, and that expression of both DUOX2 and VEGF-A mRNAs is significantly increased in surgically-resected pancreatic cancers compared to the adjacent normal pancreas.
Collapse
Affiliation(s)
- Yongzhong Wu
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Smitha Antony
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Agnes Juhasz
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jiamo Lu
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Guojian Jiang
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Han Liu
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Melinda Hollingshead
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Diana C. Haines
- Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Michaela S. Panter
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Krishnendu Roy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - James H. Doroshow
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
63
|
Longo V, Brunetti O, Gnoni A, Cascinu S, Gasparini G, Lorusso V, Ribatti D, Silvestris N. Angiogenesis in pancreatic ductal adenocarcinoma: A controversial issue. Oncotarget 2016; 7:58649-58658. [PMID: 27462915 PMCID: PMC5295459 DOI: 10.18632/oncotarget.10765] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/13/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) occurs in the majority of cases with early loco-regional spread and distant metastases at diagnosis, leading to dismal prognosis with a 5-year overall survival rate moderately over than 5%. This malignancy is largely resistant to chemotherapy and radiation, but the reasons of the refractoriness to the therapies is still unknown. Evidence is accumulating to indicate that the PDAC microenvironment and vascularity strongly contribute to the clinical features of this disease. In particular, PDAC is characterized by excessive dense extracellular matrix deposition associated to vasculature collapse and hypoxia with low drug delivery, explaining at least partly the low efficacy of antiangiogenic drugs in this cancer. Strategies aimed to modulate tumor stroma favoring vasculature perfusion and chemotherapeutics delivery are under investigation.
Collapse
Affiliation(s)
- Vito Longo
- Department of Medical Oncology, Hospital of Taranto, Taranto, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Antonio Gnoni
- Department of Medical Oncology, Hospital "Vito Fazi" of Lecce, Lecce, Italy
| | | | | | - Vito Lorusso
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Cancer Institute "Giovanni Paolo II", Bari, Italy
| |
Collapse
|
64
|
Dükel M, Streitfeld WS, Tang TCC, Backman LRF, Ai L, May WS, Brown KD. The Breast Cancer Tumor Suppressor TRIM29 Is Expressed via ATM-dependent Signaling in Response to Hypoxia. J Biol Chem 2016; 291:21541-21552. [PMID: 27535224 DOI: 10.1074/jbc.m116.730960] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/17/2016] [Indexed: 02/01/2023] Open
Abstract
Reduced ATM function has been linked to breast cancer risk, and the TRIM29 protein is an emerging breast cancer tumor suppressor. Here we show that, in cultured breast tumor and non-tumorigenic mammary epithelial cells, TRIM29 is up-regulated in response to hypoxic stress but not DNA damage. Hypoxia-induced up-regulation of TRIM29 is dependent upon ATM and HIF1α and occurs through increased transcription of the TRIM29 gene. Basal expression of TRIM29 is also down-regulated in cells expressing diminished levels of ATM, and findings suggest that this occurs through basal NF-κB activity as knockdown of the NF-κB subunit RelA suppresses TRIM29 abundance. We have previously shown that the activity of the TWIST1 oncogene is antagonized by TRIM29 and now show that TRIM29 is necessary to block the up-regulation of TWIST1 that occurs in response to hypoxic stress. This study establishes TRIM29 as a hypoxia-induced tumor suppressor gene and provides a novel molecular mechanism for ATM-dependent breast cancer suppression.
Collapse
Affiliation(s)
- Muzaffer Dükel
- From the Departments of Biochemistry and Molecular Biology and
| | - W Scott Streitfeld
- Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | | | | | - Lingbao Ai
- From the Departments of Biochemistry and Molecular Biology and
| | - W Stratford May
- Medicine, University of Florida College of Medicine, Gainesville, Florida 32610
| | - Kevin D Brown
- From the Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
65
|
Park KJ, Yu MO, Park DH, Park JY, Chung YG, Kang SH. Role of vincristine in the inhibition of angiogenesis in glioblastoma. Neurol Res 2016; 38:871-9. [PMID: 27472259 DOI: 10.1080/01616412.2016.1211231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Vincristine, a microtubule-destabilizing drug, was found to exhibit anti-angiogenic effects and anti-tumoral activity. However, the precise mechanism by which vincristine inhibits angiogenesis in glioblastomas is not well understood. Our aim was to investigate whether vincristine affects vascular endothelial growth factor (VEGF) expression in glioblastoma cells and determine whether it is mediated by the downregulation of hypoxia-inducible factor-1α (HIF-1α). METHODS We investigated the expression of HIF-1α in glioblastoma tissues resected from patients and in human glioblastoma cell lines using immunohistochemistry, Western blot analysis, and immunocytochemistry. In addition to an MTT assay assessing the effect of vincristine on cell proliferation and viability, the effects of vincristine on VEGF mRNA expression and HIF-1α protein were examined using real-time RT-PCR and Western blot analysis under 1% O2 (hypoxia). RESULTS HIF-1α was expressed in the majority of glioblastoma tissues and was detected mainly in the nucleus. Strong immunoreactivity for HIF- 1 α was found often in the hypercellular zones. Under hypoxic conditions, HIF-1α protein levels in the glioblastoma cell lines increased, primarily localizing into the nucleus similar to glioblastoma tissues. Exposure of glioblastoma cells to vincristine resulted in enrichment of the G2-M fraction of the cell cycle, which suggests that vincristine-mediated growth inhibition of glioblastoma is correlated with mitotic inhibition. Using doses lower than those found to reduce the viability and proliferation of cells by 50% (IC50), vincristine decreased both the expression of VEGF mRNA and the level of HIF-1α protein in hypoxic glioblastoma cells. In addition, following exposure to vincristine, the expression of VEGF mRNA was correlated with HIF-1α protein levels. CONCLUSIONS Our results suggest that the mechanism by which vincristine elicits an anti-angiogenic effect in glioblastomas under hypoxic conditions might be mediated, in part, by HIF-1α inhibition.
Collapse
Affiliation(s)
- Kyung-Jae Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Mi Ok Yu
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Dong-Hyuk Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Jung-Yul Park
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Yong-Gu Chung
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| | - Shin-Hyuk Kang
- a Department of Neurosurgery, College of Medicine , Korea University Medical Center, Korea University , Seoul , Korea
| |
Collapse
|
66
|
Nielsen MFB, Mortensen MB, Detlefsen S. Key players in pancreatic cancer-stroma interaction: Cancer-associated fibroblasts, endothelial and inflammatory cells. World J Gastroenterol 2016; 22:2678-2700. [PMID: 26973408 PMCID: PMC4777992 DOI: 10.3748/wjg.v22.i9.2678] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/19/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) is the most aggressive type of common cancers, and in 2014, nearly 40000 patients died from the disease in the United States. Pancreatic ductal adenocarcinoma, which accounts for the majority of PC cases, is characterized by an intense stromal desmoplastic reaction surrounding the cancer cells. Cancer-associated fibroblasts (CAFs) are the main effector cells in the desmoplastic reaction, and pancreatic stellate cells are the most important source of CAFs. However, other important components of the PC stroma are inflammatory cells and endothelial cells. The aim of this review is to describe the complex interplay between PC cells and the cellular and non-cellular components of the tumour stroma. Published data have indicated that the desmoplastic stroma protects PC cells against chemotherapy and radiation therapy and that it might promote the proliferation and migration of PC cells. However, in animal studies, experimental depletion of the desmoplastic stroma and CAFs has led to more aggressive cancers. Hence, the precise role of the tumour stroma in PC remains to be elucidated. However, it is likely that a context-dependent therapeutic modification, rather than pure depletion, of the PC stroma holds potential for the development of new treatment strategies for PC patients.
Collapse
|
67
|
Metran-Nascente C, Yeung I, Vines DC, Metser U, Dhani NC, Green D, Milosevic M, Jaffray D, Hedley DW. Measurement of Tumor Hypoxia in Patients with Advanced Pancreatic Cancer Based on 18F-Fluoroazomyin Arabinoside Uptake. J Nucl Med 2016; 57:361-6. [PMID: 26769863 DOI: 10.2967/jnumed.115.167650] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/23/2015] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Pancreatic cancers are thought to be unusually hypoxic, which might sensitize them to drugs that are activated under hypoxic conditions. In order to develop this idea in the clinic, a minimally invasive technique for measuring the oxygenation status of pancreatic cancers is needed. METHODS We tested the potential for minimally invasive imaging of hypoxia in pancreatic cancer patients, using the 2-nitroimidazole PET tracer (18)F-fluoroazomycin arabinoside (or (18)F-1-α-D-[5-fluoro-5-deoxyarabinofuranosyl]-2-nitroimidazole [(18)F-FAZA]). Dynamic and static scans were obtained in 21 patients with either locally advanced or metastatic disease. The hypoxic fraction was determined in the 2-h static scans as the percentage of voxels with SUVs more than 3 SDs from the mean values obtained for skeletal muscle. RESULTS Hypoxia was detected in 15 of 20 evaluable patients, with the hypoxic fraction ranging from less than 5% to greater than 50%. Compartmental analysis of the dynamic scans allowed us to approximate the tumor perfusion as mL/min/g of tissue, a value that is independent of the extent of hypoxia derived from tracer uptake in the 2-h static scan. There was no significant correlation between tumor perfusion and hypoxia; nor did we see an association between tumor volume and hypoxia. CONCLUSION Although pancreatic cancers can be highly hypoxic, a substantial proportion appears to be well oxygenated. Therefore, we suggest that a minimally invasive technique such as the one described in this study be used for patient stratification in future clinical trials of hypoxia-targeting agents.
Collapse
Affiliation(s)
- Cristiane Metran-Nascente
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Ivan Yeung
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; and
| | - Douglass C Vines
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; and
| | - Ur Metser
- Department of Medical Imaging, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Neesha C Dhani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - David Green
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; and
| | - Michael Milosevic
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; and
| | - David Jaffray
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada; and
| | - David W Hedley
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
68
|
Dhani N, Fyles A, Hedley D, Milosevic M. The clinical significance of hypoxia in human cancers. Semin Nucl Med 2015; 45:110-21. [PMID: 25704384 DOI: 10.1053/j.semnuclmed.2014.11.002] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hypoxia is present to some extent in most solid malignant human cancers because of an imbalance between the limited oxygen delivery capacity of the abnormal vasculature and the high oxygen consumption of tumor cells. This drives a complex and dynamic compensatory response to enable continued cell survival, including genomic changes leading to selection of hypoxia-adapted cells with a propensity to invade locally, metastasize, and recur following surgery or radiotherapy. There is indisputable clinical evidence from numerous observational and therapeutic studies across a range of tumor types to implicate hypoxia as a key determinant of cancer behavior and treatment outcome. Despite this, hypoxia-targeted treatment has failed to influence clinical practice. This is explained, in part, by emerging findings to indicate that hypoxia is not equally important in all patients even when present to the same extent. The impact of hypoxia on patient outcome and the benefit of hypoxia-targeted treatments are greatest in situations where hypoxia is a primary biological driver of disease behavior-patients with tumors having a "hypoxic driver" phenotype. The challenge for the clinical and scientific communities moving forward is to develop robust precision cancer medicine strategies for identifying these patients in the setting of other etiologic, genomic, and host-tumor factors, considering not only the state of the tumor at diagnosis but also changing patient and tumor characteristics over time.
Collapse
Affiliation(s)
- Neesha Dhani
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Anthony Fyles
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - David Hedley
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Department of Medicine, University of Toronto, Toronto, Canada
| | - Michael Milosevic
- Department of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada; Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada.
| |
Collapse
|
69
|
He J, Ning C, Wang Y, Ma T, Huang H, Ge Y, Liu J, Jiang Y. Natural plant flavonoid apigenin directly disrupts Hsp90/Cdc37 complex and inhibits pancreatic cancer cell growth and migration. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.06.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
70
|
Archetti M. Heterogeneity and proliferation of invasive cancer subclones in game theory models of the Warburg effect. Cell Prolif 2015; 48:259-69. [PMID: 25643821 PMCID: PMC4964921 DOI: 10.1111/cpr.12169] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 10/06/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES The Warburg effect, a switch from aerobic energy production to anaerobic glycolysis, promotes tumour proliferation and motility by inducing acidification of the tumour microenvironment. Therapies that reduce acidity could impair tumour growth and invasiveness. I analysed the dynamics of cell proliferation and of resistance to therapies that target acidity, in a population of cells, under the Warburg effect. MATERIALS AND METHODS The dynamics of mutant cells with increased glycolysis and motility has been assessed in a multi-player game with collective interactions in the framework of evolutionary game theory. Perturbations of the level of acidity in the microenvironment have been used to simulate the effect of therapies that target glycolysis. RESULTS The non-linear effects of glycolysis induce frequency-dependent clonal selection leading to coexistence of glycolytic and non-glycolytic cells within a tumour. Mutants with increased motility can invade such a polymorphic population and spread within the tumour. While reducing acidity may produce a sudden reduction in tumour cell proliferation, frequency-dependent selection enables it to adapt to the new conditions and can enable the tumour to restore its original levels of growth and invasiveness. CONCLUSIONS The acidity produced by glycolysis acts as a non-linear public good that leads to coexistence of cells with high and low glycolysis within the tumour. Such a heterogeneous population can easily adapt to changes in acidity. Therapies that target acidity can only be effective in the long term if the cost of glycolysis is high, that is, under non-limiting oxygen concentrations. Their efficacy, therefore, is reduced when combined with therapies that impair angiogenesis.
Collapse
Affiliation(s)
- M. Archetti
- School of Biological SciencesUniversity of East AngliaNorwichNR4 7TJUK
| |
Collapse
|
71
|
Paraneoplastic Ma Antigen-Like 1 as a Potential Prognostic Biomarker in Human Pancreatic Ductal Adenocarcinoma. Pancreas 2015; 44:106-15. [PMID: 25251443 DOI: 10.1097/mpa.0000000000000220] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The present study aimed to identify novel useful clinical biomarker at early stages and to elucidate the molecular background of carcinogenesis in human pancreatic ductal adenocarcinomas (PDACs). METHODS Proteomes of dissected PDACs and adjacent nontumor pancreatic tissues from formalin-fixed and paraffin-embedded sections from 10 patients were analyzed using QSTAR Elite liquid chromatography-tandem mass spectrometry, ProteinPilot Software, and Ingenuity Pathway Analysis. Expression of potential biomarker candidates was validated immunohistochemically in 50 PDAC patients, followed by survival analyses and statistical comparison of protein expression with clinicopathologic variables. RESULTS Eight hundred five proteins displaying significant quantitative changes were identified in human PDACs by liquid chromatography-tandem mass spectrometry. Based on altered expression of downstream molecules, Ingenuity Pathway Analysis predicted up-regulation and/or activation of nuclear factor β-catenin, SOX11, enolase 1, NFE2L2, SP1, SMAD1, SMAD2, SMAD3, SMAD4, HIF-1, and others. From proteome analysis, paraneoplastic Ma antigen-like 1 was selected as a potential biomarker of human PDAC. Furthermore, paraneoplastic neuronal Ma antigen-like 1 immunohistochemical evaluation in 50 PDAC patients revealed that its positive expression was significantly associated with the better overall survival (log-rank test; P = 0.009) and histological differentiation of PDACs (well, moderate, and poor; P = 0.027) as compared with patients with negative expression. CONCLUSION Paraneoplastic Ma antigen-like 1 is suggested as a novel potential clinically useful prognostic biomarker for patients with PDAC.
Collapse
|
72
|
Zhen L, Shijie N, Shuijun Z. Tumor PHD2 expression is correlated with clinical features and prognosis of patients with HCC receiving liver resection. Medicine (Baltimore) 2014; 93:e179. [PMID: 25546659 PMCID: PMC4602600 DOI: 10.1097/md.0000000000000179] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The role of prolyl hydroxylase domain protein 2 (PHD2) in carcinogenesis has been studied in a variety of cancer types. However, the association between PHD2 and human hepatocellular carcinoma (HCC) has not been documented. A total of 220 patients with primary HCC who underwent a curative liver resection were enrolled in this study. The tumor samples were obtained during the surgical procedure from each patient for PHD2 immunohistological staining. All the patients were followed up and the disease-free survival (DFS) and overall survival (OS) were evaluated. We found that that high PHD2 expression was significantly associated with higher stage (stages III + IV) (odds ratio [OR] = 5.576, P < 0.001), larger tumor size (> 5 cm) (OR = 6.176, P < 0.001), poorer tumor differentiation (OR = 1.424, P = 0.003), and higher serum alpha fetoprotein (AFP) level (OR = 6.861, P < 0.001). Compared to those with high PHD2 expressions, patients with low PHD2 expression had significantly longer DFS and OS periods (both P < 0.001). Cox regression analyses revealed that higher levels of PHD2, tumor size, tumor stage, as well as serum AFP level were predictors for a worse prognosis in patients with HCC. PHD2 expression in the tumors is associated with the clinical features and prognosis of patients with HCC; it may be used as a histological marker for HCC.
Collapse
Affiliation(s)
- Li Zhen
- From the Department of Colorectal and Anal Surgery (LZ); Department of Vascular Surgery (NS); and Department of Hepatobiliary Surgery (ZS), The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | | | | |
Collapse
|
73
|
Villa JC, Chiu D, Brandes AH, Escorcia FE, Villa CH, Maguire WF, Hu CJ, de Stanchina E, Simon MC, Sisodia SS, Scheinberg DA, Li YM. Nontranscriptional role of Hif-1α in activation of γ-secretase and notch signaling in breast cancer. Cell Rep 2014; 8:1077-92. [PMID: 25131208 DOI: 10.1016/j.celrep.2014.07.028] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 03/04/2014] [Accepted: 07/16/2014] [Indexed: 12/11/2022] Open
Abstract
γ-Secretase is composed of four proteins that are obligatory for protease activity: presenilin, nicastrin, Aph1, and Pen-2. Despite the progress toward understanding the function of these individual subunits, there is no information available pertaining to the modulation of γ-secretase in response to environmental changes in cells. Here, we show that hypoxia upregulates γ-secretase activity through a direct interaction with Hif-1α, revealing an unconventional function for Hif-1α as an enzyme subunit, which is distinct from its canonical role as a transcription factor. Moreover, hypoxia-induced cell invasion and metastasis are alleviated by either γ-secretase inhibitors or a dominant-negative Notch coactivator, indicating that γ-secretase/Notch signaling plays an essential role in controlling these cellular processes. The present study reveals a mechanism in which γ-secretase can achieve temporal control through conditional interactions with regulatory proteins, such as Hif-1α, under select physiological and pathological conditions.
Collapse
Affiliation(s)
- Jennifer C Villa
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Danica Chiu
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Alissa H Brandes
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Freddy E Escorcia
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Carlos H Villa
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - William F Maguire
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Cheng-Jun Hu
- Molecular Biology Graduate Program, School of Dental Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Elisa de Stanchina
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Sangram S Sisodia
- The Center for Molecular Neurobiology, The University of Chicago, Chicago, IL 60637, USA
| | - David A Scheinberg
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
74
|
Novel thalidomide analogs: Anti-angiogenic and apoptotic effects on Hep-G2 and MCF-7 cancer cell lines. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.biomag.2014.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
75
|
Dong X, Sun B, Zhao X, Liu Z, Gu Q, Zhang D, Zhao N, Wang J, Chi J. Expression of relative-protein of hypoxia-inducible factor-1α in vasculogenesis of mouse embryo. ACTA ACUST UNITED AC 2014; 21:4. [PMID: 25984487 PMCID: PMC4376343 DOI: 10.1186/2241-5793-21-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Accepted: 01/20/2014] [Indexed: 01/12/2023]
Abstract
Background Physiological vasculogenesis in embryonic tissues share some important features with pathological neoangiogenesis in tumors. Linearly Patterned Programmed Cell Necrosis (LPPCN) and Vasculogenic Mimicry (VM) have been reported in tumors. The term VM refers to the aggressive tumor cells with CD31-negative phenotype to form Periodic Αcid Schiff (PAS)-positive network, that mimics the pattern of embryonic vasculogenic networks. LPPCN had been observed in our laboratory, and served as a spatial infrastructure for VM and endothelium-dependent vessel formation. Studies have been shown that hypoxia-inducible factor-1α (HIF-1α) can induce tumor cells to form vessel-like tubes and express genes associated with VM. Therefore, an analogous investigation has been carried out to determine if these patterns existed in mouse embryonic vasculogenesis. Results In this essay, the results demonstrated that the number of Linearly Patterned Cell Αpoptosis (LPCA), embryo Vasculogenic Μimicry (embryo VM), endothelium-dependent vessels, and relative-protein of HIF-1α expression all showed time-dependent tendencies on E5.5-E9.5 (p < 0.05). The proteins CD133, VEGF, Twist, E-cadherin, and Vimentin showed local plexus distribution on E6.5-E7.5 (p < 0.05). Conclusions LPCA and embryo VM existed in embryonic vasculogenesis. The relative protein of HIF-1α regulated the mouse embryonic vasculogenesis.
Collapse
Affiliation(s)
- Xueyi Dong
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Baocun Sun
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Xiulan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Zhiyong Liu
- Department of Pathology, Tianjin Cancer Hospital, Tianjin Medical University, Tianjin, 300060 China
| | - Qiang Gu
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Danfang Zhang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Nan Zhao
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Jinjing Wang
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China ; Department of Pathology, Tianjin General Hospital, Tianjin Medical University, Tianjin, 300052 China
| | - Jiadong Chi
- Department of Pathology, Tianjin Medical University, Tianjin, 300070 China
| |
Collapse
|
76
|
A Novel Mechanism for Cross-Adaptation between Heat and Altitude Acclimation: The Role of Heat Shock Protein 90. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/121402] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Heat shock protein 90 (HSP90) is a member of a family of molecular chaperone proteins which can be upregulated by various stressors including heat stress leading to increases in HSP90 protein expression. Its primary functions include (1) renaturing and denaturing of damaged proteins caused by heat stress and (2) interacting with client proteins to induce cell signaling for gene expression. The latter function is of interest because, in cancer cells, HSP90 has been reported to interact with the transcription hypoxic-inducible factor 1α (HIF1α). In a normoxic environment, HIF1α is degraded and therefore has limited physiological function. In contrast, in a hypoxic environment, stabilized HIF1α acts to promote erythropoiesis and angiogenesis. Since HSP90 interacts with HIF1α, and HSP90 can be upregulated from heat acclimation in humans, we present a proposal that heat acclimation can mimic molecular adaptations to those of altitude exposure. Specifically, we propose that heat acclimation increases HSP90 which then stabilizes HIF1α in a normoxic environment. This has many implications since HIF1α regulates red blood cell and vasculature formation. In this paper we will discuss (1) the functional roles of HSP90 and HIF1α, (2) the interaction between HSP90 and other client proteins including HIF1α, and (3) results from in vitro studies that may suggest how the relationship between HSP90 and HIF1α might be applied to individuals preparing to make altitude sojourns.
Collapse
|
77
|
Sibenaller ZA, Welsh JL, Du C, Witmer JR, Schrock HE, Du J, Buettner GR, Goswami PC, Cieslak JA, Cullen JJ. Extracellular superoxide dismutase suppresses hypoxia-inducible factor-1α in pancreatic cancer. Free Radic Biol Med 2014; 69:357-66. [PMID: 24509158 PMCID: PMC3981470 DOI: 10.1016/j.freeradbiomed.2014.02.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 01/31/2014] [Accepted: 02/03/2014] [Indexed: 12/31/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription factor that governs cellular responses to reduced oxygen availability by mediating crucial homeostatic processes and is a major survival determinant for tumor cells growing in a low-oxygen environment. Clinically, HIF-1α seems to be important in pancreatic cancer, as HIF-1α correlates with metastatic status of the tumor. Extracellular superoxide dismutase (EcSOD) inhibits pancreatic cancer cell growth by scavenging nonmitochondrial superoxide. We hypothesized that EcSOD overexpression leads to changes in the O2(-)/H2O2 balance modulating the redox status affecting signal transduction pathways. Both transient and stable overexpression of EcSOD suppressed the hypoxic accumulation of HIF-1α in human pancreatic cancer cells. This suppression of HIF-1α had a strong inverse correlation with levels of EcSOD protein. Coexpression of the hydrogen peroxide-removing protein glutathione peroxidase did not prevent the EcSOD-induced suppression of HIF-1α, suggesting that the degradation of HIF-1α observed with high EcSOD overexpression is possibly due to a low steady-state level of superoxide. Hypoxic induction of vascular endothelial growth factor (VEGF) was also suppressed with increased EcSOD. Intratumoral injections of an adenoviral vector containing the EcSOD gene into preestablished pancreatic tumors suppressed both VEGF levels and tumor growth. These results demonstrate that the transcription factor HIF-1α and its important gene target VEGF can be modulated by the antioxidant enzyme EcSOD.
Collapse
Affiliation(s)
- Zita A Sibenaller
- Department of Surgery, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Jessemae L Welsh
- Department of Surgery, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Changbin Du
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Jordan R Witmer
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Hannah E Schrock
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Juan Du
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Garry R Buettner
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA
| | - Prabhat C Goswami
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA
| | - John A Cieslak
- Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Joseph J Cullen
- Department of Surgery, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Department of Radiation Oncology, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Iowa City, IA 52242, USA; Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
78
|
Archetti M. Evolutionary dynamics of the Warburg effect: Glycolysis as a collective action problem among cancer cells. J Theor Biol 2014; 341:1-8. [DOI: 10.1016/j.jtbi.2013.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Revised: 09/07/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
|
79
|
Camaj P, Jäckel C, Krebs S, De Toni EN, Blum H, Jauch KW, Nelson PJ, Bruns CJ. Hypoxia-independent gene expression mediated by SOX9 promotes aggressive pancreatic tumor biology. Mol Cancer Res 2013; 12:421-32. [PMID: 24302456 DOI: 10.1158/1541-7786.mcr-13-0351] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Pancreatic cancer aggressiveness is characterized by its high capacity for local invasion, ability to promote angiogenesis, and potential to metastasize. Hypoxia is known to represent a crucial step in the development of aggressive malignant features of many human cancers. However, micrometastatic tumors are not typically subjected to hypoxic events during early stages of dissemination; therefore, it is unclear how these tumors are able to maintain their aggressive phenotype. Thus, the identification of regulators of hypoxia-related genes in aggressive/metastatic tumors represents a fundamental step for the design of future therapies to treat pancreatic cancer. To this end, transcriptomic profiles were compared between the nonmetastatic pancreatic cancer cell line FG (LMET) and its angiogenic/metastatic derivate L3.6pl (HMET) under normoxic or hypoxic conditions. Cluster analysis revealed a number of transcripts that were induced by hypoxia in nonmetastatic cancer cells. Strikingly, this cluster was determined to be constitutively activated under normoxia in the metastatic cancer cells and could not be further induced by hypoxia. A subset of these transcripts were regulated by the transcription factor SOX9 in the aggressive-metastatic cells, but driven by hypoxia-inducible factor-1α (HIF-1α) in the parental nonmetastatic cell line. Moreover, these transcripts were enriched in cancer-related networks including: WNT, CXCR4, retinoic acid, and (FAK) focal adhesion kinase, gene PTK2 signaling pathways. In functional assays, inhibition of SOX9 expression in HMET cells led to increased apoptosis and reduced migration in vitro and a significant reduction in primary tumor growth, angiogenesis, and metastasis following orthotopic tumor cell injection. At the molecular level, the control of SOX9 expression was associated with changes in the methylation status of the SOX9 promoter. Finally, SOX9 upregulation was verified in a series of tumor specimens of patients with pancreatic carcinoma. IMPLICATIONS SOX9 represents a novel target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Peter Camaj
- Klinikum Großhadern, VH02 426, Marchioninistr. 15, 81377 München, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Chen H, Guan Y, Yuan G, Zhang Q, Jing N. A perylene derivative regulates HIF-1α and Stat3 signaling pathways. Bioorg Med Chem 2013; 22:1496-505. [PMID: 24485121 DOI: 10.1016/j.bmc.2013.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/07/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
Abstract
It is becoming increasingly evident that improving the cure rate of many cancers will require treatment regimens hit more than one validated tumor targets. Developing an anti-cancer agent that targets two oncoproteins simultaneously is a promising strategy for accomplishing this goal. It would be expected to promote drug efficacy, reduce therapy-resistant without introducing additional toxic side effects. HIF-1α is a key regulator of the cellular response to hypoxia and is involved in tumor angiogenesis and cancer cell survival, glucose metabolism, and invasion. Stat3 has several oncogenic functions, including suppression of anti-tumor immune responses and promotion of inflammation. Recently, we have identified the perylene derivative, TEL03, as a dual inhibitor that targets both HIF-1α and Stat3. TEL03 blocks the expression of both HIF-1α and Stat3, regulated oncogenes (e.g., Bcl-2, VEGF, Glut1, and others) in cancer cells, and induces cancer cell apoptosis. The results demonstrated that: (i) TEL03 blocks Stat3 phosphorylation, and inhibits Stat3 transcriptional activity; and (ii) interferes the binding of HIF-1α to p300/CBP inducing its degradation by proteasomes under hypoxic conditions. Our in vivo tests showed that as a dual inhibitor, TEL03 dramatically inhibited tumor growth, and provided the evidence that targeting both HIF-1α and Stat3 simultaneously could be a promising strategy for breast and pancreatic cancer therapies.
Collapse
Affiliation(s)
- Han Chen
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Yongli Guan
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gu Yuan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Qiang Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Naijie Jing
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
81
|
Fifis T, Nguyen L, Malcontenti-Wilson C, Chan LS, Nunes Costa PL, Daruwalla J, Nikfarjam M, Muralidharan V, Waltham M, Thompson EW, Christophi C. Treatment with the vascular disruptive agent OXi4503 induces an immediate and widespread epithelial to mesenchymal transition in the surviving tumor. Cancer Med 2013; 2:595-610. [PMID: 24403226 PMCID: PMC3892792 DOI: 10.1002/cam4.109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/17/2013] [Accepted: 06/19/2013] [Indexed: 12/14/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is considered an important mechanism in tumor resistance to drug treatments; however, in vivo observation of this process has been limited. In this study we demonstrated an immediate and widespread EMT involving all surviving tumor cells following treatment of a mouse model of colorectal liver metastases with the vascular disruptive agent OXi4503. EMT was characterized by significant downregulation of E-cadherin, relocation and nuclear accumulation of β-catenin as well as significant upregulation of ZEB1 and vimentin. Concomitantly, significant temporal upregulation in hypoxia and the pro-angiogenic growth factors hypoxia-inducible factor 1-alpha, hepatocyte growth factor, vascular endothelial growth factor and transforming growth factor-beta were seen within the surviving tumor. The process of EMT was transient and by 5 days after treatment tumor cell reversion to epithelial morphology was evident. This reversal, termed mesenchymal to epithelial transition (MET) is a process implicated in the development of new metastases but has not been observed in vivo histologically. Similar EMT changes were observed in response to other antitumor treatments including chemotherapy, thermal ablation, and antiangiogenic treatments in our mouse colorectal metastasis model and in a murine orthotopic breast cancer model after OXi4503 treatment. These results suggest that EMT may be an early mechanism adopted by tumors in response to injury and hypoxic stress, such that inhibition of EMT in combination with other therapies could play a significant role in future cancer therapy.
Collapse
Affiliation(s)
- Theodora Fifis
- Department of Surgery, University of Melbourne, Austin Health, Heidelberg, Victoria, 3084, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Obacz J, Pastorekova S, Vojtesek B, Hrstka R. Cross-talk between HIF and p53 as mediators of molecular responses to physiological and genotoxic stresses. Mol Cancer 2013; 12:93. [PMID: 23945296 PMCID: PMC3844392 DOI: 10.1186/1476-4598-12-93] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/10/2013] [Indexed: 01/06/2023] Open
Abstract
Abnormal rates of growth together with metastatic potential and lack of susceptibility to cellular signals leading to apoptosis are widely investigated characteristics of tumors that develop via genetic or epigenetic mechanisms. Moreover, in the growing tumor, cells are exposed to insufficient nutrient supply, low oxygen availability (hypoxia) and/or reactive oxygen species. These physiological stresses force them to switch into more adaptable and aggressive phenotypes. This paper summarizes the role of two key mediators of cellular stress responses, namely p53 and HIF, which significantly affect cancer progression and compromise treatment outcomes. Furthermore, it describes cross-talk between these factors.
Collapse
Affiliation(s)
- Joanna Obacz
- Masaryk Memorial Cancer Institute, Regional Centre for Applied Molecular Oncology, Zluty kopec 7, 65653 Brno, Czech Republic.
| | | | | | | |
Collapse
|
83
|
Ma J, Zhang Q, Chen S, Fang B, Yang Q, Chen C, Miele L, Sarkar FH, Xia J, Wang Z. Mitochondrial dysfunction promotes breast cancer cell migration and invasion through HIF1α accumulation via increased production of reactive oxygen species. PLoS One 2013; 8:e69485. [PMID: 23922721 PMCID: PMC3726697 DOI: 10.1371/journal.pone.0069485] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 06/10/2013] [Indexed: 01/12/2023] Open
Abstract
Although mitochondrial dysfunction has been observed in various types of human cancer cells, the molecular mechanism underlying mitochondrial dysfunction mediated tumorigenesis remains largely elusive. To further explore the function of mitochondria and their involvement in the pathogenic mechanisms of cancer development, mitochondrial dysfunction clones of breast cancer cells were generated by rotenone treatment, a specific inhibitor of mitochondrial electron transport complex I. These clones were verified by mitochondrial respiratory defect measurement. Moreover, those clones exhibited increased reactive oxygen species (ROS), and showed higher migration and invasive behaviors compared with their parental cells. Furthermore, antioxidant N-acetyl cysteine, PEG-catalase, and mito-TEMPO effectively inhibited cell migration and invasion in these clones. Notably, ROS regulated malignant cellular behavior was in part mediated through upregulation of hypoxia-inducible factor-1 α and vascular endothelial growth factor. Our results suggest that mitochondrial dysfunction promotes cancer cell motility partly through HIF1α accumulation mediated via increased production of reactive oxygen species.
Collapse
Affiliation(s)
- Jia Ma
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China
| | - Qing Zhang
- Department of Orthopedics, The Center Hospital of Bengbu, Anhui, China
| | - Sulian Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China
| | - Binbin Fang
- Research Center of Clinical Laboratory Science, Bengbu Medical College, Bengbu, Anhui, China
| | - Qingling Yang
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China
| | - Changjie Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China
| | - Lucio Miele
- University of Mississippi Cancer Institute, Jackson, Mississippi, United States of America
| | - Fazlul H. Sarkar
- Department of Pathology and Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Jun Xia
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui, China
- * E-mail: (ZW); (JX)
| | - Zhiwei Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, United States of America
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- * E-mail: (ZW); (JX)
| |
Collapse
|
84
|
Spivak-Kroizman TR, Hostetter G, Posner R, Aziz M, Hu C, Demeure MJ, Von Hoff D, Hingorani SR, Palculict TB, Izzo J, Kiriakova GM, Abdelmelek M, Bartholomeusz G, James BP, Powis G. Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res 2013; 73:3235-47. [PMID: 23633488 DOI: 10.1158/0008-5472.can-11-1433] [Citation(s) in RCA: 158] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pancreatic cancer is characterized by a desmoplastic reaction that creates a dense fibroinflammatory microenvironment, promoting hypoxia and limiting cancer drug delivery due to decreased blood perfusion. Here, we describe a novel tumor-stroma interaction that may help explain the prevalence of desmoplasia in this cancer. Specifically, we found that activation of hypoxia-inducible factor-1α (HIF-1α) by tumor hypoxia strongly activates secretion of the sonic hedgehog (SHH) ligand by cancer cells, which in turn causes stromal fibroblasts to increase fibrous tissue deposition. In support of this finding, elevated levels of HIF-1α and SHH in pancreatic tumors were determined to be markers of decreased patient survival. Repeated cycles of hypoxia and desmoplasia amplified each other in a feed forward loop that made tumors more aggressive and resistant to therapy. This loop could be blocked by HIF-1α inhibition, which was sufficient to block SHH production and hedgehog signaling. Taken together, our findings suggest that increased HIF-1α produced by hypoxic tumors triggers the desmoplasic reaction in pancreatic cancer, which is then amplified by a feed forward loop involving cycles of decreased blood flow and increased hypoxia. Our findings strengthen the rationale for testing HIF inhibitors and may therefore represent a novel therapeutic option for pancreatic cancer.
Collapse
|
85
|
Lu XX, Chen YT, Feng B, Mao XB, Yu B, Chu XY. Expression and clinical significance of CD73 and hypoxia-inducible factor-1α in gastric carcinoma. World J Gastroenterol 2013; 19:1912-1918. [PMID: 23569336 PMCID: PMC3613106 DOI: 10.3748/wjg.v19.i12.1912] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 09/28/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of CD73 and hypoxia-inducible factor-1α (HIF-1α) in human gastric carcinoma, and explore their clinical significance and prognostic value.
METHODS: CD73 and HIF-1α expressions were detected by immunohistochemistry in consecutive sections of tissue samples from 68 gastric carcinoma patients. The peritumor tissues 2 cm away from the tumor were obtained and served as controls. The presence of CD73 and HIF-1α was analyzed by immunohistochemistry using the Envision technique.
RESULTS: CD73 and HIF-1α expressions in gastric carcinoma were significantly higher than those in gastric mucosal tissues as control (P < 0.001) and showed a close correlation (Spearman r = 0.390, P = 0.001). Overexpression of CD73 was positively correlated with differentiation of tumor (P = 0.000), histopathology (P = 0.041), depth of invasion (P < 0.001), nodal status (P = 0.003), metastasis (P = 0.013), and the American Joint Committee on Cancer (AJCC) stage (P < 0.001). High expression of HIF-1α was positively correlated with tumor diameter (P = 0.031), depth of invasion (P = 0.022), and AJCC stage (P = 0.035). The overall survival rate was low in the patients with high expression of CD73 (P < 0.001). Moreover, CD73+/HIF-1α+ patients had the worst prognosis (P < 0.001). CD73 expression was proven to be an independent predictor for patients with gastric carcinoma by both multivariate Cox regression analysis (P = 0.021) and receiver operating characteristic curves (P = 0.001).
CONCLUSION: CD73 expression correlates closely with HIF-1α expression in gastric carcinoma. CD73 could be an independent prognostic indicator for gastric carcinoma.
Collapse
|
86
|
Iovanna JL, Marks DL, Fernandez-Zapico ME, Urrutia R. Mechanistic insights into self-reinforcing processes driving abnormal histogenesis during the development of pancreatic cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1078-86. [PMID: 23375449 DOI: 10.1016/j.ajpath.2012.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 12/16/2012] [Accepted: 12/24/2012] [Indexed: 12/28/2022]
Abstract
Pancreatic ductal adenocarcinoma, one of the most feared lethal and painful diseases, is increasing in incidence. The poor prognosis of pancreatic ductal adenocarcinoma-affected patients primarily is owing to our inability to develop effective therapies. Mechanistic studies of genetic, epigenetic, and cell-to-cell signaling events are providing clues to molecular pathways that can be targeted in an attempt to cure this disease. The current review article seeks to draw inferences from available mechanistic knowledge to build a theoretical framework that can facilitate these approaches. This conceptual model considers pancreatic cancer as a tissue disease rather than an isolated epithelial cell problem, which develops and progresses in large part as a result of three positive feedback loops: i) genetic and epigenetic changes in epithelial cells modulate their interaction with mesenchymal cells to generate a dynamically changing process of abnormal histogenesis, which drives more changes; ii) the faulty tissue architecture of neoplastic lesions results in unsynchronized secretion of signaling molecules by cells, which generates an environment that is poor in oxygen and nutrients; and iii) the increased metabolic needs of rapidly dividing cells serve as an evolutionary pressure for them to adapt to this adverse microenvironment, leading to the emergence of resistant clones. We discuss how these concepts can guide mechanistic studies, as well as aid in the design of novel experimental therapeutics.
Collapse
Affiliation(s)
- Juan L Iovanna
- Cancer Research Center of Marseille, Inserm U1068, CNRS, UMR7258, Institute Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | | | | | | |
Collapse
|
87
|
Provenzano PP, Hingorani SR. Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer. Br J Cancer 2013; 108:1-8. [PMID: 23299539 PMCID: PMC3553539 DOI: 10.1038/bjc.2012.569] [Citation(s) in RCA: 240] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinomas (PDAs) are notoriously aggressive and resistant to treatment. They distinguish themselves further by their robust fibroinflammatory, or desmoplastic, stromal reaction and degree of hypovascularity. Recent findings have revealed multiple mechanisms of stromal complicity in disease pathogenesis and resistance. In this review, we focus on altered physicomechanics as one mechanism of what we term as ‘stromal resistance’ in PDA. Extremely high interstitial fluid pressures and a dense extracellular matrix combine to limit the delivery and distribution of therapeutic agents. We discuss the genesis and consequences of altered fluid dynamics in PDA and strategies to restore them.
Collapse
Affiliation(s)
- P P Provenzano
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | |
Collapse
|
88
|
Hata H, Tatemichi M, Nakadate T. Involvement of annexin A8 in the properties of pancreatic cancer. Mol Carcinog 2012; 53:181-91. [PMID: 23001853 DOI: 10.1002/mc.21961] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 08/09/2012] [Accepted: 08/23/2012] [Indexed: 11/10/2022]
Abstract
Although Annexin A8 (ANXA8), a member of a superfamily of calcium and phospholipid binding proteins, is physiologically expressed in a tissue-specific manner, recent microarray studies reported that ANXA8 was also ectopically expressed in pancreatic cancers. We investigated the molecular mechanism of expression of ANXA8 in cancer cells and its functional role in pancreatic cancer cells. ANXA8 was diversely expressed in human cancer cell lines. Expression was enhanced by treatment with 5-aza-dC and butyrate, and correlated with methylation status at CpG in the promoter-exon 1 region. Inhibition of ANXA8 using siRNA in BxPC-3 cells which express ANXA8 at a high level elevated caspase-3 and -7 activities. In in vitro invasion assay, inhibition of ANXA8 using siRNA in BxPC-3 reduced the numbers of migrating cells, and down-regulated HIF-1α mRNA transcription. Overexpression of ANXA8 increased the number of viable cells and BrdU incorporation in PANC-1 cells, which express ANXA8 at a low level. Expression of ANXA8 was induced under conditions of nutrient deprivation, and overexpression of ANXA8 showed resistance against serum starvation in PANC-1 cells. In a promoter assay, co-transfection with the expression vector of ANXA8 and the vector of a reporter gene containing the promoter of HIF-1α enhanced HIF-1α promoter activity. In contrast, this effect of ANXA8 was inhibited by administration of BAPTA-AM, an intracellular Ca²⁺ chelator. These results suggest that ectopic ANXA8 expression in cancer cells might involve an epigenetic mechanism. ANXA8 might play an important role in calcium fluctuation-mediated HIF-1α transcriptional activation and cell viability.
Collapse
Affiliation(s)
- Harumi Hata
- Department of Hygiene and Preventive Medicine, Showa University, School of Medicine, Tokyo, Japan
| | | | | |
Collapse
|
89
|
Development of a combination drug-eluting bead: towards enhanced efficacy for locoregional tumour therapies. Anticancer Drugs 2012; 23:355-69. [PMID: 22241169 DOI: 10.1097/cad.0b013e32835006d2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Drug-eluting beads (DEBs) are becoming a mainstay locoregional therapy for hepatic malignancies but are currently loaded with single drugs alone. Here, we wished to prepare DEB containing different drug combinations, to screen their efficacy using an in-vitro cell culture assay and to include any promising combinations that demonstrate additive efficacy in an in-vivo model of locoregional tumour treatment. A modified in-vitro assay was used based upon the use of 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) with either HepG2 liver cancer or PSN1 pancreatic cancer cell lines. The comparative cytotoxicity of DEB combinations prepared containing doxorubicin, irinotecan, topotecan and rapamycin was evaluated. Those combinations that demonstrated an additive cytotoxicity effect were investigated in vivo using a nude mouse xenograft model of pancreatic cancer. Although many of the DEB combinations showed either no effect or a slight antagonistic effect, the combination of doxorubicin and rapamycin DEBs demonstrated synergistic activity. On the basis of these findings, a method was developed to prepare a doxorubicin/rapamycin dual-loaded DEB, which was shown to possess the same drug-loading capacities, drug elution properties and HepG2 cell cytotoxicity synergy as the single drug-loaded DEB combination. Evaluation of this dual-loaded combination DEB versus the respective single drug-loaded DEBs in a mouse xenograft model of pancreatic cancer showed an equivalent tumour volume reduction as the doxorubicin DEB, but with less toxicity than the rapamycin DEB. The doxorubicin/rapamycin combination DEB offers great potential for enhanced efficacy in the locoregional treatment of malignant tumours.
Collapse
|
90
|
Yang S, Zhao Z, Wu R, Lu H, Zhang X, Huan C, Wang C, Wu X, Guan G. Expression and biological relationship of vascular endothelial growth factor-A and matrix metalloproteinase-9 in gastric carcinoma. J Int Med Res 2012; 39:2076-85. [PMID: 22289522 DOI: 10.1177/147323001103900603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Protein expression of vascular endothelial growth factor A (VEGF-A) and matrix metalloproteinase 9 (MMP-9) was studied in gastric carcinoma patients in relation to clinicopathological characteristics and prognosis. Fifty-four samples of gastric carcinoma tissue and 15 samples of adjacent normal gastric mucosal tissue were examined immunohistochemically. Expression rates of VEGF-A (66.7%) and MMP-9 (63.0%) in carcinoma tissue were significantly higher than in normal tissue (6.7% for both proteins). VEGF-A and MMP9 expression was associated with tumour size, invasion depth, lymph node metastasis, degree of histological differentiation and pathological stage, but not age or sex. VEGF-A expression was positively correlated with that of MMP-9. Expression of VEGF-A and MMP-9 were each inversely correlated with 5-year survival. VEGF-A and MMP-9 were overexpressed in tumours compared with normal tissue; they may act together to increase carcinogenesis and the progression, invasion and metastasis of gastric carcinoma, and could be used as biomarkers for the prognosis of gastric carcinoma.
Collapse
Affiliation(s)
- S Yang
- Department of Oncology, The Union Hospital of Fujian Medical University, Fuzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Isobe T, Aoyagi K, Koufuji K, Shirouzu K, Kawahara A, Taira T, Kage M. Clinicopathological significance of hypoxia-inducible factor-1 alpha (HIF-1α) expression in gastric cancer. Int J Clin Oncol 2012; 18:293-304. [PMID: 22350022 DOI: 10.1007/s10147-012-0378-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 01/09/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hypoxia is a common feature of rapidly growing solid tumors. Therefore, cellular adaptation to hypoxia and altered glucose metabolism are fundamental to the biology of cancer cells. Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor for more than 60 genes recognized to control the delivery of oxygen and nutrients through the induction of angiogenesis and glycolysis under hypoxic conditions. Therefore, inhibition of the expression of HIF-1α can be expected to be potentially tumor-specific molecular target-based therapy. In this study, we evaluated the significance of HIF-1α expression in relationship to clinicopathological factors, prognosis, vascular endothelial growth factor (VEGF) expression, and microvessel density (MVD). METHODS Paraffin-embedded tumor specimens from 128 patients who underwent gastrectomy at Kurume University from 2004 to 2005 were used to assess the clinical significance of HIF-1α expression. We used the ABC method to perform an immunohistochemical analysis of the HIF-1α and VEGF expression. RESULTS Eighty-four (65.6%) of gastric cancer specimens were positive for HIF-1α expression. Multivariate analysis showed that histology, depth of invasion, VEGF expression, and MVD were significantly associated with HIF-1α expression. On relapse-free and overall survival curves, the HIF-1α-negative group was significantly higher than the HIF-1α-positive group. Moreover, HIF-1α(+)/VEGF(+) patients had the worst prognosis. HIF-1α expression was identified as a significant predictor of relapse-free survival and overall survival by multivariate Cox's proportional hazard analyses. CONCLUSION Overexpression of HIF-1α was found to be an indicator of poor prognosis for patients with gastric cancer and was significantly correlated with histology, depth of invasion, VEGF, and MVD.
Collapse
Affiliation(s)
- Taro Isobe
- Department of Surgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | | | | | | | | | | | | |
Collapse
|
92
|
Wang Z, Shi Q, Wang Z, Gu Y, Shen Y, Sun M, Deng M, Zhang H, Fang J, Zhang S, Xie F. Clinicopathologic correlation of cancer stem cell markers CD44, CD24, VEGF and HIF-1α in ductal carcinoma in situ and invasive ductal carcinoma of breast: an immunohistochemistry-based pilot study. Pathol Res Pract 2011; 207:505-13. [PMID: 21802218 DOI: 10.1016/j.prp.2011.06.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 05/30/2011] [Accepted: 06/15/2011] [Indexed: 10/17/2022]
Abstract
CD24(-/low)CD44(+) cells have been identified as putative cancer stem cells (CSCs) in breast cancer. However, the expression of these markers, as well as their association with clinical parameters or tumor microenvironment of breast cancer, remains largely unknown. In the present study, we examined the expression of CD44, CD24, VEGF, and HIF-1α in human breast tumor tissues and assessed their clinicopathological correlations. We investigated tissue samples, including 117 cases of invasive ductal carcinoma (IDCa), 14 cases of ductal carcinoma in situ (DCIS), and 15 cases of intraductal hyperplasia (IDH) from breast tissues. The expression of CD44, CD24, HIF-1α, and VEGF was evaluated using immunohistochemical staining. CD24, CD44, HIF-1α, and VEGF were expressed in 49 (41.9%), 51 (43.6%), 32 (27.4%), and 97 cases (82.9%), respectively, in IDCa. CD24(-/low)CD44(+) cells were noted in 48 (41.3%) cases. The levels of CD24 and VEGF expression correlated positively with tumor malignancy (P<0.05). Meanwhile, the expression of CD24, CD44, and VEGF correlated significantly positively with increasing tumor grade (P<0.05). In addition, associations between CD44 and VEGF, CD24 and VEGF, HIF-1α and VEGF, CD24(-/low)CD44(+) and VEGF, CD24(-/low)CD44(+) and HIF-1α were also observed (P<0.05). The HIF-1α expression level was relatively higher in early stage breast cancer patients with CD24(-/low)CD44(+) cells. Taken together, our results suggest that CD24 and VEGF may play important roles in breast tumorigenesis and progression, while HIF-1α may play a role in the early stage of breast carcinogenesis.
Collapse
Affiliation(s)
- Zhen Wang
- The Ultrasonagraphy Center of Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215004, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Su Y, Loos M, Giese N, Metzen E, Büchler MW, Friess H, Kornberg A, Büchler P. Prolyl hydroxylase-2 (PHD2) exerts tumor-suppressive activity in pancreatic cancer. Cancer 2011; 118:960-72. [PMID: 21792862 DOI: 10.1002/cncr.26344] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Revised: 05/10/2011] [Accepted: 05/16/2011] [Indexed: 01/31/2023]
Abstract
BACKGROUND Pancreatic cancer is 1 of the most common and poorly treated tumors. In search of new therapeutic approaches, the oxygen sensors prolyl hydroxylases (PHD) are potential targets. PHD2 is considered the key oxygen sensor-regulating hypoxia-inducible factor (HIF). Currently, there is conflicting evidence regarding the exact role of PHD2 in tumorigenesis. The objective of this study was to investigate the role of PHD2 in pancreatic cancer growth and progression. METHODS PHD2 expression was analyzed by quantitative real-time polymerase chain reaction analysis and immunohistochemistry in human tissue specimens and cell lines. Knockdown of PHD2 was done by using short-interfering RNAs (siRNAs) specific against PHD2, and PHD2 overexpression was achieved by stable combinational DNA transfection. In vivo, an orthotopic murine model was used. Angiogenic cytokines were assessed with enzyme-linked immunosorbent assays, and invasion was studied with Matrigel assays. RESULTS PHD2 expression was not altered substantially in cancer tissues and their metastases. Lymph node-negative tissues had higher levels of PHD2 than lymph node-positive tissues. PHD2 was hypoxia-inducible in pancreatic cancer cell lines and regulated cell growth through cyclin D1 down-regulation samples with PHD2 suppression and through p21 up-regulation in samples with of PHD2 overexpression. In vivo, PHD2 caused tumor growth retardation and reduced tumor invasion by inhibiting angiogenesis. This observation was caused by the suppression of angiogenic cytokines and tumor invasion. CONCLUSIONS The current results indicated that PHD2 plays an important role in pancreatic tumorigenesis. In summary, the authors concluded that PHD2 may function as a tumor suppressor gene in pancreatic cancer and, thus, may define a potential target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yun Su
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
94
|
Human pancreatic cancer stem cells: implications for how we treat pancreatic cancer. Transl Oncol 2011; 1:14-8. [PMID: 18607507 DOI: 10.1593/tlo.08013] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 02/08/2008] [Accepted: 02/11/2008] [Indexed: 02/08/2023] Open
Abstract
Pancreatic cancer has the worst prognosis of any major malignancy, with an annual death rate that approximates the annual incidence rate. Delayed diagnosis, relative chemotherapy and radiation resistance and an intrinsic biologic aggressiveness all contribute to the abysmal prognosis associated with pancreatic cancer. Answers to the frustrating effort to find effective therapies for pancreatic cancer may be gained through a renewed perspective on tumorigenesis as a process governed by a select population of cells, termed cancer stem cells (CSCs). Cancer stem cells, like their normal counterparts, have the properties of self-renewal and multilineage differentiation and possess inherently heightened DNA damage response and repair mechanisms that make them difficult to eradicate. Initially discovered in leukemias, researchers have identified CSCs in several solid-organ malignancies including breast, brain, prostate, and colon cancers. We have recently identified a CSC population in human pancreatic cancers. These pancreatic CSC represent 0.5% to 1.0% of all pancreatic cancer cells and express the cell surface markers CD44, CD24, and epithelial-specific antigen. Pancreatic CSCs have been shown to be resistant to standard chemotherapy and radiation, and devising specific therapies to target this distinct cell population is likely needed to identify effective therapies to treat this dismal disease.
Collapse
|
95
|
Li CL, Cui YF, Du XF, Tai S. Clinical significance of matrix metalloproteinases-3 and vascular endothelial growth factor expression in pancreatic carcinoma. Shijie Huaren Xiaohua Zazhi 2011; 19:1574-1578. [DOI: 10.11569/wcjd.v19.i15.1574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of vascular endothelial growth factor (VEGF) and matrix metalloproteinases-3 (MMP-3) in pancreatic carcinoma (PC) and to analyze their relationship with tumor angiogenesis, invasion and metastasis.
METHODS: The protein expression of VEGF and MMP-3 and microvessel density (MVD) were detected by immunohistochemistry in 56 PC specimens and 56 normal pancreatic tissue specimens.
RESULTS: The positive rates of both MMP-3 and VEGF were significantly higher in PC than in normal pancreatic tissue (75.00% vs 3.57%, 67.85% vs 1.78%, both P < 0.01). There was a positive correlation between the expression of VEGF and that of MMP-3. Higher expression of MMP-3 was detected in late-stage PC and PC with lymph node metastasis. The expression of VEGF was correlated with clinical stage (P < 0.05). MVD was correlated with expression of MMP-3 and VEGF in PC (both P < 0.05).
CONCLUSION: MMP-3 and VEGF may promote tumor angiogenesis and play an important role in the development and progression of PC. Detection of MMP-3 expression could be used to predict early metastasis of PC. Lower expression of VEGF may be related to lower arterial blood supply in PC.
Collapse
|
96
|
Angst E, Chen M, Mojadidi M, Hines OJ, Reber HA, Eibl G. Bioluminescence imaging of angiogenesis in a murine orthotopic pancreatic cancer model. Mol Imaging Biol 2011; 12:570-5. [PMID: 20376570 PMCID: PMC2917614 DOI: 10.1007/s11307-010-0310-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Angiogenesis is essential for physiological processes as well as for carcinogenesis. New approaches to cancer therapy include targeting angiogenesis. One target is VEGF-A and its receptor VEGFR2. In this study, we sought to investigate pancreatic cancer angiogenesis in a genetically modified VEGFR2-luc-KI mouse. PROCEDURES Live in vivo bioluminescence imaging of angiogenesis was performed continuously until sacrifice in subcutaneous tumors as well as in orthotopically transplanted tumors. Tumor tissue was immunostained for CD-31 and VEGFR2. RESULTS Peritumoral angiogenesis measured by light emission was detected beginning at week 3 following subcutaneous injection. In the orthotopic model, light emission began at day 4, which likely corresponds to wound healing, and continued throughout the experimental period during tumor growth. Peritumoral CD-31 vessel- and VEGFR2-staining were positive. CONCLUSIONS The VEGFR2-luc-KI mouse is a valuable tool to demonstrate tumor angiogenesis and seems to be suitable to evaluate anti-angiogenic approaches in pancreatic cancer.
Collapse
Affiliation(s)
- Eliane Angst
- Hirshberg Laboratory for Pancreatic Cancer Research, Department of Surgery, UCLA Center for Excellence in Pancreatic Diseases, David Geffen School of Medicine, University of California-Los Angeles, 675 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| | | | | | | | | | | |
Collapse
|
97
|
Melstrom LG, Salabat MR, Ding XZ, Strouch MJ, Grippo PJ, Mirzoeva S, Pelling JC, Bentrem DJ. Apigenin down-regulates the hypoxia response genes: HIF-1α, GLUT-1, and VEGF in human pancreatic cancer cells. J Surg Res 2010; 167:173-81. [PMID: 21227456 DOI: 10.1016/j.jss.2010.10.041] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 10/22/2010] [Accepted: 10/28/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND The flavonoid apigenin exhibits anti-proliferative and anti-angiogenic activities. Our objective was to evaluate the effect of apigenin on hypoxia responsive genes important in pancreatic cancer cell proliferation. MATERIALS AND METHODS Immunohistochemistry for GLUT-1 expression was conducted on human pancreatic cancer samples and adjacent controls. Real-time RT-PCR, Western blot analysis, and enzyme-linked immunosorbent assay (ELISA) were conducted on CD18 and S2-013 human pancreatic cancer cells treated with apigenin (0-50 μM) in normoxic and hypoxic conditions to evaluate HIF-1α, GLUT-1, and VEGF mRNA and protein expression and secretion. RESULTS GLUT-1 expression was significantly increased in pancreatic adenocarcinoma samples versus adjacent controls (P < 0.001). Hypoxic conditions induced HIF-1α, GLUT-1, and VEGF protein expression in both CD18 and S2-013 pancreatic cancer cells. Apigenin (50 μM) blocked hypoxia induced up-regulation of all three proteins in both cell lines. Apigenin also impeded hypoxia-mediated induction of GLUT-1 and VEGF mRNA in both cell lines (P < 0.05). CONCLUSIONS Apigenin inhibits HIF-1α, GLUT-1, and VEGF mRNA and protein expression in pancreatic cancer cells in both normoxic and hypoxic conditions. This may account for the mechanism of apigenin's anti-proliferative and anti-angiogenic effects and further supports the potential of apigenin as a future chemopreventive agent for pancreatic cancer.
Collapse
Affiliation(s)
- Laleh G Melstrom
- Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
98
|
Kang SH, Yu MO, Park KJ, Chi SG, Park DH, Chung YG. Activated STAT3 Regulates Hypoxia-Induced Angiogenesis and Cell Migration in Human Glioblastoma. Neurosurgery 2010; 67:1386-95; discussion 1395. [DOI: 10.1227/neu.0b013e3181f1c0cd] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Affiliation(s)
- Shin-Hyuk Kang
- Department of Neurosurgery, Korea University Anam Hospital, College of Medicine, Korea University, Seoul, Korea
| | - Mi Ok Yu
- Department of Neurosurgery, College of Medicine, and School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Kyung-Jae Park
- Department of Neurosurgery, Korea University Anam Hospital, College of Medicine, Korea University, Seoul, Korea
| | - Sung-Gil Chi
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Dong-Hyuk Park
- Department of Neurosurgery, Korea University Anam Hospital, College of Medicine, Korea University, Seoul, Korea
| | - Yong-Gu Chung
- Department of Neurosurgery, Korea University Anam Hospital, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
99
|
Su Y, Loos M, Giese N, Hines OJ, Diebold I, Görlach A, Metzen E, Pastorekova S, Friess H, Büchler P. PHD3 regulates differentiation, tumour growth and angiogenesis in pancreatic cancer. Br J Cancer 2010; 103:1571-9. [PMID: 20978507 PMCID: PMC2990580 DOI: 10.1038/sj.bjc.6605936] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose: Tumour hypoxia activates hypoxia-inducible factor-1 (HIF-1) and indluences angiogenesis, cell survival and invasion. Prolyl hydroxylase-3 (PHD3) regulates degradation of HIF-1α. The effects of PHD3 in tumour growth are largely unknown. Experimental design: PHD3 expression was analysed in human pancreatic cancer tissues and cancer cell lines by real-time quantitative PCR and immunohistochemistry. PHD3 overexpression was established by stable transfection and downregulation by short interfering RNA technology. VEGF was quantified by enzyme-linked immunosorbent assay. Matrigel invasion assays were performed to examine tumour cell invasion. Apoptosis was measured by annexin-V staining and caspase-3 assays. The effect of PHD3 on tumour growth in vivo was evaluated in an established orthotopic murine model. Results: PHD3 was upregulated in well-differentiated human tumours and cell lines, and regulated hypoxic VEGF secretion. PHD3 overexpression mediated tumour cell growth and invasion by induction of apoptosis in a nerve growth factor-dependent manner by the activation of caspase-3 and phosphorylation of focal adhesion kinase HIF-1 independently. In vivo, PHD3 inhibited tumour growth by abrogation of tumour angiogenesis. Conclusion: Our results indicate essential functions of PHD3 in tumour growth, apoptosis and angiogenesis and through HIF-1-dependent and HIF-1-independent pathways.
Collapse
Affiliation(s)
- Y Su
- Department of General Surgery, University of Heidelberg, Im Neuenheimer Feld 110, Heidelberg 69120, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Talaat RM. Soluble angiogenesis factors in sera of Egyptian patients with hepatitis C virus infection: correlation with disease severity. Viral Immunol 2010; 23:151-7. [PMID: 20373995 DOI: 10.1089/vim.2009.0089] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) infection causes chronic hepatitis, which gradually progresses to liver cirrhosis and subsequently to hepatocellular carcinoma (HCC). Angiogenesis plays a major role in chronic inflammation and may have prognostic value in disease progression. This study was designed to evaluate vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), and tumor necrosis factor-alpha (TNF-alpha) as prognostic factors of disease progression in Egyptian patients with different stages of HCV-related cirrhosis and HCC. VEGF, PDGF, and TNF-alpha were measured using enzyme-linked immunosorbent assay (ELISA) in 82 HCV-infected patients (20 mild, 20 moderate, and 20 severe cirrhosis patients, and 22 HCC patients), and 20 healthy controls. Our results showed comparable increases in VEGF and PDGF levels in those with increasing clinical stages of disease, with maximal production seen in HCC patients. A gradual elevation of TNF-alpha levels was seen also in HCV-infected patients at different stages of disease and HCC. A statistically significantly positive correlation between serum levels of VEGF, PDGF, and TNF-alpha, and grade of disease was recorded. Thus assessment of these parameters in those with different stages of disease may be helpful in choosing the best treatment strategy, and indicate that anti-angiogenic therapy may be useful.
Collapse
Affiliation(s)
- Roba M Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), Menofia University, Sadat City, Egypt.
| |
Collapse
|