51
|
Ko YS, Cho SJ, Park J, Kim Y, Choi YJ, Pyo JS, Jang BG, Park JW, Kim WH, Lee BL. Loss of FOXO1 promotes gastric tumour growth and metastasis through upregulation of human epidermal growth factor receptor 2/neu expression. Br J Cancer 2015; 113:1186-96. [PMID: 26448177 PMCID: PMC4647872 DOI: 10.1038/bjc.2015.273] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/04/2015] [Accepted: 07/01/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The biological significance of FOXO1, a member of the forkhead box O transcription factor family, in gastric cancer (GC) remains unclear. The present study provides direct evidence of the role of FOXO1 in tumour growth and metastasis of GC in relation to human epidermal growth factor receptor 2 (HER2). METHODS The expressions of FOXO1 and HER2 were modulated in GC cell lines (SNU-638, MKN45, SNU-216 and NCI-N87) by stable transfections. The effects of transfection on GC phenotypes were evaluated in vitro and in animal models. In addition, the relationship between FOXO1 and HER2 was analysed using GC clinical specimens, cell lines and xenografts. RESULTS FOXO1 silencing in GC cells increased colony formation and mesenchymal transition in vitro, as well as tumour growth and metastasis in nude mice, whereas HER2 silencing induced the opposite results.. Furthermore, an inverse relationship between FOXO1 and HER2 was found in clinical specimens of GC, GC cells and GC xenograft tumours. Although a negative crosstalk between these two molecules was shown, double knockdown of both FOXO1 and HER2 in GC cells revealed that HER2 silencing reversed the FOXO1 shRNA-induced migration and invasion even without the FOXO1 restoration. CONCLUSIONS Our results indicate that loss of FOXO1 promotes GC growth and metastasis by upregulating HER2 expression and that the HER2 expression is more critical to the induction of GC cell metastasis. The present study provides evidence that the FOXO1/HER2 pathway may regulate GC progression in a subgroup of GC patients.
Collapse
Affiliation(s)
- Young San Ko
- Department of Anatomy, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, South Korea
| | - Sung Jin Cho
- Department of Anatomy, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, South Korea
| | - Jinju Park
- Tumour Biology (Cancer Research Institute), Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Yong Joon Choi
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 110-746, South Korea
| | - Bo Gun Jang
- Department of Pathology, Jeju National University Hospital, Jeju 690-767, South Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul 110-799, South Korea.,Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Woo Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul 110-799, South Korea
| | - Byung Lan Lee
- Department of Anatomy, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 110-799, South Korea.,Tumour Biology (Cancer Research Institute), Seoul National University College of Medicine, Seoul 110-799, South Korea.,Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, South Korea
| |
Collapse
|
52
|
HIF-1α Plays a Critical Role in the Gestational Sidestream Smoke-Induced Bronchopulmonary Dysplasia in Mice. PLoS One 2015; 10:e0137757. [PMID: 26361040 PMCID: PMC4567349 DOI: 10.1371/journal.pone.0137757] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/20/2015] [Indexed: 11/24/2022] Open
Abstract
Rationale Smoking during pregnancy increases the risk of bronchopulmonary dysplasia (BPD) and, in mice, gestational exposure to sidestream cigarette smoke (SS) induces BPD-like condition characterized by alveolar simplification, impaired angiogenesis, and suppressed surfactant protein production. Normal fetal development occurs in a hypoxic environment and nicotinic acetylcholine receptors (nAChRs) regulate the hypoxia-inducible factor (HIF)-1α that controls apoptosis and angiogenesis. To understand SS-induced BPD, we hypothesized that gestational SS affected alveolar development through HIF-1α. Methods Pregnant BALB/c mice were exposed to air (control) or SS throughout the gestational period and the 7-day-old lungs of the progeny were examined. Results Gestational SS increased apoptosis of alveolar and airway epithelial cells. This response was associated with increased alveolar volumes, higher levels of proapoptotic factors (FOXO3a, HIPK2, p53, BIM, BIK, and BAX) and the antiangiogenic factor (GAX), and lower levels of antiapoptotic factors (Akt-PI3K, NF-κB, HIF-1α, and Bcl-2) in the lung. Although gestational SS increased the cells containing the proangiogenic bombesin-like-peptide, it markedly decreased the expression of its receptor GRPR in the lung. The effects of SS on apoptosis were attenuated by the nAChR antagonist mecamylamine. Conclusions Gestational SS-induced BPD is potentially regulated by nAChRs and associated with downregulation of HIF-1α, increased apoptosis of epithelial cells, and increased alveolar volumes. Thus, in mice, exposure to sidestream tobacco smoke during pregnancy promotes BPD-like condition that is potentially mediated through the nAChR/HIF-1α pathway.
Collapse
|
53
|
Wu MH, Hsiao KY, Tsai SJ. Endometriosis and possible inflammation markers. Gynecol Minim Invasive Ther 2015. [DOI: 10.1016/j.gmit.2015.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
54
|
Manu KA, Shanmugam MK, Ramachandran L, Li F, Siveen KS, Chinnathambi A, Zayed M, Alharbi SA, Arfuso F, Kumar AP, Ahn KS, Sethi G. Isorhamnetin augments the anti-tumor effect of capeciatbine through the negative regulation of NF-κB signaling cascade in gastric cancer. Cancer Lett 2015; 363:28-36. [DOI: 10.1016/j.canlet.2015.03.033] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/18/2015] [Accepted: 03/18/2015] [Indexed: 01/28/2023]
|
55
|
Abstract
Oxygen is the basic molecule which supports life and it truly is "god's gift to life." Despite its immense importance, research on "oxygen biology" has never received the light of the day and has been limited to physiological and biochemical studies. It seems that in modern day biology, oxygen research is summarized in one word "hypoxia." Scientists have focused on hypoxia-induced transcriptomics and molecular-cellular alterations exclusively in disease models. Interestingly, the potential of oxygen to control the basic principles of biology like homeostatic maintenance, transcription, replication, and protein folding among many others, at the molecular level, has been completely ignored. Here, we present a perspective on the crucial role played by oxygen in regulation of basic biological phenomena. Our conclusion highlights the importance of establishing novel research areas like oxygen biology, as there is great potential in this field for basic science discoveries and clinical benefits to the society.
Collapse
|
56
|
Kim SY, Ko YS, Park J, Choi Y, Park JW, Kim Y, Pyo JS, Yoo YB, Lee JS, Lee BL. Forkhead Transcription Factor FOXO1 Inhibits Angiogenesis in Gastric Cancer in Relation to SIRT1. Cancer Res Treat 2015; 48:345-54. [PMID: 25761483 PMCID: PMC4720104 DOI: 10.4143/crt.2014.247] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 12/05/2014] [Indexed: 01/26/2023] Open
Abstract
Purpose We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expression in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. Materials and Methods Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. Results In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1α (HIF-1α) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1α activation and vascular endothelial growth factor (VEGF) expression. In addition, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. Conclusion Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic conditions via inactivation of the HIF-1α–VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.
Collapse
Affiliation(s)
- Sue Youn Kim
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Young San Ko
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea
| | - Jinju Park
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yiseul Choi
- Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Wan Park
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Younghoon Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Bok Yoo
- Department of Anatomy, Dankook University School of Medicine, Cheonan, Korea
| | - Jae-Seon Lee
- Department of Biomedical Sciences, Inha University College of Medicine, Incheon, Korea
| | - Byung Lan Lee
- Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea ; Tumour Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea ; Ischemic/Hypoxic Disease Institute Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
57
|
Oral delivery of prolyl hydroxylase inhibitor: AKB-4924 promotes localized mucosal healing in a mouse model of colitis. Inflamm Bowel Dis 2015; 21:267-75. [PMID: 25545377 DOI: 10.1097/mib.0000000000000277] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pharmacological induction of hypoxia-inducible factor (HIF), a global transcriptional regulator of the hypoxic response, by prolyl hydroxylase inhibitors (PHDi) is protective in murine models of colitis, and epithelial cells are critical for the observed therapeutic efficacy. Because systemic HIF activation may lead to potentially negative off-target effects, we hypothesized that targeting epithelial HIF through oral delivery of PHDi would be sufficient to protect against colitis in a mouse model. METHODS Using a chemically induced trinitrobenzene sulfonic acid murine model of colitis, we compared the efficacy of oral and intraperitoneal (i.p.) delivery of the PHDi; AKB-4924 in preventing colitis, as measured by endoscopy, histology, barrier integrity, and immune profiling. Furthermore, we measured potential off-target effects, examining HIF and HIF target genes in the heart and kidney, as well as erythropoietin and hematocrit levels. RESULTS Oral administration of AKB-4924 exhibited mucosal protection comparable i.p. dosing. Oral delivery of PHDi led to reduced colonic epithelial HIF stabilization compared with i.p. delivery, but this was still sufficient to induce transcription of downstream HIF targets. Furthermore, oral delivery of PHDi led to reduced stabilization of HIF and activation of HIF targets in extraintestinal organs. CONCLUSIONS Oral delivery of PHDi therapies to this intestinal mucosa protects against colitis in animal models and represents a potential therapeutic strategy for inflammatory bowel disease, which also precludes unwanted extraintestinal effects.
Collapse
|
58
|
Li P, Li Z. Effects of NF-κB and hypoxia on the biological behavior of Y79 retinoblastoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1725-1730. [PMID: 25973060 PMCID: PMC4396231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/28/2015] [Indexed: 06/04/2023]
Abstract
We aimed to investigate the influence of nuclear factor-κB (NF-κB) on the biological behavior of Y79 retinoblastoma cells exposed to hypoxia and its possible mechanism. The cells were administrated with hypoxia, and/or 5 μM pyrrolidine dithiocarbamate (PDTC) (a selective NF-κB inhibitor) to inhibit the NF-κB activity, expressions of NF-κB was measured by western blot, and the translocation of NF-κB was detected. To examine the proliferation of Y79 cells, MTT assay was applied. Transwell assay was used to detect the invasion and migration ability of cells. The expressions of molecules involved in invasion was analyzed including HIF-1α, MMP-2, 9, and VEGF. We found that hypoxia significantly activated NF-κB activity. While once the NF-κB was inhibited, the proliferation, invasion and migration ability of Y79 cells were also blocked. Interestingly, the expressions of invasion-involved molecules elevated by hypoxia induction were also decreased when NF-κB was inhibited. Hypoxia could significantly change the adhesive and invasive ability of Y79 retinoblastoma cells, NF-κB signal might be one of the main mediators for these hypoxia induced cell changes of biological behavior via downregulation of HIF-1α and the invasion related molecules, and the mechanism still needs further investigation.
Collapse
Affiliation(s)
- Peng Li
- Department of Ophthalmology, Tianjin Eye HospitalTianjin 300020, China
| | - Zhaohui Li
- Department of Ophthalmology, General Hospital of PLABeijing 100853, China
| |
Collapse
|
59
|
Ikemori RY, Machado CML, Furuzawa KM, Nonogaki S, Osinaga E, Umezawa K, de Carvalho MA, Verinaud L, Chammas R. Galectin-3 up-regulation in hypoxic and nutrient deprived microenvironments promotes cell survival. PLoS One 2014; 9:e111592. [PMID: 25369297 PMCID: PMC4219723 DOI: 10.1371/journal.pone.0111592] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 10/06/2014] [Indexed: 01/20/2023] Open
Abstract
Galectin-3 (gal-3) is a β-galactoside binding protein related to many tumoral aspects, e.g. angiogenesis, cell growth and motility and resistance to cell death. Evidence has shown its upregulation upon hypoxia, a common feature in solid tumors such as glioblastoma multiformes (GBM). This tumor presents a unique feature described as pseudopalisading cells, which accumulate large amounts of gal-3. Tumor cells far from hypoxic/nutrient deprived areas express little, if any gal-3. Here, we have shown that the hybrid glioma cell line, NG97ht, recapitulates GBM growth forming gal-3 positive pseudopalisades even when cells are grafted subcutaneously in nude mice. In vitro experiments were performed exposing these cells to conditions mimicking tumor areas that display oxygen and nutrient deprivation. Results indicated that gal-3 transcription under hypoxic conditions requires previous protein synthesis and is triggered in a HIF-1α and NF-κB dependent manner. In addition, a significant proportion of cells die only when exposed simultaneously to hypoxia and nutrient deprivation and demonstrate ROS induction. Inhibition of gal-3 expression using siRNA led to protein knockdown followed by a 1.7–2.2 fold increase in cell death. Similar results were also found in a human GBM cell line, T98G. In vivo, U87MG gal-3 knockdown cells inoculated subcutaneously in nude mice demonstrated decreased tumor growth and increased time for tumor engraftment. These results indicate that gal-3 protected cells from cell death under hypoxia and nutrient deprivation in vitro and that gal-3 is a key factor in tumor growth and engraftment in hypoxic and nutrient-deprived microenvironments. Overexpression of gal-3, thus, is part of an adaptive program leading to tumor cell survival under these stressing conditions.
Collapse
Affiliation(s)
- Rafael Yamashita Ikemori
- Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
- * E-mail: (RYI); (RC)
| | - Camila Maria Longo Machado
- Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
- Laboratório de Investigação Médica em Medicina Nuclear – LIM43, São Paulo, SP, Brazil
| | - Karina Mie Furuzawa
- Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
| | - Suely Nonogaki
- Departamento de Patologia do Instituto Adolfo Lutz, São Paulo, SP, Brazil
| | - Eduardo Osinaga
- Facultad de Medicina de La Universidad de La Republica, Montevideo, Uruguay
| | | | | | - Liana Verinaud
- Departamento de Microbiologia e Imunologia, Instituto de Biologia, UNICAMP, Campinas, SP, Brazil
| | - Roger Chammas
- Faculdade de Medicina da Universidade de São Paulo, Instituto do Câncer do Estado de São Paulo, São Paulo, SP, Brazil
- * E-mail: (RYI); (RC)
| |
Collapse
|
60
|
Zhang H, Sun L, Xiao X, Xie R, Liu C, Wang Y, Wei Y, Zhang H, Liu L. Krüppel-like factor 8 contributes to hypoxia-induced MDR in gastric cancer cells. Cancer Sci 2014; 105:1109-15. [PMID: 25040744 PMCID: PMC4462403 DOI: 10.1111/cas.12483] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/31/2022] Open
Abstract
We previously reported that hypoxia-induced MDR in gastric cancer (GC) cells is hypoxia-inducible factor-1 (HIF-1)-dependent. However, the exact mechanisms are still unknown. Our previous study revealed that Krüppel-like factor 8 (KLF8), a novel transcription factor, was associated with malignant phenotype in GC cells. KLF8 is overexpressed in clear cell renal carcinoma lacking von Hippel-Lindau protein function, which resulted in HIF-1 stabilization. Given this association, we hypothesized that KLF8 contributed to hypoxia-induced MDR in GC cells. Initial experiments revealed that hypoxia could increase KLF8 and HIF-1α expressions in GC cells, and KLF8 levels in GC drug-resistant cell lines were higher than in parental cell lines. Subsequent experiments showed that in normoxia, exogenous KLF8 could promote the MDR phenotype; however, blocking KLF8 expression could effectively reverse the MDR phenotype induced by hypoxia. Overexpressed KLF8 increased resistance-associated gene MDR1 mRNA levels, Bcl-2 and P-gp protein levels, and decreased Bax and caspase-3 protein levels in GC cells, and knockout KLF8 reversed these effects. Dual luciferase reporter and ChIP assays showed that KLF8 could promote MDR1 transcriptional activity by combining with KLF8 binding sites located in the upstream of MDR1 transcriptional start site. These results suggest that KLF8 is involved in hypoxia-induced MDR through inhibiting apoptosis and increasing the drug release rate by directly regulating MDR1 transcription. This study aims to discuss whether KLF8 involves in hypoxia-induced multi-drug resistance and its mechanism. Our results showed that hypoxia could increase KLF8 expression in gastric cancer cells. Meanwhile, we found that KLF8 contributed to hypoxia-induced multi-drug resistance via regulating MDR1 directly. Through this research, we found a new target gene of KLF8 and further clarified the mechanism of multi-drug resistance happened in gastric cancer.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Oncology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China; Department of Tuberculosis Control and Prevention, Xi'an Center for Disease Control and Prevention, Xi'an, China
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Yang T, Zhang X, Wang M, Zhang J, Huang F, Cai J, Zhang Q, Mao F, Zhu W, Qian H, Xu W. Activation of mesenchymal stem cells by macrophages prompts human gastric cancer growth through NF-κB pathway. PLoS One 2014; 9:e97569. [PMID: 24824968 PMCID: PMC4019592 DOI: 10.1371/journal.pone.0097569] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/21/2014] [Indexed: 01/13/2023] Open
Abstract
Accumulating evidence indicate that macrophages activate mesenchymal stem cells (MSCs) to acquire pro-inflammatory phenotype. However, the role of MSCs activated by macrophages in gastric cancer remains largely unknown. In this study, we found that MSCs were activated by macrophages to produce increased levels of inflammatory cytokines. Cell colony formation and transwell migration assays revealed that supernatants from the activated MSCs could promote both gastric epithelial cell and gastric cancer cell proliferation and migration. In addition, the expression of epithelial-mesenchymal transition (EMT), angiogenesis, and stemness-related genes was increased in activated MSCs. The phosphorylated forms of NF-κB, ERK and STAT3 in gastric cells were increased by active MSCs. Inhibition of NF-κB activation by PDTC blocked the effect of activated MSCs on gastric cancer cells. Co-injection of activated MSCs with gastric cancer cells could accelerate gastric cancer growth. Moreover, human peripheral blood monocytes derived macrophages also activated MSCs to prompt gastric cancer cell proliferation and migration. Taken together, our findings suggest that MSCs activated by macrophage acquire pro-inflammatory phenotype and prompt gastric cancer growth in an NF-κB-dependent manner, which provides new evidence for the modulation of MSCs by tumor microenvironment and further insight to the role of stromal cells in gastric carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Tingting Yang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Xu Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Mei Wang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Jie Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Feng Huang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Jie Cai
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Qiang Zhang
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Fei Mao
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Wei Zhu
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
| | - Hui Qian
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
- * E-mail: (WX); (HQ)
| | - Wenrong Xu
- School of Medical Science and Laboratory Medicine, Jiangsu University Zhenjiang, Jiangsu, China
- The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- * E-mail: (WX); (HQ)
| |
Collapse
|
62
|
Periasamy J, Muthuswami M, Rao DB, Tan P, Ganesan K. Stratification and delineation of gastric cancer signaling by in vitro transcription factor activity profiling and integrative genomics. Cell Signal 2014; 26:880-94. [PMID: 24462706 DOI: 10.1016/j.cellsig.2014.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 01/12/2023]
Abstract
Integrative functional genomic approaches are helpful in delineating the complex dysregulations in cancers. In the present study, in vitro activity profiling of 45 signaling pathway driven transcription factors in eight gastric cancer cell lines and direct comparison with genome-wide profiles of gastric tumors were performed and the integration resulted in the identification of three categories of factors/pathways: i) highly activated signaling pathways that stem from mutations are the critical oncogenic drivers, ii) constitutively activated stress responsive pathways which are activated not due to genetic alterations, and iii) consistently down-regulated nuclear receptor responsive factors. This functional profiling helps in discriminating therapeutic targets and signaling interactions.
Collapse
Affiliation(s)
- Jayaprakash Periasamy
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Muthulakshmi Muthuswami
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Divya Bhaskar Rao
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Patrick Tan
- Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore
| | - Kumaresan Ganesan
- Cancer Genetics Laboratory, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India.
| |
Collapse
|
63
|
Yu DA, Yoon J, Ko YS, Park J, Kim SY, Kim MA, Kim JH, Jung J, Cheon Y, Lee HS, Kim WH, Lee BL. Forkhead transcription factor FOXO1 inhibits nuclear factor-κB in gastric cancer. APMIS 2014; 122:848-55. [DOI: 10.1111/apm.12247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 12/18/2013] [Indexed: 12/28/2022]
Affiliation(s)
- Da-Ae Yu
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Jiyeon Yoon
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Young San Ko
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Jinju Park
- Cancer Research Institute; Department of Tumor Biology; Seoul National University College of Medicine; Seoul South Korea
| | - Sue Youn Kim
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
| | - Min A Kim
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Ji Hun Kim
- Department of Pathology; Asan Medical Center; University of Ulsan College of Medicine; Seoul South Korea
| | - Jieun Jung
- Department of Nanobiomedical Science; Dankook University; Cheonan South Korea
| | - Younghee Cheon
- Department of Pediatrics; College of Medicine; Yeungnam University; Daegu South Korea
| | - Hye Seung Lee
- Department of Pathology; Seoul National University Bundang Hospital; Seongnam South Korea
| | - Woo Ho Kim
- Department of Pathology; Seoul National University College of Medicine; Seoul South Korea
| | - Byung Lan Lee
- Department of Anatomy; Seoul National University College of Medicine; Seoul South Korea
- Cancer Research Institute; Department of Tumor Biology; Seoul National University College of Medicine; Seoul South Korea
- Ischemic/Hypoxic Disease Institute Medical Research Center; Seoul National University College of Medicine; Seoul South Korea
| |
Collapse
|
64
|
Park KW, Kim SJ, Oh SY. Clinicopathologic significance of nuclear factor-κB and vascular endothelial growth factor expression in advanced gastric cancer patients. Oncol Res Treat 2014; 37:183-90. [PMID: 24732642 DOI: 10.1159/000360777] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 02/04/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Nuclear factor-κB (NF-κB) and vascular endothelial growth factor (VEGF) are involved in cell proliferation, invasion, angiogenesis, and metastasis. The principal objective of this study was to assess the prognostic significance of NF-κB and VEGF expression in gastric cancer. METHODS Tumor tissues of 154 patients with gastric cancer, all of whom underwent potentially curative resection, were immunohistochemically evaluated using monoclonal antibodies against NF-κB and VEGF. RESULTS The positivity rates of NF-κB and VEGF staining were 44.2% and 39.6%, respectively. NF-κB expression in tumor tissues was significantly correlated with VEGF expression (p < 0.001). VEGF expression was related to Lauren's classification (p = 0.002), differentiation (p = 0.043), depth of invasion (p = 0.005), carcinoembryonic antigen expression (p = 0.032), and stage (p = 0.026). Univariate analysis demonstrated that NF-κB expression was significantly related to both the 5-year disease-free survival (65.2% vs. 46.4%, p = 0.007) and the 5-year overall survival (60.0% vs. 42.5%, p = 0.014). Multivariate analysis verified that NF-κB was independently associated with disease-free survival (hazard ratio: 2.082, p = 0.005) and overall survival (hazard ratio: 1.841, p = 0.008). CONCLUSION NF-κB expression in tumor tissue is associated with poor survival in gastric cancer patients.
Collapse
Affiliation(s)
- Keon Woo Park
- Division of Hematology-Oncology, Department of Medicine, Dankook University Hospital, Cheonan, Korea
| | | | | |
Collapse
|
65
|
Chen L, Shi Y, Yuan J, Han Y, Qin R, Wu Q, Jia B, Wei B, Wei L, Dai G, Jiao S. HIF-1 alpha overexpression correlates with poor overall survival and disease-free survival in gastric cancer patients post-gastrectomy. PLoS One 2014; 9:e90678. [PMID: 24614305 PMCID: PMC3948685 DOI: 10.1371/journal.pone.0090678] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 02/03/2014] [Indexed: 12/16/2022] Open
Abstract
Background Overall, gastric cancer prognosis remains poor. Detailed characterization of molecular markers that govern gastric cancer pathogenesis is warranted to establish innovative therapeutic options. HIF-1α overexpression has been linked to poor gastric cancer prognosis. However, though researched for years, the prognostic role of HIF-1α in gastric cancer is still controversial. Hence, the objective of the present study was to analyze the prognostic values of HIF-1α, TGF-β, VEGF and pERK1/2 in gastric cancer patients following gastrectomy. Methods This study included 446 patients with confirmed gastric cancer who underwent gastrectomy in a single Chinese Cancer Center between 2005 and 2006. Clinicopathologic features, as well as immunohistochemical analysis of TGF-β, HIF-1α, VEGF and pERK1/2 were determined. Long-term survival of these patients was analyzed using univariate and multivariate analyses. Results HIF-1α overexpression was more frequent in patients with hepatic metastases (71.6% versus 43.0% in those without hepatic metastases, P = 0.000, χ2 = 23.086) and more frequent in patients with peritoneum cavity metastasis (62.3% versus 43.0% in those without such metastasis, P = 0.000, χ2 = 13.691). In univariate analysis, patients with HIF-1α overexpression had a shorter disease-free survival (DFS) and overall survival (OS) than patients with weak-expression (DFS: NA VS. 16.8 m, P = 0.000, χ2 = 74.937; OS: NA VS. 25.5 m, P = 0.000, χ2 = 90.594). Importantly, HIF-1α overexpression was a promising prognostic marker for poor survival by multivariate analysis (DFS: HR 2.766, 95%CI 2.136–2.583, P = 0.000; OS: HR 3.529, 95%CI 2.663–4.667, P = 0.000). Conclusions HIF-1α overexpression could be considered a useful independent prognostic biomarker in gastric cancer after gastrectomy, and is correlated to both a poor overall survival and disease-free survival in these patients. HIF-1α expression can be used to stratify patients at higher risk for poor prognosis, and is potentially an important therapeutic target in gastric cancer patients.
Collapse
Affiliation(s)
- Li Chen
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yan Shi
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jing Yuan
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yalin Han
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rui Qin
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Qian Wu
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Baoqing Jia
- Department of Surgical oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Bo Wei
- Department of General surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixin Wei
- Department of Pathology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guanghai Dai
- Department of Comprehensive Treatment Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Medical oncology, Chinese People's Liberation Army General Hospital, Beijing, China
- * E-mail:
| |
Collapse
|
66
|
Wang J, Zhang J, Zhou C, Chen L, Yu Q. An insertion/deletion polymorphism within the proximal promoter of EGLN2 is associated with susceptibility for gastric cancer in the Chinese population. Genet Test Mol Biomarkers 2014; 18:269-73. [PMID: 24517638 DOI: 10.1089/gtmb.2013.0438] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Gastric cancer (GC) is among the most common human malignancies and the second leading cause of cancer-related death worldwide. Accumulated evidence from molecular genetics indicates that an individual's genetic factors are involved in their susceptibility to GC. Hypoxia is a common feature of cancer and the hypoxia-inducible factor (HIF), a transcription factor that regulates oxygen homeostasis, plays key roles in the growth of solid tumors and regulating cellular responses to hypoxia. Prolyl hydroxylase (PHD1, also known as EGLN2) is one of the three enzymes capable of hydroxylating the alpha subunit of HIF and results in polyubiquitinylation and proteasomal degradation of HIF. A case-control study, including 415 GC patients and 830 healthy controls, was conducted to investigate the association between GC susceptibility with a 4-bp insertion/deletion polymorphism (rs10680577) in the proximal promoter of EGLN2. Logistic regression analysis showed that the heterozygote and the homozygote 4-bp del/del confer a significantly increased risk of GC after controlling for other covariates (adjusted odds ratio [OR]=1.35, 95% confidence interval [CI] 1.05-1.75, p=0.017; OR=2.19, 95% CI 1.15-4.18, p=0.009, respectively). Carriage of the 4-bp deletion allele was associated with a greatly increased risk of developing the disease (OR=1.38, 95% CI 1.12-1.70, p=0.002). Moreover, stratification analysis showed that the association was more prominent in smokers (adjusted OR=2.09, 95% CI=1.40-3.12, p for heterogeneity=0.01). Our data suggested that common genetic polymorphisms in EGLN2 may influence GC risk in the Chinese population. Considering the relative small sample size, replication in other populations with a larger sample size and further functional analysis are required for fully understanding the roles of EGLN2 polymorphisms in predisposition for GC.
Collapse
Affiliation(s)
- Jian Wang
- 1 Department of Oncology, The Fifth People's Hospital of Changshu , Changshu, People's Republic of China
| | | | | | | | | |
Collapse
|
67
|
Wu G, Yan WF, Zhu YZ, Sun PC. Hypoxia-inducible factor-1α (HIF-1α) C1772T polymorphism significantly contributes to the risk of malignancy from a meta-analysis. Tumour Biol 2014; 35:4113-22. [PMID: 24425105 DOI: 10.1007/s13277-013-1538-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 12/11/2013] [Indexed: 12/20/2022] Open
Abstract
Although the association between hypoxia-inducible factor-1α (HIF-1α) C1772T polymorphism and risk of malignancy has been widely studied, results from published studies remained controversial. Therefore, the relationship between them was further assessed in this meta-analysis. The databases of PubMed, Embase, and Wanfang were searched, and odds ratio with 95% confidence interval (OR and 95% CI) were used to assess the strength of the association. A total of 38 case-control studies with 23,876 participants were included. Overall, the T allele of HIF-1α C1772T was significantly associated with increased risk of malignancy development (OR and 95% CI 1.18 (1.00-1.38), P = 0.048 for T carriers vs. CC; 1.22 (1.05-1.41), P = 0.010 for T carriers vs. C carriers). When subgroup analyses were conducted, T allele was further found to be associated with increased risk of malignancy development for Asians rather than Caucasians (OR and 95% CI 1.36 (1.10-1.67), P = 0.004 for Asians) and for population-based studies (OR and 95% CI 1.19 (1.01-1.41), P = 0.040). Between-study heterogeneity existed in genetic comparison models, and meta-regression indicated that the participants' ethnicities and types of malignancy might be the sources of heterogeneity. No publication bias was found. In conclusion, this study indicated that HIF-1α C1772T polymorphism was significantly associated with increased risk of malignancy development for Asians. More studies were further required to focus on the relationship between HIF-1α C1772T polymorphism and risk of a specific type of tumor.
Collapse
Affiliation(s)
- Gang Wu
- Department of General Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No.7 Weiwu Road, Zhengzhou, China
| | | | | | | |
Collapse
|
68
|
Tsuboi K, Matsuo Y, Shamoto T, Shibata T, Koide S, Morimoto M, Guha S, Sung B, Aggarwal BB, Takahashi H, Takeyama H. Zerumbone inhibits tumor angiogenesis via NF-κB in gastric cancer. Oncol Rep 2014; 31:57-64. [PMID: 24220661 DOI: 10.3892/or.2013.2842] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/21/2013] [Indexed: 11/06/2022] Open
Abstract
Zerumbone derived from a subtropical ginger, Zingiber zerumbet Smith, was previously reported to have antitumor growth and anti-inflammatory properties in some types of cancer. However, the effects of zerumbone against cancer angiogenesis have not been fully elucidated. In this study, we clarified the role of zerumbone in gastric cancer angiogenesis. We examined the expression of vascular endothelial growth factor (VEGF) in gastric cancer cell lines both in the basal state and following zerumbone treatment by real-time RT-PCR and enzyme-linked immunosorbent assay (ELISA). Changes in gastric cancer cell proliferation in response to zerumbone treatment were measured by WST-1 assay. Additionally, the effects of zerumbone on NF-κB activity were examined in AGS cells. Finally, the effects of zerumbone on angiogenesis in AGS cells were measured by in vitro angiogenesis assay in which human umbilical vein endothelial cells (HUVECs) and fibroblasts were cocultured with AGS cells. Among the 6 gastric cancer cell lines tested, AGS cells exhibited the highest expression of VEGF. Cell proliferation, VEGF expression and NF-κB activity in AGS cells were all significantly inhibited by zerumbone. Moreover, the tube formation area of HUVECs was increased by coculture with AGS cells, and this effect was inhibited by zerumbone. Both VEGF expression and NF-κB activity in AGS cells were reduced by treatment with zerumbone, thereby inhibiting angiogenesis. Thus, zerumbone may become a new anti-angiogenic and antitumor drug in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Guan Z, Ding C, Du Y, Zhang K, Zhu JN, Zhang T, He D, Xu S, Wang X, Fan J. HAF drives the switch of HIF-1α to HIF-2α by activating the NF-κB pathway, leading to malignant behavior of T24 bladder cancer cells. Int J Oncol 2013; 44:393-402. [PMID: 24316875 PMCID: PMC3898811 DOI: 10.3892/ijo.2013.2210] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/19/2013] [Indexed: 12/14/2022] Open
Abstract
Hypoxia is a characteristic feature of solid tumors, leading to malignant behavior. During this process, HIF family members (HIFs) and the NF-κB pathway are activated. In addition, the hypoxia-associated factor (HAF) is reported to participate in the regulation of HIFs. However, the precise relationship among HIFs, HAF and the NF-κB pathway in bladder cancer (BC) remains unknown. In the current investigation, T24 BC cells were exposed to hypoxia, or by plasmid transfection to overexpress HAF or RelA (P65) to demonstrate their roles. The results indicate that hypoxia leads to the elevation of HAF plus activation of the NF-κB pathway, accompanied by the switch of HIF-1α to HIF-2α, resulting in the enhanced ability of malignancy in T24 cells. In order to further demonstrate the significance of this switch, HIF-1α and HIF-2α were co-transfected into T24 cells with HIF-β, respectively. The following results indicate that the T24hif-2α/β cells show enhanced ability of malignancy, accompanied by the maintenance of stem-cell markers, but the T24hif-1α/β cells show higher expression of metabolism-related genes. Boyden assays and wound-healing assays indicate the enhanced ability of malignancy for T24hif-2α/β. Thus, we conclude that on the hypoxic microenvironment, the switching of HIF-1α to HIF-2α, which is driven by HAF through activating the NF-κB pathway, contributes to the malignancy of T24 cells, accompanied by the maintenance of stem-cell markers. This provides us an avenue for understanding the progression of bladder cancer.
Collapse
Affiliation(s)
- Zhenfeng Guan
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Chen Ding
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Yiqing Du
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Kai Zhang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Jian Ning Zhu
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Tingting Zhang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shan Xu
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Xinyang Wang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
70
|
Baldo BA, Pham NH. Adverse reactions to targeted and non-targeted chemotherapeutic drugs with emphasis on hypersensitivity responses and the invasive metastatic switch. Cancer Metastasis Rev 2013; 32:723-61. [PMID: 24043487 PMCID: PMC7102343 DOI: 10.1007/s10555-013-9447-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
More than 100 drugs are used to treat the many different cancers. They can be divided into agents with relatively broad, non-targeted specificity and targeted drugs developed on the basis of a more refined understanding of individual cancers and directed at specific molecular targets on different cancer cells. Individual drugs in both groups have been classified on the basis of their mechanism of action in killing cancer cells. The targeted drugs include proteasome inhibitors, toxic chimeric proteins and signal transduction inhibitors such as tyrosine kinase (non-receptor and receptor), serine/threonine kinase, histone deacetylase and mammalian target of rapamycin inhibitors. Increasingly used targeted vascular (VEGF) and platelet-derived endothelial growth factor blockade can provoke a range of pathological consequences. Many of the non-targeted drugs are cytotoxic, suppressing haematopoiesis as well as provoking cutaneous eruptions and vascular, lung and liver injury. Cytotoxic side effects of the targeted drugs occur less often and usually with less severity, but they show their own unusual adverse effects including, for example, a lengthened QT interval, a characteristic papulopustular rash, nail disorders and a hand-foot skin reaction variant. The term hypersensitivity is widely used across a number of disciplines but not always with the same definition in mind, and the terminology needs to be standardised. This is particularly apparent in cancer chemotherapy where anti-neoplastic drug-induced thrombocytopenia, neutropenia, anaemia, vascular disorders, liver injury and lung disease as well as many dermatological manifestations sometimes have an immune basis. The most insidious of all adverse consequences of targeted therapies, however, are tumour adaptation, increased malignancy and the invasive metastatic switch seen with anti-angiogenic drugs that inhibit the VEGF-A pathway. Adverse reactions to 44 non-targeted and 33 targeted, frequently used, chemotherapeutic drugs are presented together with discussions of diagnosis, premedications, desensitizations and importance of understanding the mechanisms underlying the various drug-induced reactions. There is need for wide-ranging acceptance of what constitutes a hypersensitivity reaction and for allergists to be more involved in the diagnosis, treatment and prevention of chemotherapeutic drug-induced hypersensitivity reactions.
Collapse
Affiliation(s)
- Brian A Baldo
- Molecular Immunology Unit, Kolling Institute of Medical Research, Royal North Shore Hospital of Sydney, Sydney, New South Wales, Australia,
| | | |
Collapse
|
71
|
Yang S, Yu M, Sun L, Xiao W, Yang X, Sun L, Zhang C, Ma Y, Yang H, Liu Y, Lu D, Teitelbaum DH, Yang H. Interferon-γ-induced intestinal epithelial barrier dysfunction by NF-κB/HIF-1α pathway. J Interferon Cytokine Res 2013; 34:195-203. [PMID: 24237301 DOI: 10.1089/jir.2013.0044] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Interferon-γ (IFN-γ) plays an important role in intestinal barrier dysfunction. However, the mechanisms are not fully understood. As hypoxia-inducible factor-1 (HIF-1) is a critical determinant response to hypoxia and inflammation, which has been shown to be deleterious to intestinal barrier function, we hypothesized that IFN-γ induces loss of barrier function through the regulation of HIF-1α activation and function. In this study, we detected the expressions of HIF-1α and tight junction proteins in IFN-γ-treated T84 intestinal epithelial cell line. IFN-γ led to an increase of HIF-1α expression in time- and dose-dependent manners but did not change the expression of HIF-1β. The IFN-γ-induced increase in HIF-1α was associated with an activation of NF-κB. Treatment with the NF-κB inhibitor, pyrolidinedithiocarbamate (PDTC), significantly suppressed the activation of NF-κB and the expression of HIF-1α. In addition, IFN-γ also increased intestinal epithelial permeability and depletion of tight junction proteins; inhibition of NF-κB or HIF-1α prevented the increase in intestinal permeability and alteration in tight junction protein expressions. Interestingly, we demonstrated that a significant portion of IFN-γ activation NF-kB and modulation tight junction expression is mediated through HIF-1α. Taken together, this study suggested that IFN-γ induced the loss of epithelial barrier function and disruption of tight junction proteins, by upregulation of HIF-1α expression through NF-κB pathway.
Collapse
Affiliation(s)
- Songwei Yang
- 1 Department of General Surgery, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Cheng J, Fan XM. Role of cyclooxygenase-2 in gastric cancer development and progression. World J Gastroenterol 2013; 19:7361-7368. [PMID: 24259966 PMCID: PMC3831217 DOI: 10.3748/wjg.v19.i42.7361] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 09/12/2013] [Accepted: 09/29/2013] [Indexed: 02/06/2023] Open
Abstract
Although the incidence of gastric cancer has been declining in recent decades, it remains a major public health issue as the second leading cause of cancer death worldwide. In China, gastric cancer is still the main cause of death in patients with malignant tumors. Most patients are diagnosed at an advanced stage and mortality is high. Cyclooxygenase-2 (COX-2) is a rate-limiting enzyme in prostanoid synthesis and plays an important role in the development and progression of gastric cancer. The expression of COX-2 in gastric cancer is upregulated and its molecular mechanisms have been investigated. Helicobacter pylori infection, tumor suppressor gene mutation and the activation of nuclear factor-kappa B may be responsible for the elevated expression of COX-2 in gastric cancer. The mechanisms of COX-2 in the development and progression of gastric cancer are probably through promoting the proliferation of gastric cancer cells, while inhibiting apoptosis, assisting angiogenesis and lymphatic metastasis, and participating in cancer invasion and immunosuppression. This review is intended to discuss, comment and summarize recent research progress on the role of COX-2 in gastric cancer development and progression, and elucidate the molecular mechanisms which might be involved in the carcinogenesis.
Collapse
|
73
|
Simvastatin sensitizes human gastric cancer xenograft in nude mice to capecitabine by suppressing nuclear factor-kappa B-regulated gene products. J Mol Med (Berl) 2013; 92:267-76. [DOI: 10.1007/s00109-013-1095-0] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 09/21/2013] [Accepted: 10/14/2013] [Indexed: 01/28/2023]
|
74
|
Su JH, Chen YC, El-Shazly M, Du YC, Su CW, Tsao CW, Liu LL, Chou Y, Chang WB, Su YD, Chiang MY, Yeh YT, Lu MC. Towards the small and the beautiful: a small dibromotyrosine derivative from Pseudoceratina sp. sponge exhibits potent apoptotic effect through targeting IKK/NFκB signaling pathway. Mar Drugs 2013; 11:3168-85. [PMID: 24065159 PMCID: PMC3801119 DOI: 10.3390/md11093168] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 08/09/2013] [Accepted: 08/09/2013] [Indexed: 12/21/2022] Open
Abstract
A dibromotyrosine derivative, (1'R,5'S,6'S)-2-(3',5'-dibromo-1',6'-dihydroxy-4'-oxocyclohex-2'-enyl) acetonitrile (DT), was isolated from the sponge Pseudoceratina sp., and was found to exhibit a significant cytotoxic activity against leukemia K562 cells. Despite the large number of the isolated bromotyrosine derivatives, studies focusing on their biological mechanism of action are scarce. In the current study we designed a set of experiments to reveal the underlying mechanism of DT cytotoxic activity against K562 cells. First, the results of MTT cytotoxic and the annexin V-FITC/PI apoptotic assays, indicated that the DT cytotoxic activity is mediated through induction of apoptosis. This effect was also supported by caspases-3 and -9 activation as well as PARP cleavage. DT induced generation of reactive oxygen species (ROS) and the disruption of mitochondrial membrane potential (MMP) as indicated by flow cytometric assay. The involvement of ROS generation in the apoptotic activity of DT was further corroborated by the pretreatment of K562 cells with N-acetyl-cysteine (NAC), a ROS scavenger, which prevented apoptosis and the disruption of MMP induced by DT. Results of cell-free system assay suggested that DT can act as a topoisomerase II catalytic inhibitor, unlike the clinical anticancer drug, etoposide, which acts as a topoisomerase poison. Additionally, we found that DT treatment can block IKK/NFκB pathway and activate PI3K/Akt pathway. These findings suggest that the cytotoxic effect of DT is associated with mitochondrial dysfunction-dependent apoptosis which is mediated through oxidative stress. Therefore, DT represents an interesting reference point for the development of new cytotoxic agent targeting IKK/NFκB pathway.
Collapse
Affiliation(s)
- Jui-Hsin Su
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 944, Taiwan; E-Mail:
| | - Yu-Cheng Chen
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
| | - Mohamed El-Shazly
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (M.E.-S.); (Y.-C.D.)
- Department of Pharmacognosy and Natural Products Chemistry, Faculty of Pharmacy, Ain-Shams University, Organization of African Unity Street, Abassia, Cairo 11566, Egypt
| | - Ying-Chi Du
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; E-Mails: (M.E.-S.); (Y.-C.D.)
| | - Chiang-Wen Su
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 944, Taiwan; E-Mail:
| | - Chia-Wei Tsao
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
| | - Li-Lian Liu
- Institute of Marine Biology, National Sun Yat-sen University, Kaohsiung, Taiwan; E-Mails: (L.-L.L.); (Y.C.)
| | - Yalan Chou
- Institute of Marine Biology, National Sun Yat-sen University, Kaohsiung, Taiwan; E-Mails: (L.-L.L.); (Y.C.)
| | - Wen-Been Chang
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
| | - Yin-Di Su
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
- Department of Marine Biotechnology and Resources and Asia-Pacific Ocean Research Center, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Michael Y. Chiang
- Department of Chemistry, National Sun Yat-sen University, Kaohsiung 804, Taiwan; E-Mail:
| | - Yao-Tsung Yeh
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung 831, Taiwan; E-Mail:
| | - Mei-Chin Lu
- National Museum of Marine Biology & Aquarium, Pingtung 944, Taiwan; E-Mails: (J.-H.S.); (Y.-C.C.); (C.-W.T.); (W.-B.C.); (Y.-D.S.)
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 944, Taiwan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +886-888-250-37; Fax: +886-888-250-87
| |
Collapse
|
75
|
Qiu F, Shi CH, Zheng J, Liu YB. Periostin mediates the increased pro-angiogenic activity of gastric cancer cells under hypoxic conditions. J Biochem Mol Toxicol 2013; 27:364-9. [PMID: 23728938 DOI: 10.1002/jbt.21498] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/03/2013] [Indexed: 12/29/2022]
Abstract
This study was conducted to investigate the biological role of periostin in gastric cancer (GC) under hypoxia. Western blot analysis revealed that along with an upregulation of hypoxia-inducible factor-1alpha, there was a time-dependent induction of periostin in MKN-45 cells under hypoxia (2% O2 ), increasing by eightfold as compared to normoxic cells. Pretreatment with 30 µM PD98059, an inhibitor of ERK1/2, significantly reduced hypoxia-stimulated periostin expression (P < 0.01). Periostin knockdown in MKN-45 cells was achieved by specific small interfering RNA (siRNA). The conditioned medium from periostin siRNA-transfected MKN-45 cells induced significantly less (P < 0.01) endothelial tube formation than control siRNA-transfected cells. Additionally, periostin silencing markedly decreased the mRNA expression and secretion of vascular endothelial growth factor (VEGF) in hypoxic MKN-45 cells. Thus, our data suggest that periostin is a hypoxia-response gene and mediates a cross talk between GC and endothelial cells under hypoxia, partially through regulation of the VEGF expression.
Collapse
Affiliation(s)
- Feng Qiu
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | | | | | | |
Collapse
|
76
|
Anbo N, Ogi K, Sogabe Y, Shimanishi M, Kaneko T, Dehari H, Miyazaki A, Hiratsuka H. Suppression of NF-κB/p65 Inhibits the Proliferation in Oral Squamous Cancer Cells. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.44100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
77
|
Bao B, Azmi AS, Ali S, Ahmad A, Li Y, Banerjee S, Kong D, Sarkar FH. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1826:272-96. [PMID: 22579961 PMCID: PMC3788359 DOI: 10.1016/j.bbcan.2012.04.008] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/25/2012] [Accepted: 04/28/2012] [Indexed: 12/13/2022]
Abstract
Hypoxia is one of the fundamental biological phenomena that are intricately associated with the development and aggressiveness of a variety of solid tumors. Hypoxia-inducible factors (HIF) function as a master transcription factor, which regulates hypoxia responsive genes and has been recognized to play critical roles in tumor invasion, metastasis, and chemo-radiation resistance, and contributes to increased cell proliferation, survival, angiogenesis and metastasis. Therefore, tumor hypoxia with deregulated expression of HIF and its biological consequence lead to poor prognosis of patients diagnosed with solid tumors, resulting in higher mortality, suggesting that understanding of the molecular relationship of hypoxia with other cellular features of tumor aggressiveness would be invaluable for developing newer targeted therapy for solid tumors. It has been well recognized that cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) phenotypic cells are associated with therapeutic resistance and contribute to aggressive tumor growth, invasion, metastasis and believed to be the cause of tumor recurrence. Interestingly, hypoxia and HIF signaling pathway are known to play an important role in the regulation and sustenance of CSCs and EMT phenotype. However, the molecular relationship between HIF signaling pathway with the biology of CSCs and EMT remains unclear although NF-κB, PI3K/Akt/mTOR, Notch, Wnt/β-catenin, and Hedgehog signaling pathways have been recognized as important regulators of CSCs and EMT. In this article, we will discuss the state of our knowledge on the role of HIF-hypoxia signaling pathway and its kinship with CSCs and EMT within the tumor microenvironment. We will also discuss the potential role of hypoxia-induced microRNAs (miRNAs) in tumor development and aggressiveness, and finally discuss the potential effects of nutraceuticals on the biology of CSCs and EMT in the context of tumor hypoxia.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Asfar S. Azmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Shadan Ali
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Yiwei Li
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
78
|
All-trans retinoic acid protects renal tubular epithelial cells against hypoxia induced injury in vitro. Transplant Proc 2012; 45:497-502. [PMID: 23267795 DOI: 10.1016/j.transproceed.2012.02.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/14/2012] [Indexed: 01/23/2023]
Abstract
BACKGROUND It has been reported that the all-trans retinoic acid (atRA)-mediated protective effects in various cells are related to the inhibition of nuclear factor (NF)-κB activities. There exists some evidence that an increase in vascular endothelial growth factor (VEGF), which is expressed by proximal tubular epithelial cells and regulated by NFκB, may play a critical role in maintaining peritubular capillary endothelium in renal disease. By stimulating the production of VEGF, hypoxia is involved in tubulointerstitial fibrosis processes in various renal diseases. METHODS NRK52E cells survival rate was proportional to absorbance in dimethyl-thiazol-diphenyltetrazoliumbromide tests. Quantitative real-time polymerase chain reaction and Western blot were performed to assay the expression of VEGF, p65, and Scpep1. The activation of NFκB was determined by electrophoretic mobility shift assay. Co-immunoprecipitation analysis demonstrates that whether the Scpep1 and NFκB protein interacted. RESULTS We demonstrated that the hypoxia-mimicking agent CoCl2 triggered hypoxia injury of rat proximal tubular epithelial cells and significantly reduced cell viability. Addition of atRA increased the cell survival rate. Under CoCl(2)-mimicking hypoxic conditions, the expression of VEGF and p65 increased. The addition of atRA significantly attenuated the expression of VEGF and p65. There was a similar variation of NFκB/DNA binding activities. atRA not only activated distinct pathways to stimulate the expression of Scpep1, a retinoid-inducible gene, under normoxic conditions, but also acted as a CoCl(2)-mimicking hypoxia. CONCLUSION The protective effects of atRA against hypoxia-induced injury might be involved in suppression of VEGF expression via stimulating Scpep1 distinct pathways and inhibiting the NFκB pathway.
Collapse
|
79
|
Fujita N, Gogate SS, Chiba K, Toyama Y, Shapiro IM, Risbud MV. Prolyl hydroxylase 3 (PHD3) modulates catabolic effects of tumor necrosis factor-α (TNF-α) on cells of the nucleus pulposus through co-activation of nuclear factor κB (NF-κB)/p65 signaling. J Biol Chem 2012; 287:39942-53. [PMID: 22948157 DOI: 10.1074/jbc.m112.375964] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Recent studies suggest a differential role of prolyl hydroxylase (PHD) isoforms in controlling hypoxia-inducible factor (HIF)-α degradation and activity in nucleus pulposus (NP) cells. However, the regulation and function of PHDs under inflammatory conditions that characterize disc disease are not yet known. Here, we show that in NP cells, TNF-α and IL-1β induce PHD3 expression through NF-κB. Lentiviral delivery of Sh-p65 and Sh-IKKβ confirms that cytokine-mediated PHD3 expression is NF-κB-dependent. It is noteworthy that although both cytokines induce HIF activity, mechanistic studies using Sh-HIF-1α and PHD3 promoter/enhancer constructs harboring well characterized hypoxia response element (HRE) show lack of HIF involvement in cytokine-mediated PHD3 expression. Loss-of-function studies clearly indicate that PHD3 serves as a co-activator of NF-κB signaling activity in NP cells; PHD3 interacts with, and co-localizes with, p65. We observed that when PHD3 is silenced, there is a significant decrease in TNF-α-induced expression of catabolic markers that include ADAMTS5, syndecan4, MMP13, and COX2, and at the same time, there is restoration of aggrecan and collagen type II expression. It is noteworthy that hydroxylase function of PHDs is not required for mediating cytokine-dependent gene expression. These findings show that by enhancing the activity of inflammatory cytokines, PHD3 may serve a critical role in degenerative disc disease.
Collapse
Affiliation(s)
- Nobuyuki Fujita
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | |
Collapse
|
80
|
Vakili H, Jin Y, Cattini PA. Negative regulation of human growth hormone gene expression by insulin is dependent on hypoxia-inducible factor binding in primary non-tumor pituitary cells. J Biol Chem 2012; 287:33282-92. [PMID: 22833680 DOI: 10.1074/jbc.m112.380949] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Insulin controls growth hormone (GH) production at multiple levels, including via a direct effect on pituitary somatotrophs. There are no data, however, on the regulation of the intact human (h) GH gene (hGH1) by insulin in non-tumor pituitary cells, but the proximal promoter region (nucleotides -496/+1) responds negatively to insulin in transfected pituitary tumor cells. A DNA-protein interaction was also induced by insulin at nucleotides -308/-235. Here, we confirmed the presence of a hypoxia-inducible factor 1 (HIF-1) binding site within these sequences (-264/-259) and investigated whether HIF-1 is associated with insulin regulation of "endogenous" hGH1. In the absence of primary human pituitary cells, transgenic mice expressing the intact hGH locus in a somatotroph-specific manner were generated. A significant and dose-dependent decrease in hGH and mouse GH RNA levels was detected in primary pituitary cell cultures from these mice with insulin treatment. Increasing HIF-1α availability with a hypoxia mimetic significantly decreased hGH RNA levels and was accompanied by recruitment of HIF-1α to the hGH1 promoter in situ as seen with insulin. Both inhibition of HIF-1 DNA binding by echinomycin and RNA interference of HIF-1α synthesis blunted the negative effect of insulin on hGH1 but not mGH. The insulin response is also sensitive to histone deacetylase inhibition/trichostatin A and associated with a decrease in H3/H4 hyperacetylation in the proximal hGH1 promoter region. These data are consistent with HIF-1-dependent down-regulation of hGH1 by insulin via chromatin remodeling specifically in the proximal promoter region.
Collapse
Affiliation(s)
- Hana Vakili
- Department of Physiology, Division of Endocrine and Metabolic diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | |
Collapse
|
81
|
Hypoxia-inducible factor-1α and excision repair cross-complementing 1 in patients with small cell lung cancer who received front-line platinum-based chemotherapy: a retrospective study. J Thorac Oncol 2012; 7:528-34. [PMID: 22258474 DOI: 10.1097/jto.0b013e3182417830] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Hypoxia-inducible factor-1α (HIF-1α), which plays an essential role in the adaptive response of cells to hypoxia, is associated with aggressive tumor behavior. Furthermore, a relationship between excision repair cross-complementing 1 (ERCC1) expression and platinum resistance has been reported in patients with various malignancies. The aim of this study was to investigate the expression of HIF-1α and ERCC1 and to elucidate the clinical significance of their expression in patients with small cell lung cancer (SCLC) treated with front-line platinum-based chemotherapy. METHODS SCLC biopsy samples were obtained before front-line platinum-based chemotherapy from 111 patients with SCLC (limited disease, 29; extensive disease [ED], 82) between January 2002 and December 2009 at Gyeongsang National University Hospital. The expression levels of HIF-1α and ERCC1 were assessed by immunohistochemistry. RESULTS High expression levels of ERCC1 and HIF-1α were observed in 49 (44.1%) and 71 (64.0%) of 111 patients, respectively. Expression of ERCC1 and HIF-1α was not significantly associated with age, sex, Eastern Cooperative Oncology Group performance status, weight loss, or response to treatment, regardless of stage. In ED-SCLC, low expression in the HIF-1α group showed statistically better survival compared with high expression in the HIF-1α group (p = 0.018). Multivariate analysis revealed that response to front-line platinum-based chemotherapy (p < 0.001), good Eastern Cooperative Oncology Group performance status (0-1) (p = 0.002), and low expression of HIF-1α (p = 0.004) were independent predictors of better overall survival in ED-SCLC. CONCLUSIONS Low expression of HIF-1α may be a useful predictor of better overall survival in ED-SCLC patients treated with front-line platinum-based chemotherapy.
Collapse
|
82
|
Maione F, Capano S, Regano D, Zentilin L, Giacca M, Casanovas O, Bussolino F, Serini G, Giraudo E. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J Clin Invest 2012; 122:1832-48. [PMID: 22484816 DOI: 10.1172/jci58976] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 02/22/2012] [Indexed: 12/27/2022] Open
Abstract
Cancer development, progression, and metastasis are highly dependent on angiogenesis. The use of antiangiogenic drugs has been proposed as a novel strategy to interfere with tumor growth, but cancer cells respond by developing strategies to escape these treatments. In particular, animal models show that antiangiogenic drugs currently used in clinical settings reduce tumor tissue oxygenation and trigger molecular events that foster cancer resistance to therapy. Here, we show that semaphorin 3A (Sema3A) expression overcomes the proinvasive and prometastatic resistance observed upon angiogenesis reduction by the small-molecule tyrosine inhibitor sunitinib in both pancreatic neuroendocrine tumors (PNETs) in RIP-Tag2 mice and cervical carcinomas in HPV16/E2 mice. By improving cancer tissue oxygenation and extending the normalization window, Sema3A counteracted sunitinib-induced activation of HIF-1α, Met tyrosine kinase receptor, epithelial-mesenchymal transition (EMT), and other hypoxia-dependent signaling pathways. Sema3A also reduced tumor hypoxia and halted cancer dissemination induced by DC101, a specific inhibitor of the VEGF pathway. As a result, reexpressing Sema3A in cancer cells converts metastatic PNETs and cervical carcinomas into benign lesions. We therefore suggest that this strategy could be developed to safely harnesses the therapeutic potential of the antiangiogenic treatment.
Collapse
Affiliation(s)
- Federica Maione
- Laboratory of Transgenic Mouse Models, University of Torino School of Medicine, Candiolo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Torres MP, Rachagani S, Purohit V, Pandey P, Joshi S, Moore ED, Johansson SL, Singh PK, Ganti AK, Batra SK. Graviola: a novel promising natural-derived drug that inhibits tumorigenicity and metastasis of pancreatic cancer cells in vitro and in vivo through altering cell metabolism. Cancer Lett 2012; 323:29-40. [PMID: 22475682 DOI: 10.1016/j.canlet.2012.03.031] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 02/25/2012] [Accepted: 03/26/2012] [Indexed: 02/07/2023]
Abstract
Pancreatic tumors are resistant to conventional chemotherapies. The present study was aimed at evaluating the potential of a novel plant-derived product as a therapeutic agent for pancreatic cancer (PC). The effects of an extract from the tropical tree Annona Muricata, commonly known as Graviola, was evaluated for cytotoxicity, cell metabolism, cancer-associated protein/gene expression, tumorigenicity, and metastatic properties of PC cells. Our experiments revealed that Graviola induced necrosis of PC cells by inhibiting cellular metabolism. The expression of molecules related to hypoxia and glycolysis in PC cells (i.e. HIF-1α, NF-κB, GLUT1, GLUT4, HKII, and LDHA) were downregulated in the presence of the extract. In vitro functional assays further confirmed the inhibition of tumorigenic properties of PC cells. Overall, the compounds that are naturally present in a Graviola extract inhibited multiple signaling pathways that regulate metabolism, cell cycle, survival, and metastatic properties in PC cells. Collectively, alterations in these parameters led to a decrease in tumorigenicity and metastasis of orthotopically implanted pancreatic tumors, indicating promising characteristics of the natural product against this lethal disease.
Collapse
Affiliation(s)
- María P Torres
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE 68198-5870, USA
| | | | - Vinee Purohit
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE 68198-5870, USA
| | - Poomy Pandey
- Department of Environmental, Agricultural & Occupational Health, Omaha, NE 68198-5870, USA
| | - Suhasini Joshi
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198-5870, USA
| | - Erik D Moore
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198-5870, USA
| | - Sonny L Johansson
- Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE 68198-5870, USA; Department of Pathology and Microbiology, Omaha, NE 68198-5870, USA
| | - Pankaj K Singh
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE 68198-5870, USA
| | - Apar K Ganti
- Department of Internal Medicine VA Nebraska-Western Iowa Health Care System and University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, Omaha, NE 68198-5870, USA; Department of Pathology and Microbiology, Omaha, NE 68198-5870, USA.
| |
Collapse
|
84
|
Partyka R, Gonciarz M, Jałowiecki P, Kokocińska D, Byrczek T. VEGF and metalloproteinase 2 (MMP 2) expression in gastric cancer tissue. Med Sci Monit 2012; 18:BR130-4. [PMID: 22460086 PMCID: PMC3560834 DOI: 10.12659/msm.882614] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Accepted: 04/26/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Neoplasms are the second leading cause of death in Poland after vessel diseases, despite the huge progress in medical sciences in the last 20 years. Recently, gastric cancer morbidity has decreased, but mortality is still at a high level. MATERIAL/METHODS Tissues from 24 patients with a histopathologically diagnosed mucosal and adenomucosal gastric cancer were tested. Patients were divided into 2 equal groups: patients without metastases (G1) and patients with metastases in the liver (G2). In all tested tissues of G1 and G2, the expression of VEGF (vascular endothelial growth factor) and metalloproteinase 2, respectively, were estimated. RESULTS Results revealed a statistically significant increase in the VEGF expression for G1 and G2 in relation to the margin (p1<0.001; p2<0.001). The increase of gene expression for VEGF did not significantly differ statistically in G1 and G2. The obtained results revealed a statistically significant difference in the increase of gene expression for MMP-2 in G1 in relation to the margin (p<0.05) and a very high one in G2 in relation to the average margin value (p<0.001). A highly statistically significant correlation was obtained for VEGF and MMP-2 in the tissue of patients with metastases (p<0.001; r=0.714). The highly elevated expression of MMP-2 in the tissue of gastric cancer in patients with metastases confirms its participation in the invasiveness of the neoplasmatic process. CONCLUSIONS The highly significant correlation between VEGF and MMP-2 suggests a connection between both mechanisms in the progression of gastric cancer.
Collapse
Affiliation(s)
- Robert Partyka
- Clinical Division of Anesthesiology and Intensive Treatment of the Department of Anesthesiology, Intensive Treatment and Emergency Medicine, Medical University of Silesia in Katowice, Katowice, Poland.
| | | | | | | | | |
Collapse
|
85
|
Manu KA, Shanmugam MK, Ramachandran L, Li F, Fong CW, Kumar AP, Tan P, Sethi G. First evidence that γ-tocotrienol inhibits the growth of human gastric cancer and chemosensitizes it to capecitabine in a xenograft mouse model through the modulation of NF-κB pathway. Clin Cancer Res 2012; 18:2220-9. [PMID: 22351692 DOI: 10.1158/1078-0432.ccr-11-2470] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Because of poor prognosis and development of resistance against chemotherapeutic drugs, the existing treatment modalities for gastric cancer are ineffective. Hence, novel agents that are safe and effective are urgently needed. Whether γ-tocotrienol can sensitize gastric cancer to capecitabine in vitro and in a xenograft mouse model was investigated. EXPERIMENTAL DESIGN The effect of γ-tocotrienol on proliferation of gastric cancer cell lines was examined by mitochondrial dye uptake assay, apoptosis by esterase staining, NF-κB activation by DNA-binding assay, and gene expression by Western blotting. The effect of γ-tocotrienol on the growth and chemosensitization was also examined in subcutaneously implanted tumors in nude mice. RESULTS γ-Tocotrienol inhibited the proliferation of various gastric cancer cell lines, potentiated the apoptotic effects of capecitabine, inhibited the constitutive activation of NF-κB, and suppressed the NF-κB-regulated expression of COX-2, cyclin D1, Bcl-2, CXCR4, VEGF, and matrix metalloproteinase-9 (MMP-9). In a xenograft model of human gastric cancer in nude mice, we found that administration of γ-tocotrienol alone (1 mg/kg body weight, intraperitoneally 3 times/wk) significantly suppressed the growth of the tumor and this effect was further enhanced by capecitabine. Both the markers of proliferation index Ki-67 and for microvessel density CD31 were downregulated in tumor tissue by the combination of capecitabine and γ-tocotrienol. As compared with vehicle control, γ-tocotrienol also suppressed the NF-κB activation and the expression of cyclin D1, COX-2, intercellular adhesion molecule-1 (ICAM-1), MMP-9, survivin, Bcl-xL, and XIAP. CONCLUSIONS Overall our results show that γ-tocotrienol can potentiate the effects of capecitabine through suppression of NF-κB-regulated markers of proliferation, invasion, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Kanjoormana A Manu
- Department of Pharmacology, Yong Loo Lin School of Medicine, Cancer Science Institute of Singapore, National University of Singapore
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Abstract
Gastric cancer is a main cause of cancer death worldwide. Despite the knowledge that Helicobacter pylori constitutes the main cause of gastric cancer, the mechanisms for gastric carcinogenesis are still elucidated. Cancer development and progression depend on the balance between cell survival and death signals. Common cell survival signaling pathways are activated by carcinogens as well as by inflammatory cytokines, which contribute substantially to tumorigenesis. Nuclear factor-kappaB (NF-κB) is a major cell survival signal. Recent studies with cell culture systems and animal models have certified the links between NF-κB and gastric carcinogenesis and resistance to chemotherapy. Inhibition of NF-κB activation can enhance the sensitivity of cancer cells to chemotherapeutic drugs. In this review, we summarize recent progress in understanding the role of the NF-κB pathway in gastric cancer development as well as in modulating NF-κB for gastric cancer prevention and therapy.
Collapse
|
87
|
Dabkeviciene D, Sasnauskiene A, Leman E, Kvietkauskaite R, Daugelaviciene N, Stankevicius V, Jurgelevicius V, Juodka B, Kirveliene V. mTHPC-mediated photodynamic treatment up-regulates the cytokines VEGF and IL-1alpha. Photochem Photobiol 2012; 88:432-9. [PMID: 22171990 DOI: 10.1111/j.1751-1097.2011.01062.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Photodynamic therapy (PDT) of cancer induces oxidative stress, which intervenes in the expression of cytokines by tumor cells. The cytokines might have either a positive or a negative impact on tumor eradication. Here, we studied the expression of cytokines vascular endothelial growth factor (VEGF) and interleukin-1alpha (IL-1alpha) in the human epidermoid carcinoma A-431 cells following m-tetra(3-hydroxyphenyl)-chlorin (mTHPC)-mediated PDT in vitro and assessed the IL-1alpha effect on VEGF expression. Quantitative polymerase chain reaction and enzyme-linked immunosorbent assay revealed the enhanced production of VEGF and IL-1alpha both on mRNA and protein levels by mTHPC-loaded cells after light exposure. The silencing of IL1A by small interfering RNA resulted in decreased production of IL-1alpha and a reduced amount of VEGF. Furthermore, exogenous recombinant IL-1alpha stimulated the VEGF expression after PDT. Thus, in addition to the cytotoxic action on the A-431 cells, mTHPC-mediated PDT stimulated the production of VEGF and IL-1alpha, and IL-1alpha contributed to the VEGF overexpression. These data establish IL-1alpha as a possible target of combined cancer treatment.
Collapse
Affiliation(s)
- Daiva Dabkeviciene
- Department of Biochemistry and Biophysics, Vilnius University, Vilnius, Lithuania
| | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Kuo WH, Shih CM, Lin CW, Cheng WE, Chen SC, Chen W, Lee YL. Association of hypoxia inducible factor-1α polymorphisms with susceptibility to non-small-cell lung cancer. Transl Res 2012; 159:42-50. [PMID: 22153809 DOI: 10.1016/j.trsl.2011.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 08/25/2011] [Accepted: 09/13/2011] [Indexed: 01/22/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) is a key regulator of cellular response to hypoxia and has been suggested to play an important role in tumorigenesis and metastasis. The aim of this study was to investigate the role of HIF-1α-1772 C/T (P582S) and -1790 G/A (A588T) polymorphisms in the susceptibility to and severity of non-small-cell lung cancer (NSCLC). Using a case-control study design and polymerase chain reaction-restriction fragment-length polymorphism (PCR-RFLP) analysis, the allele frequencies and genotype distributions of each single nucleotide polymorphism in 285 NSCLC cases and 300 gender-matched controls were compared. The distribution of the genotype frequencies of HIF-1α-1772 C/T and -1790 G/A were significantly different between the NSCLC and the controls. Logistic regression analysis revealed that higher odds ratios (ORs) for lung cancer were observed for individuals with HIF-1α-1772 T/T genotype against CC/CT genotypes (an OR of 4.04, 95% confidence interval [CI] = 2.02-8.08, P = 0.0001), and HIF-1α-1790 A/A genotype against GG/GA genotypes (an OR of 4.42, 95% CI 2.22-8.78, P < 0.0001). There were no relationship between HIF-1α-1772 C/T or -1790 G/A allele distribution and disease severity of NSCLC (P > 0.05). However, those patients carrying a HIF-1α-1772 T/T genotype or a HIF-1α-1790 A/A had a tendency toward inferior prognosis compared with other patients.
Collapse
Affiliation(s)
- Wu-Hsien Kuo
- Department of Medicine, Armed-Force Taichung General Hospital, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
89
|
Otjacques E, Binsfeld M, Noel A, Beguin Y, Cataldo D, Caers J. Biological aspects of angiogenesis in multiple myeloma. Int J Hematol 2011; 94:505-18. [PMID: 22086206 DOI: 10.1007/s12185-011-0963-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 10/17/2011] [Accepted: 10/20/2011] [Indexed: 11/26/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by the aberrant expansion of malignant plasma cells within the bone marrow (BM). One of the hallmarks of this disease is the close interaction between myeloma cells and neighboring cells within the BM. Angiogenesis, through the activation of endothelial cells, plays an essential role in MM biology. In the current review, we describe the angiogenesis process in MM by identifying the interacting cells, the pro- and anti-angiogenic cytokines modulated, and the extracellular matrix degrading proteases liable to participate in the pathophysiology. Finally, we highlight the impact of hypoxia (through hypoxia-inducible factor-1) and constitutive activation of nuclear factor-κB in this tumor-induced neo-vascularization.
Collapse
Affiliation(s)
- Eléonore Otjacques
- Laboratory of Hematology, GIGA-Research, University of Liège, Sart-Tilman, Belgium
| | | | | | | | | | | |
Collapse
|
90
|
Tammali R, Saxena A, Srivastava SK, Ramana KV. Aldose reductase inhibition prevents hypoxia-induced increase in hypoxia-inducible factor-1alpha (HIF-1alpha) and vascular endothelial growth factor (VEGF) by regulating 26 S proteasome-mediated protein degradation in human colon cancer cells. J Biol Chem 2011; 286:24089-100. [PMID: 21576240 PMCID: PMC3129190 DOI: 10.1074/jbc.m111.219733] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/10/2011] [Indexed: 01/28/2023] Open
Abstract
The development of intratumoral hypoxia, a hallmark of rapidly progressing solid tumors, renders tumor cells resistant to chemotherapy and radiation therapy. We have recently shown that inhibition of aldose reductase (AR), an enzyme that catalyzes the reduction of lipid aldehydes and their glutathione conjugates, prevents human colon cancer cell growth in culture as well as in nude mouse xenografts by inhibiting the NF-κB-dependent activation of oxidative stress-mediated inflammatory and carcinogenic markers. However, the role of AR in mediating hypoxic stress signals is not known. We therefore investigated the molecular mechanisms by which AR inhibition prevents the hypoxia-induced human colon cancer cells growth and invasion. Our results indicate that AR inhibition by the pharmacological inhibitor fidarestat or ablation by AR-specific siRNA prevents hypoxia-induced proliferation of HT29, SW480, and Caco-2 colon cancer cells. Furthermore, hypoxia-induced increase in the level of HIF-1α in colon cancer cells was significantly decreased by AR inhibition. During hypoxic conditions, treatment of HT29 cells with the AR inhibitor fidarestat significantly decreased the expression of vascular endothelial growth factor, a down target of HIF-1α, at both mRNA and protein levels and also prevented the activation of PI3K/AKT, GSK3β, Snail, and lysyl oxidase. Furthermore, inhibition of hypoxia-induced HIF-1α protein accumulation by AR inhibition was abolished in the presence of MG132, a potent inhibitor of the 26 S proteasome. In addition, AR inhibition also prevented the hypoxia-induced inflammatory molecules such as Cox-2 and PGE2 and expression of extracellular matrix proteins such as MMP2, vimentin, uPAR, and lysyl oxidase 2. In conclusion, our results indicate that AR mediates hypoxic signals, leading to tumor progression and invasion.
Collapse
Affiliation(s)
- Ravinder Tammali
- From the Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Ashish Saxena
- From the Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Satish K. Srivastava
- From the Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555
| | - Kota V. Ramana
- From the Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77555
| |
Collapse
|