1
|
Wang Q, Suo Y, Tian X. 5-Aminolaevulinic Acid-Mediated Photodynamic Therapy Combined with Tirapazamine Enhances Efficacy in Ovarian Cancer. Biomedicines 2025; 13:724. [PMID: 40149700 PMCID: PMC11939993 DOI: 10.3390/biomedicines13030724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Ovarian cancer is a common gynaecological malignancy. Photodynamic therapy (PDT) mediated by 5-aminolaevulinic acid (5-ALA-PDT) is widely used in clinical practice. However, hypoxia may impact the efficacy of this treatment. In the present study, we combined the bioreductively active drug tirapazamine (TPZ) with PDT to explore its potential in enhancing ovarian cancer cell death. Methods: A cell counting kit-8 assay was used to determine cytotoxicity under different intervention conditions. The distribution of protoporphyrin IX, a metabolite of 5-ALA, was observed using in vivo fluorescence imaging. The effect of the combined treatment was assessed by measuring changes in tumour size following the corresponding interventions and by haematoxylin and eosin staining of tumour tissues. Immunohistochemical staining was used to detect the expression levels of relevant proteins. Results: TPZ exhibited no cytotoxicity under normoxic conditions but was activated under hypoxic conditions, inducing cytotoxic effects that were enhanced when combined with PDT. Over time, protoporphyrin IX achieved systemic distribution, and high drug concentrations were maintained within the tumour. The combination therapy suppressed tumour growth, and pathological staining showed that necrotic tumour areas were significantly enlarged after treatment. The enhanced therapeutic effect may be attributable to the inhibition of the hypoxia-inducible factor-1α/vascular endothelial growth factor axis and PI3K/Akt/mTOR pathway. Conclusions: 5-ALA-PDT combined with TPZ can overcome both the hypoxic state of ovarian cancer tissues and the increased hypoxia induced by PDT, thereby inhibiting tumour growth.
Collapse
Affiliation(s)
- Qian Wang
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| | - Yuping Suo
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
- Department of Gynaecology and Obstetrics, Shanxi Provincial People’s Hospital, Taiyuan 030012, China
| | - Xiaojuan Tian
- Fifth Clinical Medical College, Shanxi Medical University, Taiyuan 030012, China; (Q.W.); (X.T.)
| |
Collapse
|
2
|
Li J, Xian L, Zhu Z, Wang Y, Zhang W, Zheng R, Xue W, Li J. Role of CELF2 in ferroptosis: Potential targets for cancer therapy (Review). Int J Mol Med 2023; 52:88. [PMID: 37594127 PMCID: PMC10500222 DOI: 10.3892/ijmm.2023.5291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023] Open
Abstract
Ferroptosis is a novel form of regulated cellular necrosis that plays a critical role in promoting cancer progression and developing drug resistance. The main characteristic of ferroptosis is iron‑dependent lipid peroxidation caused by excess intracellular levels of reactive oxygen species. CUGBP ELAV‑like family number 2 (CELF2) is an RNA‑binding protein that is downregulated in various types of cancer and is associated with poor patient prognoses. CELF2 can directly bind mRNA to a variety of ferroptosis control factors; however, direct evidence of the regulatory role of CELF2 in ferroptosis is currently limited. The aim of the present review was to summarise the findings of previous studies on CELF2 and its role in regulating cellular redox homeostasis. The present review may provide insight into the possible mechanisms through which CELF2 affects ferroptosis and to provide recommendations for future studies.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zifeng Zhu
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Wang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wenlei Zhang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruipeng Zheng
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wang Xue
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiarui Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
3
|
Aboouf MA, Thiersch M, Soliz J, Gassmann M, Schneider Gasser EM. The Brain at High Altitude: From Molecular Signaling to Cognitive Performance. Int J Mol Sci 2023; 24:10179. [PMID: 37373327 DOI: 10.3390/ijms241210179] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/13/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
The brain requires over one-fifth of the total body oxygen demand for normal functioning. At high altitude (HA), the lower atmospheric oxygen pressure inevitably challenges the brain, affecting voluntary spatial attention, cognitive processing, and attention speed after short-term, long-term, or lifespan exposure. Molecular responses to HA are controlled mainly by hypoxia-inducible factors. This review aims to summarize the cellular, metabolic, and functional alterations in the brain at HA with a focus on the role of hypoxia-inducible factors in controlling the hypoxic ventilatory response, neuronal survival, metabolism, neurogenesis, synaptogenesis, and plasticity.
Collapse
Affiliation(s)
- Mostafa A Aboouf
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Markus Thiersch
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Jorge Soliz
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC G1V 4G5, Canada
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Edith M Schneider Gasser
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zürich, 8057 Zurich, Switzerland
- Institute Universitaire de Cardiologie et de Pneumologie de Québec (IUCPQ), Faculty of Medicine, Université Laval, Québec, QC G1V 4G5, Canada
- Neuroscience Center Zurich, University of Zurich and ETH Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
4
|
Pu J, Huang Y, Fang Q, Wang J, Li W, Xu Z, Wu X, Lu Y, Wei H. Hypoxia-induced Fascin-1 upregulation is regulated by Akt/Rac1 axis and enhances malignant properties of liver cancer cells via mediating actin cytoskeleton rearrangement and Hippo/YAP activation. Cell Death Discov 2021; 7:385. [PMID: 34897283 PMCID: PMC8665929 DOI: 10.1038/s41420-021-00778-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/12/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
In solid tumors, hypoxia facilitates malignant progression of cancer cells by triggering epithelial-mesenchymal transition (EMT) and cancer stemness. Fascin-1, an actin-bundling protein, takes part in the formation of many actin-based cellular structures. In the present study, we explored the potential functions of hypoxia-induced upregulation of Fascin-1 in liver cancer. Transcriptome RNA-sequencing was conducted to identify hypoxia-related genes. The potential functions of Fascin-1 were evaluated by western blot, transwell migration and invasion assays, sphere-formation assay, tumor xenograft growth, gelatin zymography analysis, immunofluorescence, cell viability assay, soft agar assay, and flow cytometry. We found that Fascin-1 was upregulated by hypoxia in liver cancer cell lines, elevated in liver cancer patients and correlated with larger tumor size, lymph node metastasis, distant metastasis, and shorter overall survival. Knockdown of Fascin-1 suppressed migration, invasion, EMT, stemness, and tumor xenograft growth of liver cancer cells under both normoxia and hypoxia conditions, while forced Fascin-1 expression showed opposite effects. Moreover, hypoxia-induced upregulation of Fascin-1 was regulated by the Akt/Rac1 signaling, and inhibition of Akt/Rac1 signaling by EHop-016 and MK-2206 restrained migration, invasion, EMT, and stemness of liver cancer cells under hypoxia. Furthermore, Fascin-1 knockdown suppressed MMP-2 and MMP-9 expression, impaired actin cytoskeleton rearrangement, inactivated Hippo/YAP signaling, and increased Sorafenib sensitivity in liver cancer cells. Our study provided a novel insight of Fascin-1 in regulating migration, invasion, EMT, and stemness of liver cancer cells under normoxia and hypoxia conditions.
Collapse
Affiliation(s)
- Jian Pu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Youguan Huang
- Graduate College of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Quan Fang
- Graduate College of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Jianchu Wang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Wenchuan Li
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Zuoming Xu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Xianjian Wu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Yuan Lu
- Department of Hepatobiliary Surgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Huamei Wei
- Department of Pathology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| |
Collapse
|
5
|
Kumari M, Krishnamurthy PT, Sola P. Targeted Drug Therapy to Overcome Chemoresistance in Triple-negative Breast Cancer. Curr Cancer Drug Targets 2021; 20:559-572. [PMID: 32370716 DOI: 10.2174/1568009620666200506110850] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023]
Abstract
Triple-negative Breast Cancer (TNBC) is the most aggressive and prevailing breast cancer subtype. The chemotherapeutics used in the treatment of TNBC suffer from chemoresistance, dose-limiting toxicities and off-target side effects. As a result, conventional chemotherapeutics are unable to prevent tumor growth, metastasis and result in failure of therapy. Various new targets such as BCSCs surface markers (CD44, CD133, ALDH1), signaling pathways (IL-6/JAK/STAT3, notch), pro and anti-apoptotic proteins (Bcl-2, Bcl-xL, DR4, DR5), hypoxic factors (HIF-1α, HIF-2α) and drug efflux transporters (ABCC1, ABCG2 and ABCB1) have been exploited to treat TNBC. Further, to improve the efficacy and safety of conventional chemotherapeutics, researchers have tried to deliver anticancer agents specifically to the TNBCs using nanocarrier based drug delivery. In this review, an effort has been made to highlight the various factors responsible for the chemoresistance in TNBC, novel molecular targets of TNBC and nano-delivery systems employed to achieve sitespecific drug delivery to improve efficacy and reduce off-target side effects.
Collapse
Affiliation(s)
- Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, (A Constituent College of JSS Academy of Higher Education & Research), Ooty, Tamilnadu, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, (A Constituent College of JSS Academy of Higher Education & Research), Ooty, Tamilnadu, India
| | - Piyong Sola
- Department of Pharmacology, JSS College of Pharmacy, (A Constituent College of JSS Academy of Higher Education & Research), Ooty, Tamilnadu, India
| |
Collapse
|
6
|
Kierans SJ, Taylor CT. Regulation of glycolysis by the hypoxia-inducible factor (HIF): implications for cellular physiology. J Physiol 2020; 599:23-37. [PMID: 33006160 DOI: 10.1113/jp280572] [Citation(s) in RCA: 509] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
Under conditions of hypoxia, most eukaryotic cells can shift their primary metabolic strategy from predominantly mitochondrial respiration towards increased glycolysis to maintain ATP levels. This hypoxia-induced reprogramming of metabolism is key to satisfying cellular energetic requirements during acute hypoxic stress. At a transcriptional level, this metabolic switch can be regulated by several pathways including the hypoxia inducible factor-1α (HIF-1α) which induces an increased expression of glycolytic enzymes. While this increase in glycolytic flux is beneficial for maintaining bioenergetic homeostasis during hypoxia, the pathways mediating this increase can also be exploited by cancer cells to promote tumour survival and growth, an area which has been extensively studied. It has recently become appreciated that increased glycolytic metabolism in hypoxia may also have profound effects on cellular physiology in hypoxic immune and endothelial cells. Therefore, understanding the mechanisms central to mediating this reprogramming are of importance from both physiological and pathophysiological standpoints. In this review, we highlight the role of HIF-1α in the regulation of hypoxic glycolysis and its implications for physiological processes such as angiogenesis and immune cell effector function.
Collapse
Affiliation(s)
- S J Kierans
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - C T Taylor
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Ireland.,School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
7
|
Valvezan AJ, Manning BD. Molecular logic of mTORC1 signalling as a metabolic rheostat. Nat Metab 2019; 1:321-333. [PMID: 32694720 DOI: 10.1038/s42255-019-0038-7] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/24/2019] [Indexed: 02/05/2023]
Abstract
The protein kinase complex mechanistic target of rapamycin complex 1 (mTORC1) serves as a key conduit between growth signals and the metabolic processes underlying cell growth. The activation state of mTORC1 is controlled by intracellular nutrients and energy, as well as exogenous hormones and growth factors, thereby integrating local and systemic growth signals. Here we discuss the molecular logic of the mTORC1 signalling network and its importance in coupling growth signals to the control of cellular metabolism. After activation, mTORC1 promotes the conversion of available nutrients and energy into the major macromolecular species contributing to cellular mass, including proteins, nucleic acids and lipids, while suppressing the autophagic recycling of these macromolecules back into their nutrient constituents. Given that uncoupling of mTORC1 from its normal regulatory inputs contributes to many diseases-including cancer, genetic tumour syndromes, metabolic diseases, autoimmune diseases and neurological disorders-understanding the molecular logic of the mTORC1 network and how to modulate it may present therapeutic opportunities for treatment of a broad range of diseases and potentially even for the extension of lifespan.
Collapse
Affiliation(s)
- Alexander J Valvezan
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brendan D Manning
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
8
|
Xie Y, Shi X, Sheng K, Han G, Li W, Zhao Q, Jiang B, Feng J, Li J, Gu Y. PI3K/Akt signaling transduction pathway, erythropoiesis and glycolysis in hypoxia (Review). Mol Med Rep 2018; 19:783-791. [PMID: 30535469 PMCID: PMC6323245 DOI: 10.3892/mmr.2018.9713] [Citation(s) in RCA: 234] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
The purpose of this review is to summarize the research progress of PI3K/Akt signaling pathway in erythropoiesis and glycolysis. Phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) is activated by numerous genes and leads to protein kinase B (Akt) binding to the cell membrane, with the help of phosphoinositide-dependent kinase, in the PI3K/Akt signal transduction pathway. Threonine and serine phosphorylation contribute to Akt translocation from the cytoplasm to the nucleus and further mediates enzymatic biological effects, including those involved in cell proliferation, apoptosis inhibition, cell migration, vesicle transport and cell cancerous transformation. As a key downstream protein of the PI3K/Akt signaling pathway, hypoxia-inducible factor (HIF)-1 is closely associated with the concentration of oxygen in the environment. Maintaining stable levels of HIF-1 protein is critical under normoxic conditions; however, HIF-1 levels quickly increase under hypoxic conditions. HIF-1α is involved in the acute hypoxic response associated with erythropoietin, whereas HIF-2α is associated with the response to chronic hypoxia. Furthermore, PI3K/Akt can reduce the synthesis of glycogen and increase glycolysis. Inhibition of glycogen synthase kinase 3β activity by phosphorylation of its N-terminal serine increases accumulation of cyclin D1, which promotes the cell cycle and improves cell proliferation through the PI3K/Akt signaling pathway. The PI3K/Akt signaling pathway is closely associated with a variety of enzymatic biological effects and glucose metabolism.
Collapse
Affiliation(s)
- Youbang Xie
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Xuefeng Shi
- Department of Respiratory Medicine, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Kuo Sheng
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Guoxiong Han
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Wenqian Li
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Qiangqiang Zhao
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Baili Jiang
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Jianming Feng
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Jianping Li
- Department of Hematology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| | - Yuhai Gu
- Department of Respiratory Medicine, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, P.R. China
| |
Collapse
|
9
|
Sheng W, Mao H, Wang C, Yang N, Zhang Z, Han J. Dehydrocostus Lactone Enhances Chemotherapeutic Potential of Doxorubicin in Lung Cancer by Inducing Cell Death and Limiting Metastasis. Med Sci Monit 2018; 24:7850-7861. [PMID: 30388099 PMCID: PMC6228117 DOI: 10.12659/msm.911410] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Natural compounds have been utilized in inhibiting metastasis alone or in combination with other anti-tumor agents. Dehydrocostus lactone (DHC), a natural sesquiterpene lactone, was used to investigate its effect on proliferation of lung cancer cells and on the anti-angiogenic efficacy of doxorubicin. MATERIAL AND METHODS Cell proliferation was assessed by MTT assay and clonogenic assay. Apoptosis and migration were assessed by flow cytometry and wound-healing assay, respectively. Western blotting and qPCR were performed for gene and protein expression analysis. Matrigel plug assay was performed for angiogenesis assessment. RESULTS Results of the study show that DHC inhibited the survival and proliferation of lung cancer cells (A549 and H460) and enhanced the growth-inhibitory properties of DOX. Cotreatment of DHC enhanced the apoptosis-inducing effects of DOX by activating caspase-9 and caspase-3 followed by cleavage of PARP. Treatment of A549 and H460 cells with DHC caused suppression of HIF-1α, Akt and pAkt, GSK-3β and pGSK-3β, as well as ERK, pERK, mTOR, and p-mTOR. DHC enhanced the effect of DOX by inhibiting migration of A549 cells as observed by wound-healing assay. DHC caused synergistic inhibition of MMP-2 and MMP-9 genes when treated in combination with DOX. DHC further enhanced the anti-angiogenic properties of DOX in mice implanted with Matrigel plugs. DHC suppressed the proliferation of lung cancer cells and enhanced the anti-angiogenic properties of DOX. CONCLUSIONS The putative mechanism behind the metastasis-limiting effects of DHC may involve the suppression of Akt/GSK-3β and inhibition of MMP-2 and MMP-9 in lung cancer cells.
Collapse
Affiliation(s)
- Wei Sheng
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Hongyan Mao
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Chuanxi Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Ning Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Zhe Zhang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| | - Junqing Han
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
10
|
Zhang XR, Wang SY, Sun W, Wei C. Isoliquiritigenin inhibits proliferation and metastasis of MKN28 gastric cancer cells by suppressing the PI3K/AKT/mTOR signaling pathway. Mol Med Rep 2018; 18:3429-3436. [PMID: 30066879 DOI: 10.3892/mmr.2018.9318] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 06/19/2018] [Indexed: 11/06/2022] Open
Abstract
Isoliquiritigenin (ISL) is a flavonoid extracted from licorice root, which is known to serve important antitumor roles in numerous types of cancers; however, its effect on gastric cancer remains to be elucidated. The present study aimed to explore the roles and underlying mechanisms of ISL in MKN28 gastric cancer cells. MKN28 cell proliferation was measured using the Cell Counting Kit‑8 (CCK8) assay. A Transwell assay was used to determine the effects of ISL on the migration and invasion of MKN28 cells. Apoptosis was assessed by flow cytometry, and the expression levels of apoptosis‑, autophagy‑ and signaling pathway‑related proteins were detected by western blot analysis. The results of the CCK8 assay demonstrated that ISL significantly inhibited the proliferation of MKN28 cells (P<0.05). Transwell assays demonstrated that the migration and invasion of MKN28 cells were significantly inhibited following treatment with ISL (P<0.05). Flow cytometric analysis indicated that ISL induced apoptosis of MKN28 cells. In addition, western blot analysis revealed that the ratio of microtubule‑associated proteins 1A/1B light chain 3B (LC3)II/LC3I was upregulated, as was Beclin 1 expression; however, p62 was downregulated following ISL pretreatment, thus suggesting that ISL triggered autophagy in MKN28 cells. In addition, the phosphorylation levels of protein kinase B (AKT) and mammalian target of rapamycin (mTOR) were significantly reduced following ISL treatment. These results indicated that ISL may influence apoptosis and autophagy in MKN28 cells by suppressing the phosphoinositide 3‑kinase/AKT/mTOR signaling pathway. In conclusion, the findings of the present study suggested that ISL may inhibit MKN28 cell proliferation, migration and invasion by inducing apoptosis and autophagy, implying potential as a therapeutic agent for gastric cancer.
Collapse
Affiliation(s)
- Xiu-Rong Zhang
- Department of Traditional Chinese Medicine, Maternal and Child Health Care of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Shi-Yao Wang
- Department of Chinese Medicine, The Second Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong 510000, P.R. China
| | - Wen Sun
- Department of Research, Beijing Splinger Medical Research Institute, Jinan, Shandong 250021, P.R. China
| | - Chao Wei
- Department of Ophthalmology, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
11
|
Zhang Y, Liu Y, Zou J, Yan L, Du W, Zhang Y, Sun H, Lu P, Geng S, Gu R, Zhang H, Bi Z. Tetrahydrocurcumin induces mesenchymal-epithelial transition and suppresses angiogenesis by targeting HIF-1α and autophagy in human osteosarcoma. Oncotarget 2017; 8:91134-91149. [PMID: 29207631 PMCID: PMC5710911 DOI: 10.18632/oncotarget.19845] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/24/2017] [Indexed: 01/10/2023] Open
Abstract
Human osteosarcoma is considered a malignant tumor with poor prognosis that readily metastasizes. Tetrahydrocurcumin (THC) has been reported to have anti-tumor activity in numerous tumors. In addition, hypoxia-inducible factor-1α (HIF-1α) has been demonstrated to be associated with tumor metastasis by regulating epithelial-mesenchymal transition (EMT). However, the role of THC in osteosarcoma remains uncertain. Therefore, this study aimed to elucidate the potential mechanisms. We found that THC significantly reduced the growth of osteosarcoma cells and suppressed migration and invasion, as tested in a nude mouse lung metastasis model. Additionally, the mesenchymal-epithelial transition (MET) process was facilitated by THC. Mechanistically, our study showed that HIF-1α had a pivotal role in the anti-metastatic effect of THC. Importantly, HIF-1α expression was downregulated by THC by inhibiting Akt/mTOR and p38 MAPK pathways. Moreover, THC exhibited a remarkable inhibitory effect on HIF-1α expression and angiogenesis under hypoxic conditions. Furthermore, THC activated autophagy and induced MET and suppressed angiogenesis in a HIF-1α-related manner. Taken together, our findings suggest that THC suppresses metastasis and invasion and this may be associated with HIF-1α and autophagy, which would potentially provide therapeutic strategies for human osteosarcoma.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, P.R. China
| | - Ying Liu
- Department of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China.,Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Jilong Zou
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, P.R. China
| | - Lixin Yan
- Department of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China.,Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Wei Du
- School of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang, P.R. China
| | - Yafeng Zhang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Hanliang Sun
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Peng Lu
- Department of Orthopaedics, Baoquanling Central Hospital, Baoquanling, Heilongjiang, P.R. China
| | - Shuo Geng
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, P.R. China
| | - Rui Gu
- Department of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China.,Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Hongyue Zhang
- Department of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P.R. China.,Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang, P.R. China
| | - Zhenggang Bi
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Nangang District, Harbin, P.R. China
| |
Collapse
|
12
|
Narayanan S, Loganathan G, Dhanasekaran M, Tucker W, Patel A, Subhashree V, Mokshagundam S, Hughes MG, Williams SK, Balamurugan AN. Intra-islet endothelial cell and β-cell crosstalk: Implication for islet cell transplantation. World J Transplant 2017; 7:117-128. [PMID: 28507914 PMCID: PMC5409911 DOI: 10.5500/wjt.v7.i2.117] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/28/2017] [Accepted: 03/24/2017] [Indexed: 02/05/2023] Open
Abstract
The intra-islet microvasculature is a critical interface between the blood and islet endocrine cells governing a number of cellular and pathophysiological processes associated with the pancreatic tissue. A growing body of evidence indicates a strong functional and physical interdependency of β-cells with endothelial cells (ECs), the building blocks of islet microvasculature. Intra-islet ECs, actively regulate vascular permeability and appear to play a role in fine-tuning blood glucose sensing and regulation. These cells also tend to behave as “guardians”, controlling the expression and movement of a number of important immune mediators, thereby strongly contributing to the physiology of islets. This review will focus on the molecular signalling and crosstalk between the intra-islet ECs and β-cells and how their relationship can be a potential target for intervention strategies in islet pathology and islet transplantation.
Collapse
|
13
|
Hicks KC, Patel TB. Sprouty2 Protein Regulates Hypoxia-inducible Factor-α (HIFα) Protein Levels and Transcription of HIFα-responsive Genes. J Biol Chem 2016; 291:16787-801. [PMID: 27281823 DOI: 10.1074/jbc.m116.714139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Indexed: 12/14/2022] Open
Abstract
The α-subunits of hypoxia-inducible factors (HIF1α and HIF2α) promote transcription of genes that regulate glycolysis and cell survival and growth. Sprouty2 (Spry2) is a modulator of receptor tyrosine kinase signaling and inhibits cell proliferation by a number of different mechanisms. Because of the seemingly opposite actions of HIFα subunits and Spry2 on cellular processes, we investigated whether Spry2 regulates the levels of HIF1α and HIF2α proteins. In cell lines from different types of tumors in which the decreased protein levels of Spry2 have been associated with poor prognosis, silencing of Spry2 elevated HIF1α protein levels. Increases in HIF1α and HIF2α protein levels due to silencing of Spry2 also up-regulated HIFα target genes. Using HIF1α as a prototype, we show that Spry2 decreases HIF1α stability and enhances the ubiquitylation of HIF1α by a von Hippel-Lindau protein (pVHL)-dependent mechanism. Spry2 also exists in a complex with HIF1α. Because Spry2 can also associate with pVHL, using a mutant form of Spry2 (3P/3A-Spry2) that binds HIF1α, but not pVHL, we show that WT-Spry2, but not the 3P/3A-Spry2 decreases HIF1α protein levels. In accordance, expression of WT-Spry2, but not 3P/3A-Spry2 results in a decrease in HIF1α-sensitive glucose uptake. Together our data suggest that Spry2 acts as a scaffold to bring more pVHL/associated E3 ligase in proximity of HIF1α and increase its ubiquitylation and degradation. This represents a novel action for Spry2 in modulating biological processes regulated by HIFα subunits.
Collapse
Affiliation(s)
- Kristin C Hicks
- From the Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, Illinois 60153, and
| | - Tarun B Patel
- the Albany College of Pharmacy and Health Sciences, Albany, New York 12208
| |
Collapse
|
14
|
Interaction between hypoxia, AKT and HIF-1 signaling in HNSCC and NSCLC: implications for future treatment strategies. Future Sci OA 2016; 2:FSO84. [PMID: 28031935 PMCID: PMC5137923 DOI: 10.4155/fso.15.84] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/03/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Hypoxia is a negative prognostic factor and this study investigated the relationship between hypoxia, hypoxia inducible factor 1 (HIF-1) and AKT signaling in head and neck squamous cell carcinoma (HNSCC) and non-small-cell lung cancer (NSCLC). RESULTS/METHODOLOGY pAKT was induced by hypoxia (0.5% O2) in a part of HNSCC (3/4) and squamous (2/3) and adenocarcinoma (1/3) NSCLS lines. AKT-inhibitor MK-2206 reduced hypoxic HIF-1 signaling in most HNSCC cell lines. This reduction did not correlate with hypoxic induction of pAKT or with sensitivity to MK-2206 under hypoxia. Patient biopsies revealed a hypoxia-induced expression pattern of pAKT in HNSCC (n = 16), which was not observed in squamous cell (n = 34) or adenocarcinoma (n = 41) NSCLC. CONCLUSION The interaction between hypoxia, HIF-1 and AKT signaling varies between tumor types and histologies, which could significantly affect response to targeted therapies.
Collapse
|
15
|
Oxidative Dimerization of PHD2 is Responsible for its Inactivation and Contributes to Metabolic Reprogramming via HIF-1α Activation. Sci Rep 2016; 6:18928. [PMID: 26740011 PMCID: PMC4703963 DOI: 10.1038/srep18928] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/30/2015] [Indexed: 12/20/2022] Open
Abstract
Prolyl hydroxylase domain protein 2 (PHD2) belongs to an evolutionarily conserved superfamily of 2-oxoglutarate and Fe(II)-dependent dioxygenases that mediates homeostatic responses to oxygen deprivation by mediating hypoxia-inducible factor-1α (HIF-1α) hydroxylation and degradation. Although oxidative stress contributes to the inactivation of PHD2, the precise molecular mechanism of PHD2 inactivation independent of the levels of co-factors is not understood. Here, we identified disulfide bond-mediated PHD2 homo-dimer formation in response to oxidative stress caused by oxidizing agents and oncogenic H-rasV12 signalling. Cysteine residues in the double-stranded β-helix fold that constitutes the catalytic site of PHD isoforms appeared responsible for the oxidative dimerization. Furthermore, we demonstrated that disulfide bond-mediated PHD2 dimerization is associated with the stabilization and activation of HIF-1α under oxidative stress. Oncogenic H-rasV12 signalling facilitates the accumulation of HIF-1α in the nucleus and promotes aerobic glycolysis and lactate production. Moreover, oncogenic H-rasV12 does not trigger aerobic glycolysis in antioxidant-treated or PHD2 knocked-down cells, suggesting the participation of the ROS-mediated PHD2 inactivation in the oncogenic H-rasV12-mediated metabolic reprogramming. We provide here a better understanding of the mechanism by which disulfide bond-mediated PHD2 dimerization and inactivation result in the activation of HIF-1α and aerobic glycolysis in response to oxidative stress.
Collapse
|
16
|
Chaudagar KK, Mehta AA. Effect of atorvastatin on the angiogenic responsiveness of coronary endothelial cells in normal and streptozotocin (STZ) induced diabetic rats. Can J Physiol Pharmacol 2014; 92:338-49. [PMID: 24708217 DOI: 10.1139/cjpp-2013-0391] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atorvastatin, a lipid lowering agent, possesses various pleiotropic vasculoprotective effects, but its role in coronary angiogenesis is still controversial. Our objective was to study the effects of atorvastatin on the angiogenic responsiveness of coronary endothelial cells (cEC) from normal and diabetic rats. Male Wistar rats were distributed among 9 groups; (i) normal rats, (ii) 30 day diabetic rats, (iii) 60 day diabetic rats, (iv) normal rats administered a low dose of atorvastatin (1 mg/kg body mass, per oral (p.o.), for 15 days); (v) 30 day diabetic rats administered a low dose of atorvastatin; (vi) 60 day diabetic rats administered a low dose of atorvastatin; (vii) normal rats administered a high dose of atorvastatin (5 mg/kg, p.o., for 15 days); (viii) 30 day diabetic rats administered a high dose of atorvastatin; (ix) 60 day diabetic rats administered a high dose of atorvastatin. Each group was further divided into 2 subgroups, (i) sham ischemia-reperfusion and (ii) rats hearts that underwent ischemia-reperfusion. Angiogenic responsiveness the and nitric oxide (NO) releasing properties of the subgroups of cECs were studied using a chorioallantoic membrane assay and the Griess method, respectively. Atorvastatin treatment significantly increased VEGF-induced angiogenic responsiveness and the NO-releasing properties of cECs from all of the subgroups, compared with their respective non-treated subgroups except for the late-phase diabetic rat hearts that underwent ischemia-reperfusion, and the high dose of atorvastatin treatment groups. These effects of atorvastatin were significantly inhibited by pretreatment of cECs with l-NAME, wortmannin, and chelerythrine. Thus, treatment with a low dose of atorvastatin improves the angiogenic responsiveness of the cECs from normal and diabetic rats, in the presence of VEGF, via activation of eNOS-NO release.
Collapse
Affiliation(s)
- Kiranj K Chaudagar
- a Department of Pharmacology, L.M. College of Pharmacy, Opp. Gujarat University, Navarangpura, Ahmedabad 380009, India
| | | |
Collapse
|
17
|
Joshi S, Singh AR, Zulcic M, Durden DL. A macrophage-dominant PI3K isoform controls hypoxia-induced HIF1α and HIF2α stability and tumor growth, angiogenesis, and metastasis. Mol Cancer Res 2014; 12:1520-31. [PMID: 25103499 DOI: 10.1158/1541-7786.mcr-13-0682] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Tumor growth, progression, and response to the hypoxic tumor microenvironment involve the action of hypoxia-inducible transcription factors, HIF1 and HIF2. HIF is a heterodimeric transcription factor containing an inducible HIFα subunit and a constitutively expressed HIFβ subunit. The signaling pathways operational in macrophages regulating hypoxia-induced HIFα stabilization remain the subject of intense investigation. Here, it was discovered that the PTEN/PI3K/AKT signaling axis controls hypoxia-induced HIF1α (HIF1A) and HIF2α (EPAS1) stability in macrophages. Using genetic mouse models and pan-PI3K as well as isoform-specific inhibitors, inhibition of the PI3K/AKT pathway blocked the accumulation of HIFα protein and its primary transcriptional target VEGF in response to hypoxia. Moreover, blocking the PI3K/AKT signaling axis promoted the hypoxic degradation of HIFα via the 26S proteasome. Mechanistically, a macrophage-dominant PI3K isoform (p110γ) directed tumor growth, angiogenesis, metastasis, and the HIFα/VEGF axis. Moreover, a pan-PI3K inhibitor (SF1126) blocked tumor-induced angiogenesis and inhibited VEGF and other proangiogenic factors secreted by macrophages. These data define a novel molecular mechanism by which PTEN/PI3K/AKT regulates the proteasome-dependent stability of HIFα under hypoxic conditions, a signaling pathway in macrophages that controls tumor-induced angiogenesis and metastasis. IMPLICATIONS This study indicates that PI3K inhibitors are excellent candidates for the treatment of cancers where macrophages promote tumor progression.
Collapse
Affiliation(s)
- Shweta Joshi
- UCSD Department of Pediatrics, Moores Cancer Center, University of California, La Jolla, California
| | - Alok R Singh
- UCSD Department of Pediatrics, Moores Cancer Center, University of California, La Jolla, California
| | - Muamera Zulcic
- UCSD Department of Pediatrics, Moores Cancer Center, University of California, La Jolla, California
| | - Donald L Durden
- UCSD Department of Pediatrics, Moores Cancer Center, University of California, La Jolla, California. Division of Pediatric Hematology-Oncology, UCSD Rady Children's Hospital, San Diego, California. SignalRx Pharmaceuticals, San Diego, California.
| |
Collapse
|
18
|
Secades P, de Santa-María IS, Merlo A, Suarez C, Chiara MD. In vitro study of normoxic epidermal growth factor receptor-induced hypoxia-inducible factor-1-alpha, vascular endothelial growth factor, and BNIP3 expression in head and neck squamous cell carcinoma cell lines: Implications for anti-epidermal growth factor receptor therapy. Head Neck 2014; 37:1150-62. [PMID: 24798801 DOI: 10.1002/hed.23733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 02/26/2014] [Accepted: 04/28/2014] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We previously showed that activation of epidermal growth factor receptor (EGFR) induces hypoxia inducible factor-1α (HIF-1α) in head and neck squamous cell carcinoma (HNSCC) cells. In this study, we have furthered this by investigating the mechanism of HIF-1α activation by epidermal growth factor (EGF) and its association with the sensitivity to gefitinib. METHODS EGFR/HIF-1α signaling was tested by immunoblot, polymerase chain reaction (PCR), cell proliferation, and apoptosis assays. RESULTS HIF-1α accumulated in cells overexpressing EGF and phosphorylated epidermal growth factor receptor (pEGFR), phosphatidylinositol-3-kinase (pPI3K), and mitogen-activated protein kinase (pMAPK). EGF-induced expression of HIF-1α and its targets, vascular endothelial growth factor (VEGF) and BNIP3, were blocked by gefitinib and PI3K-inhibitors and MAPK-inhibitors. HIF-1α-siRNAs abrogated EGF-induced BNIP3 but not VEGF expression. Gefitinib inhibited cell proliferation and induced apoptosis more strongly in cells with constitutively active EGFR/HIF-1α signaling than in cells lacking activation of these pathways. HIF-1α-siRNA treatment reduced sensitivity to gefitinib. CONCLUSION The search for molecular predictors of sensitivity to gefitinib in HNSCC should be extended to the activation status of EGFR-downstream pathways, phosphorylated protein kinase B, pMAPK, and HIF-1α.
Collapse
Affiliation(s)
- Pablo Secades
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Inés Saenz de Santa-María
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Anna Merlo
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - Carlos Suarez
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| | - María-Dolores Chiara
- Servicio de Otorrinolaringología, Hospital Universitario Central de Asturias, Instituto Universitario de Oncología del Principado de Asturias, Universidad de Oviedo, Oviedo, Spain
| |
Collapse
|
19
|
Joshi S, Singh AR, Durden DL. MDM2 regulates hypoxic hypoxia-inducible factor 1α stability in an E3 ligase, proteasome, and PTEN-phosphatidylinositol 3-kinase-AKT-dependent manner. J Biol Chem 2014; 289:22785-22797. [PMID: 24982421 DOI: 10.1074/jbc.m114.587493] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia-inducible factor 1 (HIF1) is a heterodimeric transcription factor containing an inducibly expressed HIF1α subunit and a constitutively expressed HIF1β subunit. Under hypoxic conditions, the HIF1α subunit accumulates because of a decrease in the rate of proteolytic degradation, and the resulting HIF1α-HIF1β heterodimers undergo post-translational modifications that promote transactivation. Previous reports suggest that amplified signaling through PI3K enhances HIF1-dependent gene expression; however, its role is controversial, and the mechanism is unclear. Using genetically engineered PTEN-deficient cell lines, we demonstrate that PTEN specifically inhibited the accumulation of HIF1α in response to hypoxia. Furthermore, we report that in glioblastoma cell lines, inhibition of PI3K pathway, using pan as well as isoform-specific PI3K inhibitors SF1126, PF4691502, BEZ-235, GDC0941, and TGX221 blocked the induction of HIF1α protein and its targets vascular endothelial growth factor, HK1, and GLUT1 mRNA in response to hypoxia. Herein, we describe the first evidence that HIF1α can be degraded under hypoxic conditions via the 26 S proteasome and that MDM2 is the E3 ligase that induces the hypoxic degradation of HIF1α. Moreover, the action of MDM2 on HIF1α under hypoxia occurs in the cytoplasm and is controlled by the PTEN-PI3K-AKT signaling axis. These data strongly suggest a new role for PTEN in the regulation of HIF1α and importantly that PI3K-AKT activation is required for the hypoxic stabilization of HIF1α and that hypoxia alone is not sufficient to render HIF1α resistant to proteasomal cleavage and degradation. Moreover, these findings suggest new therapeutic considerations for PI3K and/or AKT inhibitors for cancer therapeutics.
Collapse
Affiliation(s)
- Shweta Joshi
- Department of Pediatrics, Moores Cancer Center, UC San Diego Health System, La Jolla, California 92093 and
| | - Alok R Singh
- Department of Pediatrics, Moores Cancer Center, UC San Diego Health System, La Jolla, California 92093 and
| | - Donald L Durden
- Department of Pediatrics, Moores Cancer Center, UC San Diego Health System, La Jolla, California 92093 and; University of California at San Diego Rady Children's Hospital, San Diego, California 92123.
| |
Collapse
|
20
|
Schaible EV, Windschügl J, Bobkiewicz W, Kaburov Y, Dangel L, Krämer T, Huang C, Sebastiani A, Luh C, Werner C, Engelhard K, Thal SC, Schäfer MK. 2-Methoxyestradiol confers neuroprotection and inhibits a maladaptive HIF-1α response after traumatic brain injury in mice. J Neurochem 2014; 129:940-54. [DOI: 10.1111/jnc.12708] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 02/27/2014] [Accepted: 03/05/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Eva-Verena Schaible
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Julia Windschügl
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Wiesia Bobkiewicz
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Yordan Kaburov
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Larissa Dangel
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Tobias Krämer
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Changsheng Huang
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Anne Sebastiani
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Clara Luh
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Christian Werner
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Kristin Engelhard
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| | - Serge C. Thal
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
| | - Michael K.E. Schäfer
- Department of Anesthesiology; University Medical Center; Johannes Gutenberg-University; Mainz Germany
- Focus Program Translational Neuroscience (FTN); Johannes Gutenberg-University; Mainz Germany
| |
Collapse
|
21
|
Sui W, Zhang Y, Wang Z, Wang Z, Jia Q, Wu L, Zhang W. Antitumor effect of a selective COX-2 inhibitor, celecoxib, may be attributed to angiogenesis inhibition through modulating the PTEN/PI3K/Akt/HIF-1 pathway in an H₂₂ murine hepatocarcinoma model. Oncol Rep 2014; 31:2252-60. [PMID: 24647425 DOI: 10.3892/or.2014.3093] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 02/04/2014] [Indexed: 11/06/2022] Open
Abstract
Celecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor, has recently been shown to affect the development of different types of cancer. The present study utilized a murine H22 hepatocarcinoma model to investigate the molecular mechanisms involved in celecoxib-induced inhibition of tumor angiogenesis. Tumor-bearing mice were randomly divided into five groups: i) control; ii) low-dose celecoxib (50 mg/kg); iii) high-dose celecoxib (200 mg/kg); iv) 5-fluorouracil (5-FU), (20 mg/kg) and v) combination of 5-FU and celecoxib (50 mg/kg). The antitumor effect of celecoxib was determined by measuring tumor volume. Tumor angiogenesis was evaluated by microvessel density (MVD). Tumor histology and immunostaining for CD34 in endothelial cells were performed to detect MVD. The expression levels of phosphatase and tensin homologue deleted from chromosome 10 (PTEN), phosphatidylinositol 3-kinase (PI3K), phospho‑Akt (P-Akt), COX-2, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor-A (VEGF-A) were detected by ELISA, immunohistochemistry and western blotting, respectively. We discovered substantial growth delay in murine H22 hepatoma as a result of celecoxib treatment. The inhibition rate of tumor growth induced by high-dose and low-dose celecoxib was 49.3 and 37.0%, respectively (P<0.05). The expression of PI3K, P-Akt, COX-2, HIF-1α, VEGF-A and PTEN in tumor tissues treated with celecoxib was demonstrated by immunohistochemistry, and the MVD was decreased in a dose-dependent manner (P<0.05). Reduced PI3K and P-Akt was particularly apparent in the high-dose celecoxib group (P<0.05). ELISA and western blotting data showed that the expression of PI3K, P-Akt, COX-2, HIF-1α and VEGF-A were reduced and PTEN was increased after treatment with celecoxib. In conclusion, the impact of celecoxib-induced tumor growth delay of murine H22 hepatocarcinoma may correlate with the inhibition of angiogenesis by reducing PI3K, P-Akt, COX-2, HIF-1α and VEGF-A expression and increasing PTEN expression in tumor tissue.
Collapse
Affiliation(s)
- Wenwen Sui
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Yueying Zhang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Zhaopeng Wang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Zhaoxia Wang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Qing Jia
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories and Division of Thoracic Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Weidong Zhang
- Key Laboratory for Modern Medicine and Technology of Shandong Province, Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong, P.R. China
| |
Collapse
|
22
|
Chaudagar KK, Mehta AA. Effect of telmisartan on VEGF-induced and VEGF-independent angiogenic responsiveness of coronary endothelial cells in normal and streptozotocin (STZ)-induced diabetic rats. Clin Exp Hypertens 2014; 36:557-66. [DOI: 10.3109/10641963.2014.881842] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
Muh CR, Joshi S, Singh AR, Kesari S, Durden DL, Makale MT. PTEN status mediates 2ME2 anti-tumor efficacy in preclinical glioblastoma models: role of HIF1α suppression. J Neurooncol 2013; 116:89-97. [PMID: 24162827 DOI: 10.1007/s11060-013-1283-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/13/2013] [Indexed: 10/26/2022]
Abstract
Glioblastoma (GBM) is the most common brain cancer and is highly lethal in both adults and children. 2-methoxyestradiol (2ME2) is a microtubule inhibitor that potently inhibits HIF1α, GBM angiogenesis and tumor growth in preclinical models. In patients, 2ME2 exhibits low toxicity and promising but inconsistent efficacy. Given its preclinical potency and its tolerability in patients, we sought to determine whether 2ME2 therapy could be enhanced by addressing resistance via combination therapy, and with biomarkers to identify responsive glioma subgroups. We demonstrate that the PTEN-PI3K axis regulates HIF1α in glioma models. We utilized isogenic-pairs of glioma cell lines, deficient in PTEN or stably reconstituted with PTEN, to determine the role of PTEN in 2ME2 sensitivity in vitro and in vivo. Chou-Talalay synergy studies reveal significant synergy when a pan-PI3K inhibitor is combined with 2ME2. This synergistic activity was correlated with a synergistic suppression of HIF1α accumulation under hypoxic conditions in glioma models. In vivo, 2ME2 markedly inhibited tumor-induced angiogenesis and significantly reduced tumor growth only in a PTEN reconstituted GBM models in both subcutaneous and orthotopic intracranial mouse models. Collectively, these results: (1) suggest that PTEN status predicts sensitivity to 2ME2 and (2) justify exploration of 2ME2 combined with pan-PI3K inhibitors for the treatment of this intractable brain cancer.
Collapse
Affiliation(s)
- Carrie R Muh
- Department of Neurosurgery and Pediatrics, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | | | |
Collapse
|
24
|
Validation of an Engineered Cell Model for In Vitro and In Vivo HIF-1α Evaluation by Different Imaging Modalities. Mol Imaging Biol 2013; 16:210-23. [DOI: 10.1007/s11307-013-0669-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
25
|
Zhang S, Sun Y, Yuan Z, Li Y, Li X, Gong Z, Peng Y. Heat shock protein 90β inhibits apoptosis of intestinal epithelial cells induced by hypoxia through stabilizing phosphorylated Akt. BMB Rep 2013; 46:47-52. [PMID: 23351384 PMCID: PMC4133828 DOI: 10.5483/bmbrep.2013.46.1.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intestinal epithelial cell (IEC) apoptosis induced by hypoxia compromise intestinal epithelium barrier function. Both Akt and Hsp90 have cytoprotective function. However, the specific role
of Akt and Hsp90β in IEC apoptosis induced by hypoxia has not been explored. We confirmed that hypoxia-induced apoptosis was reduced by Hsp90β overexpression but enhanced by decreasing Hsp90β expression. Hsp90β overexpression enhanced BAD phosphorylation and thus reduced mitochondrial release of cytochrome C. Reducing Hsp90β expression had opposite effects. The protective effect of Hsp90β against apoptosis was negated by LY294002, an Akt inhibitor. Further study showed that Akt phosphorylation was enhanced by Hsp90β, which was not due to the activation of upstream PI3K and PDK1 but because of stabilization of pAkt via direct interaction between Hsp90β and pAkt. These results demonstrate that Hsp90β may play a significant role in protecting IECs from hypoxia-induced apoptosis via stabilizing pAkt to phosphorylate
BAD and reduce cytochrome C release. [BMB Reports 2013;46(1): 47-52]
Collapse
Affiliation(s)
- Shuai Zhang
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | |
Collapse
|
26
|
Kilic-Eren M, Boylu T, Tabor V. Targeting PI3K/Akt represses Hypoxia inducible factor-1α activation and sensitizes Rhabdomyosarcoma and Ewing's sarcoma cells for apoptosis. Cancer Cell Int 2013; 13:36. [PMID: 23590596 PMCID: PMC3637483 DOI: 10.1186/1475-2867-13-36] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 04/10/2013] [Indexed: 12/23/2022] Open
Abstract
Background Hypoxia inducible factor-1 α (HIF-1α) has been identified as an important novel target in apoptosis resistance of pediatric tumors such as Rhabdomyosarcoma (RMS) and Ewing’s sarcoma (ES). Evidence suggests that PI3K/Akt signaling plays a role in regulation of HIF-1α activation as well as apoptosis resistance in various adult tumors. However the relevance of PI3K/Akt signaling in HIF-1bα activation and apoptosis resistance in childhood tumors has not been addressed yet. Thus, this study was to investigate whether PI3K/Akt signaling is involved in hypoxia induced activation of HIF-1α as well as in resistance to hypoxia-induced apoptosis in childhood tumors such as RMS and ES. Methods Constitutive activation of PI3K/Akt signaling was analyzed by Western blotting. Hypoxic activation of HIF-1α was determined by Western Blot analysis and electrophoretic mobility shift assay. Apoptosis was determined by flow cytometric analysis of the propidium iodine stained nuclei of cells treated with PI3K inhibitor LY294002 in combination with either TNF-related apoptosis-inducing ligand (TRAIL) or doxorubicin. Results This study demonstrated that PI3K/Akt signaling was constitutively activated in RMS and ES cell lines, A204 and A673, respectively. Targeting PI3K/Akt signaling by the inhibitor LY294002 (30 μM) significantly decreased the protein expression as well as DNA binding activity of HIF-1α and restored the apoptosis-inducing ability of cells in hypoxia Additionally, pretreatment with LY294002 sensitized A204 and A673 cells to TRAIL or doxorubicin induced apoptosis under hypoxia. Conclusion These results suggest that the constitutively active PI3K/Akt signaling contributes to hypoxic activation of HIF-1α as well as HIF1α-mediated apoptosis resistance in RMS and ES cells under hypoxia.
Collapse
Affiliation(s)
- Mehtap Kilic-Eren
- Department of Medical Biology, Faculty of Medicine, Adnan Menderes University, Aydin, Turkey
| | - Tulin Boylu
- Department of Histology and Embryology, Faculty of Medicine, Adnan Menderes University, Aydin, Turkey
| | - Vedrana Tabor
- Present address: Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
27
|
Gordon LK, Kiyohara M, Fu M, Braun J, Dhawan P, Chan A, Goodglick L, Wadehra M. EMP2 regulates angiogenesis in endometrial cancer cells through induction of VEGF. Oncogene 2013; 32:5369-76. [PMID: 23334331 PMCID: PMC3898317 DOI: 10.1038/onc.2012.622] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 10/19/2012] [Accepted: 11/02/2012] [Indexed: 01/20/2023]
Abstract
Understanding tumor-induced angiogenesis is a challenging problem with important consequences for the diagnosis and treatment of cancer. In this study, we define a novel function for epithelial membrane protein-2 (EMP2) in the control of angiogenesis. EMP2 functions as an oncogene in endometrial cancer, and its expression has been linked to decreased survival. Using endometrial cancer xenografts, modulation of EMP2 expression resulted in profound changes to the tumor microvasculature. Under hypoxic conditions, upregulation of EMP2 promoted vascular endothelial growth factors (VEGF) expression through a HIF-1α-dependent pathway and resulted in successful capillary-like tube formation. In contrast, reduction of EMP2 correlated with reduced HIF-1α and VEGF expression with the net consequence of poorly vascularized tumors in vivo. We have previously shown that targeting of EMP2 using diabodies in endometrial cancer resulted in a reduction of tumor load, and since then we have constructed a fully human EMP2 IgG1. Treatment of endometrial cancer cells with EMP2-IgG1 reduced tumor load with a significant improvement in survival. These results support the role of EMP2 in the control of the tumor microenvironment and confirm the cytotoxic effects observed by EMP2 treatment in vivo.
Collapse
Affiliation(s)
- L K Gordon
- Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Sevoflurane postconditioning involves an up-regulation of HIF-1α and HO-1 expression via PI3K/Akt pathway in a rat model of focal cerebral ischemia. Brain Res 2012; 1463:63-74. [PMID: 22580326 DOI: 10.1016/j.brainres.2012.04.050] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/25/2012] [Accepted: 04/26/2012] [Indexed: 11/20/2022]
Abstract
Administration of sevoflurane at the onset of reperfusion has been confirmed to provide a cerebral protection. However, little is known about the mechanism. In this study, we tested the hypothesis that sevoflurane postconditioning induces neuroprotection through the up-regulation hypoxia inducible factor-1α (HIF-1α) and heme oxygenase-1 (HO-1) involving phosphatidylinositol-3-kinase (PI3K)/Akt pathway. In the first experiment, male Sprague-Dawley rats were subjected to focal cerebral ischemia. Postconditioning was performed by exposure to 2.5% sevoflurane immediately at the onset of reperfusion. The mRNA and protein expression of HIF-1α and its target gene, HO-1, intact neurons and the activity of caspase-3 was evaluated at 6, 24 and 72h after reperfusion. In the second experiment, we investigated the relationship between PI3K/Akt pathway and the expression of HIF-1α and HO-1 in the neuroprotection induced by sevoflurane. Cerebral infarct volume, apoptotic neuron and the expression of HIF-1α, HO-1 and p-Akt were evaluated at 24h after reperfusion. Compared with the control group, sevoflurane postconditiong significantly ameliorated neuronal injury, up-regulated mRNA and protein levels of HIF-1α and HO-1, inhibited the activity of caspase-3, and decreased the number of TUNEL-positive cells and infarct sizes. However, the selective PI3K inhibitor, wortmannin not only partly eliminated the neuroprotection of sevoflurane as shown by reducing infarct size and apoptotic neuronal cells, but also reversed the elevation of HIF-1α, HO-1 and p-Akt expression in the ischemic penumbra induced by sevoflurane. Therefore, our data demonstrate that the cerebral protection from sevoflurane postconditioning is partly mediated by PI3K/Akt pathway via the up-regulation of HIF-1α and HO-1.
Collapse
|
29
|
Conde E, Alegre L, Blanco-Sánchez I, Sáenz-Morales D, Aguado-Fraile E, Ponte B, Ramos E, Sáiz A, Jiménez C, Ordoñez A, López-Cabrera M, del Peso L, de Landázuri MO, Liaño F, Selgas R, Sanchez-Tomero JA, García-Bermejo ML. Hypoxia inducible factor 1-alpha (HIF-1 alpha) is induced during reperfusion after renal ischemia and is critical for proximal tubule cell survival. PLoS One 2012; 7:e33258. [PMID: 22432008 PMCID: PMC3303832 DOI: 10.1371/journal.pone.0033258] [Citation(s) in RCA: 130] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 02/06/2012] [Indexed: 01/17/2023] Open
Abstract
Acute tubular necrosis (ATN) caused by ischemia/reperfusion (I/R) during renal transplantation delays allograft function. Identification of factors that mediate protection and/or epithelium recovery could help to improve graft outcome. We studied the expression, regulation and role of hypoxia inducible factor 1-alpha (HIF-1 α), using in vitro and in vivo experimental models of I/R as well as human post-transplant renal biopsies. We found that HIF-1 α is stabilized in proximal tubule cells during ischemia and unexpectedly in late reperfusion, when oxygen tension is normal. Both inductions lead to gene expression in vitro and in vivo. In vitro interference of HIF-1 α promoted cell death and in vivo interference exacerbated tissue damage and renal dysfunction. In pos-transplant human biopsies, HIF-1 α was expressed only in proximal tubules which exhibited normal renal structure with a significant negative correlation with ATN grade. In summary, using experimental models and human biopsies, we identified a novel HIF-1 α induction during reperfusion with a potential critical role in renal transplant.
Collapse
Affiliation(s)
- Elisa Conde
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Laura Alegre
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
- Department of Nephrology, Instituto de Investigación La Princesa (IP), Madrid, Spain
| | - Ignacio Blanco-Sánchez
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - David Sáenz-Morales
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Elia Aguado-Fraile
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Belén Ponte
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Edurne Ramos
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Ana Sáiz
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Carlos Jiménez
- Department of Nephrology, Hospital La Paz (IdIPaz), Madrid, Spain
| | - Angel Ordoñez
- Department of Immunology, Instituto de Investigación La Princesa (IP), Madrid, Spain
| | | | - Luis del Peso
- HIV Unit, Department of Biochemistry, Hospital La Paz (IdiPAZ), Autónoma University School of Medicine, Institute of Biomedical Research Alberto Sols, CSIC-UAM, Madrid, Spain
| | | | - Fernando Liaño
- Department of Nephrology, Hospital Ramón y Cajal, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
| | - Rafael Selgas
- Department of Nephrology, Hospital La Paz (IdIPaz), Madrid, Spain
| | | | - María Laura García-Bermejo
- Department of System Disorders and Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Alcalá University, Madrid, Spain
- * E-mail:
| |
Collapse
|
30
|
Sood A, Salih S, Roh D, Lacharme-Lora L, Parry M, Hardiman B, Keehan R, Grummer R, Winterhager E, Gokhale PJ, Andrews PW, Abbott C, Forbes K, Westwood M, Aplin JD, Ingham E, Papageorgiou I, Berry M, Liu J, Dick AD, Garland RJ, Williams N, Singh R, Simon AK, Lewis M, Ham J, Roger L, Baird DM, Crompton LA, Caldwell MA, Swalwell H, Birch-Machin M, Lopez-Castejon G, Randall A, Lin H, Suleiman MS, Evans WH, Newson R, Case CP. Signalling of DNA damage and cytokines across cell barriers exposed to nanoparticles depends on barrier thickness. NATURE NANOTECHNOLOGY 2011; 6:824-33. [PMID: 22056725 DOI: 10.1038/nnano.2011.188] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Accepted: 09/28/2011] [Indexed: 05/28/2023]
Abstract
The use of nanoparticles in medicine is ever increasing, and it is important to understand their targeted and non-targeted effects. We have previously shown that nanoparticles can cause DNA damage to cells cultured below a cellular barrier without crossing this barrier. Here, we show that this indirect DNA damage depends on the thickness of the cellular barrier, and it is mediated by signalling through gap junction proteins following the generation of mitochondrial free radicals. Indirect damage was seen across both trophoblast and corneal barriers. Signalling, including cytokine release, occurred only across bilayer and multilayer barriers, but not across monolayer barriers. Indirect toxicity was also observed in mice and using ex vivo explants of the human placenta. If the importance of barrier thickness in signalling is a general feature for all types of barriers, our results may offer a principle with which to limit the adverse effects of nanoparticle exposure and offer new therapeutic approaches.
Collapse
Affiliation(s)
- A Sood
- Bristol Musculoskeletal Research Unit, Clinical Science at North Bristol University of Bristol, Avon Orthopaedic Centre, Southmead Hospital, Bristol
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Rapid non-genomic signalling by 17β-oestradiol through c-Src involves mTOR-dependent expression of HIF-1α in breast cancer cells. Br J Cancer 2011; 105:953-60. [PMID: 21897387 PMCID: PMC3185958 DOI: 10.1038/bjc.2011.349] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Hypoxia-inducible factor 1 (HIF1) has been implicated in regulating many of the genes responsible for angiogenesis, erythropoiesis, glucose metabolism and cancer pathogenesis. In this study, we demonstrate that exposure of human breast cancer lines to 17β-oestradiol (E2) rapidly induced the expression of HIF-1α, the regulated subunit of HIF1, in normoxic condition. Hypoxia-inducible factor-1α is normally degraded in normoxia through ubiquitination-mediated proteolysis, whereas hypoxia modulates HIF-1α level by inhibiting ubiquitination-mediated degradation. Methods: Oestradiol-induced accumulation of HIF-1α in breast cancer lines was detected by western blot analysis and its promoter activity was measured by HIF1 reporter assay. Molecular signalling of oestradiol-mediated HIF-1α expression was studied using specific pharmacological inhibitors and small interference RNA by co-immunoprecipitation and western blotting analysis. Results: Oestradiol has been observed to rapidly activate the nongenomic signalling cascade leading to HIF-1α protein synthesis. The results define a signalling pathway in breast cancer cells whereby oestradiol induces a rapid protein–protein interaction of ERα-c-Src-PI3K, resulting in the activation of PI3K/AKT pathway leading to mammalian target of rapamycin (mTOR) phosphorylation. The mTOR then stimulates translation by phosphorylating p70 S6 kinase and 4EB-P1, modulating HIF-1α protein synthesis. Oestradiol-stimulated HIF-1α activity was inhibited by either siRNA or pharmacological inhibitors to ERα, c-Src, PI3K and mTOR, providing a mechanism for the modulation of HIF-1α protein synthesis. Conclusion: These results show oestradiol-induced expression of HIF-1α, downstream of the ERα/c-Src/PI3K/AKT/mTOR pathway in human breast cancer cells.
Collapse
|
32
|
Human embryonic stem cell neural differentiation and enhanced cell survival promoted by hypoxic preconditioning. Cell Death Dis 2011; 1:e22. [PMID: 21364630 PMCID: PMC3032335 DOI: 10.1038/cddis.2009.22] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Transplantation of neural progenitors derived from human embryonic stem cells (hESCs) provides a potential therapy for ischemic stroke. However, poor graft survival within the host environment has hampered the benefits and applications of cell-based therapies. The present investigation tested a preconditioning strategy to enhance hESC tolerance, thereby improving graft survival and the therapeutic potential of hESC transplantation. UC06 hESCs underwent neural induction and terminal differentiation for up to 30 days, becoming neural lineage cells, exhibiting extensive neurites and axonal projections, generating synapses and action potentials. To induce a cytoprotective phenotype, hESC-derived neurospheres were cultured at 0.1% oxygen for 12 h, dissociated and plated for terminal differentiation under 21% oxygen. Immunocytochemistry and electrophysiology demonstrated the 'hypoxic preconditioning' promoted neuronal differentiation. Western blotting revealed significantly upregulated oxygen-sensitive transcription factors hypoxia-inducible factor (HIF)-1α and HIF-2α, while producing a biphasic response within HIF targets, including erythropoietin, vascular endothelial growth factor and Bcl-2 family members, during hypoxia and subsequent reoxygenation. This cytoprotective phenotype resulted in a 50% increase in both total and neural precursor cell survival after either hydrogen peroxide insult or oxygen-glucose deprivation. Cellular protection was maintained for at least 5 days and corresponded to upregulation of neuroprotective proteins. These results suggest that hypoxic preconditioning could be used to improve the effectiveness of human neural precursor transplantation therapies.
Collapse
|
33
|
Kato H, Seto K, Kobayashi N, Yoshinaga K, Meyer T, Takei M. CCK-2/gastrin receptor signaling pathway is significant for gemcitabine-induced gene expression of VEGF in pancreatic carcinoma cells. Life Sci 2011; 89:603-8. [PMID: 21839751 DOI: 10.1016/j.lfs.2011.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 07/15/2011] [Accepted: 07/21/2011] [Indexed: 12/24/2022]
Abstract
AIMS As activation and overexpression of the cholecystokinin-2 (CCK-2)/gastrin receptor can lead to carcinogenesis, it has been explored as a therapeutic target in pancreatic cancer. We demonstrated that Z-360, a CCK-2/gastrin receptor antagonist, combined with gemcitabine prolonged survival and reduced gemcitabine-induced vascular endothelial growth factor (VEGF) expression in a pancreatic carcinoma orthotopic xenograft mouse. In this study, we investigated the role of the CCK-2/gastrin signaling pathway on gemcitabine-induced VEGF expression in PANC-1 human pancreatic carcinoma cells. MAIN METHODS In PANC-1 cells treated with Z-360, anti-gastrin IgG or kinase inhibitors, the gene expression levels were analyzed by quantitative real-time RT-PCR, and the protein levels of Akt and phosphorylated Akt (p-Akt) in cellular extracts were measured by ELISA. KEY FINDINGS Gemcitabine-induced expression of VEGF and hypoxia-inducible factor-1 alpha (HIF-1 alpha) were suppressed by the treatment with an anti-gastrin antibody. In addition, VEGF and HIF-1 alpha gene expression was inhibited by treatment with an inhibitor of phosphatidylinositol 3-kinase (PI3K), which is involved in the downstream signaling pathway of the CCK-2/gastrin receptor, and was also suppressed by treatment with Z-360. Moreover, although Akt phosphorylation was increased by treatment with gemcitabine, this elevation was partially, but significantly, inhibited by an exposure of Z-360. SIGNIFICANCE Gemcitabine might induce gene expression of VEGF via the PI3K/Akt signaling pathway in the downstream of the CCK-2/gastrin receptor. The suppression of the CCK-2/gastrin signaling pathway by treatment with Z-360 could be a useful approach for potentiating prolonged survival of pancreatic cancer patients receiving gemcitabine therapy.
Collapse
Affiliation(s)
- Hiroki Kato
- Department of Clinical Research, Zeria Pharmaceutical Co., Ltd., Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Zhang L, Liu Q, Lu L, Zhao X, Gao X, Wang Y. Astragaloside IV stimulates angiogenesis and increases hypoxia-inducible factor-1α accumulation via phosphatidylinositol 3-kinase/Akt pathway. J Pharmacol Exp Ther 2011; 338:485-91. [PMID: 21576377 DOI: 10.1124/jpet.111.180992] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Astragaloside IV is the major active constituent of Astragalus membranaceus, which has been widely used for the treatment of cardiovascular diseases in China. The aim of this study was to determine the angiogenic effect of astragaloside IV and its underlying mechanism. We used the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay, Western blotting, real-time polymerase chain reaction, and immunofluorescence to detect the effect of astragaloside IV on proliferation of human umbilical vein endothelial cells (HUVECs), the phospho-Akt protein level, hypoxia-inducible factor-1α (HIF-1α) accumulation, vascular endothelial growth factor mRNA expression, and applied cell migration, tube formation, and chick chorioallantoic membrane assays to study the angiogenic effect of astragaloside IV. Results indicate that astragaloside IV promoted cell proliferation and stimulated HIF-1α accumulation during hypoxia. Mechanism studies revealed that astragaloside IV did not affect the degradation of HIF-1α protein or the level of HIF-1α mRNA. In contrast, astragaloside IV apparently activated the phosphatidylinositol 3-kinase (PI3K)/Akt pathway, which regulates HIF-1α protein synthesis. Moreover, astragaloside IV also stimulated cell migration, increased tube formation, and promoted angiogenesis in the chick chorioallantoic membrane assay. All angiogenic effects of astragaloside IV were reversed by the PI3K inhibitor. Taken together, our data collectively reveal that astragaloside IV is a novel regulator of HIF-1α and angiogenesis through the PI3K/Akt pathway in HUVECs that are exposed to hypoxia.
Collapse
Affiliation(s)
- Ling Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | | | | | | | | | | |
Collapse
|
35
|
Nakamura M, Kitagawa Y, Yamazaki Y, Hata H, Kotsuji M, Fujibayashi Y, Okazawa H, Yonekura Y, Sano K. Increased glucose metabolism by FDG-PET correlates with reduced tumor angiogenesis in oral squamous cell carcinoma. Odontology 2011; 100:87-94. [PMID: 21567121 DOI: 10.1007/s10266-011-0024-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 02/06/2011] [Indexed: 01/15/2023]
Abstract
Hypoxia is known to have been related with angiogenesis and glycolysis, and may have an influence on tumor treatment effect. Because glucose utilization is higher in malignant cells than that in normal cells, dynamic glucose metabolism of tumor has been evaluated by means of [(18)F]-fluorodeoxyglucose positron emission tomography (FDG-PET). To investigate the significance of tumor vascularization in oral squamous cell carcinoma, we compared tumor angiogenesis with the FDG-PET findings. Twenty patients underwent FDG-PET. For the quantitative evaluation of FDG uptake in each tumor, the mean standardized uptake value (SUV) was calculated. Microvessel structures labeled with CD34 antigen were investigated in pretreatment biopsy specimens. Using an image analyzer, we calculated the following microvessel parameters: the ratio of the total number of microvessels (TN) to tumor area (TA), the ratio of the total microvessel perimeter (TP) to the TA, and the ratio of the tumor tissue area more than 150 μm distant from each microvessel (hypoxic ratio, %). The SUV was compared with the above parameters. Simple regression analysis revealed a statistical significance between the SUV and the TN:TA ratio (p = 0.046), as well as between the SUV and the TP:TA ratio (p = 0.0206). The SUV was found to be inversely related to the TN:TA and TP:TA ratios. Elevated glucose metabolism assessed by FDG-PET correlated with reduced vascularization. Higher glucose metabolism might therefore reflect a state of hypoxia.
Collapse
Affiliation(s)
- Mikiko Nakamura
- Division of Dentistry and Oral Surgery, Department of Sensory and Locomotor Medicine, School of Medicine, University of Fukui, Fukui, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Otto Warburg pioneered quantitative investigations of cancer cell metabolism, as well as photosynthesis and respiration. Warburg and co-workers showed in the 1920s that, under aerobic conditions, tumour tissues metabolize approximately tenfold more glucose to lactate in a given time than normal tissues, a phenomenon known as the Warburg effect. However, this increase in aerobic glycolysis in cancer cells is often erroneously thought to occur instead of mitochondrial respiration and has been misinterpreted as evidence for damage to respiration instead of damage to the regulation of glycolysis. In fact, many cancers exhibit the Warburg effect while retaining mitochondrial respiration. We re-examine Warburg's observations in relation to the current concepts of cancer metabolism as being intimately linked to alterations of mitochondrial DNA, oncogenes and tumour suppressors, and thus readily exploitable for cancer therapy.
Collapse
Affiliation(s)
- Willem H Koppenol
- Institute of Inorganic Chemistry, Swiss Federal Institute of Technology, Zurich, Switzerland. koppenol@inorg. chem.ethz.ch
| | | | | |
Collapse
|
37
|
Lin HH, Tsai CW, Chou FP, Wang CJ, Hsuan SW, Wang CK, Chen JH. Andrographolide down-regulates hypoxia-inducible factor-1α in human non-small cell lung cancer A549 cells. Toxicol Appl Pharmacol 2011; 250:336-45. [DOI: 10.1016/j.taap.2010.11.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Revised: 11/16/2010] [Accepted: 11/25/2010] [Indexed: 12/27/2022]
|
38
|
Pistollato F, Rampazzo E, Persano L, Abbadi S, Frasson C, Denaro L, D’Avella D, Panchision DM, Puppa AD, Scienza R, Basso G. Interaction of hypoxia-inducible factor-1α and Notch signaling regulates medulloblastoma precursor proliferation and fate. Stem Cells 2010; 28:1918-29. [PMID: 20827750 PMCID: PMC3474900 DOI: 10.1002/stem.518] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Medulloblastoma (MDB) is the most common brain malignancy of childhood. It is currently thought that MDB arises from aberrantly functioning stem cells in the cerebellum that fail to maintain proper control of self-renewal. Additionally, it has been reported that MDB cells display higher endogenous Notch signaling activation, known to promote the survival and proliferation of neoplastic neural stem cells and to inhibit their differentiation. Although interaction between hypoxia-inducible factor-1α (HIF-1α) and Notch signaling is required to maintain normal neural precursors in an undifferentiated state, an interaction has not been identified in MDB. Here, we investigate whether hypoxia, through HIF-1α stabilization, modulates Notch1 signaling in primary MDB-derived cells. Our results indicate that MDB-derived precursor cells require hypoxic conditions for in vitro expansion, whereas acute exposure to 20% oxygen induces tumor cell differentiation and death through inhibition of Notch signaling. Importantly, stimulating Notch1 activation with its ligand Dll4 under hypoxic conditions leads to expansion of MDB-derived CD133(+) and nestin(+) precursors, suggesting a regulatory effect on stem cells. In contrast, MDB cells undergo neuronal differentiation when treated with γ-secretase inhibitor, which prevents Notch activation. These results suggest that hypoxia, by maintaining Notch1 in its active form, preserves MDB stem cell viability and expansion.
Collapse
Affiliation(s)
- Francesca Pistollato
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| | - Elena Rampazzo
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| | - Luca Persano
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| | - Sara Abbadi
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| | - Chiara Frasson
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| | - Luca Denaro
- Department of Neurosurgery, University of Padova, Padova, Italy
| | | | - David M. Panchision
- Division of Neuroscience and Basic Behavioral Science, National Institute of Mental Health, National Institutes of Health, Bethesda, MD
| | | | - Renato Scienza
- Department of Neurosurgery, University of Padova, Padova, Italy
| | - Giuseppe Basso
- SSD Clinical and Experimental Hematology, Department of Paediatrics, University of Padova, Padova, Italy
| |
Collapse
|
39
|
Lee KH. CaMKII Inhibitor KN-62 Blunts Tumor Response to Hypoxia by Inhibiting HIF-1α in Hepatoma Cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:331-6. [PMID: 21165333 DOI: 10.4196/kjpp.2010.14.5.331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 10/08/2010] [Accepted: 10/18/2010] [Indexed: 12/22/2022]
Abstract
In rapidly growing tumors, hypoxia commonly develops due to the imbalance between O(2) consumption and supply. Hypoxia Inducible Factor (HIF)-1α is a transcription factor responsible for tumor growth and angiogenesis in the hypoxic microenvironment; thus, its inhibition is regarded as a promising strategy for cancer therapy. Given that CamKII or PARP inhibitors are emerging anticancer agents, we investigated if they have the potential to be developed as new HIF-1α-targeting drugs. When treating various cancer cells with the inhibitors, we found that a CamKII inhibitor, KN-62, effectively suppressed HIF-1α specifically in hepatoma cells. To examine the effect of KN-62 on HIF-1α-driven gene expression, we analyzed the EPO-enhancer reporter activity and mRNA levels of HIF-1α downstream genes, such as EPO, LOX and CA9. Both the reporter activity and the mRNA expression were repressed by KN-62. We also found that KN-62 suppressed HIF-1α by impairing synthesis of HIF-1α protein. Based on these results, we propose that KN-62 is a candidate as a HIF-1α-targeting anticancer agent.
Collapse
Affiliation(s)
- Kyoung-Hwa Lee
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 110-799, Korea
| |
Collapse
|
40
|
Guo W, Yang Z, Xia Q, Liu J, Yu Y, Li J, Zuo Z, Zhang D, Li X, Shi X, Huang C. Arsenite stabilizes HIF-1α protein through p85α-mediated up-regulation of inducible Hsp70 protein expression. Cell Mol Life Sci 2010; 68:475-88. [PMID: 20835880 DOI: 10.1007/s00018-010-0459-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Revised: 06/09/2010] [Accepted: 07/06/2010] [Indexed: 01/21/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) has been reported to regulate over 100 gene expressions in response to hypoxia and other stress conditions. In the present study, we found that arsenite could induce HIF-1α protein accumulation in both mouse epidermal Cl41 cells and mouse embryonic fibroblasts (MEFs). Knockout of p85α, a regulatory subunit of PI-3K, in MEFs (p85α(-/-)) dramatically decreased the arsenite-induced HIF-1α accumulation, indicating that p85α is crucial for arsenite effects on the stabilization of HIF-1α protein. Our further studies suggest that arsenite could induce inducible Hsp70 expression, and transfection of inducible Hsp70 into p85α(-/-) MEFs could restore HIF-1α protein accumulation. Moreover, the results using EMSA and Supershift assays indicate that p85α is crucial for arsenite-induced activation of the heat-shock transcription factor 1 (HSF-1), which is responsible for transcription of inducible Hsp70. Taken together, p85α-mediated HIF-1α stabilization upon arsenite exposure is specifically through HSF-1 activation and subsequent up-regulation of the inducible Hsp70 expression.
Collapse
Affiliation(s)
- Wei Guo
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Pistollato F, Abbadi S, Rampazzo E, Viola G, Della Puppa A, Cavallini L, Frasson C, Persano L, Panchision DM, Basso G. Hypoxia and succinate antagonize 2-deoxyglucose effects on glioblastoma. Biochem Pharmacol 2010; 80:1517-27. [PMID: 20705058 DOI: 10.1016/j.bcp.2010.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 08/03/2010] [Accepted: 08/03/2010] [Indexed: 02/04/2023]
Abstract
Glioblastoma multiforme (GBM) are highly proliferative brain tumors characterized by a hypoxic microenvironment which controls GBM stem cell maintenance. Tumor hypoxia promotes also elevated glycolytic rate; thus, limiting glucose metabolism is a potential approach to inhibit tumor growth. Here we investigate the effects mediated by 2-deoxyglucose (2-DG), a glucose analogue, on primary GBM-derived cells maintained under hypoxia. Our results indicate that hypoxia protects GBM cells from the apoptotic effect elicited by 2-DG, which raises succinate dehydrogenase activity thus promoting succinate level decrease. As a consequence hypoxia inducible factor-1α (HIF-1α) degradation occurs and this induces GBM cells to acquire a neuronal committed phenotype. By adding succinate these effects are reverted, as succinate stabilizes HIF-1α and increases GBM stem cell fraction particularly under hypoxia, thus preserving the tumor stem cell niche. 2-DG inhibits anaerobic glycolysis altering GBM cell phenotype by forcing tumor cells into mitochondrial metabolism and by inducing differentiation.
Collapse
Affiliation(s)
- Francesca Pistollato
- Hemato-Oncology Laboratory, Department of Pediatrics, University of Padova, Via Giustiniani 3, Padova 35128, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Song G, Ouyang G, Mao Y, Ming Y, Bao S, Hu T. Osteopontin promotes gastric cancer metastasis by augmenting cell survival and invasion through Akt-mediated HIF-1alpha up-regulation and MMP9 activation. J Cell Mol Med 2010; 13:1706-1718. [PMID: 19602039 DOI: 10.1111/j.1582-4934.2008.00540.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Osteopontin (OPN) is a secreted, integrin-binding matrix phosphorylated glycoprotein. OPN has been shown to facilitate the progression and metastasis of malignancies and has prognostic value in several types of cancer, including gastric cancer. However, the functional mechanism of OPN mediated metastatic growth in gastric cancer remains unclear. Here, using multiple in vitro and in vivo models, we report that OPN strongly promoted the progression and metastasis of gastric cancer. Immunohistochemical staining revealed that OPN, matrix metalloproteinase (MMP)9 and hypoxia-inducible factor (HIF)-1alpha have statistically significant different expression patterns between well- and poorly differentiated tissue samples (P < 0.05). Correlations existed between OPN and MMP9, and between OPN and HIF-1 (r(1) = 0.872, p(1) < 0.01 and r(2) = 0.878, p(2) < 0.01). Furthermore, OPN dramatically increased colony formation and invasion of gastric cancer cells in vitro and promoted tumour growth and metastasis in vivo. In addition, OPN potently protected gastric cancer cells from serum depletion-induced apoptosis. Further study shows that OPN activated phosphoinositide 3-kinase/Akt survival pathway and up-regulated HIF-1alpha via binding to v3 integrins in gastric cancer cells. Moreover, we found that OPN could activate MMP9 and upregulate MMP2. Taken together, our results suggest that the survival-promoting function is crucial for OPN to promote the development of gastric cancer, and HIF-1 and MMP9 may play key roles during this process. Thus, targeting OPN and its related signalling network may develop an effective therapeutic approach for the management of gastric cancer.
Collapse
Affiliation(s)
- Gang Song
- Cancer Research Center, Xiamen University Medical College, Xiamen, China.,The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Gaoliang Ouyang
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yubin Mao
- Cancer Research Center, Xiamen University Medical College, Xiamen, China
| | - Yanlin Ming
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shideng Bao
- The Key Laboratory of Ministry of Education for Cell Biology and Tumor Cell Engineering, School of Life Sciences, Xiamen University, Xiamen, China
| | - Tianhui Hu
- Cancer Research Center, Xiamen University Medical College, Xiamen, China
| |
Collapse
|
43
|
Shin HW, Cho CH, Kim TY, Park JW. Sunitinib deregulates tumor adaptation to hypoxia by inhibiting HIF-1α synthesis in HT-29 colon cancer cells. Biochem Biophys Res Commun 2010; 398:205-11. [DOI: 10.1016/j.bbrc.2010.06.060] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 06/12/2010] [Indexed: 11/15/2022]
|
44
|
Dooms C, Verbeken E, Stroobants S, Vansteenkiste J. Biological correlates of the maximum 18-fluoro-2-deoxy-glucose uptake on positron emission tomography in non-small cell lung carcinoma after induction chemotherapy. J Thorac Oncol 2010; 4:1221-5. [PMID: 19641474 DOI: 10.1097/jto.0b013e3181ab0997] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The magnitude of change in quantitative 18-fluoro-2-deoxy-glucose (FDG) uptake in the primary tumor after induction chemotherapy (IC) strongly correlates with the histologic response. We aimed to investigate the mechanisms that drive persistent FDG uptake in non-small cell lung carcinoma treated with IC. PATIENTS AND METHODS Baseline and repeat positron emission tomography using 18-fluoro-2-deoxy-glucose after IC and completely resected primary tumor after IC was available in 30 patients. The surgical specimens were quantitatively analyzed for tumor architecture on hematoxylin and eosin staining. Immunohistochemical staining was assessed for microvessel density, markers of cell proliferation (Ki-67), hypoxia (HIF-1alpha), extracellular acidification (CAIX), and p-Akt. RESULTS The percentage viable tumor cells and Ki-67 length density after IC were significantly lower in metabolic responders compared with nonresponders. Hypoxia-related and acidity-related markers did not show a significant difference between metabolic responders and nonresponders. CONCLUSIONS Tumor response after IC is related to both a decline in the number of viable tumor cells and FDG consumption. Although surrogate markers for tumor hypoxia and acidity showed a trend for correlation with increased glycolysis in pretreated stage IIIA-N2 non-small cell lung carcinoma, these markers did not contribute to the recognition of metabolic responders versus nonresponders. We observed that only surrogate markers of cell proliferation correlate with a metabolic response after IC.
Collapse
Affiliation(s)
- Christophe Dooms
- Department of Pulmonology and Leuven Lung Cancer Group, University Hospital Gasthuisberg, Leuven, Belgium.
| | | | | | | |
Collapse
|
45
|
[mTOR inhibitors: temsirolimus and everolimus in the treatment of renal cell carcinoma]. Bull Cancer 2010; 97:45-51. [PMID: 20418203 DOI: 10.1684/bdc.2010.1069] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
mTOR signaling pathway (mammalian target of rapamycin) is a major pathway in cell physiology and malignant behavior implicated in cell growth, cell proliferation, cell metabolism, protein synthesis and angiogenesis. Temsirolimus has shown in a randomized phase III trial for patients with poor risk feature of metastatic renal cell carcinoma, a significant gain in overall survival compared to this obtained with alpha interferon (7.3 à 10.9 months; HR: 0.73; P < 0.0069). Everolimus has shown in a randomized phase III trial for patients with metastatic renal cell carcinoma having failed under VEGFR tyrosine kinase inhibitor a significant gain in progression-free survival compared to this obtained with placebo VEGFR (1,8 à 4,6 months; HR: 0.33; P < 0.001). Temsirolimus and everolimus are now part of the reference treatments in renal cell carcinoma. This paper is a review of these two drugs in this setting.
Collapse
|
46
|
Cho YS, Lee KH, Park JW. Pyrithione-zinc Prevents UVB-induced Epidermal Hyperplasia by Inducing HIF-1alpha. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2010; 14:91-7. [PMID: 20473380 DOI: 10.4196/kjpp.2010.14.2.91] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 03/24/2010] [Accepted: 04/10/2010] [Indexed: 11/15/2022]
Abstract
Epidermal keratinocytes overgrow in response to ultraviolet-B (UVB), which may be associated with skin photoaging and cancer development. Recently, we found that HIF-1alpha controls the keratinocyte cell cycle and thereby contributes to epidermal homeostasis. A further study demonstrated that HIF-1alpha is down-regulated by UVB and that this process is involved in UVB-induced skin hyperplasia. Therefore, we hypothesized that the forced expression of HIF-1alpha in keratinocytes would prevent UVB-induced keratinocyte overgrowth. Among several agents known to induce HIF-1alpha, pyrithione-zinc (Py-Zn) overcame the UVB suppression of HIF-1alpha in cultured keratinocytes. Mechanistically, Py-Zn blocked the degradation of HIF-1alpha protein in keratinocytes, while it did not affect the synthesis of HIF-1alpha. Moreover, the p21 cell cycle inhibitor was down-regulated after UVB exposure, but was robustly induced by Py-Zn. In mice repeatedly irradiated with UVB, the epidermis became hyperplastic and HIF-1alpha disappeared from nuclei of epidermal keratinocytes. However, a cream containing Py-Zn effectively prevented the skin thickening and up-regulated HIF-1alpha to the normal level. These results suggest that Py-Zn is a potential agent to prevent UVB-induced photoaging and skin cancer development. This work also provides insight into a molecular target for treatment of UVB-induced skin diseases.
Collapse
Affiliation(s)
- Young-Suk Cho
- Department of Pharmacology, Chungbuk National University College of Medicine, Cheongju 361-763, Korea
| | | | | |
Collapse
|
47
|
Kirkali Z, Tüzel E. Systemic therapy of kidney cancer: tyrosine kinase inhibitors, antiangiogenesis or IL-2? Future Oncol 2010; 5:871-88. [PMID: 19663736 DOI: 10.2217/fon.09.51] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The von Hippel-Lindau (VHL) tumor suppressor gene is mutated in at least 50% of sporadic clear-cell renal cell carcinoma (RCC). This leads to a pseudohypoxic state in which the pVHL complex does not degrade hypoxia-inducible factor. Subsequent intracellular hypoxia-inducible factor accumulation results in increased production of growth factors such as VEGF, responsible for angiogenesis, tumor proliferation and mitogenesis. Recently, a number of strategies have been designed to specifically target these pathways. The VEGF, its related receptor and the mammalian target of rapamycin (mTOR) signal transduction pathway, in particular, have been utilized as therapeutic targets. Clinical trials have demonstrated the survival benefit of these agents, particularly in clear-cell RCC. Today, sunitinib is recommended as first-line therapy for intermediate- or low-risk patients with metastatic RCC. Sorafenib is advised as second-line therapy, whereas temsirolimus is generally recommended as first-line treatment in high-risk patients. Everolimus is the new standard following sunitinib. High-risk patients appeared to benefit less than low-risk patients from bevacizumab plus IFN-alpha therapy. High-dose IL-2 has been proven effective in prolonging progression-free survival or overall survival, depending on risk group selection criteria. Although novel agents show a consistent effect as measured by objective response, no currently available data demonstrate that these agents will cure any patient.
Collapse
Affiliation(s)
- Ziya Kirkali
- Professor of Urology, Dokuz Eylul University School of Medicine, Department of Urology, Inciralti, Izmir, Turkey.
| | | |
Collapse
|
48
|
Ward CS, Venkatesh HS, Chaumeil MM, Brandes AH, Vancriekinge M, Dafni H, Sukumar S, Nelson SJ, Vigneron DB, Kurhanewicz J, James CD, Haas-Kogan DA, Ronen SM. Noninvasive detection of target modulation following phosphatidylinositol 3-kinase inhibition using hyperpolarized 13C magnetic resonance spectroscopy. Cancer Res 2010; 70:1296-305. [PMID: 20145128 PMCID: PMC2822895 DOI: 10.1158/0008-5472.can-09-2251] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Numerous mechanism-based anticancer drugs that target the phosphatidylinositol 3-kinase (PI3K) pathway are in clinical trials. However, it remains challenging to assess responses by traditional imaging methods. Here, we show for the first time the efficacy of hyperpolarized (13)C magnetic resonance spectroscopy (MRS) in detecting the effect of PI3K inhibition by monitoring hyperpolarized [1-(13)C]lactate levels produced from hyperpolarized [1-(13)C]pyruvate through lactate dehydrogenase (LDH) activity. In GS-2 glioblastoma cells, PI3K inhibition by LY294002 or everolimus caused hyperpolarized lactate to drop to 42 +/- 12% and to 76 +/- 5%, respectively. In MDA-MB-231 breast cancer cells, hyperpolarized lactate dropped to 71 +/- 15% after treatment with LY294002. These reductions were correlated with reductions in LDH activity to 48 +/- 4%, 63 +/- 4%, and 69 +/- 12%, respectively, and were associated with a drop in levels of LDHA mRNA and LDHA and hypoxia-inducible factor-1alpha proteins. Supporting these findings, tumor growth inhibition achieved by everolimus in murine GS-2 xenografts was associated with a drop in the hyperpolarized lactate-to-pyruvate ratio detected by in vivo MRS imaging, whereas an increase in this ratio occurred with tumor growth in control animals. Taken together, our findings illustrate the application of hyperpolarized (13)C MRS of pyruvate to monitor alterations in LDHA activity and expression caused by PI3K pathway inhibition, showing the potential of this method for noninvasive imaging of drug target modulation.
Collapse
Affiliation(s)
- Christopher S Ward
- Department of Radiology and Biomedical Imaging , University of California at San Francisco, San Francisco, California 94158, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Solid tumors usually occur and progress in a hypoxic environment, suggesting that tumor cells are resistant to apoptosis and are associated to an increased angiogenesis, which makes them more aggressive, with invasive capacity and resistant to treatment. The genetic and biological mechanisms underlying this phenomenon are still unclear, but many studies suggest a role of HIF in this process. Under hypoxic conditions, the alpha subunit is not destroyed, and will activate transcription of a set of genes contributing to tumor aggressiveness. Its expression is associated to an increased metastatic potential that has been shown in both animal studies and human tumors. Tumor hypoxia has emerged as a key factor in tumor progression and is associated to a poor prognosis, particularly in kidney and prostate tumors. The purpose of this study was to review the significance of hypoxia in carcinogenesis and tumor progression by reviewing the current knowledge on the subject and the mechanisms of action and activation of HIF-1a Tumor hypoxia has emerged as a key factor in tumor progression and is associated to a poor prognosis, particularly in kidney and prostate tumors. The purpose of this study was to review the significance of hypoxia in carcinogenesis and tumor progression by reviewing the current knowledge on the subject and the mechanisms of action and activation of HIF-1a.
Collapse
|
50
|
Fijalkowska I, Xu W, Comhair SAA, Janocha AJ, Mavrakis LA, Krishnamachary B, Zhen L, Mao T, Richter A, Erzurum SC, Tuder RM. Hypoxia inducible-factor1alpha regulates the metabolic shift of pulmonary hypertensive endothelial cells. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1130-8. [PMID: 20110409 DOI: 10.2353/ajpath.2010.090832] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Severe pulmonary hypertension is irreversible and often fatal. Abnormal proliferation and resistance to apoptosis of endothelial cells (ECs) and hypertrophy of smooth muscle cells in this disease are linked to decreased mitochondria and preferential energy generation by glycolysis. We hypothesized this metabolic shift of pulmonary hypertensive ECs is due to greater hypoxia inducible-factor1alpha (HIF-1alpha) expression caused by low levels of nitric oxide combined with low superoxide dismutase activity. We show that cultured ECs from patients with idiopathic pulmonary arterial hypertension (IPAH-ECs) have greater HIF-1alpha expression and transcriptional activity than controls under normoxia or hypoxia, and pulmonary arteries from affected patients have increased expression of HIF-1alpha and its target carbonic anhydrase IX. Decreased expression of manganese superoxide dismutase (MnSOD) in IPAH-ECs paralleled increased HIF-1alpha levels and small interfering (SI) RNA knockdown of MnSOD, but not of the copper-zinc SOD, increased HIF-1 protein expression and hypoxia response element (HRE)-driven luciferase activity in normoxic ECs. MnSOD siRNA also reduced nitric oxide production in supernatants of IPAH-ECs. Conversely, low levels of a nitric oxide donor reduced HIF-1alpha expression in normoxic IPAH-ECs. Finally, mitochondria numbers increased in IPAH-ECs with knockdown of HIF-1alpha. These findings indicate that alterations of nitric oxide and MnSOD contribute to pathological HIF-1alpha expression and account for lower numbers of mitochondria in IPAH-ECs.
Collapse
Affiliation(s)
- Iwona Fijalkowska
- Department of Pathology, Johns Hopkins Univesity School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|